User login
Estimating Minimally Important Differences for the Worst Pain Rating of the Brief Pain Inventory–Short Form
Original research
Susan D. Mathias MPH
Abstract
The Brief Pain Inventory–Short Form (BPI-SF) is widely used for assessing pain in clinical and research studies. The worst pain rating is often the primary outcome of interest; yet, no published data are available on its minimally important difference (MID). Breast cancer patients with bone metastases enrolled in a randomized, double-blind, phase III study comparing denosumab with zoledronic acid for preventing skeletal related events and completed the BPI-SF, FACT-B, and EQ-5D at baseline, week 5, and monthly through the end of the study. Anchor- and distribution-based MID estimates were computed. Data from 1,564 patients were available. Spearman correlation coefficients for anchors ranged from 0.33–0.65. Mean change scores for worst pain ratings corresponding to one-category improvement in each anchor were 0.26–1.04 for BPI-SF current pain, −1.40 to −2.42 for EQ-5D Index score, 1.71–1.98 for EQ-5D Pain item, −2.22 to −0.51 for FACT-B TOI, −1.61 to −0.16 for FACT-G Physical, and −1.31 to −0.12 for FACT-G total. Distribution-based results were 1 SEM = 1.6, 0.5 effect size = 1.4, and Guyatt's statistic = 1.4. Combining anchor- and distribution-based results yielded a two-point MID estimate. An MID estimate of two points is useful for interpreting how much change in worst pain is considered clinically meaningful.
Article Outline
- Methods
- Study Design
- Outcome Measures and Assessment Intervals
- Anchor-Based Analysis
- Distribution-Based Analysis
- Integrating Anchor-Based and Distribution-Based Mid Estimates
The MID may be estimated through distribution-based methods and/or anchor-based methods. Distribution-based methods are based on the distribution of the data. Examples of distribution-based methods include effect size measures, the standard error of measurement (SEM), one-half times the standard deviation, and the responsiveness index.[2] and [3] Anchor-based methods are based on the association between the PRO measure and an interpretable external measure, such as a global rating of change or a response to treatment. These methods may result in somewhat different estimates, and no particular estimate is considered the most valid.[2], [3] and [4] Therefore, researchers are encouraged to use more than one method and to present a range of MID estimates.
A frequently used PRO measure for the assessment of pain is the Brief Pain Inventory–Short Form (BPI-SF). The foundation of the BPI-SF is the Wisconsin Brief Pain Questionnaire, which was developed over 25 years ago based on interviews with cancer patients, expert opinion, and then-current psychometric standards.5 Over time, the Wisconsin Brief Pain Questionnaire evolved into the Brief Pain Inventory, which was later reduced to a shorter version, the BPI-SF. Today, the BPI-SF is the standard for clinical and research use. It has been used in over 400 studies, including psychometric evaluations and clinical applications with a wide range of conditions (e.g., cancer pain, fibromyalgia, neuropathic pain, and joint diseases).6
The BPI-SF includes two domains: pain severity and pain interference. The pain severity domain, the focus of this report, includes items specific to pain at “worst,” “least,” “average,” and “now” (current pain), with a numerical response scale ranging from 0 (no pain) to 10 (pain as bad as you can imagine). In clinical trials, the worst pain item has been used alone as a measure of pain severity.6 Its use as a single item is supported by a consensus panel on outcome measures for chronic pain clinical trials.7 In addition, the Food and Drug Administration's (FDA) guidance on PROs states that a single-item PRO measure of pain severity is appropriate for assessing the effect of a treatment on pain.8 Although extensive psychometric evaluation of the BPI-SF has been conducted, no estimates of the MID are available for the BPI-SF worst pain item. Establishing the MID for the BPI-SF worst pain item is important because it will provide a clinically relevant reference to interpret changes in pain scores. Therefore, the objective of this current report was to estimate the MID of the worst pain item of the BPI-SF.
Methods
Study Design
Patients with advanced breast cancer and bone metastases were enrolled in an international, randomized, double-blind, double-dummy, active-controlled phase III study comparing denosumab with zoledronic acid for delaying or preventing skeletal related events. Patients were eligible to participate if they had histologically or cytologically confirmed breast adenocarcinoma; current or prior radiologic, computed tomography, or magnetic resonance imaging evidence of at least one bone metastasis; and an Eastern Cooperative Oncology Group (ECOG) performance status of 0, 1, or 2. Patients with current or prior intravenous bisphosphonate administration were excluded. Patients completed PRO assessments, including the BPI-SF, at baseline, week 5, and every 4 weeks thereafter until the end of the study. Assessments were scheduled to take place prior to any study procedures and prior to study drug administration. Although data collection continued, PRO analyses for efficacy were truncated when approximately 30% of patients dropped out of the study due to death, disease progression, or withdrawn consent.
Outcome Measures and Assessment Intervals
A number of outcome measures were assessed in the study and considered for use as anchors for evaluating the MID of the BPI-SF worst pain item, including one clinician-reported measure (ECOG Performance Status) and several PRO measures: the EuroQoL 5 Dimensions (EQ-5D) Index score, the Functional Assessment of Cancer Therapy-Breast Cancer (FACT-B), and the BPI-SF current pain rating.
The ECOG Performance Status, which assesses how a patient's disease or its treatment is progressing and how the disease affects the daily living abilities of the patient, is a single-item, six-point, clinician-rated assessment of performance ranging from 0 (fully active, no restrictions) to 5 (dead).9 The EQ-5D Index score is a measure of health status, which assesses five dimensions: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Each dimension is comprised of three response options: no problems, some/moderate problems, and extreme problems. Responses are converted to a weighted health state index, with scores ranging from −0.594 (worst health) to 1.0 (full health). The single item on pain from the EQ-5D was also evaluated separately as an anchor. The FACT-B includes the four core FACT-General (FACT-G) dimensions of physical well-being, social/family well-being, emotional well-being, and functional well-being, for which scale scores and a total score can be computed. In addition, the FACT-B includes a breast cancer–specific subscale.10 The FACT-B Trial Outcome Index (TOI) is the sum of the physical well-being score, the functional well-being score, and the breast cancer subscale. The four FACT-G scale scores, the FACT-G total score, the FACT-B TOI, and a single-item overall quality-of-life (QOL) rating from the functional well-being section were all evaluated as potential anchors. The single-item overall QOL item from the functional well-being scale was selected to balance out the single item on pain that was selected from the EQ-5D, by serving as a more general potential anchor in breadth and scope. For all of these FACT outcome measures, a higher score indicates better health-related QOL. Finally, the current pain rating from the BPI-SF, ranging from 0 (no pain) to 10 (pain as bad as you can imagine), was also considered as an anchor because it was hypothesized to be highly correlated with the worst pain rating and because it would assist in understanding the behavior of other potential anchors.
Several assessment intervals were considered for evaluation of the MID for the BPI-SF worst pain item: baseline to week 5, baseline to week 13, and baseline to week 25. The analysis for each time interval included only those patients with complete baseline and end-of-interval (i.e., week 5, week 13, or week 25) assessments on the BPI-SF worst pain item and the relevant anchor of interest. In addition, a post hoc confirmatory analysis was conducted using a longer interval of time, from baseline to week 49. No imputation of missing data was performed. Analysis was performed on pooled data, regardless of treatment assignment.
Anchor-Based Analysis
The usefulness of an anchor depends on the correlation of the PRO change score and the anchor.11 Therefore, to select the most appropriate anchors and time interval for estimating the MID for the BPI-SF worst pain item, Spearman correlation coefficients were calculated between changes in the BPI-SF worst pain rating and changes in potential anchors across each of the potential time intervals. The time interval with the highest correlations and the anchors with statistically significant (P < 0.05) a priori specified correlations above 0.30 were selected for inclusion in the MID analysis.12
A one-category change was defined as a one-point change for the BPI-SF current pain item, a one-point change for the EQ-5D pain item, a three-point change for the FACT-G Physical Well-Being scale,13 a six-point change for the FACT-G total and FACT-B TOI scores,14 and a 0.20 change for the EQ-5D Index score. For the selected interval and anchors, the mean change in BPI-SF worst pain item that corresponds to a one-category increase and decrease in each anchor was calculated. In addition, ordinary least squares regression models were used to regress changes in BPI-SF worst pain ratings on changes of each of the anchors.[15] and [16] The regression models included main effects for change in each anchor and an interaction term expressing the change in anchor-by-baseline anchor.
Distribution-Based Analysis
The following distribution-based measures were calculated for the BPI-SF worst pain item: (1) the SEM, (2) effect size (Cohen's d), and (3) Guyatt's statistic. The SEM is a measure of the precision of a test instrument. It is calculated on the basis of sample data using the sample standard deviation and the sample reliability coefficient. While the standard deviation and the reliability of a measure are sample-dependent, their relationship (and hence the SEM) remains relatively constant across samples. Therefore, the SEM is considered to be an attribute of the measure and not a characteristic of the sample per se.17 Threshold values of 1 SEM have been suggested for defining clinically meaningful differences.18 The reliability coefficient was estimated for the BPI-SF worst pain item by calculating the intraclass correlation coefficients (ICCs) using two intervals of time. One used 7 days (days 1–8), a more typical interval for assessing reproducibility, while the other approach used a later interval, from week 105 to week 109. (Note: The 1-month interval was dictated by the schedule of assessments.) For both ICC values, only those patients whose FACT-B overall QOL ratings changed by 10% or less during the respective intervals were included. The 10% criterion was selected after reviewing the full distribution of change scores and their associated sample sizes, to arrive at a reasonable sample size of approximately 100 subjects.
Cohen's d, alternatively referred to as the “standardized effect size,” is calculated by dividing the difference between the baseline and week-25 scores by the standard deviation at baseline.19 The effect size represents individual change in terms of the number of baseline standard deviations. A value of 0.20 is a small effect, 0.50 is a medium effect, and 0.80 is a large effect. Effect sizes of 0.20, 0.50, and 0.80 were calculated in this study.
Guyatt's statistic, also referred to as the “responsiveness statistic,” is calculated by dividing the difference between baseline and week-25 change by the standard deviation of change observed for a group of stable patients.20 The denominator of the responsiveness statistics adjusts for spurious change due to measurement error. Values of 0.20 and 0.50 have been used to represent “small” and “medium” changes, respectively.21 Values representing 0.20 and 0.50 were calculated in this study. Stable patients were defined as those whose ECOG Performance rating did not change during the assessment interval. A different variable was used in defining the stable population for purposes of calculating the SEM and Guyatt's statistic because both variables were not consistently collected on the same schedule of assessments.
Integrating Anchor-Based and Distribution-Based Mid Estimates
The minimal detectable change (MDC) for the worst pain item was established by comparing distribution-based estimates. The MDC represents the smallest change that can be reliably distinguished from random fluctuation and, thus, the lower bound for establishing the MID.11 If the MID were lower than the MDC, then the instrument would not be capable of distinguishing the MID. The SEM was considered the primary distribution-based estimate because it takes into account the reliability of the measure and, thus, estimates the precision of the instrument.11 Other distribution-based measures were also considered in establishing the MDC. Standardized effect size was considered a secondary distribution-based estimate because of its reliance on interperson variability, which is generally higher and less consistent than intraperson variability. Anchor-based estimates of the MID range were then compared. A final MID range was established that is greater than the MDC and integrates estimates from the various anchors.
Results
Patient Population
Demographic and clinical characteristics for patients included in the baseline to week 25 interval are presented in Table 1. Data from 1,564 of 2,049 patients who participated in the study and had valid (i.e., nonmissing) baseline and end-of-interval scores for the BPI-SF and anchors were used in these analyses. Patients were predominantly female with an average age of 57.2 ± 11.2 years. The majority of patients were white (80.9%). Average pain scores at baseline were 2.45 ± 2.51, with a full range of scores (0–10) being used. Clinical results from the study have been presented previously.22
CHARACTERISTIC, n (%) | STUDY SAMPLE (n = 1,564) |
---|---|
Gender | |
Female | 1,550 (99.1) |
Male | 14 (0.9) |
Age, mean years ± SD (range) | 57.2 ± 11.2 (27.1–91.2) |
Race | |
White | 1,265 (80.9) |
Black | 38 (2.4) |
Hispanic | 92 (5.9) |
Japanese | 119 (7.6) |
Asian | 28 (1.8) |
Other | 22 (1.4) |
Demographic characteristics including the breakdown by gender, age, and race for the study sample are shown.
Anchor-Based Analysis
Spearman correlations between changes in the BPI-SF worst pain item and changes in potential anchors are presented in Table 2. For all potential anchors, the highest correlations with the BPI-SF worst pain rating were obtained at the baseline to week 25 interval. All potential anchors correlated significantly (P < 0.001) with the BPI-SF worst pain rating with the exception of the FACT-G Social/Family Well-Being scale. However, correlations were low (<0.30) for several potential anchors: ECOG Performance Status, FACT-B Overall QOL item, FACT-G Emotional Well-Being, and FACT-G Functional Well-Being. Therefore, the week 25 interval and the following anchors were selected for the MID analysis: BPI-SF current pain rating, EQ-5D Index score, EQ-5D Pain item, FACT-B TOI, FACT-G Physical Well-Being, and FACT-G total score. Correlation coefficients between the changes in the selected anchors and changes in the BPI-SF worst pain ratings range from 0.329–0.647.
Bolded correlations represent the highest correlations with anchors where correlation r ≥ 0.300.
Spearman correlation coefficients between changes in BPI-SF worst pain rating and changes in each of the 11 potential anchors that were considered are provided. The data are displayed for three intervals of time including baseline to week 5, baseline to week 13, and baseline to week 25. Using a cut point of r ≥ 0.300, only those correlations that are bolded meet the criteria of acceptability.
Mean changes in the BPI-SF worst pain rating that correspond to a one-category change in anchors from baseline to week 25 are presented in Table 3. BPI-SF current pain ratings >5 and EQ-5D Index scores <0.40 were excluded from their respective analysis due to small sample sizes. A one-category increase in the anchor scores was associated with an absolute value of change in the BPI-SF worst pain item ranging from 0.26–2.42. A one-category decrease in the anchor score was associated with an absolute value of change in the BPI-SF worst pain item ranging from 0.56–3.16. Changes associated with improvement and worsening in anchors were not symmetrical, nor was there a consistent trend across anchors. For example, for the EQ-5D pain item, the magnitude of change in BPI-SF worst pain was greater for a one-category increase in the anchor than for a one-category decrease in the anchor. In contrast, for the EQ-5D Index score, the magnitude of change in BPI-SF worst pain was greater for a one-category decrease in the anchor than for a one-category increase in the anchor.
ANCHOR | ONE CATEGORYA INCREASE IN ANCHOR | ONE CATEGORY DECREASE IN ANCHOR |
---|---|---|
BPI-SF Current Pain rating | 0.26–1.04 | −0.89 to −1.66 |
EQ 5D Index score | −2.42 to −1.40 | 0.56–1.63 |
EQ 5D Pain item | 1.71–1.98 | −3.16 to −2.56 |
FACT-B TOI | −2.22 to −0.51 | −0.56 to 0.77 |
FACT-G Physical Well-Being | −1.61 to −0.16 | −0.79 to 0.46 |
FACT-G total | −1.31 to −0.12 | −0.97 to 0.57 |
The range of mean changes in BPI-SF worst pain ratings (using the interval from baseline to week 25) for the six anchors that met the correlation criteria in Table 2 are provided. Mean changes are displayed for one-category increases and one-category decreases in anchor.
a One category (increase or decrease) represents 0.20 points for EQ-5D Index score, one point for BPI-SF current pain rating and EQ-5D pain item, three points for FACT-G Physical Well-Being, and six points for FACT-G total and FACT-B TOI.
The regression of changes in anchors on changes in the BPI-SF worst pain item is shown in Table 4. Changes in each anchor are significantly (P < 0.05) associated with changes in BPI-SF worst pain rating. A one-point increase in BPI-SF current pain rating and EQ-5D Pain item is associated with a 0.817 and 1.805 increase in BPI-SF worst pain, respectively, while a one-point increase in EQ-5D Index score, FACT-B TOI, FACT-G Physical Well-Being, and FACT-G total is associated with a 3.548, 0.098, 0.163, and 0.048 decrease in BPI-SF worst pain rating, respectively. Likewise, a two-point increase in BPI-SF current pain rating and EQ-5D Pain item is associated with a 1.634 and 3.610 increase in BPI-SF worst pain, respectively, while a two-point increase in EQ-5D Index score, FACT-B TOI, FACT-G Physical Well-Being, and FACT-G total is associated with a 7.096, 0.196, 0.326, and 0.096 decrease in BPI-SF worst pain rating, respectively. The change in anchor-by-baseline anchor interaction was statistically significant only for BPI current pain and FACT-G Physical Well-Being. The interaction tests whether the anchor–BPI-SF slope differs as a function of baseline anchor score; therefore, a lack of significance suggests that the association between BPI-SF worst pain and other anchors does not differ by baseline anchor rating.
VARIABLE | PREDICTOR | b | β | SIG. |
---|---|---|---|---|
Change in BPI current pain | Main effect | 0.817 | 0.724 | <0.001 |
Interaction with baseline anchor | −0.024 | −0.107 | 0.001 | |
Change in EQ-5D Health State Index | Main effect | −3.548 | −0.349 | <0.001 |
Interaction with baseline anchor | 0.220 | 0.021 | 0.465 | |
Change in EQ-5D Pain item | Main effect | 1.805 | 0.352 | <0.001 |
Interaction with baseline anchor | 0.207 | 0.080 | 0.261 | |
Change in FACT-B TOI | Main effect | −0.098 | −0.406 | <0.001 |
Interaction with baseline anchor | 0.000 | 0.028 | 0.756 | |
Change in FACT-G Physical Well-Being | Main effect | −0.163 | −0.321 | <0.001 |
Interaction with baseline anchor | −0.004 | −0.133 | 0.024 | |
Change in FACT-G total score | Main effect | −0.048 | −0.231 | 0.025 |
Interaction with baseline anchor | 0.000 | −0.130 | 0.209 |
b, regression coefficient; β, standardized regression coefficient; Sig., significance level.
Possible ranges: BPI Pain Right Now 0 (least) to 10 (most), EQ-5D Health State Index scores −0.594 (worst) to 1.00 (best), EQ-5D Pain item scores 1 (none) to 3 (severe), FACT-B TOI scores 4 (worst) to 92 (best), FACT-G Physical Well-Being scores 0 (worst) to 28 (best), FACT-G total score 8 (worst) to 108 (best), BPI Worst Pain item 0 (least) to 10 (most).
Changes in all anchors are significantly (P < 0.05) associated with changes in BPI-SF worst pain ratings. A one-point increase in BPI-SF current pain rating and EQ-5D pain item is associated with increases (positive b score) in the BPI-SF worst pain rating, and a one-point increase in EQ-5D Index, FACT-B TOI, FACT-G Physical Well-Being, and FACT-G total scores is associated with decreases (negative b score) in the BPI-SF worst pain ratings. The change in anchor-by-baseline anchor interaction was statistically significant only for the BPI current pain and FACT-G PWB items.
A post hoc confirmatory analysis was done replicating these analyses using data from the baseline to week 49 interval (n = 1,250). Results indicate a slightly stronger correlation between the anchors and the change scores. (Spearman's correlations range from 0.372 for FACT-TOI to 0.644 for BPI-SF current pain rating.) Mean change scores of BPI-SF worst pain ratings by each of the six anchors and regression coefficients were similar to those for the baseline to week 25 interval. For instance, mean change scores for the EQ-5D Pain item for stable patients ranged from 0.25–0.56, 1.58–295 for an improvement of one category, and 1.75–2.80 for a worsening of one category compared with 0.50–0.51, 1.71–1.98, and 2.56–3.16, respectively, for the baseline to week 25 interval.
Distribution-Based Analysis
The distribution-based estimates for the BPI-SF worst pain rating are presented in Table 5. There appears to be consistency with the 1 SEM estimates, the 0.50 effect size, and the 0.50 Guyatt's statistic.
The results from the three distribution-based approaches presented in this table will be combined with those of the anchor-based results to estimate the MID.
a The standard error of measurement is a measure of the precision of a test instrument. It is calculated on the basis of sample data using the sample standard deviation and the sample reliability coefficient. Intraclass correlation coefficients (ICCs) for BPI-SF worst pain rating from day 1 to day 8 and week 105 to week 109 in patients whose FACT-B overall QOL ratings change by <10% are 0.685 (n = 926) and 0.800 (n = 109), respectively.b Alternatively referred to as Cohen's d, the effect size is calculated by dividing the difference between the pretest and posttest scores by the standard deviation at pretest. The standard deviation of BPI-SF worst pain rating at baseline (n = 1,877) is 2.849.c Alternatively referred to as the responsiveness statistic, Guyatt's statistic is calculated by dividing the difference between pretest and posttest changes by the standard deviation of change observed for a group of stable patients. The standard deviation of change in BPI-SF worst pain rating from baseline to week 25 in patients whose ECOG performance rating does not change (n = 1,120) is 2.833.
Integrating Anchor-Based and Distribution-Based Mid Estimates
The distribution-based analyses suggest that the MDC for the worst pain rating, defined as the smallest change that can be reliably differentiated from random fluctuation, is between 1.3 and 1.6 points (see Table 5). This represents the lower bound for establishing the MID.
The results from regression analyses can be used to translate changes between anchors and corresponding changes in BPI-SF worst pain. This strategy can be particularly informative when the MID for an anchor is known. This is the case for the EQ-5D Health State Index, where the MID has been estimated at 0.06 for U.S. Index scores and 0.07 for U.K. Index scores.23 A one-point change in EQ-5D Index translates to a change of −3.548 in BPI-SF worst pain, so a 0.07-point change in EQ-5D Index (the MID for the measure) corresponds to a change of −0.248 in BPI-SF worst pain. In contrast, a one-point change in BPI-SF worst pain (which is smaller than the MID based upon the distribution-based analyses) translates to a change of 0.036 for the EQ-5D Index score (considerably smaller than the MID of 0.07). However, a two-point change in BPI-SF worst pain rating corresponds to a 0.072 change in EQ-5D Index score, which is almost identical to the MID for that measure. This suggests that a two-point change may be a reasonable estimate for the MID of the BPI-SF worst pain rating.
Discussion
Data from both distribution-based and anchor-based approaches were used to develop estimates of the MID for the BPI-SF worst pain rating. Results from these approaches are similar, providing reasonably strong support for establishing a two-point MID for the BPI-SF worst pain rating. Further, the results suggest that this estimate of MID is, for the most part, independent of baseline BPI-SF worst pain ratings. However, there is some evidence to suggest that the direction of change (improvement or worsening) may be important to consider. A number of reports have suggested that a smaller change may be required to be considered clinically important when a patient is improving compared with worsening.13 Also, when considered as a percentage, a one-point change in any scale has a different value for an increase versus a decrease; eg, a change from 2 to 3 is an increase of 50%, while a change from 3 to 2 is a decrease of 33%. Nonetheless, these findings provide important information to researchers for interpreting changes in the BPI-SF worst pain ratings.
In addition, although not specific to the BPI worst pain rating, the findings of this study are consistent with other published MID analyses for a similar item. A recent review of three studies concluded that, for a numerical rating scale of pain intensity ranging 0–10 similar in content to the BPI-SF worst pain rating, changes of around two points represent “meaningful,” “much better,” or “much improved” reductions in chronic pain.24
Several factors contribute to the overall strength of the current results. First, as frequently recommended in the literature,11 both anchor-based and distribution-based methods were used to estimate the MID for the worst pain rating. Second, analyses were based on a large sample, totaling over 1,500 patients for the baseline to week 25 assessment interval. A larger sample size will generally provide a broader distribution of responses, which will likely increase the generalizability of the results. Third, multiple anchors were used to evaluate changes in BPI-SF worst pain ratings. Fourth, analyses were performed across several assessment intervals to determine the strongest relationship between BPI-SF ratings and other anchors. Finally, the regression analyses provide important information about whether baseline differences influence the relationship between BPI-SF and other PRO measures.
Nevertheless, these analyses are not without certain limitations. The sample for the current analyses consisted entirely of breast cancer patients. It is unclear to what extent these results will be relevant for other patient populations. Further research is needed to determine whether the MID for the BPI-SF worst pain rating established in this sample has broader applicability. Also, it must be noted that the recall period varied across assessments. The BPI-SF focuses on the past 24 hours, the FACT uses the past week, and the EQ-5D uses the present moment. It is unclear to what extent these differences in recall periods may have influenced the current results. Finally, the baseline to week 25 interval was used to determine the MID for the BPI-SF worst pain rating based on the higher correlations for this interval. Data from baseline to week 49 are consistent with these results, providing some confirmatory evidence to suggest that these MID estimates are stable.
In conclusion, the findings of the present analyses suggest that the MID estimate for the BPI-SF worst pain rating is two points. This value provides guidance to researchers using the BPI-SF worst pain rating on how to interpret baseline differences as well as change scores in the BPI-SF worst pain rating. Additional analyses could be done in other populations to confirm these findings.
References1
1 K.W. Wyrwich, M. Bullinger and N. Aaronson et al., Estimating clinically significant differences in quality of life outcomes, Qual Life Res 14 (2005), pp. 285–295. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (119)
2 S.D. Mathias, S.K. Gao, M. Rutstein, C.F. Snyder, A.W. Wu and D. Cella, Evaluating clinically meaningful change on the ITP-PAQ: preliminary estimates of minimal important differences, Curr Med Res Opin 25 (2) (2009), pp. 375–383. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)
3 K.J. Yost, M.V. Sorensen, E.A. Hahn, G.A. Glendenning, A. Gnanasakthy and D. Cella, Using multiple anchor- and distribution-based estimates to evaluate clinically meaningful change on the Functional Assessment of Cancer Therapy-Biologic Response Modifiers (FACT-BRM) instrument, Value Health 8 (2) (2005), pp. 117–127. Abstract | | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (28)
4 R.D. Hays and J.M. Woolley, The concept of clinically meaningful difference in health-related quality-of-life research: How meaningful is it?, Pharmacoeconomics 18 (5) (2000), pp. 419–423. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (177)
5 R.L. Daut, C.S. Cleeland and R.C. Flanery, Development of the Wisconsin Brief Pain Questionnaire to assess pain in cancer and other diseases, Pain 17 (2) (1983), pp. 197–210. Abstract | | View Record in Scopus | Cited By in Scopus (543)
6 C. Cleeland, Brief Pain Inventory User Guide, University of Texas M. D. Anderson Cancer Center, Houston (2009).
7 R.H. Dworkin, D.C. Turk and J.T. Farrar et al., Core outcome measures for chronic pain clinical trials: IMMPACT recommendations, Pain 113 (1–2) (2005), pp. 9–19. Article | | View Record in Scopus | Cited By in Scopus (380)
8 U.S. Department of Health and Human Services Food and Drug Administration (FDA), Guidance for Industry: Patient-Reported Outcome Measures: Use in Medical Product Development to Support Labeling Claims, FDA, Silver Spring, MD (2009).
9 M.M. Oken, R.H. Creech and D.C. Tormey et al., Toxicity and response criteria of the Eastern Cooperative Oncology Group, Am J Clin Oncol 5 (6) (1982), pp. 649–655. View Record in Scopus | Cited By in Scopus (1968)
10 M.J. Brady, D.F. Cella, F. Mo and A.E. Bonomi et al., Reliability and validity of the Functional Assessment of Cancer Therapy–Breast Cancer Quality of Life instrument, J Clin Oncol 15 (1997), pp. 974–986. View Record in Scopus | Cited By in Scopus (360)
11 R.D. Crosby, R.L. Kolotkin and G.R. Williams, Defining clinically meaningful change in health-related quality of life, J Clin Epidemiol 56 (5) (2003), pp. 395–407. Article | | View Record in Scopus | Cited By in Scopus (233)
12 D. Revicki, R.D. Hays, D. Cella and J. Sloan, Recommended methods for determining responsiveness and minimally important differences for patient-reported outcomes, J Clin Epidemiol 61 (2) (2008), pp. 102–109. Article | | View Record in Scopus | Cited By in Scopus (121)
13 D. Cella, E.A. Hahn and K. Dineen, Meaningful change in cancer-specific quality of life scores: differences between improvement and worsening, Qual Life Res 11 (3) (2002), pp. 207–221. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (137)
14 D.T. Eton, D. Cella and K.J. Yost et al., A combination of distribution- and anchor-based approaches determined the minimally important differences (MIDs) for four endpoints in a breast cancer scale, J Clin Epidemiol 57 (2004), pp. 898–910. Article | | View Record in Scopus | Cited By in Scopus (68)
15 S. Weibe, S. Matijevic, M. Eliasziw and P.A. Derry, Clinically important change in quality of life in epilepsy, J Neurol Neurosurg Psychiatry 73 (2002), pp. 116–120.
16 K.L. Miller, J.G. Walt and D.R. Mink et al., Minimal clinically important difference for the ocular surface disease index, Arch Ophthalmol 128 (1) (2010), pp. 94–101. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
17 K.W. Wyrwich, W.M. Tierney and F.D. Wolinsky, Further evidence supporting an SEM-based criterion for identifying meaningful intra-individual changes in health-related quality of life, J Clin Epidemiol 52 (9) (1999), pp. 861–873. Article | | View Record in Scopus | Cited By in Scopus (272)
18 F.D. Wolinsky, G.J. Wan and W.M. Tierney, Changes in the SF-36 in 12 months in a clinical sample of disadvantaged older adults, Med Care 36 (11) (1998), pp. 1589–1598. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (33)
19 J. Cohen, Statistical Power Analysis for the Behavioral Sciences (2nd ed.), Lawrence Erlbaum, Hillsdale, NJ (1988).
20 G.H. Guyatt, C. Bombardier and P.X. Tugwell, Measuring disease-specific quality of life in clinical trials, CMAJ 134 (8) (1986), pp. 889–895. View Record in Scopus | Cited By in Scopus (324)
21 G.R. Norman, P. Stratford and G. Regehr, Methodological problems in the retrospective computation of responsiveness to change: the lesson of Cronbach, J Clin Epidemiol 50 (8) (1997), pp. 869–879. Article | | View Record in Scopus | Cited By in Scopus (230)
22 A. Stopeck, J. Body and Y. Fujiwara et al., Denosumab versus zoledronic acid for the treatment of breast cancer patients with bone metastases: results of a randomized phase 3 study, Eur J Cancer Suppl 7 (2009), p. 2. Abstract |
23 A.S. Pickard, M.P. Neary and D. Cella, Estimation of minimally important differences in EQ-5D utility and VAS scores in cancer, Health Qual Life Outcomes 5 (2007), p. 70. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
24 R.H. Dworkin, D.C. Turk and K.W. Wyrwich et al., Interpreting the clinical importance of treatment outcomes in chronic pain clinical trials: IMMPACT recommendations, J Pain 9 (2) (2008), pp. 105–121. Article | | View Record in Scopus | Cited By in Scopus (190)
Correspondence to: Susan D. Mathias, Health Outcomes Solutions, PO Box 2343, Winter Park, FL 32790; telephone: (407) 643-9016; fax: (866) 384-0194
Original research
Susan D. Mathias MPH
Abstract
The Brief Pain Inventory–Short Form (BPI-SF) is widely used for assessing pain in clinical and research studies. The worst pain rating is often the primary outcome of interest; yet, no published data are available on its minimally important difference (MID). Breast cancer patients with bone metastases enrolled in a randomized, double-blind, phase III study comparing denosumab with zoledronic acid for preventing skeletal related events and completed the BPI-SF, FACT-B, and EQ-5D at baseline, week 5, and monthly through the end of the study. Anchor- and distribution-based MID estimates were computed. Data from 1,564 patients were available. Spearman correlation coefficients for anchors ranged from 0.33–0.65. Mean change scores for worst pain ratings corresponding to one-category improvement in each anchor were 0.26–1.04 for BPI-SF current pain, −1.40 to −2.42 for EQ-5D Index score, 1.71–1.98 for EQ-5D Pain item, −2.22 to −0.51 for FACT-B TOI, −1.61 to −0.16 for FACT-G Physical, and −1.31 to −0.12 for FACT-G total. Distribution-based results were 1 SEM = 1.6, 0.5 effect size = 1.4, and Guyatt's statistic = 1.4. Combining anchor- and distribution-based results yielded a two-point MID estimate. An MID estimate of two points is useful for interpreting how much change in worst pain is considered clinically meaningful.
Article Outline
- Methods
- Study Design
- Outcome Measures and Assessment Intervals
- Anchor-Based Analysis
- Distribution-Based Analysis
- Integrating Anchor-Based and Distribution-Based Mid Estimates
The MID may be estimated through distribution-based methods and/or anchor-based methods. Distribution-based methods are based on the distribution of the data. Examples of distribution-based methods include effect size measures, the standard error of measurement (SEM), one-half times the standard deviation, and the responsiveness index.[2] and [3] Anchor-based methods are based on the association between the PRO measure and an interpretable external measure, such as a global rating of change or a response to treatment. These methods may result in somewhat different estimates, and no particular estimate is considered the most valid.[2], [3] and [4] Therefore, researchers are encouraged to use more than one method and to present a range of MID estimates.
A frequently used PRO measure for the assessment of pain is the Brief Pain Inventory–Short Form (BPI-SF). The foundation of the BPI-SF is the Wisconsin Brief Pain Questionnaire, which was developed over 25 years ago based on interviews with cancer patients, expert opinion, and then-current psychometric standards.5 Over time, the Wisconsin Brief Pain Questionnaire evolved into the Brief Pain Inventory, which was later reduced to a shorter version, the BPI-SF. Today, the BPI-SF is the standard for clinical and research use. It has been used in over 400 studies, including psychometric evaluations and clinical applications with a wide range of conditions (e.g., cancer pain, fibromyalgia, neuropathic pain, and joint diseases).6
The BPI-SF includes two domains: pain severity and pain interference. The pain severity domain, the focus of this report, includes items specific to pain at “worst,” “least,” “average,” and “now” (current pain), with a numerical response scale ranging from 0 (no pain) to 10 (pain as bad as you can imagine). In clinical trials, the worst pain item has been used alone as a measure of pain severity.6 Its use as a single item is supported by a consensus panel on outcome measures for chronic pain clinical trials.7 In addition, the Food and Drug Administration's (FDA) guidance on PROs states that a single-item PRO measure of pain severity is appropriate for assessing the effect of a treatment on pain.8 Although extensive psychometric evaluation of the BPI-SF has been conducted, no estimates of the MID are available for the BPI-SF worst pain item. Establishing the MID for the BPI-SF worst pain item is important because it will provide a clinically relevant reference to interpret changes in pain scores. Therefore, the objective of this current report was to estimate the MID of the worst pain item of the BPI-SF.
Methods
Study Design
Patients with advanced breast cancer and bone metastases were enrolled in an international, randomized, double-blind, double-dummy, active-controlled phase III study comparing denosumab with zoledronic acid for delaying or preventing skeletal related events. Patients were eligible to participate if they had histologically or cytologically confirmed breast adenocarcinoma; current or prior radiologic, computed tomography, or magnetic resonance imaging evidence of at least one bone metastasis; and an Eastern Cooperative Oncology Group (ECOG) performance status of 0, 1, or 2. Patients with current or prior intravenous bisphosphonate administration were excluded. Patients completed PRO assessments, including the BPI-SF, at baseline, week 5, and every 4 weeks thereafter until the end of the study. Assessments were scheduled to take place prior to any study procedures and prior to study drug administration. Although data collection continued, PRO analyses for efficacy were truncated when approximately 30% of patients dropped out of the study due to death, disease progression, or withdrawn consent.
Outcome Measures and Assessment Intervals
A number of outcome measures were assessed in the study and considered for use as anchors for evaluating the MID of the BPI-SF worst pain item, including one clinician-reported measure (ECOG Performance Status) and several PRO measures: the EuroQoL 5 Dimensions (EQ-5D) Index score, the Functional Assessment of Cancer Therapy-Breast Cancer (FACT-B), and the BPI-SF current pain rating.
The ECOG Performance Status, which assesses how a patient's disease or its treatment is progressing and how the disease affects the daily living abilities of the patient, is a single-item, six-point, clinician-rated assessment of performance ranging from 0 (fully active, no restrictions) to 5 (dead).9 The EQ-5D Index score is a measure of health status, which assesses five dimensions: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Each dimension is comprised of three response options: no problems, some/moderate problems, and extreme problems. Responses are converted to a weighted health state index, with scores ranging from −0.594 (worst health) to 1.0 (full health). The single item on pain from the EQ-5D was also evaluated separately as an anchor. The FACT-B includes the four core FACT-General (FACT-G) dimensions of physical well-being, social/family well-being, emotional well-being, and functional well-being, for which scale scores and a total score can be computed. In addition, the FACT-B includes a breast cancer–specific subscale.10 The FACT-B Trial Outcome Index (TOI) is the sum of the physical well-being score, the functional well-being score, and the breast cancer subscale. The four FACT-G scale scores, the FACT-G total score, the FACT-B TOI, and a single-item overall quality-of-life (QOL) rating from the functional well-being section were all evaluated as potential anchors. The single-item overall QOL item from the functional well-being scale was selected to balance out the single item on pain that was selected from the EQ-5D, by serving as a more general potential anchor in breadth and scope. For all of these FACT outcome measures, a higher score indicates better health-related QOL. Finally, the current pain rating from the BPI-SF, ranging from 0 (no pain) to 10 (pain as bad as you can imagine), was also considered as an anchor because it was hypothesized to be highly correlated with the worst pain rating and because it would assist in understanding the behavior of other potential anchors.
Several assessment intervals were considered for evaluation of the MID for the BPI-SF worst pain item: baseline to week 5, baseline to week 13, and baseline to week 25. The analysis for each time interval included only those patients with complete baseline and end-of-interval (i.e., week 5, week 13, or week 25) assessments on the BPI-SF worst pain item and the relevant anchor of interest. In addition, a post hoc confirmatory analysis was conducted using a longer interval of time, from baseline to week 49. No imputation of missing data was performed. Analysis was performed on pooled data, regardless of treatment assignment.
Anchor-Based Analysis
The usefulness of an anchor depends on the correlation of the PRO change score and the anchor.11 Therefore, to select the most appropriate anchors and time interval for estimating the MID for the BPI-SF worst pain item, Spearman correlation coefficients were calculated between changes in the BPI-SF worst pain rating and changes in potential anchors across each of the potential time intervals. The time interval with the highest correlations and the anchors with statistically significant (P < 0.05) a priori specified correlations above 0.30 were selected for inclusion in the MID analysis.12
A one-category change was defined as a one-point change for the BPI-SF current pain item, a one-point change for the EQ-5D pain item, a three-point change for the FACT-G Physical Well-Being scale,13 a six-point change for the FACT-G total and FACT-B TOI scores,14 and a 0.20 change for the EQ-5D Index score. For the selected interval and anchors, the mean change in BPI-SF worst pain item that corresponds to a one-category increase and decrease in each anchor was calculated. In addition, ordinary least squares regression models were used to regress changes in BPI-SF worst pain ratings on changes of each of the anchors.[15] and [16] The regression models included main effects for change in each anchor and an interaction term expressing the change in anchor-by-baseline anchor.
Distribution-Based Analysis
The following distribution-based measures were calculated for the BPI-SF worst pain item: (1) the SEM, (2) effect size (Cohen's d), and (3) Guyatt's statistic. The SEM is a measure of the precision of a test instrument. It is calculated on the basis of sample data using the sample standard deviation and the sample reliability coefficient. While the standard deviation and the reliability of a measure are sample-dependent, their relationship (and hence the SEM) remains relatively constant across samples. Therefore, the SEM is considered to be an attribute of the measure and not a characteristic of the sample per se.17 Threshold values of 1 SEM have been suggested for defining clinically meaningful differences.18 The reliability coefficient was estimated for the BPI-SF worst pain item by calculating the intraclass correlation coefficients (ICCs) using two intervals of time. One used 7 days (days 1–8), a more typical interval for assessing reproducibility, while the other approach used a later interval, from week 105 to week 109. (Note: The 1-month interval was dictated by the schedule of assessments.) For both ICC values, only those patients whose FACT-B overall QOL ratings changed by 10% or less during the respective intervals were included. The 10% criterion was selected after reviewing the full distribution of change scores and their associated sample sizes, to arrive at a reasonable sample size of approximately 100 subjects.
Cohen's d, alternatively referred to as the “standardized effect size,” is calculated by dividing the difference between the baseline and week-25 scores by the standard deviation at baseline.19 The effect size represents individual change in terms of the number of baseline standard deviations. A value of 0.20 is a small effect, 0.50 is a medium effect, and 0.80 is a large effect. Effect sizes of 0.20, 0.50, and 0.80 were calculated in this study.
Guyatt's statistic, also referred to as the “responsiveness statistic,” is calculated by dividing the difference between baseline and week-25 change by the standard deviation of change observed for a group of stable patients.20 The denominator of the responsiveness statistics adjusts for spurious change due to measurement error. Values of 0.20 and 0.50 have been used to represent “small” and “medium” changes, respectively.21 Values representing 0.20 and 0.50 were calculated in this study. Stable patients were defined as those whose ECOG Performance rating did not change during the assessment interval. A different variable was used in defining the stable population for purposes of calculating the SEM and Guyatt's statistic because both variables were not consistently collected on the same schedule of assessments.
Integrating Anchor-Based and Distribution-Based Mid Estimates
The minimal detectable change (MDC) for the worst pain item was established by comparing distribution-based estimates. The MDC represents the smallest change that can be reliably distinguished from random fluctuation and, thus, the lower bound for establishing the MID.11 If the MID were lower than the MDC, then the instrument would not be capable of distinguishing the MID. The SEM was considered the primary distribution-based estimate because it takes into account the reliability of the measure and, thus, estimates the precision of the instrument.11 Other distribution-based measures were also considered in establishing the MDC. Standardized effect size was considered a secondary distribution-based estimate because of its reliance on interperson variability, which is generally higher and less consistent than intraperson variability. Anchor-based estimates of the MID range were then compared. A final MID range was established that is greater than the MDC and integrates estimates from the various anchors.
Results
Patient Population
Demographic and clinical characteristics for patients included in the baseline to week 25 interval are presented in Table 1. Data from 1,564 of 2,049 patients who participated in the study and had valid (i.e., nonmissing) baseline and end-of-interval scores for the BPI-SF and anchors were used in these analyses. Patients were predominantly female with an average age of 57.2 ± 11.2 years. The majority of patients were white (80.9%). Average pain scores at baseline were 2.45 ± 2.51, with a full range of scores (0–10) being used. Clinical results from the study have been presented previously.22
CHARACTERISTIC, n (%) | STUDY SAMPLE (n = 1,564) |
---|---|
Gender | |
Female | 1,550 (99.1) |
Male | 14 (0.9) |
Age, mean years ± SD (range) | 57.2 ± 11.2 (27.1–91.2) |
Race | |
White | 1,265 (80.9) |
Black | 38 (2.4) |
Hispanic | 92 (5.9) |
Japanese | 119 (7.6) |
Asian | 28 (1.8) |
Other | 22 (1.4) |
Demographic characteristics including the breakdown by gender, age, and race for the study sample are shown.
Anchor-Based Analysis
Spearman correlations between changes in the BPI-SF worst pain item and changes in potential anchors are presented in Table 2. For all potential anchors, the highest correlations with the BPI-SF worst pain rating were obtained at the baseline to week 25 interval. All potential anchors correlated significantly (P < 0.001) with the BPI-SF worst pain rating with the exception of the FACT-G Social/Family Well-Being scale. However, correlations were low (<0.30) for several potential anchors: ECOG Performance Status, FACT-B Overall QOL item, FACT-G Emotional Well-Being, and FACT-G Functional Well-Being. Therefore, the week 25 interval and the following anchors were selected for the MID analysis: BPI-SF current pain rating, EQ-5D Index score, EQ-5D Pain item, FACT-B TOI, FACT-G Physical Well-Being, and FACT-G total score. Correlation coefficients between the changes in the selected anchors and changes in the BPI-SF worst pain ratings range from 0.329–0.647.
Bolded correlations represent the highest correlations with anchors where correlation r ≥ 0.300.
Spearman correlation coefficients between changes in BPI-SF worst pain rating and changes in each of the 11 potential anchors that were considered are provided. The data are displayed for three intervals of time including baseline to week 5, baseline to week 13, and baseline to week 25. Using a cut point of r ≥ 0.300, only those correlations that are bolded meet the criteria of acceptability.
Mean changes in the BPI-SF worst pain rating that correspond to a one-category change in anchors from baseline to week 25 are presented in Table 3. BPI-SF current pain ratings >5 and EQ-5D Index scores <0.40 were excluded from their respective analysis due to small sample sizes. A one-category increase in the anchor scores was associated with an absolute value of change in the BPI-SF worst pain item ranging from 0.26–2.42. A one-category decrease in the anchor score was associated with an absolute value of change in the BPI-SF worst pain item ranging from 0.56–3.16. Changes associated with improvement and worsening in anchors were not symmetrical, nor was there a consistent trend across anchors. For example, for the EQ-5D pain item, the magnitude of change in BPI-SF worst pain was greater for a one-category increase in the anchor than for a one-category decrease in the anchor. In contrast, for the EQ-5D Index score, the magnitude of change in BPI-SF worst pain was greater for a one-category decrease in the anchor than for a one-category increase in the anchor.
ANCHOR | ONE CATEGORYA INCREASE IN ANCHOR | ONE CATEGORY DECREASE IN ANCHOR |
---|---|---|
BPI-SF Current Pain rating | 0.26–1.04 | −0.89 to −1.66 |
EQ 5D Index score | −2.42 to −1.40 | 0.56–1.63 |
EQ 5D Pain item | 1.71–1.98 | −3.16 to −2.56 |
FACT-B TOI | −2.22 to −0.51 | −0.56 to 0.77 |
FACT-G Physical Well-Being | −1.61 to −0.16 | −0.79 to 0.46 |
FACT-G total | −1.31 to −0.12 | −0.97 to 0.57 |
The range of mean changes in BPI-SF worst pain ratings (using the interval from baseline to week 25) for the six anchors that met the correlation criteria in Table 2 are provided. Mean changes are displayed for one-category increases and one-category decreases in anchor.
a One category (increase or decrease) represents 0.20 points for EQ-5D Index score, one point for BPI-SF current pain rating and EQ-5D pain item, three points for FACT-G Physical Well-Being, and six points for FACT-G total and FACT-B TOI.
The regression of changes in anchors on changes in the BPI-SF worst pain item is shown in Table 4. Changes in each anchor are significantly (P < 0.05) associated with changes in BPI-SF worst pain rating. A one-point increase in BPI-SF current pain rating and EQ-5D Pain item is associated with a 0.817 and 1.805 increase in BPI-SF worst pain, respectively, while a one-point increase in EQ-5D Index score, FACT-B TOI, FACT-G Physical Well-Being, and FACT-G total is associated with a 3.548, 0.098, 0.163, and 0.048 decrease in BPI-SF worst pain rating, respectively. Likewise, a two-point increase in BPI-SF current pain rating and EQ-5D Pain item is associated with a 1.634 and 3.610 increase in BPI-SF worst pain, respectively, while a two-point increase in EQ-5D Index score, FACT-B TOI, FACT-G Physical Well-Being, and FACT-G total is associated with a 7.096, 0.196, 0.326, and 0.096 decrease in BPI-SF worst pain rating, respectively. The change in anchor-by-baseline anchor interaction was statistically significant only for BPI current pain and FACT-G Physical Well-Being. The interaction tests whether the anchor–BPI-SF slope differs as a function of baseline anchor score; therefore, a lack of significance suggests that the association between BPI-SF worst pain and other anchors does not differ by baseline anchor rating.
VARIABLE | PREDICTOR | b | β | SIG. |
---|---|---|---|---|
Change in BPI current pain | Main effect | 0.817 | 0.724 | <0.001 |
Interaction with baseline anchor | −0.024 | −0.107 | 0.001 | |
Change in EQ-5D Health State Index | Main effect | −3.548 | −0.349 | <0.001 |
Interaction with baseline anchor | 0.220 | 0.021 | 0.465 | |
Change in EQ-5D Pain item | Main effect | 1.805 | 0.352 | <0.001 |
Interaction with baseline anchor | 0.207 | 0.080 | 0.261 | |
Change in FACT-B TOI | Main effect | −0.098 | −0.406 | <0.001 |
Interaction with baseline anchor | 0.000 | 0.028 | 0.756 | |
Change in FACT-G Physical Well-Being | Main effect | −0.163 | −0.321 | <0.001 |
Interaction with baseline anchor | −0.004 | −0.133 | 0.024 | |
Change in FACT-G total score | Main effect | −0.048 | −0.231 | 0.025 |
Interaction with baseline anchor | 0.000 | −0.130 | 0.209 |
b, regression coefficient; β, standardized regression coefficient; Sig., significance level.
Possible ranges: BPI Pain Right Now 0 (least) to 10 (most), EQ-5D Health State Index scores −0.594 (worst) to 1.00 (best), EQ-5D Pain item scores 1 (none) to 3 (severe), FACT-B TOI scores 4 (worst) to 92 (best), FACT-G Physical Well-Being scores 0 (worst) to 28 (best), FACT-G total score 8 (worst) to 108 (best), BPI Worst Pain item 0 (least) to 10 (most).
Changes in all anchors are significantly (P < 0.05) associated with changes in BPI-SF worst pain ratings. A one-point increase in BPI-SF current pain rating and EQ-5D pain item is associated with increases (positive b score) in the BPI-SF worst pain rating, and a one-point increase in EQ-5D Index, FACT-B TOI, FACT-G Physical Well-Being, and FACT-G total scores is associated with decreases (negative b score) in the BPI-SF worst pain ratings. The change in anchor-by-baseline anchor interaction was statistically significant only for the BPI current pain and FACT-G PWB items.
A post hoc confirmatory analysis was done replicating these analyses using data from the baseline to week 49 interval (n = 1,250). Results indicate a slightly stronger correlation between the anchors and the change scores. (Spearman's correlations range from 0.372 for FACT-TOI to 0.644 for BPI-SF current pain rating.) Mean change scores of BPI-SF worst pain ratings by each of the six anchors and regression coefficients were similar to those for the baseline to week 25 interval. For instance, mean change scores for the EQ-5D Pain item for stable patients ranged from 0.25–0.56, 1.58–295 for an improvement of one category, and 1.75–2.80 for a worsening of one category compared with 0.50–0.51, 1.71–1.98, and 2.56–3.16, respectively, for the baseline to week 25 interval.
Distribution-Based Analysis
The distribution-based estimates for the BPI-SF worst pain rating are presented in Table 5. There appears to be consistency with the 1 SEM estimates, the 0.50 effect size, and the 0.50 Guyatt's statistic.
The results from the three distribution-based approaches presented in this table will be combined with those of the anchor-based results to estimate the MID.
a The standard error of measurement is a measure of the precision of a test instrument. It is calculated on the basis of sample data using the sample standard deviation and the sample reliability coefficient. Intraclass correlation coefficients (ICCs) for BPI-SF worst pain rating from day 1 to day 8 and week 105 to week 109 in patients whose FACT-B overall QOL ratings change by <10% are 0.685 (n = 926) and 0.800 (n = 109), respectively.b Alternatively referred to as Cohen's d, the effect size is calculated by dividing the difference between the pretest and posttest scores by the standard deviation at pretest. The standard deviation of BPI-SF worst pain rating at baseline (n = 1,877) is 2.849.c Alternatively referred to as the responsiveness statistic, Guyatt's statistic is calculated by dividing the difference between pretest and posttest changes by the standard deviation of change observed for a group of stable patients. The standard deviation of change in BPI-SF worst pain rating from baseline to week 25 in patients whose ECOG performance rating does not change (n = 1,120) is 2.833.
Integrating Anchor-Based and Distribution-Based Mid Estimates
The distribution-based analyses suggest that the MDC for the worst pain rating, defined as the smallest change that can be reliably differentiated from random fluctuation, is between 1.3 and 1.6 points (see Table 5). This represents the lower bound for establishing the MID.
The results from regression analyses can be used to translate changes between anchors and corresponding changes in BPI-SF worst pain. This strategy can be particularly informative when the MID for an anchor is known. This is the case for the EQ-5D Health State Index, where the MID has been estimated at 0.06 for U.S. Index scores and 0.07 for U.K. Index scores.23 A one-point change in EQ-5D Index translates to a change of −3.548 in BPI-SF worst pain, so a 0.07-point change in EQ-5D Index (the MID for the measure) corresponds to a change of −0.248 in BPI-SF worst pain. In contrast, a one-point change in BPI-SF worst pain (which is smaller than the MID based upon the distribution-based analyses) translates to a change of 0.036 for the EQ-5D Index score (considerably smaller than the MID of 0.07). However, a two-point change in BPI-SF worst pain rating corresponds to a 0.072 change in EQ-5D Index score, which is almost identical to the MID for that measure. This suggests that a two-point change may be a reasonable estimate for the MID of the BPI-SF worst pain rating.
Discussion
Data from both distribution-based and anchor-based approaches were used to develop estimates of the MID for the BPI-SF worst pain rating. Results from these approaches are similar, providing reasonably strong support for establishing a two-point MID for the BPI-SF worst pain rating. Further, the results suggest that this estimate of MID is, for the most part, independent of baseline BPI-SF worst pain ratings. However, there is some evidence to suggest that the direction of change (improvement or worsening) may be important to consider. A number of reports have suggested that a smaller change may be required to be considered clinically important when a patient is improving compared with worsening.13 Also, when considered as a percentage, a one-point change in any scale has a different value for an increase versus a decrease; eg, a change from 2 to 3 is an increase of 50%, while a change from 3 to 2 is a decrease of 33%. Nonetheless, these findings provide important information to researchers for interpreting changes in the BPI-SF worst pain ratings.
In addition, although not specific to the BPI worst pain rating, the findings of this study are consistent with other published MID analyses for a similar item. A recent review of three studies concluded that, for a numerical rating scale of pain intensity ranging 0–10 similar in content to the BPI-SF worst pain rating, changes of around two points represent “meaningful,” “much better,” or “much improved” reductions in chronic pain.24
Several factors contribute to the overall strength of the current results. First, as frequently recommended in the literature,11 both anchor-based and distribution-based methods were used to estimate the MID for the worst pain rating. Second, analyses were based on a large sample, totaling over 1,500 patients for the baseline to week 25 assessment interval. A larger sample size will generally provide a broader distribution of responses, which will likely increase the generalizability of the results. Third, multiple anchors were used to evaluate changes in BPI-SF worst pain ratings. Fourth, analyses were performed across several assessment intervals to determine the strongest relationship between BPI-SF ratings and other anchors. Finally, the regression analyses provide important information about whether baseline differences influence the relationship between BPI-SF and other PRO measures.
Nevertheless, these analyses are not without certain limitations. The sample for the current analyses consisted entirely of breast cancer patients. It is unclear to what extent these results will be relevant for other patient populations. Further research is needed to determine whether the MID for the BPI-SF worst pain rating established in this sample has broader applicability. Also, it must be noted that the recall period varied across assessments. The BPI-SF focuses on the past 24 hours, the FACT uses the past week, and the EQ-5D uses the present moment. It is unclear to what extent these differences in recall periods may have influenced the current results. Finally, the baseline to week 25 interval was used to determine the MID for the BPI-SF worst pain rating based on the higher correlations for this interval. Data from baseline to week 49 are consistent with these results, providing some confirmatory evidence to suggest that these MID estimates are stable.
In conclusion, the findings of the present analyses suggest that the MID estimate for the BPI-SF worst pain rating is two points. This value provides guidance to researchers using the BPI-SF worst pain rating on how to interpret baseline differences as well as change scores in the BPI-SF worst pain rating. Additional analyses could be done in other populations to confirm these findings.
References1
1 K.W. Wyrwich, M. Bullinger and N. Aaronson et al., Estimating clinically significant differences in quality of life outcomes, Qual Life Res 14 (2005), pp. 285–295. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (119)
2 S.D. Mathias, S.K. Gao, M. Rutstein, C.F. Snyder, A.W. Wu and D. Cella, Evaluating clinically meaningful change on the ITP-PAQ: preliminary estimates of minimal important differences, Curr Med Res Opin 25 (2) (2009), pp. 375–383. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)
3 K.J. Yost, M.V. Sorensen, E.A. Hahn, G.A. Glendenning, A. Gnanasakthy and D. Cella, Using multiple anchor- and distribution-based estimates to evaluate clinically meaningful change on the Functional Assessment of Cancer Therapy-Biologic Response Modifiers (FACT-BRM) instrument, Value Health 8 (2) (2005), pp. 117–127. Abstract | | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (28)
4 R.D. Hays and J.M. Woolley, The concept of clinically meaningful difference in health-related quality-of-life research: How meaningful is it?, Pharmacoeconomics 18 (5) (2000), pp. 419–423. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (177)
5 R.L. Daut, C.S. Cleeland and R.C. Flanery, Development of the Wisconsin Brief Pain Questionnaire to assess pain in cancer and other diseases, Pain 17 (2) (1983), pp. 197–210. Abstract | | View Record in Scopus | Cited By in Scopus (543)
6 C. Cleeland, Brief Pain Inventory User Guide, University of Texas M. D. Anderson Cancer Center, Houston (2009).
7 R.H. Dworkin, D.C. Turk and J.T. Farrar et al., Core outcome measures for chronic pain clinical trials: IMMPACT recommendations, Pain 113 (1–2) (2005), pp. 9–19. Article | | View Record in Scopus | Cited By in Scopus (380)
8 U.S. Department of Health and Human Services Food and Drug Administration (FDA), Guidance for Industry: Patient-Reported Outcome Measures: Use in Medical Product Development to Support Labeling Claims, FDA, Silver Spring, MD (2009).
9 M.M. Oken, R.H. Creech and D.C. Tormey et al., Toxicity and response criteria of the Eastern Cooperative Oncology Group, Am J Clin Oncol 5 (6) (1982), pp. 649–655. View Record in Scopus | Cited By in Scopus (1968)
10 M.J. Brady, D.F. Cella, F. Mo and A.E. Bonomi et al., Reliability and validity of the Functional Assessment of Cancer Therapy–Breast Cancer Quality of Life instrument, J Clin Oncol 15 (1997), pp. 974–986. View Record in Scopus | Cited By in Scopus (360)
11 R.D. Crosby, R.L. Kolotkin and G.R. Williams, Defining clinically meaningful change in health-related quality of life, J Clin Epidemiol 56 (5) (2003), pp. 395–407. Article | | View Record in Scopus | Cited By in Scopus (233)
12 D. Revicki, R.D. Hays, D. Cella and J. Sloan, Recommended methods for determining responsiveness and minimally important differences for patient-reported outcomes, J Clin Epidemiol 61 (2) (2008), pp. 102–109. Article | | View Record in Scopus | Cited By in Scopus (121)
13 D. Cella, E.A. Hahn and K. Dineen, Meaningful change in cancer-specific quality of life scores: differences between improvement and worsening, Qual Life Res 11 (3) (2002), pp. 207–221. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (137)
14 D.T. Eton, D. Cella and K.J. Yost et al., A combination of distribution- and anchor-based approaches determined the minimally important differences (MIDs) for four endpoints in a breast cancer scale, J Clin Epidemiol 57 (2004), pp. 898–910. Article | | View Record in Scopus | Cited By in Scopus (68)
15 S. Weibe, S. Matijevic, M. Eliasziw and P.A. Derry, Clinically important change in quality of life in epilepsy, J Neurol Neurosurg Psychiatry 73 (2002), pp. 116–120.
16 K.L. Miller, J.G. Walt and D.R. Mink et al., Minimal clinically important difference for the ocular surface disease index, Arch Ophthalmol 128 (1) (2010), pp. 94–101. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
17 K.W. Wyrwich, W.M. Tierney and F.D. Wolinsky, Further evidence supporting an SEM-based criterion for identifying meaningful intra-individual changes in health-related quality of life, J Clin Epidemiol 52 (9) (1999), pp. 861–873. Article | | View Record in Scopus | Cited By in Scopus (272)
18 F.D. Wolinsky, G.J. Wan and W.M. Tierney, Changes in the SF-36 in 12 months in a clinical sample of disadvantaged older adults, Med Care 36 (11) (1998), pp. 1589–1598. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (33)
19 J. Cohen, Statistical Power Analysis for the Behavioral Sciences (2nd ed.), Lawrence Erlbaum, Hillsdale, NJ (1988).
20 G.H. Guyatt, C. Bombardier and P.X. Tugwell, Measuring disease-specific quality of life in clinical trials, CMAJ 134 (8) (1986), pp. 889–895. View Record in Scopus | Cited By in Scopus (324)
21 G.R. Norman, P. Stratford and G. Regehr, Methodological problems in the retrospective computation of responsiveness to change: the lesson of Cronbach, J Clin Epidemiol 50 (8) (1997), pp. 869–879. Article | | View Record in Scopus | Cited By in Scopus (230)
22 A. Stopeck, J. Body and Y. Fujiwara et al., Denosumab versus zoledronic acid for the treatment of breast cancer patients with bone metastases: results of a randomized phase 3 study, Eur J Cancer Suppl 7 (2009), p. 2. Abstract |
23 A.S. Pickard, M.P. Neary and D. Cella, Estimation of minimally important differences in EQ-5D utility and VAS scores in cancer, Health Qual Life Outcomes 5 (2007), p. 70. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
24 R.H. Dworkin, D.C. Turk and K.W. Wyrwich et al., Interpreting the clinical importance of treatment outcomes in chronic pain clinical trials: IMMPACT recommendations, J Pain 9 (2) (2008), pp. 105–121. Article | | View Record in Scopus | Cited By in Scopus (190)
Correspondence to: Susan D. Mathias, Health Outcomes Solutions, PO Box 2343, Winter Park, FL 32790; telephone: (407) 643-9016; fax: (866) 384-0194
Original research
Susan D. Mathias MPH
Abstract
The Brief Pain Inventory–Short Form (BPI-SF) is widely used for assessing pain in clinical and research studies. The worst pain rating is often the primary outcome of interest; yet, no published data are available on its minimally important difference (MID). Breast cancer patients with bone metastases enrolled in a randomized, double-blind, phase III study comparing denosumab with zoledronic acid for preventing skeletal related events and completed the BPI-SF, FACT-B, and EQ-5D at baseline, week 5, and monthly through the end of the study. Anchor- and distribution-based MID estimates were computed. Data from 1,564 patients were available. Spearman correlation coefficients for anchors ranged from 0.33–0.65. Mean change scores for worst pain ratings corresponding to one-category improvement in each anchor were 0.26–1.04 for BPI-SF current pain, −1.40 to −2.42 for EQ-5D Index score, 1.71–1.98 for EQ-5D Pain item, −2.22 to −0.51 for FACT-B TOI, −1.61 to −0.16 for FACT-G Physical, and −1.31 to −0.12 for FACT-G total. Distribution-based results were 1 SEM = 1.6, 0.5 effect size = 1.4, and Guyatt's statistic = 1.4. Combining anchor- and distribution-based results yielded a two-point MID estimate. An MID estimate of two points is useful for interpreting how much change in worst pain is considered clinically meaningful.
Article Outline
- Methods
- Study Design
- Outcome Measures and Assessment Intervals
- Anchor-Based Analysis
- Distribution-Based Analysis
- Integrating Anchor-Based and Distribution-Based Mid Estimates
The MID may be estimated through distribution-based methods and/or anchor-based methods. Distribution-based methods are based on the distribution of the data. Examples of distribution-based methods include effect size measures, the standard error of measurement (SEM), one-half times the standard deviation, and the responsiveness index.[2] and [3] Anchor-based methods are based on the association between the PRO measure and an interpretable external measure, such as a global rating of change or a response to treatment. These methods may result in somewhat different estimates, and no particular estimate is considered the most valid.[2], [3] and [4] Therefore, researchers are encouraged to use more than one method and to present a range of MID estimates.
A frequently used PRO measure for the assessment of pain is the Brief Pain Inventory–Short Form (BPI-SF). The foundation of the BPI-SF is the Wisconsin Brief Pain Questionnaire, which was developed over 25 years ago based on interviews with cancer patients, expert opinion, and then-current psychometric standards.5 Over time, the Wisconsin Brief Pain Questionnaire evolved into the Brief Pain Inventory, which was later reduced to a shorter version, the BPI-SF. Today, the BPI-SF is the standard for clinical and research use. It has been used in over 400 studies, including psychometric evaluations and clinical applications with a wide range of conditions (e.g., cancer pain, fibromyalgia, neuropathic pain, and joint diseases).6
The BPI-SF includes two domains: pain severity and pain interference. The pain severity domain, the focus of this report, includes items specific to pain at “worst,” “least,” “average,” and “now” (current pain), with a numerical response scale ranging from 0 (no pain) to 10 (pain as bad as you can imagine). In clinical trials, the worst pain item has been used alone as a measure of pain severity.6 Its use as a single item is supported by a consensus panel on outcome measures for chronic pain clinical trials.7 In addition, the Food and Drug Administration's (FDA) guidance on PROs states that a single-item PRO measure of pain severity is appropriate for assessing the effect of a treatment on pain.8 Although extensive psychometric evaluation of the BPI-SF has been conducted, no estimates of the MID are available for the BPI-SF worst pain item. Establishing the MID for the BPI-SF worst pain item is important because it will provide a clinically relevant reference to interpret changes in pain scores. Therefore, the objective of this current report was to estimate the MID of the worst pain item of the BPI-SF.
Methods
Study Design
Patients with advanced breast cancer and bone metastases were enrolled in an international, randomized, double-blind, double-dummy, active-controlled phase III study comparing denosumab with zoledronic acid for delaying or preventing skeletal related events. Patients were eligible to participate if they had histologically or cytologically confirmed breast adenocarcinoma; current or prior radiologic, computed tomography, or magnetic resonance imaging evidence of at least one bone metastasis; and an Eastern Cooperative Oncology Group (ECOG) performance status of 0, 1, or 2. Patients with current or prior intravenous bisphosphonate administration were excluded. Patients completed PRO assessments, including the BPI-SF, at baseline, week 5, and every 4 weeks thereafter until the end of the study. Assessments were scheduled to take place prior to any study procedures and prior to study drug administration. Although data collection continued, PRO analyses for efficacy were truncated when approximately 30% of patients dropped out of the study due to death, disease progression, or withdrawn consent.
Outcome Measures and Assessment Intervals
A number of outcome measures were assessed in the study and considered for use as anchors for evaluating the MID of the BPI-SF worst pain item, including one clinician-reported measure (ECOG Performance Status) and several PRO measures: the EuroQoL 5 Dimensions (EQ-5D) Index score, the Functional Assessment of Cancer Therapy-Breast Cancer (FACT-B), and the BPI-SF current pain rating.
The ECOG Performance Status, which assesses how a patient's disease or its treatment is progressing and how the disease affects the daily living abilities of the patient, is a single-item, six-point, clinician-rated assessment of performance ranging from 0 (fully active, no restrictions) to 5 (dead).9 The EQ-5D Index score is a measure of health status, which assesses five dimensions: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Each dimension is comprised of three response options: no problems, some/moderate problems, and extreme problems. Responses are converted to a weighted health state index, with scores ranging from −0.594 (worst health) to 1.0 (full health). The single item on pain from the EQ-5D was also evaluated separately as an anchor. The FACT-B includes the four core FACT-General (FACT-G) dimensions of physical well-being, social/family well-being, emotional well-being, and functional well-being, for which scale scores and a total score can be computed. In addition, the FACT-B includes a breast cancer–specific subscale.10 The FACT-B Trial Outcome Index (TOI) is the sum of the physical well-being score, the functional well-being score, and the breast cancer subscale. The four FACT-G scale scores, the FACT-G total score, the FACT-B TOI, and a single-item overall quality-of-life (QOL) rating from the functional well-being section were all evaluated as potential anchors. The single-item overall QOL item from the functional well-being scale was selected to balance out the single item on pain that was selected from the EQ-5D, by serving as a more general potential anchor in breadth and scope. For all of these FACT outcome measures, a higher score indicates better health-related QOL. Finally, the current pain rating from the BPI-SF, ranging from 0 (no pain) to 10 (pain as bad as you can imagine), was also considered as an anchor because it was hypothesized to be highly correlated with the worst pain rating and because it would assist in understanding the behavior of other potential anchors.
Several assessment intervals were considered for evaluation of the MID for the BPI-SF worst pain item: baseline to week 5, baseline to week 13, and baseline to week 25. The analysis for each time interval included only those patients with complete baseline and end-of-interval (i.e., week 5, week 13, or week 25) assessments on the BPI-SF worst pain item and the relevant anchor of interest. In addition, a post hoc confirmatory analysis was conducted using a longer interval of time, from baseline to week 49. No imputation of missing data was performed. Analysis was performed on pooled data, regardless of treatment assignment.
Anchor-Based Analysis
The usefulness of an anchor depends on the correlation of the PRO change score and the anchor.11 Therefore, to select the most appropriate anchors and time interval for estimating the MID for the BPI-SF worst pain item, Spearman correlation coefficients were calculated between changes in the BPI-SF worst pain rating and changes in potential anchors across each of the potential time intervals. The time interval with the highest correlations and the anchors with statistically significant (P < 0.05) a priori specified correlations above 0.30 were selected for inclusion in the MID analysis.12
A one-category change was defined as a one-point change for the BPI-SF current pain item, a one-point change for the EQ-5D pain item, a three-point change for the FACT-G Physical Well-Being scale,13 a six-point change for the FACT-G total and FACT-B TOI scores,14 and a 0.20 change for the EQ-5D Index score. For the selected interval and anchors, the mean change in BPI-SF worst pain item that corresponds to a one-category increase and decrease in each anchor was calculated. In addition, ordinary least squares regression models were used to regress changes in BPI-SF worst pain ratings on changes of each of the anchors.[15] and [16] The regression models included main effects for change in each anchor and an interaction term expressing the change in anchor-by-baseline anchor.
Distribution-Based Analysis
The following distribution-based measures were calculated for the BPI-SF worst pain item: (1) the SEM, (2) effect size (Cohen's d), and (3) Guyatt's statistic. The SEM is a measure of the precision of a test instrument. It is calculated on the basis of sample data using the sample standard deviation and the sample reliability coefficient. While the standard deviation and the reliability of a measure are sample-dependent, their relationship (and hence the SEM) remains relatively constant across samples. Therefore, the SEM is considered to be an attribute of the measure and not a characteristic of the sample per se.17 Threshold values of 1 SEM have been suggested for defining clinically meaningful differences.18 The reliability coefficient was estimated for the BPI-SF worst pain item by calculating the intraclass correlation coefficients (ICCs) using two intervals of time. One used 7 days (days 1–8), a more typical interval for assessing reproducibility, while the other approach used a later interval, from week 105 to week 109. (Note: The 1-month interval was dictated by the schedule of assessments.) For both ICC values, only those patients whose FACT-B overall QOL ratings changed by 10% or less during the respective intervals were included. The 10% criterion was selected after reviewing the full distribution of change scores and their associated sample sizes, to arrive at a reasonable sample size of approximately 100 subjects.
Cohen's d, alternatively referred to as the “standardized effect size,” is calculated by dividing the difference between the baseline and week-25 scores by the standard deviation at baseline.19 The effect size represents individual change in terms of the number of baseline standard deviations. A value of 0.20 is a small effect, 0.50 is a medium effect, and 0.80 is a large effect. Effect sizes of 0.20, 0.50, and 0.80 were calculated in this study.
Guyatt's statistic, also referred to as the “responsiveness statistic,” is calculated by dividing the difference between baseline and week-25 change by the standard deviation of change observed for a group of stable patients.20 The denominator of the responsiveness statistics adjusts for spurious change due to measurement error. Values of 0.20 and 0.50 have been used to represent “small” and “medium” changes, respectively.21 Values representing 0.20 and 0.50 were calculated in this study. Stable patients were defined as those whose ECOG Performance rating did not change during the assessment interval. A different variable was used in defining the stable population for purposes of calculating the SEM and Guyatt's statistic because both variables were not consistently collected on the same schedule of assessments.
Integrating Anchor-Based and Distribution-Based Mid Estimates
The minimal detectable change (MDC) for the worst pain item was established by comparing distribution-based estimates. The MDC represents the smallest change that can be reliably distinguished from random fluctuation and, thus, the lower bound for establishing the MID.11 If the MID were lower than the MDC, then the instrument would not be capable of distinguishing the MID. The SEM was considered the primary distribution-based estimate because it takes into account the reliability of the measure and, thus, estimates the precision of the instrument.11 Other distribution-based measures were also considered in establishing the MDC. Standardized effect size was considered a secondary distribution-based estimate because of its reliance on interperson variability, which is generally higher and less consistent than intraperson variability. Anchor-based estimates of the MID range were then compared. A final MID range was established that is greater than the MDC and integrates estimates from the various anchors.
Results
Patient Population
Demographic and clinical characteristics for patients included in the baseline to week 25 interval are presented in Table 1. Data from 1,564 of 2,049 patients who participated in the study and had valid (i.e., nonmissing) baseline and end-of-interval scores for the BPI-SF and anchors were used in these analyses. Patients were predominantly female with an average age of 57.2 ± 11.2 years. The majority of patients were white (80.9%). Average pain scores at baseline were 2.45 ± 2.51, with a full range of scores (0–10) being used. Clinical results from the study have been presented previously.22
CHARACTERISTIC, n (%) | STUDY SAMPLE (n = 1,564) |
---|---|
Gender | |
Female | 1,550 (99.1) |
Male | 14 (0.9) |
Age, mean years ± SD (range) | 57.2 ± 11.2 (27.1–91.2) |
Race | |
White | 1,265 (80.9) |
Black | 38 (2.4) |
Hispanic | 92 (5.9) |
Japanese | 119 (7.6) |
Asian | 28 (1.8) |
Other | 22 (1.4) |
Demographic characteristics including the breakdown by gender, age, and race for the study sample are shown.
Anchor-Based Analysis
Spearman correlations between changes in the BPI-SF worst pain item and changes in potential anchors are presented in Table 2. For all potential anchors, the highest correlations with the BPI-SF worst pain rating were obtained at the baseline to week 25 interval. All potential anchors correlated significantly (P < 0.001) with the BPI-SF worst pain rating with the exception of the FACT-G Social/Family Well-Being scale. However, correlations were low (<0.30) for several potential anchors: ECOG Performance Status, FACT-B Overall QOL item, FACT-G Emotional Well-Being, and FACT-G Functional Well-Being. Therefore, the week 25 interval and the following anchors were selected for the MID analysis: BPI-SF current pain rating, EQ-5D Index score, EQ-5D Pain item, FACT-B TOI, FACT-G Physical Well-Being, and FACT-G total score. Correlation coefficients between the changes in the selected anchors and changes in the BPI-SF worst pain ratings range from 0.329–0.647.
Bolded correlations represent the highest correlations with anchors where correlation r ≥ 0.300.
Spearman correlation coefficients between changes in BPI-SF worst pain rating and changes in each of the 11 potential anchors that were considered are provided. The data are displayed for three intervals of time including baseline to week 5, baseline to week 13, and baseline to week 25. Using a cut point of r ≥ 0.300, only those correlations that are bolded meet the criteria of acceptability.
Mean changes in the BPI-SF worst pain rating that correspond to a one-category change in anchors from baseline to week 25 are presented in Table 3. BPI-SF current pain ratings >5 and EQ-5D Index scores <0.40 were excluded from their respective analysis due to small sample sizes. A one-category increase in the anchor scores was associated with an absolute value of change in the BPI-SF worst pain item ranging from 0.26–2.42. A one-category decrease in the anchor score was associated with an absolute value of change in the BPI-SF worst pain item ranging from 0.56–3.16. Changes associated with improvement and worsening in anchors were not symmetrical, nor was there a consistent trend across anchors. For example, for the EQ-5D pain item, the magnitude of change in BPI-SF worst pain was greater for a one-category increase in the anchor than for a one-category decrease in the anchor. In contrast, for the EQ-5D Index score, the magnitude of change in BPI-SF worst pain was greater for a one-category decrease in the anchor than for a one-category increase in the anchor.
ANCHOR | ONE CATEGORYA INCREASE IN ANCHOR | ONE CATEGORY DECREASE IN ANCHOR |
---|---|---|
BPI-SF Current Pain rating | 0.26–1.04 | −0.89 to −1.66 |
EQ 5D Index score | −2.42 to −1.40 | 0.56–1.63 |
EQ 5D Pain item | 1.71–1.98 | −3.16 to −2.56 |
FACT-B TOI | −2.22 to −0.51 | −0.56 to 0.77 |
FACT-G Physical Well-Being | −1.61 to −0.16 | −0.79 to 0.46 |
FACT-G total | −1.31 to −0.12 | −0.97 to 0.57 |
The range of mean changes in BPI-SF worst pain ratings (using the interval from baseline to week 25) for the six anchors that met the correlation criteria in Table 2 are provided. Mean changes are displayed for one-category increases and one-category decreases in anchor.
a One category (increase or decrease) represents 0.20 points for EQ-5D Index score, one point for BPI-SF current pain rating and EQ-5D pain item, three points for FACT-G Physical Well-Being, and six points for FACT-G total and FACT-B TOI.
The regression of changes in anchors on changes in the BPI-SF worst pain item is shown in Table 4. Changes in each anchor are significantly (P < 0.05) associated with changes in BPI-SF worst pain rating. A one-point increase in BPI-SF current pain rating and EQ-5D Pain item is associated with a 0.817 and 1.805 increase in BPI-SF worst pain, respectively, while a one-point increase in EQ-5D Index score, FACT-B TOI, FACT-G Physical Well-Being, and FACT-G total is associated with a 3.548, 0.098, 0.163, and 0.048 decrease in BPI-SF worst pain rating, respectively. Likewise, a two-point increase in BPI-SF current pain rating and EQ-5D Pain item is associated with a 1.634 and 3.610 increase in BPI-SF worst pain, respectively, while a two-point increase in EQ-5D Index score, FACT-B TOI, FACT-G Physical Well-Being, and FACT-G total is associated with a 7.096, 0.196, 0.326, and 0.096 decrease in BPI-SF worst pain rating, respectively. The change in anchor-by-baseline anchor interaction was statistically significant only for BPI current pain and FACT-G Physical Well-Being. The interaction tests whether the anchor–BPI-SF slope differs as a function of baseline anchor score; therefore, a lack of significance suggests that the association between BPI-SF worst pain and other anchors does not differ by baseline anchor rating.
VARIABLE | PREDICTOR | b | β | SIG. |
---|---|---|---|---|
Change in BPI current pain | Main effect | 0.817 | 0.724 | <0.001 |
Interaction with baseline anchor | −0.024 | −0.107 | 0.001 | |
Change in EQ-5D Health State Index | Main effect | −3.548 | −0.349 | <0.001 |
Interaction with baseline anchor | 0.220 | 0.021 | 0.465 | |
Change in EQ-5D Pain item | Main effect | 1.805 | 0.352 | <0.001 |
Interaction with baseline anchor | 0.207 | 0.080 | 0.261 | |
Change in FACT-B TOI | Main effect | −0.098 | −0.406 | <0.001 |
Interaction with baseline anchor | 0.000 | 0.028 | 0.756 | |
Change in FACT-G Physical Well-Being | Main effect | −0.163 | −0.321 | <0.001 |
Interaction with baseline anchor | −0.004 | −0.133 | 0.024 | |
Change in FACT-G total score | Main effect | −0.048 | −0.231 | 0.025 |
Interaction with baseline anchor | 0.000 | −0.130 | 0.209 |
b, regression coefficient; β, standardized regression coefficient; Sig., significance level.
Possible ranges: BPI Pain Right Now 0 (least) to 10 (most), EQ-5D Health State Index scores −0.594 (worst) to 1.00 (best), EQ-5D Pain item scores 1 (none) to 3 (severe), FACT-B TOI scores 4 (worst) to 92 (best), FACT-G Physical Well-Being scores 0 (worst) to 28 (best), FACT-G total score 8 (worst) to 108 (best), BPI Worst Pain item 0 (least) to 10 (most).
Changes in all anchors are significantly (P < 0.05) associated with changes in BPI-SF worst pain ratings. A one-point increase in BPI-SF current pain rating and EQ-5D pain item is associated with increases (positive b score) in the BPI-SF worst pain rating, and a one-point increase in EQ-5D Index, FACT-B TOI, FACT-G Physical Well-Being, and FACT-G total scores is associated with decreases (negative b score) in the BPI-SF worst pain ratings. The change in anchor-by-baseline anchor interaction was statistically significant only for the BPI current pain and FACT-G PWB items.
A post hoc confirmatory analysis was done replicating these analyses using data from the baseline to week 49 interval (n = 1,250). Results indicate a slightly stronger correlation between the anchors and the change scores. (Spearman's correlations range from 0.372 for FACT-TOI to 0.644 for BPI-SF current pain rating.) Mean change scores of BPI-SF worst pain ratings by each of the six anchors and regression coefficients were similar to those for the baseline to week 25 interval. For instance, mean change scores for the EQ-5D Pain item for stable patients ranged from 0.25–0.56, 1.58–295 for an improvement of one category, and 1.75–2.80 for a worsening of one category compared with 0.50–0.51, 1.71–1.98, and 2.56–3.16, respectively, for the baseline to week 25 interval.
Distribution-Based Analysis
The distribution-based estimates for the BPI-SF worst pain rating are presented in Table 5. There appears to be consistency with the 1 SEM estimates, the 0.50 effect size, and the 0.50 Guyatt's statistic.
The results from the three distribution-based approaches presented in this table will be combined with those of the anchor-based results to estimate the MID.
a The standard error of measurement is a measure of the precision of a test instrument. It is calculated on the basis of sample data using the sample standard deviation and the sample reliability coefficient. Intraclass correlation coefficients (ICCs) for BPI-SF worst pain rating from day 1 to day 8 and week 105 to week 109 in patients whose FACT-B overall QOL ratings change by <10% are 0.685 (n = 926) and 0.800 (n = 109), respectively.b Alternatively referred to as Cohen's d, the effect size is calculated by dividing the difference between the pretest and posttest scores by the standard deviation at pretest. The standard deviation of BPI-SF worst pain rating at baseline (n = 1,877) is 2.849.c Alternatively referred to as the responsiveness statistic, Guyatt's statistic is calculated by dividing the difference between pretest and posttest changes by the standard deviation of change observed for a group of stable patients. The standard deviation of change in BPI-SF worst pain rating from baseline to week 25 in patients whose ECOG performance rating does not change (n = 1,120) is 2.833.
Integrating Anchor-Based and Distribution-Based Mid Estimates
The distribution-based analyses suggest that the MDC for the worst pain rating, defined as the smallest change that can be reliably differentiated from random fluctuation, is between 1.3 and 1.6 points (see Table 5). This represents the lower bound for establishing the MID.
The results from regression analyses can be used to translate changes between anchors and corresponding changes in BPI-SF worst pain. This strategy can be particularly informative when the MID for an anchor is known. This is the case for the EQ-5D Health State Index, where the MID has been estimated at 0.06 for U.S. Index scores and 0.07 for U.K. Index scores.23 A one-point change in EQ-5D Index translates to a change of −3.548 in BPI-SF worst pain, so a 0.07-point change in EQ-5D Index (the MID for the measure) corresponds to a change of −0.248 in BPI-SF worst pain. In contrast, a one-point change in BPI-SF worst pain (which is smaller than the MID based upon the distribution-based analyses) translates to a change of 0.036 for the EQ-5D Index score (considerably smaller than the MID of 0.07). However, a two-point change in BPI-SF worst pain rating corresponds to a 0.072 change in EQ-5D Index score, which is almost identical to the MID for that measure. This suggests that a two-point change may be a reasonable estimate for the MID of the BPI-SF worst pain rating.
Discussion
Data from both distribution-based and anchor-based approaches were used to develop estimates of the MID for the BPI-SF worst pain rating. Results from these approaches are similar, providing reasonably strong support for establishing a two-point MID for the BPI-SF worst pain rating. Further, the results suggest that this estimate of MID is, for the most part, independent of baseline BPI-SF worst pain ratings. However, there is some evidence to suggest that the direction of change (improvement or worsening) may be important to consider. A number of reports have suggested that a smaller change may be required to be considered clinically important when a patient is improving compared with worsening.13 Also, when considered as a percentage, a one-point change in any scale has a different value for an increase versus a decrease; eg, a change from 2 to 3 is an increase of 50%, while a change from 3 to 2 is a decrease of 33%. Nonetheless, these findings provide important information to researchers for interpreting changes in the BPI-SF worst pain ratings.
In addition, although not specific to the BPI worst pain rating, the findings of this study are consistent with other published MID analyses for a similar item. A recent review of three studies concluded that, for a numerical rating scale of pain intensity ranging 0–10 similar in content to the BPI-SF worst pain rating, changes of around two points represent “meaningful,” “much better,” or “much improved” reductions in chronic pain.24
Several factors contribute to the overall strength of the current results. First, as frequently recommended in the literature,11 both anchor-based and distribution-based methods were used to estimate the MID for the worst pain rating. Second, analyses were based on a large sample, totaling over 1,500 patients for the baseline to week 25 assessment interval. A larger sample size will generally provide a broader distribution of responses, which will likely increase the generalizability of the results. Third, multiple anchors were used to evaluate changes in BPI-SF worst pain ratings. Fourth, analyses were performed across several assessment intervals to determine the strongest relationship between BPI-SF ratings and other anchors. Finally, the regression analyses provide important information about whether baseline differences influence the relationship between BPI-SF and other PRO measures.
Nevertheless, these analyses are not without certain limitations. The sample for the current analyses consisted entirely of breast cancer patients. It is unclear to what extent these results will be relevant for other patient populations. Further research is needed to determine whether the MID for the BPI-SF worst pain rating established in this sample has broader applicability. Also, it must be noted that the recall period varied across assessments. The BPI-SF focuses on the past 24 hours, the FACT uses the past week, and the EQ-5D uses the present moment. It is unclear to what extent these differences in recall periods may have influenced the current results. Finally, the baseline to week 25 interval was used to determine the MID for the BPI-SF worst pain rating based on the higher correlations for this interval. Data from baseline to week 49 are consistent with these results, providing some confirmatory evidence to suggest that these MID estimates are stable.
In conclusion, the findings of the present analyses suggest that the MID estimate for the BPI-SF worst pain rating is two points. This value provides guidance to researchers using the BPI-SF worst pain rating on how to interpret baseline differences as well as change scores in the BPI-SF worst pain rating. Additional analyses could be done in other populations to confirm these findings.
References1
1 K.W. Wyrwich, M. Bullinger and N. Aaronson et al., Estimating clinically significant differences in quality of life outcomes, Qual Life Res 14 (2005), pp. 285–295. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (119)
2 S.D. Mathias, S.K. Gao, M. Rutstein, C.F. Snyder, A.W. Wu and D. Cella, Evaluating clinically meaningful change on the ITP-PAQ: preliminary estimates of minimal important differences, Curr Med Res Opin 25 (2) (2009), pp. 375–383. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)
3 K.J. Yost, M.V. Sorensen, E.A. Hahn, G.A. Glendenning, A. Gnanasakthy and D. Cella, Using multiple anchor- and distribution-based estimates to evaluate clinically meaningful change on the Functional Assessment of Cancer Therapy-Biologic Response Modifiers (FACT-BRM) instrument, Value Health 8 (2) (2005), pp. 117–127. Abstract | | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (28)
4 R.D. Hays and J.M. Woolley, The concept of clinically meaningful difference in health-related quality-of-life research: How meaningful is it?, Pharmacoeconomics 18 (5) (2000), pp. 419–423. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (177)
5 R.L. Daut, C.S. Cleeland and R.C. Flanery, Development of the Wisconsin Brief Pain Questionnaire to assess pain in cancer and other diseases, Pain 17 (2) (1983), pp. 197–210. Abstract | | View Record in Scopus | Cited By in Scopus (543)
6 C. Cleeland, Brief Pain Inventory User Guide, University of Texas M. D. Anderson Cancer Center, Houston (2009).
7 R.H. Dworkin, D.C. Turk and J.T. Farrar et al., Core outcome measures for chronic pain clinical trials: IMMPACT recommendations, Pain 113 (1–2) (2005), pp. 9–19. Article | | View Record in Scopus | Cited By in Scopus (380)
8 U.S. Department of Health and Human Services Food and Drug Administration (FDA), Guidance for Industry: Patient-Reported Outcome Measures: Use in Medical Product Development to Support Labeling Claims, FDA, Silver Spring, MD (2009).
9 M.M. Oken, R.H. Creech and D.C. Tormey et al., Toxicity and response criteria of the Eastern Cooperative Oncology Group, Am J Clin Oncol 5 (6) (1982), pp. 649–655. View Record in Scopus | Cited By in Scopus (1968)
10 M.J. Brady, D.F. Cella, F. Mo and A.E. Bonomi et al., Reliability and validity of the Functional Assessment of Cancer Therapy–Breast Cancer Quality of Life instrument, J Clin Oncol 15 (1997), pp. 974–986. View Record in Scopus | Cited By in Scopus (360)
11 R.D. Crosby, R.L. Kolotkin and G.R. Williams, Defining clinically meaningful change in health-related quality of life, J Clin Epidemiol 56 (5) (2003), pp. 395–407. Article | | View Record in Scopus | Cited By in Scopus (233)
12 D. Revicki, R.D. Hays, D. Cella and J. Sloan, Recommended methods for determining responsiveness and minimally important differences for patient-reported outcomes, J Clin Epidemiol 61 (2) (2008), pp. 102–109. Article | | View Record in Scopus | Cited By in Scopus (121)
13 D. Cella, E.A. Hahn and K. Dineen, Meaningful change in cancer-specific quality of life scores: differences between improvement and worsening, Qual Life Res 11 (3) (2002), pp. 207–221. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (137)
14 D.T. Eton, D. Cella and K.J. Yost et al., A combination of distribution- and anchor-based approaches determined the minimally important differences (MIDs) for four endpoints in a breast cancer scale, J Clin Epidemiol 57 (2004), pp. 898–910. Article | | View Record in Scopus | Cited By in Scopus (68)
15 S. Weibe, S. Matijevic, M. Eliasziw and P.A. Derry, Clinically important change in quality of life in epilepsy, J Neurol Neurosurg Psychiatry 73 (2002), pp. 116–120.
16 K.L. Miller, J.G. Walt and D.R. Mink et al., Minimal clinically important difference for the ocular surface disease index, Arch Ophthalmol 128 (1) (2010), pp. 94–101. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
17 K.W. Wyrwich, W.M. Tierney and F.D. Wolinsky, Further evidence supporting an SEM-based criterion for identifying meaningful intra-individual changes in health-related quality of life, J Clin Epidemiol 52 (9) (1999), pp. 861–873. Article | | View Record in Scopus | Cited By in Scopus (272)
18 F.D. Wolinsky, G.J. Wan and W.M. Tierney, Changes in the SF-36 in 12 months in a clinical sample of disadvantaged older adults, Med Care 36 (11) (1998), pp. 1589–1598. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (33)
19 J. Cohen, Statistical Power Analysis for the Behavioral Sciences (2nd ed.), Lawrence Erlbaum, Hillsdale, NJ (1988).
20 G.H. Guyatt, C. Bombardier and P.X. Tugwell, Measuring disease-specific quality of life in clinical trials, CMAJ 134 (8) (1986), pp. 889–895. View Record in Scopus | Cited By in Scopus (324)
21 G.R. Norman, P. Stratford and G. Regehr, Methodological problems in the retrospective computation of responsiveness to change: the lesson of Cronbach, J Clin Epidemiol 50 (8) (1997), pp. 869–879. Article | | View Record in Scopus | Cited By in Scopus (230)
22 A. Stopeck, J. Body and Y. Fujiwara et al., Denosumab versus zoledronic acid for the treatment of breast cancer patients with bone metastases: results of a randomized phase 3 study, Eur J Cancer Suppl 7 (2009), p. 2. Abstract |
23 A.S. Pickard, M.P. Neary and D. Cella, Estimation of minimally important differences in EQ-5D utility and VAS scores in cancer, Health Qual Life Outcomes 5 (2007), p. 70. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
24 R.H. Dworkin, D.C. Turk and K.W. Wyrwich et al., Interpreting the clinical importance of treatment outcomes in chronic pain clinical trials: IMMPACT recommendations, J Pain 9 (2) (2008), pp. 105–121. Article | | View Record in Scopus | Cited By in Scopus (190)
Correspondence to: Susan D. Mathias, Health Outcomes Solutions, PO Box 2343, Winter Park, FL 32790; telephone: (407) 643-9016; fax: (866) 384-0194
Symptom Experience in Patients with Gynecological Cancers: The Development of Symptom Clusters through Patient Narratives
Original research
Violeta Lopez RN, PhD
Abstract
The vast majority of the increasing cancer literature on physical and psychological symptom clusters is quantitative, attempting either to model clusters through statistical techniques or to test priori clusters for their strength of relationship. Narrative symptom clusters can be particularly sensitive outcomes that can generate conceptually meaningful hypotheses for symptom cluster research. We conducted a study to explore the explanation of patients about the development and coexistence of symptoms and how patients attempted to self-manage them. We collected 12-month qualitative longitudinal data over four assessment points consisting of 39 interview data sets from 10 participants with gynecological cancer. Participants' experiences highlighted the presence of physical and psychological symptom clusters, complicating the patients' symptom experience that often lasted 1 year. While some complementary and self-management approaches were used to manage symptoms, few options and interventions were discussed. The cancer care team may be able to develop strategies for a more thorough patient assessment of symptoms reported as the most bothersome and patient-centered sensitive interventions that encompass the physiological, psychological, sociocultural, and behavioral components of the symptom experience essential for effective symptom management.
Article Outline
The physical effects on women after being diagnosed with gynecological cancer are often attributed not only to the symptoms arising from the disease itself but, most importantly, from the side effects of treatment such as surgery, chemotherapy, and radiotherapy.[3], [4] and [5] Symptoms such as fatigue, frequency of urination, bleeding, weight loss, and ascites are commonly experienced by patients, particularly those with ovarian cancers.6 Once diagnosed, gynecological cancer patients often go on to face a prolonged course of treatments which contribute to further symptoms such as chemotherapy-induced alopecia,7 dermatological toxicity,8 fatigue, sleep disturbance,9 nausea, vomiting, and sexual dysfunction.10 Portenoy et al.11 reported that ovarian cancer patients alone experienced a mean of 10.2 symptoms with a range of 0–25 concurrent symptoms. Similarly, 13.4 concurrent symptoms were reported in a study of 49 women undergoing chemotherapy, which caused disruption to the patients' quality of life.6
The psychological state of patients with gynecological cancers has also been investigated, particularly in association with increased risks of psychological morbidity such as anxiety and depression.2 In a longitudinal study of women with ovarian cancer, Gonçalves et al.12 found that neuroticism was associated with persistent psychological morbidity and suggested the need for routine and regular psychological screening for cancer patients. Newly diagnosed women with gynecological cancer also appeared to experience diverse psychological symptomatology that persisted over the first 6 weeks after the diagnosis.2
The relationship between symptom experience, distress produced, and quality of life has also been pursued, of particular interest being the direct correlation between improvement of symptoms and increased quality of life. Ferrell et al.3 found that ovarian cancer patients not only experienced distress but often differently ordered the importance of symptoms at different phases of their illness. They also found that these patients utilized resourcefulness and innovative ideas to manage their symptoms. These authors suggested that symptom experience may be associated with, and can be mediated by, the influence of variables such as disease state, demographic and clinical characteristics, or individual and psychological factors.3 It is therefore unsurprising that treatment-induced symptoms have been a major concern of most studies to gather information about symptoms arising from residual treatment or disease progression as well as frequency and types of symptoms.5 To date, longitudinal studies have yet to be undertaken to gather information prospectively about gynecological cancer patients' symptom experiences. Consequently, the patients' personal experiences of physical and psychological symptoms, such as their concerns, perceptions, and responses to symptoms, remain largely unexplored. Such information is important in the development of interventions for symptom management and the provision of supportive care. Also, while some literature exists in relation to ovarian cancer symptoms, minimal related work has focused on other types of gynecological cancer, suggesting a gap in the literature.
The aim of our study was to explore the physical and psychological symptom experience in patients with gynecological cancer undergoing radiotherapy and/or chemotherapy over the first year from diagnosis. Specific objectives of the study were to (1) qualitatively assess the possible relationships among symptoms resulting from cancer treatments in patients with gynecological cancer, as understood by patients, and (2) explore how patients with gynecological cancer manage the symptoms they experience.
Methods
A descriptive qualitative longitudinal design using face-to-face interviews was used in this study. Qualitative descriptive methods serve to provide descriptions of facts about a phenomenon.13 Sandelowski14 elucidates that qualitative descriptive research methods lend themselves to the data to produce comprehensively and accurately detailed summaries of different participants' experiences of the same event. Interviews were conducted by an experienced qualitative researcher. Interviews were conducted prospectively over four time periods: beginning of treatment (T1) and three (T2), six (T3), and 12 months (T4) later. This time frame was chosen as these are the critical times over which patients with cancer most commonly experience symptoms as a result of treatments or disease progression.15 Leventhal and Johnson's16 self-regulation theory was used as the study's theoretical framework, assisting us in developing the interview guide around symptom identification, exploration of meaning and consequence, and attempts to control or manage it. Their self-regulation theory suggests that symptoms activate a cognitive search process, which results in the construction or elaboration of illness representation. These representations then serve as standards against which new information is matched and evaluated. Comparisons of current sensations with cognitive representations allow for interpretation of new symptoms and for evaluation of the seriousness of current symptoms. Hence, fear behaviors (distress) or instrumental behaviors (coping) are the result of simultaneous parallel psychophysiological processes in response to the threatening experience. The response may be different from individual to individual, based on past experience and the cognitive processes involved, as may the strategies used to cope with the experience. Dodd et al17 simplified the symptom experience as including an individual's perception of a symptom, evaluation of the meaning of a symptom, and response to a symptom.
After approval from the ethics committee, patients were recruited from a large specialist oncology center in the UK a few weeks after diagnosis and prior to commencement of adjuvant treatment. Patients were provided with information about the study, and written consent was obtained. Ten patients were recruited from a list of consecutive newly diagnosed patients through purposeful sampling, and five declined participation, primarily due to the long-term commitment necessary for the study and being too upset with the diagnosis. Maximum variation was used13 to capture core experiences and central, shared aspects or impacts of having a gynecological cancer rather than confining to specific aspects of different types of gynecological cancer. The sample included patients with any type of gynecological cancer and those receiving chemotherapy and/or radiotherapy. Patients with cognitive impairment, metastasis with central nervous system involvement, or life expectancy of less than 6 months at recruitment or who were unable to carry out the interview were excluded. Patients initially were provided with brief information from their oncologist; upon showing an interest, potential participants were provided with a detailed information sheet and had a discussion with the research nurse. Upon agreement, patients signed a consent form and the first interview was scheduled. Participants were followed up for one year. Past experience, judgment on the quality of the data obtained, and data saturation were the key determinants in the decision to have a sample size of 10 over four times (=40 possible transcripts) with the possibility of recruiting more if data were not saturated with the initial sample, although in our study this did not need to take place.
An interview guide was used, starting with a broad question, such as “How have you been feeling physically this last week?” This was followed by questions relating to the psychological symptoms participants experienced, how these related to their physical symptom experience, what they thought when a symptom occurred, what impact the symptoms had on their life, and how they managed the symptoms. New issues identified in the early interviews were incorporated into the interview guide for subsequent interviews. Each interview lasted about an hour to an hour and a half. Interviews were conducted in the patients' homes. Information about sociodemographic characteristics including age, education, and marital status was obtained from patients, who completed an initial sociodemographic form. Disease- and treatment-related information (diagnosis, treatment received, stage of cancer) was obtained from the patients' medical notes. Interviews were recorded and transcribed verbatim.
Data were analyzed line by line using content analysis to code the content of each interview and to map major categories. Categories were compared by two of the researchers, the project lead investigator and another independent person. The analyzed categories were compared and discussed until agreement was reached. Symptoms that were expressed in T1 were grouped together if more than two participants spontaneously mentioned an association between at least two of the symptoms. In T2–T4 we continued this process, focusing primarily on changes in the initial cluster. Symptoms were grouped together as patients discussed them, and if patients reported the same symptom in different contexts, this was coded separately. No participant was asked for specific symptoms as the questions in the interview were broad to allow for important aspects of the symptom experience in each woman and each interview to surface. A final consensus was sought after comparisons and discussions for all categories.13
Credibility of the qualitative data was maintained by ensuring voluntary participation. Analyzed data were constantly discussed and checked by two independent persons, which acted as a constant peer-review process to ensure the analyzed data were true findings and free from potential bias. All interviews were audiotaped, and participants' verbatim quotes were provided to represent categories and subcategories identified, which further ensured reliability by reducing the risk of selective data filtering by the investigators through recall or summation. Consistency was maintained by comparing initial categories within and across the data gathered from the participants to ensure repeatability of the categories. Field notes were reviewed as a kind of inquiry audit to prevent potential bias and to ensure the stability of data.
Results
Patient Characteristics
All 10 participants completed the interviews at the four time points, over the course of one year. One interview transcript (at T3) was subsequently found to be unusable, due to tape recorder malfunction, and was excluded from the analysis, thus leaving a total of 39 data sets of interviews over four time points. The mean age of the group was 62.8 years (SD = 7.7, range 51–72). Most were married (n = 6), two were separated, and two were widowed. The majority (n = 7) had secondary/high school education. Six were retired, two were homemakers, and two reported technical/manual work. Half of the participants (5/10) had ovarian cancer, while the rest had uterine (1/10), cervical (2/10), or endometrial (1/10) cancer and one had both uterine and cervical cancer. Seven participants had surgery. Of the 10 participants, six had chemotherapy, three had radiotherapy, and one had chemotherapy followed by radiotherapy. Chemotherapy included carboplatin (n = 2), carboplatin and paclitaxel (Taxol) (n = 5), and cisplatin (n = 1). Half the patients were at an early disease stage (stage 1 or 2, n = 5), three were at stage 3, one was at stage 4, and the stage was unknown in one patient.
Qualitative Data
Patients identified symptoms in an interlinked manner rather than in isolation, suggesting some symptom clustering. There was always a key symptom mentioned together with several others that were reported as co-occurring or resulting symptoms. These associations and explanations helped patients to make sense of the symptoms and rationalize or legitimize the complexity of the symptom relationships and the difficulty in having control over the symptoms. Participants gave meaning to the physical symptoms experienced alongside psychological responses and how they managed to alleviate them. The meaning element was fairly stable across times as women discussed primarily the occurrence of symptoms, their impact on their lives, and their struggle to cope with them. What was an evident change in the perception, however, for most symptoms was the frustration from the “chronicity” of symptoms and the differential impact of symptoms at different times in their disease trajectory. Symptoms were described as co-occurring with one influencing others, giving an understanding of the formation of symptom clusters. Four major narrative symptom clusters emerged from the data.
Tiredness, sleeplessness, pain, depression, and weakness
The most common symptom experienced by all patients that persisted over the 12-month period was tiredness, which was also related with sleep disturbance associated with pain, tingling sensation of the hands and feet, and anxiety. Tiredness was experienced throughout the year as recounted by several participants:
“Basically, it's after the effects of the treatment that I was feeling tired cos I'm having radiotherapy every day and chemotherapy once a week.” (GYP 01 at T1)“Just me body felt tired, me body felt tired of it [radiotherapy]. I felt rotten. I couldn't do anything. It's depressing.” (GYP 12 at T2)
“Physically, I'm alright. I felt alright except I get tired easily, but other than that, I feel alright.” (GYP06 at T3)
“What I do find is that it's depressing to feel physically tired all the time and I don't know why.” (GYP04 at T4)
Difficulties with sleep were also associated with depression. For some (4/10) participants, feelings of depression occurred as they went through the treatment, as two participants described:
“I've actually felt quite depressed this last week and actually burst into tears, which is something I haven't done before. But it was sort of overwhelming—these symptoms.” (GYP08 at T3)“These symptoms are affecting my sleep. If I can't sleep, I get tired and I can't do anything the next day, then I get depressed.” (GYP06 at T3)
Depression was often the result of uncertainty and fear. Half of the participants expressed feelings of uncertainty lasting until the twelfth month, as highlighted by two of them:
“It's very uncertain you know. It's a very uncertain way of life. You don't know … like I go and see … I go back every three months for check-up. You're living your life for check-ups. So every four months you're thinking … ‘Is everything alright?’ You live your life for those four months' check-ups. The frightening bit is the uncertainty of it and it's depressing just thinking of what will happen next.” (GYP04 at T3)“I can't sleep thinking about what will happen all the time. The uncertainty keeps you awake.” (GYP012 at T3)
However, this feeling seemed to subside by T4 as they became more in control and able to cope by not dwelling on it and just accepting the disease, its treatment, and symptoms. This marked shift in coping styles in T4 characterized the patients' increased positive response to symptoms at this stage.
When participants were tired, they also complained of weakness. The expressions used included “muscle tone” and “muscle strength” weakness. However, a slight improvement in this symptom was observed from T3 onward. Tiredness and its related symptoms affected the participants' ability to get on with usual routine housework; however, support from husbands, family, and friends helped the participants to cope with their symptoms:
“I've got such a strong group of friends and relatives and they all live around me … so all the family are around me and they'll all come at least once a day.” (GYP11 at T4)
Very few management options were discussed. For both tiredness and weakness, participants used a variety of self-management approaches such as taking a rest even for half an hour each day or doing some physical fitness like walking and pilates from the third month onward. To get their muscle tone back to normal, some participants expressed their desire to “get fit,” so they walked or went cycling. Two participants reported taking herbal medicines, such as echinacea, after surgery and before chemotherapy. They perceived this as helpful in building up their immune system. Pain was managed by taking painkillers, as prescribed by their doctors. Praying was also reported as a strategy used to combat the anxieties related to the illness.
Hair loss, ocular changes, body image, identity experience, and anxiety
Hair loss, including body hair such as eyebrows and eyelashes, was reported by four participants, all of whom had received chemotherapy. In the beginning, participants did not want to wear wigs and were anxious that it portrayed a symbol to others that they had cancer:
“I do not want my family to see me without hair as they will know I have cancer and then they will start to worry about me.” (GYP10 at T1)
Later on, they no longer cared whether they wore a wig or not, especially when they themselves and others accepted the situation. Such an experience was described as follows:
“I noticed my hair falling and got me a wig the first day.” (GYP11 at T1)“I noticed that … the hairs in my nose, I think have all disappeared as well.” (GYP10 at T2)
“My hair came back like Shirley Temple, curly but it all came back slate gray and I didn't like it.” (GYP11 at T4)
On a couple of occasions, negative feelings were externalized through talking about someone else, often a famous person. This may have facilitated the expression of difficult emotions. Such a transference is depicted below:
“My hair is more or less gone but this time my husband just shaved it all off. I bung my hat on and that's it. I feel sorry for young girls. It must be horrendous cos I think of Kylie [Minogue, pop singer], for someone like her to lose her hair must have been terrible.” (GYP03 at T3)
Although participants understood that hair loss was an expected and common consequence of chemotherapy, its impact in some patients was more difficult to accept. Losing hair was seen as a realization that they had cancer or increased one's identity as a cancer patient. The same patient also talked about hair loss and anxiety:
“I think one of the most difficult things is that you might be feeling alright physically and then you've got a bald head. When I put my wig on, I feel alright. But no hair is a big thing—even though people say you looked alright without it, you don't.” (GYP03 at T3)
In some participants, hair loss, especially eyelashes, was connected with blurred vision as they believed that eyelashes protected their eyes, as reported by one (1/10) participant at T2 and three (3/9) participants at T3, around the end of their treatment.
“My eyes seem as though they're a bit blurred. I feel like I need eyeglasses. It's a thing that annoyed me most. It just felt like there's a film on your eyes. I wonder if it's something to do with hair loss.” (GYP10 at T2)
One participant was worried about her blurred vision being associated with other health problems:
“I've had slightly sort of, not blurred vision but zig-zaggy vision that made me a bit … ‘Oh dear,' you know … have I got a brain tumor? … a strange sort of thing that upsets my balance.” (GYP08 at T3)
Gastrointestinal problems: nausea, loss of appetite, taste changes, bowel function, weight changes, and distress
Nausea, appetite changes, and changes in bowel function (diarrhea or constipation) were the most common symptoms reported in relation to gastrointestinal system problems that distressed patients. Patients who reported these occurrences received either chemotherapy or radiotherapy, and one received both. However, these symptoms were of limited extent as women reported them as mild or of less importance and perceived them as more manageable. Nausea was only reported by one participant at T1 and was relieved by prescribed medications. Another participant reported loss of taste throughout the year of the study. Loss of appetite was reported by one participant at T2 and T3. Weight loss, which three of the participants attributed to diarrhea, was also reported:
“My tummy's a bit off, had a bit of diarrhea, but that's the norm.” (GYP05 at T1)“I lost weight but then again, I don't know if that's down to eating then going to the toilet.” (GYP04 at T3)
However, weight gain was reported by six participants at T3 and five participants at T4, which was the key distressing nutritional problem described, although for some it was seen in a more positive way:
“I put on weight since the radium. But it's a small price to pay isn't it? A bit of weight for all that you've gone through.” (GYP15 at T4)
Numbness and tingling sensations in the hands and feet, restlessness, sleeplessness, and depression
A common physical problem experienced by three of the participants who all received chemotherapy was tingling sensations of the hands and feet, which increased over time. At T4, three out of 10 participants still experienced numbness and tingling sensations as described by one participant:
“I was worst after my last treatment, all sorts, my feet, my fingers were really bad … always tingly. My feet they're numb and I get cramps. It's weird, they get too cold. It's depressing especially if I can't sleep.” (GYP10 at T3)“I still got funny toes and fingers. They feel fat and podgy. It's depressing. It's difficult to explain, it feels like because they're not dead but … I know I've got them, if that makes any sense. I find it difficult to spread them.” (GYP10 at T4)
Participants sometimes related the sensation to achy joint pains, as if they were getting the flu. This sensation also made them feel restless at night, contributing to sleeplessness. One participant related this to the side effect of paclitaxel (Taxol); therefore, her medication was changed to liposomal doxorubicin. Two participants tried to self-manage the feeling of coldness and numbness of their feet and fingers by soaking them in hot water. For those participants who experienced sleeplessness, due to feelings of numbness and tingling sensations in the hands and feet, wearing bed socks or soaking them in warm water, as well as using reiki and massage, were the management strategies described.
Discussion
This study explored the explanations of patients about the development and coexistence of symptoms and how patients attempted to self-manage them. This is one of the few studies in the literature, and the only one in gynecological cancer, which has explored clusters of symptoms in a narrative manner. Its longitudinal nature, unusual in qualitative research due to the inherent issues in the analysis of such data, was another strength of the study as it allowed us to explore shifts in the symptom experience, perception, and meaning over time (although meaning was fairly stable and participants talked little about it). The vast majority of the increasing literature on symptom clusters is quantitative, attempting either to model clusters through statistical techniques or to test priori clusters for their strength of relationship. However, such clusters may be biased, not only from the technique used but also from the content of self-reports utilized to collect the data. The narrative symptom clusters could rectify problems with statistical measures as they reflect the unique patient experience in the patients' own words and can assist in the development of (patient-centered rather than statistically based) symptom clusters that can then be tested quantitatively with larger samples. Hence, narrative symptom clusters can be particularly sensitive outcomes and can generate conceptually meaningful hypotheses for symptom cluster research.
Key symptoms experienced by the participants were tiredness, pain, body image changes, gastrointestinal changes, and peripheral neuropathy associated with chemotherapy, which concur with past studies of primarily ovarian cancer patients.[9] and [10] Out of the four clusters identified, one is applicable to all patients irrespective of treatment and two are clearly linked with chemotherapy. Symptoms varied in intensity but tended to subside in a year's time for the majority of patients. Acceptance brought about self-management strategies to overcome both the physical and psychological effects of cancer and its treatment, but most important was the support they received from families and friends. In addition, the fact that some symptoms decreased over time may be due to some symptoms being linked to the time since the end of treatment; such symptoms could have naturally resolved after completion of treatment. However, we have limited information on the natural history of symptoms in patients with different types of gynecological cancer.
As with other studies, the most common symptom experienced by these participants was tiredness,[3], [9] and [18] often associated with sleep disturbance due to pain, peripheral neuropathy, change in bowel function, and depression. Because these women complained of tiredness throughout the year, having social support from their husbands, families, friends, and neighbors helped them carry on with their usual household roles. This highlighted the important role caregivers play in supporting patients with cancer. Participants clearly differentiated between the symptom of tiredness/fatigue (a complex symptom involving physical, mental, and motivational aspects) from weakness (which is related more to muscle strength). This differentiation is evident in the literature,19 and while they may be related symptoms, they should be assessed separately as they may necessitate different management strategies. Dodd et al20have shown the clustering of the symptoms of fatigue, sleep disturbance, pain, and depression in breast cancer patients, similarly to the work of Liu et al.21 This quantitative work and our narrative cluster strongly support the existence and clinical relevance of this symptom cluster.
Loss of weight in the beginning was due to gastrointestinal disturbance including nausea, loss of taste, and change in bowel function. These findings concur with the literature, where such symptoms are prevalent up to one year posttreatment.22 However, as time passed, the participants regained their weight, often above their prediagnosis level. Such a gastrointestinal symptom cluster has also been supported in the quantitative literature on symptom clusters, although the relevant items within the cluster very much depend on the items included in the data-collection scale.23 Our own work with 143 patients over one year (n = 504 symptom assessments) has also identified a gastrointestinal cluster, with the key symptom being weight loss, together with loss of appetite and difficulties swallowing, experienced by up to one-quarter of a heterogeneous sample of cancer patients at the one-year time point.24 The attempts highlighted by the majority of participants to control their weight suggest that this is an important issue for these women. The inability to control weight may be frustrating and a key stressor in women with gynecological cancer. This assertion needs further investigation as the information we have about this topic to date derives almost exclusively from breast cancer patients. Weight control may be an important component of survivorship in these women, and it should be incorporated in the follow-up care of patients beyond breast cancer.25 While the use of medication was mentioned with regard to the presence of gastrointestinal symptoms, no interventions have taken place with taste changes and other nutritional concerns. Interventions around the experience and enjoyment of eating and food should be an important research focus in the future, as should work around weight gain, for which we currently have limited information.
Concern about hair loss was mentioned by only four participants. They were concerned mainly that it identified them to others as a cancer patient as baldness became the main element of the cancer patients' everyday life and identity.7 Participants were not concerned about their self-image but rather more concerned about protecting others (particularly family) and being treated differently. For these participants, they accepted that loss of hair was a side effect of treatment and viewed regrowth of hair as a positive effect, which concurred with the study by Sun et al.10 Ocular changes reported by three participants (out of 10) during treatment and up to six months later are an underreported issue in the literature. This is despite the established association between some types of chemotherapy (ie, cyclophosphamide, cisplatin) and ocular changes. More focus should be directed to this area, as well as to identifying reversible and irreversible ocular changes in the survivorship period. The narrative clustering of symptoms such as hair loss and ocular changes (connected with being “visible” cancer patients) with body image, identity, and anxiety is an interesting clustering of physical and psychological interrelated symptoms, with body image being the key symptom in this cluster. Our past work has highlighted the relationship between body image changes and “disliking” self in up to 20% of the sample, although these did not cluster together after the six-month assessment point (end of treatments) and were most visible during the chemotherapy period.24With the exception of using wigs, patients did not mention using any interventions regarding the multiple symptoms experienced within this symptom cluster, suggesting that this is an important area of research in the future.
Many of the physical symptoms reported were interrelated with descriptions of depression, uncertainty, body image, and identity as a cancer patient. While causal relationships cannot be ascertained from such a qualitative design, it is evident that there is a close relationship between the presence of physical symptoms, psychological status, and the impact of them in life. This is confirmed in a systematic review of studies with ovarian cancer patients26 as well as other studies that support the association of symptoms of fatigue, pain, anxiety, and depression with quality of life.27 A better understanding of these relationships is paramount in symptom-management efforts, particularly as it is recognized that interventions need to be multimodal and to target more than one concurrent symptom, a clear message that comes from the symptom cluster research.28 For the majority of the participants, when symptoms were not managed well, they experienced psychological responses such as depression, commonly seen in the literature. Participants in our study accepted that they would experience these symptoms, although their occurrence and intensity differed from patient to patient. Some participants were able to tolerate treatment with little physical discomfort, while in others symptoms prevented them from resuming usual functional activities and social roles, thus leading to feelings of depression. It would be interesting to identify in future research the factors that allow some patients to live well with cancer, moving away from the current research model of ill-health to a model of “wellness.”
Furthermore, a key distressing symptom that was associated with impairments in a variety of life areas was peripheral neuropathy in patients receiving chemotherapy. This cluster has some similarities with the tiredness cluster (i.e. the, presence of sleep difficulties or depression), but women talked about it as a separate experience from tiredness. Peripheral neuropathy is a difficult symptom to manage in practice and may necessitate dose reductions or chemotherapy discontinuation; therefore, the development of management strategies for this symptom is imperative. Women complained of sleeping difficulties when they experienced numbness and tingling sensations in their hands and feet, and it was a frustrating symptom that was also associated with depression. This is another symptom cluster that merits further research, and it is only emerging in the literature. Our past work on symptom clusters has also identified the presence of a “hand–foot” symptom cluster, which consistently increased over time and suggested a chronic nature, although not all symptoms identified in the present study were part of the latter quantitative evaluation.24
It was surprising to see the minimal range of interventions used to manage symptoms, both self-management and formal ones directed by the clinicians. The latter had minimal presence in the women's descriptions, with the exception of pain management, antiemetics, and antidiarrheal medication. Specific physical responses to treatment were dealt with by changing the type of chemotherapy and resorting to the use of herbal medicine for symptom management, such as echinacea and red clover, or other simple self-management techniques, such as soaking the feet in warm water, eating healthy food, and carrying out regular exercise. The use of complementary therapies was also found in the study by Ferrell et al,3 complementing the medical care provided to help control the symptoms.6 However, all of these attempts seemed to be ad hoc, with no clear understanding of processes and possible outcomes or any guidance about their use from health-care professionals. Possible reasons for this ad hoc and unsatisfactory underutilization of symptom-management interventions may include the “acceptance” by patients that some symptoms are part of their treatment, the British cultural norm of not complaining, limited confidence from both clinicians and patients on nonpharmacological interventions with variable quality of evidence of effectiveness, distance from patients' homes to the specialist service to provide supportive care, limited understanding from the clinicians of the impact of symptoms on patients' lives, and clinician time constraints.
Although we purposely included women with a range of gynecological cancer diagnoses in order to gain an understanding of broadly applicable issues related to the physical and psychological symptom experiences of patients, the small sample size limits the generalizability of the results. These symptom clusters will need further evaluation with statistical modeling. Also, the existence of symptom clusters in radiotherapy may not have surfaced well in our study with the inclusion of only three patients receiving radiotherapy, and this needs to be explored in future research. The length of treatment for each patient was variable, and knowledge of this would have enhanced the interpretability of the findings; however, this information was not available to us. Finally, although we wanted to focus on treatment-related symptoms, some of the symptoms (ie, anxiety and depression) may have multiple possible etiologies; and this needs to be considered in the interpretation of the findings.
Conclusion
Our study provides information on symptom experiences from the patients' perspective, which could lead to a better understanding of how patients perceive, assess, monitor, and manage their symptoms. This is particularly useful as the majority of the symptom literature focuses on the experience of patients with ovarian cancer. This is also important background information in developing strategies or interventions that are patient-centered and sensitive to the needs of patients that has relevance to the current policy framework. It also highlights the need for a more thorough patient assessment, to assess which symptoms are most bothersome and how symptoms are interrelated, and has implications for the physiological, psychological, sociocultural, and behavioral components of the symptom experience essential for effective symptom management. While we have identified the presence and experience of some well-documented symptoms, we have also highlighted areas of importance for patients and some underreported symptoms that merit further research in the future. Narrative symptom clusters, such as those identified in the present study, can provide a stronger conceptual basis in the symptom cluster modeling work and can assist in identifying patient-relevant and clinically meaningful groups of symptoms that can be the focus of future cluster research.
References1
1 R.L. Johnson, M.A. Gold and K.F. Wyche, Distress in women with gynecologic cancer, Psychooncology 19 (2010), pp. 665–668. View Record in Scopus | Cited By in Scopus (1)
2 R.W. Petersen, G. Graham and J.A. Quinlivan, Psychological changes after a gynaecologic cancer, J Obstet Gynecol Res 31 (2006), pp. 152–157.
3 B. Ferrell, S. Smith, C. Cullinane and C. Melancon, Symptom concerns of women with ovarian cancer, J Pain Symptom Manage 25 (2003), pp. 528–538. Article | | View Record in Scopus | Cited By in Scopus (16)
4 J. Hipkins, M. Whitworth, N. Tarrier and G. Jayson, Social support, anxiety and depression after chemotherapy for ovarian cancer: a prospective study, Br J Health Psychol 9 (pt 4) (2004), pp. 569–581. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (36)
5 P.D. Williams, U. Piamjariyakul, K.A. Ducey, J. Badura, D. Boltz, K. Olberding, A. Wingate and A.R. Williams, Cancer treatment, symptom monitoring, and self-care in adults, Cancer Nurs 29 (2006), pp. 347–355. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
6 H.S. Donovan, E.M. Hartenbach and M.W. Method, Patient–provider communication and perceived control for women experiencing multiple symptoms associated with ovarian cancer, Gynecol Oncol 99 (2005), pp. 404–411. Article | | View Record in Scopus | Cited By in Scopus (17)
7 S. Rosman, Cancer and stigma: experience of patients with chemotherapy-induced alopecia, Patient Educ Couns 52 (2004), pp. 333–339. Article | | View Record in Scopus | Cited By in Scopus (33)
8 M. Hackbarth, N. Haas, C. Fotopoulou, W. Lichtenegger and J. Schouli, Chemotherapy-induced dermatological toxicity: frequencies and impact on quality of life in women's cancer: results of a prospective study, Support Care Cancer 16 (2008), pp. 267–273. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
9 A.E. Kayl and C.A. Meyers, Side-effects of chemotherapy and quality of life in ovarian and breast cancer patients, Curr Opin Obstet Gynecol 18 (2006), pp. 24–28. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
10 C.C. Sun, D.C. Bodurka, C.B. Weaver, R. Rasu, J.K. Wolf, M.W. Beyers, J.A. Smith and J.T. Wharton, Rankings and symptom assessments of side effects from chemotherapy: insights from experiences patients with ovarian cancer, Support Care Cancer 13 (2005), pp. 219–227. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (53)
11 R.K. Portenoy, H.T. Thaler, A.B. Kornblith, J. McCarthy-Lepore, H. Friedlander-Klar, N. Coyle, T. Smart-Curley, N. Kemeny, L. Norton, W. Hoskins and H. Scher, Symptom prevalence, characteristics and distress in a cancer population, Qual Life Res 3 (1994), pp. 183–189. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (281)
12 V. Gonçalves, G. Jayson and N. Tarrier, A longitudinal investigation of psychological morbidity in patients with ovarian cancer, Br J Cancer 99 (2008), pp. 1794–1801. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
13 M.Q. Patton, Qualitative Evaluation and Research Methods (2nd ed.), Sage Publications, Newbury Park, CA (1990).
14 M. Sandelowski, Real qualitative researchers do not count: the use of numbers in qualitative research, Res Nurs Health 24 (2001), pp. 230–240. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
15 S.S. Hwang, V.T. Chang, D.L. Fairclough, J. Cogswell and B. Kasimis, Longitudinal quality of life in advanced cancer patients: pilot study results from a VA Medical Cancer Center, J Pain Symptom Manage 25 (2003), pp. 225–235. Article | | View Record in Scopus | Cited By in Scopus (46)
16 H. Leventhal and J.E. Johnson, Laboratory and field experimentation: development of a theory of self-regulation. In: J.P. Wooldridge, M.H. Schmitt, J.K. Skipper and R.C. Leonard, Editors, Behavioral Science and Nursing Theory, Mosby, St. Louis (1983), pp. 189–264.
17 M. Dodd, S. Janson, N. Facione, J. Faucett and E.S. Froelicher et al., Advancing the science of symptom management, J Adv Nurs 33 (2001), pp. 668–676. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (173)
18 C.C. Sun, D.C. Bodurka, M.L. Donato, E.B. Rubenstein, C.L. Borden, K. Basen-Engquist, M.S. Munsell, J.J. Kavanagh and D.M. Gershenson, Patient preferences regarding side effects of chemotherapy for ovarian cancer: do they change over time?, Gynecol Oncol 87 (2002), pp. 118–128. Abstract | | View Record in Scopus | Cited By in Scopus (22)
19 S.C. Teunissen, W. Wesker, C. Kruitwagen, H.C. de Haes, E.E. Voest and A. de Graeff, Symptom prevalence in patients with incurable cancer: a systematic review, J Pain Symptom Manage 34 (2007), pp. 94–104. Article | | View Record in Scopus | Cited By in Scopus (67)
20 M.J. Dodd, M.H. Cho, B.A. Cooper and C. Miaskowski, The effect of symptom clusters on functional status and quality of life in women with breast cancer, Eur J Oncol Nurs 14 (2010), pp. 101–110. Article | | View Record in Scopus | Cited By in Scopus (6)
21 L. Liu, L. Fiorentino, L. Natarajan, B.A. Parker, P.J. Mills, G.R. Sadler, J.E. Dimsdale, M. Rissling, F. He and S. Ancoli-Israel, Pre-treatment symptom cluster in breast cancer patients is associated with worse sleep, fatigue and depression during chemotherapy, Psychooncology 18 (2009), pp. 187–194. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (0)
22 H. Tong, E. Isenring and P. Yates, The prevalence of nutrition impact symptoms and their relationship to quality of life and clinical outcomes in medical oncology patients, Support Care Cancer 17 (2009), pp. 83–90. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
23 H.J. Kim, A.M. Barsevick, L. Tulman and P.A. McDermott, Treatment-related symptom clusters in breast cancer: a secondary analysis, J Pain Symptom Manage 36 (2008), pp. 468–479. Article | | View Record in Scopus | Cited By in Scopus (12)
24 A. Molassiotis, Y. Wengstrom and N. Kearney, Symptom cluster patterns during the first year from the diagnosis with cancer, J Pain Symptom Manage 39 (2010), pp. 847–858. Article | | View Record in Scopus | Cited By in Scopus (1)
25 L. Muraca, D. Leung, A. Clark, M.A. Beduz and P. Goodwin, Breast cancer survivors: taking charge of lifestyle choices after treatment, Eur J Oncol Nurs (2010) 10.1016/e.ejon.2009.12.001.
26 E. Arden-Close, Y. Gidron and R. Moss-Morris, Psychological distress and its correlates in ovarian cancer: a systematic review, Psychooncology 17 (2008), pp. 1061–1072. View Record in Scopus | Cited By in Scopus (7)
27 W.K.W. So, G. Marsh, W.M. Ling, F.Y. Leung, J.C.K. Lo, M. Yeung and G.K.H. Li, The symptom cluster of fatigue, pain anxiety, and depression and the effect on the quality of life of women receiving treatment for breast cancer: a multicentre study, Oncol Nurs Forum 36 (2009), pp. E205–E214. Full Text via CrossRef
28 M.J. Dodd, C. Miaskowski and S.M. Paul, Symptom clusters and their effect on functional status of patients with cancer, Oncol Nurs Forum 28 (2001), pp. 465–470. View Record in Scopus | Cited By in Scopus (192)
Correspondence to: Violeta Lopez, TCH, RCNMP, Building 6, Level 3, East Wing, Yamba Drive, Garran, 2605, Canberra, Australian Capital Territory (ACT), Australia; telephone: +612 6244 2333; fax: +612 6244 2573
Original research
Violeta Lopez RN, PhD
Abstract
The vast majority of the increasing cancer literature on physical and psychological symptom clusters is quantitative, attempting either to model clusters through statistical techniques or to test priori clusters for their strength of relationship. Narrative symptom clusters can be particularly sensitive outcomes that can generate conceptually meaningful hypotheses for symptom cluster research. We conducted a study to explore the explanation of patients about the development and coexistence of symptoms and how patients attempted to self-manage them. We collected 12-month qualitative longitudinal data over four assessment points consisting of 39 interview data sets from 10 participants with gynecological cancer. Participants' experiences highlighted the presence of physical and psychological symptom clusters, complicating the patients' symptom experience that often lasted 1 year. While some complementary and self-management approaches were used to manage symptoms, few options and interventions were discussed. The cancer care team may be able to develop strategies for a more thorough patient assessment of symptoms reported as the most bothersome and patient-centered sensitive interventions that encompass the physiological, psychological, sociocultural, and behavioral components of the symptom experience essential for effective symptom management.
Article Outline
The physical effects on women after being diagnosed with gynecological cancer are often attributed not only to the symptoms arising from the disease itself but, most importantly, from the side effects of treatment such as surgery, chemotherapy, and radiotherapy.[3], [4] and [5] Symptoms such as fatigue, frequency of urination, bleeding, weight loss, and ascites are commonly experienced by patients, particularly those with ovarian cancers.6 Once diagnosed, gynecological cancer patients often go on to face a prolonged course of treatments which contribute to further symptoms such as chemotherapy-induced alopecia,7 dermatological toxicity,8 fatigue, sleep disturbance,9 nausea, vomiting, and sexual dysfunction.10 Portenoy et al.11 reported that ovarian cancer patients alone experienced a mean of 10.2 symptoms with a range of 0–25 concurrent symptoms. Similarly, 13.4 concurrent symptoms were reported in a study of 49 women undergoing chemotherapy, which caused disruption to the patients' quality of life.6
The psychological state of patients with gynecological cancers has also been investigated, particularly in association with increased risks of psychological morbidity such as anxiety and depression.2 In a longitudinal study of women with ovarian cancer, Gonçalves et al.12 found that neuroticism was associated with persistent psychological morbidity and suggested the need for routine and regular psychological screening for cancer patients. Newly diagnosed women with gynecological cancer also appeared to experience diverse psychological symptomatology that persisted over the first 6 weeks after the diagnosis.2
The relationship between symptom experience, distress produced, and quality of life has also been pursued, of particular interest being the direct correlation between improvement of symptoms and increased quality of life. Ferrell et al.3 found that ovarian cancer patients not only experienced distress but often differently ordered the importance of symptoms at different phases of their illness. They also found that these patients utilized resourcefulness and innovative ideas to manage their symptoms. These authors suggested that symptom experience may be associated with, and can be mediated by, the influence of variables such as disease state, demographic and clinical characteristics, or individual and psychological factors.3 It is therefore unsurprising that treatment-induced symptoms have been a major concern of most studies to gather information about symptoms arising from residual treatment or disease progression as well as frequency and types of symptoms.5 To date, longitudinal studies have yet to be undertaken to gather information prospectively about gynecological cancer patients' symptom experiences. Consequently, the patients' personal experiences of physical and psychological symptoms, such as their concerns, perceptions, and responses to symptoms, remain largely unexplored. Such information is important in the development of interventions for symptom management and the provision of supportive care. Also, while some literature exists in relation to ovarian cancer symptoms, minimal related work has focused on other types of gynecological cancer, suggesting a gap in the literature.
The aim of our study was to explore the physical and psychological symptom experience in patients with gynecological cancer undergoing radiotherapy and/or chemotherapy over the first year from diagnosis. Specific objectives of the study were to (1) qualitatively assess the possible relationships among symptoms resulting from cancer treatments in patients with gynecological cancer, as understood by patients, and (2) explore how patients with gynecological cancer manage the symptoms they experience.
Methods
A descriptive qualitative longitudinal design using face-to-face interviews was used in this study. Qualitative descriptive methods serve to provide descriptions of facts about a phenomenon.13 Sandelowski14 elucidates that qualitative descriptive research methods lend themselves to the data to produce comprehensively and accurately detailed summaries of different participants' experiences of the same event. Interviews were conducted by an experienced qualitative researcher. Interviews were conducted prospectively over four time periods: beginning of treatment (T1) and three (T2), six (T3), and 12 months (T4) later. This time frame was chosen as these are the critical times over which patients with cancer most commonly experience symptoms as a result of treatments or disease progression.15 Leventhal and Johnson's16 self-regulation theory was used as the study's theoretical framework, assisting us in developing the interview guide around symptom identification, exploration of meaning and consequence, and attempts to control or manage it. Their self-regulation theory suggests that symptoms activate a cognitive search process, which results in the construction or elaboration of illness representation. These representations then serve as standards against which new information is matched and evaluated. Comparisons of current sensations with cognitive representations allow for interpretation of new symptoms and for evaluation of the seriousness of current symptoms. Hence, fear behaviors (distress) or instrumental behaviors (coping) are the result of simultaneous parallel psychophysiological processes in response to the threatening experience. The response may be different from individual to individual, based on past experience and the cognitive processes involved, as may the strategies used to cope with the experience. Dodd et al17 simplified the symptom experience as including an individual's perception of a symptom, evaluation of the meaning of a symptom, and response to a symptom.
After approval from the ethics committee, patients were recruited from a large specialist oncology center in the UK a few weeks after diagnosis and prior to commencement of adjuvant treatment. Patients were provided with information about the study, and written consent was obtained. Ten patients were recruited from a list of consecutive newly diagnosed patients through purposeful sampling, and five declined participation, primarily due to the long-term commitment necessary for the study and being too upset with the diagnosis. Maximum variation was used13 to capture core experiences and central, shared aspects or impacts of having a gynecological cancer rather than confining to specific aspects of different types of gynecological cancer. The sample included patients with any type of gynecological cancer and those receiving chemotherapy and/or radiotherapy. Patients with cognitive impairment, metastasis with central nervous system involvement, or life expectancy of less than 6 months at recruitment or who were unable to carry out the interview were excluded. Patients initially were provided with brief information from their oncologist; upon showing an interest, potential participants were provided with a detailed information sheet and had a discussion with the research nurse. Upon agreement, patients signed a consent form and the first interview was scheduled. Participants were followed up for one year. Past experience, judgment on the quality of the data obtained, and data saturation were the key determinants in the decision to have a sample size of 10 over four times (=40 possible transcripts) with the possibility of recruiting more if data were not saturated with the initial sample, although in our study this did not need to take place.
An interview guide was used, starting with a broad question, such as “How have you been feeling physically this last week?” This was followed by questions relating to the psychological symptoms participants experienced, how these related to their physical symptom experience, what they thought when a symptom occurred, what impact the symptoms had on their life, and how they managed the symptoms. New issues identified in the early interviews were incorporated into the interview guide for subsequent interviews. Each interview lasted about an hour to an hour and a half. Interviews were conducted in the patients' homes. Information about sociodemographic characteristics including age, education, and marital status was obtained from patients, who completed an initial sociodemographic form. Disease- and treatment-related information (diagnosis, treatment received, stage of cancer) was obtained from the patients' medical notes. Interviews were recorded and transcribed verbatim.
Data were analyzed line by line using content analysis to code the content of each interview and to map major categories. Categories were compared by two of the researchers, the project lead investigator and another independent person. The analyzed categories were compared and discussed until agreement was reached. Symptoms that were expressed in T1 were grouped together if more than two participants spontaneously mentioned an association between at least two of the symptoms. In T2–T4 we continued this process, focusing primarily on changes in the initial cluster. Symptoms were grouped together as patients discussed them, and if patients reported the same symptom in different contexts, this was coded separately. No participant was asked for specific symptoms as the questions in the interview were broad to allow for important aspects of the symptom experience in each woman and each interview to surface. A final consensus was sought after comparisons and discussions for all categories.13
Credibility of the qualitative data was maintained by ensuring voluntary participation. Analyzed data were constantly discussed and checked by two independent persons, which acted as a constant peer-review process to ensure the analyzed data were true findings and free from potential bias. All interviews were audiotaped, and participants' verbatim quotes were provided to represent categories and subcategories identified, which further ensured reliability by reducing the risk of selective data filtering by the investigators through recall or summation. Consistency was maintained by comparing initial categories within and across the data gathered from the participants to ensure repeatability of the categories. Field notes were reviewed as a kind of inquiry audit to prevent potential bias and to ensure the stability of data.
Results
Patient Characteristics
All 10 participants completed the interviews at the four time points, over the course of one year. One interview transcript (at T3) was subsequently found to be unusable, due to tape recorder malfunction, and was excluded from the analysis, thus leaving a total of 39 data sets of interviews over four time points. The mean age of the group was 62.8 years (SD = 7.7, range 51–72). Most were married (n = 6), two were separated, and two were widowed. The majority (n = 7) had secondary/high school education. Six were retired, two were homemakers, and two reported technical/manual work. Half of the participants (5/10) had ovarian cancer, while the rest had uterine (1/10), cervical (2/10), or endometrial (1/10) cancer and one had both uterine and cervical cancer. Seven participants had surgery. Of the 10 participants, six had chemotherapy, three had radiotherapy, and one had chemotherapy followed by radiotherapy. Chemotherapy included carboplatin (n = 2), carboplatin and paclitaxel (Taxol) (n = 5), and cisplatin (n = 1). Half the patients were at an early disease stage (stage 1 or 2, n = 5), three were at stage 3, one was at stage 4, and the stage was unknown in one patient.
Qualitative Data
Patients identified symptoms in an interlinked manner rather than in isolation, suggesting some symptom clustering. There was always a key symptom mentioned together with several others that were reported as co-occurring or resulting symptoms. These associations and explanations helped patients to make sense of the symptoms and rationalize or legitimize the complexity of the symptom relationships and the difficulty in having control over the symptoms. Participants gave meaning to the physical symptoms experienced alongside psychological responses and how they managed to alleviate them. The meaning element was fairly stable across times as women discussed primarily the occurrence of symptoms, their impact on their lives, and their struggle to cope with them. What was an evident change in the perception, however, for most symptoms was the frustration from the “chronicity” of symptoms and the differential impact of symptoms at different times in their disease trajectory. Symptoms were described as co-occurring with one influencing others, giving an understanding of the formation of symptom clusters. Four major narrative symptom clusters emerged from the data.
Tiredness, sleeplessness, pain, depression, and weakness
The most common symptom experienced by all patients that persisted over the 12-month period was tiredness, which was also related with sleep disturbance associated with pain, tingling sensation of the hands and feet, and anxiety. Tiredness was experienced throughout the year as recounted by several participants:
“Basically, it's after the effects of the treatment that I was feeling tired cos I'm having radiotherapy every day and chemotherapy once a week.” (GYP 01 at T1)“Just me body felt tired, me body felt tired of it [radiotherapy]. I felt rotten. I couldn't do anything. It's depressing.” (GYP 12 at T2)
“Physically, I'm alright. I felt alright except I get tired easily, but other than that, I feel alright.” (GYP06 at T3)
“What I do find is that it's depressing to feel physically tired all the time and I don't know why.” (GYP04 at T4)
Difficulties with sleep were also associated with depression. For some (4/10) participants, feelings of depression occurred as they went through the treatment, as two participants described:
“I've actually felt quite depressed this last week and actually burst into tears, which is something I haven't done before. But it was sort of overwhelming—these symptoms.” (GYP08 at T3)“These symptoms are affecting my sleep. If I can't sleep, I get tired and I can't do anything the next day, then I get depressed.” (GYP06 at T3)
Depression was often the result of uncertainty and fear. Half of the participants expressed feelings of uncertainty lasting until the twelfth month, as highlighted by two of them:
“It's very uncertain you know. It's a very uncertain way of life. You don't know … like I go and see … I go back every three months for check-up. You're living your life for check-ups. So every four months you're thinking … ‘Is everything alright?’ You live your life for those four months' check-ups. The frightening bit is the uncertainty of it and it's depressing just thinking of what will happen next.” (GYP04 at T3)“I can't sleep thinking about what will happen all the time. The uncertainty keeps you awake.” (GYP012 at T3)
However, this feeling seemed to subside by T4 as they became more in control and able to cope by not dwelling on it and just accepting the disease, its treatment, and symptoms. This marked shift in coping styles in T4 characterized the patients' increased positive response to symptoms at this stage.
When participants were tired, they also complained of weakness. The expressions used included “muscle tone” and “muscle strength” weakness. However, a slight improvement in this symptom was observed from T3 onward. Tiredness and its related symptoms affected the participants' ability to get on with usual routine housework; however, support from husbands, family, and friends helped the participants to cope with their symptoms:
“I've got such a strong group of friends and relatives and they all live around me … so all the family are around me and they'll all come at least once a day.” (GYP11 at T4)
Very few management options were discussed. For both tiredness and weakness, participants used a variety of self-management approaches such as taking a rest even for half an hour each day or doing some physical fitness like walking and pilates from the third month onward. To get their muscle tone back to normal, some participants expressed their desire to “get fit,” so they walked or went cycling. Two participants reported taking herbal medicines, such as echinacea, after surgery and before chemotherapy. They perceived this as helpful in building up their immune system. Pain was managed by taking painkillers, as prescribed by their doctors. Praying was also reported as a strategy used to combat the anxieties related to the illness.
Hair loss, ocular changes, body image, identity experience, and anxiety
Hair loss, including body hair such as eyebrows and eyelashes, was reported by four participants, all of whom had received chemotherapy. In the beginning, participants did not want to wear wigs and were anxious that it portrayed a symbol to others that they had cancer:
“I do not want my family to see me without hair as they will know I have cancer and then they will start to worry about me.” (GYP10 at T1)
Later on, they no longer cared whether they wore a wig or not, especially when they themselves and others accepted the situation. Such an experience was described as follows:
“I noticed my hair falling and got me a wig the first day.” (GYP11 at T1)“I noticed that … the hairs in my nose, I think have all disappeared as well.” (GYP10 at T2)
“My hair came back like Shirley Temple, curly but it all came back slate gray and I didn't like it.” (GYP11 at T4)
On a couple of occasions, negative feelings were externalized through talking about someone else, often a famous person. This may have facilitated the expression of difficult emotions. Such a transference is depicted below:
“My hair is more or less gone but this time my husband just shaved it all off. I bung my hat on and that's it. I feel sorry for young girls. It must be horrendous cos I think of Kylie [Minogue, pop singer], for someone like her to lose her hair must have been terrible.” (GYP03 at T3)
Although participants understood that hair loss was an expected and common consequence of chemotherapy, its impact in some patients was more difficult to accept. Losing hair was seen as a realization that they had cancer or increased one's identity as a cancer patient. The same patient also talked about hair loss and anxiety:
“I think one of the most difficult things is that you might be feeling alright physically and then you've got a bald head. When I put my wig on, I feel alright. But no hair is a big thing—even though people say you looked alright without it, you don't.” (GYP03 at T3)
In some participants, hair loss, especially eyelashes, was connected with blurred vision as they believed that eyelashes protected their eyes, as reported by one (1/10) participant at T2 and three (3/9) participants at T3, around the end of their treatment.
“My eyes seem as though they're a bit blurred. I feel like I need eyeglasses. It's a thing that annoyed me most. It just felt like there's a film on your eyes. I wonder if it's something to do with hair loss.” (GYP10 at T2)
One participant was worried about her blurred vision being associated with other health problems:
“I've had slightly sort of, not blurred vision but zig-zaggy vision that made me a bit … ‘Oh dear,' you know … have I got a brain tumor? … a strange sort of thing that upsets my balance.” (GYP08 at T3)
Gastrointestinal problems: nausea, loss of appetite, taste changes, bowel function, weight changes, and distress
Nausea, appetite changes, and changes in bowel function (diarrhea or constipation) were the most common symptoms reported in relation to gastrointestinal system problems that distressed patients. Patients who reported these occurrences received either chemotherapy or radiotherapy, and one received both. However, these symptoms were of limited extent as women reported them as mild or of less importance and perceived them as more manageable. Nausea was only reported by one participant at T1 and was relieved by prescribed medications. Another participant reported loss of taste throughout the year of the study. Loss of appetite was reported by one participant at T2 and T3. Weight loss, which three of the participants attributed to diarrhea, was also reported:
“My tummy's a bit off, had a bit of diarrhea, but that's the norm.” (GYP05 at T1)“I lost weight but then again, I don't know if that's down to eating then going to the toilet.” (GYP04 at T3)
However, weight gain was reported by six participants at T3 and five participants at T4, which was the key distressing nutritional problem described, although for some it was seen in a more positive way:
“I put on weight since the radium. But it's a small price to pay isn't it? A bit of weight for all that you've gone through.” (GYP15 at T4)
Numbness and tingling sensations in the hands and feet, restlessness, sleeplessness, and depression
A common physical problem experienced by three of the participants who all received chemotherapy was tingling sensations of the hands and feet, which increased over time. At T4, three out of 10 participants still experienced numbness and tingling sensations as described by one participant:
“I was worst after my last treatment, all sorts, my feet, my fingers were really bad … always tingly. My feet they're numb and I get cramps. It's weird, they get too cold. It's depressing especially if I can't sleep.” (GYP10 at T3)“I still got funny toes and fingers. They feel fat and podgy. It's depressing. It's difficult to explain, it feels like because they're not dead but … I know I've got them, if that makes any sense. I find it difficult to spread them.” (GYP10 at T4)
Participants sometimes related the sensation to achy joint pains, as if they were getting the flu. This sensation also made them feel restless at night, contributing to sleeplessness. One participant related this to the side effect of paclitaxel (Taxol); therefore, her medication was changed to liposomal doxorubicin. Two participants tried to self-manage the feeling of coldness and numbness of their feet and fingers by soaking them in hot water. For those participants who experienced sleeplessness, due to feelings of numbness and tingling sensations in the hands and feet, wearing bed socks or soaking them in warm water, as well as using reiki and massage, were the management strategies described.
Discussion
This study explored the explanations of patients about the development and coexistence of symptoms and how patients attempted to self-manage them. This is one of the few studies in the literature, and the only one in gynecological cancer, which has explored clusters of symptoms in a narrative manner. Its longitudinal nature, unusual in qualitative research due to the inherent issues in the analysis of such data, was another strength of the study as it allowed us to explore shifts in the symptom experience, perception, and meaning over time (although meaning was fairly stable and participants talked little about it). The vast majority of the increasing literature on symptom clusters is quantitative, attempting either to model clusters through statistical techniques or to test priori clusters for their strength of relationship. However, such clusters may be biased, not only from the technique used but also from the content of self-reports utilized to collect the data. The narrative symptom clusters could rectify problems with statistical measures as they reflect the unique patient experience in the patients' own words and can assist in the development of (patient-centered rather than statistically based) symptom clusters that can then be tested quantitatively with larger samples. Hence, narrative symptom clusters can be particularly sensitive outcomes and can generate conceptually meaningful hypotheses for symptom cluster research.
Key symptoms experienced by the participants were tiredness, pain, body image changes, gastrointestinal changes, and peripheral neuropathy associated with chemotherapy, which concur with past studies of primarily ovarian cancer patients.[9] and [10] Out of the four clusters identified, one is applicable to all patients irrespective of treatment and two are clearly linked with chemotherapy. Symptoms varied in intensity but tended to subside in a year's time for the majority of patients. Acceptance brought about self-management strategies to overcome both the physical and psychological effects of cancer and its treatment, but most important was the support they received from families and friends. In addition, the fact that some symptoms decreased over time may be due to some symptoms being linked to the time since the end of treatment; such symptoms could have naturally resolved after completion of treatment. However, we have limited information on the natural history of symptoms in patients with different types of gynecological cancer.
As with other studies, the most common symptom experienced by these participants was tiredness,[3], [9] and [18] often associated with sleep disturbance due to pain, peripheral neuropathy, change in bowel function, and depression. Because these women complained of tiredness throughout the year, having social support from their husbands, families, friends, and neighbors helped them carry on with their usual household roles. This highlighted the important role caregivers play in supporting patients with cancer. Participants clearly differentiated between the symptom of tiredness/fatigue (a complex symptom involving physical, mental, and motivational aspects) from weakness (which is related more to muscle strength). This differentiation is evident in the literature,19 and while they may be related symptoms, they should be assessed separately as they may necessitate different management strategies. Dodd et al20have shown the clustering of the symptoms of fatigue, sleep disturbance, pain, and depression in breast cancer patients, similarly to the work of Liu et al.21 This quantitative work and our narrative cluster strongly support the existence and clinical relevance of this symptom cluster.
Loss of weight in the beginning was due to gastrointestinal disturbance including nausea, loss of taste, and change in bowel function. These findings concur with the literature, where such symptoms are prevalent up to one year posttreatment.22 However, as time passed, the participants regained their weight, often above their prediagnosis level. Such a gastrointestinal symptom cluster has also been supported in the quantitative literature on symptom clusters, although the relevant items within the cluster very much depend on the items included in the data-collection scale.23 Our own work with 143 patients over one year (n = 504 symptom assessments) has also identified a gastrointestinal cluster, with the key symptom being weight loss, together with loss of appetite and difficulties swallowing, experienced by up to one-quarter of a heterogeneous sample of cancer patients at the one-year time point.24 The attempts highlighted by the majority of participants to control their weight suggest that this is an important issue for these women. The inability to control weight may be frustrating and a key stressor in women with gynecological cancer. This assertion needs further investigation as the information we have about this topic to date derives almost exclusively from breast cancer patients. Weight control may be an important component of survivorship in these women, and it should be incorporated in the follow-up care of patients beyond breast cancer.25 While the use of medication was mentioned with regard to the presence of gastrointestinal symptoms, no interventions have taken place with taste changes and other nutritional concerns. Interventions around the experience and enjoyment of eating and food should be an important research focus in the future, as should work around weight gain, for which we currently have limited information.
Concern about hair loss was mentioned by only four participants. They were concerned mainly that it identified them to others as a cancer patient as baldness became the main element of the cancer patients' everyday life and identity.7 Participants were not concerned about their self-image but rather more concerned about protecting others (particularly family) and being treated differently. For these participants, they accepted that loss of hair was a side effect of treatment and viewed regrowth of hair as a positive effect, which concurred with the study by Sun et al.10 Ocular changes reported by three participants (out of 10) during treatment and up to six months later are an underreported issue in the literature. This is despite the established association between some types of chemotherapy (ie, cyclophosphamide, cisplatin) and ocular changes. More focus should be directed to this area, as well as to identifying reversible and irreversible ocular changes in the survivorship period. The narrative clustering of symptoms such as hair loss and ocular changes (connected with being “visible” cancer patients) with body image, identity, and anxiety is an interesting clustering of physical and psychological interrelated symptoms, with body image being the key symptom in this cluster. Our past work has highlighted the relationship between body image changes and “disliking” self in up to 20% of the sample, although these did not cluster together after the six-month assessment point (end of treatments) and were most visible during the chemotherapy period.24With the exception of using wigs, patients did not mention using any interventions regarding the multiple symptoms experienced within this symptom cluster, suggesting that this is an important area of research in the future.
Many of the physical symptoms reported were interrelated with descriptions of depression, uncertainty, body image, and identity as a cancer patient. While causal relationships cannot be ascertained from such a qualitative design, it is evident that there is a close relationship between the presence of physical symptoms, psychological status, and the impact of them in life. This is confirmed in a systematic review of studies with ovarian cancer patients26 as well as other studies that support the association of symptoms of fatigue, pain, anxiety, and depression with quality of life.27 A better understanding of these relationships is paramount in symptom-management efforts, particularly as it is recognized that interventions need to be multimodal and to target more than one concurrent symptom, a clear message that comes from the symptom cluster research.28 For the majority of the participants, when symptoms were not managed well, they experienced psychological responses such as depression, commonly seen in the literature. Participants in our study accepted that they would experience these symptoms, although their occurrence and intensity differed from patient to patient. Some participants were able to tolerate treatment with little physical discomfort, while in others symptoms prevented them from resuming usual functional activities and social roles, thus leading to feelings of depression. It would be interesting to identify in future research the factors that allow some patients to live well with cancer, moving away from the current research model of ill-health to a model of “wellness.”
Furthermore, a key distressing symptom that was associated with impairments in a variety of life areas was peripheral neuropathy in patients receiving chemotherapy. This cluster has some similarities with the tiredness cluster (i.e. the, presence of sleep difficulties or depression), but women talked about it as a separate experience from tiredness. Peripheral neuropathy is a difficult symptom to manage in practice and may necessitate dose reductions or chemotherapy discontinuation; therefore, the development of management strategies for this symptom is imperative. Women complained of sleeping difficulties when they experienced numbness and tingling sensations in their hands and feet, and it was a frustrating symptom that was also associated with depression. This is another symptom cluster that merits further research, and it is only emerging in the literature. Our past work on symptom clusters has also identified the presence of a “hand–foot” symptom cluster, which consistently increased over time and suggested a chronic nature, although not all symptoms identified in the present study were part of the latter quantitative evaluation.24
It was surprising to see the minimal range of interventions used to manage symptoms, both self-management and formal ones directed by the clinicians. The latter had minimal presence in the women's descriptions, with the exception of pain management, antiemetics, and antidiarrheal medication. Specific physical responses to treatment were dealt with by changing the type of chemotherapy and resorting to the use of herbal medicine for symptom management, such as echinacea and red clover, or other simple self-management techniques, such as soaking the feet in warm water, eating healthy food, and carrying out regular exercise. The use of complementary therapies was also found in the study by Ferrell et al,3 complementing the medical care provided to help control the symptoms.6 However, all of these attempts seemed to be ad hoc, with no clear understanding of processes and possible outcomes or any guidance about their use from health-care professionals. Possible reasons for this ad hoc and unsatisfactory underutilization of symptom-management interventions may include the “acceptance” by patients that some symptoms are part of their treatment, the British cultural norm of not complaining, limited confidence from both clinicians and patients on nonpharmacological interventions with variable quality of evidence of effectiveness, distance from patients' homes to the specialist service to provide supportive care, limited understanding from the clinicians of the impact of symptoms on patients' lives, and clinician time constraints.
Although we purposely included women with a range of gynecological cancer diagnoses in order to gain an understanding of broadly applicable issues related to the physical and psychological symptom experiences of patients, the small sample size limits the generalizability of the results. These symptom clusters will need further evaluation with statistical modeling. Also, the existence of symptom clusters in radiotherapy may not have surfaced well in our study with the inclusion of only three patients receiving radiotherapy, and this needs to be explored in future research. The length of treatment for each patient was variable, and knowledge of this would have enhanced the interpretability of the findings; however, this information was not available to us. Finally, although we wanted to focus on treatment-related symptoms, some of the symptoms (ie, anxiety and depression) may have multiple possible etiologies; and this needs to be considered in the interpretation of the findings.
Conclusion
Our study provides information on symptom experiences from the patients' perspective, which could lead to a better understanding of how patients perceive, assess, monitor, and manage their symptoms. This is particularly useful as the majority of the symptom literature focuses on the experience of patients with ovarian cancer. This is also important background information in developing strategies or interventions that are patient-centered and sensitive to the needs of patients that has relevance to the current policy framework. It also highlights the need for a more thorough patient assessment, to assess which symptoms are most bothersome and how symptoms are interrelated, and has implications for the physiological, psychological, sociocultural, and behavioral components of the symptom experience essential for effective symptom management. While we have identified the presence and experience of some well-documented symptoms, we have also highlighted areas of importance for patients and some underreported symptoms that merit further research in the future. Narrative symptom clusters, such as those identified in the present study, can provide a stronger conceptual basis in the symptom cluster modeling work and can assist in identifying patient-relevant and clinically meaningful groups of symptoms that can be the focus of future cluster research.
References1
1 R.L. Johnson, M.A. Gold and K.F. Wyche, Distress in women with gynecologic cancer, Psychooncology 19 (2010), pp. 665–668. View Record in Scopus | Cited By in Scopus (1)
2 R.W. Petersen, G. Graham and J.A. Quinlivan, Psychological changes after a gynaecologic cancer, J Obstet Gynecol Res 31 (2006), pp. 152–157.
3 B. Ferrell, S. Smith, C. Cullinane and C. Melancon, Symptom concerns of women with ovarian cancer, J Pain Symptom Manage 25 (2003), pp. 528–538. Article | | View Record in Scopus | Cited By in Scopus (16)
4 J. Hipkins, M. Whitworth, N. Tarrier and G. Jayson, Social support, anxiety and depression after chemotherapy for ovarian cancer: a prospective study, Br J Health Psychol 9 (pt 4) (2004), pp. 569–581. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (36)
5 P.D. Williams, U. Piamjariyakul, K.A. Ducey, J. Badura, D. Boltz, K. Olberding, A. Wingate and A.R. Williams, Cancer treatment, symptom monitoring, and self-care in adults, Cancer Nurs 29 (2006), pp. 347–355. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
6 H.S. Donovan, E.M. Hartenbach and M.W. Method, Patient–provider communication and perceived control for women experiencing multiple symptoms associated with ovarian cancer, Gynecol Oncol 99 (2005), pp. 404–411. Article | | View Record in Scopus | Cited By in Scopus (17)
7 S. Rosman, Cancer and stigma: experience of patients with chemotherapy-induced alopecia, Patient Educ Couns 52 (2004), pp. 333–339. Article | | View Record in Scopus | Cited By in Scopus (33)
8 M. Hackbarth, N. Haas, C. Fotopoulou, W. Lichtenegger and J. Schouli, Chemotherapy-induced dermatological toxicity: frequencies and impact on quality of life in women's cancer: results of a prospective study, Support Care Cancer 16 (2008), pp. 267–273. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
9 A.E. Kayl and C.A. Meyers, Side-effects of chemotherapy and quality of life in ovarian and breast cancer patients, Curr Opin Obstet Gynecol 18 (2006), pp. 24–28. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
10 C.C. Sun, D.C. Bodurka, C.B. Weaver, R. Rasu, J.K. Wolf, M.W. Beyers, J.A. Smith and J.T. Wharton, Rankings and symptom assessments of side effects from chemotherapy: insights from experiences patients with ovarian cancer, Support Care Cancer 13 (2005), pp. 219–227. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (53)
11 R.K. Portenoy, H.T. Thaler, A.B. Kornblith, J. McCarthy-Lepore, H. Friedlander-Klar, N. Coyle, T. Smart-Curley, N. Kemeny, L. Norton, W. Hoskins and H. Scher, Symptom prevalence, characteristics and distress in a cancer population, Qual Life Res 3 (1994), pp. 183–189. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (281)
12 V. Gonçalves, G. Jayson and N. Tarrier, A longitudinal investigation of psychological morbidity in patients with ovarian cancer, Br J Cancer 99 (2008), pp. 1794–1801. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
13 M.Q. Patton, Qualitative Evaluation and Research Methods (2nd ed.), Sage Publications, Newbury Park, CA (1990).
14 M. Sandelowski, Real qualitative researchers do not count: the use of numbers in qualitative research, Res Nurs Health 24 (2001), pp. 230–240. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
15 S.S. Hwang, V.T. Chang, D.L. Fairclough, J. Cogswell and B. Kasimis, Longitudinal quality of life in advanced cancer patients: pilot study results from a VA Medical Cancer Center, J Pain Symptom Manage 25 (2003), pp. 225–235. Article | | View Record in Scopus | Cited By in Scopus (46)
16 H. Leventhal and J.E. Johnson, Laboratory and field experimentation: development of a theory of self-regulation. In: J.P. Wooldridge, M.H. Schmitt, J.K. Skipper and R.C. Leonard, Editors, Behavioral Science and Nursing Theory, Mosby, St. Louis (1983), pp. 189–264.
17 M. Dodd, S. Janson, N. Facione, J. Faucett and E.S. Froelicher et al., Advancing the science of symptom management, J Adv Nurs 33 (2001), pp. 668–676. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (173)
18 C.C. Sun, D.C. Bodurka, M.L. Donato, E.B. Rubenstein, C.L. Borden, K. Basen-Engquist, M.S. Munsell, J.J. Kavanagh and D.M. Gershenson, Patient preferences regarding side effects of chemotherapy for ovarian cancer: do they change over time?, Gynecol Oncol 87 (2002), pp. 118–128. Abstract | | View Record in Scopus | Cited By in Scopus (22)
19 S.C. Teunissen, W. Wesker, C. Kruitwagen, H.C. de Haes, E.E. Voest and A. de Graeff, Symptom prevalence in patients with incurable cancer: a systematic review, J Pain Symptom Manage 34 (2007), pp. 94–104. Article | | View Record in Scopus | Cited By in Scopus (67)
20 M.J. Dodd, M.H. Cho, B.A. Cooper and C. Miaskowski, The effect of symptom clusters on functional status and quality of life in women with breast cancer, Eur J Oncol Nurs 14 (2010), pp. 101–110. Article | | View Record in Scopus | Cited By in Scopus (6)
21 L. Liu, L. Fiorentino, L. Natarajan, B.A. Parker, P.J. Mills, G.R. Sadler, J.E. Dimsdale, M. Rissling, F. He and S. Ancoli-Israel, Pre-treatment symptom cluster in breast cancer patients is associated with worse sleep, fatigue and depression during chemotherapy, Psychooncology 18 (2009), pp. 187–194. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (0)
22 H. Tong, E. Isenring and P. Yates, The prevalence of nutrition impact symptoms and their relationship to quality of life and clinical outcomes in medical oncology patients, Support Care Cancer 17 (2009), pp. 83–90. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
23 H.J. Kim, A.M. Barsevick, L. Tulman and P.A. McDermott, Treatment-related symptom clusters in breast cancer: a secondary analysis, J Pain Symptom Manage 36 (2008), pp. 468–479. Article | | View Record in Scopus | Cited By in Scopus (12)
24 A. Molassiotis, Y. Wengstrom and N. Kearney, Symptom cluster patterns during the first year from the diagnosis with cancer, J Pain Symptom Manage 39 (2010), pp. 847–858. Article | | View Record in Scopus | Cited By in Scopus (1)
25 L. Muraca, D. Leung, A. Clark, M.A. Beduz and P. Goodwin, Breast cancer survivors: taking charge of lifestyle choices after treatment, Eur J Oncol Nurs (2010) 10.1016/e.ejon.2009.12.001.
26 E. Arden-Close, Y. Gidron and R. Moss-Morris, Psychological distress and its correlates in ovarian cancer: a systematic review, Psychooncology 17 (2008), pp. 1061–1072. View Record in Scopus | Cited By in Scopus (7)
27 W.K.W. So, G. Marsh, W.M. Ling, F.Y. Leung, J.C.K. Lo, M. Yeung and G.K.H. Li, The symptom cluster of fatigue, pain anxiety, and depression and the effect on the quality of life of women receiving treatment for breast cancer: a multicentre study, Oncol Nurs Forum 36 (2009), pp. E205–E214. Full Text via CrossRef
28 M.J. Dodd, C. Miaskowski and S.M. Paul, Symptom clusters and their effect on functional status of patients with cancer, Oncol Nurs Forum 28 (2001), pp. 465–470. View Record in Scopus | Cited By in Scopus (192)
Correspondence to: Violeta Lopez, TCH, RCNMP, Building 6, Level 3, East Wing, Yamba Drive, Garran, 2605, Canberra, Australian Capital Territory (ACT), Australia; telephone: +612 6244 2333; fax: +612 6244 2573
Original research
Violeta Lopez RN, PhD
Abstract
The vast majority of the increasing cancer literature on physical and psychological symptom clusters is quantitative, attempting either to model clusters through statistical techniques or to test priori clusters for their strength of relationship. Narrative symptom clusters can be particularly sensitive outcomes that can generate conceptually meaningful hypotheses for symptom cluster research. We conducted a study to explore the explanation of patients about the development and coexistence of symptoms and how patients attempted to self-manage them. We collected 12-month qualitative longitudinal data over four assessment points consisting of 39 interview data sets from 10 participants with gynecological cancer. Participants' experiences highlighted the presence of physical and psychological symptom clusters, complicating the patients' symptom experience that often lasted 1 year. While some complementary and self-management approaches were used to manage symptoms, few options and interventions were discussed. The cancer care team may be able to develop strategies for a more thorough patient assessment of symptoms reported as the most bothersome and patient-centered sensitive interventions that encompass the physiological, psychological, sociocultural, and behavioral components of the symptom experience essential for effective symptom management.
Article Outline
The physical effects on women after being diagnosed with gynecological cancer are often attributed not only to the symptoms arising from the disease itself but, most importantly, from the side effects of treatment such as surgery, chemotherapy, and radiotherapy.[3], [4] and [5] Symptoms such as fatigue, frequency of urination, bleeding, weight loss, and ascites are commonly experienced by patients, particularly those with ovarian cancers.6 Once diagnosed, gynecological cancer patients often go on to face a prolonged course of treatments which contribute to further symptoms such as chemotherapy-induced alopecia,7 dermatological toxicity,8 fatigue, sleep disturbance,9 nausea, vomiting, and sexual dysfunction.10 Portenoy et al.11 reported that ovarian cancer patients alone experienced a mean of 10.2 symptoms with a range of 0–25 concurrent symptoms. Similarly, 13.4 concurrent symptoms were reported in a study of 49 women undergoing chemotherapy, which caused disruption to the patients' quality of life.6
The psychological state of patients with gynecological cancers has also been investigated, particularly in association with increased risks of psychological morbidity such as anxiety and depression.2 In a longitudinal study of women with ovarian cancer, Gonçalves et al.12 found that neuroticism was associated with persistent psychological morbidity and suggested the need for routine and regular psychological screening for cancer patients. Newly diagnosed women with gynecological cancer also appeared to experience diverse psychological symptomatology that persisted over the first 6 weeks after the diagnosis.2
The relationship between symptom experience, distress produced, and quality of life has also been pursued, of particular interest being the direct correlation between improvement of symptoms and increased quality of life. Ferrell et al.3 found that ovarian cancer patients not only experienced distress but often differently ordered the importance of symptoms at different phases of their illness. They also found that these patients utilized resourcefulness and innovative ideas to manage their symptoms. These authors suggested that symptom experience may be associated with, and can be mediated by, the influence of variables such as disease state, demographic and clinical characteristics, or individual and psychological factors.3 It is therefore unsurprising that treatment-induced symptoms have been a major concern of most studies to gather information about symptoms arising from residual treatment or disease progression as well as frequency and types of symptoms.5 To date, longitudinal studies have yet to be undertaken to gather information prospectively about gynecological cancer patients' symptom experiences. Consequently, the patients' personal experiences of physical and psychological symptoms, such as their concerns, perceptions, and responses to symptoms, remain largely unexplored. Such information is important in the development of interventions for symptom management and the provision of supportive care. Also, while some literature exists in relation to ovarian cancer symptoms, minimal related work has focused on other types of gynecological cancer, suggesting a gap in the literature.
The aim of our study was to explore the physical and psychological symptom experience in patients with gynecological cancer undergoing radiotherapy and/or chemotherapy over the first year from diagnosis. Specific objectives of the study were to (1) qualitatively assess the possible relationships among symptoms resulting from cancer treatments in patients with gynecological cancer, as understood by patients, and (2) explore how patients with gynecological cancer manage the symptoms they experience.
Methods
A descriptive qualitative longitudinal design using face-to-face interviews was used in this study. Qualitative descriptive methods serve to provide descriptions of facts about a phenomenon.13 Sandelowski14 elucidates that qualitative descriptive research methods lend themselves to the data to produce comprehensively and accurately detailed summaries of different participants' experiences of the same event. Interviews were conducted by an experienced qualitative researcher. Interviews were conducted prospectively over four time periods: beginning of treatment (T1) and three (T2), six (T3), and 12 months (T4) later. This time frame was chosen as these are the critical times over which patients with cancer most commonly experience symptoms as a result of treatments or disease progression.15 Leventhal and Johnson's16 self-regulation theory was used as the study's theoretical framework, assisting us in developing the interview guide around symptom identification, exploration of meaning and consequence, and attempts to control or manage it. Their self-regulation theory suggests that symptoms activate a cognitive search process, which results in the construction or elaboration of illness representation. These representations then serve as standards against which new information is matched and evaluated. Comparisons of current sensations with cognitive representations allow for interpretation of new symptoms and for evaluation of the seriousness of current symptoms. Hence, fear behaviors (distress) or instrumental behaviors (coping) are the result of simultaneous parallel psychophysiological processes in response to the threatening experience. The response may be different from individual to individual, based on past experience and the cognitive processes involved, as may the strategies used to cope with the experience. Dodd et al17 simplified the symptom experience as including an individual's perception of a symptom, evaluation of the meaning of a symptom, and response to a symptom.
After approval from the ethics committee, patients were recruited from a large specialist oncology center in the UK a few weeks after diagnosis and prior to commencement of adjuvant treatment. Patients were provided with information about the study, and written consent was obtained. Ten patients were recruited from a list of consecutive newly diagnosed patients through purposeful sampling, and five declined participation, primarily due to the long-term commitment necessary for the study and being too upset with the diagnosis. Maximum variation was used13 to capture core experiences and central, shared aspects or impacts of having a gynecological cancer rather than confining to specific aspects of different types of gynecological cancer. The sample included patients with any type of gynecological cancer and those receiving chemotherapy and/or radiotherapy. Patients with cognitive impairment, metastasis with central nervous system involvement, or life expectancy of less than 6 months at recruitment or who were unable to carry out the interview were excluded. Patients initially were provided with brief information from their oncologist; upon showing an interest, potential participants were provided with a detailed information sheet and had a discussion with the research nurse. Upon agreement, patients signed a consent form and the first interview was scheduled. Participants were followed up for one year. Past experience, judgment on the quality of the data obtained, and data saturation were the key determinants in the decision to have a sample size of 10 over four times (=40 possible transcripts) with the possibility of recruiting more if data were not saturated with the initial sample, although in our study this did not need to take place.
An interview guide was used, starting with a broad question, such as “How have you been feeling physically this last week?” This was followed by questions relating to the psychological symptoms participants experienced, how these related to their physical symptom experience, what they thought when a symptom occurred, what impact the symptoms had on their life, and how they managed the symptoms. New issues identified in the early interviews were incorporated into the interview guide for subsequent interviews. Each interview lasted about an hour to an hour and a half. Interviews were conducted in the patients' homes. Information about sociodemographic characteristics including age, education, and marital status was obtained from patients, who completed an initial sociodemographic form. Disease- and treatment-related information (diagnosis, treatment received, stage of cancer) was obtained from the patients' medical notes. Interviews were recorded and transcribed verbatim.
Data were analyzed line by line using content analysis to code the content of each interview and to map major categories. Categories were compared by two of the researchers, the project lead investigator and another independent person. The analyzed categories were compared and discussed until agreement was reached. Symptoms that were expressed in T1 were grouped together if more than two participants spontaneously mentioned an association between at least two of the symptoms. In T2–T4 we continued this process, focusing primarily on changes in the initial cluster. Symptoms were grouped together as patients discussed them, and if patients reported the same symptom in different contexts, this was coded separately. No participant was asked for specific symptoms as the questions in the interview were broad to allow for important aspects of the symptom experience in each woman and each interview to surface. A final consensus was sought after comparisons and discussions for all categories.13
Credibility of the qualitative data was maintained by ensuring voluntary participation. Analyzed data were constantly discussed and checked by two independent persons, which acted as a constant peer-review process to ensure the analyzed data were true findings and free from potential bias. All interviews were audiotaped, and participants' verbatim quotes were provided to represent categories and subcategories identified, which further ensured reliability by reducing the risk of selective data filtering by the investigators through recall or summation. Consistency was maintained by comparing initial categories within and across the data gathered from the participants to ensure repeatability of the categories. Field notes were reviewed as a kind of inquiry audit to prevent potential bias and to ensure the stability of data.
Results
Patient Characteristics
All 10 participants completed the interviews at the four time points, over the course of one year. One interview transcript (at T3) was subsequently found to be unusable, due to tape recorder malfunction, and was excluded from the analysis, thus leaving a total of 39 data sets of interviews over four time points. The mean age of the group was 62.8 years (SD = 7.7, range 51–72). Most were married (n = 6), two were separated, and two were widowed. The majority (n = 7) had secondary/high school education. Six were retired, two were homemakers, and two reported technical/manual work. Half of the participants (5/10) had ovarian cancer, while the rest had uterine (1/10), cervical (2/10), or endometrial (1/10) cancer and one had both uterine and cervical cancer. Seven participants had surgery. Of the 10 participants, six had chemotherapy, three had radiotherapy, and one had chemotherapy followed by radiotherapy. Chemotherapy included carboplatin (n = 2), carboplatin and paclitaxel (Taxol) (n = 5), and cisplatin (n = 1). Half the patients were at an early disease stage (stage 1 or 2, n = 5), three were at stage 3, one was at stage 4, and the stage was unknown in one patient.
Qualitative Data
Patients identified symptoms in an interlinked manner rather than in isolation, suggesting some symptom clustering. There was always a key symptom mentioned together with several others that were reported as co-occurring or resulting symptoms. These associations and explanations helped patients to make sense of the symptoms and rationalize or legitimize the complexity of the symptom relationships and the difficulty in having control over the symptoms. Participants gave meaning to the physical symptoms experienced alongside psychological responses and how they managed to alleviate them. The meaning element was fairly stable across times as women discussed primarily the occurrence of symptoms, their impact on their lives, and their struggle to cope with them. What was an evident change in the perception, however, for most symptoms was the frustration from the “chronicity” of symptoms and the differential impact of symptoms at different times in their disease trajectory. Symptoms were described as co-occurring with one influencing others, giving an understanding of the formation of symptom clusters. Four major narrative symptom clusters emerged from the data.
Tiredness, sleeplessness, pain, depression, and weakness
The most common symptom experienced by all patients that persisted over the 12-month period was tiredness, which was also related with sleep disturbance associated with pain, tingling sensation of the hands and feet, and anxiety. Tiredness was experienced throughout the year as recounted by several participants:
“Basically, it's after the effects of the treatment that I was feeling tired cos I'm having radiotherapy every day and chemotherapy once a week.” (GYP 01 at T1)“Just me body felt tired, me body felt tired of it [radiotherapy]. I felt rotten. I couldn't do anything. It's depressing.” (GYP 12 at T2)
“Physically, I'm alright. I felt alright except I get tired easily, but other than that, I feel alright.” (GYP06 at T3)
“What I do find is that it's depressing to feel physically tired all the time and I don't know why.” (GYP04 at T4)
Difficulties with sleep were also associated with depression. For some (4/10) participants, feelings of depression occurred as they went through the treatment, as two participants described:
“I've actually felt quite depressed this last week and actually burst into tears, which is something I haven't done before. But it was sort of overwhelming—these symptoms.” (GYP08 at T3)“These symptoms are affecting my sleep. If I can't sleep, I get tired and I can't do anything the next day, then I get depressed.” (GYP06 at T3)
Depression was often the result of uncertainty and fear. Half of the participants expressed feelings of uncertainty lasting until the twelfth month, as highlighted by two of them:
“It's very uncertain you know. It's a very uncertain way of life. You don't know … like I go and see … I go back every three months for check-up. You're living your life for check-ups. So every four months you're thinking … ‘Is everything alright?’ You live your life for those four months' check-ups. The frightening bit is the uncertainty of it and it's depressing just thinking of what will happen next.” (GYP04 at T3)“I can't sleep thinking about what will happen all the time. The uncertainty keeps you awake.” (GYP012 at T3)
However, this feeling seemed to subside by T4 as they became more in control and able to cope by not dwelling on it and just accepting the disease, its treatment, and symptoms. This marked shift in coping styles in T4 characterized the patients' increased positive response to symptoms at this stage.
When participants were tired, they also complained of weakness. The expressions used included “muscle tone” and “muscle strength” weakness. However, a slight improvement in this symptom was observed from T3 onward. Tiredness and its related symptoms affected the participants' ability to get on with usual routine housework; however, support from husbands, family, and friends helped the participants to cope with their symptoms:
“I've got such a strong group of friends and relatives and they all live around me … so all the family are around me and they'll all come at least once a day.” (GYP11 at T4)
Very few management options were discussed. For both tiredness and weakness, participants used a variety of self-management approaches such as taking a rest even for half an hour each day or doing some physical fitness like walking and pilates from the third month onward. To get their muscle tone back to normal, some participants expressed their desire to “get fit,” so they walked or went cycling. Two participants reported taking herbal medicines, such as echinacea, after surgery and before chemotherapy. They perceived this as helpful in building up their immune system. Pain was managed by taking painkillers, as prescribed by their doctors. Praying was also reported as a strategy used to combat the anxieties related to the illness.
Hair loss, ocular changes, body image, identity experience, and anxiety
Hair loss, including body hair such as eyebrows and eyelashes, was reported by four participants, all of whom had received chemotherapy. In the beginning, participants did not want to wear wigs and were anxious that it portrayed a symbol to others that they had cancer:
“I do not want my family to see me without hair as they will know I have cancer and then they will start to worry about me.” (GYP10 at T1)
Later on, they no longer cared whether they wore a wig or not, especially when they themselves and others accepted the situation. Such an experience was described as follows:
“I noticed my hair falling and got me a wig the first day.” (GYP11 at T1)“I noticed that … the hairs in my nose, I think have all disappeared as well.” (GYP10 at T2)
“My hair came back like Shirley Temple, curly but it all came back slate gray and I didn't like it.” (GYP11 at T4)
On a couple of occasions, negative feelings were externalized through talking about someone else, often a famous person. This may have facilitated the expression of difficult emotions. Such a transference is depicted below:
“My hair is more or less gone but this time my husband just shaved it all off. I bung my hat on and that's it. I feel sorry for young girls. It must be horrendous cos I think of Kylie [Minogue, pop singer], for someone like her to lose her hair must have been terrible.” (GYP03 at T3)
Although participants understood that hair loss was an expected and common consequence of chemotherapy, its impact in some patients was more difficult to accept. Losing hair was seen as a realization that they had cancer or increased one's identity as a cancer patient. The same patient also talked about hair loss and anxiety:
“I think one of the most difficult things is that you might be feeling alright physically and then you've got a bald head. When I put my wig on, I feel alright. But no hair is a big thing—even though people say you looked alright without it, you don't.” (GYP03 at T3)
In some participants, hair loss, especially eyelashes, was connected with blurred vision as they believed that eyelashes protected their eyes, as reported by one (1/10) participant at T2 and three (3/9) participants at T3, around the end of their treatment.
“My eyes seem as though they're a bit blurred. I feel like I need eyeglasses. It's a thing that annoyed me most. It just felt like there's a film on your eyes. I wonder if it's something to do with hair loss.” (GYP10 at T2)
One participant was worried about her blurred vision being associated with other health problems:
“I've had slightly sort of, not blurred vision but zig-zaggy vision that made me a bit … ‘Oh dear,' you know … have I got a brain tumor? … a strange sort of thing that upsets my balance.” (GYP08 at T3)
Gastrointestinal problems: nausea, loss of appetite, taste changes, bowel function, weight changes, and distress
Nausea, appetite changes, and changes in bowel function (diarrhea or constipation) were the most common symptoms reported in relation to gastrointestinal system problems that distressed patients. Patients who reported these occurrences received either chemotherapy or radiotherapy, and one received both. However, these symptoms were of limited extent as women reported them as mild or of less importance and perceived them as more manageable. Nausea was only reported by one participant at T1 and was relieved by prescribed medications. Another participant reported loss of taste throughout the year of the study. Loss of appetite was reported by one participant at T2 and T3. Weight loss, which three of the participants attributed to diarrhea, was also reported:
“My tummy's a bit off, had a bit of diarrhea, but that's the norm.” (GYP05 at T1)“I lost weight but then again, I don't know if that's down to eating then going to the toilet.” (GYP04 at T3)
However, weight gain was reported by six participants at T3 and five participants at T4, which was the key distressing nutritional problem described, although for some it was seen in a more positive way:
“I put on weight since the radium. But it's a small price to pay isn't it? A bit of weight for all that you've gone through.” (GYP15 at T4)
Numbness and tingling sensations in the hands and feet, restlessness, sleeplessness, and depression
A common physical problem experienced by three of the participants who all received chemotherapy was tingling sensations of the hands and feet, which increased over time. At T4, three out of 10 participants still experienced numbness and tingling sensations as described by one participant:
“I was worst after my last treatment, all sorts, my feet, my fingers were really bad … always tingly. My feet they're numb and I get cramps. It's weird, they get too cold. It's depressing especially if I can't sleep.” (GYP10 at T3)“I still got funny toes and fingers. They feel fat and podgy. It's depressing. It's difficult to explain, it feels like because they're not dead but … I know I've got them, if that makes any sense. I find it difficult to spread them.” (GYP10 at T4)
Participants sometimes related the sensation to achy joint pains, as if they were getting the flu. This sensation also made them feel restless at night, contributing to sleeplessness. One participant related this to the side effect of paclitaxel (Taxol); therefore, her medication was changed to liposomal doxorubicin. Two participants tried to self-manage the feeling of coldness and numbness of their feet and fingers by soaking them in hot water. For those participants who experienced sleeplessness, due to feelings of numbness and tingling sensations in the hands and feet, wearing bed socks or soaking them in warm water, as well as using reiki and massage, were the management strategies described.
Discussion
This study explored the explanations of patients about the development and coexistence of symptoms and how patients attempted to self-manage them. This is one of the few studies in the literature, and the only one in gynecological cancer, which has explored clusters of symptoms in a narrative manner. Its longitudinal nature, unusual in qualitative research due to the inherent issues in the analysis of such data, was another strength of the study as it allowed us to explore shifts in the symptom experience, perception, and meaning over time (although meaning was fairly stable and participants talked little about it). The vast majority of the increasing literature on symptom clusters is quantitative, attempting either to model clusters through statistical techniques or to test priori clusters for their strength of relationship. However, such clusters may be biased, not only from the technique used but also from the content of self-reports utilized to collect the data. The narrative symptom clusters could rectify problems with statistical measures as they reflect the unique patient experience in the patients' own words and can assist in the development of (patient-centered rather than statistically based) symptom clusters that can then be tested quantitatively with larger samples. Hence, narrative symptom clusters can be particularly sensitive outcomes and can generate conceptually meaningful hypotheses for symptom cluster research.
Key symptoms experienced by the participants were tiredness, pain, body image changes, gastrointestinal changes, and peripheral neuropathy associated with chemotherapy, which concur with past studies of primarily ovarian cancer patients.[9] and [10] Out of the four clusters identified, one is applicable to all patients irrespective of treatment and two are clearly linked with chemotherapy. Symptoms varied in intensity but tended to subside in a year's time for the majority of patients. Acceptance brought about self-management strategies to overcome both the physical and psychological effects of cancer and its treatment, but most important was the support they received from families and friends. In addition, the fact that some symptoms decreased over time may be due to some symptoms being linked to the time since the end of treatment; such symptoms could have naturally resolved after completion of treatment. However, we have limited information on the natural history of symptoms in patients with different types of gynecological cancer.
As with other studies, the most common symptom experienced by these participants was tiredness,[3], [9] and [18] often associated with sleep disturbance due to pain, peripheral neuropathy, change in bowel function, and depression. Because these women complained of tiredness throughout the year, having social support from their husbands, families, friends, and neighbors helped them carry on with their usual household roles. This highlighted the important role caregivers play in supporting patients with cancer. Participants clearly differentiated between the symptom of tiredness/fatigue (a complex symptom involving physical, mental, and motivational aspects) from weakness (which is related more to muscle strength). This differentiation is evident in the literature,19 and while they may be related symptoms, they should be assessed separately as they may necessitate different management strategies. Dodd et al20have shown the clustering of the symptoms of fatigue, sleep disturbance, pain, and depression in breast cancer patients, similarly to the work of Liu et al.21 This quantitative work and our narrative cluster strongly support the existence and clinical relevance of this symptom cluster.
Loss of weight in the beginning was due to gastrointestinal disturbance including nausea, loss of taste, and change in bowel function. These findings concur with the literature, where such symptoms are prevalent up to one year posttreatment.22 However, as time passed, the participants regained their weight, often above their prediagnosis level. Such a gastrointestinal symptom cluster has also been supported in the quantitative literature on symptom clusters, although the relevant items within the cluster very much depend on the items included in the data-collection scale.23 Our own work with 143 patients over one year (n = 504 symptom assessments) has also identified a gastrointestinal cluster, with the key symptom being weight loss, together with loss of appetite and difficulties swallowing, experienced by up to one-quarter of a heterogeneous sample of cancer patients at the one-year time point.24 The attempts highlighted by the majority of participants to control their weight suggest that this is an important issue for these women. The inability to control weight may be frustrating and a key stressor in women with gynecological cancer. This assertion needs further investigation as the information we have about this topic to date derives almost exclusively from breast cancer patients. Weight control may be an important component of survivorship in these women, and it should be incorporated in the follow-up care of patients beyond breast cancer.25 While the use of medication was mentioned with regard to the presence of gastrointestinal symptoms, no interventions have taken place with taste changes and other nutritional concerns. Interventions around the experience and enjoyment of eating and food should be an important research focus in the future, as should work around weight gain, for which we currently have limited information.
Concern about hair loss was mentioned by only four participants. They were concerned mainly that it identified them to others as a cancer patient as baldness became the main element of the cancer patients' everyday life and identity.7 Participants were not concerned about their self-image but rather more concerned about protecting others (particularly family) and being treated differently. For these participants, they accepted that loss of hair was a side effect of treatment and viewed regrowth of hair as a positive effect, which concurred with the study by Sun et al.10 Ocular changes reported by three participants (out of 10) during treatment and up to six months later are an underreported issue in the literature. This is despite the established association between some types of chemotherapy (ie, cyclophosphamide, cisplatin) and ocular changes. More focus should be directed to this area, as well as to identifying reversible and irreversible ocular changes in the survivorship period. The narrative clustering of symptoms such as hair loss and ocular changes (connected with being “visible” cancer patients) with body image, identity, and anxiety is an interesting clustering of physical and psychological interrelated symptoms, with body image being the key symptom in this cluster. Our past work has highlighted the relationship between body image changes and “disliking” self in up to 20% of the sample, although these did not cluster together after the six-month assessment point (end of treatments) and were most visible during the chemotherapy period.24With the exception of using wigs, patients did not mention using any interventions regarding the multiple symptoms experienced within this symptom cluster, suggesting that this is an important area of research in the future.
Many of the physical symptoms reported were interrelated with descriptions of depression, uncertainty, body image, and identity as a cancer patient. While causal relationships cannot be ascertained from such a qualitative design, it is evident that there is a close relationship between the presence of physical symptoms, psychological status, and the impact of them in life. This is confirmed in a systematic review of studies with ovarian cancer patients26 as well as other studies that support the association of symptoms of fatigue, pain, anxiety, and depression with quality of life.27 A better understanding of these relationships is paramount in symptom-management efforts, particularly as it is recognized that interventions need to be multimodal and to target more than one concurrent symptom, a clear message that comes from the symptom cluster research.28 For the majority of the participants, when symptoms were not managed well, they experienced psychological responses such as depression, commonly seen in the literature. Participants in our study accepted that they would experience these symptoms, although their occurrence and intensity differed from patient to patient. Some participants were able to tolerate treatment with little physical discomfort, while in others symptoms prevented them from resuming usual functional activities and social roles, thus leading to feelings of depression. It would be interesting to identify in future research the factors that allow some patients to live well with cancer, moving away from the current research model of ill-health to a model of “wellness.”
Furthermore, a key distressing symptom that was associated with impairments in a variety of life areas was peripheral neuropathy in patients receiving chemotherapy. This cluster has some similarities with the tiredness cluster (i.e. the, presence of sleep difficulties or depression), but women talked about it as a separate experience from tiredness. Peripheral neuropathy is a difficult symptom to manage in practice and may necessitate dose reductions or chemotherapy discontinuation; therefore, the development of management strategies for this symptom is imperative. Women complained of sleeping difficulties when they experienced numbness and tingling sensations in their hands and feet, and it was a frustrating symptom that was also associated with depression. This is another symptom cluster that merits further research, and it is only emerging in the literature. Our past work on symptom clusters has also identified the presence of a “hand–foot” symptom cluster, which consistently increased over time and suggested a chronic nature, although not all symptoms identified in the present study were part of the latter quantitative evaluation.24
It was surprising to see the minimal range of interventions used to manage symptoms, both self-management and formal ones directed by the clinicians. The latter had minimal presence in the women's descriptions, with the exception of pain management, antiemetics, and antidiarrheal medication. Specific physical responses to treatment were dealt with by changing the type of chemotherapy and resorting to the use of herbal medicine for symptom management, such as echinacea and red clover, or other simple self-management techniques, such as soaking the feet in warm water, eating healthy food, and carrying out regular exercise. The use of complementary therapies was also found in the study by Ferrell et al,3 complementing the medical care provided to help control the symptoms.6 However, all of these attempts seemed to be ad hoc, with no clear understanding of processes and possible outcomes or any guidance about their use from health-care professionals. Possible reasons for this ad hoc and unsatisfactory underutilization of symptom-management interventions may include the “acceptance” by patients that some symptoms are part of their treatment, the British cultural norm of not complaining, limited confidence from both clinicians and patients on nonpharmacological interventions with variable quality of evidence of effectiveness, distance from patients' homes to the specialist service to provide supportive care, limited understanding from the clinicians of the impact of symptoms on patients' lives, and clinician time constraints.
Although we purposely included women with a range of gynecological cancer diagnoses in order to gain an understanding of broadly applicable issues related to the physical and psychological symptom experiences of patients, the small sample size limits the generalizability of the results. These symptom clusters will need further evaluation with statistical modeling. Also, the existence of symptom clusters in radiotherapy may not have surfaced well in our study with the inclusion of only three patients receiving radiotherapy, and this needs to be explored in future research. The length of treatment for each patient was variable, and knowledge of this would have enhanced the interpretability of the findings; however, this information was not available to us. Finally, although we wanted to focus on treatment-related symptoms, some of the symptoms (ie, anxiety and depression) may have multiple possible etiologies; and this needs to be considered in the interpretation of the findings.
Conclusion
Our study provides information on symptom experiences from the patients' perspective, which could lead to a better understanding of how patients perceive, assess, monitor, and manage their symptoms. This is particularly useful as the majority of the symptom literature focuses on the experience of patients with ovarian cancer. This is also important background information in developing strategies or interventions that are patient-centered and sensitive to the needs of patients that has relevance to the current policy framework. It also highlights the need for a more thorough patient assessment, to assess which symptoms are most bothersome and how symptoms are interrelated, and has implications for the physiological, psychological, sociocultural, and behavioral components of the symptom experience essential for effective symptom management. While we have identified the presence and experience of some well-documented symptoms, we have also highlighted areas of importance for patients and some underreported symptoms that merit further research in the future. Narrative symptom clusters, such as those identified in the present study, can provide a stronger conceptual basis in the symptom cluster modeling work and can assist in identifying patient-relevant and clinically meaningful groups of symptoms that can be the focus of future cluster research.
References1
1 R.L. Johnson, M.A. Gold and K.F. Wyche, Distress in women with gynecologic cancer, Psychooncology 19 (2010), pp. 665–668. View Record in Scopus | Cited By in Scopus (1)
2 R.W. Petersen, G. Graham and J.A. Quinlivan, Psychological changes after a gynaecologic cancer, J Obstet Gynecol Res 31 (2006), pp. 152–157.
3 B. Ferrell, S. Smith, C. Cullinane and C. Melancon, Symptom concerns of women with ovarian cancer, J Pain Symptom Manage 25 (2003), pp. 528–538. Article | | View Record in Scopus | Cited By in Scopus (16)
4 J. Hipkins, M. Whitworth, N. Tarrier and G. Jayson, Social support, anxiety and depression after chemotherapy for ovarian cancer: a prospective study, Br J Health Psychol 9 (pt 4) (2004), pp. 569–581. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (36)
5 P.D. Williams, U. Piamjariyakul, K.A. Ducey, J. Badura, D. Boltz, K. Olberding, A. Wingate and A.R. Williams, Cancer treatment, symptom monitoring, and self-care in adults, Cancer Nurs 29 (2006), pp. 347–355. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
6 H.S. Donovan, E.M. Hartenbach and M.W. Method, Patient–provider communication and perceived control for women experiencing multiple symptoms associated with ovarian cancer, Gynecol Oncol 99 (2005), pp. 404–411. Article | | View Record in Scopus | Cited By in Scopus (17)
7 S. Rosman, Cancer and stigma: experience of patients with chemotherapy-induced alopecia, Patient Educ Couns 52 (2004), pp. 333–339. Article | | View Record in Scopus | Cited By in Scopus (33)
8 M. Hackbarth, N. Haas, C. Fotopoulou, W. Lichtenegger and J. Schouli, Chemotherapy-induced dermatological toxicity: frequencies and impact on quality of life in women's cancer: results of a prospective study, Support Care Cancer 16 (2008), pp. 267–273. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
9 A.E. Kayl and C.A. Meyers, Side-effects of chemotherapy and quality of life in ovarian and breast cancer patients, Curr Opin Obstet Gynecol 18 (2006), pp. 24–28. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
10 C.C. Sun, D.C. Bodurka, C.B. Weaver, R. Rasu, J.K. Wolf, M.W. Beyers, J.A. Smith and J.T. Wharton, Rankings and symptom assessments of side effects from chemotherapy: insights from experiences patients with ovarian cancer, Support Care Cancer 13 (2005), pp. 219–227. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (53)
11 R.K. Portenoy, H.T. Thaler, A.B. Kornblith, J. McCarthy-Lepore, H. Friedlander-Klar, N. Coyle, T. Smart-Curley, N. Kemeny, L. Norton, W. Hoskins and H. Scher, Symptom prevalence, characteristics and distress in a cancer population, Qual Life Res 3 (1994), pp. 183–189. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (281)
12 V. Gonçalves, G. Jayson and N. Tarrier, A longitudinal investigation of psychological morbidity in patients with ovarian cancer, Br J Cancer 99 (2008), pp. 1794–1801. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
13 M.Q. Patton, Qualitative Evaluation and Research Methods (2nd ed.), Sage Publications, Newbury Park, CA (1990).
14 M. Sandelowski, Real qualitative researchers do not count: the use of numbers in qualitative research, Res Nurs Health 24 (2001), pp. 230–240. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
15 S.S. Hwang, V.T. Chang, D.L. Fairclough, J. Cogswell and B. Kasimis, Longitudinal quality of life in advanced cancer patients: pilot study results from a VA Medical Cancer Center, J Pain Symptom Manage 25 (2003), pp. 225–235. Article | | View Record in Scopus | Cited By in Scopus (46)
16 H. Leventhal and J.E. Johnson, Laboratory and field experimentation: development of a theory of self-regulation. In: J.P. Wooldridge, M.H. Schmitt, J.K. Skipper and R.C. Leonard, Editors, Behavioral Science and Nursing Theory, Mosby, St. Louis (1983), pp. 189–264.
17 M. Dodd, S. Janson, N. Facione, J. Faucett and E.S. Froelicher et al., Advancing the science of symptom management, J Adv Nurs 33 (2001), pp. 668–676. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (173)
18 C.C. Sun, D.C. Bodurka, M.L. Donato, E.B. Rubenstein, C.L. Borden, K. Basen-Engquist, M.S. Munsell, J.J. Kavanagh and D.M. Gershenson, Patient preferences regarding side effects of chemotherapy for ovarian cancer: do they change over time?, Gynecol Oncol 87 (2002), pp. 118–128. Abstract | | View Record in Scopus | Cited By in Scopus (22)
19 S.C. Teunissen, W. Wesker, C. Kruitwagen, H.C. de Haes, E.E. Voest and A. de Graeff, Symptom prevalence in patients with incurable cancer: a systematic review, J Pain Symptom Manage 34 (2007), pp. 94–104. Article | | View Record in Scopus | Cited By in Scopus (67)
20 M.J. Dodd, M.H. Cho, B.A. Cooper and C. Miaskowski, The effect of symptom clusters on functional status and quality of life in women with breast cancer, Eur J Oncol Nurs 14 (2010), pp. 101–110. Article | | View Record in Scopus | Cited By in Scopus (6)
21 L. Liu, L. Fiorentino, L. Natarajan, B.A. Parker, P.J. Mills, G.R. Sadler, J.E. Dimsdale, M. Rissling, F. He and S. Ancoli-Israel, Pre-treatment symptom cluster in breast cancer patients is associated with worse sleep, fatigue and depression during chemotherapy, Psychooncology 18 (2009), pp. 187–194. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (0)
22 H. Tong, E. Isenring and P. Yates, The prevalence of nutrition impact symptoms and their relationship to quality of life and clinical outcomes in medical oncology patients, Support Care Cancer 17 (2009), pp. 83–90. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
23 H.J. Kim, A.M. Barsevick, L. Tulman and P.A. McDermott, Treatment-related symptom clusters in breast cancer: a secondary analysis, J Pain Symptom Manage 36 (2008), pp. 468–479. Article | | View Record in Scopus | Cited By in Scopus (12)
24 A. Molassiotis, Y. Wengstrom and N. Kearney, Symptom cluster patterns during the first year from the diagnosis with cancer, J Pain Symptom Manage 39 (2010), pp. 847–858. Article | | View Record in Scopus | Cited By in Scopus (1)
25 L. Muraca, D. Leung, A. Clark, M.A. Beduz and P. Goodwin, Breast cancer survivors: taking charge of lifestyle choices after treatment, Eur J Oncol Nurs (2010) 10.1016/e.ejon.2009.12.001.
26 E. Arden-Close, Y. Gidron and R. Moss-Morris, Psychological distress and its correlates in ovarian cancer: a systematic review, Psychooncology 17 (2008), pp. 1061–1072. View Record in Scopus | Cited By in Scopus (7)
27 W.K.W. So, G. Marsh, W.M. Ling, F.Y. Leung, J.C.K. Lo, M. Yeung and G.K.H. Li, The symptom cluster of fatigue, pain anxiety, and depression and the effect on the quality of life of women receiving treatment for breast cancer: a multicentre study, Oncol Nurs Forum 36 (2009), pp. E205–E214. Full Text via CrossRef
28 M.J. Dodd, C. Miaskowski and S.M. Paul, Symptom clusters and their effect on functional status of patients with cancer, Oncol Nurs Forum 28 (2001), pp. 465–470. View Record in Scopus | Cited By in Scopus (192)
Correspondence to: Violeta Lopez, TCH, RCNMP, Building 6, Level 3, East Wing, Yamba Drive, Garran, 2605, Canberra, Australian Capital Territory (ACT), Australia; telephone: +612 6244 2333; fax: +612 6244 2573
Evaluating the “Good Death” Concept from Iranian Bereaved Family Members' Perspective
Original research
Sedigheh Iranmanesh PhDa, Habibollah Hosseini doctoral student
Abstract
Improving end-of-life care demands that first you define what constitutes a good death for different cultures. This study was conducted to evaluate a good death concept from the Iranian bereaved family members' perspective. A descriptive, cross-sectional study was designed using a Good Death Inventory (GDI) questionnaire to evaluate 150 bereaved family members. Data were analyzed by SPSS. Based on the results, the highest scores belonged to the domains “being respected as an individual,” “natural death,” “religious and spiritual comfort,” and “control over the future.” The domain perceived by family members as less important was “unawareness of death.” Providing a good death requires professional caregivers to be sensitive and pay attention to the preferences of each unique person's perceptions. In order to implement holistic care, caregivers must be aware of patients' spiritual needs. Establishing a specific unit in a hospital and individually treating each patient as a valued family member could be the best way to improve the quality of end-of-life care that is missing in Iran.
Article Outline
From a review of different studies, the core quality of a good death varies among cultures. In a qualitative study, Griggs4 analyzed perceptions of a “good death” among community nurses in England. Nurses identified several key themes for a good death, such as: symptom control, patient choice, honesty, spirituality, interprofessional relationships, effective preparation, organization, and provision of seamless care. American researchers concluded that a good death involves respect for the individual's autonomy with open communication among family members.5 Vig and Pearlman6 also reported that “good death” has an individual meaning for Americans and does not have a consensual meaning. In Ghana, Van der Greest7 found that a good death is integrated with a peaceful death, meaning peace with others, being at peace with one's own life and soul, dying in the fullness of time, dying at home, and being surrounded by relatives. For the Japanese, Hattori et al.8 found that a good death is a multidimensional, individual experience based on personal and sociocultural domains of life that incorporate the person's past, present, and future. In Norway, Ruland and Moore9 conducted research on the theory of a peaceful end of life which has five major concepts: not being in pain, experience of comfort, experience of dignity/respect, being at peace, and closeness to significant others/persons who care. In Thailand, people commonly used “peaceful death” instead of “good death.” Kongsuwan and Locsin10 reported that Thai intensive care unit nurses perceived peaceful death as awareness of dying, creating a caring environment, and promoting end-of-life care. In Muslim society, Tayeb et al11 identified three domains related to a good death: religion and faith, self-esteem and personal image, and satisfaction about family security.
After reviewing these studies, we determined there is no universal definition of good death and it is based on sociocultural context. The subject of death and dying has a religious and sociocultural background, yet Iranian health-care providers mainly depend on Western references. Moreover, upon reviewing the literature in Iran, no published study related to defining the concept of “good death” was located. This descriptive study was thus designed to determine what constitutes a good death in the Iranian context.
Context
Iran is one of the most ancient world civilizations and part of the Middle East culture. The population is approximately 67 million, and of this 51% is less than 20 years old and 6.5% is 65 or older.12 The majority (99.4%) of the people in Iran consider themselves as religious,13 and religious beliefs strongly and explicitly deal with death.14
Iranians are familiar with death. Besides the Iran–Iraq war and natural disasters in recent years, the major causes (65%) of death among Iranians are heart disease, cancer, and accidents.15 Apart from chronic disease, accidents seem to be a significant cause of death among Iranian people. In Iran, the overall national curriculum for registered nursing education includes just a few hours of academic education about death. End-of-life care remains a new topic in the Iranian health-care system. Hospice care units, which are common in Western countries, are not available in Iran.
Most religions are represented in this country; however, Islam is the most prevalent. Sareming16 indicates that Muslims are taught that Allah gives birth and death. Allah determines the appointed term for every human. Only Allah knows when, where, and how a person will die. For a Muslim, death is the transition from the earthly form of existence to the next.17 Tayeb et al11 explained that Muslims prefer to approach death with a certainty that someone is there to prompt them with the Shahadah, reciting a chapter of the Quran, dying in a position facing Mecca, and dying in a holy place such as a mosque.
Method
Design
There was approval from the heads of hospitals prior to the collection of data. The study employed a descriptive design and was conducted in two hospitals that had oncology units in southeast Iran.
Participants
Referring to the hospitals' and patients' documents, 150 bereaved family members of patients who died within 1 year were identified. They were called by the researcher and asked to participate in this study.
Background Information
At first, a questionnaire was designed in order to obtain background information which was assumed to influence the good death concept. It included questions about gender, age, marital status, previous studies about death, and level of education.
Instruments
The good death concept was evaluated using the Good Death Inventory (GDI). The GDI was designed by Miyashita et al18 for evaluating a good death from the bereaved family members' perspective. This scale has 51 items. The items are graded from 1 to 7 (1 = strongly disagree to 7 = strongly agree). A factor analysis made by Miyashita et al18 on research made in a Japanese setting revealed that the questions could be divided into 18 domains: (1) physical and psychological comfort, (2) dying in a favorite place, (3) good relationship with medical staff, (4) maintaining hope and pleasure, (5) not being a burden to others, (6) good relationship with family, (7) physical and cognitive control, (8) environmental comfort, (9) being respected as an individual, (10) life completion, (11) natural death, (12) preparation for death, (13) role accomplishment and contributing to others, (14) unawareness of death, (15) fighting against cancer, (16) pride and beauty, (17) control over the future, and (18) religious and spiritual comfort.
For translation from English into Farsi, the standard forward–backward procedure was applied. Translation of the items and the response categories was independently performed by two professional translators, and then temporary versions were provided. Afterward they were back-translated into English, and after a careful cultural adaptation the final versions were provided. Translated questionnaires went through pilot testing. Suggestions by family members were combined into the final versions.
Reliability and validity
The translated scale was originally developed and tested in a Japanese cultural context, which is different from the research contexts, so the validity and reliability of both scales were rechecked. A factor analysis (rotated component matrix) on the results was done in order to examine the context validity of the GDI. The concession of the items was similar to the Japanese results, and 18 components were identified. The validity of the scale was assessed through a content validity discussion. Scholars of statistics and nursing care have reviewed the content of the scale from religious and cultural aspects of death and agreed upon a reasonable content validity. To reassess the reliability of the translated scale, alpha coefficients of internal consistency and 3-week test–retest coefficients (n = 30) of stability were computed. The alpha coefficient for GDI was 0.68. The 3-week test–retest coefficient of stability for the GDI was 0.79. Therefore, the translated scale presented an acceptable reliability.
Data Collection and Analysis
Accompanied by a letter including some information about the aim of the study, the questionnaires were handed out by the second author to 150 family members who were introduced by the matron of two hospitals over 2 months (May/June 2010) in southeast Iran. Some oral information about the study was also given by the third author. Participation in the study was voluntary and anonymous. We distributed 150 sets of questionnaires. In all collected data, 98% of all questions were answered. Data from the questionnaires were analyzed using the Statistical Package for Social Scientists (SPSS, Inc., Chicago, IL). A Kolmogorov-Smirnov test indicated that the data were sampled from a population with normal distribution. Descriptive statistics of the sample and measures that were computed included frequencies, means, and reliability. Cross-table analysis (Spearman's test) was used to examine relationships among demographic factors and scores on the GDI.
Results
Participants
A descriptive analysis of the background information revealed that the participants belonged to the age group of 16–68 years, with a mean age of 33 years, and were mainly female (81%). About 68% were married, and the majority had an academic degree. Regarding personal study about death, 36.9% had read some things about death previously.
Findings
Descriptive analysis indicated that the highest scores belonged to the domains “being respected as an individual” (mean = 6.55), “natural death” (mean = 6.36), “religious and spiritual comfort” (mean = 6.02), and “control over the future” (mean = 6.55) (Table 1).
The domains and the components perceived as important by bereaved family members were (1) physical and psychological comfort, (2) dying in a favorite place, (3) maintaining hope and pleasure, (5) not being a burden to others, (6) good relationship with family, (7) physical and cognitive control, (8) environmental comfort, and (9) life completion. The domain perceived by family members as less important was “unawareness of death” (mean = 3.05).
Significant differences were found between some domains of a good death and demographic characters of family members. Older participants were more likely to perceive a good death as “being respected as an individual” and “having good relationships with family members.” Among participants, those who had a higher level of education were more likely to view a good death as “being respected as an individual” and “pride and beauty.” There was a negative correlation between level of education and “unawareness of death” (Table 2).
Discussion
According to the factor analysis, 18 domains contributing to a good death were identified. However, the domains of the “good death” concept that were perceived as important by bereaved family members were similar to those in Japan. This finding thus indicates that these perceptions are foundational elements of a good death, regardless of ethnicity or cultural differences.
The results indicated that most family members are likely to view a good death as “being respected as an individual” and having “control over the future.” According to Murata,19 approaching death can cause a sense that life is meaningless and a loss of the patient's well-being founded on temporality, relationships, and autonomy. Providing a good death means that dying patients are able and allowed to participate in the same human interactions that are important throughout life and appreciating patients as unique and “whole persons,” not only as “diseases” or cases.20 It means supporting patients' well-being through positive stimulation, for example, offering beautiful views and tasty meals.21 A good death is also perceived by family members as “religious and spiritual comfort.” Ghavamzadeh and Bahar14 claimed that among Iranians religious beliefs strongly and explicitly deal with the fact of death. This finding reflects the result of Tayeb et al,11 who found that Muslims believe that death is closely linked to faith. They appreciated the importance of access to any needed spiritual or emotional support. Steinhauser et al.20 also found that 89% of American patients and 85% of their families emphasize that a good death is “being at peace with God” and “prayer.”
Participants perceived a good death as a “natural death.” Johnson et al22 claimed that death without “machines,” “tubes,” and “lines” is considered more dignified and aesthetically pleasing. Withdrawal or withholding of treatment of the highly invasive and technological sort is conceptualized as restoring patient dignity and, to a small degree, personhood.22 Many deaths were not considered “good” because of inherent problems within a culture of care that usually strives to prolong life and prevent death.23 Similarly, Miyashita et al18 reported that most Japanese view unnecessary life-prolonging treatments such as vasopressors, antibiotics, and artificial hydration as barriers to achieving a good death. The domain perceived by family members as less important was “unawareness of death.” This is consistent with Steinhauser et al's20 finding that 96% of American patients emphasized “knowing what to expect about one's physical condition” achieves a good death. This is inconsistent with Tang et al's24 claims that in many traditional cultures (eg, most Asian countries and a few European cultures), in an effort to protect the patient from despair and a feeling of hopelessness, family caregivers often exclude patients from the process of information exchange. This is also in contrast to Miyashita et al's[18] and [25] findings, where many Japanese do not want to know the seriousness of their condition. Our findings could be explained by the other results of this study. The results indicated that the majority of participants had a high level of education. The other findings showed there is a negative correlation between level of education and “unawareness of death.” Since the majority of participants were well-educated, it can be concluded that they were less likely to view a good death as “unawareness of death.” This has also been found by Montazeri et al.26
The results showed that the family members' age was correlated with some aspects of a good death. Miyashita et al18 also found that the older the family member, the more positively he or she would look on the patient's death. They claimed that death at younger ages tended to be evaluated as a bad death. This could be explained by their earlier study, where they found that age and psychosocial maturity inversely related to death anxiety.27 Based on the results, level of education positively influenced some domains of a good death. There was a negative correlation between level of education and “unawareness of death,” with Montazeri et al26 finding that Iranian patients with a low level of education were more likely to not know the diagnosis.
Conclusion
According to the results of this study, providing a good death requires professional caregivers to be sensitive and pay attention to the preferences of each unique person's perceptions through her or his senses. This includes views, tastes, sounds, smells, and bodily contact. The ability of a dying person to see a sunset may seem petty but is important in providing high-quality care for people at the end of their lives. The same goes for the other senses. These circumstances deserve attention in all educational programs and especially in programs dealing with end-of-life care. In order to implement holistic care, caregivers must pay attention to patients' spiritual needs. Establishing a specific palliative care unit in a hospital and meeting each patient as a unique being and part of a family could be the best way to improve the quality of end-of-life care that is missing in Iran. It requires cultural preparation and public education through the media and by well-educated staff. Since demographic variables influenced the evaluation of a good death from the bereaved family members' perspective, public education needs different strategies.
Limitation
All data in this study were collected by use of self-report questionnaires. The dependence on self-report aspects in this study may have caused an overestimation of some of the findings due to variance, which is common in different methods. The respondents were predominantly female, which limits the generalization of the results for male respondents. Moreover, the convenience sample of Iranian bereaved family members, which is not representative of the entire Iranian population, could weaken the generalization of the findings. Further research is necessary to illuminate the concept of a good death as perceived by the general Iranian population.
References
1 I.M. Proot, H.H. Abu-Saad, H.F.J.M. Crebolder, K.A. Luker and G.A.M. Widdershoven, Vulnerability of family caregivers in terminal palliative care at home; balancing between burden and capacity, Scand J Caring Sci 17 (2003), pp. 113–121. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (53)
2 D.L. Patrick, R.A. Engelberg and J.R. Curtis, Evaluating the quality of dying and death, J Pain Symptom Manage 22 (2001), pp. 717–726. Article | | View Record in Scopus | Cited By in Scopus (96)
3 , Longman Dictionary of Contemporary English, Pearson Education, Canada (2003).
4 C.H. Griggs, Community nurses' perceptions of a good death: a qualitative exploratory study, Int J Palliat Nurs 16 (2010), pp. 140–149. View Record in Scopus | Cited By in Scopus (0)
5 J.E. Winland-Brown, Public's perceptions of a good death and assisted suicide, Issues Interdisc Care 3 (2001), pp. 137–144. View Record in Scopus | Cited By in Scopus (1)
6 E.K. Vig and R.A. Pearlman, Good and bad dying from the perspective of terminally ill men, Arch Intern Med 164 (2004), pp. 977–981. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (27)
7 S. Van der Greest, Dying peacefully: considering good death and bad death in Kwahu-Tafo, Ghana, Soc Sci Med 58 (2004), pp. 899–911.
8 K. Hattori, M.A. McCubbin and D.N. Ishida, Concept analysis of good death in the Japanese community, J Nurs Scholarsh 38 (2006), pp. 165–170. View Record in Scopus | Cited By in Scopus (9)
9 C.M. Ruland and S.M. Moore, Theory construction based on standards of care: a proposed theory of the peaceful end of life, Nurs Outlook 46 (1998), pp. 169–175. Article | | View Record in Scopus | Cited By in Scopus (13)
10 W. Kongsuwan and R.C. Locsin, Promoting peaceful death in the intensive care unit in Thailand, Int Nurs Rev 56 (2009), pp. 116–122. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
11 M.A. Tayeb, E. Al-zamel, M.M. Fareed and H.A. Abouellail, A ”good death”: perspectives of Muslim patients and health care providers, Ann Saudi Med 30 (2010), pp. 215–221. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1)
12 WHO www.who.Int/countries/en/#s (2008).
13 European Values Study Group and World Values Surveys Association, European and World Values Surveys, four-wave integrated data file, 1981–2004 http://www.worldvaluessurvey.org/services/index.html.
14 A. Ghavamzadeh and B. Bahar, Communication with the cancer patients in Iran, information and truth, Ann N Y Acad Sci 809 (1997), pp. 261–265. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
15 Cancer Institute of Iran www.cancer-institute.ac.ir Access date is June 2010.
16 N. Sareming, Islamic teaching on death and practices toward the dying person, J Soc Sci Hum 3 (1997), pp. 75–91.
17 A. Sheikh, Death and dying: A Muslim perspective, J R Soc Med 91 (1998), pp. 138–140. View Record in Scopus | Cited By in Scopus (16)
18 M. Miyashita, T. Morita, K. Sato, K. Hirai, Y. Shima and Y. Uchitomi, Good Death Inventory: a measure for evaluating good death from the bereaved family members' perspective, J Pain Symptom Manage 35 (2008), pp. 486–498. Article | | View Record in Scopus | Cited By in Scopus (10)
19 H. Murata, Spiritual pain and its care in patients with terminal cancer: construction of a conceptual framework by philosophical approach, Palliat Support Care 1 (2003), pp. 15–21. View Record in Scopus | Cited By in Scopus (12)
20 K.E. Steinhauser, E.C. Clipp, M. McNeilly, N.A. Christakis, L.M. McIntyre and J.A. Tulsky, In search of a good death: observations of patients, families, and providers, Ann Intern Med 132 (2000), pp. 825–832. View Record in Scopus | Cited By in Scopus (372)
21 S. Iranmanesh, T. Haggstrom, K. Axelsson and S. Savenstedt, Swedish nurses' experiences of caring for dying people: a holistic approach, Holist Nurs Pract 23 (2009), pp. 243–252. View Record in Scopus | Cited By in Scopus (1)
22 N. Johnson, D. Cook, M. Giacomini and D. Willms, Towards a good death: end of life narratives constructed in an intensive care unit, Cult Med Psychiatry 24 (2000), pp. 275–295. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
23 R.L. Beckstrand, L.C. Callister and K.T. Kirchhoff, Providing a ”good death”: critical care nurses' suggestions for improving end-of-life care, Am J Crit Care 15 (2006), pp. 38–45. View Record in Scopus | Cited By in Scopus (49)
24 S.T. Tang, T.W. Liu, M.S. Lai, L.N. Liu, C.H. Chen and S.L. Koong, Congruence of knowledge, experiences, and preferences for disclosure of diagnosis and prognosis between terminally-ill cancer patients and their family caregivers in Taiwan, Cancer Invest 24 (2006), pp. 360–366. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (21)
25 M. Miyashita, M. Sanjo, K. Morita and Y. Uchitomi, Good death in cancer care: a nationwide quantitative study, Ann Oncol 18 (2007), pp. 1090–1097. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (31)
26 A. Montazeri, A. Tavoli, M.A. Mohagheghi, R. Roshan and Z. Tavoli, Disclosure of cancer diagnosis and quality of life in cancer patients: should it be the same everywhere, BMC Cancer 9 (2009), pp. 1–21.
27 R.J. Russac, C. Gatliff, M. Reece and D. Spottswood, Death anxiety across the adult years: an examination of age and gender effects, J Death Stud 31 (2007), pp. 549–561. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)
Original research
Sedigheh Iranmanesh PhDa, Habibollah Hosseini doctoral student
Abstract
Improving end-of-life care demands that first you define what constitutes a good death for different cultures. This study was conducted to evaluate a good death concept from the Iranian bereaved family members' perspective. A descriptive, cross-sectional study was designed using a Good Death Inventory (GDI) questionnaire to evaluate 150 bereaved family members. Data were analyzed by SPSS. Based on the results, the highest scores belonged to the domains “being respected as an individual,” “natural death,” “religious and spiritual comfort,” and “control over the future.” The domain perceived by family members as less important was “unawareness of death.” Providing a good death requires professional caregivers to be sensitive and pay attention to the preferences of each unique person's perceptions. In order to implement holistic care, caregivers must be aware of patients' spiritual needs. Establishing a specific unit in a hospital and individually treating each patient as a valued family member could be the best way to improve the quality of end-of-life care that is missing in Iran.
Article Outline
From a review of different studies, the core quality of a good death varies among cultures. In a qualitative study, Griggs4 analyzed perceptions of a “good death” among community nurses in England. Nurses identified several key themes for a good death, such as: symptom control, patient choice, honesty, spirituality, interprofessional relationships, effective preparation, organization, and provision of seamless care. American researchers concluded that a good death involves respect for the individual's autonomy with open communication among family members.5 Vig and Pearlman6 also reported that “good death” has an individual meaning for Americans and does not have a consensual meaning. In Ghana, Van der Greest7 found that a good death is integrated with a peaceful death, meaning peace with others, being at peace with one's own life and soul, dying in the fullness of time, dying at home, and being surrounded by relatives. For the Japanese, Hattori et al.8 found that a good death is a multidimensional, individual experience based on personal and sociocultural domains of life that incorporate the person's past, present, and future. In Norway, Ruland and Moore9 conducted research on the theory of a peaceful end of life which has five major concepts: not being in pain, experience of comfort, experience of dignity/respect, being at peace, and closeness to significant others/persons who care. In Thailand, people commonly used “peaceful death” instead of “good death.” Kongsuwan and Locsin10 reported that Thai intensive care unit nurses perceived peaceful death as awareness of dying, creating a caring environment, and promoting end-of-life care. In Muslim society, Tayeb et al11 identified three domains related to a good death: religion and faith, self-esteem and personal image, and satisfaction about family security.
After reviewing these studies, we determined there is no universal definition of good death and it is based on sociocultural context. The subject of death and dying has a religious and sociocultural background, yet Iranian health-care providers mainly depend on Western references. Moreover, upon reviewing the literature in Iran, no published study related to defining the concept of “good death” was located. This descriptive study was thus designed to determine what constitutes a good death in the Iranian context.
Context
Iran is one of the most ancient world civilizations and part of the Middle East culture. The population is approximately 67 million, and of this 51% is less than 20 years old and 6.5% is 65 or older.12 The majority (99.4%) of the people in Iran consider themselves as religious,13 and religious beliefs strongly and explicitly deal with death.14
Iranians are familiar with death. Besides the Iran–Iraq war and natural disasters in recent years, the major causes (65%) of death among Iranians are heart disease, cancer, and accidents.15 Apart from chronic disease, accidents seem to be a significant cause of death among Iranian people. In Iran, the overall national curriculum for registered nursing education includes just a few hours of academic education about death. End-of-life care remains a new topic in the Iranian health-care system. Hospice care units, which are common in Western countries, are not available in Iran.
Most religions are represented in this country; however, Islam is the most prevalent. Sareming16 indicates that Muslims are taught that Allah gives birth and death. Allah determines the appointed term for every human. Only Allah knows when, where, and how a person will die. For a Muslim, death is the transition from the earthly form of existence to the next.17 Tayeb et al11 explained that Muslims prefer to approach death with a certainty that someone is there to prompt them with the Shahadah, reciting a chapter of the Quran, dying in a position facing Mecca, and dying in a holy place such as a mosque.
Method
Design
There was approval from the heads of hospitals prior to the collection of data. The study employed a descriptive design and was conducted in two hospitals that had oncology units in southeast Iran.
Participants
Referring to the hospitals' and patients' documents, 150 bereaved family members of patients who died within 1 year were identified. They were called by the researcher and asked to participate in this study.
Background Information
At first, a questionnaire was designed in order to obtain background information which was assumed to influence the good death concept. It included questions about gender, age, marital status, previous studies about death, and level of education.
Instruments
The good death concept was evaluated using the Good Death Inventory (GDI). The GDI was designed by Miyashita et al18 for evaluating a good death from the bereaved family members' perspective. This scale has 51 items. The items are graded from 1 to 7 (1 = strongly disagree to 7 = strongly agree). A factor analysis made by Miyashita et al18 on research made in a Japanese setting revealed that the questions could be divided into 18 domains: (1) physical and psychological comfort, (2) dying in a favorite place, (3) good relationship with medical staff, (4) maintaining hope and pleasure, (5) not being a burden to others, (6) good relationship with family, (7) physical and cognitive control, (8) environmental comfort, (9) being respected as an individual, (10) life completion, (11) natural death, (12) preparation for death, (13) role accomplishment and contributing to others, (14) unawareness of death, (15) fighting against cancer, (16) pride and beauty, (17) control over the future, and (18) religious and spiritual comfort.
For translation from English into Farsi, the standard forward–backward procedure was applied. Translation of the items and the response categories was independently performed by two professional translators, and then temporary versions were provided. Afterward they were back-translated into English, and after a careful cultural adaptation the final versions were provided. Translated questionnaires went through pilot testing. Suggestions by family members were combined into the final versions.
Reliability and validity
The translated scale was originally developed and tested in a Japanese cultural context, which is different from the research contexts, so the validity and reliability of both scales were rechecked. A factor analysis (rotated component matrix) on the results was done in order to examine the context validity of the GDI. The concession of the items was similar to the Japanese results, and 18 components were identified. The validity of the scale was assessed through a content validity discussion. Scholars of statistics and nursing care have reviewed the content of the scale from religious and cultural aspects of death and agreed upon a reasonable content validity. To reassess the reliability of the translated scale, alpha coefficients of internal consistency and 3-week test–retest coefficients (n = 30) of stability were computed. The alpha coefficient for GDI was 0.68. The 3-week test–retest coefficient of stability for the GDI was 0.79. Therefore, the translated scale presented an acceptable reliability.
Data Collection and Analysis
Accompanied by a letter including some information about the aim of the study, the questionnaires were handed out by the second author to 150 family members who were introduced by the matron of two hospitals over 2 months (May/June 2010) in southeast Iran. Some oral information about the study was also given by the third author. Participation in the study was voluntary and anonymous. We distributed 150 sets of questionnaires. In all collected data, 98% of all questions were answered. Data from the questionnaires were analyzed using the Statistical Package for Social Scientists (SPSS, Inc., Chicago, IL). A Kolmogorov-Smirnov test indicated that the data were sampled from a population with normal distribution. Descriptive statistics of the sample and measures that were computed included frequencies, means, and reliability. Cross-table analysis (Spearman's test) was used to examine relationships among demographic factors and scores on the GDI.
Results
Participants
A descriptive analysis of the background information revealed that the participants belonged to the age group of 16–68 years, with a mean age of 33 years, and were mainly female (81%). About 68% were married, and the majority had an academic degree. Regarding personal study about death, 36.9% had read some things about death previously.
Findings
Descriptive analysis indicated that the highest scores belonged to the domains “being respected as an individual” (mean = 6.55), “natural death” (mean = 6.36), “religious and spiritual comfort” (mean = 6.02), and “control over the future” (mean = 6.55) (Table 1).
The domains and the components perceived as important by bereaved family members were (1) physical and psychological comfort, (2) dying in a favorite place, (3) maintaining hope and pleasure, (5) not being a burden to others, (6) good relationship with family, (7) physical and cognitive control, (8) environmental comfort, and (9) life completion. The domain perceived by family members as less important was “unawareness of death” (mean = 3.05).
Significant differences were found between some domains of a good death and demographic characters of family members. Older participants were more likely to perceive a good death as “being respected as an individual” and “having good relationships with family members.” Among participants, those who had a higher level of education were more likely to view a good death as “being respected as an individual” and “pride and beauty.” There was a negative correlation between level of education and “unawareness of death” (Table 2).
Discussion
According to the factor analysis, 18 domains contributing to a good death were identified. However, the domains of the “good death” concept that were perceived as important by bereaved family members were similar to those in Japan. This finding thus indicates that these perceptions are foundational elements of a good death, regardless of ethnicity or cultural differences.
The results indicated that most family members are likely to view a good death as “being respected as an individual” and having “control over the future.” According to Murata,19 approaching death can cause a sense that life is meaningless and a loss of the patient's well-being founded on temporality, relationships, and autonomy. Providing a good death means that dying patients are able and allowed to participate in the same human interactions that are important throughout life and appreciating patients as unique and “whole persons,” not only as “diseases” or cases.20 It means supporting patients' well-being through positive stimulation, for example, offering beautiful views and tasty meals.21 A good death is also perceived by family members as “religious and spiritual comfort.” Ghavamzadeh and Bahar14 claimed that among Iranians religious beliefs strongly and explicitly deal with the fact of death. This finding reflects the result of Tayeb et al,11 who found that Muslims believe that death is closely linked to faith. They appreciated the importance of access to any needed spiritual or emotional support. Steinhauser et al.20 also found that 89% of American patients and 85% of their families emphasize that a good death is “being at peace with God” and “prayer.”
Participants perceived a good death as a “natural death.” Johnson et al22 claimed that death without “machines,” “tubes,” and “lines” is considered more dignified and aesthetically pleasing. Withdrawal or withholding of treatment of the highly invasive and technological sort is conceptualized as restoring patient dignity and, to a small degree, personhood.22 Many deaths were not considered “good” because of inherent problems within a culture of care that usually strives to prolong life and prevent death.23 Similarly, Miyashita et al18 reported that most Japanese view unnecessary life-prolonging treatments such as vasopressors, antibiotics, and artificial hydration as barriers to achieving a good death. The domain perceived by family members as less important was “unawareness of death.” This is consistent with Steinhauser et al's20 finding that 96% of American patients emphasized “knowing what to expect about one's physical condition” achieves a good death. This is inconsistent with Tang et al's24 claims that in many traditional cultures (eg, most Asian countries and a few European cultures), in an effort to protect the patient from despair and a feeling of hopelessness, family caregivers often exclude patients from the process of information exchange. This is also in contrast to Miyashita et al's[18] and [25] findings, where many Japanese do not want to know the seriousness of their condition. Our findings could be explained by the other results of this study. The results indicated that the majority of participants had a high level of education. The other findings showed there is a negative correlation between level of education and “unawareness of death.” Since the majority of participants were well-educated, it can be concluded that they were less likely to view a good death as “unawareness of death.” This has also been found by Montazeri et al.26
The results showed that the family members' age was correlated with some aspects of a good death. Miyashita et al18 also found that the older the family member, the more positively he or she would look on the patient's death. They claimed that death at younger ages tended to be evaluated as a bad death. This could be explained by their earlier study, where they found that age and psychosocial maturity inversely related to death anxiety.27 Based on the results, level of education positively influenced some domains of a good death. There was a negative correlation between level of education and “unawareness of death,” with Montazeri et al26 finding that Iranian patients with a low level of education were more likely to not know the diagnosis.
Conclusion
According to the results of this study, providing a good death requires professional caregivers to be sensitive and pay attention to the preferences of each unique person's perceptions through her or his senses. This includes views, tastes, sounds, smells, and bodily contact. The ability of a dying person to see a sunset may seem petty but is important in providing high-quality care for people at the end of their lives. The same goes for the other senses. These circumstances deserve attention in all educational programs and especially in programs dealing with end-of-life care. In order to implement holistic care, caregivers must pay attention to patients' spiritual needs. Establishing a specific palliative care unit in a hospital and meeting each patient as a unique being and part of a family could be the best way to improve the quality of end-of-life care that is missing in Iran. It requires cultural preparation and public education through the media and by well-educated staff. Since demographic variables influenced the evaluation of a good death from the bereaved family members' perspective, public education needs different strategies.
Limitation
All data in this study were collected by use of self-report questionnaires. The dependence on self-report aspects in this study may have caused an overestimation of some of the findings due to variance, which is common in different methods. The respondents were predominantly female, which limits the generalization of the results for male respondents. Moreover, the convenience sample of Iranian bereaved family members, which is not representative of the entire Iranian population, could weaken the generalization of the findings. Further research is necessary to illuminate the concept of a good death as perceived by the general Iranian population.
References
1 I.M. Proot, H.H. Abu-Saad, H.F.J.M. Crebolder, K.A. Luker and G.A.M. Widdershoven, Vulnerability of family caregivers in terminal palliative care at home; balancing between burden and capacity, Scand J Caring Sci 17 (2003), pp. 113–121. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (53)
2 D.L. Patrick, R.A. Engelberg and J.R. Curtis, Evaluating the quality of dying and death, J Pain Symptom Manage 22 (2001), pp. 717–726. Article | | View Record in Scopus | Cited By in Scopus (96)
3 , Longman Dictionary of Contemporary English, Pearson Education, Canada (2003).
4 C.H. Griggs, Community nurses' perceptions of a good death: a qualitative exploratory study, Int J Palliat Nurs 16 (2010), pp. 140–149. View Record in Scopus | Cited By in Scopus (0)
5 J.E. Winland-Brown, Public's perceptions of a good death and assisted suicide, Issues Interdisc Care 3 (2001), pp. 137–144. View Record in Scopus | Cited By in Scopus (1)
6 E.K. Vig and R.A. Pearlman, Good and bad dying from the perspective of terminally ill men, Arch Intern Med 164 (2004), pp. 977–981. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (27)
7 S. Van der Greest, Dying peacefully: considering good death and bad death in Kwahu-Tafo, Ghana, Soc Sci Med 58 (2004), pp. 899–911.
8 K. Hattori, M.A. McCubbin and D.N. Ishida, Concept analysis of good death in the Japanese community, J Nurs Scholarsh 38 (2006), pp. 165–170. View Record in Scopus | Cited By in Scopus (9)
9 C.M. Ruland and S.M. Moore, Theory construction based on standards of care: a proposed theory of the peaceful end of life, Nurs Outlook 46 (1998), pp. 169–175. Article | | View Record in Scopus | Cited By in Scopus (13)
10 W. Kongsuwan and R.C. Locsin, Promoting peaceful death in the intensive care unit in Thailand, Int Nurs Rev 56 (2009), pp. 116–122. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
11 M.A. Tayeb, E. Al-zamel, M.M. Fareed and H.A. Abouellail, A ”good death”: perspectives of Muslim patients and health care providers, Ann Saudi Med 30 (2010), pp. 215–221. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1)
12 WHO www.who.Int/countries/en/#s (2008).
13 European Values Study Group and World Values Surveys Association, European and World Values Surveys, four-wave integrated data file, 1981–2004 http://www.worldvaluessurvey.org/services/index.html.
14 A. Ghavamzadeh and B. Bahar, Communication with the cancer patients in Iran, information and truth, Ann N Y Acad Sci 809 (1997), pp. 261–265. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
15 Cancer Institute of Iran www.cancer-institute.ac.ir Access date is June 2010.
16 N. Sareming, Islamic teaching on death and practices toward the dying person, J Soc Sci Hum 3 (1997), pp. 75–91.
17 A. Sheikh, Death and dying: A Muslim perspective, J R Soc Med 91 (1998), pp. 138–140. View Record in Scopus | Cited By in Scopus (16)
18 M. Miyashita, T. Morita, K. Sato, K. Hirai, Y. Shima and Y. Uchitomi, Good Death Inventory: a measure for evaluating good death from the bereaved family members' perspective, J Pain Symptom Manage 35 (2008), pp. 486–498. Article | | View Record in Scopus | Cited By in Scopus (10)
19 H. Murata, Spiritual pain and its care in patients with terminal cancer: construction of a conceptual framework by philosophical approach, Palliat Support Care 1 (2003), pp. 15–21. View Record in Scopus | Cited By in Scopus (12)
20 K.E. Steinhauser, E.C. Clipp, M. McNeilly, N.A. Christakis, L.M. McIntyre and J.A. Tulsky, In search of a good death: observations of patients, families, and providers, Ann Intern Med 132 (2000), pp. 825–832. View Record in Scopus | Cited By in Scopus (372)
21 S. Iranmanesh, T. Haggstrom, K. Axelsson and S. Savenstedt, Swedish nurses' experiences of caring for dying people: a holistic approach, Holist Nurs Pract 23 (2009), pp. 243–252. View Record in Scopus | Cited By in Scopus (1)
22 N. Johnson, D. Cook, M. Giacomini and D. Willms, Towards a good death: end of life narratives constructed in an intensive care unit, Cult Med Psychiatry 24 (2000), pp. 275–295. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
23 R.L. Beckstrand, L.C. Callister and K.T. Kirchhoff, Providing a ”good death”: critical care nurses' suggestions for improving end-of-life care, Am J Crit Care 15 (2006), pp. 38–45. View Record in Scopus | Cited By in Scopus (49)
24 S.T. Tang, T.W. Liu, M.S. Lai, L.N. Liu, C.H. Chen and S.L. Koong, Congruence of knowledge, experiences, and preferences for disclosure of diagnosis and prognosis between terminally-ill cancer patients and their family caregivers in Taiwan, Cancer Invest 24 (2006), pp. 360–366. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (21)
25 M. Miyashita, M. Sanjo, K. Morita and Y. Uchitomi, Good death in cancer care: a nationwide quantitative study, Ann Oncol 18 (2007), pp. 1090–1097. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (31)
26 A. Montazeri, A. Tavoli, M.A. Mohagheghi, R. Roshan and Z. Tavoli, Disclosure of cancer diagnosis and quality of life in cancer patients: should it be the same everywhere, BMC Cancer 9 (2009), pp. 1–21.
27 R.J. Russac, C. Gatliff, M. Reece and D. Spottswood, Death anxiety across the adult years: an examination of age and gender effects, J Death Stud 31 (2007), pp. 549–561. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)
Original research
Sedigheh Iranmanesh PhDa, Habibollah Hosseini doctoral student
Abstract
Improving end-of-life care demands that first you define what constitutes a good death for different cultures. This study was conducted to evaluate a good death concept from the Iranian bereaved family members' perspective. A descriptive, cross-sectional study was designed using a Good Death Inventory (GDI) questionnaire to evaluate 150 bereaved family members. Data were analyzed by SPSS. Based on the results, the highest scores belonged to the domains “being respected as an individual,” “natural death,” “religious and spiritual comfort,” and “control over the future.” The domain perceived by family members as less important was “unawareness of death.” Providing a good death requires professional caregivers to be sensitive and pay attention to the preferences of each unique person's perceptions. In order to implement holistic care, caregivers must be aware of patients' spiritual needs. Establishing a specific unit in a hospital and individually treating each patient as a valued family member could be the best way to improve the quality of end-of-life care that is missing in Iran.
Article Outline
From a review of different studies, the core quality of a good death varies among cultures. In a qualitative study, Griggs4 analyzed perceptions of a “good death” among community nurses in England. Nurses identified several key themes for a good death, such as: symptom control, patient choice, honesty, spirituality, interprofessional relationships, effective preparation, organization, and provision of seamless care. American researchers concluded that a good death involves respect for the individual's autonomy with open communication among family members.5 Vig and Pearlman6 also reported that “good death” has an individual meaning for Americans and does not have a consensual meaning. In Ghana, Van der Greest7 found that a good death is integrated with a peaceful death, meaning peace with others, being at peace with one's own life and soul, dying in the fullness of time, dying at home, and being surrounded by relatives. For the Japanese, Hattori et al.8 found that a good death is a multidimensional, individual experience based on personal and sociocultural domains of life that incorporate the person's past, present, and future. In Norway, Ruland and Moore9 conducted research on the theory of a peaceful end of life which has five major concepts: not being in pain, experience of comfort, experience of dignity/respect, being at peace, and closeness to significant others/persons who care. In Thailand, people commonly used “peaceful death” instead of “good death.” Kongsuwan and Locsin10 reported that Thai intensive care unit nurses perceived peaceful death as awareness of dying, creating a caring environment, and promoting end-of-life care. In Muslim society, Tayeb et al11 identified three domains related to a good death: religion and faith, self-esteem and personal image, and satisfaction about family security.
After reviewing these studies, we determined there is no universal definition of good death and it is based on sociocultural context. The subject of death and dying has a religious and sociocultural background, yet Iranian health-care providers mainly depend on Western references. Moreover, upon reviewing the literature in Iran, no published study related to defining the concept of “good death” was located. This descriptive study was thus designed to determine what constitutes a good death in the Iranian context.
Context
Iran is one of the most ancient world civilizations and part of the Middle East culture. The population is approximately 67 million, and of this 51% is less than 20 years old and 6.5% is 65 or older.12 The majority (99.4%) of the people in Iran consider themselves as religious,13 and religious beliefs strongly and explicitly deal with death.14
Iranians are familiar with death. Besides the Iran–Iraq war and natural disasters in recent years, the major causes (65%) of death among Iranians are heart disease, cancer, and accidents.15 Apart from chronic disease, accidents seem to be a significant cause of death among Iranian people. In Iran, the overall national curriculum for registered nursing education includes just a few hours of academic education about death. End-of-life care remains a new topic in the Iranian health-care system. Hospice care units, which are common in Western countries, are not available in Iran.
Most religions are represented in this country; however, Islam is the most prevalent. Sareming16 indicates that Muslims are taught that Allah gives birth and death. Allah determines the appointed term for every human. Only Allah knows when, where, and how a person will die. For a Muslim, death is the transition from the earthly form of existence to the next.17 Tayeb et al11 explained that Muslims prefer to approach death with a certainty that someone is there to prompt them with the Shahadah, reciting a chapter of the Quran, dying in a position facing Mecca, and dying in a holy place such as a mosque.
Method
Design
There was approval from the heads of hospitals prior to the collection of data. The study employed a descriptive design and was conducted in two hospitals that had oncology units in southeast Iran.
Participants
Referring to the hospitals' and patients' documents, 150 bereaved family members of patients who died within 1 year were identified. They were called by the researcher and asked to participate in this study.
Background Information
At first, a questionnaire was designed in order to obtain background information which was assumed to influence the good death concept. It included questions about gender, age, marital status, previous studies about death, and level of education.
Instruments
The good death concept was evaluated using the Good Death Inventory (GDI). The GDI was designed by Miyashita et al18 for evaluating a good death from the bereaved family members' perspective. This scale has 51 items. The items are graded from 1 to 7 (1 = strongly disagree to 7 = strongly agree). A factor analysis made by Miyashita et al18 on research made in a Japanese setting revealed that the questions could be divided into 18 domains: (1) physical and psychological comfort, (2) dying in a favorite place, (3) good relationship with medical staff, (4) maintaining hope and pleasure, (5) not being a burden to others, (6) good relationship with family, (7) physical and cognitive control, (8) environmental comfort, (9) being respected as an individual, (10) life completion, (11) natural death, (12) preparation for death, (13) role accomplishment and contributing to others, (14) unawareness of death, (15) fighting against cancer, (16) pride and beauty, (17) control over the future, and (18) religious and spiritual comfort.
For translation from English into Farsi, the standard forward–backward procedure was applied. Translation of the items and the response categories was independently performed by two professional translators, and then temporary versions were provided. Afterward they were back-translated into English, and after a careful cultural adaptation the final versions were provided. Translated questionnaires went through pilot testing. Suggestions by family members were combined into the final versions.
Reliability and validity
The translated scale was originally developed and tested in a Japanese cultural context, which is different from the research contexts, so the validity and reliability of both scales were rechecked. A factor analysis (rotated component matrix) on the results was done in order to examine the context validity of the GDI. The concession of the items was similar to the Japanese results, and 18 components were identified. The validity of the scale was assessed through a content validity discussion. Scholars of statistics and nursing care have reviewed the content of the scale from religious and cultural aspects of death and agreed upon a reasonable content validity. To reassess the reliability of the translated scale, alpha coefficients of internal consistency and 3-week test–retest coefficients (n = 30) of stability were computed. The alpha coefficient for GDI was 0.68. The 3-week test–retest coefficient of stability for the GDI was 0.79. Therefore, the translated scale presented an acceptable reliability.
Data Collection and Analysis
Accompanied by a letter including some information about the aim of the study, the questionnaires were handed out by the second author to 150 family members who were introduced by the matron of two hospitals over 2 months (May/June 2010) in southeast Iran. Some oral information about the study was also given by the third author. Participation in the study was voluntary and anonymous. We distributed 150 sets of questionnaires. In all collected data, 98% of all questions were answered. Data from the questionnaires were analyzed using the Statistical Package for Social Scientists (SPSS, Inc., Chicago, IL). A Kolmogorov-Smirnov test indicated that the data were sampled from a population with normal distribution. Descriptive statistics of the sample and measures that were computed included frequencies, means, and reliability. Cross-table analysis (Spearman's test) was used to examine relationships among demographic factors and scores on the GDI.
Results
Participants
A descriptive analysis of the background information revealed that the participants belonged to the age group of 16–68 years, with a mean age of 33 years, and were mainly female (81%). About 68% were married, and the majority had an academic degree. Regarding personal study about death, 36.9% had read some things about death previously.
Findings
Descriptive analysis indicated that the highest scores belonged to the domains “being respected as an individual” (mean = 6.55), “natural death” (mean = 6.36), “religious and spiritual comfort” (mean = 6.02), and “control over the future” (mean = 6.55) (Table 1).
The domains and the components perceived as important by bereaved family members were (1) physical and psychological comfort, (2) dying in a favorite place, (3) maintaining hope and pleasure, (5) not being a burden to others, (6) good relationship with family, (7) physical and cognitive control, (8) environmental comfort, and (9) life completion. The domain perceived by family members as less important was “unawareness of death” (mean = 3.05).
Significant differences were found between some domains of a good death and demographic characters of family members. Older participants were more likely to perceive a good death as “being respected as an individual” and “having good relationships with family members.” Among participants, those who had a higher level of education were more likely to view a good death as “being respected as an individual” and “pride and beauty.” There was a negative correlation between level of education and “unawareness of death” (Table 2).
Discussion
According to the factor analysis, 18 domains contributing to a good death were identified. However, the domains of the “good death” concept that were perceived as important by bereaved family members were similar to those in Japan. This finding thus indicates that these perceptions are foundational elements of a good death, regardless of ethnicity or cultural differences.
The results indicated that most family members are likely to view a good death as “being respected as an individual” and having “control over the future.” According to Murata,19 approaching death can cause a sense that life is meaningless and a loss of the patient's well-being founded on temporality, relationships, and autonomy. Providing a good death means that dying patients are able and allowed to participate in the same human interactions that are important throughout life and appreciating patients as unique and “whole persons,” not only as “diseases” or cases.20 It means supporting patients' well-being through positive stimulation, for example, offering beautiful views and tasty meals.21 A good death is also perceived by family members as “religious and spiritual comfort.” Ghavamzadeh and Bahar14 claimed that among Iranians religious beliefs strongly and explicitly deal with the fact of death. This finding reflects the result of Tayeb et al,11 who found that Muslims believe that death is closely linked to faith. They appreciated the importance of access to any needed spiritual or emotional support. Steinhauser et al.20 also found that 89% of American patients and 85% of their families emphasize that a good death is “being at peace with God” and “prayer.”
Participants perceived a good death as a “natural death.” Johnson et al22 claimed that death without “machines,” “tubes,” and “lines” is considered more dignified and aesthetically pleasing. Withdrawal or withholding of treatment of the highly invasive and technological sort is conceptualized as restoring patient dignity and, to a small degree, personhood.22 Many deaths were not considered “good” because of inherent problems within a culture of care that usually strives to prolong life and prevent death.23 Similarly, Miyashita et al18 reported that most Japanese view unnecessary life-prolonging treatments such as vasopressors, antibiotics, and artificial hydration as barriers to achieving a good death. The domain perceived by family members as less important was “unawareness of death.” This is consistent with Steinhauser et al's20 finding that 96% of American patients emphasized “knowing what to expect about one's physical condition” achieves a good death. This is inconsistent with Tang et al's24 claims that in many traditional cultures (eg, most Asian countries and a few European cultures), in an effort to protect the patient from despair and a feeling of hopelessness, family caregivers often exclude patients from the process of information exchange. This is also in contrast to Miyashita et al's[18] and [25] findings, where many Japanese do not want to know the seriousness of their condition. Our findings could be explained by the other results of this study. The results indicated that the majority of participants had a high level of education. The other findings showed there is a negative correlation between level of education and “unawareness of death.” Since the majority of participants were well-educated, it can be concluded that they were less likely to view a good death as “unawareness of death.” This has also been found by Montazeri et al.26
The results showed that the family members' age was correlated with some aspects of a good death. Miyashita et al18 also found that the older the family member, the more positively he or she would look on the patient's death. They claimed that death at younger ages tended to be evaluated as a bad death. This could be explained by their earlier study, where they found that age and psychosocial maturity inversely related to death anxiety.27 Based on the results, level of education positively influenced some domains of a good death. There was a negative correlation between level of education and “unawareness of death,” with Montazeri et al26 finding that Iranian patients with a low level of education were more likely to not know the diagnosis.
Conclusion
According to the results of this study, providing a good death requires professional caregivers to be sensitive and pay attention to the preferences of each unique person's perceptions through her or his senses. This includes views, tastes, sounds, smells, and bodily contact. The ability of a dying person to see a sunset may seem petty but is important in providing high-quality care for people at the end of their lives. The same goes for the other senses. These circumstances deserve attention in all educational programs and especially in programs dealing with end-of-life care. In order to implement holistic care, caregivers must pay attention to patients' spiritual needs. Establishing a specific palliative care unit in a hospital and meeting each patient as a unique being and part of a family could be the best way to improve the quality of end-of-life care that is missing in Iran. It requires cultural preparation and public education through the media and by well-educated staff. Since demographic variables influenced the evaluation of a good death from the bereaved family members' perspective, public education needs different strategies.
Limitation
All data in this study were collected by use of self-report questionnaires. The dependence on self-report aspects in this study may have caused an overestimation of some of the findings due to variance, which is common in different methods. The respondents were predominantly female, which limits the generalization of the results for male respondents. Moreover, the convenience sample of Iranian bereaved family members, which is not representative of the entire Iranian population, could weaken the generalization of the findings. Further research is necessary to illuminate the concept of a good death as perceived by the general Iranian population.
References
1 I.M. Proot, H.H. Abu-Saad, H.F.J.M. Crebolder, K.A. Luker and G.A.M. Widdershoven, Vulnerability of family caregivers in terminal palliative care at home; balancing between burden and capacity, Scand J Caring Sci 17 (2003), pp. 113–121. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (53)
2 D.L. Patrick, R.A. Engelberg and J.R. Curtis, Evaluating the quality of dying and death, J Pain Symptom Manage 22 (2001), pp. 717–726. Article | | View Record in Scopus | Cited By in Scopus (96)
3 , Longman Dictionary of Contemporary English, Pearson Education, Canada (2003).
4 C.H. Griggs, Community nurses' perceptions of a good death: a qualitative exploratory study, Int J Palliat Nurs 16 (2010), pp. 140–149. View Record in Scopus | Cited By in Scopus (0)
5 J.E. Winland-Brown, Public's perceptions of a good death and assisted suicide, Issues Interdisc Care 3 (2001), pp. 137–144. View Record in Scopus | Cited By in Scopus (1)
6 E.K. Vig and R.A. Pearlman, Good and bad dying from the perspective of terminally ill men, Arch Intern Med 164 (2004), pp. 977–981. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (27)
7 S. Van der Greest, Dying peacefully: considering good death and bad death in Kwahu-Tafo, Ghana, Soc Sci Med 58 (2004), pp. 899–911.
8 K. Hattori, M.A. McCubbin and D.N. Ishida, Concept analysis of good death in the Japanese community, J Nurs Scholarsh 38 (2006), pp. 165–170. View Record in Scopus | Cited By in Scopus (9)
9 C.M. Ruland and S.M. Moore, Theory construction based on standards of care: a proposed theory of the peaceful end of life, Nurs Outlook 46 (1998), pp. 169–175. Article | | View Record in Scopus | Cited By in Scopus (13)
10 W. Kongsuwan and R.C. Locsin, Promoting peaceful death in the intensive care unit in Thailand, Int Nurs Rev 56 (2009), pp. 116–122. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
11 M.A. Tayeb, E. Al-zamel, M.M. Fareed and H.A. Abouellail, A ”good death”: perspectives of Muslim patients and health care providers, Ann Saudi Med 30 (2010), pp. 215–221. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1)
12 WHO www.who.Int/countries/en/#s (2008).
13 European Values Study Group and World Values Surveys Association, European and World Values Surveys, four-wave integrated data file, 1981–2004 http://www.worldvaluessurvey.org/services/index.html.
14 A. Ghavamzadeh and B. Bahar, Communication with the cancer patients in Iran, information and truth, Ann N Y Acad Sci 809 (1997), pp. 261–265. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
15 Cancer Institute of Iran www.cancer-institute.ac.ir Access date is June 2010.
16 N. Sareming, Islamic teaching on death and practices toward the dying person, J Soc Sci Hum 3 (1997), pp. 75–91.
17 A. Sheikh, Death and dying: A Muslim perspective, J R Soc Med 91 (1998), pp. 138–140. View Record in Scopus | Cited By in Scopus (16)
18 M. Miyashita, T. Morita, K. Sato, K. Hirai, Y. Shima and Y. Uchitomi, Good Death Inventory: a measure for evaluating good death from the bereaved family members' perspective, J Pain Symptom Manage 35 (2008), pp. 486–498. Article | | View Record in Scopus | Cited By in Scopus (10)
19 H. Murata, Spiritual pain and its care in patients with terminal cancer: construction of a conceptual framework by philosophical approach, Palliat Support Care 1 (2003), pp. 15–21. View Record in Scopus | Cited By in Scopus (12)
20 K.E. Steinhauser, E.C. Clipp, M. McNeilly, N.A. Christakis, L.M. McIntyre and J.A. Tulsky, In search of a good death: observations of patients, families, and providers, Ann Intern Med 132 (2000), pp. 825–832. View Record in Scopus | Cited By in Scopus (372)
21 S. Iranmanesh, T. Haggstrom, K. Axelsson and S. Savenstedt, Swedish nurses' experiences of caring for dying people: a holistic approach, Holist Nurs Pract 23 (2009), pp. 243–252. View Record in Scopus | Cited By in Scopus (1)
22 N. Johnson, D. Cook, M. Giacomini and D. Willms, Towards a good death: end of life narratives constructed in an intensive care unit, Cult Med Psychiatry 24 (2000), pp. 275–295. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
23 R.L. Beckstrand, L.C. Callister and K.T. Kirchhoff, Providing a ”good death”: critical care nurses' suggestions for improving end-of-life care, Am J Crit Care 15 (2006), pp. 38–45. View Record in Scopus | Cited By in Scopus (49)
24 S.T. Tang, T.W. Liu, M.S. Lai, L.N. Liu, C.H. Chen and S.L. Koong, Congruence of knowledge, experiences, and preferences for disclosure of diagnosis and prognosis between terminally-ill cancer patients and their family caregivers in Taiwan, Cancer Invest 24 (2006), pp. 360–366. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (21)
25 M. Miyashita, M. Sanjo, K. Morita and Y. Uchitomi, Good death in cancer care: a nationwide quantitative study, Ann Oncol 18 (2007), pp. 1090–1097. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (31)
26 A. Montazeri, A. Tavoli, M.A. Mohagheghi, R. Roshan and Z. Tavoli, Disclosure of cancer diagnosis and quality of life in cancer patients: should it be the same everywhere, BMC Cancer 9 (2009), pp. 1–21.
27 R.J. Russac, C. Gatliff, M. Reece and D. Spottswood, Death anxiety across the adult years: an examination of age and gender effects, J Death Stud 31 (2007), pp. 549–561. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)
Feasibility and acceptance of a telehealth intervention to promote symptom management during treatment for head and neck cancer
Treatment for head and neck cancer is most often a rigorous regimen of combination therapies, producing a multitude of distressing symptoms and side effects. While it is nearly impossible to circumvent the physical and psychosocial insults caused by such treatment, some interventions directed toward educating and supporting patients during active treatment have met with success.[1], [2], [3] and [4] Conversely, other efforts have demonstrated little impact[5] and [6] or have been poorly received,7 pointing to the need for effective, acceptable means to provide support during such difficult treatment.
Over the past 10 years, telemedicine technology has enabled innovative approaches for improving patient education, assessment, support, and communication during treatment for both acute and chronic diseases. A recent policy white paper8 described telemedicine technology as including “the electronic acquisition, processing, dissemination, storage, retrieval, and exchange of information for the purpose of promoting health, preventing disease, treating the sick, managing chronic illness, rehabilitating the disabled, and protecting public health and safety” (p. 2). This same paper suggests that national telemedicine initiatives are essential to health-care reform based upon their proven cost–effectiveness and clinical efficacy. However, cost savings and clinical effectiveness will be unrealized outcomes if the interventions are not feasible in practice or acceptable to the targeted population.
In the arena of cancer care, telephone-based systems have been used to report and monitor cancer symptoms with favorable compliance noted even when patients are expected to initiate calls on a regular basis.[9], [10], [11] and [12] Favorable acceptance ratings have also been reported by both patients and clinicians regarding computerized systems used to assess symptoms and quality of life (QOL) in cancer patients.[13], [14], [15], [16], [17], [18] and [19] In the United Kingdom, a handheld computer system was successfully used to monitor and support patients receiving chemotherapy for lung or colorectal cancer,20 and a study testing a dialogic model of cancer care expecting patients to respond to telehealth messaging on a daily basis over 6 months reported an 84% cooperation rate.21 In these studies, the majority of patients reported ease of use and acceptability of the technology. Survey research has found both urban and rural cancer patients to be receptive to medical and psychiatric services provided via telehealth.22
Published reports describing use of telehealth and computerized interventions during head and neck cancer treatment are less prevalent. Touch-screen computers were successfully used in the Netherlands to collect QOL and distress data from head and neck cancer patients.16 Videoconferencing has been used successfully to overcome geographical barriers to patient assessment[23], [24] and [25] and to provide speech–language pathology services to people living with head and neck cancers in remote areas. Reported use of telehealth management appears promising for providing timely access to care for those who are geographically isolated.26
A research group based in the Netherlands developed and tested a comprehensive electronic health information support system for use in head and neck cancer care.27 The system had four patient-related functions: facilitating communication between patients and health-care providers, providing information about the disease and its treatment, connecting patients with other patients similarly diagnosed, and monitoring patients after hospital discharge. The system was found to be well-accepted and appreciated by participating patients, and its use enabled early identification and direct intervention for patient problems.27 A clinical trial of the telehealth application showed improved QOL in five of 22 studied parameters for the treatment group.28 However, 20 of the 59 patients eligible for the intervention group refused participation; 11 (55%) of these stated computer-related concerns as their reason for nonparticipation.
Knowing that head and neck cancer patients experience a high burden of illness and often have significant communication, socioeconomic, and geographic barriers to care, our team developed a telehealth intervention using a simple telemessaging device to circumvent communication barriers and perceived technical challenges associated with computer-based systems to provide education and support to patients in their own home and on their own time schedule.29 Overall, we hypothesized that patients receiving the intervention would experience less symptom distress, improved QOL, increased self-efficacy, and greater satisfaction with symptom management than those in the control group. However, as a first step toward examining the efficacy and effectiveness of this intervention, this study examined both quantitative and qualitative indicators of its feasibility and acceptance among patients undergoing treatment for head and neck cancer.
Methods
Design
Subsequent to study approval by the University of Louisville's Human Subjects Protection Office, a randomized clinical trial comparing the telehealth intervention to standard care was conducted using a two-group parallel design. This study reports on the intervention's feasibility and acceptance in the treatment group of 44 patients.
Site
Participants were recruited from patients receiving care from the Multidisciplinary Head and Neck Cancer Team at the James Graham Brown Cancer Center (JGBCC) over a 2-year period (June 2006 through June 2008). The team consisted of head and neck surgeons, medical oncologists, radiation oncologists, nurses, a pathologist, a speech therapist, a registered dietician, a psychologist, and a social worker. This team developed a comprehensive assessment and treatment plan during each patient's initial visit to the clinic and coordinated patient care throughout the treatment process.
Sample
Patients eligible for study participation met the following inclusion criteria: (1) initial diagnosis of head or neck cancer including cancers of the oral cavity, salivary glands, paranasal sinuses and nasal cavity, pharynx, and larynx; (2) involvement in a treatment plan including one or more modalities (ie, surgery, chemotherapy, radiation, or any combination); (3) capacity to give independent informed consent; and (4) ability to speak, read, and comprehend English at the eighth-grade level or above. Patients were excluded from participation if they had no land telephone line, had a thought disorder, were incarcerated, or had compromised cognitive functioning.
All patients scheduled for assessment received an explanation of the research study via print materials prior to their first clinic visit. During their first scheduled clinic visit, all patients identified as eligible were approached by a member of the research study staff, who briefly explained the study and asked if they might be interested in study participation. Because of the stress and content of this first clinic visit, interested patients were contacted later by phone to schedule an additional visit to review the study and obtain informed consent.
During the informed consent meeting, the study procedures were explained in detail. If the patient agreed and signed a consent form, a randomization grid which considered the patient's particular treatment plan was used to assign the patient to either the control or the experimental group. Baseline data were also collected during this first visit.
Description of the Intervention
The technology selected for implementing the intervention was the Health Buddy® System, a commercially available, proprietary system produced and maintained by Robert Bosch Healthcare Palo Alto, CA. The Health Buddy, the appliance used for interaction between the participant and the health-care provider, is a user-friendly, easily visible, electrical device that attaches to the user's land phone line (see Figure 1). Questions and information are displayed on the liquid crystal display (LCD) screen of the 6 × 9–inch appliance. The individual responds to questions by pressing one of the four large buttons below the screen. The research team selected the technology provider based on the ability of the technology to perform in accordance with the research objectives.
Symptom control algorithms developed using participatory action research (surveys of current and past patients and clinicians) and evidence-based practice were programmed into the telehealth messaging system (see article by Head et al,29 which details the algorithm topic selection and development process). The algorithms addressed 29 different symptoms and side effects of treatment, consisting of approximately 100 questions accompanied by related educational and supportive responses. Patients were asked three to five questions daily related to the symptoms anticipated during their treatment scenario. Depending upon their response, they would receive specific information related to symptom self-management, including recommendations as to when to contact their clinicians. The algorithms were constructed with the goal of encouraging self-efficacy and independent action on the part of the participant. See Figure 2 for an example of the branching algorithms.
Participants randomly assigned to the treatment group immediately had the Health Buddy connected to a land telephone line in their home. Most (40%) chose to place it in their kitchen, while another 26% placed it in their bedrooms; most often, it was in a highly visible location, serving to remind the participant to respond. Research study staff delivered, installed, and demonstrated how to operate the equipment. Installation was simple and required only minutes. A tutorial programmed into the Health Buddy taught participants how to reply to questions appearing on the monitor using the four large keys below the possible answers or a rating scale which would appear depending on the type of question asked.
During the early hours of the morning, the device would automatically call a toll-free number. Responses to the previous day's questions were uploaded, and questions and related information for the next day were downloaded over the telephone line onto a secure server. Phone service was never disrupted by the device; if the phone was in use, the system would connect later to retrieve and download information. Once new content was transferred, a green light on the device would flash to alert the participant that new questions were available for response. Once the participant pressed any of the keys, the new algorithms would begin appearing on the monitor screen.
Participants were instructed to begin responding on the first day they received treatment or on the first day after returning home from surgery. They were asked to continue responding daily (unless hospitalized for treatment) throughout the treatment period and for approximately 2 weeks posttreatment as treatment-induced symptoms continue during that period of time. Study staff contacted participants when treatment was complete and scheduled a date to pick up the appliance and end daily responding. Daily patient responses required 5–10 minutes.
Participant responses could be viewed by study staff via Internet access 1 day after being answered. Responses were monitored daily by study nurses. Symptoms unrelieved over time or symptoms targeted as requiring immediate intervention (ie, serious consideration of suicide) would result in the study nurse contacting the patient directly by phone and/or contacting clinicians to assure immediate intervention. However, it is important to note that this direct intervention by study staff was infrequent as most symptoms were addressed independently by the participant as desired. If a participant had not reported a period of planned hospitalization and did not respond for 3 consecutive days, study staff would contact the patient by phone to ascertain the reason for noncompliance.
Measures
The following indicators were selected as measures of acceptance (accrual rate), feasibility (utilization, nurse-initiated contacts), and/or satisfaction (satisfaction ratings). Narrative responses and a poststudy survey provided additional data examining acceptance, feasibility, and satisfaction with the intervention. In addition, demographic and medical information as well as measures assessing primary study outcomes were collected from each participant. Table 1 lists all measures and the study time point when they were administered.
MEASURES | PRETREATMENT | DURING TREATMENT | POSTTREATMENT | CUMULATIVE |
---|---|---|---|---|
Demographics | X (baseline) | X | ||
Accrual rate | X | |||
Utilization rate | X | |||
FACT-H&N | X (baseline) | X (mid-tx) | X (2–4 weeks post-tx) | |
MSAS | X (baseline) | X (mid-tx) | X (2–4 weeks post-tx) | |
Satisfaction with technology | X | |||
Nurse-initiated contacts | X | |||
Exit interview | X (end of treatment) | |||
Poststudy written survey | X (60–90 days post-tx) |
tx = treatment
Accrual rate
The number of individuals assessed for study eligibility, reasons for exclusion or noncompletion, and numbers included in the analysis were all recorded to examine acceptance of the intervention and identify issues with the intervention or technology affecting participation.
Utilization
Feasibility was operationalized as device utilization using the percentage of days on which a participant responded to the Health Buddy. This was calculated using the number of days the participant responded to the telehealth device divided by the number of days the participant had the device and was expected to respond. These data were maintained and provided by the telehealth provider (Robert Bosch Healthcare).
Nurse-initiated contacts with participants and/or clinicians
The number of occasions on which a nurse decided to intervene was used as an indicator of feasibility under the premise that the goal of the intervention was to support and encourage patient-driven efforts to seek care for persistent or troubling symptoms. If a patient reported a symptom, he or she was given management information and encouraged to discuss problems further with the clinician either by phone or during clinic visits. If a patient continued to report an unresolved symptom or if the symptom required immediate intervention (ie, suicide threat), the research nurse reviewing responses would contact the patient and/or clinician to ascertain why and/or assist with its resolution. These nurse-initiated contacts should be infrequent if the intervention is achieving the goal of developing patient self-efficacy.
Satisfaction ratings
Items assessing satisfaction with the technology were also administered to participants via the telehealth messaging device. Questions related to satisfaction with the initial setup of the telehealth appliance were asked at the beginning of the intervention. Ongoing satisfaction with the device, messaging content, and the health-care provider were assessed every 90 days. The specific questions asked are detailed in Table 4.
Narrative data
Upon completion of the intervention, participants in the treatment group completed an exit interview using open-ended questions regarding the utility of the intervention, relevance of the algorithms, value or burden of item repetition in the algorithms, symptoms or problems experienced that were not addressed by the intervention, and general comments.
Poststudy survey
A final survey was mailed to participants several months after completion of the study, asking for additional feedback about the impact of the intervention. Specifically, participants (both treatment and control groups) were asked about their overall satisfaction with the treatment and services at the cancer center, their satisfaction with information received about their treatment, the response(s) received when they attempted contact with the health-care team after hours, the amount of support received, their current smoking and alcohol usage, and several demographic questions not earlier assessed or available through record review (years of education, highest degree, income range). Those receiving the intervention were also asked about the impact of the Health Buddy on their care and actions taken in response to the algorithms.
Demographic and medical information
Demographic information was collected using the initial survey, and information about the participant's medical history, condition, treatments received, treatment timing, complications, comorbidities, and treatment response was collected via retrospective medical record review subsequent to completion of the clinical trial.
Outcome measures
While outcomes of the clinical trial are not the subject of this article, the results of QOL and symptom burden measures for the treatment group only are included here because of their relationship with device utilization. The two measures included the Functional Assessment of Cancer Therapy–Head and Neck Scale and the Memorial Symptom Assessment Scale and were administered at baseline (before beginning treatment), mid-treatment, and posttreatment.
• Functional Assessment of Cancer Therapy–Head and Neck Scale (FACT-H&N). The FACT-G (general) is a multidimensional QOL instrument designed for use with all cancer patients. The instrument has 28 items divided into four subscales: Functional Well-Being, Physical Well-Being, Social Well-Being, and Emotional Well-Being. This generic core questionnaire was found to meet or exceed requirements for use in oncology based upon ease of administration, brevity, reliability, validity, and responsiveness to clinical change.30 Added to the core questionnaire is the head and neck–specific subscale, consisting of 11 items specific to this cancer site. A Trial Outcome Index (TOI) is also scored and is the result of the physical, functional, and cancer-specific subscales. List et al31 found the FACT-H&N to be reliable and sensitive to differences in functioning for patients with head and neck cancers (Cronbach's alpha was 0.89 for total FACT-G and 0.63 for the head and neck subscale in this study of 151 patients). Additionally, head and neck cancer patients found the FACT-H&N relevant to their problems and easy to understand, and it was preferred over other validated head and neck cancer QOL questionnaires.32 The FACT-H&N was chosen for this study because it (1) is nonspecific related to a treatment modality or subsite among head and neck cancers, (2) allows comparison across cancer diagnoses while still probing issues specific to head and neck cancer, (3) is short and can be completed quickly, (4) includes the psychosocial domains of social/family and emotion subscales as well as physical and functional areas, and (5) is self-administered.
• Memorial Symptom Assessment Scale (MSAS). This multidimensional scale measures the prevalence, severity, and distress associated with the most common symptoms experienced by cancer patients. Physical and emotional subscale scores as well as a Global Distress Index (GDI, considered to be a measure of total symptom burden) can be generated from patient responses. The MSAS has demonstrated validity and reliability in both in- and outpatient cancer populations.[33], [34] and [35] Initial psychometric analysis by Portenoy et al34 used factor analysis to define two subscales: psychological symptoms and physical symptoms with Cronbach alpha coefficients of 0.88 and 0.83, respectively; convergent validity was also established. It was chosen for this study because of its proven ability to measure both the presence and the intensity of experienced symptoms.[33], [35], [36], [37] and [38]
Data Analysis
Quantitative data were documented and analyzed using the Statistical Package for the Social Sciences (SPSS, Inc., Chicago, IL), version 16. Descriptive statistics were calculated to describe the sample and assess study outcomes, including feasibility and acceptability of the intervention. To ascertain relationships between usage of the device and demographic and medical information, a series of correlational analyses using Spearman's rho were conducted. This nonparametric test was chosen over Pearson's r because of the small sample size, the lack of a normal distribution for several of the variables, and the ordinal nature of several of the variables. Multiple regression analyses were also planned, but lack of significant bivariate correlations precluded multivariate analysis.
Qualitative responses to open-ended questions were analyzed to identify themes and direct quotations illustrating those themes.
Descriptive analysis of the treatment group's responses to the outcome measures (QOL and symptom burden) was done to ascertain changes over the course of the intervention using the mean scores at baseline, during treatment, and posttreatment.
Results
Description of Participants
Participants randomly assigned to the intervention group (n = 45) were an average age of 59 years (±11.7), and most were covered by private (34%) or public (48%) insurance. On average, participants had completed 13.5 years (±3.0) of formal education. Thirty-nine (87%) of the participants were male and 91% were Caucasian.
With regard to medical information, participants were predominantly diagnosed with stage II cancers of the head and neck (36%). The most prevalent site was the larynx (12 patients), followed by the tongue and the base of the tongue (seven patients) and unknown primary (seven patients). The vast majority received chemotherapy (32, or 71%) and/or radiation (42, or 93%).
Additional details regarding demographic and medical characteristics of the sample are provided in Table 2.
FREQUENCY | VALID PERCENT | |
---|---|---|
Gender (n = 44) | ||
Male | 39 | 88.6 |
Female | 5 | 11.3 |
Race (n = 44) | ||
Caucasian | 40 | 90.9 |
African American | 4 | 9.0 |
Tumor stage (n = 44) | ||
I | 7 | 15.9 |
II | 15 | 34.0 |
III | 11 | 25.0 |
IV | 4 | 9.0 |
Unable to determine | 5 | 11.4 |
Unknown | 2 | 4.5 |
Site of cancer (n = 44) | ||
Larynx | 12 | 27.2 |
Tongue, base of tongue | 7 | 15.9 |
Unknown primary | 7 | 15.9 |
Tonsillar | 4 | 9.0 |
Other H&N sites | 14 | 31.8 |
Insurance status (n = 44) | ||
No insurance | 8 | 18.2 |
Medicaid | 1 | 2.3 |
Medicare | 2 | 4.5 |
Medicaid and Medicare | 1 | 2.3 |
Medicare and supplement | 9 | 20.5 |
Medicare and VA benefits | 2 | 4.5 |
Veteran benefits only | 6 | 13.6 |
Private insurance | 15 | 34.1 |
Highest educational degree (n = 20)a | ||
Less than high school | 3 | 15.0 |
High school or GED | 9 | 45.0 |
Associate's/bachelor's degree | 4 | 20.0 |
Masters, PhD, or MD | 2 | 10.0 |
Other | 2 | 10.0 |
Income range (n = 18)a | ||
$20,000 or less | 5 | 27.8 |
$20,001–50,000 | 5 | 27.8 |
$70,001–100,000 | 5 | 27.8 |
Over $100,000 | 3 | 16.7 |
Percent of poverty in zip code area (n = 44) | ||
2.8–5.1% | 11 | 25.0 |
5.9–8.6% | 11 | 25.0 |
9.0–11.9% | 10 | 22.7 |
12.3–45.9% | 12 | 27.2 |
Feasibility and Acceptability
Accrual rate
Of the 185 patients assessed for eligibility during the 2-year recruitment period, 105 were excluded. See Figure 3 for a detailed depiction of study accrual for both the treatment and control groups. Thirty-three (31%) were excluded because they did not have a land phone line, a requirement for transmitting the algorithms to the Health Buddy appliance. Most of these had cell phones only. No potential participants refused participation due to issues related to operation of the technology itself.
Device utilization
Participants used the telehealth device for an average of 70.7 days (±26.7), which constituted 86.3% (±15.0) of the total days available for use. Of note, the median percentage of use was 94.2% and the modal percentage was 100%, indicating that the vast majority of participants consistently used the telehealth device. The participant with the lowest usage rate used the device 46% of the days available.
By far, the most common reason for Health Buddy nonresponse was patient hospitalization. Two subjects traveled out of town frequently on weekends and would leave the Health Buddy at home. One subject had accidentally unhooked the Health Buddy, and a home visit was made to reconnect the device into the patient's phone line.
Nurse-initiated contacts with participants and/or clinicians
Of the 45 enrolled patients, 33 required additional contact with a research nurse (see Table 3). The most common reasons patients were contacted were nonresponse for 3 consecutive days (38.3%), repeated reporting of high levels of unrelieved pain (30%), and suicidal thoughts (10%). In all, 120 calls were placed: one call for every 25.9 response days. In every case, the problem was resolved.
NUMBER OF PATIENTS | PROBLEM | OUTGOING CALLS | RESOLUTION |
---|---|---|---|
15 | No response on Health Buddy for 3 consecutive days | 46 | Patient teaching |
17 | Pain-related issues | 36 | Advocacy/referral/patient teaching |
5 | Suicidal thoughts | 12 | Advocacy/referral |
7 | G-tube problems | 8 | Patient teaching |
5 | Sadness/depression | 6 | Advocacy/referral |
3 | Multiple symptoms | 3 | Advocacy/referral |
3 | Nausea/vomiting | 4 | Referral/patient teaching |
2 | Coughing/excessive secretions | 2 | Patient teaching |
2 | Constipation | 2 | Patient teaching |
1 | Stomatitis | 1 | Referral |
Satisfaction ratings
Responses to surveys programmed into the Health Buddy system are displayed in Table 4. Overall, respondents responded favorably, finding the installation to be easy, the content to be helpful, and the overall experience to be positive.
PERCENT OF RESPONDENTS | |
---|---|
Installation satisfaction | |
Installation problems? | |
Yes | 2 |
No | 98 |
Any difficulty completing the first training questions? | |
Yes | 7 |
No | 94 |
Length of installation? | |
2–5 minutes | 52 |
6–10 minutes | 41 |
11–15 minutes | 4 |
16–20 minutes | 2 |
Content satisfaction | |
Overall, I think the Health Buddy questions are | |
Very easy | 44 |
Somewhat easy | 16 |
Neutral | 32 |
Somewhat difficult | 4 |
Difficult | 4 |
Repeating questions reinforced knowledge and understanding | |
Strongly agree | 56 |
Somewhat agree | 28 |
Neutral | 12 |
Somewhat disagree | 4 |
Strongly disagree | 0 |
Understanding of my health condition | |
Much better | 64 |
Somewhat better | 20 |
Neutral | 16 |
Somewhat worse | 0 |
Much worse | 0 |
Managing my health condition | |
Much better | 52 |
Somewhat better | 44 |
Neutral | 4 |
Somewhat worse | 0 |
Much worse | 0 |
Recommend the device to others | |
Very willing | 80 |
Somewhat willing | 12 |
Neutral | 4 |
Somewhat unwilling | 0 |
Very unwilling | 4 |
Overall satisfaction | |
Satisfaction with device | |
Very satisfied | 45 |
Satisfied | 35 |
Somewhat satisfied | 15 |
Not very satisfied | 5 |
Satisfaction with the communication between you and your doctor or nurse | |
More satisfied | 65 |
No difference | 30 |
Less satisfied | 5 |
Ease of using the device | |
Very easy | 85 |
Easy | 15 |
Not easy | 0 |
Overall experience with the device | |
Positive | 85 |
Neutral | 15 |
Negative | 0 |
Continue to use the device | |
Very likely | 40 |
Likely | 40 |
Somewhat likely | 15 |
Not very likely | 0 |
Narrative comments
During the exit interview, participants were asked, “How was having the Health Buddy helpful to you?” Responses could be categorized into two major themes: (1) the Health Buddy provided needed information and (2) the Health Buddy improved my self-management during treatment.
Statements made related to the information provided included the following:
- • It gave me information on what could be expected from treatment
• It was a constant reminder of things to watch for
• It kept me abreast of my total condition at all times
• It gave good directions so I didn't have to ask at the cancer center
• It gave good suggestions on treatments (home remedies) such as gargles, care of feeding tube, exhaustion, and everyday symptoms
Statements made indicative that the Health Buddy improved self-management included the following:
- • I learned what I could do to make myself feel better
• It helped me manage my symptoms
• It taught me about symptom management and how to handle problems
• It let me know whether to contact a doctor or use self-care
• It gave me who to call for problems and some things to try
• It kept me aware of what I needed to do in order to make the period easier
• It reminded me to take my meds and exercise
Additionally, some participants noted the support they felt from having the Health Buddy interventions during treatment in saying the following:
- • It kind of helped my depression through acknowledging it and giving me something to do
• It made me feel I was not the only one who had experience with these things
• It comforted me because I knew what was going to happen
Poststudy survey
Twenty (45%) of the 44 patients who received the intervention responded to the mailed poststudy survey. When asked if they felt they received better care because they had the device, 13 of the 20 (65%) responded that they did. Eighteen (90%) of the treatment group responders stated they were very satisfied with their care (one stated “somewhat satisfied”) and 20 (100%) said they would recommend the cancer center for treatment. Nineteen (95%) stated they received adequate support during treatment.
Outcome Measures
Mean scores on the FACT-H&N and subscales and the MSAS and subscales taken pre-, during, and posttreatment are displayed in Table 5. As expected, average QOL scores declined during treatment, while symptom distress increased, with a return to near baseline scores posttreatment.
SCALE/SUBSCALE | PRETREATMENT | DURING TREATMENT | POSTTREATMENT |
---|---|---|---|
Total FACT-H&N | 100.3 | 85.6 | 101.5 |
FACT-G | 74.3 | 69.4 | 78.5 |
Trial Outcome Index | 62.6 | 46.0 | 65.0 |
Physical Well-Being | 21.2 | 17.6 | 21.1 |
Functional Well-Being | 15.6 | 12.5 | 17.4 |
Emotional Well-Being | 21.1 | 22.3 | 22.2 |
Social Well-Being | 21.1 | 22.3 | 22.2 |
Total MSAS | 0.7 | 1.1 | 0.8 |
Global Distress Index | 1.1 | 1.8 | 1.3 |
Physical | 0.7 | 1.5 | 1.1 |
Psychological | 1.1 | 1.2 | 0.8 |
Correlations
The relationships between percentage usage per patient and the following variables were evaluated: age, income, years of education, tumor stage, and percent poverty in patient's zip code. Percent poverty in zip code area was intended to be a surrogate measure of the patient's socioeconomic status. Results are displayed in Table 6. No significant correlations were noted, although years of education and percentage poverty in zip code showed a trend toward significance.
VARIABLE (VS % USAGE) | RELATIONSHIP | |
---|---|---|
SPEARMAN'S RHO RS | SIGNIFICANCE (ONE-TAILED) | |
Percent poverty in zip code | 0.213 | 0.083 |
Age | 0.146 | 0.173 |
Years of education | −0.325 | 0.081 |
Income | −0.292 | 0.120 |
Tumor stage | 0.196 | 0.122 |
Physical Well-Being (during treatment) | 0.310 | 0.048 |
Emotional Well-Being (during treatment) | 0.315 | 0.042 |
Although a multivariate model was planned, the lack of significant bivariate correlations precluded the need for multivariate analysis.
When percent usage was correlated with FACT-H&N total and subscales taken at baseline, during active treatment, and posttreatment, significant positive correlations were found between the percentage used and the Physical Well-Being subscale score during treatment (Spearman's rho = 0.310, P = 0.048) and between percentage used and the Emotional Well-Being subscale during treatment (Spearman's rho = 0.315, P = 0.042).
There were no significant correlations between percentage usage and the scores on the MSAS.
Discussion
Both qualitative and quantitative measures indicate that using telehealth to support symptom management during aggressive cancer treatment is both feasible and well-accepted. Patient users were not intimidated by this particular technology as it was simple to set up and use and required no previous computer training to operate. The Health Buddy was viewed as providing important and useful information. Overall, users felt that it improved their ability to self-manage their disease and the side effects of treatment and provided a sense of support and security.
Unlike other studies which use telehealth devices to monitor patient symptoms, our goal was to increase patient self-management of the symptoms experienced during intensive medical treatment, therefore avoiding increased burden on the medical system. The fact that the research nurse overseeing the responses needed to intervene only once every 25.9 days speaks to the ability of the intervention to have a positive impact on utilization of medical services.
The lack of significant relationships between usage and descriptive variables such as age and years of education suggests that the intervention was equally acceptable to all subgroups. Factors such as age, previous computer literacy, educational obtainment, and socioeconomic status did not significantly differentiate our study population in terms of compliance as verified by usage percentages.
The significant relationships found between the percentage used and the subscale scores on Physical Well-Being and Emotional Well-Being during treatment may indicate that increased use of the telemessaging intervention during treatment resulted in better physical and emotional aspects of QOL.
The high rate of daily compliance with the intervention in spite of differentiating personal variables and the severity of the treatment regimen may have been due to one or a combination of the following factors:
- • the simplicity of the technology
• the visibility of the appliance (often placed in the kitchen or living area of the home) and its flashing green light as cues to the need to respond
• the usefulness of the information provided
• the use of simple messaging language presented in an encouraging, positive manner
• affirmations related to application of the symptom management protocols suggested
• curiosity related to the day's messaging and the motivational saying which always appeared at the end
• knowledge that someone was reviewing the responses, tracking and intervening when the participant did not respond for several days
Our study supports the benefits of telehealth interventions noted by providers in a study by Sandberg et al39: opportunities for more frequent contact, greater relaxation and information due to the ability to interact in one's own home, increased accessibility by those frequently underserved, and timely medical information and monitoring. Similar to the study done in the Netherlands,[27] and [28] this study noted technological problems as the primary disadvantage; but in our intervention, we had no problems with the technology or equipment.
Although computerized technology served as a barrier to previous telehealth research, the lack of a land-based phone line was a factor preventing participation in the current study. Indeed, many participants maintained only wireless communication devices, which were not compatible with the version of the Health Buddy that was employed in this study. However, improvements in the technology since completion of this study now allow for wireless access to the appliance or provision of an independent wireless messaging device for those without such access in their own homes.
Although the data generally support the feasibility and acceptability of the telehealth-based intervention, the results should be interpreted in the context of a few study limitations. In particular, the sample size was somewhat small, and data pertaining to the socioeconomic status of participants were not available for all participants. Second, the study did not include measures of the patient's direct interactions with health-care providers during the study or specific data related to their health-care utilization (eg, emergency room visits, preventable inpatient hospitalizations, emergency calls to clinicians). The collection of more exhaustive measures of health-care utilization was limited by resources but is planned for subsequent studies. Finally, concerns regarding subject burden limited assessment of the usability of the telehealth device.
Although compliance with utilization expectations and completion of study measures was excellent during the course of the intervention, response to the follow-up survey mailed several months later was less than 50%. This low response rate was most likely due to several factors: (1) this survey was sent at the conclusion of the entire study (by this time, patients were 0–21 months past their active participation); (2) it was a mailed survey with no additional contact or follow-up effort to increase response rate; (3) participants may have felt that they had already shared their opinions in the exit interview and may have felt overburdened by study measures at this point; and (4) participants may have died, moved, or been medically unable to respond. This lack of response did limit our ability to evaluate the longitudinal impact of the intervention.
Conclusions
This telehealth intervention proved to be an acceptable and feasible means to educate and support patients during aggressive treatment for head and neck cancer. Patient compliance with telehealth interventions during periods of extreme symptom burden and declining QOL is feasible if simple technology cues the patient to participate, offers positive support and relevant education, and is targeted or tailored to their specific condition.
1 C.D. Llewellyn, M. McGurk and J. Weinman, Are psycho-social and behavioural factors related to health related-quality of life in patients with head and neck cancer?: A systematic review, Oral Oncol 41 (5) (2005), pp. 440–454. Article | | View Record in Scopus | Cited By in Scopus (24)
2 K.T. Vakharia, M.J. Ali and S.J. Wang, Quality-of-life impact of participation in a head and neck cancer support group, Otolaryngol Head Neck Surg 136 (3) (2007), pp. 405–410. Article | | View Record in Scopus | Cited By in Scopus (6)
3 P.J. Allison et al., Results of a feasibility study for a psycho-educational intervention in head and neck cancer, Psychooncology 13 (2004), pp. 482–485. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
4 L.H. Karnell et al., Influence of social support on health-related quality of life outcomes in head and neck cancer, Head Neck 29 (2) (2007), pp. 143–146. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (19)
5 K.M. Petruson, E.M. Silander and E.B. Hammerlid, Effects of psychosocial intervention on quality of life in patients with head and neck cancer, Head Neck 25 (7) (2003), pp. 576–584. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)
6 J.R.J. deLeeuw et al., Negative and positive influences of social support on depression in patients with head and neck cancer: a prospective study, Psychooncology 9 (2000), pp. 20–28. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)
7 J. Ostroff et al., Interest in and barriers to participation in multiple family groups among head and neck cancer survivors and their primary family caregivers, Fam Process 43 (2) (2004), pp. 195–208. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
8 R.L. Bashshur et al., National telemedicine initiatives: essential to healthcare reform, Telemed J E Health 15 (6) (2009), pp. 1–11.
9 K. Davis et al., An innovative symptom monitoring tool for people with advanced lung cancer: a pilot demonstration, J Support Oncol 5 (8) (2007), pp. 381–387. View Record in Scopus | Cited By in Scopus (8)
10 K.H. Mooney et al., Telephone-linked care for cancer symptom monitoring: A pilot study, Cancer Pract 10 (3) (2002), pp. 147–154. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (30)
11 R.H. Friedman et al., The virtual visit: using telecommunications technology to take care of patients, J Am Med Inform Assoc 4 (1997), pp. 413–425. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
12 A. Weaver et al., Application of mobile phone technology for managing chemotherapy-associated side-effects, Ann Oncol 18 (11) (2007), pp. 1887–1892. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
13 D. Berry et al., Computerized symptom and quality-of-life assessment for patients with cancer: Part 1: Development and pilot testing, Oncol Nurs Forum 31(5 (2004), pp. E75–E83. Full Text via CrossRef
14 K. Mullen, D. Berry and B. Zierler, Computerized symptom and quality-of-life assessment for patients with cancer: Part II: Acceptability and usability, Oncol Nurs Forum 31 (5) (2004), pp. E84–E89. Full Text via CrossRef
15 B. Fortner et al., The Cancer Care Monitor: psychometric content evaluation and pilot testing of a computer administered system for symptom screening and quality of life in adult cancer patients, J Pain Symptom Manage 26 (6) (2003), pp. 1077–1092. Article | | View Record in Scopus | Cited By in Scopus (43)
16 R. de Bree et al., Touch screen computer-assisted health-related quality of life and distress data collection in head and neck cancer patients, Clin Otolaryngol 33 (2) (2008), pp. 138–142. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
17 H. Huang et al., Developing a computerized data collection and decision support system for cancer pain management, Comput Inform Nurs 21 (4) (2003), pp. 206–217. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
18 D.J. Wilkie et al., Usability of a computerized pain report in the general public with pain and people with cancer pain, J Pain Symptom Manage 25 (3) (2003), pp. 213–224. Article | | View Record in Scopus | Cited By in Scopus (35)
19 K. Kroenke et al., Effect of telecare management on pain and depression in patients with cancer: a randomized trial, JAMA 304 (2) (2010), pp. 163–171. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
20 N. Kearney et al., Utilizing handheld computers to monitor and support patients receiving chemotherapy: results of a UK-based feasibility study, Support Care Cancer 14 (7) (2006), pp. 742–752. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
21 N.R. Chumbler et al., Remote patient–provider communication and quality of life: empirical test of a dialogic model of cancer care, J Telemed Telecare 13 (2007), pp. 20–25. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
22 A.L. Grubaugh et al., Attitudes toward medical and mental health care delivered via telehealth applications among rural and urban primary care patients, J Nerv Ment Dis 196 (2) (2008), pp. 166–170. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
23 J. Stalfors et al., Accuracy of tele-oncology compared with face-to-face consultation in head and neck cancer case conferences, J Telemed Telecare 7 (2001), pp. 338–343. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
24 C. Dorrian et al., Head and neck cancer assessment by flexible endoscopy and telemedicine, J Telemed Telecare 15 (2009), pp. 118–121. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
25 J. Stalfors et al., Haptic palpation of head and neck cancer patients—implications for education and telemedicine, Stud Health Technol Inform 81 (2001), pp. 471–474. View Record in Scopus | Cited By in Scopus (8)
26 C. Myers, Telehealth applications in head and neck oncology, J Speech Lang Pathol Audiol 29 (3) (2005), pp. 125–127.
27 J.L. van den Brink et al., Involving the patient: a prospective study on use, appreciation and effectiveness of an information system in head and neck cancer care, Int J Med Inform 74 (10) (2005), pp. 839–849. Article | | View Record in Scopus | Cited By in Scopus (14)
28 J.L. van den Brink et al., Impact on quality of life of a telemedicine system supporting head and neck cancer patients: a controlled trial during the postoperative period at home, J Am Med Inform Assoc 14 (2) (2007), pp. 198–205. Article | | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)
29 B. Head et al., Development of a telehhealth intervention for head and neck cancer patients, Telemed J E Health 15 (1) (2009), pp. 100–108. View Record in Scopus | Cited By in Scopus (1)
30 D.F. Cella et al., The Functional Assessment of Cancer Therapy (FACT) scale: development and validation of the general measure, J Clin Oncol 11 (3) (1993), pp. 570–579. View Record in Scopus | Cited By in Scopus (1626)
31 M.A. List et al., The Performance Status scale for head and neck cancer patients and the Functional Assessment of Cancer Therapy-Head and Neck scale: A study of utility and validity, Cancer 77 (11) (1996), pp. 2294–2301. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (169)
32 H.M. Mehanna and R.P. Morton, Patients' views on the utility of quality of life questionnaires in head and neck cancer: a randomised trial, Clin Otolaryngol 31 (4) (2006), pp. 310–316. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
33 V. Chang et al., The Memorial Symptom Assessment Scale short form, Cancer 89 (2000), pp. 1162–1171. Full Text via CrossRef
34 R.K. Portenoy et al., The Memorial Symptom Assessment Scale: an instrument for the evaluation of symptom prevalence, characteristics and distress, Eur J Cancer 30A (9) (1994), pp. 1326–1336. Abstract | | View Record in Scopus | Cited By in Scopus (449)
35 J.E. Tranmer et al., Measuring the symptom experience of seriously ill cancer and noncancer hospitalized patients near the end of life with the Memorial Symptom Assessment Scale, J Pain Symptom Manage 25 (5) (2003), pp. 420–429. Article | | View Record in Scopus | Cited By in Scopus (81)
36 V.T. Chang et al., Symptom and quality of life survey of medical oncology patients at a Veterans Affairs medical center: a role for symptom assessment, Cancer 88 (5) (2000), pp. 1175–1183. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (121)
37 J.F. Nelson et al., The symptom burden of chronic critical illness, Crit Care Med 32 (7) (2004), pp. 1527–1534. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (55)
38 L.B. Harrison et al., Detailed quality of life assessment in patients treated with primary radiotherapy for squamous cell cancer of the base of the tongue, Head Neck 19 (3) (1997), pp. 169–175. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (132)
39 J. Sandberg et al., A qualitative study of the experiences and satisfaction of direct telemedicine providers in diabetes case management, Telemed J E Health 15 (8) (2009), pp. 742–750. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1)
Treatment for head and neck cancer is most often a rigorous regimen of combination therapies, producing a multitude of distressing symptoms and side effects. While it is nearly impossible to circumvent the physical and psychosocial insults caused by such treatment, some interventions directed toward educating and supporting patients during active treatment have met with success.[1], [2], [3] and [4] Conversely, other efforts have demonstrated little impact[5] and [6] or have been poorly received,7 pointing to the need for effective, acceptable means to provide support during such difficult treatment.
Over the past 10 years, telemedicine technology has enabled innovative approaches for improving patient education, assessment, support, and communication during treatment for both acute and chronic diseases. A recent policy white paper8 described telemedicine technology as including “the electronic acquisition, processing, dissemination, storage, retrieval, and exchange of information for the purpose of promoting health, preventing disease, treating the sick, managing chronic illness, rehabilitating the disabled, and protecting public health and safety” (p. 2). This same paper suggests that national telemedicine initiatives are essential to health-care reform based upon their proven cost–effectiveness and clinical efficacy. However, cost savings and clinical effectiveness will be unrealized outcomes if the interventions are not feasible in practice or acceptable to the targeted population.
In the arena of cancer care, telephone-based systems have been used to report and monitor cancer symptoms with favorable compliance noted even when patients are expected to initiate calls on a regular basis.[9], [10], [11] and [12] Favorable acceptance ratings have also been reported by both patients and clinicians regarding computerized systems used to assess symptoms and quality of life (QOL) in cancer patients.[13], [14], [15], [16], [17], [18] and [19] In the United Kingdom, a handheld computer system was successfully used to monitor and support patients receiving chemotherapy for lung or colorectal cancer,20 and a study testing a dialogic model of cancer care expecting patients to respond to telehealth messaging on a daily basis over 6 months reported an 84% cooperation rate.21 In these studies, the majority of patients reported ease of use and acceptability of the technology. Survey research has found both urban and rural cancer patients to be receptive to medical and psychiatric services provided via telehealth.22
Published reports describing use of telehealth and computerized interventions during head and neck cancer treatment are less prevalent. Touch-screen computers were successfully used in the Netherlands to collect QOL and distress data from head and neck cancer patients.16 Videoconferencing has been used successfully to overcome geographical barriers to patient assessment[23], [24] and [25] and to provide speech–language pathology services to people living with head and neck cancers in remote areas. Reported use of telehealth management appears promising for providing timely access to care for those who are geographically isolated.26
A research group based in the Netherlands developed and tested a comprehensive electronic health information support system for use in head and neck cancer care.27 The system had four patient-related functions: facilitating communication between patients and health-care providers, providing information about the disease and its treatment, connecting patients with other patients similarly diagnosed, and monitoring patients after hospital discharge. The system was found to be well-accepted and appreciated by participating patients, and its use enabled early identification and direct intervention for patient problems.27 A clinical trial of the telehealth application showed improved QOL in five of 22 studied parameters for the treatment group.28 However, 20 of the 59 patients eligible for the intervention group refused participation; 11 (55%) of these stated computer-related concerns as their reason for nonparticipation.
Knowing that head and neck cancer patients experience a high burden of illness and often have significant communication, socioeconomic, and geographic barriers to care, our team developed a telehealth intervention using a simple telemessaging device to circumvent communication barriers and perceived technical challenges associated with computer-based systems to provide education and support to patients in their own home and on their own time schedule.29 Overall, we hypothesized that patients receiving the intervention would experience less symptom distress, improved QOL, increased self-efficacy, and greater satisfaction with symptom management than those in the control group. However, as a first step toward examining the efficacy and effectiveness of this intervention, this study examined both quantitative and qualitative indicators of its feasibility and acceptance among patients undergoing treatment for head and neck cancer.
Methods
Design
Subsequent to study approval by the University of Louisville's Human Subjects Protection Office, a randomized clinical trial comparing the telehealth intervention to standard care was conducted using a two-group parallel design. This study reports on the intervention's feasibility and acceptance in the treatment group of 44 patients.
Site
Participants were recruited from patients receiving care from the Multidisciplinary Head and Neck Cancer Team at the James Graham Brown Cancer Center (JGBCC) over a 2-year period (June 2006 through June 2008). The team consisted of head and neck surgeons, medical oncologists, radiation oncologists, nurses, a pathologist, a speech therapist, a registered dietician, a psychologist, and a social worker. This team developed a comprehensive assessment and treatment plan during each patient's initial visit to the clinic and coordinated patient care throughout the treatment process.
Sample
Patients eligible for study participation met the following inclusion criteria: (1) initial diagnosis of head or neck cancer including cancers of the oral cavity, salivary glands, paranasal sinuses and nasal cavity, pharynx, and larynx; (2) involvement in a treatment plan including one or more modalities (ie, surgery, chemotherapy, radiation, or any combination); (3) capacity to give independent informed consent; and (4) ability to speak, read, and comprehend English at the eighth-grade level or above. Patients were excluded from participation if they had no land telephone line, had a thought disorder, were incarcerated, or had compromised cognitive functioning.
All patients scheduled for assessment received an explanation of the research study via print materials prior to their first clinic visit. During their first scheduled clinic visit, all patients identified as eligible were approached by a member of the research study staff, who briefly explained the study and asked if they might be interested in study participation. Because of the stress and content of this first clinic visit, interested patients were contacted later by phone to schedule an additional visit to review the study and obtain informed consent.
During the informed consent meeting, the study procedures were explained in detail. If the patient agreed and signed a consent form, a randomization grid which considered the patient's particular treatment plan was used to assign the patient to either the control or the experimental group. Baseline data were also collected during this first visit.
Description of the Intervention
The technology selected for implementing the intervention was the Health Buddy® System, a commercially available, proprietary system produced and maintained by Robert Bosch Healthcare Palo Alto, CA. The Health Buddy, the appliance used for interaction between the participant and the health-care provider, is a user-friendly, easily visible, electrical device that attaches to the user's land phone line (see Figure 1). Questions and information are displayed on the liquid crystal display (LCD) screen of the 6 × 9–inch appliance. The individual responds to questions by pressing one of the four large buttons below the screen. The research team selected the technology provider based on the ability of the technology to perform in accordance with the research objectives.
Symptom control algorithms developed using participatory action research (surveys of current and past patients and clinicians) and evidence-based practice were programmed into the telehealth messaging system (see article by Head et al,29 which details the algorithm topic selection and development process). The algorithms addressed 29 different symptoms and side effects of treatment, consisting of approximately 100 questions accompanied by related educational and supportive responses. Patients were asked three to five questions daily related to the symptoms anticipated during their treatment scenario. Depending upon their response, they would receive specific information related to symptom self-management, including recommendations as to when to contact their clinicians. The algorithms were constructed with the goal of encouraging self-efficacy and independent action on the part of the participant. See Figure 2 for an example of the branching algorithms.
Participants randomly assigned to the treatment group immediately had the Health Buddy connected to a land telephone line in their home. Most (40%) chose to place it in their kitchen, while another 26% placed it in their bedrooms; most often, it was in a highly visible location, serving to remind the participant to respond. Research study staff delivered, installed, and demonstrated how to operate the equipment. Installation was simple and required only minutes. A tutorial programmed into the Health Buddy taught participants how to reply to questions appearing on the monitor using the four large keys below the possible answers or a rating scale which would appear depending on the type of question asked.
During the early hours of the morning, the device would automatically call a toll-free number. Responses to the previous day's questions were uploaded, and questions and related information for the next day were downloaded over the telephone line onto a secure server. Phone service was never disrupted by the device; if the phone was in use, the system would connect later to retrieve and download information. Once new content was transferred, a green light on the device would flash to alert the participant that new questions were available for response. Once the participant pressed any of the keys, the new algorithms would begin appearing on the monitor screen.
Participants were instructed to begin responding on the first day they received treatment or on the first day after returning home from surgery. They were asked to continue responding daily (unless hospitalized for treatment) throughout the treatment period and for approximately 2 weeks posttreatment as treatment-induced symptoms continue during that period of time. Study staff contacted participants when treatment was complete and scheduled a date to pick up the appliance and end daily responding. Daily patient responses required 5–10 minutes.
Participant responses could be viewed by study staff via Internet access 1 day after being answered. Responses were monitored daily by study nurses. Symptoms unrelieved over time or symptoms targeted as requiring immediate intervention (ie, serious consideration of suicide) would result in the study nurse contacting the patient directly by phone and/or contacting clinicians to assure immediate intervention. However, it is important to note that this direct intervention by study staff was infrequent as most symptoms were addressed independently by the participant as desired. If a participant had not reported a period of planned hospitalization and did not respond for 3 consecutive days, study staff would contact the patient by phone to ascertain the reason for noncompliance.
Measures
The following indicators were selected as measures of acceptance (accrual rate), feasibility (utilization, nurse-initiated contacts), and/or satisfaction (satisfaction ratings). Narrative responses and a poststudy survey provided additional data examining acceptance, feasibility, and satisfaction with the intervention. In addition, demographic and medical information as well as measures assessing primary study outcomes were collected from each participant. Table 1 lists all measures and the study time point when they were administered.
MEASURES | PRETREATMENT | DURING TREATMENT | POSTTREATMENT | CUMULATIVE |
---|---|---|---|---|
Demographics | X (baseline) | X | ||
Accrual rate | X | |||
Utilization rate | X | |||
FACT-H&N | X (baseline) | X (mid-tx) | X (2–4 weeks post-tx) | |
MSAS | X (baseline) | X (mid-tx) | X (2–4 weeks post-tx) | |
Satisfaction with technology | X | |||
Nurse-initiated contacts | X | |||
Exit interview | X (end of treatment) | |||
Poststudy written survey | X (60–90 days post-tx) |
tx = treatment
Accrual rate
The number of individuals assessed for study eligibility, reasons for exclusion or noncompletion, and numbers included in the analysis were all recorded to examine acceptance of the intervention and identify issues with the intervention or technology affecting participation.
Utilization
Feasibility was operationalized as device utilization using the percentage of days on which a participant responded to the Health Buddy. This was calculated using the number of days the participant responded to the telehealth device divided by the number of days the participant had the device and was expected to respond. These data were maintained and provided by the telehealth provider (Robert Bosch Healthcare).
Nurse-initiated contacts with participants and/or clinicians
The number of occasions on which a nurse decided to intervene was used as an indicator of feasibility under the premise that the goal of the intervention was to support and encourage patient-driven efforts to seek care for persistent or troubling symptoms. If a patient reported a symptom, he or she was given management information and encouraged to discuss problems further with the clinician either by phone or during clinic visits. If a patient continued to report an unresolved symptom or if the symptom required immediate intervention (ie, suicide threat), the research nurse reviewing responses would contact the patient and/or clinician to ascertain why and/or assist with its resolution. These nurse-initiated contacts should be infrequent if the intervention is achieving the goal of developing patient self-efficacy.
Satisfaction ratings
Items assessing satisfaction with the technology were also administered to participants via the telehealth messaging device. Questions related to satisfaction with the initial setup of the telehealth appliance were asked at the beginning of the intervention. Ongoing satisfaction with the device, messaging content, and the health-care provider were assessed every 90 days. The specific questions asked are detailed in Table 4.
Narrative data
Upon completion of the intervention, participants in the treatment group completed an exit interview using open-ended questions regarding the utility of the intervention, relevance of the algorithms, value or burden of item repetition in the algorithms, symptoms or problems experienced that were not addressed by the intervention, and general comments.
Poststudy survey
A final survey was mailed to participants several months after completion of the study, asking for additional feedback about the impact of the intervention. Specifically, participants (both treatment and control groups) were asked about their overall satisfaction with the treatment and services at the cancer center, their satisfaction with information received about their treatment, the response(s) received when they attempted contact with the health-care team after hours, the amount of support received, their current smoking and alcohol usage, and several demographic questions not earlier assessed or available through record review (years of education, highest degree, income range). Those receiving the intervention were also asked about the impact of the Health Buddy on their care and actions taken in response to the algorithms.
Demographic and medical information
Demographic information was collected using the initial survey, and information about the participant's medical history, condition, treatments received, treatment timing, complications, comorbidities, and treatment response was collected via retrospective medical record review subsequent to completion of the clinical trial.
Outcome measures
While outcomes of the clinical trial are not the subject of this article, the results of QOL and symptom burden measures for the treatment group only are included here because of their relationship with device utilization. The two measures included the Functional Assessment of Cancer Therapy–Head and Neck Scale and the Memorial Symptom Assessment Scale and were administered at baseline (before beginning treatment), mid-treatment, and posttreatment.
• Functional Assessment of Cancer Therapy–Head and Neck Scale (FACT-H&N). The FACT-G (general) is a multidimensional QOL instrument designed for use with all cancer patients. The instrument has 28 items divided into four subscales: Functional Well-Being, Physical Well-Being, Social Well-Being, and Emotional Well-Being. This generic core questionnaire was found to meet or exceed requirements for use in oncology based upon ease of administration, brevity, reliability, validity, and responsiveness to clinical change.30 Added to the core questionnaire is the head and neck–specific subscale, consisting of 11 items specific to this cancer site. A Trial Outcome Index (TOI) is also scored and is the result of the physical, functional, and cancer-specific subscales. List et al31 found the FACT-H&N to be reliable and sensitive to differences in functioning for patients with head and neck cancers (Cronbach's alpha was 0.89 for total FACT-G and 0.63 for the head and neck subscale in this study of 151 patients). Additionally, head and neck cancer patients found the FACT-H&N relevant to their problems and easy to understand, and it was preferred over other validated head and neck cancer QOL questionnaires.32 The FACT-H&N was chosen for this study because it (1) is nonspecific related to a treatment modality or subsite among head and neck cancers, (2) allows comparison across cancer diagnoses while still probing issues specific to head and neck cancer, (3) is short and can be completed quickly, (4) includes the psychosocial domains of social/family and emotion subscales as well as physical and functional areas, and (5) is self-administered.
• Memorial Symptom Assessment Scale (MSAS). This multidimensional scale measures the prevalence, severity, and distress associated with the most common symptoms experienced by cancer patients. Physical and emotional subscale scores as well as a Global Distress Index (GDI, considered to be a measure of total symptom burden) can be generated from patient responses. The MSAS has demonstrated validity and reliability in both in- and outpatient cancer populations.[33], [34] and [35] Initial psychometric analysis by Portenoy et al34 used factor analysis to define two subscales: psychological symptoms and physical symptoms with Cronbach alpha coefficients of 0.88 and 0.83, respectively; convergent validity was also established. It was chosen for this study because of its proven ability to measure both the presence and the intensity of experienced symptoms.[33], [35], [36], [37] and [38]
Data Analysis
Quantitative data were documented and analyzed using the Statistical Package for the Social Sciences (SPSS, Inc., Chicago, IL), version 16. Descriptive statistics were calculated to describe the sample and assess study outcomes, including feasibility and acceptability of the intervention. To ascertain relationships between usage of the device and demographic and medical information, a series of correlational analyses using Spearman's rho were conducted. This nonparametric test was chosen over Pearson's r because of the small sample size, the lack of a normal distribution for several of the variables, and the ordinal nature of several of the variables. Multiple regression analyses were also planned, but lack of significant bivariate correlations precluded multivariate analysis.
Qualitative responses to open-ended questions were analyzed to identify themes and direct quotations illustrating those themes.
Descriptive analysis of the treatment group's responses to the outcome measures (QOL and symptom burden) was done to ascertain changes over the course of the intervention using the mean scores at baseline, during treatment, and posttreatment.
Results
Description of Participants
Participants randomly assigned to the intervention group (n = 45) were an average age of 59 years (±11.7), and most were covered by private (34%) or public (48%) insurance. On average, participants had completed 13.5 years (±3.0) of formal education. Thirty-nine (87%) of the participants were male and 91% were Caucasian.
With regard to medical information, participants were predominantly diagnosed with stage II cancers of the head and neck (36%). The most prevalent site was the larynx (12 patients), followed by the tongue and the base of the tongue (seven patients) and unknown primary (seven patients). The vast majority received chemotherapy (32, or 71%) and/or radiation (42, or 93%).
Additional details regarding demographic and medical characteristics of the sample are provided in Table 2.
FREQUENCY | VALID PERCENT | |
---|---|---|
Gender (n = 44) | ||
Male | 39 | 88.6 |
Female | 5 | 11.3 |
Race (n = 44) | ||
Caucasian | 40 | 90.9 |
African American | 4 | 9.0 |
Tumor stage (n = 44) | ||
I | 7 | 15.9 |
II | 15 | 34.0 |
III | 11 | 25.0 |
IV | 4 | 9.0 |
Unable to determine | 5 | 11.4 |
Unknown | 2 | 4.5 |
Site of cancer (n = 44) | ||
Larynx | 12 | 27.2 |
Tongue, base of tongue | 7 | 15.9 |
Unknown primary | 7 | 15.9 |
Tonsillar | 4 | 9.0 |
Other H&N sites | 14 | 31.8 |
Insurance status (n = 44) | ||
No insurance | 8 | 18.2 |
Medicaid | 1 | 2.3 |
Medicare | 2 | 4.5 |
Medicaid and Medicare | 1 | 2.3 |
Medicare and supplement | 9 | 20.5 |
Medicare and VA benefits | 2 | 4.5 |
Veteran benefits only | 6 | 13.6 |
Private insurance | 15 | 34.1 |
Highest educational degree (n = 20)a | ||
Less than high school | 3 | 15.0 |
High school or GED | 9 | 45.0 |
Associate's/bachelor's degree | 4 | 20.0 |
Masters, PhD, or MD | 2 | 10.0 |
Other | 2 | 10.0 |
Income range (n = 18)a | ||
$20,000 or less | 5 | 27.8 |
$20,001–50,000 | 5 | 27.8 |
$70,001–100,000 | 5 | 27.8 |
Over $100,000 | 3 | 16.7 |
Percent of poverty in zip code area (n = 44) | ||
2.8–5.1% | 11 | 25.0 |
5.9–8.6% | 11 | 25.0 |
9.0–11.9% | 10 | 22.7 |
12.3–45.9% | 12 | 27.2 |
Feasibility and Acceptability
Accrual rate
Of the 185 patients assessed for eligibility during the 2-year recruitment period, 105 were excluded. See Figure 3 for a detailed depiction of study accrual for both the treatment and control groups. Thirty-three (31%) were excluded because they did not have a land phone line, a requirement for transmitting the algorithms to the Health Buddy appliance. Most of these had cell phones only. No potential participants refused participation due to issues related to operation of the technology itself.
Device utilization
Participants used the telehealth device for an average of 70.7 days (±26.7), which constituted 86.3% (±15.0) of the total days available for use. Of note, the median percentage of use was 94.2% and the modal percentage was 100%, indicating that the vast majority of participants consistently used the telehealth device. The participant with the lowest usage rate used the device 46% of the days available.
By far, the most common reason for Health Buddy nonresponse was patient hospitalization. Two subjects traveled out of town frequently on weekends and would leave the Health Buddy at home. One subject had accidentally unhooked the Health Buddy, and a home visit was made to reconnect the device into the patient's phone line.
Nurse-initiated contacts with participants and/or clinicians
Of the 45 enrolled patients, 33 required additional contact with a research nurse (see Table 3). The most common reasons patients were contacted were nonresponse for 3 consecutive days (38.3%), repeated reporting of high levels of unrelieved pain (30%), and suicidal thoughts (10%). In all, 120 calls were placed: one call for every 25.9 response days. In every case, the problem was resolved.
NUMBER OF PATIENTS | PROBLEM | OUTGOING CALLS | RESOLUTION |
---|---|---|---|
15 | No response on Health Buddy for 3 consecutive days | 46 | Patient teaching |
17 | Pain-related issues | 36 | Advocacy/referral/patient teaching |
5 | Suicidal thoughts | 12 | Advocacy/referral |
7 | G-tube problems | 8 | Patient teaching |
5 | Sadness/depression | 6 | Advocacy/referral |
3 | Multiple symptoms | 3 | Advocacy/referral |
3 | Nausea/vomiting | 4 | Referral/patient teaching |
2 | Coughing/excessive secretions | 2 | Patient teaching |
2 | Constipation | 2 | Patient teaching |
1 | Stomatitis | 1 | Referral |
Satisfaction ratings
Responses to surveys programmed into the Health Buddy system are displayed in Table 4. Overall, respondents responded favorably, finding the installation to be easy, the content to be helpful, and the overall experience to be positive.
PERCENT OF RESPONDENTS | |
---|---|
Installation satisfaction | |
Installation problems? | |
Yes | 2 |
No | 98 |
Any difficulty completing the first training questions? | |
Yes | 7 |
No | 94 |
Length of installation? | |
2–5 minutes | 52 |
6–10 minutes | 41 |
11–15 minutes | 4 |
16–20 minutes | 2 |
Content satisfaction | |
Overall, I think the Health Buddy questions are | |
Very easy | 44 |
Somewhat easy | 16 |
Neutral | 32 |
Somewhat difficult | 4 |
Difficult | 4 |
Repeating questions reinforced knowledge and understanding | |
Strongly agree | 56 |
Somewhat agree | 28 |
Neutral | 12 |
Somewhat disagree | 4 |
Strongly disagree | 0 |
Understanding of my health condition | |
Much better | 64 |
Somewhat better | 20 |
Neutral | 16 |
Somewhat worse | 0 |
Much worse | 0 |
Managing my health condition | |
Much better | 52 |
Somewhat better | 44 |
Neutral | 4 |
Somewhat worse | 0 |
Much worse | 0 |
Recommend the device to others | |
Very willing | 80 |
Somewhat willing | 12 |
Neutral | 4 |
Somewhat unwilling | 0 |
Very unwilling | 4 |
Overall satisfaction | |
Satisfaction with device | |
Very satisfied | 45 |
Satisfied | 35 |
Somewhat satisfied | 15 |
Not very satisfied | 5 |
Satisfaction with the communication between you and your doctor or nurse | |
More satisfied | 65 |
No difference | 30 |
Less satisfied | 5 |
Ease of using the device | |
Very easy | 85 |
Easy | 15 |
Not easy | 0 |
Overall experience with the device | |
Positive | 85 |
Neutral | 15 |
Negative | 0 |
Continue to use the device | |
Very likely | 40 |
Likely | 40 |
Somewhat likely | 15 |
Not very likely | 0 |
Narrative comments
During the exit interview, participants were asked, “How was having the Health Buddy helpful to you?” Responses could be categorized into two major themes: (1) the Health Buddy provided needed information and (2) the Health Buddy improved my self-management during treatment.
Statements made related to the information provided included the following:
- • It gave me information on what could be expected from treatment
• It was a constant reminder of things to watch for
• It kept me abreast of my total condition at all times
• It gave good directions so I didn't have to ask at the cancer center
• It gave good suggestions on treatments (home remedies) such as gargles, care of feeding tube, exhaustion, and everyday symptoms
Statements made indicative that the Health Buddy improved self-management included the following:
- • I learned what I could do to make myself feel better
• It helped me manage my symptoms
• It taught me about symptom management and how to handle problems
• It let me know whether to contact a doctor or use self-care
• It gave me who to call for problems and some things to try
• It kept me aware of what I needed to do in order to make the period easier
• It reminded me to take my meds and exercise
Additionally, some participants noted the support they felt from having the Health Buddy interventions during treatment in saying the following:
- • It kind of helped my depression through acknowledging it and giving me something to do
• It made me feel I was not the only one who had experience with these things
• It comforted me because I knew what was going to happen
Poststudy survey
Twenty (45%) of the 44 patients who received the intervention responded to the mailed poststudy survey. When asked if they felt they received better care because they had the device, 13 of the 20 (65%) responded that they did. Eighteen (90%) of the treatment group responders stated they were very satisfied with their care (one stated “somewhat satisfied”) and 20 (100%) said they would recommend the cancer center for treatment. Nineteen (95%) stated they received adequate support during treatment.
Outcome Measures
Mean scores on the FACT-H&N and subscales and the MSAS and subscales taken pre-, during, and posttreatment are displayed in Table 5. As expected, average QOL scores declined during treatment, while symptom distress increased, with a return to near baseline scores posttreatment.
SCALE/SUBSCALE | PRETREATMENT | DURING TREATMENT | POSTTREATMENT |
---|---|---|---|
Total FACT-H&N | 100.3 | 85.6 | 101.5 |
FACT-G | 74.3 | 69.4 | 78.5 |
Trial Outcome Index | 62.6 | 46.0 | 65.0 |
Physical Well-Being | 21.2 | 17.6 | 21.1 |
Functional Well-Being | 15.6 | 12.5 | 17.4 |
Emotional Well-Being | 21.1 | 22.3 | 22.2 |
Social Well-Being | 21.1 | 22.3 | 22.2 |
Total MSAS | 0.7 | 1.1 | 0.8 |
Global Distress Index | 1.1 | 1.8 | 1.3 |
Physical | 0.7 | 1.5 | 1.1 |
Psychological | 1.1 | 1.2 | 0.8 |
Correlations
The relationships between percentage usage per patient and the following variables were evaluated: age, income, years of education, tumor stage, and percent poverty in patient's zip code. Percent poverty in zip code area was intended to be a surrogate measure of the patient's socioeconomic status. Results are displayed in Table 6. No significant correlations were noted, although years of education and percentage poverty in zip code showed a trend toward significance.
VARIABLE (VS % USAGE) | RELATIONSHIP | |
---|---|---|
SPEARMAN'S RHO RS | SIGNIFICANCE (ONE-TAILED) | |
Percent poverty in zip code | 0.213 | 0.083 |
Age | 0.146 | 0.173 |
Years of education | −0.325 | 0.081 |
Income | −0.292 | 0.120 |
Tumor stage | 0.196 | 0.122 |
Physical Well-Being (during treatment) | 0.310 | 0.048 |
Emotional Well-Being (during treatment) | 0.315 | 0.042 |
Although a multivariate model was planned, the lack of significant bivariate correlations precluded the need for multivariate analysis.
When percent usage was correlated with FACT-H&N total and subscales taken at baseline, during active treatment, and posttreatment, significant positive correlations were found between the percentage used and the Physical Well-Being subscale score during treatment (Spearman's rho = 0.310, P = 0.048) and between percentage used and the Emotional Well-Being subscale during treatment (Spearman's rho = 0.315, P = 0.042).
There were no significant correlations between percentage usage and the scores on the MSAS.
Discussion
Both qualitative and quantitative measures indicate that using telehealth to support symptom management during aggressive cancer treatment is both feasible and well-accepted. Patient users were not intimidated by this particular technology as it was simple to set up and use and required no previous computer training to operate. The Health Buddy was viewed as providing important and useful information. Overall, users felt that it improved their ability to self-manage their disease and the side effects of treatment and provided a sense of support and security.
Unlike other studies which use telehealth devices to monitor patient symptoms, our goal was to increase patient self-management of the symptoms experienced during intensive medical treatment, therefore avoiding increased burden on the medical system. The fact that the research nurse overseeing the responses needed to intervene only once every 25.9 days speaks to the ability of the intervention to have a positive impact on utilization of medical services.
The lack of significant relationships between usage and descriptive variables such as age and years of education suggests that the intervention was equally acceptable to all subgroups. Factors such as age, previous computer literacy, educational obtainment, and socioeconomic status did not significantly differentiate our study population in terms of compliance as verified by usage percentages.
The significant relationships found between the percentage used and the subscale scores on Physical Well-Being and Emotional Well-Being during treatment may indicate that increased use of the telemessaging intervention during treatment resulted in better physical and emotional aspects of QOL.
The high rate of daily compliance with the intervention in spite of differentiating personal variables and the severity of the treatment regimen may have been due to one or a combination of the following factors:
- • the simplicity of the technology
• the visibility of the appliance (often placed in the kitchen or living area of the home) and its flashing green light as cues to the need to respond
• the usefulness of the information provided
• the use of simple messaging language presented in an encouraging, positive manner
• affirmations related to application of the symptom management protocols suggested
• curiosity related to the day's messaging and the motivational saying which always appeared at the end
• knowledge that someone was reviewing the responses, tracking and intervening when the participant did not respond for several days
Our study supports the benefits of telehealth interventions noted by providers in a study by Sandberg et al39: opportunities for more frequent contact, greater relaxation and information due to the ability to interact in one's own home, increased accessibility by those frequently underserved, and timely medical information and monitoring. Similar to the study done in the Netherlands,[27] and [28] this study noted technological problems as the primary disadvantage; but in our intervention, we had no problems with the technology or equipment.
Although computerized technology served as a barrier to previous telehealth research, the lack of a land-based phone line was a factor preventing participation in the current study. Indeed, many participants maintained only wireless communication devices, which were not compatible with the version of the Health Buddy that was employed in this study. However, improvements in the technology since completion of this study now allow for wireless access to the appliance or provision of an independent wireless messaging device for those without such access in their own homes.
Although the data generally support the feasibility and acceptability of the telehealth-based intervention, the results should be interpreted in the context of a few study limitations. In particular, the sample size was somewhat small, and data pertaining to the socioeconomic status of participants were not available for all participants. Second, the study did not include measures of the patient's direct interactions with health-care providers during the study or specific data related to their health-care utilization (eg, emergency room visits, preventable inpatient hospitalizations, emergency calls to clinicians). The collection of more exhaustive measures of health-care utilization was limited by resources but is planned for subsequent studies. Finally, concerns regarding subject burden limited assessment of the usability of the telehealth device.
Although compliance with utilization expectations and completion of study measures was excellent during the course of the intervention, response to the follow-up survey mailed several months later was less than 50%. This low response rate was most likely due to several factors: (1) this survey was sent at the conclusion of the entire study (by this time, patients were 0–21 months past their active participation); (2) it was a mailed survey with no additional contact or follow-up effort to increase response rate; (3) participants may have felt that they had already shared their opinions in the exit interview and may have felt overburdened by study measures at this point; and (4) participants may have died, moved, or been medically unable to respond. This lack of response did limit our ability to evaluate the longitudinal impact of the intervention.
Conclusions
This telehealth intervention proved to be an acceptable and feasible means to educate and support patients during aggressive treatment for head and neck cancer. Patient compliance with telehealth interventions during periods of extreme symptom burden and declining QOL is feasible if simple technology cues the patient to participate, offers positive support and relevant education, and is targeted or tailored to their specific condition.
Treatment for head and neck cancer is most often a rigorous regimen of combination therapies, producing a multitude of distressing symptoms and side effects. While it is nearly impossible to circumvent the physical and psychosocial insults caused by such treatment, some interventions directed toward educating and supporting patients during active treatment have met with success.[1], [2], [3] and [4] Conversely, other efforts have demonstrated little impact[5] and [6] or have been poorly received,7 pointing to the need for effective, acceptable means to provide support during such difficult treatment.
Over the past 10 years, telemedicine technology has enabled innovative approaches for improving patient education, assessment, support, and communication during treatment for both acute and chronic diseases. A recent policy white paper8 described telemedicine technology as including “the electronic acquisition, processing, dissemination, storage, retrieval, and exchange of information for the purpose of promoting health, preventing disease, treating the sick, managing chronic illness, rehabilitating the disabled, and protecting public health and safety” (p. 2). This same paper suggests that national telemedicine initiatives are essential to health-care reform based upon their proven cost–effectiveness and clinical efficacy. However, cost savings and clinical effectiveness will be unrealized outcomes if the interventions are not feasible in practice or acceptable to the targeted population.
In the arena of cancer care, telephone-based systems have been used to report and monitor cancer symptoms with favorable compliance noted even when patients are expected to initiate calls on a regular basis.[9], [10], [11] and [12] Favorable acceptance ratings have also been reported by both patients and clinicians regarding computerized systems used to assess symptoms and quality of life (QOL) in cancer patients.[13], [14], [15], [16], [17], [18] and [19] In the United Kingdom, a handheld computer system was successfully used to monitor and support patients receiving chemotherapy for lung or colorectal cancer,20 and a study testing a dialogic model of cancer care expecting patients to respond to telehealth messaging on a daily basis over 6 months reported an 84% cooperation rate.21 In these studies, the majority of patients reported ease of use and acceptability of the technology. Survey research has found both urban and rural cancer patients to be receptive to medical and psychiatric services provided via telehealth.22
Published reports describing use of telehealth and computerized interventions during head and neck cancer treatment are less prevalent. Touch-screen computers were successfully used in the Netherlands to collect QOL and distress data from head and neck cancer patients.16 Videoconferencing has been used successfully to overcome geographical barriers to patient assessment[23], [24] and [25] and to provide speech–language pathology services to people living with head and neck cancers in remote areas. Reported use of telehealth management appears promising for providing timely access to care for those who are geographically isolated.26
A research group based in the Netherlands developed and tested a comprehensive electronic health information support system for use in head and neck cancer care.27 The system had four patient-related functions: facilitating communication between patients and health-care providers, providing information about the disease and its treatment, connecting patients with other patients similarly diagnosed, and monitoring patients after hospital discharge. The system was found to be well-accepted and appreciated by participating patients, and its use enabled early identification and direct intervention for patient problems.27 A clinical trial of the telehealth application showed improved QOL in five of 22 studied parameters for the treatment group.28 However, 20 of the 59 patients eligible for the intervention group refused participation; 11 (55%) of these stated computer-related concerns as their reason for nonparticipation.
Knowing that head and neck cancer patients experience a high burden of illness and often have significant communication, socioeconomic, and geographic barriers to care, our team developed a telehealth intervention using a simple telemessaging device to circumvent communication barriers and perceived technical challenges associated with computer-based systems to provide education and support to patients in their own home and on their own time schedule.29 Overall, we hypothesized that patients receiving the intervention would experience less symptom distress, improved QOL, increased self-efficacy, and greater satisfaction with symptom management than those in the control group. However, as a first step toward examining the efficacy and effectiveness of this intervention, this study examined both quantitative and qualitative indicators of its feasibility and acceptance among patients undergoing treatment for head and neck cancer.
Methods
Design
Subsequent to study approval by the University of Louisville's Human Subjects Protection Office, a randomized clinical trial comparing the telehealth intervention to standard care was conducted using a two-group parallel design. This study reports on the intervention's feasibility and acceptance in the treatment group of 44 patients.
Site
Participants were recruited from patients receiving care from the Multidisciplinary Head and Neck Cancer Team at the James Graham Brown Cancer Center (JGBCC) over a 2-year period (June 2006 through June 2008). The team consisted of head and neck surgeons, medical oncologists, radiation oncologists, nurses, a pathologist, a speech therapist, a registered dietician, a psychologist, and a social worker. This team developed a comprehensive assessment and treatment plan during each patient's initial visit to the clinic and coordinated patient care throughout the treatment process.
Sample
Patients eligible for study participation met the following inclusion criteria: (1) initial diagnosis of head or neck cancer including cancers of the oral cavity, salivary glands, paranasal sinuses and nasal cavity, pharynx, and larynx; (2) involvement in a treatment plan including one or more modalities (ie, surgery, chemotherapy, radiation, or any combination); (3) capacity to give independent informed consent; and (4) ability to speak, read, and comprehend English at the eighth-grade level or above. Patients were excluded from participation if they had no land telephone line, had a thought disorder, were incarcerated, or had compromised cognitive functioning.
All patients scheduled for assessment received an explanation of the research study via print materials prior to their first clinic visit. During their first scheduled clinic visit, all patients identified as eligible were approached by a member of the research study staff, who briefly explained the study and asked if they might be interested in study participation. Because of the stress and content of this first clinic visit, interested patients were contacted later by phone to schedule an additional visit to review the study and obtain informed consent.
During the informed consent meeting, the study procedures were explained in detail. If the patient agreed and signed a consent form, a randomization grid which considered the patient's particular treatment plan was used to assign the patient to either the control or the experimental group. Baseline data were also collected during this first visit.
Description of the Intervention
The technology selected for implementing the intervention was the Health Buddy® System, a commercially available, proprietary system produced and maintained by Robert Bosch Healthcare Palo Alto, CA. The Health Buddy, the appliance used for interaction between the participant and the health-care provider, is a user-friendly, easily visible, electrical device that attaches to the user's land phone line (see Figure 1). Questions and information are displayed on the liquid crystal display (LCD) screen of the 6 × 9–inch appliance. The individual responds to questions by pressing one of the four large buttons below the screen. The research team selected the technology provider based on the ability of the technology to perform in accordance with the research objectives.
Symptom control algorithms developed using participatory action research (surveys of current and past patients and clinicians) and evidence-based practice were programmed into the telehealth messaging system (see article by Head et al,29 which details the algorithm topic selection and development process). The algorithms addressed 29 different symptoms and side effects of treatment, consisting of approximately 100 questions accompanied by related educational and supportive responses. Patients were asked three to five questions daily related to the symptoms anticipated during their treatment scenario. Depending upon their response, they would receive specific information related to symptom self-management, including recommendations as to when to contact their clinicians. The algorithms were constructed with the goal of encouraging self-efficacy and independent action on the part of the participant. See Figure 2 for an example of the branching algorithms.
Participants randomly assigned to the treatment group immediately had the Health Buddy connected to a land telephone line in their home. Most (40%) chose to place it in their kitchen, while another 26% placed it in their bedrooms; most often, it was in a highly visible location, serving to remind the participant to respond. Research study staff delivered, installed, and demonstrated how to operate the equipment. Installation was simple and required only minutes. A tutorial programmed into the Health Buddy taught participants how to reply to questions appearing on the monitor using the four large keys below the possible answers or a rating scale which would appear depending on the type of question asked.
During the early hours of the morning, the device would automatically call a toll-free number. Responses to the previous day's questions were uploaded, and questions and related information for the next day were downloaded over the telephone line onto a secure server. Phone service was never disrupted by the device; if the phone was in use, the system would connect later to retrieve and download information. Once new content was transferred, a green light on the device would flash to alert the participant that new questions were available for response. Once the participant pressed any of the keys, the new algorithms would begin appearing on the monitor screen.
Participants were instructed to begin responding on the first day they received treatment or on the first day after returning home from surgery. They were asked to continue responding daily (unless hospitalized for treatment) throughout the treatment period and for approximately 2 weeks posttreatment as treatment-induced symptoms continue during that period of time. Study staff contacted participants when treatment was complete and scheduled a date to pick up the appliance and end daily responding. Daily patient responses required 5–10 minutes.
Participant responses could be viewed by study staff via Internet access 1 day after being answered. Responses were monitored daily by study nurses. Symptoms unrelieved over time or symptoms targeted as requiring immediate intervention (ie, serious consideration of suicide) would result in the study nurse contacting the patient directly by phone and/or contacting clinicians to assure immediate intervention. However, it is important to note that this direct intervention by study staff was infrequent as most symptoms were addressed independently by the participant as desired. If a participant had not reported a period of planned hospitalization and did not respond for 3 consecutive days, study staff would contact the patient by phone to ascertain the reason for noncompliance.
Measures
The following indicators were selected as measures of acceptance (accrual rate), feasibility (utilization, nurse-initiated contacts), and/or satisfaction (satisfaction ratings). Narrative responses and a poststudy survey provided additional data examining acceptance, feasibility, and satisfaction with the intervention. In addition, demographic and medical information as well as measures assessing primary study outcomes were collected from each participant. Table 1 lists all measures and the study time point when they were administered.
MEASURES | PRETREATMENT | DURING TREATMENT | POSTTREATMENT | CUMULATIVE |
---|---|---|---|---|
Demographics | X (baseline) | X | ||
Accrual rate | X | |||
Utilization rate | X | |||
FACT-H&N | X (baseline) | X (mid-tx) | X (2–4 weeks post-tx) | |
MSAS | X (baseline) | X (mid-tx) | X (2–4 weeks post-tx) | |
Satisfaction with technology | X | |||
Nurse-initiated contacts | X | |||
Exit interview | X (end of treatment) | |||
Poststudy written survey | X (60–90 days post-tx) |
tx = treatment
Accrual rate
The number of individuals assessed for study eligibility, reasons for exclusion or noncompletion, and numbers included in the analysis were all recorded to examine acceptance of the intervention and identify issues with the intervention or technology affecting participation.
Utilization
Feasibility was operationalized as device utilization using the percentage of days on which a participant responded to the Health Buddy. This was calculated using the number of days the participant responded to the telehealth device divided by the number of days the participant had the device and was expected to respond. These data were maintained and provided by the telehealth provider (Robert Bosch Healthcare).
Nurse-initiated contacts with participants and/or clinicians
The number of occasions on which a nurse decided to intervene was used as an indicator of feasibility under the premise that the goal of the intervention was to support and encourage patient-driven efforts to seek care for persistent or troubling symptoms. If a patient reported a symptom, he or she was given management information and encouraged to discuss problems further with the clinician either by phone or during clinic visits. If a patient continued to report an unresolved symptom or if the symptom required immediate intervention (ie, suicide threat), the research nurse reviewing responses would contact the patient and/or clinician to ascertain why and/or assist with its resolution. These nurse-initiated contacts should be infrequent if the intervention is achieving the goal of developing patient self-efficacy.
Satisfaction ratings
Items assessing satisfaction with the technology were also administered to participants via the telehealth messaging device. Questions related to satisfaction with the initial setup of the telehealth appliance were asked at the beginning of the intervention. Ongoing satisfaction with the device, messaging content, and the health-care provider were assessed every 90 days. The specific questions asked are detailed in Table 4.
Narrative data
Upon completion of the intervention, participants in the treatment group completed an exit interview using open-ended questions regarding the utility of the intervention, relevance of the algorithms, value or burden of item repetition in the algorithms, symptoms or problems experienced that were not addressed by the intervention, and general comments.
Poststudy survey
A final survey was mailed to participants several months after completion of the study, asking for additional feedback about the impact of the intervention. Specifically, participants (both treatment and control groups) were asked about their overall satisfaction with the treatment and services at the cancer center, their satisfaction with information received about their treatment, the response(s) received when they attempted contact with the health-care team after hours, the amount of support received, their current smoking and alcohol usage, and several demographic questions not earlier assessed or available through record review (years of education, highest degree, income range). Those receiving the intervention were also asked about the impact of the Health Buddy on their care and actions taken in response to the algorithms.
Demographic and medical information
Demographic information was collected using the initial survey, and information about the participant's medical history, condition, treatments received, treatment timing, complications, comorbidities, and treatment response was collected via retrospective medical record review subsequent to completion of the clinical trial.
Outcome measures
While outcomes of the clinical trial are not the subject of this article, the results of QOL and symptom burden measures for the treatment group only are included here because of their relationship with device utilization. The two measures included the Functional Assessment of Cancer Therapy–Head and Neck Scale and the Memorial Symptom Assessment Scale and were administered at baseline (before beginning treatment), mid-treatment, and posttreatment.
• Functional Assessment of Cancer Therapy–Head and Neck Scale (FACT-H&N). The FACT-G (general) is a multidimensional QOL instrument designed for use with all cancer patients. The instrument has 28 items divided into four subscales: Functional Well-Being, Physical Well-Being, Social Well-Being, and Emotional Well-Being. This generic core questionnaire was found to meet or exceed requirements for use in oncology based upon ease of administration, brevity, reliability, validity, and responsiveness to clinical change.30 Added to the core questionnaire is the head and neck–specific subscale, consisting of 11 items specific to this cancer site. A Trial Outcome Index (TOI) is also scored and is the result of the physical, functional, and cancer-specific subscales. List et al31 found the FACT-H&N to be reliable and sensitive to differences in functioning for patients with head and neck cancers (Cronbach's alpha was 0.89 for total FACT-G and 0.63 for the head and neck subscale in this study of 151 patients). Additionally, head and neck cancer patients found the FACT-H&N relevant to their problems and easy to understand, and it was preferred over other validated head and neck cancer QOL questionnaires.32 The FACT-H&N was chosen for this study because it (1) is nonspecific related to a treatment modality or subsite among head and neck cancers, (2) allows comparison across cancer diagnoses while still probing issues specific to head and neck cancer, (3) is short and can be completed quickly, (4) includes the psychosocial domains of social/family and emotion subscales as well as physical and functional areas, and (5) is self-administered.
• Memorial Symptom Assessment Scale (MSAS). This multidimensional scale measures the prevalence, severity, and distress associated with the most common symptoms experienced by cancer patients. Physical and emotional subscale scores as well as a Global Distress Index (GDI, considered to be a measure of total symptom burden) can be generated from patient responses. The MSAS has demonstrated validity and reliability in both in- and outpatient cancer populations.[33], [34] and [35] Initial psychometric analysis by Portenoy et al34 used factor analysis to define two subscales: psychological symptoms and physical symptoms with Cronbach alpha coefficients of 0.88 and 0.83, respectively; convergent validity was also established. It was chosen for this study because of its proven ability to measure both the presence and the intensity of experienced symptoms.[33], [35], [36], [37] and [38]
Data Analysis
Quantitative data were documented and analyzed using the Statistical Package for the Social Sciences (SPSS, Inc., Chicago, IL), version 16. Descriptive statistics were calculated to describe the sample and assess study outcomes, including feasibility and acceptability of the intervention. To ascertain relationships between usage of the device and demographic and medical information, a series of correlational analyses using Spearman's rho were conducted. This nonparametric test was chosen over Pearson's r because of the small sample size, the lack of a normal distribution for several of the variables, and the ordinal nature of several of the variables. Multiple regression analyses were also planned, but lack of significant bivariate correlations precluded multivariate analysis.
Qualitative responses to open-ended questions were analyzed to identify themes and direct quotations illustrating those themes.
Descriptive analysis of the treatment group's responses to the outcome measures (QOL and symptom burden) was done to ascertain changes over the course of the intervention using the mean scores at baseline, during treatment, and posttreatment.
Results
Description of Participants
Participants randomly assigned to the intervention group (n = 45) were an average age of 59 years (±11.7), and most were covered by private (34%) or public (48%) insurance. On average, participants had completed 13.5 years (±3.0) of formal education. Thirty-nine (87%) of the participants were male and 91% were Caucasian.
With regard to medical information, participants were predominantly diagnosed with stage II cancers of the head and neck (36%). The most prevalent site was the larynx (12 patients), followed by the tongue and the base of the tongue (seven patients) and unknown primary (seven patients). The vast majority received chemotherapy (32, or 71%) and/or radiation (42, or 93%).
Additional details regarding demographic and medical characteristics of the sample are provided in Table 2.
FREQUENCY | VALID PERCENT | |
---|---|---|
Gender (n = 44) | ||
Male | 39 | 88.6 |
Female | 5 | 11.3 |
Race (n = 44) | ||
Caucasian | 40 | 90.9 |
African American | 4 | 9.0 |
Tumor stage (n = 44) | ||
I | 7 | 15.9 |
II | 15 | 34.0 |
III | 11 | 25.0 |
IV | 4 | 9.0 |
Unable to determine | 5 | 11.4 |
Unknown | 2 | 4.5 |
Site of cancer (n = 44) | ||
Larynx | 12 | 27.2 |
Tongue, base of tongue | 7 | 15.9 |
Unknown primary | 7 | 15.9 |
Tonsillar | 4 | 9.0 |
Other H&N sites | 14 | 31.8 |
Insurance status (n = 44) | ||
No insurance | 8 | 18.2 |
Medicaid | 1 | 2.3 |
Medicare | 2 | 4.5 |
Medicaid and Medicare | 1 | 2.3 |
Medicare and supplement | 9 | 20.5 |
Medicare and VA benefits | 2 | 4.5 |
Veteran benefits only | 6 | 13.6 |
Private insurance | 15 | 34.1 |
Highest educational degree (n = 20)a | ||
Less than high school | 3 | 15.0 |
High school or GED | 9 | 45.0 |
Associate's/bachelor's degree | 4 | 20.0 |
Masters, PhD, or MD | 2 | 10.0 |
Other | 2 | 10.0 |
Income range (n = 18)a | ||
$20,000 or less | 5 | 27.8 |
$20,001–50,000 | 5 | 27.8 |
$70,001–100,000 | 5 | 27.8 |
Over $100,000 | 3 | 16.7 |
Percent of poverty in zip code area (n = 44) | ||
2.8–5.1% | 11 | 25.0 |
5.9–8.6% | 11 | 25.0 |
9.0–11.9% | 10 | 22.7 |
12.3–45.9% | 12 | 27.2 |
Feasibility and Acceptability
Accrual rate
Of the 185 patients assessed for eligibility during the 2-year recruitment period, 105 were excluded. See Figure 3 for a detailed depiction of study accrual for both the treatment and control groups. Thirty-three (31%) were excluded because they did not have a land phone line, a requirement for transmitting the algorithms to the Health Buddy appliance. Most of these had cell phones only. No potential participants refused participation due to issues related to operation of the technology itself.
Device utilization
Participants used the telehealth device for an average of 70.7 days (±26.7), which constituted 86.3% (±15.0) of the total days available for use. Of note, the median percentage of use was 94.2% and the modal percentage was 100%, indicating that the vast majority of participants consistently used the telehealth device. The participant with the lowest usage rate used the device 46% of the days available.
By far, the most common reason for Health Buddy nonresponse was patient hospitalization. Two subjects traveled out of town frequently on weekends and would leave the Health Buddy at home. One subject had accidentally unhooked the Health Buddy, and a home visit was made to reconnect the device into the patient's phone line.
Nurse-initiated contacts with participants and/or clinicians
Of the 45 enrolled patients, 33 required additional contact with a research nurse (see Table 3). The most common reasons patients were contacted were nonresponse for 3 consecutive days (38.3%), repeated reporting of high levels of unrelieved pain (30%), and suicidal thoughts (10%). In all, 120 calls were placed: one call for every 25.9 response days. In every case, the problem was resolved.
NUMBER OF PATIENTS | PROBLEM | OUTGOING CALLS | RESOLUTION |
---|---|---|---|
15 | No response on Health Buddy for 3 consecutive days | 46 | Patient teaching |
17 | Pain-related issues | 36 | Advocacy/referral/patient teaching |
5 | Suicidal thoughts | 12 | Advocacy/referral |
7 | G-tube problems | 8 | Patient teaching |
5 | Sadness/depression | 6 | Advocacy/referral |
3 | Multiple symptoms | 3 | Advocacy/referral |
3 | Nausea/vomiting | 4 | Referral/patient teaching |
2 | Coughing/excessive secretions | 2 | Patient teaching |
2 | Constipation | 2 | Patient teaching |
1 | Stomatitis | 1 | Referral |
Satisfaction ratings
Responses to surveys programmed into the Health Buddy system are displayed in Table 4. Overall, respondents responded favorably, finding the installation to be easy, the content to be helpful, and the overall experience to be positive.
PERCENT OF RESPONDENTS | |
---|---|
Installation satisfaction | |
Installation problems? | |
Yes | 2 |
No | 98 |
Any difficulty completing the first training questions? | |
Yes | 7 |
No | 94 |
Length of installation? | |
2–5 minutes | 52 |
6–10 minutes | 41 |
11–15 minutes | 4 |
16–20 minutes | 2 |
Content satisfaction | |
Overall, I think the Health Buddy questions are | |
Very easy | 44 |
Somewhat easy | 16 |
Neutral | 32 |
Somewhat difficult | 4 |
Difficult | 4 |
Repeating questions reinforced knowledge and understanding | |
Strongly agree | 56 |
Somewhat agree | 28 |
Neutral | 12 |
Somewhat disagree | 4 |
Strongly disagree | 0 |
Understanding of my health condition | |
Much better | 64 |
Somewhat better | 20 |
Neutral | 16 |
Somewhat worse | 0 |
Much worse | 0 |
Managing my health condition | |
Much better | 52 |
Somewhat better | 44 |
Neutral | 4 |
Somewhat worse | 0 |
Much worse | 0 |
Recommend the device to others | |
Very willing | 80 |
Somewhat willing | 12 |
Neutral | 4 |
Somewhat unwilling | 0 |
Very unwilling | 4 |
Overall satisfaction | |
Satisfaction with device | |
Very satisfied | 45 |
Satisfied | 35 |
Somewhat satisfied | 15 |
Not very satisfied | 5 |
Satisfaction with the communication between you and your doctor or nurse | |
More satisfied | 65 |
No difference | 30 |
Less satisfied | 5 |
Ease of using the device | |
Very easy | 85 |
Easy | 15 |
Not easy | 0 |
Overall experience with the device | |
Positive | 85 |
Neutral | 15 |
Negative | 0 |
Continue to use the device | |
Very likely | 40 |
Likely | 40 |
Somewhat likely | 15 |
Not very likely | 0 |
Narrative comments
During the exit interview, participants were asked, “How was having the Health Buddy helpful to you?” Responses could be categorized into two major themes: (1) the Health Buddy provided needed information and (2) the Health Buddy improved my self-management during treatment.
Statements made related to the information provided included the following:
- • It gave me information on what could be expected from treatment
• It was a constant reminder of things to watch for
• It kept me abreast of my total condition at all times
• It gave good directions so I didn't have to ask at the cancer center
• It gave good suggestions on treatments (home remedies) such as gargles, care of feeding tube, exhaustion, and everyday symptoms
Statements made indicative that the Health Buddy improved self-management included the following:
- • I learned what I could do to make myself feel better
• It helped me manage my symptoms
• It taught me about symptom management and how to handle problems
• It let me know whether to contact a doctor or use self-care
• It gave me who to call for problems and some things to try
• It kept me aware of what I needed to do in order to make the period easier
• It reminded me to take my meds and exercise
Additionally, some participants noted the support they felt from having the Health Buddy interventions during treatment in saying the following:
- • It kind of helped my depression through acknowledging it and giving me something to do
• It made me feel I was not the only one who had experience with these things
• It comforted me because I knew what was going to happen
Poststudy survey
Twenty (45%) of the 44 patients who received the intervention responded to the mailed poststudy survey. When asked if they felt they received better care because they had the device, 13 of the 20 (65%) responded that they did. Eighteen (90%) of the treatment group responders stated they were very satisfied with their care (one stated “somewhat satisfied”) and 20 (100%) said they would recommend the cancer center for treatment. Nineteen (95%) stated they received adequate support during treatment.
Outcome Measures
Mean scores on the FACT-H&N and subscales and the MSAS and subscales taken pre-, during, and posttreatment are displayed in Table 5. As expected, average QOL scores declined during treatment, while symptom distress increased, with a return to near baseline scores posttreatment.
SCALE/SUBSCALE | PRETREATMENT | DURING TREATMENT | POSTTREATMENT |
---|---|---|---|
Total FACT-H&N | 100.3 | 85.6 | 101.5 |
FACT-G | 74.3 | 69.4 | 78.5 |
Trial Outcome Index | 62.6 | 46.0 | 65.0 |
Physical Well-Being | 21.2 | 17.6 | 21.1 |
Functional Well-Being | 15.6 | 12.5 | 17.4 |
Emotional Well-Being | 21.1 | 22.3 | 22.2 |
Social Well-Being | 21.1 | 22.3 | 22.2 |
Total MSAS | 0.7 | 1.1 | 0.8 |
Global Distress Index | 1.1 | 1.8 | 1.3 |
Physical | 0.7 | 1.5 | 1.1 |
Psychological | 1.1 | 1.2 | 0.8 |
Correlations
The relationships between percentage usage per patient and the following variables were evaluated: age, income, years of education, tumor stage, and percent poverty in patient's zip code. Percent poverty in zip code area was intended to be a surrogate measure of the patient's socioeconomic status. Results are displayed in Table 6. No significant correlations were noted, although years of education and percentage poverty in zip code showed a trend toward significance.
VARIABLE (VS % USAGE) | RELATIONSHIP | |
---|---|---|
SPEARMAN'S RHO RS | SIGNIFICANCE (ONE-TAILED) | |
Percent poverty in zip code | 0.213 | 0.083 |
Age | 0.146 | 0.173 |
Years of education | −0.325 | 0.081 |
Income | −0.292 | 0.120 |
Tumor stage | 0.196 | 0.122 |
Physical Well-Being (during treatment) | 0.310 | 0.048 |
Emotional Well-Being (during treatment) | 0.315 | 0.042 |
Although a multivariate model was planned, the lack of significant bivariate correlations precluded the need for multivariate analysis.
When percent usage was correlated with FACT-H&N total and subscales taken at baseline, during active treatment, and posttreatment, significant positive correlations were found between the percentage used and the Physical Well-Being subscale score during treatment (Spearman's rho = 0.310, P = 0.048) and between percentage used and the Emotional Well-Being subscale during treatment (Spearman's rho = 0.315, P = 0.042).
There were no significant correlations between percentage usage and the scores on the MSAS.
Discussion
Both qualitative and quantitative measures indicate that using telehealth to support symptom management during aggressive cancer treatment is both feasible and well-accepted. Patient users were not intimidated by this particular technology as it was simple to set up and use and required no previous computer training to operate. The Health Buddy was viewed as providing important and useful information. Overall, users felt that it improved their ability to self-manage their disease and the side effects of treatment and provided a sense of support and security.
Unlike other studies which use telehealth devices to monitor patient symptoms, our goal was to increase patient self-management of the symptoms experienced during intensive medical treatment, therefore avoiding increased burden on the medical system. The fact that the research nurse overseeing the responses needed to intervene only once every 25.9 days speaks to the ability of the intervention to have a positive impact on utilization of medical services.
The lack of significant relationships between usage and descriptive variables such as age and years of education suggests that the intervention was equally acceptable to all subgroups. Factors such as age, previous computer literacy, educational obtainment, and socioeconomic status did not significantly differentiate our study population in terms of compliance as verified by usage percentages.
The significant relationships found between the percentage used and the subscale scores on Physical Well-Being and Emotional Well-Being during treatment may indicate that increased use of the telemessaging intervention during treatment resulted in better physical and emotional aspects of QOL.
The high rate of daily compliance with the intervention in spite of differentiating personal variables and the severity of the treatment regimen may have been due to one or a combination of the following factors:
- • the simplicity of the technology
• the visibility of the appliance (often placed in the kitchen or living area of the home) and its flashing green light as cues to the need to respond
• the usefulness of the information provided
• the use of simple messaging language presented in an encouraging, positive manner
• affirmations related to application of the symptom management protocols suggested
• curiosity related to the day's messaging and the motivational saying which always appeared at the end
• knowledge that someone was reviewing the responses, tracking and intervening when the participant did not respond for several days
Our study supports the benefits of telehealth interventions noted by providers in a study by Sandberg et al39: opportunities for more frequent contact, greater relaxation and information due to the ability to interact in one's own home, increased accessibility by those frequently underserved, and timely medical information and monitoring. Similar to the study done in the Netherlands,[27] and [28] this study noted technological problems as the primary disadvantage; but in our intervention, we had no problems with the technology or equipment.
Although computerized technology served as a barrier to previous telehealth research, the lack of a land-based phone line was a factor preventing participation in the current study. Indeed, many participants maintained only wireless communication devices, which were not compatible with the version of the Health Buddy that was employed in this study. However, improvements in the technology since completion of this study now allow for wireless access to the appliance or provision of an independent wireless messaging device for those without such access in their own homes.
Although the data generally support the feasibility and acceptability of the telehealth-based intervention, the results should be interpreted in the context of a few study limitations. In particular, the sample size was somewhat small, and data pertaining to the socioeconomic status of participants were not available for all participants. Second, the study did not include measures of the patient's direct interactions with health-care providers during the study or specific data related to their health-care utilization (eg, emergency room visits, preventable inpatient hospitalizations, emergency calls to clinicians). The collection of more exhaustive measures of health-care utilization was limited by resources but is planned for subsequent studies. Finally, concerns regarding subject burden limited assessment of the usability of the telehealth device.
Although compliance with utilization expectations and completion of study measures was excellent during the course of the intervention, response to the follow-up survey mailed several months later was less than 50%. This low response rate was most likely due to several factors: (1) this survey was sent at the conclusion of the entire study (by this time, patients were 0–21 months past their active participation); (2) it was a mailed survey with no additional contact or follow-up effort to increase response rate; (3) participants may have felt that they had already shared their opinions in the exit interview and may have felt overburdened by study measures at this point; and (4) participants may have died, moved, or been medically unable to respond. This lack of response did limit our ability to evaluate the longitudinal impact of the intervention.
Conclusions
This telehealth intervention proved to be an acceptable and feasible means to educate and support patients during aggressive treatment for head and neck cancer. Patient compliance with telehealth interventions during periods of extreme symptom burden and declining QOL is feasible if simple technology cues the patient to participate, offers positive support and relevant education, and is targeted or tailored to their specific condition.
1 C.D. Llewellyn, M. McGurk and J. Weinman, Are psycho-social and behavioural factors related to health related-quality of life in patients with head and neck cancer?: A systematic review, Oral Oncol 41 (5) (2005), pp. 440–454. Article | | View Record in Scopus | Cited By in Scopus (24)
2 K.T. Vakharia, M.J. Ali and S.J. Wang, Quality-of-life impact of participation in a head and neck cancer support group, Otolaryngol Head Neck Surg 136 (3) (2007), pp. 405–410. Article | | View Record in Scopus | Cited By in Scopus (6)
3 P.J. Allison et al., Results of a feasibility study for a psycho-educational intervention in head and neck cancer, Psychooncology 13 (2004), pp. 482–485. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
4 L.H. Karnell et al., Influence of social support on health-related quality of life outcomes in head and neck cancer, Head Neck 29 (2) (2007), pp. 143–146. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (19)
5 K.M. Petruson, E.M. Silander and E.B. Hammerlid, Effects of psychosocial intervention on quality of life in patients with head and neck cancer, Head Neck 25 (7) (2003), pp. 576–584. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)
6 J.R.J. deLeeuw et al., Negative and positive influences of social support on depression in patients with head and neck cancer: a prospective study, Psychooncology 9 (2000), pp. 20–28. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)
7 J. Ostroff et al., Interest in and barriers to participation in multiple family groups among head and neck cancer survivors and their primary family caregivers, Fam Process 43 (2) (2004), pp. 195–208. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
8 R.L. Bashshur et al., National telemedicine initiatives: essential to healthcare reform, Telemed J E Health 15 (6) (2009), pp. 1–11.
9 K. Davis et al., An innovative symptom monitoring tool for people with advanced lung cancer: a pilot demonstration, J Support Oncol 5 (8) (2007), pp. 381–387. View Record in Scopus | Cited By in Scopus (8)
10 K.H. Mooney et al., Telephone-linked care for cancer symptom monitoring: A pilot study, Cancer Pract 10 (3) (2002), pp. 147–154. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (30)
11 R.H. Friedman et al., The virtual visit: using telecommunications technology to take care of patients, J Am Med Inform Assoc 4 (1997), pp. 413–425. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
12 A. Weaver et al., Application of mobile phone technology for managing chemotherapy-associated side-effects, Ann Oncol 18 (11) (2007), pp. 1887–1892. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
13 D. Berry et al., Computerized symptom and quality-of-life assessment for patients with cancer: Part 1: Development and pilot testing, Oncol Nurs Forum 31(5 (2004), pp. E75–E83. Full Text via CrossRef
14 K. Mullen, D. Berry and B. Zierler, Computerized symptom and quality-of-life assessment for patients with cancer: Part II: Acceptability and usability, Oncol Nurs Forum 31 (5) (2004), pp. E84–E89. Full Text via CrossRef
15 B. Fortner et al., The Cancer Care Monitor: psychometric content evaluation and pilot testing of a computer administered system for symptom screening and quality of life in adult cancer patients, J Pain Symptom Manage 26 (6) (2003), pp. 1077–1092. Article | | View Record in Scopus | Cited By in Scopus (43)
16 R. de Bree et al., Touch screen computer-assisted health-related quality of life and distress data collection in head and neck cancer patients, Clin Otolaryngol 33 (2) (2008), pp. 138–142. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
17 H. Huang et al., Developing a computerized data collection and decision support system for cancer pain management, Comput Inform Nurs 21 (4) (2003), pp. 206–217. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
18 D.J. Wilkie et al., Usability of a computerized pain report in the general public with pain and people with cancer pain, J Pain Symptom Manage 25 (3) (2003), pp. 213–224. Article | | View Record in Scopus | Cited By in Scopus (35)
19 K. Kroenke et al., Effect of telecare management on pain and depression in patients with cancer: a randomized trial, JAMA 304 (2) (2010), pp. 163–171. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
20 N. Kearney et al., Utilizing handheld computers to monitor and support patients receiving chemotherapy: results of a UK-based feasibility study, Support Care Cancer 14 (7) (2006), pp. 742–752. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
21 N.R. Chumbler et al., Remote patient–provider communication and quality of life: empirical test of a dialogic model of cancer care, J Telemed Telecare 13 (2007), pp. 20–25. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
22 A.L. Grubaugh et al., Attitudes toward medical and mental health care delivered via telehealth applications among rural and urban primary care patients, J Nerv Ment Dis 196 (2) (2008), pp. 166–170. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
23 J. Stalfors et al., Accuracy of tele-oncology compared with face-to-face consultation in head and neck cancer case conferences, J Telemed Telecare 7 (2001), pp. 338–343. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
24 C. Dorrian et al., Head and neck cancer assessment by flexible endoscopy and telemedicine, J Telemed Telecare 15 (2009), pp. 118–121. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
25 J. Stalfors et al., Haptic palpation of head and neck cancer patients—implications for education and telemedicine, Stud Health Technol Inform 81 (2001), pp. 471–474. View Record in Scopus | Cited By in Scopus (8)
26 C. Myers, Telehealth applications in head and neck oncology, J Speech Lang Pathol Audiol 29 (3) (2005), pp. 125–127.
27 J.L. van den Brink et al., Involving the patient: a prospective study on use, appreciation and effectiveness of an information system in head and neck cancer care, Int J Med Inform 74 (10) (2005), pp. 839–849. Article | | View Record in Scopus | Cited By in Scopus (14)
28 J.L. van den Brink et al., Impact on quality of life of a telemedicine system supporting head and neck cancer patients: a controlled trial during the postoperative period at home, J Am Med Inform Assoc 14 (2) (2007), pp. 198–205. Article | | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)
29 B. Head et al., Development of a telehhealth intervention for head and neck cancer patients, Telemed J E Health 15 (1) (2009), pp. 100–108. View Record in Scopus | Cited By in Scopus (1)
30 D.F. Cella et al., The Functional Assessment of Cancer Therapy (FACT) scale: development and validation of the general measure, J Clin Oncol 11 (3) (1993), pp. 570–579. View Record in Scopus | Cited By in Scopus (1626)
31 M.A. List et al., The Performance Status scale for head and neck cancer patients and the Functional Assessment of Cancer Therapy-Head and Neck scale: A study of utility and validity, Cancer 77 (11) (1996), pp. 2294–2301. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (169)
32 H.M. Mehanna and R.P. Morton, Patients' views on the utility of quality of life questionnaires in head and neck cancer: a randomised trial, Clin Otolaryngol 31 (4) (2006), pp. 310–316. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
33 V. Chang et al., The Memorial Symptom Assessment Scale short form, Cancer 89 (2000), pp. 1162–1171. Full Text via CrossRef
34 R.K. Portenoy et al., The Memorial Symptom Assessment Scale: an instrument for the evaluation of symptom prevalence, characteristics and distress, Eur J Cancer 30A (9) (1994), pp. 1326–1336. Abstract | | View Record in Scopus | Cited By in Scopus (449)
35 J.E. Tranmer et al., Measuring the symptom experience of seriously ill cancer and noncancer hospitalized patients near the end of life with the Memorial Symptom Assessment Scale, J Pain Symptom Manage 25 (5) (2003), pp. 420–429. Article | | View Record in Scopus | Cited By in Scopus (81)
36 V.T. Chang et al., Symptom and quality of life survey of medical oncology patients at a Veterans Affairs medical center: a role for symptom assessment, Cancer 88 (5) (2000), pp. 1175–1183. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (121)
37 J.F. Nelson et al., The symptom burden of chronic critical illness, Crit Care Med 32 (7) (2004), pp. 1527–1534. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (55)
38 L.B. Harrison et al., Detailed quality of life assessment in patients treated with primary radiotherapy for squamous cell cancer of the base of the tongue, Head Neck 19 (3) (1997), pp. 169–175. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (132)
39 J. Sandberg et al., A qualitative study of the experiences and satisfaction of direct telemedicine providers in diabetes case management, Telemed J E Health 15 (8) (2009), pp. 742–750. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1)
1 C.D. Llewellyn, M. McGurk and J. Weinman, Are psycho-social and behavioural factors related to health related-quality of life in patients with head and neck cancer?: A systematic review, Oral Oncol 41 (5) (2005), pp. 440–454. Article | | View Record in Scopus | Cited By in Scopus (24)
2 K.T. Vakharia, M.J. Ali and S.J. Wang, Quality-of-life impact of participation in a head and neck cancer support group, Otolaryngol Head Neck Surg 136 (3) (2007), pp. 405–410. Article | | View Record in Scopus | Cited By in Scopus (6)
3 P.J. Allison et al., Results of a feasibility study for a psycho-educational intervention in head and neck cancer, Psychooncology 13 (2004), pp. 482–485. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
4 L.H. Karnell et al., Influence of social support on health-related quality of life outcomes in head and neck cancer, Head Neck 29 (2) (2007), pp. 143–146. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (19)
5 K.M. Petruson, E.M. Silander and E.B. Hammerlid, Effects of psychosocial intervention on quality of life in patients with head and neck cancer, Head Neck 25 (7) (2003), pp. 576–584. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)
6 J.R.J. deLeeuw et al., Negative and positive influences of social support on depression in patients with head and neck cancer: a prospective study, Psychooncology 9 (2000), pp. 20–28. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)
7 J. Ostroff et al., Interest in and barriers to participation in multiple family groups among head and neck cancer survivors and their primary family caregivers, Fam Process 43 (2) (2004), pp. 195–208. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
8 R.L. Bashshur et al., National telemedicine initiatives: essential to healthcare reform, Telemed J E Health 15 (6) (2009), pp. 1–11.
9 K. Davis et al., An innovative symptom monitoring tool for people with advanced lung cancer: a pilot demonstration, J Support Oncol 5 (8) (2007), pp. 381–387. View Record in Scopus | Cited By in Scopus (8)
10 K.H. Mooney et al., Telephone-linked care for cancer symptom monitoring: A pilot study, Cancer Pract 10 (3) (2002), pp. 147–154. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (30)
11 R.H. Friedman et al., The virtual visit: using telecommunications technology to take care of patients, J Am Med Inform Assoc 4 (1997), pp. 413–425. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
12 A. Weaver et al., Application of mobile phone technology for managing chemotherapy-associated side-effects, Ann Oncol 18 (11) (2007), pp. 1887–1892. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
13 D. Berry et al., Computerized symptom and quality-of-life assessment for patients with cancer: Part 1: Development and pilot testing, Oncol Nurs Forum 31(5 (2004), pp. E75–E83. Full Text via CrossRef
14 K. Mullen, D. Berry and B. Zierler, Computerized symptom and quality-of-life assessment for patients with cancer: Part II: Acceptability and usability, Oncol Nurs Forum 31 (5) (2004), pp. E84–E89. Full Text via CrossRef
15 B. Fortner et al., The Cancer Care Monitor: psychometric content evaluation and pilot testing of a computer administered system for symptom screening and quality of life in adult cancer patients, J Pain Symptom Manage 26 (6) (2003), pp. 1077–1092. Article | | View Record in Scopus | Cited By in Scopus (43)
16 R. de Bree et al., Touch screen computer-assisted health-related quality of life and distress data collection in head and neck cancer patients, Clin Otolaryngol 33 (2) (2008), pp. 138–142. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
17 H. Huang et al., Developing a computerized data collection and decision support system for cancer pain management, Comput Inform Nurs 21 (4) (2003), pp. 206–217. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
18 D.J. Wilkie et al., Usability of a computerized pain report in the general public with pain and people with cancer pain, J Pain Symptom Manage 25 (3) (2003), pp. 213–224. Article | | View Record in Scopus | Cited By in Scopus (35)
19 K. Kroenke et al., Effect of telecare management on pain and depression in patients with cancer: a randomized trial, JAMA 304 (2) (2010), pp. 163–171. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
20 N. Kearney et al., Utilizing handheld computers to monitor and support patients receiving chemotherapy: results of a UK-based feasibility study, Support Care Cancer 14 (7) (2006), pp. 742–752. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
21 N.R. Chumbler et al., Remote patient–provider communication and quality of life: empirical test of a dialogic model of cancer care, J Telemed Telecare 13 (2007), pp. 20–25. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
22 A.L. Grubaugh et al., Attitudes toward medical and mental health care delivered via telehealth applications among rural and urban primary care patients, J Nerv Ment Dis 196 (2) (2008), pp. 166–170. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
23 J. Stalfors et al., Accuracy of tele-oncology compared with face-to-face consultation in head and neck cancer case conferences, J Telemed Telecare 7 (2001), pp. 338–343. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
24 C. Dorrian et al., Head and neck cancer assessment by flexible endoscopy and telemedicine, J Telemed Telecare 15 (2009), pp. 118–121. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
25 J. Stalfors et al., Haptic palpation of head and neck cancer patients—implications for education and telemedicine, Stud Health Technol Inform 81 (2001), pp. 471–474. View Record in Scopus | Cited By in Scopus (8)
26 C. Myers, Telehealth applications in head and neck oncology, J Speech Lang Pathol Audiol 29 (3) (2005), pp. 125–127.
27 J.L. van den Brink et al., Involving the patient: a prospective study on use, appreciation and effectiveness of an information system in head and neck cancer care, Int J Med Inform 74 (10) (2005), pp. 839–849. Article | | View Record in Scopus | Cited By in Scopus (14)
28 J.L. van den Brink et al., Impact on quality of life of a telemedicine system supporting head and neck cancer patients: a controlled trial during the postoperative period at home, J Am Med Inform Assoc 14 (2) (2007), pp. 198–205. Article | | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)
29 B. Head et al., Development of a telehhealth intervention for head and neck cancer patients, Telemed J E Health 15 (1) (2009), pp. 100–108. View Record in Scopus | Cited By in Scopus (1)
30 D.F. Cella et al., The Functional Assessment of Cancer Therapy (FACT) scale: development and validation of the general measure, J Clin Oncol 11 (3) (1993), pp. 570–579. View Record in Scopus | Cited By in Scopus (1626)
31 M.A. List et al., The Performance Status scale for head and neck cancer patients and the Functional Assessment of Cancer Therapy-Head and Neck scale: A study of utility and validity, Cancer 77 (11) (1996), pp. 2294–2301. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (169)
32 H.M. Mehanna and R.P. Morton, Patients' views on the utility of quality of life questionnaires in head and neck cancer: a randomised trial, Clin Otolaryngol 31 (4) (2006), pp. 310–316. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
33 V. Chang et al., The Memorial Symptom Assessment Scale short form, Cancer 89 (2000), pp. 1162–1171. Full Text via CrossRef
34 R.K. Portenoy et al., The Memorial Symptom Assessment Scale: an instrument for the evaluation of symptom prevalence, characteristics and distress, Eur J Cancer 30A (9) (1994), pp. 1326–1336. Abstract | | View Record in Scopus | Cited By in Scopus (449)
35 J.E. Tranmer et al., Measuring the symptom experience of seriously ill cancer and noncancer hospitalized patients near the end of life with the Memorial Symptom Assessment Scale, J Pain Symptom Manage 25 (5) (2003), pp. 420–429. Article | | View Record in Scopus | Cited By in Scopus (81)
36 V.T. Chang et al., Symptom and quality of life survey of medical oncology patients at a Veterans Affairs medical center: a role for symptom assessment, Cancer 88 (5) (2000), pp. 1175–1183. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (121)
37 J.F. Nelson et al., The symptom burden of chronic critical illness, Crit Care Med 32 (7) (2004), pp. 1527–1534. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (55)
38 L.B. Harrison et al., Detailed quality of life assessment in patients treated with primary radiotherapy for squamous cell cancer of the base of the tongue, Head Neck 19 (3) (1997), pp. 169–175. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (132)
39 J. Sandberg et al., A qualitative study of the experiences and satisfaction of direct telemedicine providers in diabetes case management, Telemed J E Health 15 (8) (2009), pp. 742–750. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1)
Results of a Multicenter Open-Label Randomized Trial Evaluating Infusion Duration of Zoledronic Acid in Multiple Myeloma Patients (the ZMAX Trial)
Original research
James R. Berenson MD
Abstract
Zoledronic acid, an intravenous (IV) bisphosphonate, is a standard treatment for multiple myeloma (MM) but may exacerbate preexisting renal dysfunction. The incidence of zoledronic acid–induced renal dysfunction may correlate with infusion duration. In this randomized, multicenter, open-label study, 176 patients with MM, at least one bone lesion, and stable renal function with a serum creatinine (SCr) level <3 mg/dL received zoledronic acid 4 mg (in 250 mL) as a 15- or 30-minute IV infusion every 3–4 weeks. At month 12, 20% (17 patients) in the 15-minute and 16% (13 patients) in the 30-minute arm experienced a clinically relevant but nonsignificant SCr-level increase (P = 0.44). By 24 months, the proportion of patients with a clinically relevant SCr-level increase was similar between arms (15-minute 28% [24 patients] vs 30-minute 27% [23 patients], P = 0.9014). Median zoledronic acid end-of-infusion concentrations were higher with the shorter infusion (15-minute 249 ng/mL vs 30-minute 172 ng/mL), and prolonging the infusion beyond 15 minutes did not influence adverse events related to zoledronic acid. For patients with MM, the safety profile of IV zoledronic acid is similar between those receiving a 15- or 30-minute infusion; therefore, determining the appropriate infusion duration of zoledronic acid should be based on individual patient considerations.
Article Outline
Considerable research has focused on preventive and/or treatment strategies to reduce bone complications in MM patients. In a large, international, randomized, phase III trial of MM patients with at least one osteolytic bone lesion, zoledronic acid (Zometa), a potent intravenous (IV) bisphosphonate that inhibits osteoclast-mediated bone resorption, reduced the overall risk of developing skeletally related events (SREs) including HCM by 16% (P = 0.03) compared with standard-dose pamidronate 90 mg (Aredia), another less potent IV bisphosphonate.[5] and [6] As a result of this study and others, monthly infusion of zoledronic acid at 4 mg over at least 15 minutes has become a common treatment for MM patients with bone involvement.
The U.S. Food and Drug Administration (FDA) has approved zoledronic acid use for patients with MM, documented bone metastases from solid tumors, or HCM.[5], [6], [7] and [8] The FDA-approved dose for MM patients is 4 mg administered as an IV infusion over at least 15 minutes every 3–4 weeks for patients with a creatinine clearance (CrCl) of >60 mL/min; when treating HCM, zoledronic acid 4 mg is administered as a single IV infusion.[5], [6], [7] and [8]
Zoledronic acid is primarily excreted intact through the kidney.9 Preexisting kidney disease and receipt of multiple cycles of bisphosphonate therapy are risk factors for subsequent kidney injury.7 In animal studies, IV bisphosphonates have been shown by histology to precipitate renal tubular injury when administered as a single high dose or when administered more frequently at lower doses.[10] and [11] Additionally, renal dysfunction, as evidenced by increased serum creatinine (SCr) levels, was reported among patients treated at a dose of 4 mg with an infusion time of 5 minutes.[7] and [12] When 4 mg zoledronic acid was administered with a longer infusion time of 15 minutes in large randomized trials, no significant difference between the renal safety profiles of zoledronic acid and pamidronate was reported.6
One hypothesis about the development of kidney injury associated with zoledronic acid is that it may be related to the peak plasma concentration as determined by infusion time. Results of a study evaluating patients with MM or other cancer types and bone metastases demonstrated that prolonging the infusion time of zoledronic acid reduced the end-of-infusion peak plasma concentration (Cmax) by 35%.9 Another theory about the development of kidney dysfunction is that insoluble precipitates may form when the blood is exposed to high concentrations of bisphosphonates as this has been shown to occur in vitro.[9] and [13] Therefore, the current management of renal adverse events (AEs) related to IV bisphosphonates is based on these theories so that reducing the peak plasma concentration of zoledronic acid may prevent the possible formation of insoluble precipitates through (1) lowering the dose, (2) slowing the infusion rate, or (3) increasing the volume of infusate.[5], [12] and [14]
Because MM patients are predisposed to experience deterioration of renal function, it is critical to ensure that zoledronic acid does not contribute to, or exacerbate, a decline in kidney function. To determine if increasing the duration of zoledronic acid infusion further results in improved renal safety, a multicenter, open-label, randomized study was designed to compare a 15-minute vs a 30-minute infusion time with an increased volume of infusate from 100 to 250 mL administered every 3–4 weeks to MM patients with osteolytic bone disease.
Patients and Methods
Patient Population
Men and women (≥18 years of age) with a diagnosis of MM, at least one bone lesion on plain film radiographs, stable kidney function (defined as two SCr level determinations of <3 mg/dL obtained at least 7 days apart during the screening period), calculated CrCl of at least 30 mL/min, Eastern Cooperative Oncology Group (ECOG) performance status of 1 or less, and a life expectancy of at least 9 months were eligible. The study excluded patients with prolonged IV bisphosphonate use (defined as use of zoledronic acid longer than 3 years or pamidronate longer than 1 year [total bisphosphonate duration could not exceed 3 years]), corrected serum calcium level at first visit of <8 or ≥12 mg/dL, or diagnosis of amyloidosis. Additionally, patients who had known hypersensitivity to zoledronic acid or other bisphosphonates; were pregnant or lactating; had uncontrolled cardiovascular disease, hypertension, or type 2 diabetes mellitus; or had a history of noncompliance with medical regimens were not eligible.
Study Design
This open-label, randomized pilot study was conducted at 45 centers in the United States. Before randomization, patients were stratified based on length of time of prior bisphosphonate treatment (bisphosphonate-naive vs ≤1 year prior bisphosphonate therapy vs >1 year prior bisphosphonate therapy) and baseline calculated CrCl (>75 vs >60–75 vs ≥30–≤60 mL/min).
Treatment and Evaluation
Patients were randomized to receive zoledronic acid 4 mg as either a 15- or a 30-minute IV infusion. The volume of infusate was increased from the standard 100 to 250 mL to provide additional hydration; infusions were administered every 3–4 weeks for up to 24 months. At the time this study was developed, the 4 mg dose was used because the dose adjustments for renal dysfunction in the current FDA labeling for zoledronic acid were not yet available.7 Patients were required to take a calcium supplement containing 500 mg of calcium and a multivitamin containing 400–500 IU of vitamin D, orally, once daily, for the duration of zoledronic acid therapy.
HCM during the trial was defined as a corrected serum calcium level ≥12 mg/dL or a lower level of hypercalcemia accompanied by symptoms and/or requiring active treatment other than rehydration. If HCM occurred more than 14 days after a zoledronic acid infusion, patients could receive a zoledronic acid infusion as treatment for HCM, even if this required administration before the next scheduled dose. Patients were allowed to remain in the study provided that HCM did not persist or recur. However, zoledronic acid treatment was immediately discontinued if patients developed HCM ≤14 days after study drug infusion; these patients received HCM treatment at the discretion of their treating physician. Also, patients experiencing HCM discontinued calcium and vitamin D supplements.
Within 2 weeks before each dose, enrolled patients were assessed for increase in SCr levels. For patients experiencing a clinically relevant increase in SCr level (defined as a rise of 0.5 mg/dL or more or a doubling of baseline SCr levels), administration of zoledronic acid was suspended until the SCr level fell to within 10% of the baseline value. During the delay, SCr levels were monitored at each regularly scheduled study visit (every 3–4 weeks) or more frequently if deemed necessary by the investigator. If the SCr level fell to within 10% of the baseline value within the subsequent 12 weeks, zoledronic acid was restarted with an infusion time that was increased by 15 minutes over the starting duration. If the rise in SCr level did not resolve within 12 weeks or if the patient experienced a second clinically relevant increase in SCr level after modification of the infusion time, treatment was permanently discontinued. Otherwise, patients were followed for 24 months. A final safety assessment, including a full hematology and chemistry profile, was performed 28 days after the last infusion.
A pretreatment dental examination with appropriate preventive dentistry was suggested for all patients with known risk factors for the development of osteonecrosis of the jaw (ONJ) (eg, cancer chemotherapy, corticosteroids, poor oral hygiene, dental extraction, or dental implants). Throughout the study, patients reporting symptoms that could be consistent with ONJ were referred to a dental professional for assessment; if exposed bone was noted on dental examination, the patient was referred to an oral surgeon for further evaluation, diagnosis, and treatment. A diagnosis of ONJ required cessation of zoledronic acid therapy and study discontinuation.
Pharmacokinetic Sampling
At the first infusion visit (visit 2), pharmacokinetic (PK) parameters were measured. If PK samples were not obtained at visit 2, they could be obtained at visit 3 (otherwise, they were recorded as not done). All blood samples for PK analysis were drawn from the contralateral arm. For patients receiving the 15-minute zoledronic acid infusion, the protocol specified that PK samples were to be drawn at exactly 10 and 15 minutes from the start of the infusion; patients receiving the 30-minute zoledronic acid infusion were to have blood samples drawn at exactly 25 and 30 minutes from the start of the infusion. The second blood sample for PK analysis was taken before the study drug infusion was stopped in both groups. PK analysis was performed by Novartis Pharmaceuticals Corporation Drug Metabolism and Pharmacokinetics France (Rueil-Malmaison, France) and SGS Cephac (Geneva Switzerland), using a competitive radioimmunoassay that has a lower limit of quantification of 0.04 ng/mL and an upper limit of quantification of 40 ng/mL.
Statistical Analysis
The primary study end point was the proportion of patients with a clinically relevant increase in SCr level at 12 months. Descriptive statistics were used to summarize the primary end point; in addition, an exploratory analysis with a logistic regression model, using treatment group, prior bisphosphonate therapy, and baseline CrCl, was performed.
Additional secondary safety end points included the proportion of patients with a clinically relevant increase in SCr level at 24 months, time to first clinically relevant increase in SCr level, and the PK profile of zoledronic acid. The proportion of patients with a clinically relevant increase in SCr level at 24 months was summarized using descriptive statistics. Time to first clinically relevant increase in SCr level was analyzed using the Kaplan-Meier method at the time of the primary analysis (12 months) and at 24 months. Plasma concentration data were evaluated by treatment group and baseline kidney function using descriptive statistics. Continuous variables of baseline and demographic characteristics between treatment groups were compared using a two-sample t-test; between-group differences in discrete variables were analyzed using Pearson's chi-squared test.
The primary analysis included all randomized patients who received at least one zoledronic acid infusion and who had valid postbaseline data for assessment. All study subjects who had evaluable PK parameters were included in a secondary PK analysis. Efficacy assessments were not included in this trial.
This pilot trial was designed to obtain additional preliminary data to support the hypothesis that a longer infusion is associated with less kidney dysfunction than a shorter infusion; therefore, a sample size of 90 patients per treatment group was selected. All statistical tests employed a significance level of 0.05 against a two-sided alternative hypothesis.
The institutional review boards of participating institutions approved the study, and all patients provided written informed consent before study entry.
Results
Study Population
Between October 2004 and October 2007, 179 MM patients with SCr <3 mg/dL were randomized to receive either a 15- or a 30-minute infusion of zoledronic acid. Of these, 176 patients (88 in each group) received at least one dose of study drug. Because of protocol violations, postbaseline data from one site were excluded from analyses, leaving 85 assessable patients in the 15-minute group and 84 patients in the 30-minute group.
Overall, the study groups were representative of a general population with MM. About two-thirds of patients had received prior bisphosphonate therapy; the duration of therapy was greater than 1 year for most of these patients (Table 1). The most common concomitant therapies included dexamethasone, thalidomide, and melphalan. Although the median age, proportion of patients who were 65 years of age or older, and ratio of men to women were greater in the 15-minute infusion group, none of the differences in baseline demographics was statistically significant. All other baseline demographics and disease characteristics, including prior bisphosphonate use and baseline CrCl values, were similar between the two groups (see Table 1). During the study, six patients in the 15-minute treatment group and one patient in the 30-minute treatment group experienced HCM. Three of the six patients in the 15-minute treatment group and one patient in the 30-minute treatment group discontinued the study as a result of HCM.
NUMBER OF PATIENTS (%)a | ||
---|---|---|
CHARACTERISTIC | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 88)b | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 88)b |
Age (years) | ||
Mean (SD) | 64 | 64 |
Median | 66 | 64 |
Range | 37–91 | 27–86 |
Age category (years) | ||
<65 | 39 (44) | 47 (53) |
≥65 | 49 (56) | 41 (47) |
Sex | ||
Male | 56 (64) | 49 (56) |
Female | 32 (36) | 39 (44) |
Race | ||
White | 70 (80) | 69 (78) |
Black | 9 (10) | 13 (15) |
Asian | 1 (1) | 1 (1) |
Other | 8 (9) | 5 (6) |
Time since diagnosis (months) | ||
Mean (SD) | 12 (24) (n = 86) | 10 (14) (n = 87) |
Median | 4 | 6 |
Range | 0–186 | 0–98c |
Prior bisphosphonate use | ||
Naive | 28 (32) | 28 (32) |
≤1 year | 12 (14) | 14 (16) |
>1 year | 48 (55) | 39 (44) |
Missing | 0 (0) | 7 (8) |
Calculated CrCl (mL/min) | ||
Mean (SD) | 87 (33) | 89 (40) |
Median | 84 | 83 |
Range | 33–210 | 31–224 |
Calculated CrCl category (mL/min) | ||
CrCl ≥75 | 54 (61) | 49 (56) |
60 < CrCL < 75 | 13 (15) | 15 (17) |
30 < CrCl ≤ 60 | 21 (24) | 24 (27) |
CrCl <30 | 0 (0) | 0 (0) |
CrCl = creatinine clearance; IV = intravenous; SD = standard deviation
a Unless otherwise notedb Safety populationc One patient had a screening visit date before the date of initial diagnosis
Protocol violations and/or deviations (n = 658) occurred during this study, affecting 139 patients. The types of protocol violations/deviations were related to protocol adherence (n = 404), timing of visits (n = 210), protocol adherence/timing of visits (n = 2), exclusion criteria (n = 22), inclusion criteria (n = 10), and informed consent (n = 1); 9 violations were unclassified. Notably, one protocol adherence deviation that occurred was incorrect infusion duration despite the patient having a stable SCr level. In the 15-minute treatment group, 15% of infusions administered were longer than 15 minutes. Among the longer infusions, 7% of the infusions correctly occurred per protocol following an SCr-level increase, whereas 7% of the prolonged infusions were 20 minutes or longer in the absence of an SCr-level increase. Similarly, in the 30-minute treatment group, 5% of patients received infusions lasting at least 35 minutes in the absence of an SCr-level increase.
Renal Safety
At 12 months, slightly fewer patients (n = 13 [16%]) in the 30-minute infusion group had a clinically relevant increase in SCr level than in the 15-minute infusion group (n = 17 [20%]); but this difference was not statistically significant, and for approximately 35% of patients in each group there were no SCr data available (Table 2). The median time to a clinically relevant increase in SCr by Kaplain-Meier was not reached in either group (data not shown). Neither previous bisphosphonate use nor baseline CrCl significantly affected the results (P = 0.5837 and P = 0.9371, respectively).
NUMBER OF PATIENTS (%) | |||
---|---|---|---|
CLINICALLY RELEVANT INCREASE IN SCR | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 85)a | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 84)a | P VALUEb |
12 Months | 0.6892 | ||
Yes | 17 (20) | 13 (16) | |
No | 38 (45) | 42 (50) | |
Unknown | 30 (35) | 29 (35) | |
24 Months | 0.9750 | ||
Yes | 24 (28) | 23 (27) | |
No | 22 (26) | 23 (27) | |
Unknown | 39 (46) | 38 (45) |
CI = confidence interval; IV = intravenous; SCr = serum creatinine
a Safety population, excluding patients with protocol violationsb P value calculated based on chi-squared test
After 24 months of treatment, the proportion of patients experiencing a clinically relevant increase in SCr level was similar between treatment groups, although for approximately 45% of patients in each group there were no SCr data available (see Table 2). Moreover, the difference in time to first clinically relevant increase in SCr level was not statistically significant between the two groups (P = 0.55) (Figure 1). However, among patients with a clinically significant rise in SCr level, the median time to SCr rise was slightly longer in the 30-minute group than in the 15-minute group (22 vs 24 weeks), but this was not statistically significant.
Increases in SCr relative to baseline led to treatment discontinuation in 20 patients (24%) receiving a 15-minute infusion and 14 patients (17%) receiving a 30-minute infusion. In these cases, the treating physician either considered the SCr level too high for continued treatment or the SCr level was persistently high despite treatment interruption.
Pharmacokinetics
Median zoledronic acid concentrations, as anticipated, were higher with the 15-minute infusion time at both sampling time points (during infusion: 15-minute group 231 ng/mL [at 10 minutes] vs 30-minute group 186 ng/mL [at 25 minutes]; end-of-infusion: 15-minute group, 249 ng/mL vs 30-minute group 172 ng/mL).
Adverse Events
Overall, the incidence and severity of AEs were as anticipated for MM patients. The most commonly reported AEs included fatigue, anemia, nausea, constipation, and back pain (Table 3). Although many AEs were reported more frequently in the 30-minute infusion group, the incidence rates of AEs suspected to be related to zoledronic acid were similar between the two groups. Toxicities were graded as mild, moderate, or severe; proportions of AEs categorized by these grades were comparable. Nonfatal serious AEs (SAEs) occurred in 26% of patients receiving the 15-minute infusion and 35% of patients receiving the 30-minute infusion; however, only one patient in the 15-minute group and two patients in the 30-minute group had SAEs suspected to be related to study medication.
NUMBER OF PATIENTS (%) | |||
---|---|---|---|
TYPE OF AE | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 85) | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 84) | TOTAL (N = 169) |
Blood and lymphatic system disorders | |||
Anemia | 19 (22) | 27 (32) | 46 (27) |
Neutropenia | 6 (7) | 12 (14) | 18 (11) |
Gastrointestinal disorders | |||
Constipation | 20 (24) | 21 (25) | 41 (24) |
Diarrhea | 14 (17) | 20 (24) | 34 (20) |
Nausea | 18 (21) | 27 (32) | 45 (27) |
Vomiting | 10 (12) | 14 (17) | 24 (14) |
General disorders | |||
Fatigue | 30 (35) | 41 (49) | 71 (42) |
Pain | 7 (8) | 10 (12) | 17 (10) |
Pain in extremity | 14 (17) | 16 (19) | 30 (18) |
Peripheral edema | 13 (15) | 20 (24) | 33 (20) |
Pyrexia | 15 (18) | 19 (23) | 34 (20) |
Infections and infestations | |||
Pneumonia | 11 (13) | 7 (8) | 18 (11) |
Upper respiratory tract infection | 13 (15) | 13 (16) | 26 (15) |
Metabolism and nutrition disorders | |||
Anorexia | 8 (9) | 9 (11) | 17 (10) |
Hypokalemia | 12 (14) | 13 (15) | 25 (14) |
Musculoskeletal and connective tissue disorders | |||
Arthralgia | 10 (11) | 16 (19) | 26 (15) |
Asthenia | 9 (10) | 13 (16) | 22 (13) |
Back pain | 19 (22) | 20 (24) | 39 (23) |
Bone pain | 10 (12) | 11 (13) | 21 (12) |
Nervous system disorders | |||
Dizziness | 11 (13) | 10 (12) | 21 (12) |
Peripheral neuropathy | 7 (8) | 15 (18) | 22 (13) |
Psychiatric disorders | |||
Insomnia | 10 (12) | 14 (17) | 24 (14) |
Respiratory, thoracic, and mediastinal disorders | |||
Cough | 13 (15) | 15 (18) | 28 (17) |
Dyspnea | 15 (18) | 17 (20) | 32 (19) |
Skin and subcutaneous tissue disorders | |||
Rash | 9 (11) | 12 (14) | 21 (12) |
AE = adverse event; IV = intravenous
a Safety population excluding patients with protocol violations
The numbers of deaths, trial discontinuations, and treatment interruptions due to AEs were similar between the two groups as well. Deaths (9 [10.6%] 15-minute group vs 6 [7.1%] 30-minute group) were not suspected to be related to zoledronic acid. Eight patients in each treatment group discontinued therapy because of an AE; events leading to treatment discontinuation that were suspected to be related to zoledronic acid occurred in two patients in the 15-minute group (skeletal pain and ONJ) and one patient in the 30-minute group (jaw pain). AEs that required treatment interruption occurred in eight and nine patients in the 15-minute and 30-minute groups, respectively.
AEs of special interest included those related to kidney dysfunction, cardiac arrhythmias, SREs, and ONJ. The number of patients reporting overall kidney and urinary disorders was the same in the two treatment groups (14 patients in each group); however, acute renal failure was reported more frequently in patients receiving the 15-minute infusion compared with the 30-minute infusion (four patients [5%] vs one patient [1%] in 30-minute group). Details of these five patients are presented in Table 4. AEs related to cardiac rhythm occurred in 20 patients while on study; however, only one case of bradycardia was suspected to be related to zoledronic acid therapy (in the 30-minute group). The incidence of SREs at 2 years was comparable in the two groups (19% in 15-minute group vs 21% in 30-minute group). The time to onset of SREs was longer in the 15-minute group (222 vs 158 days), but this was not statistically significant. A total of 10 patients with suspected ONJ were identified, with three patients in the 15-minute group (all moderate) and seven patients in the 30-minute group (mild [n = 5], moderate [n = 1], severe [n = 1]). Six of these patients received bisphosphonates before entering the study (four patients received no prior bisphosphonates), but the length of previous bisphosphonate therapy varied (0–30 months). Patients with suspected ONJ were assessed by clinicians and referred to dental professionals for further evaluation.
PATIENT DEMOGRAPHICS | TYPE OF MM | MEDICAL HISTORY | CONCURRENT MEDICATIONSa | ACUTE RENAL FAILURE DETAILS | OUTCOME |
---|---|---|---|---|---|
Zoledronic acid 4 mg IV for 15 minutes | |||||
73-year-old female Caucasian | IgG | Anemia, cardiomyopathy, CHF, cholecystectomy, benign breast lump removal, CAD, DM, dyslipidemia, central venous catheterization, chronic renal failure, GERD, hypercholesterolemia, HTN, hysterectomy, mycobacterial infection, hemorrhoids, B-cell lymphoma, seborrheic keratosis, tonsillectomy | At start of study: aspirin, losartan, digoxin, hydrochlorothiazide/lorsartan, fluconazole, folic acid, atorvastatin, vitamins, warfarinDuring study: ethambutol dihydrochloride, moxifloxacin, rifabutin, fenofibrate, omeprazole, diuretics, nitroglycerin patch, angiotensin-converting enzyme inhibitors, hydroxyzine, loratadine, furosemide, vancomycin, pantoprozole, piperacillin/tazobactam, clarithromycin | Myeloma kidney mass consistent with myeloma kidney found during study; approximately 2 weeks later the patient developed severe infection that culminated in septic shock, with acute renal failure | Nephrologist considered renal insufficiency to be partly related to past history of large-cell lymphoma and chemotherapy; patient was discharged to hospice and died of acute renal failure secondary to myeloma |
71-year-old female Caucasian | IgA | Back pain, cholecystectomy, constipation, CAD, NIDDM, hypercholesterolemia, HTN, insomnia, left knee operation, neuralgia, obesity, osteoarthritis, hysterectomy, hypoacusis, seasonal allergies, urinary incontinence | At start of study: zolpidem, amitriptyline, loratidine, tolterodine l-tartrate, valsartan, metrotoprolol, furosemide, ibuprofen, clonazepam, gabapentin, liodcaine, hydrocodone/acetaminophen, quinine sulfate, simvastatin During study: calcium, multivitamins, lactulose, trazodone, hydromorphone, cyclobenzaprine, glipizide, macrogol, lorazepam, methadone, potassium, lisinopril, furosemide, meperidine, promethazine | Developed moderate acute renal failure on the day of her first dose; considered not associated with zoledronic acid | Renal ultrasound showed arterial stenosis; resolved approximately 1 month after diagnosis |
65-year-old male Caucasian | IgG | Oxycodone hypersensitivity, anemia, back pain, spine metastases, spinal compression fracture, depression, fatigue, inguinal hernia repair, spinal fusion (L1–L3) surgery, bilateral hip arthroplasty, pain, pneumonia, staphylococcal infection | At start of study: fluconazole, morphine sulfate, oxycodone/acetaminophen During study: naproxen, darbepoietin alfa, sodium ferrifluconate, calcium with vitamin D, cephalexin, dexamethasone, alginic acid, docusate, heparin, sodium polystyrene, levofloxacin, filgrastim, lansoprazole | After 5 doses of zoledronic acid, patient developed severe acute renal failure with elevated SCr; not suspected to be related to zoledronic acid | Resolved 9 days later following treatment with cephalexin and dexamethasone |
56-year-old female Caucasian | IgA | Osteolysis, cataract surgery, constipation, bone lesions, hypercholesterolemia, HTN, musculoskeletal pain, anorexia | At start of study: ibuprofen, oxycodone, propoxyphene/acetaminophen, hydrocodone/acetaminophen, valsartan, calcium/vitamin D, potassium chloride, docusate sodiumDuring study: vancomycin, acyclovir | Approximately 1 week after 9th zoledronic acid dose, patient developed acute renal failure with an increased SCr (12.5 mg/dL); not suspected to be related to zoledronic acid | Resulted from myeloma progression to plasma cell leukemia; emergency dialysis performed; catheter-related sepsis occurred approximately 1 month later, and patient died of sepsis and disease progression |
Zoledronic acid 4 mg IV for 30 minutes | |||||
80-year-old male African American | IgG | Anemia, arteriosclerotic heart disease, bilateral ankle swelling/pain, degenerative joint disease, dyspnea on exertion, fatigue, GERD, HTN, neutropenia, shoulder pain, vasovagal syncope | At start of study: aspirin, atenolol, multivitamin, doxazosin, fosinopril, hydrochlorothiazide, amlodipine besylate, simvastatinDuring study: darbepoietin alfa, warfarin sodium, furosemide, omeprazole, calcium carbonate | Approximately 1 month after 2nd dose, patient experienced increased SCr (2.9 mg/dL, 53% increase from baseline); relationship to zoledronic acid unknown | Discontinued from study after 2nd dose, and SCr remained elevated for 2 months following discontinuation |
CAD = coronary artery disease; CHF = congestive heart failure; DM = diabetes mellitus; GERD = gastroesophageal reflux disease; HTN = hypertension; MM = multiple myeloma; NIDDM = non-insulin-dependent diabetes mellitus; SCr = serum creatinine
Discussion
During the past decade, bisphosphonate therapy has become an important adjunctive treatment to prevent the emergence, or worsening, of SREs in patients with MM involving the bone.15 Kidney failure is a common and severe complication of MM that may be exacerbated by chronic administration of zoledronic acid.7 A study evaluating zoledronic acid in patients with cancer and bone metastases suggests that increasing the infusion time decreases the Cmax, which may result in fewer renal AEs.[9] and [12] This study was designed to assess whether prolonging the infusion time of zoledronic acid from the recommended 15 to 30 minutes would improve kidney safety in MM patients, as evidenced by fewer rises in SCr levels. To our knowledge, this is the only trial that has been designed to evaluate the impact of infusion duration on renal effects in this population.
The 12-month results of this pilot study showed a trend toward improved renal safety with the longer infusion time, this difference not being statistically significant. By 24 months, however, there were no differences in SCr level elevations between the two groups. The clinically relevant SCr increases observed in our study, however, differ from those reported by Rosen and colleagues,[5] and [6] who first evaluated zoledronic acid for patients with MM. In that study, 4%–11% of patients experienced kidney function deterioration, manifested by SCr increases, which is much lower than the rate observed in our study. However, several differences exist between our trial and the Rosen study. The Rosen study included both breast cancer patients with at least one bone metastasis and Durie-Salmon stage 3 MM patients with at least one osteolytic lesion, whereas our study only included MM patients with at least one bone lesion. Additionally, the criteria for defining a clinically relevant SCr increase differ between the two studies; therefore, one cannot directly compare the incidence of kidney dysfunction between these two studies. Although in our study the sample size was small, confidence intervals were wide, and protocol deviations did not permit a robust comparison, the results of this pilot study suggest that the longer infusion time of 30 minutes every 3–4 weeks for 2 years for MM patients with bone disease is also safe and well-tolerated.
As expected, PK data showed that the median zoledronic acid concentrations were greater in the samples obtained from the 15-minute group compared to those from the 30-minute group. This effect was observed in samples obtained both 5 minutes before the end of infusion and at the end of infusion.
Increasing the infusion time did not significantly alter the AE profile and was not associated with any new or unexpected AEs. The incidence rates of deaths, SAEs, treatment-related AEs, and overall AEs were generally comparable between treatment groups. Overall, the incidence rates of reported SREs and ONJ were as expected for this patient population, which are important factors when considering zoledronic acid for patients with MM, where the goal of ongoing monthly IV bisphosphonate therapy is to prevent the development of new SREs without increasing the risk of AEs, such as ONJ.
Finally, the FDA-approved current labeling for zoledronic acid recommends decreasing the dose of this bisphosphonate based on baseline kidney function.7 Because these recommendations were not in place at the time that this study was designed, whether the implementation of these dosing guidelines for patients with MM along with varying infusion durations would have impacted the results observed in our study cannot be ascertained.
In summary, the results of this study suggest that the safety profile of IV zoledronic acid is similar regardless of a 15-minute or a 30-minute infusion duration. However, because the study was not powered to detect statistical significance and the current renal dosing guidelines for zoledronic acid were not used in this study, large randomized studies, using current dosing recommendations, will be required to further assess the effects on kidney safety of prolonging the infusion time of ongoing monthly IV zoledronic acid therapy for patients with MM.
Acknowledgments
The authors thank Syntaxx Communications, Inc., specifically, Kristin Hennenfent, PharmD, MBA, BCPS, and Lisa Holle, PharmD, BCOP, who provided manuscript development and medical writing services, and Holly Matthews, BS, who provided editorial services, with support from Novartis Pharmaceuticals Corporation. We also thank all participating patients and study personnel. Research support was provided by Novartis Pharmaceuticals Corporation (East Hanover, NJ).
References
1 A. Jemal, R. Siegel and J. Xu et al., Cancer statistics, 2010, CA Cancer J Clin 60 (2010), pp. 277–300. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (543)
2 R.A. Kyle, M.A. Gertz and T.E. Witzig et al., Review of 1027 patients with newly diagnosed multiple myeloma, Mayo Clin Proc 78 (1) (2003), pp. 21–33. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (396)
3 A. Corso, P. Zappasodi and C. Pascutto et al., Urinary proteins in multiple myeloma: correlation with clinical parameters and diagnostic implications, Ann Hematol 82 (8) (2003), pp. 487–491. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
4 V. Eleutherakis-Papaiakovou, A. Bamias and D. Gika et al., Renal failure in multiple myeloma: incidence, correlations, and prognostic significance, Leuk Lymphoma 48 (2) (2007), pp. 337–341. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (35)
5 L.S. Rosen, D. Gordon and M. Kaminski et al., Zoledronic acid versus pamidronate in the treatment of skeletal metastases in patients with breast cancer or osteolytic lesions of multiple myeloma: a phase III, double-blind, comparative trial, Cancer J 7 (5) (2001), pp. 377–387. View Record in Scopus | Cited By in Scopus (461)
6 L.S. Rosen, D. Gordon and M. Kaminski et al., Long-term efficacy and safety of zoledronic acid compared with pamidronate disodium in the treatment of skeletal complications in patients with advanced multiple myeloma or breast carcinoma: a randomized, double-blind, multicenter, comparative trial, Cancer 98 (8) (2003), pp. 1735–1744. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (329)
7 , Zometa (package insert), Novartis Pharmaceuticals, Corporation, East Hanover, NJ (2008).
8 P. Major, A. Lortholary and J. Han et al., Zoledronic acid is superior to pamidronate in the treatment of hypercalcemia of malignancy: a pooled analysis of two randomized, controlled clinical trials, J Clin Oncol 19 (2) (2001), pp. 558–567. View Record in Scopus | Cited By in Scopus (325)
9 T. Chen, J. Berenson and R. Vescio et al., Pharmacokinetics and pharmacodynamics of zoledronic acid in cancer patients with bone metastases, J Clin Pharmacol 42 (11) (2002), pp. 1228–1236. View Record in Scopus | Cited By in Scopus (139)
10 T. Pfister, E. Atzpodien and F. Bauss, The renal effects of minimally nephrotoxic doses of ibandronate and zoledronate following single and intermittent intravenous administration in rats, Toxicology 191 (2003), pp. 159–167. Article | | View Record in Scopus | Cited By in Scopus (48)
11 T. Pfister, E. Aztpodien, B. Bohrmann and F. Bauss, Acute renal effects of intravenous bisphosphonates in the rat, Basic Clin Pharmacol Toxicol 97 (2005), pp. 374–381. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
12 F. Saad, D.M. Gleason and R. Murray et al., A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma, J Natl Cancer Inst 94 (19) (2002), pp. 1458–1468. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (650)
13 S. Kautiainen, S. Luurila, P. Ylitalo and R. Ylitalo, Transformation of bisphosphonates into insoluble material in human blood in vitro, Methods Find Exp Clin Pharmacol 20 (4) (1998), pp. 289–295. View Record in Scopus | Cited By in Scopus (5)
14 L.S. Rosen, D. Gordon and S. Tchekmedyian et al., Zoledronic acid versus placebo in the treatment of skeletal metastases in patients with lung cancer and other solid tumors: a phase III, double-blind, randomized trial—the Zoledronic Acid Lung Cancer and Other Solid Tumors Study Group, J Clin Oncol 21 (16) (2003), pp. 3150–3157. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (251)
15 M.A. Hussein, Multiple myeloma: most common end-organ damage and management, J Natl Compr Canc Netw 5 (2) (2007), pp. 170–178. View Record in Scopus | Cited By in Scopus (4)
Appendix
The following ZMAX Trial principal investigators participated in this study: Bart Barlogie, MD, Myeloma Institute For Research and Therapy; James Berenson, MD, Oncotherapeutics; Robert Bloom, MD, Providence Cancer Center, Clinical Trials Department; Ralph Boccia, MD, Center for Cancer and Blood Disorders; Donald Brooks, MD, Arizona Clinical Research Center, Inc.; Robert Brouillard, MD, Robert P. Brouillard, MD, and Delvyn Case, MD, Maine Center for Cancer Medicine and Blood Disorders, Pharmacy; Veena Charu, MD, Pacific Cancer Medical Center; Naveed Chowhan, MD, Cancer Care Center, Inc; Robert Collins, MD, University of Texas Southwestern Medical Center at Dallas; Thomas Cosgriff, MD, Hematology and Oncology Specialists, LLC; Jose Cruz, MD, Joe Arrington Cancer Research and Treatment Center; Surrinder Dang, MD, Oncology Specialties; Sheldon Davidson, MD, North Valley H/O; Tracy Dobbs, MD, Baptist Regional Cancer Center; Luke Dreisbach, MD, Desert Hematology Oncology Medical Group; Isaac Esseesse, MD, Hematology Oncology Associates of Central Brevard, Laboratory; Mark Fesen, MD, Hutchinson Clinic, PA; George Geils, Jr., MD, Charleston Hematology Oncology Associates, PA; Michael Greenhawt, MD, South Florida Oncology-Hematology; Manuel Guerra, MD, ORA; Rita Gupta, MD, Oncology-Hematology Associates, PA; Vicram Gupta, MD, Saint Joseph Oncology; Alexandre Hageboutros, MD, Cancer Institute of New Jersey at Cooper Hospital; Vincent Hansen, MD, Utah Hematology Oncology; David Henry, MD, Pennsylvania Oncology Hematology Associates; Benjamin Himpler, MD, Syracuse Hematology/Oncology PC; Winston Ho, MD, Hematology/Oncology Group of Orange County; William Horvath, MD, Haematology Oncology Associates of Ohio and Michigan, PC; Paul Hyman, MD, Hematology Oncology Associates of Western Suffolk; Min Kang, MD, Western Washington Oncology; Mark Keaton, MD, Augusta Oncology Associates, PC; Howard Kesselheim, MD, The Center for Cancer and Hematologic Disease; Kapisthalam Kumar, MD, Pasco Hernando Oncology Associates, PA; Edward Lee, MD, Maryland Oncology-Hematology, PA; André Liem, MD, Pacific Shore Medical Group; Timothy Lopez, MD, New Mexico Cancer Care Associates, Cancer Institute of New Mexico; Paul Michael, MD, Comprehensive Cancer Centers of Nevada; Michael Milder, MD, Swedish Cancer Institute; Barry Mirtsching, MD, Center for Oncology Research & Treatment, PA; Ruben Niesvizky, MD, New York Presbyterian Hospital; Jorge Otoya, MD, Osceola Cancer Center; Joseph Pascuzzo, MD, California Oncology of the Central Valley; Ravi Patel, MD, Comprehensive Blood and Cancer Center Lab; Allen Patton, MD, Hematology Oncology Associates, PA; Kelly Pendergrass, MD, Kansas City Cancer Center, LLC; Anthony Phillips, MD, Fox Valley Hematolgy Oncology, SC; Robert Raju, MD, Dayton Oncology and Hematology, PA; Harry Ramsey, MD, Berks Hematology Oncology Associates; Ritesh Rathore, MD, Roger Williams Hospital Medical Center; Phillip Reid, MD, Central Jersey Oncology Center; Robert Robles, MD, Bay Area Cancer Research Group, LLC; Stephen Rosenoff, MD, Oncology and Hematology Associates of Southwest Virginia, Inc; Martin Rubenstein, MD, Southbay Oncology Hematology Partners; Mansoor Saleh, MD, Georgia Cancer Specialists; Sundaresan Sambandam, MD, Hematology and Oncology Associates of RI; Mukund Shah, MD, Antelope Valley Cancer Center; David Siegel, MD, Hackensack University Medical Center; Nelida Sjak-Shie, MD, The Center for Cancer Care and Research; Michael Stone, MD, Greeley Medical Clinic; Stefano Tarantolo, MD, Nebraska Methodist Hospital; Joseph Volk, MD, Palo Verde Hematology Oncology, Ltd; Mitchell Weisberg, MD, MetCare Oncology; Ann Wierman, MD, Nevada Cancer Center; Donald Woytowitz, Jr., MD, Florida Cancer Specialists; Peter Yu, MD, Camino Medical Group.
Correspondence to: James R. Berenson, MD, Institute for Myeloma & Bone Cancer Research, 9201 West Sunset Boulevard, Suite 300, West Hollywood, CA 90069; telephone: (310) 623–1214; fax: (310) 623–1120
Original research
James R. Berenson MD, a,
Abstract
Zoledronic acid, an intravenous (IV) bisphosphonate, is a standard treatment for multiple myeloma (MM) but may exacerbate preexisting renal dysfunction. The incidence of zoledronic acid–induced renal dysfunction may correlate with infusion duration. In this randomized, multicenter, open-label study, 176 patients with MM, at least one bone lesion, and stable renal function with a serum creatinine (SCr) level <3 mg/dL received zoledronic acid 4 mg (in 250 mL) as a 15- or 30-minute IV infusion every 3–4 weeks. At month 12, 20% (17 patients) in the 15-minute and 16% (13 patients) in the 30-minute arm experienced a clinically relevant but nonsignificant SCr-level increase (P = 0.44). By 24 months, the proportion of patients with a clinically relevant SCr-level increase was similar between arms (15-minute 28% [24 patients] vs 30-minute 27% [23 patients], P = 0.9014). Median zoledronic acid end-of-infusion concentrations were higher with the shorter infusion (15-minute 249 ng/mL vs 30-minute 172 ng/mL), and prolonging the infusion beyond 15 minutes did not influence adverse events related to zoledronic acid. For patients with MM, the safety profile of IV zoledronic acid is similar between those receiving a 15- or 30-minute infusion; therefore, determining the appropriate infusion duration of zoledronic acid should be based on individual patient considerations.
Article Outline
Considerable research has focused on preventive and/or treatment strategies to reduce bone complications in MM patients. In a large, international, randomized, phase III trial of MM patients with at least one osteolytic bone lesion, zoledronic acid (Zometa), a potent intravenous (IV) bisphosphonate that inhibits osteoclast-mediated bone resorption, reduced the overall risk of developing skeletally related events (SREs) including HCM by 16% (P = 0.03) compared with standard-dose pamidronate 90 mg (Aredia), another less potent IV bisphosphonate.[5] and [6] As a result of this study and others, monthly infusion of zoledronic acid at 4 mg over at least 15 minutes has become a common treatment for MM patients with bone involvement.
The U.S. Food and Drug Administration (FDA) has approved zoledronic acid use for patients with MM, documented bone metastases from solid tumors, or HCM.[5], [6], [7] and [8] The FDA-approved dose for MM patients is 4 mg administered as an IV infusion over at least 15 minutes every 3–4 weeks for patients with a creatinine clearance (CrCl) of >60 mL/min; when treating HCM, zoledronic acid 4 mg is administered as a single IV infusion.[5], [6], [7] and [8]
Zoledronic acid is primarily excreted intact through the kidney.9 Preexisting kidney disease and receipt of multiple cycles of bisphosphonate therapy are risk factors for subsequent kidney injury.7 In animal studies, IV bisphosphonates have been shown by histology to precipitate renal tubular injury when administered as a single high dose or when administered more frequently at lower doses.[10] and [11] Additionally, renal dysfunction, as evidenced by increased serum creatinine (SCr) levels, was reported among patients treated at a dose of 4 mg with an infusion time of 5 minutes.[7] and [12] When 4 mg zoledronic acid was administered with a longer infusion time of 15 minutes in large randomized trials, no significant difference between the renal safety profiles of zoledronic acid and pamidronate was reported.6
One hypothesis about the development of kidney injury associated with zoledronic acid is that it may be related to the peak plasma concentration as determined by infusion time. Results of a study evaluating patients with MM or other cancer types and bone metastases demonstrated that prolonging the infusion time of zoledronic acid reduced the end-of-infusion peak plasma concentration (Cmax) by 35%.9 Another theory about the development of kidney dysfunction is that insoluble precipitates may form when the blood is exposed to high concentrations of bisphosphonates as this has been shown to occur in vitro.[9] and [13] Therefore, the current management of renal adverse events (AEs) related to IV bisphosphonates is based on these theories so that reducing the peak plasma concentration of zoledronic acid may prevent the possible formation of insoluble precipitates through (1) lowering the dose, (2) slowing the infusion rate, or (3) increasing the volume of infusate.[5], [12] and [14]
Because MM patients are predisposed to experience deterioration of renal function, it is critical to ensure that zoledronic acid does not contribute to, or exacerbate, a decline in kidney function. To determine if increasing the duration of zoledronic acid infusion further results in improved renal safety, a multicenter, open-label, randomized study was designed to compare a 15-minute vs a 30-minute infusion time with an increased volume of infusate from 100 to 250 mL administered every 3–4 weeks to MM patients with osteolytic bone disease.
Patients and Methods
Patient Population
Men and women (≥18 years of age) with a diagnosis of MM, at least one bone lesion on plain film radiographs, stable kidney function (defined as two SCr level determinations of <3 mg/dL obtained at least 7 days apart during the screening period), calculated CrCl of at least 30 mL/min, Eastern Cooperative Oncology Group (ECOG) performance status of 1 or less, and a life expectancy of at least 9 months were eligible. The study excluded patients with prolonged IV bisphosphonate use (defined as use of zoledronic acid longer than 3 years or pamidronate longer than 1 year [total bisphosphonate duration could not exceed 3 years]), corrected serum calcium level at first visit of <8 or ≥12 mg/dL, or diagnosis of amyloidosis. Additionally, patients who had known hypersensitivity to zoledronic acid or other bisphosphonates; were pregnant or lactating; had uncontrolled cardiovascular disease, hypertension, or type 2 diabetes mellitus; or had a history of noncompliance with medical regimens were not eligible.
Study Design
This open-label, randomized pilot study was conducted at 45 centers in the United States. Before randomization, patients were stratified based on length of time of prior bisphosphonate treatment (bisphosphonate-naive vs ≤1 year prior bisphosphonate therapy vs >1 year prior bisphosphonate therapy) and baseline calculated CrCl (>75 vs >60–75 vs ≥30–≤60 mL/min).
Treatment and Evaluation
Patients were randomized to receive zoledronic acid 4 mg as either a 15- or a 30-minute IV infusion. The volume of infusate was increased from the standard 100 to 250 mL to provide additional hydration; infusions were administered every 3–4 weeks for up to 24 months. At the time this study was developed, the 4 mg dose was used because the dose adjustments for renal dysfunction in the current FDA labeling for zoledronic acid were not yet available.7 Patients were required to take a calcium supplement containing 500 mg of calcium and a multivitamin containing 400–500 IU of vitamin D, orally, once daily, for the duration of zoledronic acid therapy.
HCM during the trial was defined as a corrected serum calcium level ≥12 mg/dL or a lower level of hypercalcemia accompanied by symptoms and/or requiring active treatment other than rehydration. If HCM occurred more than 14 days after a zoledronic acid infusion, patients could receive a zoledronic acid infusion as treatment for HCM, even if this required administration before the next scheduled dose. Patients were allowed to remain in the study provided that HCM did not persist or recur. However, zoledronic acid treatment was immediately discontinued if patients developed HCM ≤14 days after study drug infusion; these patients received HCM treatment at the discretion of their treating physician. Also, patients experiencing HCM discontinued calcium and vitamin D supplements.
Within 2 weeks before each dose, enrolled patients were assessed for increase in SCr levels. For patients experiencing a clinically relevant increase in SCr level (defined as a rise of 0.5 mg/dL or more or a doubling of baseline SCr levels), administration of zoledronic acid was suspended until the SCr level fell to within 10% of the baseline value. During the delay, SCr levels were monitored at each regularly scheduled study visit (every 3–4 weeks) or more frequently if deemed necessary by the investigator. If the SCr level fell to within 10% of the baseline value within the subsequent 12 weeks, zoledronic acid was restarted with an infusion time that was increased by 15 minutes over the starting duration. If the rise in SCr level did not resolve within 12 weeks or if the patient experienced a second clinically relevant increase in SCr level after modification of the infusion time, treatment was permanently discontinued. Otherwise, patients were followed for 24 months. A final safety assessment, including a full hematology and chemistry profile, was performed 28 days after the last infusion.
A pretreatment dental examination with appropriate preventive dentistry was suggested for all patients with known risk factors for the development of osteonecrosis of the jaw (ONJ) (eg, cancer chemotherapy, corticosteroids, poor oral hygiene, dental extraction, or dental implants). Throughout the study, patients reporting symptoms that could be consistent with ONJ were referred to a dental professional for assessment; if exposed bone was noted on dental examination, the patient was referred to an oral surgeon for further evaluation, diagnosis, and treatment. A diagnosis of ONJ required cessation of zoledronic acid therapy and study discontinuation.
Pharmacokinetic Sampling
At the first infusion visit (visit 2), pharmacokinetic (PK) parameters were measured. If PK samples were not obtained at visit 2, they could be obtained at visit 3 (otherwise, they were recorded as not done). All blood samples for PK analysis were drawn from the contralateral arm. For patients receiving the 15-minute zoledronic acid infusion, the protocol specified that PK samples were to be drawn at exactly 10 and 15 minutes from the start of the infusion; patients receiving the 30-minute zoledronic acid infusion were to have blood samples drawn at exactly 25 and 30 minutes from the start of the infusion. The second blood sample for PK analysis was taken before the study drug infusion was stopped in both groups. PK analysis was performed by Novartis Pharmaceuticals Corporation Drug Metabolism and Pharmacokinetics France (Rueil-Malmaison, France) and SGS Cephac (Geneva Switzerland), using a competitive radioimmunoassay that has a lower limit of quantification of 0.04 ng/mL and an upper limit of quantification of 40 ng/mL.
Statistical Analysis
The primary study end point was the proportion of patients with a clinically relevant increase in SCr level at 12 months. Descriptive statistics were used to summarize the primary end point; in addition, an exploratory analysis with a logistic regression model, using treatment group, prior bisphosphonate therapy, and baseline CrCl, was performed.
Additional secondary safety end points included the proportion of patients with a clinically relevant increase in SCr level at 24 months, time to first clinically relevant increase in SCr level, and the PK profile of zoledronic acid. The proportion of patients with a clinically relevant increase in SCr level at 24 months was summarized using descriptive statistics. Time to first clinically relevant increase in SCr level was analyzed using the Kaplan-Meier method at the time of the primary analysis (12 months) and at 24 months. Plasma concentration data were evaluated by treatment group and baseline kidney function using descriptive statistics. Continuous variables of baseline and demographic characteristics between treatment groups were compared using a two-sample t-test; between-group differences in discrete variables were analyzed using Pearson's chi-squared test.
The primary analysis included all randomized patients who received at least one zoledronic acid infusion and who had valid postbaseline data for assessment. All study subjects who had evaluable PK parameters were included in a secondary PK analysis. Efficacy assessments were not included in this trial.
This pilot trial was designed to obtain additional preliminary data to support the hypothesis that a longer infusion is associated with less kidney dysfunction than a shorter infusion; therefore, a sample size of 90 patients per treatment group was selected. All statistical tests employed a significance level of 0.05 against a two-sided alternative hypothesis.
The institutional review boards of participating institutions approved the study, and all patients provided written informed consent before study entry.
Results
Study Population
Between October 2004 and October 2007, 179 MM patients with SCr <3 mg/dL were randomized to receive either a 15- or a 30-minute infusion of zoledronic acid. Of these, 176 patients (88 in each group) received at least one dose of study drug. Because of protocol violations, postbaseline data from one site were excluded from analyses, leaving 85 assessable patients in the 15-minute group and 84 patients in the 30-minute group.
Overall, the study groups were representative of a general population with MM. About two-thirds of patients had received prior bisphosphonate therapy; the duration of therapy was greater than 1 year for most of these patients (Table 1). The most common concomitant therapies included dexamethasone, thalidomide, and melphalan. Although the median age, proportion of patients who were 65 years of age or older, and ratio of men to women were greater in the 15-minute infusion group, none of the differences in baseline demographics was statistically significant. All other baseline demographics and disease characteristics, including prior bisphosphonate use and baseline CrCl values, were similar between the two groups (see Table 1). During the study, six patients in the 15-minute treatment group and one patient in the 30-minute treatment group experienced HCM. Three of the six patients in the 15-minute treatment group and one patient in the 30-minute treatment group discontinued the study as a result of HCM.
NUMBER OF PATIENTS (%)a | ||
---|---|---|
CHARACTERISTIC | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 88)b | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 88)b |
Age (years) | ||
Mean (SD) | 64 | 64 |
Median | 66 | 64 |
Range | 37–91 | 27–86 |
Age category (years) | ||
<65 | 39 (44) | 47 (53) |
≥65 | 49 (56) | 41 (47) |
Sex | ||
Male | 56 (64) | 49 (56) |
Female | 32 (36) | 39 (44) |
Race | ||
White | 70 (80) | 69 (78) |
Black | 9 (10) | 13 (15) |
Asian | 1 (1) | 1 (1) |
Other | 8 (9) | 5 (6) |
Time since diagnosis (months) | ||
Mean (SD) | 12 (24) (n = 86) | 10 (14) (n = 87) |
Median | 4 | 6 |
Range | 0–186 | 0–98c |
Prior bisphosphonate use | ||
Naive | 28 (32) | 28 (32) |
≤1 year | 12 (14) | 14 (16) |
>1 year | 48 (55) | 39 (44) |
Missing | 0 (0) | 7 (8) |
Calculated CrCl (mL/min) | ||
Mean (SD) | 87 (33) | 89 (40) |
Median | 84 | 83 |
Range | 33–210 | 31–224 |
Calculated CrCl category (mL/min) | ||
CrCl ≥75 | 54 (61) | 49 (56) |
60 < CrCL < 75 | 13 (15) | 15 (17) |
30 < CrCl ≤ 60 | 21 (24) | 24 (27) |
CrCl <30 | 0 (0) | 0 (0) |
CrCl = creatinine clearance; IV = intravenous; SD = standard deviation
a Unless otherwise notedb Safety populationc One patient had a screening visit date before the date of initial diagnosis
Protocol violations and/or deviations (n = 658) occurred during this study, affecting 139 patients. The types of protocol violations/deviations were related to protocol adherence (n = 404), timing of visits (n = 210), protocol adherence/timing of visits (n = 2), exclusion criteria (n = 22), inclusion criteria (n = 10), and informed consent (n = 1); 9 violations were unclassified. Notably, one protocol adherence deviation that occurred was incorrect infusion duration despite the patient having a stable SCr level. In the 15-minute treatment group, 15% of infusions administered were longer than 15 minutes. Among the longer infusions, 7% of the infusions correctly occurred per protocol following an SCr-level increase, whereas 7% of the prolonged infusions were 20 minutes or longer in the absence of an SCr-level increase. Similarly, in the 30-minute treatment group, 5% of patients received infusions lasting at least 35 minutes in the absence of an SCr-level increase.
Renal Safety
At 12 months, slightly fewer patients (n = 13 [16%]) in the 30-minute infusion group had a clinically relevant increase in SCr level than in the 15-minute infusion group (n = 17 [20%]); but this difference was not statistically significant, and for approximately 35% of patients in each group there were no SCr data available (Table 2). The median time to a clinically relevant increase in SCr by Kaplain-Meier was not reached in either group (data not shown). Neither previous bisphosphonate use nor baseline CrCl significantly affected the results (P = 0.5837 and P = 0.9371, respectively).
NUMBER OF PATIENTS (%) | |||
---|---|---|---|
CLINICALLY RELEVANT INCREASE IN SCR | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 85)a | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 84)a | P VALUEb |
12 Months | 0.6892 | ||
Yes | 17 (20) | 13 (16) | |
No | 38 (45) | 42 (50) | |
Unknown | 30 (35) | 29 (35) | |
24 Months | 0.9750 | ||
Yes | 24 (28) | 23 (27) | |
No | 22 (26) | 23 (27) | |
Unknown | 39 (46) | 38 (45) |
CI = confidence interval; IV = intravenous; SCr = serum creatinine
a Safety population, excluding patients with protocol violationsb P value calculated based on chi-squared test
After 24 months of treatment, the proportion of patients experiencing a clinically relevant increase in SCr level was similar between treatment groups, although for approximately 45% of patients in each group there were no SCr data available (see Table 2). Moreover, the difference in time to first clinically relevant increase in SCr level was not statistically significant between the two groups (P = 0.55) (Figure 1). However, among patients with a clinically significant rise in SCr level, the median time to SCr rise was slightly longer in the 30-minute group than in the 15-minute group (22 vs 24 weeks), but this was not statistically significant.
Increases in SCr relative to baseline led to treatment discontinuation in 20 patients (24%) receiving a 15-minute infusion and 14 patients (17%) receiving a 30-minute infusion. In these cases, the treating physician either considered the SCr level too high for continued treatment or the SCr level was persistently high despite treatment interruption.
Pharmacokinetics
Median zoledronic acid concentrations, as anticipated, were higher with the 15-minute infusion time at both sampling time points (during infusion: 15-minute group 231 ng/mL [at 10 minutes] vs 30-minute group 186 ng/mL [at 25 minutes]; end-of-infusion: 15-minute group, 249 ng/mL vs 30-minute group 172 ng/mL).
Adverse Events
Overall, the incidence and severity of AEs were as anticipated for MM patients. The most commonly reported AEs included fatigue, anemia, nausea, constipation, and back pain (Table 3). Although many AEs were reported more frequently in the 30-minute infusion group, the incidence rates of AEs suspected to be related to zoledronic acid were similar between the two groups. Toxicities were graded as mild, moderate, or severe; proportions of AEs categorized by these grades were comparable. Nonfatal serious AEs (SAEs) occurred in 26% of patients receiving the 15-minute infusion and 35% of patients receiving the 30-minute infusion; however, only one patient in the 15-minute group and two patients in the 30-minute group had SAEs suspected to be related to study medication.
NUMBER OF PATIENTS (%) | |||
---|---|---|---|
TYPE OF AE | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 85) | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 84) | TOTAL (N = 169) |
Blood and lymphatic system disorders | |||
Anemia | 19 (22) | 27 (32) | 46 (27) |
Neutropenia | 6 (7) | 12 (14) | 18 (11) |
Gastrointestinal disorders | |||
Constipation | 20 (24) | 21 (25) | 41 (24) |
Diarrhea | 14 (17) | 20 (24) | 34 (20) |
Nausea | 18 (21) | 27 (32) | 45 (27) |
Vomiting | 10 (12) | 14 (17) | 24 (14) |
General disorders | |||
Fatigue | 30 (35) | 41 (49) | 71 (42) |
Pain | 7 (8) | 10 (12) | 17 (10) |
Pain in extremity | 14 (17) | 16 (19) | 30 (18) |
Peripheral edema | 13 (15) | 20 (24) | 33 (20) |
Pyrexia | 15 (18) | 19 (23) | 34 (20) |
Infections and infestations | |||
Pneumonia | 11 (13) | 7 (8) | 18 (11) |
Upper respiratory tract infection | 13 (15) | 13 (16) | 26 (15) |
Metabolism and nutrition disorders | |||
Anorexia | 8 (9) | 9 (11) | 17 (10) |
Hypokalemia | 12 (14) | 13 (15) | 25 (14) |
Musculoskeletal and connective tissue disorders | |||
Arthralgia | 10 (11) | 16 (19) | 26 (15) |
Asthenia | 9 (10) | 13 (16) | 22 (13) |
Back pain | 19 (22) | 20 (24) | 39 (23) |
Bone pain | 10 (12) | 11 (13) | 21 (12) |
Nervous system disorders | |||
Dizziness | 11 (13) | 10 (12) | 21 (12) |
Peripheral neuropathy | 7 (8) | 15 (18) | 22 (13) |
Psychiatric disorders | |||
Insomnia | 10 (12) | 14 (17) | 24 (14) |
Respiratory, thoracic, and mediastinal disorders | |||
Cough | 13 (15) | 15 (18) | 28 (17) |
Dyspnea | 15 (18) | 17 (20) | 32 (19) |
Skin and subcutaneous tissue disorders | |||
Rash | 9 (11) | 12 (14) | 21 (12) |
AE = adverse event; IV = intravenous
a Safety population excluding patients with protocol violations
The numbers of deaths, trial discontinuations, and treatment interruptions due to AEs were similar between the two groups as well. Deaths (9 [10.6%] 15-minute group vs 6 [7.1%] 30-minute group) were not suspected to be related to zoledronic acid. Eight patients in each treatment group discontinued therapy because of an AE; events leading to treatment discontinuation that were suspected to be related to zoledronic acid occurred in two patients in the 15-minute group (skeletal pain and ONJ) and one patient in the 30-minute group (jaw pain). AEs that required treatment interruption occurred in eight and nine patients in the 15-minute and 30-minute groups, respectively.
AEs of special interest included those related to kidney dysfunction, cardiac arrhythmias, SREs, and ONJ. The number of patients reporting overall kidney and urinary disorders was the same in the two treatment groups (14 patients in each group); however, acute renal failure was reported more frequently in patients receiving the 15-minute infusion compared with the 30-minute infusion (four patients [5%] vs one patient [1%] in 30-minute group). Details of these five patients are presented in Table 4. AEs related to cardiac rhythm occurred in 20 patients while on study; however, only one case of bradycardia was suspected to be related to zoledronic acid therapy (in the 30-minute group). The incidence of SREs at 2 years was comparable in the two groups (19% in 15-minute group vs 21% in 30-minute group). The time to onset of SREs was longer in the 15-minute group (222 vs 158 days), but this was not statistically significant. A total of 10 patients with suspected ONJ were identified, with three patients in the 15-minute group (all moderate) and seven patients in the 30-minute group (mild [n = 5], moderate [n = 1], severe [n = 1]). Six of these patients received bisphosphonates before entering the study (four patients received no prior bisphosphonates), but the length of previous bisphosphonate therapy varied (0–30 months). Patients with suspected ONJ were assessed by clinicians and referred to dental professionals for further evaluation.
PATIENT DEMOGRAPHICS | TYPE OF MM | MEDICAL HISTORY | CONCURRENT MEDICATIONSa | ACUTE RENAL FAILURE DETAILS | OUTCOME |
---|---|---|---|---|---|
Zoledronic acid 4 mg IV for 15 minutes | |||||
73-year-old female Caucasian | IgG | Anemia, cardiomyopathy, CHF, cholecystectomy, benign breast lump removal, CAD, DM, dyslipidemia, central venous catheterization, chronic renal failure, GERD, hypercholesterolemia, HTN, hysterectomy, mycobacterial infection, hemorrhoids, B-cell lymphoma, seborrheic keratosis, tonsillectomy | At start of study: aspirin, losartan, digoxin, hydrochlorothiazide/lorsartan, fluconazole, folic acid, atorvastatin, vitamins, warfarinDuring study: ethambutol dihydrochloride, moxifloxacin, rifabutin, fenofibrate, omeprazole, diuretics, nitroglycerin patch, angiotensin-converting enzyme inhibitors, hydroxyzine, loratadine, furosemide, vancomycin, pantoprozole, piperacillin/tazobactam, clarithromycin | Myeloma kidney mass consistent with myeloma kidney found during study; approximately 2 weeks later the patient developed severe infection that culminated in septic shock, with acute renal failure | Nephrologist considered renal insufficiency to be partly related to past history of large-cell lymphoma and chemotherapy; patient was discharged to hospice and died of acute renal failure secondary to myeloma |
71-year-old female Caucasian | IgA | Back pain, cholecystectomy, constipation, CAD, NIDDM, hypercholesterolemia, HTN, insomnia, left knee operation, neuralgia, obesity, osteoarthritis, hysterectomy, hypoacusis, seasonal allergies, urinary incontinence | At start of study: zolpidem, amitriptyline, loratidine, tolterodine l-tartrate, valsartan, metrotoprolol, furosemide, ibuprofen, clonazepam, gabapentin, liodcaine, hydrocodone/acetaminophen, quinine sulfate, simvastatin During study: calcium, multivitamins, lactulose, trazodone, hydromorphone, cyclobenzaprine, glipizide, macrogol, lorazepam, methadone, potassium, lisinopril, furosemide, meperidine, promethazine | Developed moderate acute renal failure on the day of her first dose; considered not associated with zoledronic acid | Renal ultrasound showed arterial stenosis; resolved approximately 1 month after diagnosis |
65-year-old male Caucasian | IgG | Oxycodone hypersensitivity, anemia, back pain, spine metastases, spinal compression fracture, depression, fatigue, inguinal hernia repair, spinal fusion (L1–L3) surgery, bilateral hip arthroplasty, pain, pneumonia, staphylococcal infection | At start of study: fluconazole, morphine sulfate, oxycodone/acetaminophen During study: naproxen, darbepoietin alfa, sodium ferrifluconate, calcium with vitamin D, cephalexin, dexamethasone, alginic acid, docusate, heparin, sodium polystyrene, levofloxacin, filgrastim, lansoprazole | After 5 doses of zoledronic acid, patient developed severe acute renal failure with elevated SCr; not suspected to be related to zoledronic acid | Resolved 9 days later following treatment with cephalexin and dexamethasone |
56-year-old female Caucasian | IgA | Osteolysis, cataract surgery, constipation, bone lesions, hypercholesterolemia, HTN, musculoskeletal pain, anorexia | At start of study: ibuprofen, oxycodone, propoxyphene/acetaminophen, hydrocodone/acetaminophen, valsartan, calcium/vitamin D, potassium chloride, docusate sodiumDuring study: vancomycin, acyclovir | Approximately 1 week after 9th zoledronic acid dose, patient developed acute renal failure with an increased SCr (12.5 mg/dL); not suspected to be related to zoledronic acid | Resulted from myeloma progression to plasma cell leukemia; emergency dialysis performed; catheter-related sepsis occurred approximately 1 month later, and patient died of sepsis and disease progression |
Zoledronic acid 4 mg IV for 30 minutes | |||||
80-year-old male African American | IgG | Anemia, arteriosclerotic heart disease, bilateral ankle swelling/pain, degenerative joint disease, dyspnea on exertion, fatigue, GERD, HTN, neutropenia, shoulder pain, vasovagal syncope | At start of study: aspirin, atenolol, multivitamin, doxazosin, fosinopril, hydrochlorothiazide, amlodipine besylate, simvastatinDuring study: darbepoietin alfa, warfarin sodium, furosemide, omeprazole, calcium carbonate | Approximately 1 month after 2nd dose, patient experienced increased SCr (2.9 mg/dL, 53% increase from baseline); relationship to zoledronic acid unknown | Discontinued from study after 2nd dose, and SCr remained elevated for 2 months following discontinuation |
CAD = coronary artery disease; CHF = congestive heart failure; DM = diabetes mellitus; GERD = gastroesophageal reflux disease; HTN = hypertension; MM = multiple myeloma; NIDDM = non-insulin-dependent diabetes mellitus; SCr = serum creatinine
Discussion
During the past decade, bisphosphonate therapy has become an important adjunctive treatment to prevent the emergence, or worsening, of SREs in patients with MM involving the bone.15 Kidney failure is a common and severe complication of MM that may be exacerbated by chronic administration of zoledronic acid.7 A study evaluating zoledronic acid in patients with cancer and bone metastases suggests that increasing the infusion time decreases the Cmax, which may result in fewer renal AEs.[9] and [12] This study was designed to assess whether prolonging the infusion time of zoledronic acid from the recommended 15 to 30 minutes would improve kidney safety in MM patients, as evidenced by fewer rises in SCr levels. To our knowledge, this is the only trial that has been designed to evaluate the impact of infusion duration on renal effects in this population.
The 12-month results of this pilot study showed a trend toward improved renal safety with the longer infusion time, this difference not being statistically significant. By 24 months, however, there were no differences in SCr level elevations between the two groups. The clinically relevant SCr increases observed in our study, however, differ from those reported by Rosen and colleagues,[5] and [6] who first evaluated zoledronic acid for patients with MM. In that study, 4%–11% of patients experienced kidney function deterioration, manifested by SCr increases, which is much lower than the rate observed in our study. However, several differences exist between our trial and the Rosen study. The Rosen study included both breast cancer patients with at least one bone metastasis and Durie-Salmon stage 3 MM patients with at least one osteolytic lesion, whereas our study only included MM patients with at least one bone lesion. Additionally, the criteria for defining a clinically relevant SCr increase differ between the two studies; therefore, one cannot directly compare the incidence of kidney dysfunction between these two studies. Although in our study the sample size was small, confidence intervals were wide, and protocol deviations did not permit a robust comparison, the results of this pilot study suggest that the longer infusion time of 30 minutes every 3–4 weeks for 2 years for MM patients with bone disease is also safe and well-tolerated.
As expected, PK data showed that the median zoledronic acid concentrations were greater in the samples obtained from the 15-minute group compared to those from the 30-minute group. This effect was observed in samples obtained both 5 minutes before the end of infusion and at the end of infusion.
Increasing the infusion time did not significantly alter the AE profile and was not associated with any new or unexpected AEs. The incidence rates of deaths, SAEs, treatment-related AEs, and overall AEs were generally comparable between treatment groups. Overall, the incidence rates of reported SREs and ONJ were as expected for this patient population, which are important factors when considering zoledronic acid for patients with MM, where the goal of ongoing monthly IV bisphosphonate therapy is to prevent the development of new SREs without increasing the risk of AEs, such as ONJ.
Finally, the FDA-approved current labeling for zoledronic acid recommends decreasing the dose of this bisphosphonate based on baseline kidney function.7 Because these recommendations were not in place at the time that this study was designed, whether the implementation of these dosing guidelines for patients with MM along with varying infusion durations would have impacted the results observed in our study cannot be ascertained.
In summary, the results of this study suggest that the safety profile of IV zoledronic acid is similar regardless of a 15-minute or a 30-minute infusion duration. However, because the study was not powered to detect statistical significance and the current renal dosing guidelines for zoledronic acid were not used in this study, large randomized studies, using current dosing recommendations, will be required to further assess the effects on kidney safety of prolonging the infusion time of ongoing monthly IV zoledronic acid therapy for patients with MM.
Acknowledgments
The authors thank Syntaxx Communications, Inc., specifically, Kristin Hennenfent, PharmD, MBA, BCPS, and Lisa Holle, PharmD, BCOP, who provided manuscript development and medical writing services, and Holly Matthews, BS, who provided editorial services, with support from Novartis Pharmaceuticals Corporation. We also thank all participating patients and study personnel. Research support was provided by Novartis Pharmaceuticals Corporation (East Hanover, NJ).
References
1 A. Jemal, R. Siegel and J. Xu et al., Cancer statistics, 2010, CA Cancer J Clin 60 (2010), pp. 277–300. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (543)
2 R.A. Kyle, M.A. Gertz and T.E. Witzig et al., Review of 1027 patients with newly diagnosed multiple myeloma, Mayo Clin Proc 78 (1) (2003), pp. 21–33. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (396)
3 A. Corso, P. Zappasodi and C. Pascutto et al., Urinary proteins in multiple myeloma: correlation with clinical parameters and diagnostic implications, Ann Hematol 82 (8) (2003), pp. 487–491. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
4 V. Eleutherakis-Papaiakovou, A. Bamias and D. Gika et al., Renal failure in multiple myeloma: incidence, correlations, and prognostic significance, Leuk Lymphoma 48 (2) (2007), pp. 337–341. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (35)
5 L.S. Rosen, D. Gordon and M. Kaminski et al., Zoledronic acid versus pamidronate in the treatment of skeletal metastases in patients with breast cancer or osteolytic lesions of multiple myeloma: a phase III, double-blind, comparative trial, Cancer J 7 (5) (2001), pp. 377–387. View Record in Scopus | Cited By in Scopus (461)
6 L.S. Rosen, D. Gordon and M. Kaminski et al., Long-term efficacy and safety of zoledronic acid compared with pamidronate disodium in the treatment of skeletal complications in patients with advanced multiple myeloma or breast carcinoma: a randomized, double-blind, multicenter, comparative trial, Cancer 98 (8) (2003), pp. 1735–1744. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (329)
7 , Zometa (package insert), Novartis Pharmaceuticals, Corporation, East Hanover, NJ (2008).
8 P. Major, A. Lortholary and J. Han et al., Zoledronic acid is superior to pamidronate in the treatment of hypercalcemia of malignancy: a pooled analysis of two randomized, controlled clinical trials, J Clin Oncol 19 (2) (2001), pp. 558–567. View Record in Scopus | Cited By in Scopus (325)
9 T. Chen, J. Berenson and R. Vescio et al., Pharmacokinetics and pharmacodynamics of zoledronic acid in cancer patients with bone metastases, J Clin Pharmacol 42 (11) (2002), pp. 1228–1236. View Record in Scopus | Cited By in Scopus (139)
10 T. Pfister, E. Atzpodien and F. Bauss, The renal effects of minimally nephrotoxic doses of ibandronate and zoledronate following single and intermittent intravenous administration in rats, Toxicology 191 (2003), pp. 159–167. Article | | View Record in Scopus | Cited By in Scopus (48)
11 T. Pfister, E. Aztpodien, B. Bohrmann and F. Bauss, Acute renal effects of intravenous bisphosphonates in the rat, Basic Clin Pharmacol Toxicol 97 (2005), pp. 374–381. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
12 F. Saad, D.M. Gleason and R. Murray et al., A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma, J Natl Cancer Inst 94 (19) (2002), pp. 1458–1468. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (650)
13 S. Kautiainen, S. Luurila, P. Ylitalo and R. Ylitalo, Transformation of bisphosphonates into insoluble material in human blood in vitro, Methods Find Exp Clin Pharmacol 20 (4) (1998), pp. 289–295. View Record in Scopus | Cited By in Scopus (5)
14 L.S. Rosen, D. Gordon and S. Tchekmedyian et al., Zoledronic acid versus placebo in the treatment of skeletal metastases in patients with lung cancer and other solid tumors: a phase III, double-blind, randomized trial—the Zoledronic Acid Lung Cancer and Other Solid Tumors Study Group, J Clin Oncol 21 (16) (2003), pp. 3150–3157. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (251)
15 M.A. Hussein, Multiple myeloma: most common end-organ damage and management, J Natl Compr Canc Netw 5 (2) (2007), pp. 170–178. View Record in Scopus | Cited By in Scopus (4)
Appendix
The following ZMAX Trial principal investigators participated in this study: Bart Barlogie, MD, Myeloma Institute For Research and Therapy; James Berenson, MD, Oncotherapeutics; Robert Bloom, MD, Providence Cancer Center, Clinical Trials Department; Ralph Boccia, MD, Center for Cancer and Blood Disorders; Donald Brooks, MD, Arizona Clinical Research Center, Inc.; Robert Brouillard, MD, Robert P. Brouillard, MD, and Delvyn Case, MD, Maine Center for Cancer Medicine and Blood Disorders, Pharmacy; Veena Charu, MD, Pacific Cancer Medical Center; Naveed Chowhan, MD, Cancer Care Center, Inc; Robert Collins, MD, University of Texas Southwestern Medical Center at Dallas; Thomas Cosgriff, MD, Hematology and Oncology Specialists, LLC; Jose Cruz, MD, Joe Arrington Cancer Research and Treatment Center; Surrinder Dang, MD, Oncology Specialties; Sheldon Davidson, MD, North Valley H/O; Tracy Dobbs, MD, Baptist Regional Cancer Center; Luke Dreisbach, MD, Desert Hematology Oncology Medical Group; Isaac Esseesse, MD, Hematology Oncology Associates of Central Brevard, Laboratory; Mark Fesen, MD, Hutchinson Clinic, PA; George Geils, Jr., MD, Charleston Hematology Oncology Associates, PA; Michael Greenhawt, MD, South Florida Oncology-Hematology; Manuel Guerra, MD, ORA; Rita Gupta, MD, Oncology-Hematology Associates, PA; Vicram Gupta, MD, Saint Joseph Oncology; Alexandre Hageboutros, MD, Cancer Institute of New Jersey at Cooper Hospital; Vincent Hansen, MD, Utah Hematology Oncology; David Henry, MD, Pennsylvania Oncology Hematology Associates; Benjamin Himpler, MD, Syracuse Hematology/Oncology PC; Winston Ho, MD, Hematology/Oncology Group of Orange County; William Horvath, MD, Haematology Oncology Associates of Ohio and Michigan, PC; Paul Hyman, MD, Hematology Oncology Associates of Western Suffolk; Min Kang, MD, Western Washington Oncology; Mark Keaton, MD, Augusta Oncology Associates, PC; Howard Kesselheim, MD, The Center for Cancer and Hematologic Disease; Kapisthalam Kumar, MD, Pasco Hernando Oncology Associates, PA; Edward Lee, MD, Maryland Oncology-Hematology, PA; André Liem, MD, Pacific Shore Medical Group; Timothy Lopez, MD, New Mexico Cancer Care Associates, Cancer Institute of New Mexico; Paul Michael, MD, Comprehensive Cancer Centers of Nevada; Michael Milder, MD, Swedish Cancer Institute; Barry Mirtsching, MD, Center for Oncology Research & Treatment, PA; Ruben Niesvizky, MD, New York Presbyterian Hospital; Jorge Otoya, MD, Osceola Cancer Center; Joseph Pascuzzo, MD, California Oncology of the Central Valley; Ravi Patel, MD, Comprehensive Blood and Cancer Center Lab; Allen Patton, MD, Hematology Oncology Associates, PA; Kelly Pendergrass, MD, Kansas City Cancer Center, LLC; Anthony Phillips, MD, Fox Valley Hematolgy Oncology, SC; Robert Raju, MD, Dayton Oncology and Hematology, PA; Harry Ramsey, MD, Berks Hematology Oncology Associates; Ritesh Rathore, MD, Roger Williams Hospital Medical Center; Phillip Reid, MD, Central Jersey Oncology Center; Robert Robles, MD, Bay Area Cancer Research Group, LLC; Stephen Rosenoff, MD, Oncology and Hematology Associates of Southwest Virginia, Inc; Martin Rubenstein, MD, Southbay Oncology Hematology Partners; Mansoor Saleh, MD, Georgia Cancer Specialists; Sundaresan Sambandam, MD, Hematology and Oncology Associates of RI; Mukund Shah, MD, Antelope Valley Cancer Center; David Siegel, MD, Hackensack University Medical Center; Nelida Sjak-Shie, MD, The Center for Cancer Care and Research; Michael Stone, MD, Greeley Medical Clinic; Stefano Tarantolo, MD, Nebraska Methodist Hospital; Joseph Volk, MD, Palo Verde Hematology Oncology, Ltd; Mitchell Weisberg, MD, MetCare Oncology; Ann Wierman, MD, Nevada Cancer Center; Donald Woytowitz, Jr., MD, Florida Cancer Specialists; Peter Yu, MD, Camino Medical Group.
Correspondence to: James R. Berenson, MD, Institute for Myeloma & Bone Cancer Research, 9201 West Sunset Boulevard, Suite 300, West Hollywood, CA 90069; telephone: (310) 623–1214; fax: (310) 623–1120
Original research
James R. Berenson MD, a,
Abstract
Zoledronic acid, an intravenous (IV) bisphosphonate, is a standard treatment for multiple myeloma (MM) but may exacerbate preexisting renal dysfunction. The incidence of zoledronic acid–induced renal dysfunction may correlate with infusion duration. In this randomized, multicenter, open-label study, 176 patients with MM, at least one bone lesion, and stable renal function with a serum creatinine (SCr) level <3 mg/dL received zoledronic acid 4 mg (in 250 mL) as a 15- or 30-minute IV infusion every 3–4 weeks. At month 12, 20% (17 patients) in the 15-minute and 16% (13 patients) in the 30-minute arm experienced a clinically relevant but nonsignificant SCr-level increase (P = 0.44). By 24 months, the proportion of patients with a clinically relevant SCr-level increase was similar between arms (15-minute 28% [24 patients] vs 30-minute 27% [23 patients], P = 0.9014). Median zoledronic acid end-of-infusion concentrations were higher with the shorter infusion (15-minute 249 ng/mL vs 30-minute 172 ng/mL), and prolonging the infusion beyond 15 minutes did not influence adverse events related to zoledronic acid. For patients with MM, the safety profile of IV zoledronic acid is similar between those receiving a 15- or 30-minute infusion; therefore, determining the appropriate infusion duration of zoledronic acid should be based on individual patient considerations.
Article Outline
Considerable research has focused on preventive and/or treatment strategies to reduce bone complications in MM patients. In a large, international, randomized, phase III trial of MM patients with at least one osteolytic bone lesion, zoledronic acid (Zometa), a potent intravenous (IV) bisphosphonate that inhibits osteoclast-mediated bone resorption, reduced the overall risk of developing skeletally related events (SREs) including HCM by 16% (P = 0.03) compared with standard-dose pamidronate 90 mg (Aredia), another less potent IV bisphosphonate.[5] and [6] As a result of this study and others, monthly infusion of zoledronic acid at 4 mg over at least 15 minutes has become a common treatment for MM patients with bone involvement.
The U.S. Food and Drug Administration (FDA) has approved zoledronic acid use for patients with MM, documented bone metastases from solid tumors, or HCM.[5], [6], [7] and [8] The FDA-approved dose for MM patients is 4 mg administered as an IV infusion over at least 15 minutes every 3–4 weeks for patients with a creatinine clearance (CrCl) of >60 mL/min; when treating HCM, zoledronic acid 4 mg is administered as a single IV infusion.[5], [6], [7] and [8]
Zoledronic acid is primarily excreted intact through the kidney.9 Preexisting kidney disease and receipt of multiple cycles of bisphosphonate therapy are risk factors for subsequent kidney injury.7 In animal studies, IV bisphosphonates have been shown by histology to precipitate renal tubular injury when administered as a single high dose or when administered more frequently at lower doses.[10] and [11] Additionally, renal dysfunction, as evidenced by increased serum creatinine (SCr) levels, was reported among patients treated at a dose of 4 mg with an infusion time of 5 minutes.[7] and [12] When 4 mg zoledronic acid was administered with a longer infusion time of 15 minutes in large randomized trials, no significant difference between the renal safety profiles of zoledronic acid and pamidronate was reported.6
One hypothesis about the development of kidney injury associated with zoledronic acid is that it may be related to the peak plasma concentration as determined by infusion time. Results of a study evaluating patients with MM or other cancer types and bone metastases demonstrated that prolonging the infusion time of zoledronic acid reduced the end-of-infusion peak plasma concentration (Cmax) by 35%.9 Another theory about the development of kidney dysfunction is that insoluble precipitates may form when the blood is exposed to high concentrations of bisphosphonates as this has been shown to occur in vitro.[9] and [13] Therefore, the current management of renal adverse events (AEs) related to IV bisphosphonates is based on these theories so that reducing the peak plasma concentration of zoledronic acid may prevent the possible formation of insoluble precipitates through (1) lowering the dose, (2) slowing the infusion rate, or (3) increasing the volume of infusate.[5], [12] and [14]
Because MM patients are predisposed to experience deterioration of renal function, it is critical to ensure that zoledronic acid does not contribute to, or exacerbate, a decline in kidney function. To determine if increasing the duration of zoledronic acid infusion further results in improved renal safety, a multicenter, open-label, randomized study was designed to compare a 15-minute vs a 30-minute infusion time with an increased volume of infusate from 100 to 250 mL administered every 3–4 weeks to MM patients with osteolytic bone disease.
Patients and Methods
Patient Population
Men and women (≥18 years of age) with a diagnosis of MM, at least one bone lesion on plain film radiographs, stable kidney function (defined as two SCr level determinations of <3 mg/dL obtained at least 7 days apart during the screening period), calculated CrCl of at least 30 mL/min, Eastern Cooperative Oncology Group (ECOG) performance status of 1 or less, and a life expectancy of at least 9 months were eligible. The study excluded patients with prolonged IV bisphosphonate use (defined as use of zoledronic acid longer than 3 years or pamidronate longer than 1 year [total bisphosphonate duration could not exceed 3 years]), corrected serum calcium level at first visit of <8 or ≥12 mg/dL, or diagnosis of amyloidosis. Additionally, patients who had known hypersensitivity to zoledronic acid or other bisphosphonates; were pregnant or lactating; had uncontrolled cardiovascular disease, hypertension, or type 2 diabetes mellitus; or had a history of noncompliance with medical regimens were not eligible.
Study Design
This open-label, randomized pilot study was conducted at 45 centers in the United States. Before randomization, patients were stratified based on length of time of prior bisphosphonate treatment (bisphosphonate-naive vs ≤1 year prior bisphosphonate therapy vs >1 year prior bisphosphonate therapy) and baseline calculated CrCl (>75 vs >60–75 vs ≥30–≤60 mL/min).
Treatment and Evaluation
Patients were randomized to receive zoledronic acid 4 mg as either a 15- or a 30-minute IV infusion. The volume of infusate was increased from the standard 100 to 250 mL to provide additional hydration; infusions were administered every 3–4 weeks for up to 24 months. At the time this study was developed, the 4 mg dose was used because the dose adjustments for renal dysfunction in the current FDA labeling for zoledronic acid were not yet available.7 Patients were required to take a calcium supplement containing 500 mg of calcium and a multivitamin containing 400–500 IU of vitamin D, orally, once daily, for the duration of zoledronic acid therapy.
HCM during the trial was defined as a corrected serum calcium level ≥12 mg/dL or a lower level of hypercalcemia accompanied by symptoms and/or requiring active treatment other than rehydration. If HCM occurred more than 14 days after a zoledronic acid infusion, patients could receive a zoledronic acid infusion as treatment for HCM, even if this required administration before the next scheduled dose. Patients were allowed to remain in the study provided that HCM did not persist or recur. However, zoledronic acid treatment was immediately discontinued if patients developed HCM ≤14 days after study drug infusion; these patients received HCM treatment at the discretion of their treating physician. Also, patients experiencing HCM discontinued calcium and vitamin D supplements.
Within 2 weeks before each dose, enrolled patients were assessed for increase in SCr levels. For patients experiencing a clinically relevant increase in SCr level (defined as a rise of 0.5 mg/dL or more or a doubling of baseline SCr levels), administration of zoledronic acid was suspended until the SCr level fell to within 10% of the baseline value. During the delay, SCr levels were monitored at each regularly scheduled study visit (every 3–4 weeks) or more frequently if deemed necessary by the investigator. If the SCr level fell to within 10% of the baseline value within the subsequent 12 weeks, zoledronic acid was restarted with an infusion time that was increased by 15 minutes over the starting duration. If the rise in SCr level did not resolve within 12 weeks or if the patient experienced a second clinically relevant increase in SCr level after modification of the infusion time, treatment was permanently discontinued. Otherwise, patients were followed for 24 months. A final safety assessment, including a full hematology and chemistry profile, was performed 28 days after the last infusion.
A pretreatment dental examination with appropriate preventive dentistry was suggested for all patients with known risk factors for the development of osteonecrosis of the jaw (ONJ) (eg, cancer chemotherapy, corticosteroids, poor oral hygiene, dental extraction, or dental implants). Throughout the study, patients reporting symptoms that could be consistent with ONJ were referred to a dental professional for assessment; if exposed bone was noted on dental examination, the patient was referred to an oral surgeon for further evaluation, diagnosis, and treatment. A diagnosis of ONJ required cessation of zoledronic acid therapy and study discontinuation.
Pharmacokinetic Sampling
At the first infusion visit (visit 2), pharmacokinetic (PK) parameters were measured. If PK samples were not obtained at visit 2, they could be obtained at visit 3 (otherwise, they were recorded as not done). All blood samples for PK analysis were drawn from the contralateral arm. For patients receiving the 15-minute zoledronic acid infusion, the protocol specified that PK samples were to be drawn at exactly 10 and 15 minutes from the start of the infusion; patients receiving the 30-minute zoledronic acid infusion were to have blood samples drawn at exactly 25 and 30 minutes from the start of the infusion. The second blood sample for PK analysis was taken before the study drug infusion was stopped in both groups. PK analysis was performed by Novartis Pharmaceuticals Corporation Drug Metabolism and Pharmacokinetics France (Rueil-Malmaison, France) and SGS Cephac (Geneva Switzerland), using a competitive radioimmunoassay that has a lower limit of quantification of 0.04 ng/mL and an upper limit of quantification of 40 ng/mL.
Statistical Analysis
The primary study end point was the proportion of patients with a clinically relevant increase in SCr level at 12 months. Descriptive statistics were used to summarize the primary end point; in addition, an exploratory analysis with a logistic regression model, using treatment group, prior bisphosphonate therapy, and baseline CrCl, was performed.
Additional secondary safety end points included the proportion of patients with a clinically relevant increase in SCr level at 24 months, time to first clinically relevant increase in SCr level, and the PK profile of zoledronic acid. The proportion of patients with a clinically relevant increase in SCr level at 24 months was summarized using descriptive statistics. Time to first clinically relevant increase in SCr level was analyzed using the Kaplan-Meier method at the time of the primary analysis (12 months) and at 24 months. Plasma concentration data were evaluated by treatment group and baseline kidney function using descriptive statistics. Continuous variables of baseline and demographic characteristics between treatment groups were compared using a two-sample t-test; between-group differences in discrete variables were analyzed using Pearson's chi-squared test.
The primary analysis included all randomized patients who received at least one zoledronic acid infusion and who had valid postbaseline data for assessment. All study subjects who had evaluable PK parameters were included in a secondary PK analysis. Efficacy assessments were not included in this trial.
This pilot trial was designed to obtain additional preliminary data to support the hypothesis that a longer infusion is associated with less kidney dysfunction than a shorter infusion; therefore, a sample size of 90 patients per treatment group was selected. All statistical tests employed a significance level of 0.05 against a two-sided alternative hypothesis.
The institutional review boards of participating institutions approved the study, and all patients provided written informed consent before study entry.
Results
Study Population
Between October 2004 and October 2007, 179 MM patients with SCr <3 mg/dL were randomized to receive either a 15- or a 30-minute infusion of zoledronic acid. Of these, 176 patients (88 in each group) received at least one dose of study drug. Because of protocol violations, postbaseline data from one site were excluded from analyses, leaving 85 assessable patients in the 15-minute group and 84 patients in the 30-minute group.
Overall, the study groups were representative of a general population with MM. About two-thirds of patients had received prior bisphosphonate therapy; the duration of therapy was greater than 1 year for most of these patients (Table 1). The most common concomitant therapies included dexamethasone, thalidomide, and melphalan. Although the median age, proportion of patients who were 65 years of age or older, and ratio of men to women were greater in the 15-minute infusion group, none of the differences in baseline demographics was statistically significant. All other baseline demographics and disease characteristics, including prior bisphosphonate use and baseline CrCl values, were similar between the two groups (see Table 1). During the study, six patients in the 15-minute treatment group and one patient in the 30-minute treatment group experienced HCM. Three of the six patients in the 15-minute treatment group and one patient in the 30-minute treatment group discontinued the study as a result of HCM.
NUMBER OF PATIENTS (%)a | ||
---|---|---|
CHARACTERISTIC | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 88)b | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 88)b |
Age (years) | ||
Mean (SD) | 64 | 64 |
Median | 66 | 64 |
Range | 37–91 | 27–86 |
Age category (years) | ||
<65 | 39 (44) | 47 (53) |
≥65 | 49 (56) | 41 (47) |
Sex | ||
Male | 56 (64) | 49 (56) |
Female | 32 (36) | 39 (44) |
Race | ||
White | 70 (80) | 69 (78) |
Black | 9 (10) | 13 (15) |
Asian | 1 (1) | 1 (1) |
Other | 8 (9) | 5 (6) |
Time since diagnosis (months) | ||
Mean (SD) | 12 (24) (n = 86) | 10 (14) (n = 87) |
Median | 4 | 6 |
Range | 0–186 | 0–98c |
Prior bisphosphonate use | ||
Naive | 28 (32) | 28 (32) |
≤1 year | 12 (14) | 14 (16) |
>1 year | 48 (55) | 39 (44) |
Missing | 0 (0) | 7 (8) |
Calculated CrCl (mL/min) | ||
Mean (SD) | 87 (33) | 89 (40) |
Median | 84 | 83 |
Range | 33–210 | 31–224 |
Calculated CrCl category (mL/min) | ||
CrCl ≥75 | 54 (61) | 49 (56) |
60 < CrCL < 75 | 13 (15) | 15 (17) |
30 < CrCl ≤ 60 | 21 (24) | 24 (27) |
CrCl <30 | 0 (0) | 0 (0) |
CrCl = creatinine clearance; IV = intravenous; SD = standard deviation
a Unless otherwise notedb Safety populationc One patient had a screening visit date before the date of initial diagnosis
Protocol violations and/or deviations (n = 658) occurred during this study, affecting 139 patients. The types of protocol violations/deviations were related to protocol adherence (n = 404), timing of visits (n = 210), protocol adherence/timing of visits (n = 2), exclusion criteria (n = 22), inclusion criteria (n = 10), and informed consent (n = 1); 9 violations were unclassified. Notably, one protocol adherence deviation that occurred was incorrect infusion duration despite the patient having a stable SCr level. In the 15-minute treatment group, 15% of infusions administered were longer than 15 minutes. Among the longer infusions, 7% of the infusions correctly occurred per protocol following an SCr-level increase, whereas 7% of the prolonged infusions were 20 minutes or longer in the absence of an SCr-level increase. Similarly, in the 30-minute treatment group, 5% of patients received infusions lasting at least 35 minutes in the absence of an SCr-level increase.
Renal Safety
At 12 months, slightly fewer patients (n = 13 [16%]) in the 30-minute infusion group had a clinically relevant increase in SCr level than in the 15-minute infusion group (n = 17 [20%]); but this difference was not statistically significant, and for approximately 35% of patients in each group there were no SCr data available (Table 2). The median time to a clinically relevant increase in SCr by Kaplain-Meier was not reached in either group (data not shown). Neither previous bisphosphonate use nor baseline CrCl significantly affected the results (P = 0.5837 and P = 0.9371, respectively).
NUMBER OF PATIENTS (%) | |||
---|---|---|---|
CLINICALLY RELEVANT INCREASE IN SCR | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 85)a | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 84)a | P VALUEb |
12 Months | 0.6892 | ||
Yes | 17 (20) | 13 (16) | |
No | 38 (45) | 42 (50) | |
Unknown | 30 (35) | 29 (35) | |
24 Months | 0.9750 | ||
Yes | 24 (28) | 23 (27) | |
No | 22 (26) | 23 (27) | |
Unknown | 39 (46) | 38 (45) |
CI = confidence interval; IV = intravenous; SCr = serum creatinine
a Safety population, excluding patients with protocol violationsb P value calculated based on chi-squared test
After 24 months of treatment, the proportion of patients experiencing a clinically relevant increase in SCr level was similar between treatment groups, although for approximately 45% of patients in each group there were no SCr data available (see Table 2). Moreover, the difference in time to first clinically relevant increase in SCr level was not statistically significant between the two groups (P = 0.55) (Figure 1). However, among patients with a clinically significant rise in SCr level, the median time to SCr rise was slightly longer in the 30-minute group than in the 15-minute group (22 vs 24 weeks), but this was not statistically significant.
Increases in SCr relative to baseline led to treatment discontinuation in 20 patients (24%) receiving a 15-minute infusion and 14 patients (17%) receiving a 30-minute infusion. In these cases, the treating physician either considered the SCr level too high for continued treatment or the SCr level was persistently high despite treatment interruption.
Pharmacokinetics
Median zoledronic acid concentrations, as anticipated, were higher with the 15-minute infusion time at both sampling time points (during infusion: 15-minute group 231 ng/mL [at 10 minutes] vs 30-minute group 186 ng/mL [at 25 minutes]; end-of-infusion: 15-minute group, 249 ng/mL vs 30-minute group 172 ng/mL).
Adverse Events
Overall, the incidence and severity of AEs were as anticipated for MM patients. The most commonly reported AEs included fatigue, anemia, nausea, constipation, and back pain (Table 3). Although many AEs were reported more frequently in the 30-minute infusion group, the incidence rates of AEs suspected to be related to zoledronic acid were similar between the two groups. Toxicities were graded as mild, moderate, or severe; proportions of AEs categorized by these grades were comparable. Nonfatal serious AEs (SAEs) occurred in 26% of patients receiving the 15-minute infusion and 35% of patients receiving the 30-minute infusion; however, only one patient in the 15-minute group and two patients in the 30-minute group had SAEs suspected to be related to study medication.
NUMBER OF PATIENTS (%) | |||
---|---|---|---|
TYPE OF AE | ZOLEDRONIC ACID 4 MG IV FOR 15 MINUTES (N = 85) | ZOLEDRONIC ACID 4 MG IV FOR 30 MINUTES (N = 84) | TOTAL (N = 169) |
Blood and lymphatic system disorders | |||
Anemia | 19 (22) | 27 (32) | 46 (27) |
Neutropenia | 6 (7) | 12 (14) | 18 (11) |
Gastrointestinal disorders | |||
Constipation | 20 (24) | 21 (25) | 41 (24) |
Diarrhea | 14 (17) | 20 (24) | 34 (20) |
Nausea | 18 (21) | 27 (32) | 45 (27) |
Vomiting | 10 (12) | 14 (17) | 24 (14) |
General disorders | |||
Fatigue | 30 (35) | 41 (49) | 71 (42) |
Pain | 7 (8) | 10 (12) | 17 (10) |
Pain in extremity | 14 (17) | 16 (19) | 30 (18) |
Peripheral edema | 13 (15) | 20 (24) | 33 (20) |
Pyrexia | 15 (18) | 19 (23) | 34 (20) |
Infections and infestations | |||
Pneumonia | 11 (13) | 7 (8) | 18 (11) |
Upper respiratory tract infection | 13 (15) | 13 (16) | 26 (15) |
Metabolism and nutrition disorders | |||
Anorexia | 8 (9) | 9 (11) | 17 (10) |
Hypokalemia | 12 (14) | 13 (15) | 25 (14) |
Musculoskeletal and connective tissue disorders | |||
Arthralgia | 10 (11) | 16 (19) | 26 (15) |
Asthenia | 9 (10) | 13 (16) | 22 (13) |
Back pain | 19 (22) | 20 (24) | 39 (23) |
Bone pain | 10 (12) | 11 (13) | 21 (12) |
Nervous system disorders | |||
Dizziness | 11 (13) | 10 (12) | 21 (12) |
Peripheral neuropathy | 7 (8) | 15 (18) | 22 (13) |
Psychiatric disorders | |||
Insomnia | 10 (12) | 14 (17) | 24 (14) |
Respiratory, thoracic, and mediastinal disorders | |||
Cough | 13 (15) | 15 (18) | 28 (17) |
Dyspnea | 15 (18) | 17 (20) | 32 (19) |
Skin and subcutaneous tissue disorders | |||
Rash | 9 (11) | 12 (14) | 21 (12) |
AE = adverse event; IV = intravenous
a Safety population excluding patients with protocol violations
The numbers of deaths, trial discontinuations, and treatment interruptions due to AEs were similar between the two groups as well. Deaths (9 [10.6%] 15-minute group vs 6 [7.1%] 30-minute group) were not suspected to be related to zoledronic acid. Eight patients in each treatment group discontinued therapy because of an AE; events leading to treatment discontinuation that were suspected to be related to zoledronic acid occurred in two patients in the 15-minute group (skeletal pain and ONJ) and one patient in the 30-minute group (jaw pain). AEs that required treatment interruption occurred in eight and nine patients in the 15-minute and 30-minute groups, respectively.
AEs of special interest included those related to kidney dysfunction, cardiac arrhythmias, SREs, and ONJ. The number of patients reporting overall kidney and urinary disorders was the same in the two treatment groups (14 patients in each group); however, acute renal failure was reported more frequently in patients receiving the 15-minute infusion compared with the 30-minute infusion (four patients [5%] vs one patient [1%] in 30-minute group). Details of these five patients are presented in Table 4. AEs related to cardiac rhythm occurred in 20 patients while on study; however, only one case of bradycardia was suspected to be related to zoledronic acid therapy (in the 30-minute group). The incidence of SREs at 2 years was comparable in the two groups (19% in 15-minute group vs 21% in 30-minute group). The time to onset of SREs was longer in the 15-minute group (222 vs 158 days), but this was not statistically significant. A total of 10 patients with suspected ONJ were identified, with three patients in the 15-minute group (all moderate) and seven patients in the 30-minute group (mild [n = 5], moderate [n = 1], severe [n = 1]). Six of these patients received bisphosphonates before entering the study (four patients received no prior bisphosphonates), but the length of previous bisphosphonate therapy varied (0–30 months). Patients with suspected ONJ were assessed by clinicians and referred to dental professionals for further evaluation.
PATIENT DEMOGRAPHICS | TYPE OF MM | MEDICAL HISTORY | CONCURRENT MEDICATIONSa | ACUTE RENAL FAILURE DETAILS | OUTCOME |
---|---|---|---|---|---|
Zoledronic acid 4 mg IV for 15 minutes | |||||
73-year-old female Caucasian | IgG | Anemia, cardiomyopathy, CHF, cholecystectomy, benign breast lump removal, CAD, DM, dyslipidemia, central venous catheterization, chronic renal failure, GERD, hypercholesterolemia, HTN, hysterectomy, mycobacterial infection, hemorrhoids, B-cell lymphoma, seborrheic keratosis, tonsillectomy | At start of study: aspirin, losartan, digoxin, hydrochlorothiazide/lorsartan, fluconazole, folic acid, atorvastatin, vitamins, warfarinDuring study: ethambutol dihydrochloride, moxifloxacin, rifabutin, fenofibrate, omeprazole, diuretics, nitroglycerin patch, angiotensin-converting enzyme inhibitors, hydroxyzine, loratadine, furosemide, vancomycin, pantoprozole, piperacillin/tazobactam, clarithromycin | Myeloma kidney mass consistent with myeloma kidney found during study; approximately 2 weeks later the patient developed severe infection that culminated in septic shock, with acute renal failure | Nephrologist considered renal insufficiency to be partly related to past history of large-cell lymphoma and chemotherapy; patient was discharged to hospice and died of acute renal failure secondary to myeloma |
71-year-old female Caucasian | IgA | Back pain, cholecystectomy, constipation, CAD, NIDDM, hypercholesterolemia, HTN, insomnia, left knee operation, neuralgia, obesity, osteoarthritis, hysterectomy, hypoacusis, seasonal allergies, urinary incontinence | At start of study: zolpidem, amitriptyline, loratidine, tolterodine l-tartrate, valsartan, metrotoprolol, furosemide, ibuprofen, clonazepam, gabapentin, liodcaine, hydrocodone/acetaminophen, quinine sulfate, simvastatin During study: calcium, multivitamins, lactulose, trazodone, hydromorphone, cyclobenzaprine, glipizide, macrogol, lorazepam, methadone, potassium, lisinopril, furosemide, meperidine, promethazine | Developed moderate acute renal failure on the day of her first dose; considered not associated with zoledronic acid | Renal ultrasound showed arterial stenosis; resolved approximately 1 month after diagnosis |
65-year-old male Caucasian | IgG | Oxycodone hypersensitivity, anemia, back pain, spine metastases, spinal compression fracture, depression, fatigue, inguinal hernia repair, spinal fusion (L1–L3) surgery, bilateral hip arthroplasty, pain, pneumonia, staphylococcal infection | At start of study: fluconazole, morphine sulfate, oxycodone/acetaminophen During study: naproxen, darbepoietin alfa, sodium ferrifluconate, calcium with vitamin D, cephalexin, dexamethasone, alginic acid, docusate, heparin, sodium polystyrene, levofloxacin, filgrastim, lansoprazole | After 5 doses of zoledronic acid, patient developed severe acute renal failure with elevated SCr; not suspected to be related to zoledronic acid | Resolved 9 days later following treatment with cephalexin and dexamethasone |
56-year-old female Caucasian | IgA | Osteolysis, cataract surgery, constipation, bone lesions, hypercholesterolemia, HTN, musculoskeletal pain, anorexia | At start of study: ibuprofen, oxycodone, propoxyphene/acetaminophen, hydrocodone/acetaminophen, valsartan, calcium/vitamin D, potassium chloride, docusate sodiumDuring study: vancomycin, acyclovir | Approximately 1 week after 9th zoledronic acid dose, patient developed acute renal failure with an increased SCr (12.5 mg/dL); not suspected to be related to zoledronic acid | Resulted from myeloma progression to plasma cell leukemia; emergency dialysis performed; catheter-related sepsis occurred approximately 1 month later, and patient died of sepsis and disease progression |
Zoledronic acid 4 mg IV for 30 minutes | |||||
80-year-old male African American | IgG | Anemia, arteriosclerotic heart disease, bilateral ankle swelling/pain, degenerative joint disease, dyspnea on exertion, fatigue, GERD, HTN, neutropenia, shoulder pain, vasovagal syncope | At start of study: aspirin, atenolol, multivitamin, doxazosin, fosinopril, hydrochlorothiazide, amlodipine besylate, simvastatinDuring study: darbepoietin alfa, warfarin sodium, furosemide, omeprazole, calcium carbonate | Approximately 1 month after 2nd dose, patient experienced increased SCr (2.9 mg/dL, 53% increase from baseline); relationship to zoledronic acid unknown | Discontinued from study after 2nd dose, and SCr remained elevated for 2 months following discontinuation |
CAD = coronary artery disease; CHF = congestive heart failure; DM = diabetes mellitus; GERD = gastroesophageal reflux disease; HTN = hypertension; MM = multiple myeloma; NIDDM = non-insulin-dependent diabetes mellitus; SCr = serum creatinine
Discussion
During the past decade, bisphosphonate therapy has become an important adjunctive treatment to prevent the emergence, or worsening, of SREs in patients with MM involving the bone.15 Kidney failure is a common and severe complication of MM that may be exacerbated by chronic administration of zoledronic acid.7 A study evaluating zoledronic acid in patients with cancer and bone metastases suggests that increasing the infusion time decreases the Cmax, which may result in fewer renal AEs.[9] and [12] This study was designed to assess whether prolonging the infusion time of zoledronic acid from the recommended 15 to 30 minutes would improve kidney safety in MM patients, as evidenced by fewer rises in SCr levels. To our knowledge, this is the only trial that has been designed to evaluate the impact of infusion duration on renal effects in this population.
The 12-month results of this pilot study showed a trend toward improved renal safety with the longer infusion time, this difference not being statistically significant. By 24 months, however, there were no differences in SCr level elevations between the two groups. The clinically relevant SCr increases observed in our study, however, differ from those reported by Rosen and colleagues,[5] and [6] who first evaluated zoledronic acid for patients with MM. In that study, 4%–11% of patients experienced kidney function deterioration, manifested by SCr increases, which is much lower than the rate observed in our study. However, several differences exist between our trial and the Rosen study. The Rosen study included both breast cancer patients with at least one bone metastasis and Durie-Salmon stage 3 MM patients with at least one osteolytic lesion, whereas our study only included MM patients with at least one bone lesion. Additionally, the criteria for defining a clinically relevant SCr increase differ between the two studies; therefore, one cannot directly compare the incidence of kidney dysfunction between these two studies. Although in our study the sample size was small, confidence intervals were wide, and protocol deviations did not permit a robust comparison, the results of this pilot study suggest that the longer infusion time of 30 minutes every 3–4 weeks for 2 years for MM patients with bone disease is also safe and well-tolerated.
As expected, PK data showed that the median zoledronic acid concentrations were greater in the samples obtained from the 15-minute group compared to those from the 30-minute group. This effect was observed in samples obtained both 5 minutes before the end of infusion and at the end of infusion.
Increasing the infusion time did not significantly alter the AE profile and was not associated with any new or unexpected AEs. The incidence rates of deaths, SAEs, treatment-related AEs, and overall AEs were generally comparable between treatment groups. Overall, the incidence rates of reported SREs and ONJ were as expected for this patient population, which are important factors when considering zoledronic acid for patients with MM, where the goal of ongoing monthly IV bisphosphonate therapy is to prevent the development of new SREs without increasing the risk of AEs, such as ONJ.
Finally, the FDA-approved current labeling for zoledronic acid recommends decreasing the dose of this bisphosphonate based on baseline kidney function.7 Because these recommendations were not in place at the time that this study was designed, whether the implementation of these dosing guidelines for patients with MM along with varying infusion durations would have impacted the results observed in our study cannot be ascertained.
In summary, the results of this study suggest that the safety profile of IV zoledronic acid is similar regardless of a 15-minute or a 30-minute infusion duration. However, because the study was not powered to detect statistical significance and the current renal dosing guidelines for zoledronic acid were not used in this study, large randomized studies, using current dosing recommendations, will be required to further assess the effects on kidney safety of prolonging the infusion time of ongoing monthly IV zoledronic acid therapy for patients with MM.
Acknowledgments
The authors thank Syntaxx Communications, Inc., specifically, Kristin Hennenfent, PharmD, MBA, BCPS, and Lisa Holle, PharmD, BCOP, who provided manuscript development and medical writing services, and Holly Matthews, BS, who provided editorial services, with support from Novartis Pharmaceuticals Corporation. We also thank all participating patients and study personnel. Research support was provided by Novartis Pharmaceuticals Corporation (East Hanover, NJ).
References
1 A. Jemal, R. Siegel and J. Xu et al., Cancer statistics, 2010, CA Cancer J Clin 60 (2010), pp. 277–300. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (543)
2 R.A. Kyle, M.A. Gertz and T.E. Witzig et al., Review of 1027 patients with newly diagnosed multiple myeloma, Mayo Clin Proc 78 (1) (2003), pp. 21–33. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (396)
3 A. Corso, P. Zappasodi and C. Pascutto et al., Urinary proteins in multiple myeloma: correlation with clinical parameters and diagnostic implications, Ann Hematol 82 (8) (2003), pp. 487–491. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
4 V. Eleutherakis-Papaiakovou, A. Bamias and D. Gika et al., Renal failure in multiple myeloma: incidence, correlations, and prognostic significance, Leuk Lymphoma 48 (2) (2007), pp. 337–341. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (35)
5 L.S. Rosen, D. Gordon and M. Kaminski et al., Zoledronic acid versus pamidronate in the treatment of skeletal metastases in patients with breast cancer or osteolytic lesions of multiple myeloma: a phase III, double-blind, comparative trial, Cancer J 7 (5) (2001), pp. 377–387. View Record in Scopus | Cited By in Scopus (461)
6 L.S. Rosen, D. Gordon and M. Kaminski et al., Long-term efficacy and safety of zoledronic acid compared with pamidronate disodium in the treatment of skeletal complications in patients with advanced multiple myeloma or breast carcinoma: a randomized, double-blind, multicenter, comparative trial, Cancer 98 (8) (2003), pp. 1735–1744. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (329)
7 , Zometa (package insert), Novartis Pharmaceuticals, Corporation, East Hanover, NJ (2008).
8 P. Major, A. Lortholary and J. Han et al., Zoledronic acid is superior to pamidronate in the treatment of hypercalcemia of malignancy: a pooled analysis of two randomized, controlled clinical trials, J Clin Oncol 19 (2) (2001), pp. 558–567. View Record in Scopus | Cited By in Scopus (325)
9 T. Chen, J. Berenson and R. Vescio et al., Pharmacokinetics and pharmacodynamics of zoledronic acid in cancer patients with bone metastases, J Clin Pharmacol 42 (11) (2002), pp. 1228–1236. View Record in Scopus | Cited By in Scopus (139)
10 T. Pfister, E. Atzpodien and F. Bauss, The renal effects of minimally nephrotoxic doses of ibandronate and zoledronate following single and intermittent intravenous administration in rats, Toxicology 191 (2003), pp. 159–167. Article | | View Record in Scopus | Cited By in Scopus (48)
11 T. Pfister, E. Aztpodien, B. Bohrmann and F. Bauss, Acute renal effects of intravenous bisphosphonates in the rat, Basic Clin Pharmacol Toxicol 97 (2005), pp. 374–381. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
12 F. Saad, D.M. Gleason and R. Murray et al., A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma, J Natl Cancer Inst 94 (19) (2002), pp. 1458–1468. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (650)
13 S. Kautiainen, S. Luurila, P. Ylitalo and R. Ylitalo, Transformation of bisphosphonates into insoluble material in human blood in vitro, Methods Find Exp Clin Pharmacol 20 (4) (1998), pp. 289–295. View Record in Scopus | Cited By in Scopus (5)
14 L.S. Rosen, D. Gordon and S. Tchekmedyian et al., Zoledronic acid versus placebo in the treatment of skeletal metastases in patients with lung cancer and other solid tumors: a phase III, double-blind, randomized trial—the Zoledronic Acid Lung Cancer and Other Solid Tumors Study Group, J Clin Oncol 21 (16) (2003), pp. 3150–3157. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (251)
15 M.A. Hussein, Multiple myeloma: most common end-organ damage and management, J Natl Compr Canc Netw 5 (2) (2007), pp. 170–178. View Record in Scopus | Cited By in Scopus (4)
Appendix
The following ZMAX Trial principal investigators participated in this study: Bart Barlogie, MD, Myeloma Institute For Research and Therapy; James Berenson, MD, Oncotherapeutics; Robert Bloom, MD, Providence Cancer Center, Clinical Trials Department; Ralph Boccia, MD, Center for Cancer and Blood Disorders; Donald Brooks, MD, Arizona Clinical Research Center, Inc.; Robert Brouillard, MD, Robert P. Brouillard, MD, and Delvyn Case, MD, Maine Center for Cancer Medicine and Blood Disorders, Pharmacy; Veena Charu, MD, Pacific Cancer Medical Center; Naveed Chowhan, MD, Cancer Care Center, Inc; Robert Collins, MD, University of Texas Southwestern Medical Center at Dallas; Thomas Cosgriff, MD, Hematology and Oncology Specialists, LLC; Jose Cruz, MD, Joe Arrington Cancer Research and Treatment Center; Surrinder Dang, MD, Oncology Specialties; Sheldon Davidson, MD, North Valley H/O; Tracy Dobbs, MD, Baptist Regional Cancer Center; Luke Dreisbach, MD, Desert Hematology Oncology Medical Group; Isaac Esseesse, MD, Hematology Oncology Associates of Central Brevard, Laboratory; Mark Fesen, MD, Hutchinson Clinic, PA; George Geils, Jr., MD, Charleston Hematology Oncology Associates, PA; Michael Greenhawt, MD, South Florida Oncology-Hematology; Manuel Guerra, MD, ORA; Rita Gupta, MD, Oncology-Hematology Associates, PA; Vicram Gupta, MD, Saint Joseph Oncology; Alexandre Hageboutros, MD, Cancer Institute of New Jersey at Cooper Hospital; Vincent Hansen, MD, Utah Hematology Oncology; David Henry, MD, Pennsylvania Oncology Hematology Associates; Benjamin Himpler, MD, Syracuse Hematology/Oncology PC; Winston Ho, MD, Hematology/Oncology Group of Orange County; William Horvath, MD, Haematology Oncology Associates of Ohio and Michigan, PC; Paul Hyman, MD, Hematology Oncology Associates of Western Suffolk; Min Kang, MD, Western Washington Oncology; Mark Keaton, MD, Augusta Oncology Associates, PC; Howard Kesselheim, MD, The Center for Cancer and Hematologic Disease; Kapisthalam Kumar, MD, Pasco Hernando Oncology Associates, PA; Edward Lee, MD, Maryland Oncology-Hematology, PA; André Liem, MD, Pacific Shore Medical Group; Timothy Lopez, MD, New Mexico Cancer Care Associates, Cancer Institute of New Mexico; Paul Michael, MD, Comprehensive Cancer Centers of Nevada; Michael Milder, MD, Swedish Cancer Institute; Barry Mirtsching, MD, Center for Oncology Research & Treatment, PA; Ruben Niesvizky, MD, New York Presbyterian Hospital; Jorge Otoya, MD, Osceola Cancer Center; Joseph Pascuzzo, MD, California Oncology of the Central Valley; Ravi Patel, MD, Comprehensive Blood and Cancer Center Lab; Allen Patton, MD, Hematology Oncology Associates, PA; Kelly Pendergrass, MD, Kansas City Cancer Center, LLC; Anthony Phillips, MD, Fox Valley Hematolgy Oncology, SC; Robert Raju, MD, Dayton Oncology and Hematology, PA; Harry Ramsey, MD, Berks Hematology Oncology Associates; Ritesh Rathore, MD, Roger Williams Hospital Medical Center; Phillip Reid, MD, Central Jersey Oncology Center; Robert Robles, MD, Bay Area Cancer Research Group, LLC; Stephen Rosenoff, MD, Oncology and Hematology Associates of Southwest Virginia, Inc; Martin Rubenstein, MD, Southbay Oncology Hematology Partners; Mansoor Saleh, MD, Georgia Cancer Specialists; Sundaresan Sambandam, MD, Hematology and Oncology Associates of RI; Mukund Shah, MD, Antelope Valley Cancer Center; David Siegel, MD, Hackensack University Medical Center; Nelida Sjak-Shie, MD, The Center for Cancer Care and Research; Michael Stone, MD, Greeley Medical Clinic; Stefano Tarantolo, MD, Nebraska Methodist Hospital; Joseph Volk, MD, Palo Verde Hematology Oncology, Ltd; Mitchell Weisberg, MD, MetCare Oncology; Ann Wierman, MD, Nevada Cancer Center; Donald Woytowitz, Jr., MD, Florida Cancer Specialists; Peter Yu, MD, Camino Medical Group.
Correspondence to: James R. Berenson, MD, Institute for Myeloma & Bone Cancer Research, 9201 West Sunset Boulevard, Suite 300, West Hollywood, CA 90069; telephone: (310) 623–1214; fax: (310) 623–1120
The Use of Valeriana officinalis (Valerian) in Improving Sleep in Patients Who Are Undergoing Treatment for Cancer: A Phase III Randomized, Placebo-Controlled, Double-Blind Study (NCCTG Trial, N01C5)
Original research
Debra L. Barton RN, PhD, AOCN, FAAN, a,
Abstract
Sleep disorders are a substantial problem for cancer survivors, with prevalence estimates ranging from 23% to 61%. Although numerous prescription hypnotics are available, few are approved for long-term use or have demonstrated benefit in this circumstance. Hypnotics may have unwanted side effects and are costly, and cancer survivors often wish to avoid prescription drugs. New options with limited side effects are needed. The purpose of this trial was to evaluate the efficacy of a Valerian officinalis supplement for sleep in people with cancer who were undergoing cancer treatment. Participants were randomized to receive 450 mg of valerian or placebo orally 1 hour before bedtime for 8 weeks. The primary end point was area under the curve (AUC) of the overall Pittsburgh Sleep Quality Index (PSQI). Secondary outcomes included the Functional Outcomes of Sleep Questionnaire, the Brief Fatigue Inventory (BFI), and the Profile of Mood States (POMS). Toxicity was evaluated with both self-reported numeric analogue scale questions and the Common Terminology Criteria for Adverse Events (CTCAE), version 3.0. Questionnaires were completed at baseline and at 4 and 8 weeks. A total of 227 patients were randomized into this study between March 19, 2004, and March 9, 2007, with 119 being evaluable for the primary end point. The AUC over the 8 weeks for valerian was 51.4 (SD = 16), while that for placebo was 49.7 (SD = 15), with a P value of 0.6957. A supplemental, exploratory analysis revealed that several fatigue end points, as measured by the BFI and POMS, were significantly better for those taking valerian over placebo. Participants also reported less trouble with sleep and less drowsiness on valerian than placebo. There were no significant differences in toxicities as measured by self-report or the CTCAE except for mild alkaline phosphatase increases, which were slightly more common in the placebo group. This study failed to provide data to support the hypothesis that valerian, 450 mg, at bedtime could improve sleep as measured by the PSQI. However, exploratory analyses revealed improvement in some secondary outcomes, such as fatigue. Further research with valerian exploring physiologic effects in oncology symptom management may be warranted.
Article Outline
Insomnia is present when there is repeated difficulty initiating or maintaining sleep or impairment in sleep quality that occurs despite adequate time and opportunity for sleep, and there is some form of daytime impairment as a result.10 Secondary insomnia is denoted when insomnia is prominent and develops in the setting of another primary medical or psychiatric illness or in the setting of a separate sleep disorder such as sleep apnea.[10], [11] and [12] Sleep disturbance can be associated with poor work performance, increased anxiety and depression, poor cognitive functioning, and impairment of overall quality of life (QOL).[13], [14], [15] and [16] A recent Institute of Medicine report highlighted the severe costs to individuals and society of untreated insomnia.17
Davidson and colleagues2 conducted a cross-sectional descriptive study in six malignant disease clinics from a regional cancer center in Canada. Those surveyed included patients with breast, gastrointestinal, gynecological, genitourinary, lung, and nonmelanoma skin cancers. Insomnia was defined as a report of trouble sleeping on at least 7 of the previous 28 nights, interfering with daytime functioning. More patients who had treatment within the past 6 months reported insomnia, use of sleeping pills, sleeping more than usual, or fatigue. There were no differences based on type of cancer or treatment. Baker and colleagues18 surveyed 752 adult patients who had been diagnosed with 1 of the 10 most commonly occurring cancers to identify which problems cancer survivors experience in dealing with their cancer and its treatment 1 year after diagnosis. Sleep difficulties ranked fifth on the list and were reported by 48% of the sample.
Fatigue is related to sleep disturbance. Although cancer-related fatigue is not necessarily relieved by sleep or rest, insomnia and sleep disturbances clearly contribute to fatigue issues. Fatigue and sleep disturbances are undoubtedly interwoven symptoms and may be difficult to separate. It is not known how much variance in fatigue is explained by sleep problems or in what situations sleep is a major contributor.
Pharmacological Treatments for Insomnia
Because sleep complaints are common, hypnotics are among the most commonly prescribed medications for cancer patients, being prescribed for insomnia in up to 44% of patients.19 Agents most commonly used are benzodiazepine receptor agonists, including true benzodiazepines, such as flurazepam, triazolam, quazepam, estazolam, and temazepam, and the nonbenzodiazepine agents zolpidem (Ambien®), zaleplon (Sonata®), and eszopiclone (Lunesta®), which decrease subjective time to sleep onset, improve sleep efficiency, decrease the number of awakenings, and increase total sleep duration.[20], [21], [22] and [23] Eszopiclone, extended-release formulations of zolpidem (Ambien), and ramelteon (a melatonin receptor agonist) are approved for prolonged use in patients with chronic insomnia;24 but other hypnotics lack well-established effectiveness and safety data for use beyond brief intervals in situational insomnia or as part of a combined approach using cognitive-behavioral therapy (CBT) and brief pharmacological therapy.
In general, improvements in various sleep end points with pharmacologic therapy have been modest, with mean differences in sleep latency being about 15 minutes, wake after sleep onset improving by about 26 minutes, and total sleep time improving by about 40 minutes.[22], [24] and [25] Although subjective improvements are often noted, hypnotic medications are associated with a number of risks, including residual next-day hypersomnia, dizziness, lightheadedness, impaired mental status, and increased risk of falls and hip fractures, especially in elderly patients when taking longer-acting hypnotics.[26], [27], [28], [29], [30] and [31] Clearly, better options to improve sleep are still needed.
The Use of Valeriana officinalis for Sleep
Valeriana officinalis is a perennial herb found in North America, Europe, and Asia. In the United States, it is primarily sold as a sleeping aid, while in Europe it is used for restlessness, tremors, and anxiety. There are three main chemicals that are thought to be the active components of the plant. These are the essential oils valerenic acid and valenol, valepotriates, and a few alkaloids. Herbal extracts of V. officinalis can be ground root, aqueous or aqueous-alcoholic extracts using 70% ethanol and herb-to-extract ratios of 4–7:1. Single recommended doses range from 400 to 900 mg at bedtime.32 Most sleep studies have used 400 or 450 mg for their trials, with a couple of dose-finding trials showing that 900 mg was not significantly better than 450 mg.[33] and [34] The main impact of valerian from those studies has been on sleep latency (time to fall asleep), and this has improved more in patients who had reported a longer time to fall asleep and who considered themselves poor sleepers.[33], [34], [35], [36] and [37]
Most reviews proclaim V. officinalis to be a safe herb with no drug interactions, the only adverse event being daytime sedation at higher doses.[38] and [39] Anecdotal reports of side effects include headaches, nausea, heart palpitations, and benzodiazepine-like withdrawal symptoms when stopping the agent.40 Some concern has been raised as to whether valerian might interfere with cytochrome P-450 metabolism. An article by Budzinski and colleagues reviews numerous herbs and quantitates their interaction with cytochrome P-450.41 Out of 21 herbs tested, V. officinalis ranked at the bottom of interaction potential, rating a 15 out of a possible 16 (1 being the highest, 16 being the lowest).
The cost of V. officinalis, compared to other prescription sleep aids, is less, with a 1-month supply costing around $10 per month. By contrast, zolpidem, for example, costs over $80 per month.
Therefore, based on the favorable toxicity profile, low cost, and promising but limited pilot data, this current trial was designed to evaluate 450 mg of valerian at bedtime for sleep disturbance.
Methods
The primary purpose of this trial was to assess the effect of a standardized preparation of valerian in improving sleep in patients undergoing therapy for cancer. Secondary goals were to assess its safety as well as effect on anxiety, fatigue, and activities of daily living.
Patients eligible for this trial included adults diagnosed with cancer and receiving therapy (radiation, chemotherapy, oral antitumor agents, or endocrine therapy). Patients had to report difficulty sleeping of 4 or more on a scale of 0–10, had to have a life expectancy ≥6 months, and had to have an Eastern Cooperative Oncology Group (ECOG) performance score (PS) of 0 or 1. They could not have an abnormally elevated serum glutamic-oxaloacetic transaminase (SGOT) and/or alkaline phosphatase. Patients were excluded for prior use of valerian for sleep, use of other prescription sleep aids in the past 30 days, or a diagnosis of obstructive sleep apnea or primary insomnia per Diagnostic and Statistical Manual, 4th edition (DSM-IV), criteria. Pregnant and nursing women were also excluded, as were patients with known sleep disturbance etiologies such as nighttime hot flashes, uncontrolled pain, and/or diarrhea.
Participants were randomized to receive 450 mg of oral valerian or placebo, to be taken 1 hour before bedtime for 8 weeks. The valerian used was pure ground, raw root, from one lot and standardized to contain 0.8% valerenic acid. Valerian capsules and matching placebo, a gelatin capsule, were supplied by Hi-Health (Scottsdale, AZ). Both valerian and placebo were stored in the same containers so that the placebo would acquire some of the valerian smell. Self-report booklets were completed at baseline and at weeks 4 and 8 and contained the Pittsburgh Sleep Quality Index (PSQI),42 the Profile of Moods States (POMS),43 the Functional Outcomes of Sleep Questionnaire (FOSQ),44 and the Brief Fatigue Inventory (BFI).45 Assessments were scored according to the appropriate algorithms, and total and subscale scores were transformed to a 0–100 scale, with 100 being best. Self-reported symptoms were recorded weekly using a self-report numeric analogue scale, called the Symptom Experience Diary (SED). Toxicity was also assessed every 2 weeks during a clinical research associate/nurse phone call using the Common Terminology Criteria for Adverse Events (CTCAE, v 3.0).
The primary end point was the normalized (averaged) area under the curve (AUC) of the PSQI between the two arms, compared using the Kruskal-Wallis test. Secondary analyses compared AUC scores of other assessments and toxicity incidence. Toxicity comparisons were performed using the chi-squared test or the Kruskal-Wallis test, as appropriate. As an intent-to-treat (ITT) analysis, using chi-squares tests, patients were categorized as a success if there was a 10-point improvement in the assessment score at week 4 or 8 and a failure if there was no improvement or data were missing.
All hypothesis testing was carried out using a two-sided alternative hypothesis and a 5% Type I error rate. A two-sample t-test with 100 patients per group provided 94% power to detect 50% times the standard deviation (SD) of the end point under study.46 This effect size is considered moderate and has been declared the minimally clinically significant difference for QOL end points.[47] and [48]
Results
A total of 227 patients were randomized into this study between March 19, 2004, and March 9, 2007. The consort diagram depicts the flow of data (Figure 1). Twenty-three patients withdrew before starting the study treatment. Primary end-point data were available on 119 patients (62 receiving valerian and 57 receiving placebo). Baseline characteristics and baseline patient reported outcomes were well balanced between arms with no statistically significant differences ([Table 1] and [Table 2]).
VALERIAN (N = 102) | PLACEBO (N = 100) | P | |
---|---|---|---|
Gender | 0.387 | ||
Female | 82 (80%) | 85 (85%) | |
Age (years) | 0.546 | ||
Mean (SD) | 59.5 (11.95) | 58.3 (12.71) | |
Sleep scale group | 0.963 | ||
Mildly impaired | 67 (66%) | 66 (66%) | |
Moderately or severely impaired | 35 (34%) | 34 (34%) | |
Sleep scale score | 0.841 | ||
Mean (SD) | 6.6 (1.43) | 6.6 (1.69) | |
Primary tumor site | 0.526 | ||
Breast | 64 (63%) | 66 (67%) | |
Colon | 9 (9%) | 5 (5%) | |
Prostate | 3 (3%) | 1 (1%) | |
Other | 25 (25%) | 27 (27%) | |
Tumor status | 0.322 | ||
Resected with no residual | 64 (64%) | 71 (71%) | |
Resected with known residual | 17 (17%) | 12 (13%) | |
Unresected | 19 (19%) | 13 (14%) | |
Treatment type | 0.966 | ||
Radiation therapy | 6 (5.9%) | 6 (6%) | |
Parenteral chemotherapy | 38 (37%) | 39 (39%) | |
Oral therapy | 40 (39%) | 40 (40%) | |
Combined modality | 18 (18%) | 15 (15%) | |
Concurrent radiation | 0.926 | ||
Yes | 23 (23%) | 22 (22%) | |
Concurrent cancer therapy | 0.679 | ||
Yes | 56 (55%) | 52 (53%) | |
Planned or concurrent hormone | 0.667 | ||
Yes | 51 (51%) | 53 (54%) |
VALERIAN (N = 101) | PLACEBO (N = 96) | P | |
---|---|---|---|
PSQI total1 | 0.695 | ||
Mean (SD) | 41.3 (13.92) | 42.4 (14.97) | |
POMS-SF total | 0.883 | ||
Mean (SD) | 65.0 (14.28) | 63.9 (16.46) | |
FOSQ total | 0.927 | ||
Mean (SD) | 73.7 (16.07) | 72.8 (18.37) | |
Fatigue Now | 0.285 | ||
Mean (SD) | 45.7 (24.41) | 49.4 (25.00) | |
Usual Fatigue | 0.216 | ||
Mean (SD) | 46.8 (23.27) | 51.1 (24.73) | |
Worst Fatigue | 0.522 | ||
Mean (SD) | 35.2 (24.67) | 37.9 (26.37) | |
Total Interference | 0.268 | ||
Mean (SD) | 61.4 (25.05) | 57.1 (27.37) |
The primary end point of treatment effectiveness was measured using the normalized AUC calculated using baseline, week 4, and week 8 PSQI total scores. The Wilcoxon rank-sum test P value for the total PSQI score was nonsignificant (valerian AUC = 51.4, SD = 16; placebo AUC = 49.7, SD = 15; P = 0.696) (Figure 2). Similarly the FOSQ was not significantly different between groups either overall or on any subscale score.
Supplemental and exploratory analyses using changes from baseline, however, showed a significant difference in the change from baseline in the amount of sleep at night at week 4 (P = 0.008), favoring the valerian group. Change from baseline in the categorical value for sleep latency was also significantly different at week 4, where 10% of valerian patients indicated longer time to fall asleep compared to 28% on placebo and 43% of valerian patients reported less time to fall asleep compared to 32% on placebo (P = 0.03) (Table 3). The ITT analysis indicated that about 9% more patients experienced a success on valerian relative to placebo, but this was not statistically significant. When scores on the PSQI were divided into ≤5 and >5 (this latter group representing sleep problems), there were fewer patients in the valerian group having sleep problems by week 8 (64% vs 80%, P = 0.56).
VALERIAN | PLACEBO | P | |
---|---|---|---|
Sleep quality | 0.199 | ||
Week 4 | |||
Worse | 2 (3%) | 5 (8%) | |
Same | 33 (49%) | 37 (57%) | |
Better | 33 (49%) | 23 (35%) | |
Week 8 | 0.927 | ||
Worse | 3 (5%) | 2 (3%) | |
Same | 26 (41%) | 25 (42%) | |
Better | 35 (55%) | 32 (54%) | |
Sleep latency | 0.030 | ||
Week 4 | |||
Worse | 6 (10%) | 18 (28%) | |
Same | 30 (48%) | 26 (40%) | |
Better | 27 (43%) | 21 (32%) | |
Week 8 | 0.072 | ||
Worse | 3 (5%) | 11 (18%) | |
Same | 28 (47%) | 29 (48%) | |
Better | 27 (47%) | 21 (34%) | |
Sleep duration | 0.244 | ||
Week 4 | |||
Worse | 6 (9%) | 10 (16%) | |
Same | 26 (39%) | 29 (46%) | |
Better | 34 (52%) | 24 (38%) | |
Week 8 | 0.148 | ||
Worse | 8 (13%) | 4 (7%) | |
Same | 19 (31%) | 28 (48%) | |
Better | 34 (56%) | 27 (46%) | |
Sleep efficiency | 0.295 | ||
Week 4 | |||
Worse | 7 (12%) | 13 (22%) | |
Same | 26 (43%) | 23 (39%) | |
Better | 28 (46%) | 23 (39%) | |
Week 8 | 0.758 | ||
Worse | 11 (19%) | 9 (16%) | |
Same | 19 (33%) | 22 (39%) | |
Better | 28 (48%) | 25 (45%) | |
Sleep disturbance | 0.738 | ||
Week 4 | |||
Worse | 9 (15%) | 11 (18%) | |
Same | 41 (66%) | 40 (67%) | |
Better | 12 (19%) | 9 (15%) | |
Week 8 | 0.177 | ||
Worse | 10 (16%) | 7 (13%) | |
Same | 35 (57%) | 41 (73%) | |
Better | 16 (26%) | 8 (14%) | |
Daytime dysfunction | 0.114 | ||
Week 4 | |||
Worse | 6 (9%) | 13 (19%) | |
Same | 42 (60%) | 40 (60%) | |
Better | 22 (31%) | 14 (21%) | |
Week 8 | 0.478 | ||
Worse | 6 (10%) | 8 (13%) | |
Same | 27 (43%) | 31 (50%) | |
Better | 30 (48%) | 23 (37%) |
While the POMS AUC scores indicated no difference between treatment arms, the mean change from baseline at weeks 4 and 8 was significantly different for the Fatigue-Inertia subscale at weeks 4 (P = 0.004) and 8 (P = 0.02), with the valerian arm reporting better scores (Table 4). On the BFI, the valerian arm scored significantly better than the placebo arm in the mean change from baseline at weeks 4 and 8 on the Fatigue Now (P = 0.003 and P = 0.01, respectively) and Usual Fatigue (P = 0.02 and P = 0.046, respectively) items (Table 4).
SIDE EFFECT | WEEK | VALERIAN | PLACEBO | P |
---|---|---|---|---|
BFI | ||||
Fatigue Now | Week 4 | 13.2 | 1.5 | <0.01 |
Week 8 | 22.1 | 10.5 | <0.01 | |
Usual Fatigue | Week 4 | 12.8 | 4.2 | 0.02 |
Week 8 | 19.4 | 10.0 | 0.05 | |
Worst Fatigue | Week 4 | 11.2 | 3.2 | 0.03 |
Week 8 | 14.8 | 12.4 | 0.65 | |
Activity Interference | Week 4 | 6.2 | 4.1 | 0.75 |
Week 8 | 12.3 | 10.8 | 0.75 | |
POMS | ||||
Anger-Hostility | Week 4 | 3.5 | 2.0 | 0.53 |
Week 8 | 3.9 | 4.2 | 0.89 | |
Vigor-Activity | Week 4 | 2.0 | -0.4 | 0.43 |
Week 8 | 2.0 | 4.7 | 0.34 | |
Depression-Dejection | Week 4 | 3.7 | 5.5 | 0.21 |
Week 8 | 3.7 | 5.4 | 0.25 | |
Confusion-Bewilderment | Week 4 | 4.8 | 2.6 | 0.26 |
Week 8 | 5.3 | 3.4 | 0.79 | |
Fatigue-Inertia | Week 4 | 13.9 | 2.8 | <0.01 |
Week 8 | 17.5 | 9.2 | 0.02 | |
TensionAnxiety | Week 4 | 6.3 | 5.6 | 0.85 |
Week 8 | 9.2 | 8.9 | 0.54 | |
Total score | Week 4 | 5.7 | 3.0 | 0.19 |
Week 8 | 6.9 | 6.0 | 0.90 |
In terms of toxicity, there were no significant differences between arms for the self-reported side effect items (headache, trouble waking, nausea) at baseline, week 4, or week 8 (Table 5). The valerian arm change from baseline at both weeks 4 and 8 showed significant improvement in drowsiness (P = 0.04 and P = 0.03, respectively) and sleep problems (P = 0.005 and P = 0.03, respectively) compared to placebo (Table 5). The maximum severity over time for each self-reported toxicity resulted in no significant differences between arms. There was a significant difference in the CTCAE reporting of alkaline phosphatase, with the placebo arm having a higher incidence of grade 1 toxicity (P = 0.049).
SIDE EFFECT | WEEK | VALERIAN | PLACEBO | P |
---|---|---|---|---|
Nausea | Week 4 | 3.0 | –2.1 | 0.07 |
Week 8 | 3.4 | 0.0 | 0.06 | |
Headache | Week 4 | 4.8 | 1.5 | 0.09 |
Week 8 | 6.7 | 4.6 | 0.27 | |
Trouble waking | Week 4 | 8.8 | 4.3 | 0.42 |
Week 8 | 9.5 | 5.7 | 0.36 | |
Drowsiness | Week 4 | 21.0 | 9.7 | 0.04 |
Week 8 | 24.0 | 14.0 | 0.03 | |
Sleep problems | Week 4 | 18.7 | 4.3 | <0.01 |
Week 8 | 24.0 | 13.0 | 0.03 |
Discussion
This study failed to identify any significant improvements in sleep as measured by the overall PSQI or the FOSQ in this population. This corroborates data from a recent study by Taibi and colleagues,49 who evaluated 300 mg of valerian, taken half an hour before bed. They reported that valerian did not improve any self-reported or polysomnographic sleep outcomes significantly more than placebo. The Taibi et al. study has several possible limitations, including a small sample size (n = 16), a dose lower than that used in the majority of pilot trials with promising results, and a duration of only 15 days on the study agent.
The current study is one of the few randomized placebo-controlled trials evaluating pharmacological treatment of insomnia complaints among cancer patients. Most randomized trials of treatments directed at insomnia in cancer patients compare CBT with usual care or wait-list care and find it of substantial benefit.[50], [51], [52], [53], [54], [55], [56], [57], [58] and [59] One prior trial in terminal cancer patients evaluated intravenous agents for effectiveness, and another controlled trial found mirtazapine to be effective at improving sleep complaints in cancer patients with depression.[51] and [60] Otherwise, there are no other controlled trials assessing pharmacologic agents to primarily address sleep-related complaints in cancer patients.
While there was no significant improvement in sleep quality as assessed by the PSQI, there were consistent improvements in the secondary fatigue outcomes as measured by both the BFI and the POMS Fatigue-Inertia subscale. Although caution is required in interpreting these secondary results, the raw differences in change scores between the two arms are fairly large, often over 10 points (on a 100-point scale). In addition, several other secondary end points—change from baseline related to sleep latency, amount of sleep per night, improvement in sleep problems, and less drowsiness—all support the valerian arm outperforming placebo.
There are several hypotheses related to the inconsistencies in the results. The PSQI may measure different dimensions of well-being from the BFI or POMS, the former concentrating on sleep-quality measures, while the latter two concentrate on daytime symptoms. The correlation between sleep-quality and daytime symptoms may not be very strong in this study's population. Another possibility is that there was a beta-error. Some of the data were incomplete due to the patients' inability to complete the questionnaires appropriately. The power analysis suggested 100 patients per arm were required, and only about 60 per group provided data for analysis. Another hypothesis is that the effects of valerian were too modest and limited to one aspect, perhaps sleep latency, that were not detectable with multidimensional scales such as the PSQI or the FOSQ that look at impact on activity.
There were more patients who withdrew from the placebo arm early compared to the valerian arm. The reasons for this are not known. However, patients on this trial were getting active treatment for cancer, so numerous and varied reasons could explain early withdrawals including complications from treatment, increased fatigue, and worsening sleep problems.
In summary, this trial did not provide data to support that valerian is helpful in improving sleep during cancer treatment in this population. It is not clear whether valerian may have helpful physiologic activity supporting research in oncology symptom management related to fatigue. Perhaps further exploration is warranted.
Acknowledgments
This study was conducted as a collaborative trial of the North Central Cancer Treatment Group and Mayo Clinic and was supported in part by Public Health Service grants CA-25224, CA-37404, CA-124477 (Mentorship Grant), CA-35431, CA-63848, CA-35195, CA-35133, CA-35267, CA-35269, CA-35103, CA-35101, CA-63849, CA-35119, CA-52352, CA-35448, CA-35103, CA-03011, CA-107586, CA-35261, CA-67575, CA-95968, CA-67753, and CA-35415. The content is solely the responsibility of the authors and does not necessarily represent the views of the National Cancer Institute or the National Institutes of Health.
References
1 J. Savard and C.M. Morin, Insomnia in the context of cancer: a review of a neglected problem, J Clin Oncol 19 (2001), pp. 895–908. View Record in Scopus | Cited By in Scopus (147)
2 J.R. Davidson, A.W. MacLean and M.D. Brundage et al., Sleep disturbance in cancer patients, Soc Sci Med 54 (2002), pp. 1309–1321. Article | | View Record in Scopus | Cited By in Scopus (122)
3 D.S. Hu and P.M. Silberfarb, Management of sleep problems in cancer patients, Oncology (Williston Park) 5 (1991), pp. 23–27 discussion 28. View Record in Scopus | Cited By in Scopus (24)
4 L. Fiorentino and S. Ancoli-Israel, Insomnia and its treatment in women with breast cancer, Sleep Med Rev 10 (2006), pp. 419–429. Article | | View Record in Scopus | Cited By in Scopus (13)
5 J.J. Mao, K. Armstrong and M.A. Bowman et al., Symptom burden among cancer survivors: impact of age and comorbidity, J Am Board Fam Med 20 (2007), pp. 434–443. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (24)
6 A.H. Miller, S. Ancoli-Israel and J.E. Bower et al., Neuroendocrine-immune mechanisms of behavioral comorbidities in patients with cancer, J Clin Oncol 26 (2008), pp. 971–982. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (52)
7 J. Savard, J. Villa and H. Ivers et al., Prevalence, natural course, and risk factors of insomnia comorbid with cancer over a 2-month period, J Clin Oncol 27 (31) (2009), pp. 5233–5239. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
8 G. Oxana, J. Palesh and K. Roscoe et al., Prevalence, demographics, and psychological associations of sleep disruption in patients with cancer: University of Rochester Cancer Center–Community Clinical Oncology Program, J Clin Oncol 8 (2) (2010), pp. 292–298.
9 D.C. Owen, K.P. Parker and D.B. McGuire, Comparison of subjective sleep quality in patients with cancer and healthy subjects, Oncol Nurs Forum 26 (1999), pp. 1649–1651. View Record in Scopus | Cited By in Scopus (26)
10 American Academy of Sleep Medicine, International Classification of Sleep Disorders: Diagnostic and Coding Manual (2nd ed.), American Academy of Sleep Medicine, Westchester, IL (2005).
11 A.K. Morin, C.I. Jarvis and A.M. Lynch, Therapeutic options for sleep-maintenance and sleep-onset insomnia, Pharmacotherapy 27 (2007), pp. 89–110. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (34)
12 T. Morgenthaler, M. Kramer and C. Alessi et al., Practice parameters for the psychological and behavioral treatment of insomnia: an update: An American Academy of Sleep Medicine report, Sleep 29 (2006), pp. 1415–1419. View Record in Scopus | Cited By in Scopus (81)
13 T. Roehrs and T. Roth, Sleep–wake state and memory function, Sleep 23 (suppl 3) (2000), pp. S64–S68. View Record in Scopus | Cited By in Scopus (6)
14 J. Payne, B. Piper and I. Rabinowitz et al., Biomarkers, fatigue, sleep, and depressive symptoms in women with breast cancer: a pilot study, Oncol Nurs Forum 33 (2006), pp. 775–783. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (23)
15 R.M. Benca, S. Ancoli-Israel and H. Moldofsky, Special considerations in insomnia diagnosis and management: depressed, elderly, and chronic pain populations, J Clin Psychiatry 65 (suppl 8) (2004), pp. 26–35. View Record in Scopus | Cited By in Scopus (44)
16 D.A. Katz and C.A. McHorney, The relationship between insomnia and health-related quality of life in patients with chronic illness, J Fam Pract 51 (2002), pp. 229–235. View Record in Scopus | Cited By in Scopus (145)
17 H.R. Colten, B.M. Altevogt and Institute of Medicine (U.S.) Committee on Sleep Medicine and Research, Sleep Disorders and Sleep Deprivation: An Unmet Public Health Problem, National Academies Press, Washington DC (2006).
18 F. Baker, M. Denniston and T. Smith et al., Adult cancer survivors: how are they faring?, Cancer 104 (2005), pp. 2565–2576. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (78)
19 F.C. Stiefel, A.B. Kornblith and J.C. Holland, Changes in the prescription patterns of psychotropic drugs for cancer patients during a 10-year period, Cancer 65 (4) (1990), pp. 1048–1053. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (39)
20 B. Ebert, K.A. Wafford and S. Deacon, Treating insomnia: current and investigational pharmacological approaches, Pharmacol Ther 112 (2006), pp. 612–629. Article | | View Record in Scopus | Cited By in Scopus (50)
21 H.J. Moller, Effectiveness and safety of benzodiazepines, J Clin Psychopharmacol 19 (1999), pp. 2S–11S. Full Text via CrossRef
22 J. Barbera and C. Shapiro, Benefit–risk assessment of zaleplon in the treatment of insomnia, Drug Saf 28 (2005), pp. 301–318. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (26)
23 A. Bellon, Searching for new options for treating insomnia: are melatonin and ramelteon beneficial?, J Psychiatr Pract 12 (2006), pp. 229–243. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (20)
24 T. Roth, D. Seiden and S. Sainati et al., Effects of ramelteon on patient-reported sleep latency in older adults with chronic insomnia, Sleep Med 7 (2006), pp. 312–318. Article | | View Record in Scopus | Cited By in Scopus (107)
25 A.M. Holbrook, R. Crowther and A. Lotter et al., Meta-analysis of benzodiazepine use in the treatment of insomnia, CMAJ 162 (2000), pp. 225–233. View Record in Scopus | Cited By in Scopus (215)
26 N. Hall, Taking policy action to reduce benzodiazepine use and promote self-care among seniors, J Appl Gerontol 17 (1998), pp. 318–351. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (8)
27 L.C. Johnson and D.A. Chernik, Sedative-hypnotics and human performance, Psychopharmacology (Berl) 76 (1982), pp. 101–113. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
28 W.A. Ray, M.R. Griffin and W. Downey, Benzodiazepines of long and short elimination half-life and the risk of hip fracture, JAMA 262 (1989), pp. 3303–3306.
29 A. Foy, D. O'Connell and D. Henry et al., Benzodiazepine use as a cause of cognitive impairment in elderly hospital inpatients, J Gerontol A Biol Sci Med Sci 50 (1995), pp. M99–M106. View Record in Scopus | Cited By in Scopus (73)
30 S.L. Gray, K.V. Lai and E.B. Larson, Drug-induced cognition disorders in the elderly: incidence prevention and management, Drug Saf 21 (1999), pp. 101–122. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (93)
31 L.E. Tune and F.W. Bylsma, Benzodiazepine-induced and anticholinergic-induced delirium in the elderly, Int Psychogeriatr 3 (1991), pp. 397–408. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (38)
32 P.J. Houghton, The scientific basis for the reputed activity of valerian, J Pharm Pharmacol 51 (1999), pp. 505–512. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (148)
33 G. Balderer and A.A. Borbely, Effect of valerian on human sleep, Psychopharmacology (Berl) 87 (1982), pp. 406–409.
34 P.D. Leathwood and F. Chauffard, Aqueous extract of valerian reduces latency to fall asleep in man, Planta Med 51 (1985), pp. 144–148. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (84)
35 P.D. Leathwood, F. Chauffard and E. Heck et al., Aqueous extract of valerian root (Valeriana officinalis L.) improves sleep quality in man, Pharmacol Biochem Behav 17 (1982), pp. 65–71. Abstract |
36 H. Schulz, C. Stolz and J. Muller, The effect of valerian extract on sleep polygraphy in poor sleepers: a pilot study, Pharmacopsychiatry 27 (1994), pp. 147–151. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (67)
37 O. Lindahl and L. Lindwall, Double blind study of a valerian preparation, Pharmacol Biochem Behav 32 (1989), pp. 1065–1066. Abstract | | View Record in Scopus | Cited By in Scopus (75)
38 B. Hodgson and R. Kizior, Nursing Drug Handbook, Saunders, Philadelphia (2000).
39 C.A. Thompson, USP moves forward in providing information on botanical products, Am J Health Syst Pharm 55 (1998), pp. 527–530. View Record in Scopus | Cited By in Scopus (1)
40 H. Garges, I. Varia and P. Doraiswamy et al., Cardiac complications and delirium associated with valerian root withdrawal, JAMA 280 (1998), pp. 1566–1567. View Record in Scopus | Cited By in Scopus (75)
41 J.W. Budzinski, B.C. Foster and S. Vandenhoek et al., An in vitro evaluation of human cytochrome P450 3A4 inhibition by selected commercial herbal extracts and tinctures, Phytomedicine 7 (2000), pp. 273–282. View Record in Scopus | Cited By in Scopus (176)
42 D.J. Buysse, C.F. Reynolds 3rd and T.H. Monk et al., The Pittsburgh Sleep Quality Index: a new instrument for psychiatric practice and research, Psychiatry Res 28 (1989), pp. 193–213. Abstract | | View Record in Scopus | Cited By in Scopus (2181)
43 S. Curran, M. Andrykowsky and J. Studts, Short Form of the Profile of Mood States (POMS-SF): psychometric information, Psychol Assess 7 (1995), pp. 80–83. Abstract | | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (150)
44 T.E. Weaver, A.M. Laizner and L.K. Evans et al., An instrument to measure functional status outcomes for disorders of excessive sleepiness, Sleep 20 (1997), pp. 835–843. View Record in Scopus | Cited By in Scopus (231)
45 T.R. Mendoza, X.S. Wang and C.S. Cleeland et al., The rapid assessment of fatigue severity in cancer patients: use of the Brief Fatigue Inventory, Cancer 85 (1999), pp. 1186–1196. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (349)
46 S.R. Lipsitz, G.M. Fitzmaurice and E.J. Orav et al., Performance of generalized estimating equations in practical situations, Biometrics 50 (1994), pp. 270–278. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (90)
47 J. Cohen, Statistical Power Analysis for the Behavioral Sciences, Lawrence Erlbaum, Hillsdale, NJ (1988).
48 J. Sloan, T. Symonds and D. Vargas-Chanes et al., Practical guidelines for assessing the clinical significance of health-related quality of life changes within clinical trials, Drug Inform J 37 (2003), pp. 23–31. View Record in Scopus | Cited By in Scopus (80)
49 D.M. Taibi, M.V. Vitiello and S. Barsness et al., A randomized clinical trial of valerian fails to improve self-reported, polysomnographic, and actigraphic sleep in older women with insomnia, Sleep Med 10 (2009), pp. 319–328. Article | | View Record in Scopus | Cited By in Scopus (5)
50 A. Berger, B. Kuhn and J. Farr et al., Behavioral therapy intervention trial to improve sleep quality and cancer-related fatigue, Psychooncology 18 (2009), pp. 634–646. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (14)
51 E. Cankurtaran, E. Ozalp and H. Soygur et al., Mirtazapine improves sleep and lowers anxiety and depression in cancer patients: superiority over imipramine, Support Care Cancer 16 (2008), pp. 1291–1298. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
52 C. Espie, L. Fleming and J. Cassidy et al., Randomized controlled clinical effectiveness trial of cognitive behavior therapy compared with treatment as usual for persistent insomnia in patients with cancer, J Clin Oncol 26 (28) (2008), pp. 4651–4658. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (37)
53 D. Epstein and S. Dirksen, Randomized trial of a cognitive-behavioral intervention for insomnia in breast cancer survivors, Oncol Nurs Forum 34 (5) (2007), pp. E51–E59. Full Text via CrossRef
54 J. Savard, S. Simard and I. Giguère et al., Randomized clinical trial on cognitive therapy for depression in women with metastatic breast cancer: psychological and immunological effects, Palliat Support Care 4 (3) (2006), pp. 219–237. View Record in Scopus | Cited By in Scopus (30)
55 J. Savard, S. Simard and H. Ivers et al., Randomized study on the efficacy of cognitive-behavioral therapy for insomnia secondary to breast cancer, part I: Sleep and psychological effects, J Clin Oncol 23 (25) (2005), pp. 6083–6096. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (89)
56 J. Savard, S. Simard and H. Ivers, Randomized study on the efficacy of cognitive behavioral therapy for insomnia secondary to breast cancer, part II: Immunologic effects, J Clin Oncol 23 (25) (2005), pp. 6097–6106. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (32)
57 P. Sherwood, B. Given and C. Given et al., A cognitive behavioral intervention for symptom management in patients with advanced cancer, Oncol Nurs Forum 32 (6) (2005), pp. 1190–1198. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
58 C. Quesnel, J. Savard and S. Simard et al., Efficacy of cognitive-behavioral therapy for insomnia in women treated for nonmetastatic breast cancer, J Consult Clin Psychol 71 (1) (2003), pp. 189–200. Abstract | | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (65)
59 J. Davidson, J. Waisberg and M. Brundage et al., Nonpharmacologic group treatment of insomnia: a preliminary study with cancer survivors, Psychooncology 10 (2001), pp. 389–397. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (50)
60 N. Matsuo and T. Morita, Efficacy, safety, and cost effectiveness of intravenous midazolam and flunitrazepam for primary insomnia in terminally ill patients with cancer: a retrospective multicenter audit study, J Palliat Med 10 (5) (2007), pp. 1054–1062. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
Correspondence to: Debra L. Barton, RN, PhD, AOCN, FAAN, Mayo Clinic, 200 First Street, SW, Rochester, MN 55905; telephone: 507-255-3812; fax: 507-538-8300
Original research
Debra L. Barton RN, PhD, AOCN, FAAN, a,
Abstract
Sleep disorders are a substantial problem for cancer survivors, with prevalence estimates ranging from 23% to 61%. Although numerous prescription hypnotics are available, few are approved for long-term use or have demonstrated benefit in this circumstance. Hypnotics may have unwanted side effects and are costly, and cancer survivors often wish to avoid prescription drugs. New options with limited side effects are needed. The purpose of this trial was to evaluate the efficacy of a Valerian officinalis supplement for sleep in people with cancer who were undergoing cancer treatment. Participants were randomized to receive 450 mg of valerian or placebo orally 1 hour before bedtime for 8 weeks. The primary end point was area under the curve (AUC) of the overall Pittsburgh Sleep Quality Index (PSQI). Secondary outcomes included the Functional Outcomes of Sleep Questionnaire, the Brief Fatigue Inventory (BFI), and the Profile of Mood States (POMS). Toxicity was evaluated with both self-reported numeric analogue scale questions and the Common Terminology Criteria for Adverse Events (CTCAE), version 3.0. Questionnaires were completed at baseline and at 4 and 8 weeks. A total of 227 patients were randomized into this study between March 19, 2004, and March 9, 2007, with 119 being evaluable for the primary end point. The AUC over the 8 weeks for valerian was 51.4 (SD = 16), while that for placebo was 49.7 (SD = 15), with a P value of 0.6957. A supplemental, exploratory analysis revealed that several fatigue end points, as measured by the BFI and POMS, were significantly better for those taking valerian over placebo. Participants also reported less trouble with sleep and less drowsiness on valerian than placebo. There were no significant differences in toxicities as measured by self-report or the CTCAE except for mild alkaline phosphatase increases, which were slightly more common in the placebo group. This study failed to provide data to support the hypothesis that valerian, 450 mg, at bedtime could improve sleep as measured by the PSQI. However, exploratory analyses revealed improvement in some secondary outcomes, such as fatigue. Further research with valerian exploring physiologic effects in oncology symptom management may be warranted.
Article Outline
Insomnia is present when there is repeated difficulty initiating or maintaining sleep or impairment in sleep quality that occurs despite adequate time and opportunity for sleep, and there is some form of daytime impairment as a result.10 Secondary insomnia is denoted when insomnia is prominent and develops in the setting of another primary medical or psychiatric illness or in the setting of a separate sleep disorder such as sleep apnea.[10], [11] and [12] Sleep disturbance can be associated with poor work performance, increased anxiety and depression, poor cognitive functioning, and impairment of overall quality of life (QOL).[13], [14], [15] and [16] A recent Institute of Medicine report highlighted the severe costs to individuals and society of untreated insomnia.17
Davidson and colleagues2 conducted a cross-sectional descriptive study in six malignant disease clinics from a regional cancer center in Canada. Those surveyed included patients with breast, gastrointestinal, gynecological, genitourinary, lung, and nonmelanoma skin cancers. Insomnia was defined as a report of trouble sleeping on at least 7 of the previous 28 nights, interfering with daytime functioning. More patients who had treatment within the past 6 months reported insomnia, use of sleeping pills, sleeping more than usual, or fatigue. There were no differences based on type of cancer or treatment. Baker and colleagues18 surveyed 752 adult patients who had been diagnosed with 1 of the 10 most commonly occurring cancers to identify which problems cancer survivors experience in dealing with their cancer and its treatment 1 year after diagnosis. Sleep difficulties ranked fifth on the list and were reported by 48% of the sample.
Fatigue is related to sleep disturbance. Although cancer-related fatigue is not necessarily relieved by sleep or rest, insomnia and sleep disturbances clearly contribute to fatigue issues. Fatigue and sleep disturbances are undoubtedly interwoven symptoms and may be difficult to separate. It is not known how much variance in fatigue is explained by sleep problems or in what situations sleep is a major contributor.
Pharmacological Treatments for Insomnia
Because sleep complaints are common, hypnotics are among the most commonly prescribed medications for cancer patients, being prescribed for insomnia in up to 44% of patients.19 Agents most commonly used are benzodiazepine receptor agonists, including true benzodiazepines, such as flurazepam, triazolam, quazepam, estazolam, and temazepam, and the nonbenzodiazepine agents zolpidem (Ambien®), zaleplon (Sonata®), and eszopiclone (Lunesta®), which decrease subjective time to sleep onset, improve sleep efficiency, decrease the number of awakenings, and increase total sleep duration.[20], [21], [22] and [23] Eszopiclone, extended-release formulations of zolpidem (Ambien), and ramelteon (a melatonin receptor agonist) are approved for prolonged use in patients with chronic insomnia;24 but other hypnotics lack well-established effectiveness and safety data for use beyond brief intervals in situational insomnia or as part of a combined approach using cognitive-behavioral therapy (CBT) and brief pharmacological therapy.
In general, improvements in various sleep end points with pharmacologic therapy have been modest, with mean differences in sleep latency being about 15 minutes, wake after sleep onset improving by about 26 minutes, and total sleep time improving by about 40 minutes.[22], [24] and [25] Although subjective improvements are often noted, hypnotic medications are associated with a number of risks, including residual next-day hypersomnia, dizziness, lightheadedness, impaired mental status, and increased risk of falls and hip fractures, especially in elderly patients when taking longer-acting hypnotics.[26], [27], [28], [29], [30] and [31] Clearly, better options to improve sleep are still needed.
The Use of Valeriana officinalis for Sleep
Valeriana officinalis is a perennial herb found in North America, Europe, and Asia. In the United States, it is primarily sold as a sleeping aid, while in Europe it is used for restlessness, tremors, and anxiety. There are three main chemicals that are thought to be the active components of the plant. These are the essential oils valerenic acid and valenol, valepotriates, and a few alkaloids. Herbal extracts of V. officinalis can be ground root, aqueous or aqueous-alcoholic extracts using 70% ethanol and herb-to-extract ratios of 4–7:1. Single recommended doses range from 400 to 900 mg at bedtime.32 Most sleep studies have used 400 or 450 mg for their trials, with a couple of dose-finding trials showing that 900 mg was not significantly better than 450 mg.[33] and [34] The main impact of valerian from those studies has been on sleep latency (time to fall asleep), and this has improved more in patients who had reported a longer time to fall asleep and who considered themselves poor sleepers.[33], [34], [35], [36] and [37]
Most reviews proclaim V. officinalis to be a safe herb with no drug interactions, the only adverse event being daytime sedation at higher doses.[38] and [39] Anecdotal reports of side effects include headaches, nausea, heart palpitations, and benzodiazepine-like withdrawal symptoms when stopping the agent.40 Some concern has been raised as to whether valerian might interfere with cytochrome P-450 metabolism. An article by Budzinski and colleagues reviews numerous herbs and quantitates their interaction with cytochrome P-450.41 Out of 21 herbs tested, V. officinalis ranked at the bottom of interaction potential, rating a 15 out of a possible 16 (1 being the highest, 16 being the lowest).
The cost of V. officinalis, compared to other prescription sleep aids, is less, with a 1-month supply costing around $10 per month. By contrast, zolpidem, for example, costs over $80 per month.
Therefore, based on the favorable toxicity profile, low cost, and promising but limited pilot data, this current trial was designed to evaluate 450 mg of valerian at bedtime for sleep disturbance.
Methods
The primary purpose of this trial was to assess the effect of a standardized preparation of valerian in improving sleep in patients undergoing therapy for cancer. Secondary goals were to assess its safety as well as effect on anxiety, fatigue, and activities of daily living.
Patients eligible for this trial included adults diagnosed with cancer and receiving therapy (radiation, chemotherapy, oral antitumor agents, or endocrine therapy). Patients had to report difficulty sleeping of 4 or more on a scale of 0–10, had to have a life expectancy ≥6 months, and had to have an Eastern Cooperative Oncology Group (ECOG) performance score (PS) of 0 or 1. They could not have an abnormally elevated serum glutamic-oxaloacetic transaminase (SGOT) and/or alkaline phosphatase. Patients were excluded for prior use of valerian for sleep, use of other prescription sleep aids in the past 30 days, or a diagnosis of obstructive sleep apnea or primary insomnia per Diagnostic and Statistical Manual, 4th edition (DSM-IV), criteria. Pregnant and nursing women were also excluded, as were patients with known sleep disturbance etiologies such as nighttime hot flashes, uncontrolled pain, and/or diarrhea.
Participants were randomized to receive 450 mg of oral valerian or placebo, to be taken 1 hour before bedtime for 8 weeks. The valerian used was pure ground, raw root, from one lot and standardized to contain 0.8% valerenic acid. Valerian capsules and matching placebo, a gelatin capsule, were supplied by Hi-Health (Scottsdale, AZ). Both valerian and placebo were stored in the same containers so that the placebo would acquire some of the valerian smell. Self-report booklets were completed at baseline and at weeks 4 and 8 and contained the Pittsburgh Sleep Quality Index (PSQI),42 the Profile of Moods States (POMS),43 the Functional Outcomes of Sleep Questionnaire (FOSQ),44 and the Brief Fatigue Inventory (BFI).45 Assessments were scored according to the appropriate algorithms, and total and subscale scores were transformed to a 0–100 scale, with 100 being best. Self-reported symptoms were recorded weekly using a self-report numeric analogue scale, called the Symptom Experience Diary (SED). Toxicity was also assessed every 2 weeks during a clinical research associate/nurse phone call using the Common Terminology Criteria for Adverse Events (CTCAE, v 3.0).
The primary end point was the normalized (averaged) area under the curve (AUC) of the PSQI between the two arms, compared using the Kruskal-Wallis test. Secondary analyses compared AUC scores of other assessments and toxicity incidence. Toxicity comparisons were performed using the chi-squared test or the Kruskal-Wallis test, as appropriate. As an intent-to-treat (ITT) analysis, using chi-squares tests, patients were categorized as a success if there was a 10-point improvement in the assessment score at week 4 or 8 and a failure if there was no improvement or data were missing.
All hypothesis testing was carried out using a two-sided alternative hypothesis and a 5% Type I error rate. A two-sample t-test with 100 patients per group provided 94% power to detect 50% times the standard deviation (SD) of the end point under study.46 This effect size is considered moderate and has been declared the minimally clinically significant difference for QOL end points.[47] and [48]
Results
A total of 227 patients were randomized into this study between March 19, 2004, and March 9, 2007. The consort diagram depicts the flow of data (Figure 1). Twenty-three patients withdrew before starting the study treatment. Primary end-point data were available on 119 patients (62 receiving valerian and 57 receiving placebo). Baseline characteristics and baseline patient reported outcomes were well balanced between arms with no statistically significant differences ([Table 1] and [Table 2]).
VALERIAN (N = 102) | PLACEBO (N = 100) | P | |
---|---|---|---|
Gender | 0.387 | ||
Female | 82 (80%) | 85 (85%) | |
Age (years) | 0.546 | ||
Mean (SD) | 59.5 (11.95) | 58.3 (12.71) | |
Sleep scale group | 0.963 | ||
Mildly impaired | 67 (66%) | 66 (66%) | |
Moderately or severely impaired | 35 (34%) | 34 (34%) | |
Sleep scale score | 0.841 | ||
Mean (SD) | 6.6 (1.43) | 6.6 (1.69) | |
Primary tumor site | 0.526 | ||
Breast | 64 (63%) | 66 (67%) | |
Colon | 9 (9%) | 5 (5%) | |
Prostate | 3 (3%) | 1 (1%) | |
Other | 25 (25%) | 27 (27%) | |
Tumor status | 0.322 | ||
Resected with no residual | 64 (64%) | 71 (71%) | |
Resected with known residual | 17 (17%) | 12 (13%) | |
Unresected | 19 (19%) | 13 (14%) | |
Treatment type | 0.966 | ||
Radiation therapy | 6 (5.9%) | 6 (6%) | |
Parenteral chemotherapy | 38 (37%) | 39 (39%) | |
Oral therapy | 40 (39%) | 40 (40%) | |
Combined modality | 18 (18%) | 15 (15%) | |
Concurrent radiation | 0.926 | ||
Yes | 23 (23%) | 22 (22%) | |
Concurrent cancer therapy | 0.679 | ||
Yes | 56 (55%) | 52 (53%) | |
Planned or concurrent hormone | 0.667 | ||
Yes | 51 (51%) | 53 (54%) |
VALERIAN (N = 101) | PLACEBO (N = 96) | P | |
---|---|---|---|
PSQI total1 | 0.695 | ||
Mean (SD) | 41.3 (13.92) | 42.4 (14.97) | |
POMS-SF total | 0.883 | ||
Mean (SD) | 65.0 (14.28) | 63.9 (16.46) | |
FOSQ total | 0.927 | ||
Mean (SD) | 73.7 (16.07) | 72.8 (18.37) | |
Fatigue Now | 0.285 | ||
Mean (SD) | 45.7 (24.41) | 49.4 (25.00) | |
Usual Fatigue | 0.216 | ||
Mean (SD) | 46.8 (23.27) | 51.1 (24.73) | |
Worst Fatigue | 0.522 | ||
Mean (SD) | 35.2 (24.67) | 37.9 (26.37) | |
Total Interference | 0.268 | ||
Mean (SD) | 61.4 (25.05) | 57.1 (27.37) |
The primary end point of treatment effectiveness was measured using the normalized AUC calculated using baseline, week 4, and week 8 PSQI total scores. The Wilcoxon rank-sum test P value for the total PSQI score was nonsignificant (valerian AUC = 51.4, SD = 16; placebo AUC = 49.7, SD = 15; P = 0.696) (Figure 2). Similarly the FOSQ was not significantly different between groups either overall or on any subscale score.
Supplemental and exploratory analyses using changes from baseline, however, showed a significant difference in the change from baseline in the amount of sleep at night at week 4 (P = 0.008), favoring the valerian group. Change from baseline in the categorical value for sleep latency was also significantly different at week 4, where 10% of valerian patients indicated longer time to fall asleep compared to 28% on placebo and 43% of valerian patients reported less time to fall asleep compared to 32% on placebo (P = 0.03) (Table 3). The ITT analysis indicated that about 9% more patients experienced a success on valerian relative to placebo, but this was not statistically significant. When scores on the PSQI were divided into ≤5 and >5 (this latter group representing sleep problems), there were fewer patients in the valerian group having sleep problems by week 8 (64% vs 80%, P = 0.56).
VALERIAN | PLACEBO | P | |
---|---|---|---|
Sleep quality | 0.199 | ||
Week 4 | |||
Worse | 2 (3%) | 5 (8%) | |
Same | 33 (49%) | 37 (57%) | |
Better | 33 (49%) | 23 (35%) | |
Week 8 | 0.927 | ||
Worse | 3 (5%) | 2 (3%) | |
Same | 26 (41%) | 25 (42%) | |
Better | 35 (55%) | 32 (54%) | |
Sleep latency | 0.030 | ||
Week 4 | |||
Worse | 6 (10%) | 18 (28%) | |
Same | 30 (48%) | 26 (40%) | |
Better | 27 (43%) | 21 (32%) | |
Week 8 | 0.072 | ||
Worse | 3 (5%) | 11 (18%) | |
Same | 28 (47%) | 29 (48%) | |
Better | 27 (47%) | 21 (34%) | |
Sleep duration | 0.244 | ||
Week 4 | |||
Worse | 6 (9%) | 10 (16%) | |
Same | 26 (39%) | 29 (46%) | |
Better | 34 (52%) | 24 (38%) | |
Week 8 | 0.148 | ||
Worse | 8 (13%) | 4 (7%) | |
Same | 19 (31%) | 28 (48%) | |
Better | 34 (56%) | 27 (46%) | |
Sleep efficiency | 0.295 | ||
Week 4 | |||
Worse | 7 (12%) | 13 (22%) | |
Same | 26 (43%) | 23 (39%) | |
Better | 28 (46%) | 23 (39%) | |
Week 8 | 0.758 | ||
Worse | 11 (19%) | 9 (16%) | |
Same | 19 (33%) | 22 (39%) | |
Better | 28 (48%) | 25 (45%) | |
Sleep disturbance | 0.738 | ||
Week 4 | |||
Worse | 9 (15%) | 11 (18%) | |
Same | 41 (66%) | 40 (67%) | |
Better | 12 (19%) | 9 (15%) | |
Week 8 | 0.177 | ||
Worse | 10 (16%) | 7 (13%) | |
Same | 35 (57%) | 41 (73%) | |
Better | 16 (26%) | 8 (14%) | |
Daytime dysfunction | 0.114 | ||
Week 4 | |||
Worse | 6 (9%) | 13 (19%) | |
Same | 42 (60%) | 40 (60%) | |
Better | 22 (31%) | 14 (21%) | |
Week 8 | 0.478 | ||
Worse | 6 (10%) | 8 (13%) | |
Same | 27 (43%) | 31 (50%) | |
Better | 30 (48%) | 23 (37%) |
While the POMS AUC scores indicated no difference between treatment arms, the mean change from baseline at weeks 4 and 8 was significantly different for the Fatigue-Inertia subscale at weeks 4 (P = 0.004) and 8 (P = 0.02), with the valerian arm reporting better scores (Table 4). On the BFI, the valerian arm scored significantly better than the placebo arm in the mean change from baseline at weeks 4 and 8 on the Fatigue Now (P = 0.003 and P = 0.01, respectively) and Usual Fatigue (P = 0.02 and P = 0.046, respectively) items (Table 4).
SIDE EFFECT | WEEK | VALERIAN | PLACEBO | P |
---|---|---|---|---|
BFI | ||||
Fatigue Now | Week 4 | 13.2 | 1.5 | <0.01 |
Week 8 | 22.1 | 10.5 | <0.01 | |
Usual Fatigue | Week 4 | 12.8 | 4.2 | 0.02 |
Week 8 | 19.4 | 10.0 | 0.05 | |
Worst Fatigue | Week 4 | 11.2 | 3.2 | 0.03 |
Week 8 | 14.8 | 12.4 | 0.65 | |
Activity Interference | Week 4 | 6.2 | 4.1 | 0.75 |
Week 8 | 12.3 | 10.8 | 0.75 | |
POMS | ||||
Anger-Hostility | Week 4 | 3.5 | 2.0 | 0.53 |
Week 8 | 3.9 | 4.2 | 0.89 | |
Vigor-Activity | Week 4 | 2.0 | -0.4 | 0.43 |
Week 8 | 2.0 | 4.7 | 0.34 | |
Depression-Dejection | Week 4 | 3.7 | 5.5 | 0.21 |
Week 8 | 3.7 | 5.4 | 0.25 | |
Confusion-Bewilderment | Week 4 | 4.8 | 2.6 | 0.26 |
Week 8 | 5.3 | 3.4 | 0.79 | |
Fatigue-Inertia | Week 4 | 13.9 | 2.8 | <0.01 |
Week 8 | 17.5 | 9.2 | 0.02 | |
TensionAnxiety | Week 4 | 6.3 | 5.6 | 0.85 |
Week 8 | 9.2 | 8.9 | 0.54 | |
Total score | Week 4 | 5.7 | 3.0 | 0.19 |
Week 8 | 6.9 | 6.0 | 0.90 |
In terms of toxicity, there were no significant differences between arms for the self-reported side effect items (headache, trouble waking, nausea) at baseline, week 4, or week 8 (Table 5). The valerian arm change from baseline at both weeks 4 and 8 showed significant improvement in drowsiness (P = 0.04 and P = 0.03, respectively) and sleep problems (P = 0.005 and P = 0.03, respectively) compared to placebo (Table 5). The maximum severity over time for each self-reported toxicity resulted in no significant differences between arms. There was a significant difference in the CTCAE reporting of alkaline phosphatase, with the placebo arm having a higher incidence of grade 1 toxicity (P = 0.049).
SIDE EFFECT | WEEK | VALERIAN | PLACEBO | P |
---|---|---|---|---|
Nausea | Week 4 | 3.0 | –2.1 | 0.07 |
Week 8 | 3.4 | 0.0 | 0.06 | |
Headache | Week 4 | 4.8 | 1.5 | 0.09 |
Week 8 | 6.7 | 4.6 | 0.27 | |
Trouble waking | Week 4 | 8.8 | 4.3 | 0.42 |
Week 8 | 9.5 | 5.7 | 0.36 | |
Drowsiness | Week 4 | 21.0 | 9.7 | 0.04 |
Week 8 | 24.0 | 14.0 | 0.03 | |
Sleep problems | Week 4 | 18.7 | 4.3 | <0.01 |
Week 8 | 24.0 | 13.0 | 0.03 |
Discussion
This study failed to identify any significant improvements in sleep as measured by the overall PSQI or the FOSQ in this population. This corroborates data from a recent study by Taibi and colleagues,49 who evaluated 300 mg of valerian, taken half an hour before bed. They reported that valerian did not improve any self-reported or polysomnographic sleep outcomes significantly more than placebo. The Taibi et al. study has several possible limitations, including a small sample size (n = 16), a dose lower than that used in the majority of pilot trials with promising results, and a duration of only 15 days on the study agent.
The current study is one of the few randomized placebo-controlled trials evaluating pharmacological treatment of insomnia complaints among cancer patients. Most randomized trials of treatments directed at insomnia in cancer patients compare CBT with usual care or wait-list care and find it of substantial benefit.[50], [51], [52], [53], [54], [55], [56], [57], [58] and [59] One prior trial in terminal cancer patients evaluated intravenous agents for effectiveness, and another controlled trial found mirtazapine to be effective at improving sleep complaints in cancer patients with depression.[51] and [60] Otherwise, there are no other controlled trials assessing pharmacologic agents to primarily address sleep-related complaints in cancer patients.
While there was no significant improvement in sleep quality as assessed by the PSQI, there were consistent improvements in the secondary fatigue outcomes as measured by both the BFI and the POMS Fatigue-Inertia subscale. Although caution is required in interpreting these secondary results, the raw differences in change scores between the two arms are fairly large, often over 10 points (on a 100-point scale). In addition, several other secondary end points—change from baseline related to sleep latency, amount of sleep per night, improvement in sleep problems, and less drowsiness—all support the valerian arm outperforming placebo.
There are several hypotheses related to the inconsistencies in the results. The PSQI may measure different dimensions of well-being from the BFI or POMS, the former concentrating on sleep-quality measures, while the latter two concentrate on daytime symptoms. The correlation between sleep-quality and daytime symptoms may not be very strong in this study's population. Another possibility is that there was a beta-error. Some of the data were incomplete due to the patients' inability to complete the questionnaires appropriately. The power analysis suggested 100 patients per arm were required, and only about 60 per group provided data for analysis. Another hypothesis is that the effects of valerian were too modest and limited to one aspect, perhaps sleep latency, that were not detectable with multidimensional scales such as the PSQI or the FOSQ that look at impact on activity.
There were more patients who withdrew from the placebo arm early compared to the valerian arm. The reasons for this are not known. However, patients on this trial were getting active treatment for cancer, so numerous and varied reasons could explain early withdrawals including complications from treatment, increased fatigue, and worsening sleep problems.
In summary, this trial did not provide data to support that valerian is helpful in improving sleep during cancer treatment in this population. It is not clear whether valerian may have helpful physiologic activity supporting research in oncology symptom management related to fatigue. Perhaps further exploration is warranted.
Acknowledgments
This study was conducted as a collaborative trial of the North Central Cancer Treatment Group and Mayo Clinic and was supported in part by Public Health Service grants CA-25224, CA-37404, CA-124477 (Mentorship Grant), CA-35431, CA-63848, CA-35195, CA-35133, CA-35267, CA-35269, CA-35103, CA-35101, CA-63849, CA-35119, CA-52352, CA-35448, CA-35103, CA-03011, CA-107586, CA-35261, CA-67575, CA-95968, CA-67753, and CA-35415. The content is solely the responsibility of the authors and does not necessarily represent the views of the National Cancer Institute or the National Institutes of Health.
References
1 J. Savard and C.M. Morin, Insomnia in the context of cancer: a review of a neglected problem, J Clin Oncol 19 (2001), pp. 895–908. View Record in Scopus | Cited By in Scopus (147)
2 J.R. Davidson, A.W. MacLean and M.D. Brundage et al., Sleep disturbance in cancer patients, Soc Sci Med 54 (2002), pp. 1309–1321. Article | | View Record in Scopus | Cited By in Scopus (122)
3 D.S. Hu and P.M. Silberfarb, Management of sleep problems in cancer patients, Oncology (Williston Park) 5 (1991), pp. 23–27 discussion 28. View Record in Scopus | Cited By in Scopus (24)
4 L. Fiorentino and S. Ancoli-Israel, Insomnia and its treatment in women with breast cancer, Sleep Med Rev 10 (2006), pp. 419–429. Article | | View Record in Scopus | Cited By in Scopus (13)
5 J.J. Mao, K. Armstrong and M.A. Bowman et al., Symptom burden among cancer survivors: impact of age and comorbidity, J Am Board Fam Med 20 (2007), pp. 434–443. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (24)
6 A.H. Miller, S. Ancoli-Israel and J.E. Bower et al., Neuroendocrine-immune mechanisms of behavioral comorbidities in patients with cancer, J Clin Oncol 26 (2008), pp. 971–982. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (52)
7 J. Savard, J. Villa and H. Ivers et al., Prevalence, natural course, and risk factors of insomnia comorbid with cancer over a 2-month period, J Clin Oncol 27 (31) (2009), pp. 5233–5239. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
8 G. Oxana, J. Palesh and K. Roscoe et al., Prevalence, demographics, and psychological associations of sleep disruption in patients with cancer: University of Rochester Cancer Center–Community Clinical Oncology Program, J Clin Oncol 8 (2) (2010), pp. 292–298.
9 D.C. Owen, K.P. Parker and D.B. McGuire, Comparison of subjective sleep quality in patients with cancer and healthy subjects, Oncol Nurs Forum 26 (1999), pp. 1649–1651. View Record in Scopus | Cited By in Scopus (26)
10 American Academy of Sleep Medicine, International Classification of Sleep Disorders: Diagnostic and Coding Manual (2nd ed.), American Academy of Sleep Medicine, Westchester, IL (2005).
11 A.K. Morin, C.I. Jarvis and A.M. Lynch, Therapeutic options for sleep-maintenance and sleep-onset insomnia, Pharmacotherapy 27 (2007), pp. 89–110. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (34)
12 T. Morgenthaler, M. Kramer and C. Alessi et al., Practice parameters for the psychological and behavioral treatment of insomnia: an update: An American Academy of Sleep Medicine report, Sleep 29 (2006), pp. 1415–1419. View Record in Scopus | Cited By in Scopus (81)
13 T. Roehrs and T. Roth, Sleep–wake state and memory function, Sleep 23 (suppl 3) (2000), pp. S64–S68. View Record in Scopus | Cited By in Scopus (6)
14 J. Payne, B. Piper and I. Rabinowitz et al., Biomarkers, fatigue, sleep, and depressive symptoms in women with breast cancer: a pilot study, Oncol Nurs Forum 33 (2006), pp. 775–783. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (23)
15 R.M. Benca, S. Ancoli-Israel and H. Moldofsky, Special considerations in insomnia diagnosis and management: depressed, elderly, and chronic pain populations, J Clin Psychiatry 65 (suppl 8) (2004), pp. 26–35. View Record in Scopus | Cited By in Scopus (44)
16 D.A. Katz and C.A. McHorney, The relationship between insomnia and health-related quality of life in patients with chronic illness, J Fam Pract 51 (2002), pp. 229–235. View Record in Scopus | Cited By in Scopus (145)
17 H.R. Colten, B.M. Altevogt and Institute of Medicine (U.S.) Committee on Sleep Medicine and Research, Sleep Disorders and Sleep Deprivation: An Unmet Public Health Problem, National Academies Press, Washington DC (2006).
18 F. Baker, M. Denniston and T. Smith et al., Adult cancer survivors: how are they faring?, Cancer 104 (2005), pp. 2565–2576. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (78)
19 F.C. Stiefel, A.B. Kornblith and J.C. Holland, Changes in the prescription patterns of psychotropic drugs for cancer patients during a 10-year period, Cancer 65 (4) (1990), pp. 1048–1053. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (39)
20 B. Ebert, K.A. Wafford and S. Deacon, Treating insomnia: current and investigational pharmacological approaches, Pharmacol Ther 112 (2006), pp. 612–629. Article | | View Record in Scopus | Cited By in Scopus (50)
21 H.J. Moller, Effectiveness and safety of benzodiazepines, J Clin Psychopharmacol 19 (1999), pp. 2S–11S. Full Text via CrossRef
22 J. Barbera and C. Shapiro, Benefit–risk assessment of zaleplon in the treatment of insomnia, Drug Saf 28 (2005), pp. 301–318. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (26)
23 A. Bellon, Searching for new options for treating insomnia: are melatonin and ramelteon beneficial?, J Psychiatr Pract 12 (2006), pp. 229–243. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (20)
24 T. Roth, D. Seiden and S. Sainati et al., Effects of ramelteon on patient-reported sleep latency in older adults with chronic insomnia, Sleep Med 7 (2006), pp. 312–318. Article | | View Record in Scopus | Cited By in Scopus (107)
25 A.M. Holbrook, R. Crowther and A. Lotter et al., Meta-analysis of benzodiazepine use in the treatment of insomnia, CMAJ 162 (2000), pp. 225–233. View Record in Scopus | Cited By in Scopus (215)
26 N. Hall, Taking policy action to reduce benzodiazepine use and promote self-care among seniors, J Appl Gerontol 17 (1998), pp. 318–351. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (8)
27 L.C. Johnson and D.A. Chernik, Sedative-hypnotics and human performance, Psychopharmacology (Berl) 76 (1982), pp. 101–113. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
28 W.A. Ray, M.R. Griffin and W. Downey, Benzodiazepines of long and short elimination half-life and the risk of hip fracture, JAMA 262 (1989), pp. 3303–3306.
29 A. Foy, D. O'Connell and D. Henry et al., Benzodiazepine use as a cause of cognitive impairment in elderly hospital inpatients, J Gerontol A Biol Sci Med Sci 50 (1995), pp. M99–M106. View Record in Scopus | Cited By in Scopus (73)
30 S.L. Gray, K.V. Lai and E.B. Larson, Drug-induced cognition disorders in the elderly: incidence prevention and management, Drug Saf 21 (1999), pp. 101–122. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (93)
31 L.E. Tune and F.W. Bylsma, Benzodiazepine-induced and anticholinergic-induced delirium in the elderly, Int Psychogeriatr 3 (1991), pp. 397–408. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (38)
32 P.J. Houghton, The scientific basis for the reputed activity of valerian, J Pharm Pharmacol 51 (1999), pp. 505–512. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (148)
33 G. Balderer and A.A. Borbely, Effect of valerian on human sleep, Psychopharmacology (Berl) 87 (1982), pp. 406–409.
34 P.D. Leathwood and F. Chauffard, Aqueous extract of valerian reduces latency to fall asleep in man, Planta Med 51 (1985), pp. 144–148. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (84)
35 P.D. Leathwood, F. Chauffard and E. Heck et al., Aqueous extract of valerian root (Valeriana officinalis L.) improves sleep quality in man, Pharmacol Biochem Behav 17 (1982), pp. 65–71. Abstract |
36 H. Schulz, C. Stolz and J. Muller, The effect of valerian extract on sleep polygraphy in poor sleepers: a pilot study, Pharmacopsychiatry 27 (1994), pp. 147–151. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (67)
37 O. Lindahl and L. Lindwall, Double blind study of a valerian preparation, Pharmacol Biochem Behav 32 (1989), pp. 1065–1066. Abstract | | View Record in Scopus | Cited By in Scopus (75)
38 B. Hodgson and R. Kizior, Nursing Drug Handbook, Saunders, Philadelphia (2000).
39 C.A. Thompson, USP moves forward in providing information on botanical products, Am J Health Syst Pharm 55 (1998), pp. 527–530. View Record in Scopus | Cited By in Scopus (1)
40 H. Garges, I. Varia and P. Doraiswamy et al., Cardiac complications and delirium associated with valerian root withdrawal, JAMA 280 (1998), pp. 1566–1567. View Record in Scopus | Cited By in Scopus (75)
41 J.W. Budzinski, B.C. Foster and S. Vandenhoek et al., An in vitro evaluation of human cytochrome P450 3A4 inhibition by selected commercial herbal extracts and tinctures, Phytomedicine 7 (2000), pp. 273–282. View Record in Scopus | Cited By in Scopus (176)
42 D.J. Buysse, C.F. Reynolds 3rd and T.H. Monk et al., The Pittsburgh Sleep Quality Index: a new instrument for psychiatric practice and research, Psychiatry Res 28 (1989), pp. 193–213. Abstract | | View Record in Scopus | Cited By in Scopus (2181)
43 S. Curran, M. Andrykowsky and J. Studts, Short Form of the Profile of Mood States (POMS-SF): psychometric information, Psychol Assess 7 (1995), pp. 80–83. Abstract | | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (150)
44 T.E. Weaver, A.M. Laizner and L.K. Evans et al., An instrument to measure functional status outcomes for disorders of excessive sleepiness, Sleep 20 (1997), pp. 835–843. View Record in Scopus | Cited By in Scopus (231)
45 T.R. Mendoza, X.S. Wang and C.S. Cleeland et al., The rapid assessment of fatigue severity in cancer patients: use of the Brief Fatigue Inventory, Cancer 85 (1999), pp. 1186–1196. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (349)
46 S.R. Lipsitz, G.M. Fitzmaurice and E.J. Orav et al., Performance of generalized estimating equations in practical situations, Biometrics 50 (1994), pp. 270–278. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (90)
47 J. Cohen, Statistical Power Analysis for the Behavioral Sciences, Lawrence Erlbaum, Hillsdale, NJ (1988).
48 J. Sloan, T. Symonds and D. Vargas-Chanes et al., Practical guidelines for assessing the clinical significance of health-related quality of life changes within clinical trials, Drug Inform J 37 (2003), pp. 23–31. View Record in Scopus | Cited By in Scopus (80)
49 D.M. Taibi, M.V. Vitiello and S. Barsness et al., A randomized clinical trial of valerian fails to improve self-reported, polysomnographic, and actigraphic sleep in older women with insomnia, Sleep Med 10 (2009), pp. 319–328. Article | | View Record in Scopus | Cited By in Scopus (5)
50 A. Berger, B. Kuhn and J. Farr et al., Behavioral therapy intervention trial to improve sleep quality and cancer-related fatigue, Psychooncology 18 (2009), pp. 634–646. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (14)
51 E. Cankurtaran, E. Ozalp and H. Soygur et al., Mirtazapine improves sleep and lowers anxiety and depression in cancer patients: superiority over imipramine, Support Care Cancer 16 (2008), pp. 1291–1298. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
52 C. Espie, L. Fleming and J. Cassidy et al., Randomized controlled clinical effectiveness trial of cognitive behavior therapy compared with treatment as usual for persistent insomnia in patients with cancer, J Clin Oncol 26 (28) (2008), pp. 4651–4658. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (37)
53 D. Epstein and S. Dirksen, Randomized trial of a cognitive-behavioral intervention for insomnia in breast cancer survivors, Oncol Nurs Forum 34 (5) (2007), pp. E51–E59. Full Text via CrossRef
54 J. Savard, S. Simard and I. Giguère et al., Randomized clinical trial on cognitive therapy for depression in women with metastatic breast cancer: psychological and immunological effects, Palliat Support Care 4 (3) (2006), pp. 219–237. View Record in Scopus | Cited By in Scopus (30)
55 J. Savard, S. Simard and H. Ivers et al., Randomized study on the efficacy of cognitive-behavioral therapy for insomnia secondary to breast cancer, part I: Sleep and psychological effects, J Clin Oncol 23 (25) (2005), pp. 6083–6096. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (89)
56 J. Savard, S. Simard and H. Ivers, Randomized study on the efficacy of cognitive behavioral therapy for insomnia secondary to breast cancer, part II: Immunologic effects, J Clin Oncol 23 (25) (2005), pp. 6097–6106. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (32)
57 P. Sherwood, B. Given and C. Given et al., A cognitive behavioral intervention for symptom management in patients with advanced cancer, Oncol Nurs Forum 32 (6) (2005), pp. 1190–1198. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
58 C. Quesnel, J. Savard and S. Simard et al., Efficacy of cognitive-behavioral therapy for insomnia in women treated for nonmetastatic breast cancer, J Consult Clin Psychol 71 (1) (2003), pp. 189–200. Abstract | | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (65)
59 J. Davidson, J. Waisberg and M. Brundage et al., Nonpharmacologic group treatment of insomnia: a preliminary study with cancer survivors, Psychooncology 10 (2001), pp. 389–397. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (50)
60 N. Matsuo and T. Morita, Efficacy, safety, and cost effectiveness of intravenous midazolam and flunitrazepam for primary insomnia in terminally ill patients with cancer: a retrospective multicenter audit study, J Palliat Med 10 (5) (2007), pp. 1054–1062. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
Correspondence to: Debra L. Barton, RN, PhD, AOCN, FAAN, Mayo Clinic, 200 First Street, SW, Rochester, MN 55905; telephone: 507-255-3812; fax: 507-538-8300
Original research
Debra L. Barton RN, PhD, AOCN, FAAN, a,
Abstract
Sleep disorders are a substantial problem for cancer survivors, with prevalence estimates ranging from 23% to 61%. Although numerous prescription hypnotics are available, few are approved for long-term use or have demonstrated benefit in this circumstance. Hypnotics may have unwanted side effects and are costly, and cancer survivors often wish to avoid prescription drugs. New options with limited side effects are needed. The purpose of this trial was to evaluate the efficacy of a Valerian officinalis supplement for sleep in people with cancer who were undergoing cancer treatment. Participants were randomized to receive 450 mg of valerian or placebo orally 1 hour before bedtime for 8 weeks. The primary end point was area under the curve (AUC) of the overall Pittsburgh Sleep Quality Index (PSQI). Secondary outcomes included the Functional Outcomes of Sleep Questionnaire, the Brief Fatigue Inventory (BFI), and the Profile of Mood States (POMS). Toxicity was evaluated with both self-reported numeric analogue scale questions and the Common Terminology Criteria for Adverse Events (CTCAE), version 3.0. Questionnaires were completed at baseline and at 4 and 8 weeks. A total of 227 patients were randomized into this study between March 19, 2004, and March 9, 2007, with 119 being evaluable for the primary end point. The AUC over the 8 weeks for valerian was 51.4 (SD = 16), while that for placebo was 49.7 (SD = 15), with a P value of 0.6957. A supplemental, exploratory analysis revealed that several fatigue end points, as measured by the BFI and POMS, were significantly better for those taking valerian over placebo. Participants also reported less trouble with sleep and less drowsiness on valerian than placebo. There were no significant differences in toxicities as measured by self-report or the CTCAE except for mild alkaline phosphatase increases, which were slightly more common in the placebo group. This study failed to provide data to support the hypothesis that valerian, 450 mg, at bedtime could improve sleep as measured by the PSQI. However, exploratory analyses revealed improvement in some secondary outcomes, such as fatigue. Further research with valerian exploring physiologic effects in oncology symptom management may be warranted.
Article Outline
Insomnia is present when there is repeated difficulty initiating or maintaining sleep or impairment in sleep quality that occurs despite adequate time and opportunity for sleep, and there is some form of daytime impairment as a result.10 Secondary insomnia is denoted when insomnia is prominent and develops in the setting of another primary medical or psychiatric illness or in the setting of a separate sleep disorder such as sleep apnea.[10], [11] and [12] Sleep disturbance can be associated with poor work performance, increased anxiety and depression, poor cognitive functioning, and impairment of overall quality of life (QOL).[13], [14], [15] and [16] A recent Institute of Medicine report highlighted the severe costs to individuals and society of untreated insomnia.17
Davidson and colleagues2 conducted a cross-sectional descriptive study in six malignant disease clinics from a regional cancer center in Canada. Those surveyed included patients with breast, gastrointestinal, gynecological, genitourinary, lung, and nonmelanoma skin cancers. Insomnia was defined as a report of trouble sleeping on at least 7 of the previous 28 nights, interfering with daytime functioning. More patients who had treatment within the past 6 months reported insomnia, use of sleeping pills, sleeping more than usual, or fatigue. There were no differences based on type of cancer or treatment. Baker and colleagues18 surveyed 752 adult patients who had been diagnosed with 1 of the 10 most commonly occurring cancers to identify which problems cancer survivors experience in dealing with their cancer and its treatment 1 year after diagnosis. Sleep difficulties ranked fifth on the list and were reported by 48% of the sample.
Fatigue is related to sleep disturbance. Although cancer-related fatigue is not necessarily relieved by sleep or rest, insomnia and sleep disturbances clearly contribute to fatigue issues. Fatigue and sleep disturbances are undoubtedly interwoven symptoms and may be difficult to separate. It is not known how much variance in fatigue is explained by sleep problems or in what situations sleep is a major contributor.
Pharmacological Treatments for Insomnia
Because sleep complaints are common, hypnotics are among the most commonly prescribed medications for cancer patients, being prescribed for insomnia in up to 44% of patients.19 Agents most commonly used are benzodiazepine receptor agonists, including true benzodiazepines, such as flurazepam, triazolam, quazepam, estazolam, and temazepam, and the nonbenzodiazepine agents zolpidem (Ambien®), zaleplon (Sonata®), and eszopiclone (Lunesta®), which decrease subjective time to sleep onset, improve sleep efficiency, decrease the number of awakenings, and increase total sleep duration.[20], [21], [22] and [23] Eszopiclone, extended-release formulations of zolpidem (Ambien), and ramelteon (a melatonin receptor agonist) are approved for prolonged use in patients with chronic insomnia;24 but other hypnotics lack well-established effectiveness and safety data for use beyond brief intervals in situational insomnia or as part of a combined approach using cognitive-behavioral therapy (CBT) and brief pharmacological therapy.
In general, improvements in various sleep end points with pharmacologic therapy have been modest, with mean differences in sleep latency being about 15 minutes, wake after sleep onset improving by about 26 minutes, and total sleep time improving by about 40 minutes.[22], [24] and [25] Although subjective improvements are often noted, hypnotic medications are associated with a number of risks, including residual next-day hypersomnia, dizziness, lightheadedness, impaired mental status, and increased risk of falls and hip fractures, especially in elderly patients when taking longer-acting hypnotics.[26], [27], [28], [29], [30] and [31] Clearly, better options to improve sleep are still needed.
The Use of Valeriana officinalis for Sleep
Valeriana officinalis is a perennial herb found in North America, Europe, and Asia. In the United States, it is primarily sold as a sleeping aid, while in Europe it is used for restlessness, tremors, and anxiety. There are three main chemicals that are thought to be the active components of the plant. These are the essential oils valerenic acid and valenol, valepotriates, and a few alkaloids. Herbal extracts of V. officinalis can be ground root, aqueous or aqueous-alcoholic extracts using 70% ethanol and herb-to-extract ratios of 4–7:1. Single recommended doses range from 400 to 900 mg at bedtime.32 Most sleep studies have used 400 or 450 mg for their trials, with a couple of dose-finding trials showing that 900 mg was not significantly better than 450 mg.[33] and [34] The main impact of valerian from those studies has been on sleep latency (time to fall asleep), and this has improved more in patients who had reported a longer time to fall asleep and who considered themselves poor sleepers.[33], [34], [35], [36] and [37]
Most reviews proclaim V. officinalis to be a safe herb with no drug interactions, the only adverse event being daytime sedation at higher doses.[38] and [39] Anecdotal reports of side effects include headaches, nausea, heart palpitations, and benzodiazepine-like withdrawal symptoms when stopping the agent.40 Some concern has been raised as to whether valerian might interfere with cytochrome P-450 metabolism. An article by Budzinski and colleagues reviews numerous herbs and quantitates their interaction with cytochrome P-450.41 Out of 21 herbs tested, V. officinalis ranked at the bottom of interaction potential, rating a 15 out of a possible 16 (1 being the highest, 16 being the lowest).
The cost of V. officinalis, compared to other prescription sleep aids, is less, with a 1-month supply costing around $10 per month. By contrast, zolpidem, for example, costs over $80 per month.
Therefore, based on the favorable toxicity profile, low cost, and promising but limited pilot data, this current trial was designed to evaluate 450 mg of valerian at bedtime for sleep disturbance.
Methods
The primary purpose of this trial was to assess the effect of a standardized preparation of valerian in improving sleep in patients undergoing therapy for cancer. Secondary goals were to assess its safety as well as effect on anxiety, fatigue, and activities of daily living.
Patients eligible for this trial included adults diagnosed with cancer and receiving therapy (radiation, chemotherapy, oral antitumor agents, or endocrine therapy). Patients had to report difficulty sleeping of 4 or more on a scale of 0–10, had to have a life expectancy ≥6 months, and had to have an Eastern Cooperative Oncology Group (ECOG) performance score (PS) of 0 or 1. They could not have an abnormally elevated serum glutamic-oxaloacetic transaminase (SGOT) and/or alkaline phosphatase. Patients were excluded for prior use of valerian for sleep, use of other prescription sleep aids in the past 30 days, or a diagnosis of obstructive sleep apnea or primary insomnia per Diagnostic and Statistical Manual, 4th edition (DSM-IV), criteria. Pregnant and nursing women were also excluded, as were patients with known sleep disturbance etiologies such as nighttime hot flashes, uncontrolled pain, and/or diarrhea.
Participants were randomized to receive 450 mg of oral valerian or placebo, to be taken 1 hour before bedtime for 8 weeks. The valerian used was pure ground, raw root, from one lot and standardized to contain 0.8% valerenic acid. Valerian capsules and matching placebo, a gelatin capsule, were supplied by Hi-Health (Scottsdale, AZ). Both valerian and placebo were stored in the same containers so that the placebo would acquire some of the valerian smell. Self-report booklets were completed at baseline and at weeks 4 and 8 and contained the Pittsburgh Sleep Quality Index (PSQI),42 the Profile of Moods States (POMS),43 the Functional Outcomes of Sleep Questionnaire (FOSQ),44 and the Brief Fatigue Inventory (BFI).45 Assessments were scored according to the appropriate algorithms, and total and subscale scores were transformed to a 0–100 scale, with 100 being best. Self-reported symptoms were recorded weekly using a self-report numeric analogue scale, called the Symptom Experience Diary (SED). Toxicity was also assessed every 2 weeks during a clinical research associate/nurse phone call using the Common Terminology Criteria for Adverse Events (CTCAE, v 3.0).
The primary end point was the normalized (averaged) area under the curve (AUC) of the PSQI between the two arms, compared using the Kruskal-Wallis test. Secondary analyses compared AUC scores of other assessments and toxicity incidence. Toxicity comparisons were performed using the chi-squared test or the Kruskal-Wallis test, as appropriate. As an intent-to-treat (ITT) analysis, using chi-squares tests, patients were categorized as a success if there was a 10-point improvement in the assessment score at week 4 or 8 and a failure if there was no improvement or data were missing.
All hypothesis testing was carried out using a two-sided alternative hypothesis and a 5% Type I error rate. A two-sample t-test with 100 patients per group provided 94% power to detect 50% times the standard deviation (SD) of the end point under study.46 This effect size is considered moderate and has been declared the minimally clinically significant difference for QOL end points.[47] and [48]
Results
A total of 227 patients were randomized into this study between March 19, 2004, and March 9, 2007. The consort diagram depicts the flow of data (Figure 1). Twenty-three patients withdrew before starting the study treatment. Primary end-point data were available on 119 patients (62 receiving valerian and 57 receiving placebo). Baseline characteristics and baseline patient reported outcomes were well balanced between arms with no statistically significant differences ([Table 1] and [Table 2]).
VALERIAN (N = 102) | PLACEBO (N = 100) | P | |
---|---|---|---|
Gender | 0.387 | ||
Female | 82 (80%) | 85 (85%) | |
Age (years) | 0.546 | ||
Mean (SD) | 59.5 (11.95) | 58.3 (12.71) | |
Sleep scale group | 0.963 | ||
Mildly impaired | 67 (66%) | 66 (66%) | |
Moderately or severely impaired | 35 (34%) | 34 (34%) | |
Sleep scale score | 0.841 | ||
Mean (SD) | 6.6 (1.43) | 6.6 (1.69) | |
Primary tumor site | 0.526 | ||
Breast | 64 (63%) | 66 (67%) | |
Colon | 9 (9%) | 5 (5%) | |
Prostate | 3 (3%) | 1 (1%) | |
Other | 25 (25%) | 27 (27%) | |
Tumor status | 0.322 | ||
Resected with no residual | 64 (64%) | 71 (71%) | |
Resected with known residual | 17 (17%) | 12 (13%) | |
Unresected | 19 (19%) | 13 (14%) | |
Treatment type | 0.966 | ||
Radiation therapy | 6 (5.9%) | 6 (6%) | |
Parenteral chemotherapy | 38 (37%) | 39 (39%) | |
Oral therapy | 40 (39%) | 40 (40%) | |
Combined modality | 18 (18%) | 15 (15%) | |
Concurrent radiation | 0.926 | ||
Yes | 23 (23%) | 22 (22%) | |
Concurrent cancer therapy | 0.679 | ||
Yes | 56 (55%) | 52 (53%) | |
Planned or concurrent hormone | 0.667 | ||
Yes | 51 (51%) | 53 (54%) |
VALERIAN (N = 101) | PLACEBO (N = 96) | P | |
---|---|---|---|
PSQI total1 | 0.695 | ||
Mean (SD) | 41.3 (13.92) | 42.4 (14.97) | |
POMS-SF total | 0.883 | ||
Mean (SD) | 65.0 (14.28) | 63.9 (16.46) | |
FOSQ total | 0.927 | ||
Mean (SD) | 73.7 (16.07) | 72.8 (18.37) | |
Fatigue Now | 0.285 | ||
Mean (SD) | 45.7 (24.41) | 49.4 (25.00) | |
Usual Fatigue | 0.216 | ||
Mean (SD) | 46.8 (23.27) | 51.1 (24.73) | |
Worst Fatigue | 0.522 | ||
Mean (SD) | 35.2 (24.67) | 37.9 (26.37) | |
Total Interference | 0.268 | ||
Mean (SD) | 61.4 (25.05) | 57.1 (27.37) |
The primary end point of treatment effectiveness was measured using the normalized AUC calculated using baseline, week 4, and week 8 PSQI total scores. The Wilcoxon rank-sum test P value for the total PSQI score was nonsignificant (valerian AUC = 51.4, SD = 16; placebo AUC = 49.7, SD = 15; P = 0.696) (Figure 2). Similarly the FOSQ was not significantly different between groups either overall or on any subscale score.
Supplemental and exploratory analyses using changes from baseline, however, showed a significant difference in the change from baseline in the amount of sleep at night at week 4 (P = 0.008), favoring the valerian group. Change from baseline in the categorical value for sleep latency was also significantly different at week 4, where 10% of valerian patients indicated longer time to fall asleep compared to 28% on placebo and 43% of valerian patients reported less time to fall asleep compared to 32% on placebo (P = 0.03) (Table 3). The ITT analysis indicated that about 9% more patients experienced a success on valerian relative to placebo, but this was not statistically significant. When scores on the PSQI were divided into ≤5 and >5 (this latter group representing sleep problems), there were fewer patients in the valerian group having sleep problems by week 8 (64% vs 80%, P = 0.56).
VALERIAN | PLACEBO | P | |
---|---|---|---|
Sleep quality | 0.199 | ||
Week 4 | |||
Worse | 2 (3%) | 5 (8%) | |
Same | 33 (49%) | 37 (57%) | |
Better | 33 (49%) | 23 (35%) | |
Week 8 | 0.927 | ||
Worse | 3 (5%) | 2 (3%) | |
Same | 26 (41%) | 25 (42%) | |
Better | 35 (55%) | 32 (54%) | |
Sleep latency | 0.030 | ||
Week 4 | |||
Worse | 6 (10%) | 18 (28%) | |
Same | 30 (48%) | 26 (40%) | |
Better | 27 (43%) | 21 (32%) | |
Week 8 | 0.072 | ||
Worse | 3 (5%) | 11 (18%) | |
Same | 28 (47%) | 29 (48%) | |
Better | 27 (47%) | 21 (34%) | |
Sleep duration | 0.244 | ||
Week 4 | |||
Worse | 6 (9%) | 10 (16%) | |
Same | 26 (39%) | 29 (46%) | |
Better | 34 (52%) | 24 (38%) | |
Week 8 | 0.148 | ||
Worse | 8 (13%) | 4 (7%) | |
Same | 19 (31%) | 28 (48%) | |
Better | 34 (56%) | 27 (46%) | |
Sleep efficiency | 0.295 | ||
Week 4 | |||
Worse | 7 (12%) | 13 (22%) | |
Same | 26 (43%) | 23 (39%) | |
Better | 28 (46%) | 23 (39%) | |
Week 8 | 0.758 | ||
Worse | 11 (19%) | 9 (16%) | |
Same | 19 (33%) | 22 (39%) | |
Better | 28 (48%) | 25 (45%) | |
Sleep disturbance | 0.738 | ||
Week 4 | |||
Worse | 9 (15%) | 11 (18%) | |
Same | 41 (66%) | 40 (67%) | |
Better | 12 (19%) | 9 (15%) | |
Week 8 | 0.177 | ||
Worse | 10 (16%) | 7 (13%) | |
Same | 35 (57%) | 41 (73%) | |
Better | 16 (26%) | 8 (14%) | |
Daytime dysfunction | 0.114 | ||
Week 4 | |||
Worse | 6 (9%) | 13 (19%) | |
Same | 42 (60%) | 40 (60%) | |
Better | 22 (31%) | 14 (21%) | |
Week 8 | 0.478 | ||
Worse | 6 (10%) | 8 (13%) | |
Same | 27 (43%) | 31 (50%) | |
Better | 30 (48%) | 23 (37%) |
While the POMS AUC scores indicated no difference between treatment arms, the mean change from baseline at weeks 4 and 8 was significantly different for the Fatigue-Inertia subscale at weeks 4 (P = 0.004) and 8 (P = 0.02), with the valerian arm reporting better scores (Table 4). On the BFI, the valerian arm scored significantly better than the placebo arm in the mean change from baseline at weeks 4 and 8 on the Fatigue Now (P = 0.003 and P = 0.01, respectively) and Usual Fatigue (P = 0.02 and P = 0.046, respectively) items (Table 4).
SIDE EFFECT | WEEK | VALERIAN | PLACEBO | P |
---|---|---|---|---|
BFI | ||||
Fatigue Now | Week 4 | 13.2 | 1.5 | <0.01 |
Week 8 | 22.1 | 10.5 | <0.01 | |
Usual Fatigue | Week 4 | 12.8 | 4.2 | 0.02 |
Week 8 | 19.4 | 10.0 | 0.05 | |
Worst Fatigue | Week 4 | 11.2 | 3.2 | 0.03 |
Week 8 | 14.8 | 12.4 | 0.65 | |
Activity Interference | Week 4 | 6.2 | 4.1 | 0.75 |
Week 8 | 12.3 | 10.8 | 0.75 | |
POMS | ||||
Anger-Hostility | Week 4 | 3.5 | 2.0 | 0.53 |
Week 8 | 3.9 | 4.2 | 0.89 | |
Vigor-Activity | Week 4 | 2.0 | -0.4 | 0.43 |
Week 8 | 2.0 | 4.7 | 0.34 | |
Depression-Dejection | Week 4 | 3.7 | 5.5 | 0.21 |
Week 8 | 3.7 | 5.4 | 0.25 | |
Confusion-Bewilderment | Week 4 | 4.8 | 2.6 | 0.26 |
Week 8 | 5.3 | 3.4 | 0.79 | |
Fatigue-Inertia | Week 4 | 13.9 | 2.8 | <0.01 |
Week 8 | 17.5 | 9.2 | 0.02 | |
TensionAnxiety | Week 4 | 6.3 | 5.6 | 0.85 |
Week 8 | 9.2 | 8.9 | 0.54 | |
Total score | Week 4 | 5.7 | 3.0 | 0.19 |
Week 8 | 6.9 | 6.0 | 0.90 |
In terms of toxicity, there were no significant differences between arms for the self-reported side effect items (headache, trouble waking, nausea) at baseline, week 4, or week 8 (Table 5). The valerian arm change from baseline at both weeks 4 and 8 showed significant improvement in drowsiness (P = 0.04 and P = 0.03, respectively) and sleep problems (P = 0.005 and P = 0.03, respectively) compared to placebo (Table 5). The maximum severity over time for each self-reported toxicity resulted in no significant differences between arms. There was a significant difference in the CTCAE reporting of alkaline phosphatase, with the placebo arm having a higher incidence of grade 1 toxicity (P = 0.049).
SIDE EFFECT | WEEK | VALERIAN | PLACEBO | P |
---|---|---|---|---|
Nausea | Week 4 | 3.0 | –2.1 | 0.07 |
Week 8 | 3.4 | 0.0 | 0.06 | |
Headache | Week 4 | 4.8 | 1.5 | 0.09 |
Week 8 | 6.7 | 4.6 | 0.27 | |
Trouble waking | Week 4 | 8.8 | 4.3 | 0.42 |
Week 8 | 9.5 | 5.7 | 0.36 | |
Drowsiness | Week 4 | 21.0 | 9.7 | 0.04 |
Week 8 | 24.0 | 14.0 | 0.03 | |
Sleep problems | Week 4 | 18.7 | 4.3 | <0.01 |
Week 8 | 24.0 | 13.0 | 0.03 |
Discussion
This study failed to identify any significant improvements in sleep as measured by the overall PSQI or the FOSQ in this population. This corroborates data from a recent study by Taibi and colleagues,49 who evaluated 300 mg of valerian, taken half an hour before bed. They reported that valerian did not improve any self-reported or polysomnographic sleep outcomes significantly more than placebo. The Taibi et al. study has several possible limitations, including a small sample size (n = 16), a dose lower than that used in the majority of pilot trials with promising results, and a duration of only 15 days on the study agent.
The current study is one of the few randomized placebo-controlled trials evaluating pharmacological treatment of insomnia complaints among cancer patients. Most randomized trials of treatments directed at insomnia in cancer patients compare CBT with usual care or wait-list care and find it of substantial benefit.[50], [51], [52], [53], [54], [55], [56], [57], [58] and [59] One prior trial in terminal cancer patients evaluated intravenous agents for effectiveness, and another controlled trial found mirtazapine to be effective at improving sleep complaints in cancer patients with depression.[51] and [60] Otherwise, there are no other controlled trials assessing pharmacologic agents to primarily address sleep-related complaints in cancer patients.
While there was no significant improvement in sleep quality as assessed by the PSQI, there were consistent improvements in the secondary fatigue outcomes as measured by both the BFI and the POMS Fatigue-Inertia subscale. Although caution is required in interpreting these secondary results, the raw differences in change scores between the two arms are fairly large, often over 10 points (on a 100-point scale). In addition, several other secondary end points—change from baseline related to sleep latency, amount of sleep per night, improvement in sleep problems, and less drowsiness—all support the valerian arm outperforming placebo.
There are several hypotheses related to the inconsistencies in the results. The PSQI may measure different dimensions of well-being from the BFI or POMS, the former concentrating on sleep-quality measures, while the latter two concentrate on daytime symptoms. The correlation between sleep-quality and daytime symptoms may not be very strong in this study's population. Another possibility is that there was a beta-error. Some of the data were incomplete due to the patients' inability to complete the questionnaires appropriately. The power analysis suggested 100 patients per arm were required, and only about 60 per group provided data for analysis. Another hypothesis is that the effects of valerian were too modest and limited to one aspect, perhaps sleep latency, that were not detectable with multidimensional scales such as the PSQI or the FOSQ that look at impact on activity.
There were more patients who withdrew from the placebo arm early compared to the valerian arm. The reasons for this are not known. However, patients on this trial were getting active treatment for cancer, so numerous and varied reasons could explain early withdrawals including complications from treatment, increased fatigue, and worsening sleep problems.
In summary, this trial did not provide data to support that valerian is helpful in improving sleep during cancer treatment in this population. It is not clear whether valerian may have helpful physiologic activity supporting research in oncology symptom management related to fatigue. Perhaps further exploration is warranted.
Acknowledgments
This study was conducted as a collaborative trial of the North Central Cancer Treatment Group and Mayo Clinic and was supported in part by Public Health Service grants CA-25224, CA-37404, CA-124477 (Mentorship Grant), CA-35431, CA-63848, CA-35195, CA-35133, CA-35267, CA-35269, CA-35103, CA-35101, CA-63849, CA-35119, CA-52352, CA-35448, CA-35103, CA-03011, CA-107586, CA-35261, CA-67575, CA-95968, CA-67753, and CA-35415. The content is solely the responsibility of the authors and does not necessarily represent the views of the National Cancer Institute or the National Institutes of Health.
References
1 J. Savard and C.M. Morin, Insomnia in the context of cancer: a review of a neglected problem, J Clin Oncol 19 (2001), pp. 895–908. View Record in Scopus | Cited By in Scopus (147)
2 J.R. Davidson, A.W. MacLean and M.D. Brundage et al., Sleep disturbance in cancer patients, Soc Sci Med 54 (2002), pp. 1309–1321. Article | | View Record in Scopus | Cited By in Scopus (122)
3 D.S. Hu and P.M. Silberfarb, Management of sleep problems in cancer patients, Oncology (Williston Park) 5 (1991), pp. 23–27 discussion 28. View Record in Scopus | Cited By in Scopus (24)
4 L. Fiorentino and S. Ancoli-Israel, Insomnia and its treatment in women with breast cancer, Sleep Med Rev 10 (2006), pp. 419–429. Article | | View Record in Scopus | Cited By in Scopus (13)
5 J.J. Mao, K. Armstrong and M.A. Bowman et al., Symptom burden among cancer survivors: impact of age and comorbidity, J Am Board Fam Med 20 (2007), pp. 434–443. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (24)
6 A.H. Miller, S. Ancoli-Israel and J.E. Bower et al., Neuroendocrine-immune mechanisms of behavioral comorbidities in patients with cancer, J Clin Oncol 26 (2008), pp. 971–982. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (52)
7 J. Savard, J. Villa and H. Ivers et al., Prevalence, natural course, and risk factors of insomnia comorbid with cancer over a 2-month period, J Clin Oncol 27 (31) (2009), pp. 5233–5239. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
8 G. Oxana, J. Palesh and K. Roscoe et al., Prevalence, demographics, and psychological associations of sleep disruption in patients with cancer: University of Rochester Cancer Center–Community Clinical Oncology Program, J Clin Oncol 8 (2) (2010), pp. 292–298.
9 D.C. Owen, K.P. Parker and D.B. McGuire, Comparison of subjective sleep quality in patients with cancer and healthy subjects, Oncol Nurs Forum 26 (1999), pp. 1649–1651. View Record in Scopus | Cited By in Scopus (26)
10 American Academy of Sleep Medicine, International Classification of Sleep Disorders: Diagnostic and Coding Manual (2nd ed.), American Academy of Sleep Medicine, Westchester, IL (2005).
11 A.K. Morin, C.I. Jarvis and A.M. Lynch, Therapeutic options for sleep-maintenance and sleep-onset insomnia, Pharmacotherapy 27 (2007), pp. 89–110. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (34)
12 T. Morgenthaler, M. Kramer and C. Alessi et al., Practice parameters for the psychological and behavioral treatment of insomnia: an update: An American Academy of Sleep Medicine report, Sleep 29 (2006), pp. 1415–1419. View Record in Scopus | Cited By in Scopus (81)
13 T. Roehrs and T. Roth, Sleep–wake state and memory function, Sleep 23 (suppl 3) (2000), pp. S64–S68. View Record in Scopus | Cited By in Scopus (6)
14 J. Payne, B. Piper and I. Rabinowitz et al., Biomarkers, fatigue, sleep, and depressive symptoms in women with breast cancer: a pilot study, Oncol Nurs Forum 33 (2006), pp. 775–783. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (23)
15 R.M. Benca, S. Ancoli-Israel and H. Moldofsky, Special considerations in insomnia diagnosis and management: depressed, elderly, and chronic pain populations, J Clin Psychiatry 65 (suppl 8) (2004), pp. 26–35. View Record in Scopus | Cited By in Scopus (44)
16 D.A. Katz and C.A. McHorney, The relationship between insomnia and health-related quality of life in patients with chronic illness, J Fam Pract 51 (2002), pp. 229–235. View Record in Scopus | Cited By in Scopus (145)
17 H.R. Colten, B.M. Altevogt and Institute of Medicine (U.S.) Committee on Sleep Medicine and Research, Sleep Disorders and Sleep Deprivation: An Unmet Public Health Problem, National Academies Press, Washington DC (2006).
18 F. Baker, M. Denniston and T. Smith et al., Adult cancer survivors: how are they faring?, Cancer 104 (2005), pp. 2565–2576. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (78)
19 F.C. Stiefel, A.B. Kornblith and J.C. Holland, Changes in the prescription patterns of psychotropic drugs for cancer patients during a 10-year period, Cancer 65 (4) (1990), pp. 1048–1053. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (39)
20 B. Ebert, K.A. Wafford and S. Deacon, Treating insomnia: current and investigational pharmacological approaches, Pharmacol Ther 112 (2006), pp. 612–629. Article | | View Record in Scopus | Cited By in Scopus (50)
21 H.J. Moller, Effectiveness and safety of benzodiazepines, J Clin Psychopharmacol 19 (1999), pp. 2S–11S. Full Text via CrossRef
22 J. Barbera and C. Shapiro, Benefit–risk assessment of zaleplon in the treatment of insomnia, Drug Saf 28 (2005), pp. 301–318. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (26)
23 A. Bellon, Searching for new options for treating insomnia: are melatonin and ramelteon beneficial?, J Psychiatr Pract 12 (2006), pp. 229–243. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (20)
24 T. Roth, D. Seiden and S. Sainati et al., Effects of ramelteon on patient-reported sleep latency in older adults with chronic insomnia, Sleep Med 7 (2006), pp. 312–318. Article | | View Record in Scopus | Cited By in Scopus (107)
25 A.M. Holbrook, R. Crowther and A. Lotter et al., Meta-analysis of benzodiazepine use in the treatment of insomnia, CMAJ 162 (2000), pp. 225–233. View Record in Scopus | Cited By in Scopus (215)
26 N. Hall, Taking policy action to reduce benzodiazepine use and promote self-care among seniors, J Appl Gerontol 17 (1998), pp. 318–351. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (8)
27 L.C. Johnson and D.A. Chernik, Sedative-hypnotics and human performance, Psychopharmacology (Berl) 76 (1982), pp. 101–113. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
28 W.A. Ray, M.R. Griffin and W. Downey, Benzodiazepines of long and short elimination half-life and the risk of hip fracture, JAMA 262 (1989), pp. 3303–3306.
29 A. Foy, D. O'Connell and D. Henry et al., Benzodiazepine use as a cause of cognitive impairment in elderly hospital inpatients, J Gerontol A Biol Sci Med Sci 50 (1995), pp. M99–M106. View Record in Scopus | Cited By in Scopus (73)
30 S.L. Gray, K.V. Lai and E.B. Larson, Drug-induced cognition disorders in the elderly: incidence prevention and management, Drug Saf 21 (1999), pp. 101–122. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (93)
31 L.E. Tune and F.W. Bylsma, Benzodiazepine-induced and anticholinergic-induced delirium in the elderly, Int Psychogeriatr 3 (1991), pp. 397–408. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (38)
32 P.J. Houghton, The scientific basis for the reputed activity of valerian, J Pharm Pharmacol 51 (1999), pp. 505–512. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (148)
33 G. Balderer and A.A. Borbely, Effect of valerian on human sleep, Psychopharmacology (Berl) 87 (1982), pp. 406–409.
34 P.D. Leathwood and F. Chauffard, Aqueous extract of valerian reduces latency to fall asleep in man, Planta Med 51 (1985), pp. 144–148. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (84)
35 P.D. Leathwood, F. Chauffard and E. Heck et al., Aqueous extract of valerian root (Valeriana officinalis L.) improves sleep quality in man, Pharmacol Biochem Behav 17 (1982), pp. 65–71. Abstract |
36 H. Schulz, C. Stolz and J. Muller, The effect of valerian extract on sleep polygraphy in poor sleepers: a pilot study, Pharmacopsychiatry 27 (1994), pp. 147–151. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (67)
37 O. Lindahl and L. Lindwall, Double blind study of a valerian preparation, Pharmacol Biochem Behav 32 (1989), pp. 1065–1066. Abstract | | View Record in Scopus | Cited By in Scopus (75)
38 B. Hodgson and R. Kizior, Nursing Drug Handbook, Saunders, Philadelphia (2000).
39 C.A. Thompson, USP moves forward in providing information on botanical products, Am J Health Syst Pharm 55 (1998), pp. 527–530. View Record in Scopus | Cited By in Scopus (1)
40 H. Garges, I. Varia and P. Doraiswamy et al., Cardiac complications and delirium associated with valerian root withdrawal, JAMA 280 (1998), pp. 1566–1567. View Record in Scopus | Cited By in Scopus (75)
41 J.W. Budzinski, B.C. Foster and S. Vandenhoek et al., An in vitro evaluation of human cytochrome P450 3A4 inhibition by selected commercial herbal extracts and tinctures, Phytomedicine 7 (2000), pp. 273–282. View Record in Scopus | Cited By in Scopus (176)
42 D.J. Buysse, C.F. Reynolds 3rd and T.H. Monk et al., The Pittsburgh Sleep Quality Index: a new instrument for psychiatric practice and research, Psychiatry Res 28 (1989), pp. 193–213. Abstract | | View Record in Scopus | Cited By in Scopus (2181)
43 S. Curran, M. Andrykowsky and J. Studts, Short Form of the Profile of Mood States (POMS-SF): psychometric information, Psychol Assess 7 (1995), pp. 80–83. Abstract | | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (150)
44 T.E. Weaver, A.M. Laizner and L.K. Evans et al., An instrument to measure functional status outcomes for disorders of excessive sleepiness, Sleep 20 (1997), pp. 835–843. View Record in Scopus | Cited By in Scopus (231)
45 T.R. Mendoza, X.S. Wang and C.S. Cleeland et al., The rapid assessment of fatigue severity in cancer patients: use of the Brief Fatigue Inventory, Cancer 85 (1999), pp. 1186–1196. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (349)
46 S.R. Lipsitz, G.M. Fitzmaurice and E.J. Orav et al., Performance of generalized estimating equations in practical situations, Biometrics 50 (1994), pp. 270–278. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (90)
47 J. Cohen, Statistical Power Analysis for the Behavioral Sciences, Lawrence Erlbaum, Hillsdale, NJ (1988).
48 J. Sloan, T. Symonds and D. Vargas-Chanes et al., Practical guidelines for assessing the clinical significance of health-related quality of life changes within clinical trials, Drug Inform J 37 (2003), pp. 23–31. View Record in Scopus | Cited By in Scopus (80)
49 D.M. Taibi, M.V. Vitiello and S. Barsness et al., A randomized clinical trial of valerian fails to improve self-reported, polysomnographic, and actigraphic sleep in older women with insomnia, Sleep Med 10 (2009), pp. 319–328. Article | | View Record in Scopus | Cited By in Scopus (5)
50 A. Berger, B. Kuhn and J. Farr et al., Behavioral therapy intervention trial to improve sleep quality and cancer-related fatigue, Psychooncology 18 (2009), pp. 634–646. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (14)
51 E. Cankurtaran, E. Ozalp and H. Soygur et al., Mirtazapine improves sleep and lowers anxiety and depression in cancer patients: superiority over imipramine, Support Care Cancer 16 (2008), pp. 1291–1298. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
52 C. Espie, L. Fleming and J. Cassidy et al., Randomized controlled clinical effectiveness trial of cognitive behavior therapy compared with treatment as usual for persistent insomnia in patients with cancer, J Clin Oncol 26 (28) (2008), pp. 4651–4658. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (37)
53 D. Epstein and S. Dirksen, Randomized trial of a cognitive-behavioral intervention for insomnia in breast cancer survivors, Oncol Nurs Forum 34 (5) (2007), pp. E51–E59. Full Text via CrossRef
54 J. Savard, S. Simard and I. Giguère et al., Randomized clinical trial on cognitive therapy for depression in women with metastatic breast cancer: psychological and immunological effects, Palliat Support Care 4 (3) (2006), pp. 219–237. View Record in Scopus | Cited By in Scopus (30)
55 J. Savard, S. Simard and H. Ivers et al., Randomized study on the efficacy of cognitive-behavioral therapy for insomnia secondary to breast cancer, part I: Sleep and psychological effects, J Clin Oncol 23 (25) (2005), pp. 6083–6096. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (89)
56 J. Savard, S. Simard and H. Ivers, Randomized study on the efficacy of cognitive behavioral therapy for insomnia secondary to breast cancer, part II: Immunologic effects, J Clin Oncol 23 (25) (2005), pp. 6097–6106. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (32)
57 P. Sherwood, B. Given and C. Given et al., A cognitive behavioral intervention for symptom management in patients with advanced cancer, Oncol Nurs Forum 32 (6) (2005), pp. 1190–1198. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
58 C. Quesnel, J. Savard and S. Simard et al., Efficacy of cognitive-behavioral therapy for insomnia in women treated for nonmetastatic breast cancer, J Consult Clin Psychol 71 (1) (2003), pp. 189–200. Abstract | | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (65)
59 J. Davidson, J. Waisberg and M. Brundage et al., Nonpharmacologic group treatment of insomnia: a preliminary study with cancer survivors, Psychooncology 10 (2001), pp. 389–397. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (50)
60 N. Matsuo and T. Morita, Efficacy, safety, and cost effectiveness of intravenous midazolam and flunitrazepam for primary insomnia in terminally ill patients with cancer: a retrospective multicenter audit study, J Palliat Med 10 (5) (2007), pp. 1054–1062. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
Correspondence to: Debra L. Barton, RN, PhD, AOCN, FAAN, Mayo Clinic, 200 First Street, SW, Rochester, MN 55905; telephone: 507-255-3812; fax: 507-538-8300
Pilot Study of the Prospective Identification of Changes in Cognitive Function During Chemotherapy Treatment for Advanced Ovarian Cancer
Original research
Lisa M. Hess PhD, a,
Abstract
Change in cognitive function is increasingly being recognized as an adverse outcome related to chemotherapy treatment. These changes need not be severe to impact patient functional ability and quality of life. The primary goal of this study was to determine if there is evidence of changes in the cognitive function domains of attention, processing speed, and response time among women with newly diagnosed advanced ovarian cancer who receive chemotherapy. Eligible patients were women diagnosed with stage III–IV epithelial ovarian or primary peritoneal cancer who had not yet received chemotherapy but who were prescribed a minimum of six cycles (courses) of chemotherapy treatment. Cognitive function was assessed by a computerized, Web-based assessment (attention, processing speed, and reaction time) and by patient self-report. Cognitive function was assessed at three time points: prior to the first course (baseline), course three, and course six. Medical records were reviewed to abstract information on chemotherapy treatment, concomitant medications, and blood test results (eg, hemoglobin, CA-125). Of the 27 eligible participants, 92% and 86% demonstrated cognitive impairments from baseline to course three and from baseline to course six of chemotherapy, respectively. Impairment was detected in two or more cognitive domains among 48% (12 of 25) and 41% (9 of 22) of participants at course three and course six of chemotherapy, respectively. This study shows evidence of decline in cognitive function among women being treated for ovarian cancer. There is a need for additional, prospective research to better understand the impact of chemotherapy on cognitive function among ovarian cancer patients so that effective preventive and treatment strategies can be developed.
Article Outline
Although the perception of cognitive decline is a common complaint among individuals treated with chemotherapy, it is poorly understood and limited efforts have been made to identify the extent of this problem among women with ovarian cancer. To date, the few studies documenting the neuropsychological consequences of ovarian cancer and its treatment have shown that patients report cognitive problems but that these problems were not quantifiable using objective measures due to the lack of sensitivity of standard instruments to the subtle changes that occur during cancer treatment.[5], [6] and [7]
Although studies of cognitive function among oncology patients have used instruments that have been validated in their own disciplines and with a variety of diseases, the evidence is emerging that they are not comprehensive or appropriate tools for the detection and evaluation of chemotherapy-related change in cognitive function.8 Furthermore, the likelihood of having these tests conducted in a similar manner across multiple institutions, sites, and interviewers with any degree of consistency is very low. This study was designed as a pilot study of the identification of chemotherapy-related changes in cognitive function among women with advanced ovarian cancer using a Web-based assessment tool (Headminder, Inc., New York, NY).7 The primary goal of the current study was to determine if there is evidence of changes in the cognitive function domains of attention, processing speed, and reaction time as well as self-reported changes in the memory, sensory-perception, and cognitive-intellectual domains of cognitive function during chemotherapy among women with newly diagnosed advanced ovarian cancer.
Materials and Methods
All study methods and procedures were reviewed and approved by the University of Arizona Institutional Review Board. Eligible patients included women with a histologically or pathologically confirmed diagnosis of stage III–IV epithelial ovarian or primary peritoneal cancer who were prescribed at least six courses of platinum-based therapy. Patients were excluded if they had a prior history of any cancer (other than nonmelanoma skin cancer), chemotherapy, radiation therapy, erythropoietin treatment (within the last 6 months), or severe head injury. Initially, patients were excluded if they received intraperitoneal therapy, but the protocol was later amended to permit the use of any platinum-based therapy, regardless of route of administration.
Assessment Tools
After providing informed consent, patients completed a neurocognitive battery of tests and the Functional Assessment of Cancer Therapy—Neurotoxicity (FACT-Ntx, to assess patient-reported neuropathy).[9] and [10] The neurocognitive evaluation included both a computerized, Web-based and a patient-reported assessment. The Web-based assessment was provided by HeadMinders, Inc.[7] and [11] and was a modified version of the Cognitive Stability Index. The modified battery was comprised of two warm-up tasks and three empirically-derived cognitive factors: Processing Speed (Animal Decoding and Symbol Scanning subtests), Attention (Number Recall and Number Sequencing subtests), and Reaction Time (Response Direction 1 and Response Direction 2 subtests). The subtests have been validated against traditional neuropsychological tests in healthy and clinical populations, including cancer patients.12 Cognitive domain correlations in the battery's healthy normative sample range from 0.52 to 0.74, and correlations are similar or higher in clinical populations. Test–retest reliability of the factor scores between first and second administrations ranges from 0.74 to 0.82.12 This Web-based neurocognitive assessment tool is 21 CFR Part 11– and Health on the Net (HON)–compliant to ensure patient confidentiality. Prior to undergoing the Web-based cognitive tests, all study participants completed a keyboard proficiency test as a “warm-up task” to the computerized assessment.
The patient-reported cognitive function tool used was the Patient Assessment of Own Functioning Scale (PAF).[13], [14] and [15] The PAF includes eight scales that are grouped into the nature of the ability being considered. The Memory, Sensory-Perceptual, and Cognitive-Intellectual subscales of the PAF are included in this self-assessment questionnaire. Respondents are asked to rate on a six-point scale, from almost always to almost never, how often they experience a particular kind of difficulty in their everyday lives. For this study, the Memory and Cognitive-Intellectual subscales of the PAF were used, similar to other clinical research protocols investigating cognitive changes during chemotherapy treatment.15 The PAF has been shown to be directly related to the Minnesota Multiphasic Personality Inventory (MMPI)13 and to be highly correlated with other cognitive impairment indices, such as the American College of Rheumatology neuropsychology research battery of tests.16 Of note, self-reported cognitive change has not been shown to correlate formal assessments of cognitive function among individuals who have experienced cancer.[17], [18], [19], [20] and [21]
The FACT-Ntx is a validated instrument[9] and [10] that was used to evaluate neurotoxicity. This scale includes 11 items: nine to assess neurotoxicity, one to assess bodily weakness, and one to assess anemia. Neurotoxicity may affect a patient's ability to use the keyboard in the computerized neurocognitive evaluation. This complete assessment battery of tests was completed at baseline (within 5 days of initiation of chemotherapy) and again during follow-up assessments at cycle three and cycle six of chemotherapy. The medical record was reviewed and data were abstracted related to chemotherapy medications, all concomitant medications, and blood test results (eg, hemoglobin, CA-125).
Statistical Plan
This prospective study was exploratory in nature and designed to collect pilot data to determine if there is evidence of neurocognitive change in attention, processing speed, response time, or self-reported cognitive function during the course of chemotherapy among women being treated for advanced ovarian cancer. The purpose of this study was to obtain preliminary estimates of the incidence and degree of cognitive decline to aid in the planning of future studies. While prior estimates of cognitive function were not available for this population, power analyses demonstrate that with a target recruitment goal of 30 patients, a McNemar's test has 78% power at the 0.05 level of significance to detect a significant decline in impairment in a cognitive domain if 12 patients are found to have impairment prior to course six of treatment (but not at course three) and if as few as two patients demonstrate impairment prior to course three but not at course six. This study was therefore powered to detect declines in one or more of the domains that may have occurred at less than both of the study time points following the baseline assessment.
To be considered fully evaluable, patients had to have completed at least one follow-up neurocognitive evaluation and may not have received antipsychotic neuropsychological medications during the study (eg, chlorpromazine, haloperidol, clozapine). Antidepressants and antianxiety medications (eg, serotonin/norepinephrine reuptake inhibitors or benzodiazepines) were permitted and use was recorded throughout study participation. A summary score for each cognitive domain (processing speed, reaction time, and attention) was recorded at each assessment time point using the HeadMinder Web-based assessment. This summary score was assessed by time (processing speed and reaction time), measured to the hundredth of a second, and by number of errors (attention). If a cognitive domain summary score at a follow-up assessment time declined at least one standard error of measurement (SEM) from baseline, the patient was considered to have experienced a decline at that time point. For the purposes of this article, such declines are referred to as “impairments” within the cognitive domain under investigation. A cognitive index score (CIS) was calculated as the number of cognitive domains impaired for the time point. The range of a CIS is 0–3, with zero equal to no impairment on any cognitive domain and three equal to impairment on all cognitive domains. Patients with only one cognitive domain decline (CIS = 1) at any one of the follow-up assessment time points were considered as having possible cognitive function decline. Patients with more than one cognitive domain impairment (CIS >1) at any follow-up assessment time points were considered as having evidence of cognitive function decline. The incidence of cognitive function impairment was determined by the percentage of patients who experienced any cognitive domain impairment (including possible and evidence of decline) at any follow-up assessment.
A repeated-measures analyses of variance (ANOVA) was used to further explore the neurocognitive values at the various time points during the study. Many of the neurocognitive values were not normally distributed but skewed either positively or negatively, so the square roots of the values were used in the analyses. Since this is an exploratory analysis, no corrections for multiple comparisons were performed.
The patient-reported cognitive function instrument (PAF) contains items scored on a Likert-type scale from almost never to almost always (range 0–5). Patient-reported outcomes as measured with the PAF are measured as mean scale values, ranging from 0, indicating no impairment, to 5.0, indicating complete impairment. PAF score ranges indicate low (≤1.25), medium (1.26–1.92), and high (≥1.93) levels of cognitive impairment.13 A total FACT-Ntx score was obtained; lower scores represent greater neurotoxicity, ranging from 0 (extreme neurotoxicity) to 44 (no neurotoxicity). The total score was reported, with adjustments made for missing values as described elsewhere.22
Results
Thirty patients were enrolled in this study; however, two were later deemed ineligible, and one was unable to complete the baseline neurocognitive assessment prior to chemotherapy and was withdrawn from the study, resulting in 27 patients available for assessment. Five of these patients did not complete all neurocognitive assessments. The primary reason for nonadherence to the study schedule was clinical scheduling (eg, chemotherapy was administered prior to the neurocognitive assessment). The characteristics of eligible patients are provided in Table 1. The majority of patients were receiving intravenous chemotherapy (intraperitoneal therapy was at first not permitted but later was allowable following an amendment to the protocol) and taking concomitant sleep, antianxiety, and/or antidepressant medications outside of every 3- to 4-week chemotherapy regimen (primarily zolpidem, lorazepam, sertraline, and/or trazodone).
n = 27 | |
Mean age, years (range) | 59.3 (40.3–81.5) |
Education, n (%) | |
High school or less | 3 (11.1%) |
Some college | 12 (44.4%) |
College graduate | 12 (44.4%) |
Race/ethnicity, n (%) | |
White, non-Hispanic | 25 (92.6%) |
Hispanic | 1 (3.7%) |
Native American | 1 (3.7%) |
Marital status, n (%) | |
Married/cohabitating | 19 (70.4%) |
Divorced/separated | 1 (3.7%) |
Widowed | 5 (18.5%) |
Never married | 2 (7.4%) |
Mean courses of chemotherapy, n (range) | 5.9 (4–6) |
Chemotherapy route, n (%) | |
Intraperitoneal | 5 (18.5%) |
Intravenous | 22 (81.5%) |
Concurrent medication use, n (%) | |
Antidepressant | 7 (25.9%) |
Antianxiety | 16 (59.3%) |
Sleep aids | 5 (18.5%) |
Web-Assessed Cognitive Function
Keyboard proficiency remained unchanged over time (P = 0.39). As shown in Table 2, most participants demonstrated cognitive impairments in at least one of the three cognitive domains assessed during this study (92% and 86% at course 3 and course 6, respectively). Nearly half of the study participants demonstrated impairment from baseline in two or more of the three cognitive domains assessed (Table 3). Table 4 shows a detailed summary of the subscales within the Web-based cognitive tests that comprised the CIS.This table demonstrates the statistically significant increase in test subscale errors, despite the test-taking improvements over time, as shown by reduction in testing time.
CIS | COURSE 3 | COURSE 6 |
---|---|---|
No decline (CIS = 0) | 2 (8%) | 3 (14%) |
One impairment (CIS = 1) | 11 (44%) | 10 (45%) |
Two impairments (CIS = 2) | 11 (44%) | 7 (32%) |
Three impairments (CIS = 3) | 1 (4%) | 2 (9%) |
COGNITIVE IMPAIRMENT SCALE (CIS) FACTORS | BASELINE | COURSE 3 | COURSE 6 | |||||||
---|---|---|---|---|---|---|---|---|---|---|
N | MEAN | SD | N | MEAN | SD | N | MEAN | SD | P | |
Attention | ||||||||||
Number recall (number correct) | 25 | 7.08 | 1.75 | 25 | 7.16 | 2.03 | 22 | 7.45 | 1.92 | 0.887 |
Number sequencing (number correct) | 26 | 6.23 | 0.98 | 25 | 5.96 | 2.65 | 23 | 5.61 | 2.29 | 0.476 |
Processing speed | ||||||||||
Animal decoding (number of errors) | 25 | 0.4 | 0.5 | 25 | 0.72 | 0.84 | 23 | 3.26 | 0.86 | <0.0001 |
Animal decoding (number correct) | 25 | 32.48 | 6.48 | 25 | 32.96 | 8.90 | 23 | 32.22 | 8.70 | 0.678 |
Symbol scanning (number correct) | 27 | 18.59 | 1.15 | 25 | 18.76 | 1.2 | 21 | 18.67 | 1.35 | 0.883 |
Symbol scanning (response time) | 27 | 4.38 | 1.37 | 25 | 4.26 | 1.66 | 21 | 3.61 | 0.84 | 0.002 |
Reaction time | ||||||||||
Response direction 1 (number of omissions) | 27 | 0.04 | 0.19 | 26 | 0.62 | 2.35 | 23 | 0 | 0 | 0.028 |
Response direction 1 (response time, seconds) | 27 | 0.52 | 0.06 | 26 | 0.55 | 0.22 | 23 | 0.52 | 0.07 | 0.567 |
Response direction 2 (number of omissions) | 27 | 0.63 | 1.33 | 26 | 0.5 | 2.18 | 23 | 0.43 | 0.95 | 0.135 |
Response direction 2 (response time, seconds) | 27 | 0.75 | 0.13 | 26 | 0.72 | 0.20 | 23 | 0.71 | 0.17 | 0.467 |
Response direction, shift failures (number) | 27 | 4.33 | 3.13 | 26 | 2.77 | 2.29 | 23 | 3.04 | 2.58 | 0.007 |
Patient-Reported Cognitive Function
The mean values and 95% confidence intervals of the patient-reported cognitive function outcomes are presented in Figure 1. Mean values remained within the low impairment range (less than 1.25) during chemotherapy.
Blood Chemistries and Toxicity
The mean values and 95% confidence intervals of significant differences in blood chemistries and toxicities are presented in [Figure 2] and [Figure 3]. Total patient-reported neurotoxicity increased significantly during chemotherapy (ANOVA; F = 6.851, P = 0.002), while several mean blood chemistry values decreased during chemotherapy treatment (hemoglobin F = 2.465, P = 0.09; white blood cell count F = 16.95, P < 0.001; platelets F = 13.72, P < 0.001; and CA-125 F = 4.91, P = 0.01). One study participant received a blood transfusion at the final course of chemotherapy, and two and three participants received cytokines (erythropoietin or darbepoietin) at course 3 and course 6, respectively.
Discussion
This study shows preliminary evidence that cognitive decline is a significant factor experienced by women who are treated for advanced ovarian cancer. Most participants self-reported mild declines, and these were detectable by a sensitive Web-based assessment tool. There are many potential mechanisms of cognitive decline during chemotherapy, ranging from oxidative damage to reduced blood oxygenation due to anemia to stress and anxiety. While it is outside of the scope of this small pilot study to examine the causative factors of decline, it does suggest the need for further investigation of the effect and potential mechanisms of cognitive decline in this population. While most of the prior work in cognitive function has been conducted among breast cancer patients, ovarian cancer patients appear to experience cognitive decline as well. There is a need to further understand this issue so that effective preventive or treatment strategies can be developed.
The significant increase in patient-reported neurotoxicity across each study visit may be a concern for computerized assessments that require dexterity. However, the keyboard proficiency tests did not decline over time, suggesting that the neurotoxicity reported by patients in this study was not great enough to affect their ability to use the computer keyboard. Patients appear to report higher levels of difficulty with memory (eg, forgetfulness) following diagnosis than following the initiation of chemotherapy; however, higher-level cognitive processes (eg, logic, organizational abilities, calculations) reported by patients appear to decline following the initiation of chemotherapy. Although larger, adequately powered trials are needed to determine the extent of this decline, this suggests that patients experience increasing challenges that may interfere with their ability to perform necessary tasks at work and in the household. Further work is needed to examine the duration of these effects following chemotherapy. Since the cognitive impact of chemotherapy reported by patients is mild, investigators must ensure the use of appropriate patient-reported tools that are able to detect these differences. While reported decline may occur, this is likely to remain within the mild category of traditional assessment tools. It is of benefit to use patient-reported tools such as the PAF that also permit the analysis of continuous data.
This study is limited by its design as a pilot study and was challenged by several logistical issues. Four patients were unable to complete all the neurocognitive evaluations. This was due to remote study staff, who would visit various clinics in the Tucson and Phoenix metropolitan regions in Arizona (range of travel more than 120 miles). The lack of completion was entirely due to communication and travel complications. When a patient was rescheduled to a different chemotherapy date, it was not always possible for this to be communicated to the Arizona Cancer Center researchers in a timely manner, resulting in missed visits. It is recommended for future studies that require strict timelines for study assessments (such as this cognitive function study) that the assessments be conducted by staff in those practices who can identify changes in infusion dates when they occur. This will reduce the communication barriers and rate of missed visits. This study was also not designed to be a comprehensive assessment of neurocognitive function but was focused on assessing three domains: attention, processing speed, and response time. It is possible that many other domains of cognitive function could be impacted by chemotherapy that were not evaluated in this study. Many patients were also taking antidepressant medications during the study; however, these were generally not new prescriptions and were also being taken at the baseline assessment. Nevertheless, future studies should incorporate assessments of mood, depression, and anxiety to account for the potential effect of these factors on cognitive assessment scores.
Despite these limitations, the study provides preliminary data demonstrating cognitive decline during chemotherapy among ovarian cancer patients treated in the front-line setting of advanced disease. More than 90% of all patients experienced measurable impairments in cognitive function during primary chemotherapy. More than half of all patients demonstrated impairment on two or more cognitive domains. Prior work has shown that even mild cognitive impairments can influence quality of life and the ability to perform routine daily activities (eg, taking medications, returning to work, managing household finances).23 The data emphasize the critical need to further understand the impact of chemotherapy on cognitive function among ovarian cancer patients so that effective preventive and treatment strategies can be developed. Additional research is needed to understand how long these declines may persist following chemotherapy treatment.
Acknowledgments
This study was funded by an investigator-initiated grant from Ortho Biotech, Inc., to the University of Arizona Cancer Center. Any opinions, findings, and conclusions or recommendations expressed in this material are those of the authors and do not necessarily reflect those of Ortho Biotech.
References1
1 J.S. Wefel, A.E. Kayl and C.A. Meyers, Neuropsychological dysfunction associated with cancer and cancer therapies: a conceptual review of an emerging target, Br J Cancer 90 (9) (2004), pp. 1691–1696. View Record in Scopus | Cited By in Scopus (48)
2 A.J. Saykin, T.A. Ahles and B.C. McDonald, Mechanisms of chemotherapy-induced cognitive disorders: neuropsychological, pathophysiological, and neuroimaging perspectives, Semin Clin Neuropsychiatry 8 (4) (2003), pp. 201–216. View Record in Scopus | Cited By in Scopus (63)
3 L.M. Hess and K.C. Insel, Chemotherapy-related change in cognitive function: a conceptual model, Oncol Nurs Forum 34 (5) (2007), pp. 981–994. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)
4 C.A. Meyers, Neurocognitive dysfunction in cancer patients, Oncology (Williston Park) 14 (1) (2000), pp. 75–79 discussion 9, 81–82, 85. View Record in Scopus | Cited By in Scopus (68)
5 M.L. Hensley, D.D. Correa and H. Thaler et al., Phase I/II study of weekly paclitaxel plus carboplatin and gemcitabine as first-line treatment of advanced-stage ovarian cancer: pathologic complete response and longitudinal assessment of impact on cognitive functioning, Gynecol Oncol 102 (2) (2006), pp. 270–277. Article | | View Record in Scopus | Cited By in Scopus (9)
6 C.A. Meyers and J.S. Wefel, The use of the mini-mental state examination to assess cognitive functioning in cancer trials: no ifs, ands, buts, or sensitivity, J Clin Oncol 21 (19) (2003), pp. 3557–3558. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (55)
7 J. Vardy, K. Wong and Q.L. Yi et al., Assessing cognitive function in cancer patients, Support Care Cancer 14 (11) (2006), pp. 1111–1118. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (27)
8 J. Vardy, S. Rourke and I.F. Tannock, Evaluation of cognitive function associated with chemotherapy: a review of published studies and recommendations for future research, J Clin Oncol 25 (17) (2007), pp. 2455–2463. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (58)
9 H.Q. Huang, M.F. Brady, D. Cella and G. Fleming, Validation and reduction of FACT/GOG-Ntx subscale for platinum/paclitaxel-induced neurologic symptoms: a Gynecologic Oncology Group study, Int J Gynecol Cancer 17 (2) (2007), pp. 387–393. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (16)
10 E.A. Calhoun, E.E. Welshman and C.H. Chang et al., Psychometric evaluation of the Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity (Fact/GOG-Ntx) questionnaire for patients receiving systemic chemotherapy, Int J Gynecol Cancer 13 (6) (2003), pp. 741–748. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (56)
11 D.M. Erlanger, D.J. Feldman, D. Kaplan and A. Theodoracopulos, Development and validation of the cognitive stability index, a Web-based protocol for monitoring change in cognitive function, Arch Clin Neuropsychol 15 (2000), pp. 693–694. Abstract | | Full Text via CrossRef
12 D.M. Erlanger, T. Kaushik, D. Broshek, J. Freeman, D. Feldman and J. Festa, Development and validation of a Web-based screening tool for monitoring cognitive status, J Head Trauma Rehabil 17 (5) (2002), pp. 458–476. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
13 G.J. Chelune, R.K. Heaton and R.A.W. Lehman, Neuropsychological and personality correlates of patients' complaints of disability. In: G. Goldstein and R.E. Tarter, Editors, Advances in Clinical Neuropsychology, Plenum Press, New York (1986).
14 C.E. Schwartz, E. Kozora and Q. Zeng, Towards patient collaboration in cognitive assessment: specificity, sensitivity and incremental validity of self-report, Ann Behav Med 18 (3) (1996), pp. 177–184. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (16)
15 K. Paraska and C.M. Bender, Cognitive dysfunction following adjuvant chemotherapy for breast cancer: two case studies, Oncol Nurs Forum 30 (3) (2003), pp. 473–478. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (15)
16 E. Kozora, M.C. Ellison and S. West, Depression, fatigue, and pain in systemic lupus erythematosus (SLE): relationship to the American College of Rheumatology SLE neuropsychological battery, Arthritis Rheum 55 (4) (2006), pp. 628–635. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (36)
17 T.A. Ahles, A.J. Saykin and C.T. Furstenberg et al., Neuropsychologic impact of standard-dose systemic chemotherapy in long-term survivors of breast cancer and lymphoma, J Clin Oncol 20 (2) (2002), pp. 485–493. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (251)
18 P. Klepstad, P. Hilton, J. Moen, B. Fougner, P.C. Borchgrevink and S. Kaasa, Self-reports are not related to objective assessments of cognitive function and sedation in patients with cancer pain admitted to a palliative care unit, Palliat Med 16 (6) (2002), pp. 513–519. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (25)
19 S.B. Schagen, F.S. van Dam, M.J. Muller, W. Boogerd, J. Lindeboom and P.F. Bruning, Cognitive deficits after postoperative adjuvant chemotherapy for breast carcinoma, Cancer 85 (3) (1999), pp. 640–650. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (263)
20 S.B. Schagen, M.J. Muller and W. Boogerd et al., Late effects of adjuvant chemotherapy on cognitive function: a follow-up study in breast cancer patients, Ann Oncol 13 (9) (2002), pp. 1387–1397. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (99)
21 F.S. van Dam, S.B. Schagen and M.J. Muller et al., Impairment of cognitive function in women receiving adjuvant treatment for high-risk breast cancer: high-dose versus standard-dose chemotherapy, J Natl Cancer Inst 90 (3) (1998), pp. 210–218. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (322)
22 D.L. Fairclough and D.F. Cella, Functional Assessment of Cancer Therapy (FACT-G): non-response to individual questions, Qual Life Res 5 (1996), pp. 321–329. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (75)
23 C.L. Burton, E. Strauss, D.F. Hultsch and M.A. Hunter, Cognitive functioning and everyday problem solving in older adults, Clin Neuropsychol 20 (3) (2006), pp. 432–452. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (20)
Correspondence to: Lisa M. Hess, PhD, Indiana University School of Medicine, Department of Public Health, 714 N Senate Avenue, Indianapolis, IN 46202; telephone: (317) 274-3148; Fax (317) 274-3443
Original research
Lisa M. Hess PhD, a,
Abstract
Change in cognitive function is increasingly being recognized as an adverse outcome related to chemotherapy treatment. These changes need not be severe to impact patient functional ability and quality of life. The primary goal of this study was to determine if there is evidence of changes in the cognitive function domains of attention, processing speed, and response time among women with newly diagnosed advanced ovarian cancer who receive chemotherapy. Eligible patients were women diagnosed with stage III–IV epithelial ovarian or primary peritoneal cancer who had not yet received chemotherapy but who were prescribed a minimum of six cycles (courses) of chemotherapy treatment. Cognitive function was assessed by a computerized, Web-based assessment (attention, processing speed, and reaction time) and by patient self-report. Cognitive function was assessed at three time points: prior to the first course (baseline), course three, and course six. Medical records were reviewed to abstract information on chemotherapy treatment, concomitant medications, and blood test results (eg, hemoglobin, CA-125). Of the 27 eligible participants, 92% and 86% demonstrated cognitive impairments from baseline to course three and from baseline to course six of chemotherapy, respectively. Impairment was detected in two or more cognitive domains among 48% (12 of 25) and 41% (9 of 22) of participants at course three and course six of chemotherapy, respectively. This study shows evidence of decline in cognitive function among women being treated for ovarian cancer. There is a need for additional, prospective research to better understand the impact of chemotherapy on cognitive function among ovarian cancer patients so that effective preventive and treatment strategies can be developed.
Article Outline
Although the perception of cognitive decline is a common complaint among individuals treated with chemotherapy, it is poorly understood and limited efforts have been made to identify the extent of this problem among women with ovarian cancer. To date, the few studies documenting the neuropsychological consequences of ovarian cancer and its treatment have shown that patients report cognitive problems but that these problems were not quantifiable using objective measures due to the lack of sensitivity of standard instruments to the subtle changes that occur during cancer treatment.[5], [6] and [7]
Although studies of cognitive function among oncology patients have used instruments that have been validated in their own disciplines and with a variety of diseases, the evidence is emerging that they are not comprehensive or appropriate tools for the detection and evaluation of chemotherapy-related change in cognitive function.8 Furthermore, the likelihood of having these tests conducted in a similar manner across multiple institutions, sites, and interviewers with any degree of consistency is very low. This study was designed as a pilot study of the identification of chemotherapy-related changes in cognitive function among women with advanced ovarian cancer using a Web-based assessment tool (Headminder, Inc., New York, NY).7 The primary goal of the current study was to determine if there is evidence of changes in the cognitive function domains of attention, processing speed, and reaction time as well as self-reported changes in the memory, sensory-perception, and cognitive-intellectual domains of cognitive function during chemotherapy among women with newly diagnosed advanced ovarian cancer.
Materials and Methods
All study methods and procedures were reviewed and approved by the University of Arizona Institutional Review Board. Eligible patients included women with a histologically or pathologically confirmed diagnosis of stage III–IV epithelial ovarian or primary peritoneal cancer who were prescribed at least six courses of platinum-based therapy. Patients were excluded if they had a prior history of any cancer (other than nonmelanoma skin cancer), chemotherapy, radiation therapy, erythropoietin treatment (within the last 6 months), or severe head injury. Initially, patients were excluded if they received intraperitoneal therapy, but the protocol was later amended to permit the use of any platinum-based therapy, regardless of route of administration.
Assessment Tools
After providing informed consent, patients completed a neurocognitive battery of tests and the Functional Assessment of Cancer Therapy—Neurotoxicity (FACT-Ntx, to assess patient-reported neuropathy).[9] and [10] The neurocognitive evaluation included both a computerized, Web-based and a patient-reported assessment. The Web-based assessment was provided by HeadMinders, Inc.[7] and [11] and was a modified version of the Cognitive Stability Index. The modified battery was comprised of two warm-up tasks and three empirically-derived cognitive factors: Processing Speed (Animal Decoding and Symbol Scanning subtests), Attention (Number Recall and Number Sequencing subtests), and Reaction Time (Response Direction 1 and Response Direction 2 subtests). The subtests have been validated against traditional neuropsychological tests in healthy and clinical populations, including cancer patients.12 Cognitive domain correlations in the battery's healthy normative sample range from 0.52 to 0.74, and correlations are similar or higher in clinical populations. Test–retest reliability of the factor scores between first and second administrations ranges from 0.74 to 0.82.12 This Web-based neurocognitive assessment tool is 21 CFR Part 11– and Health on the Net (HON)–compliant to ensure patient confidentiality. Prior to undergoing the Web-based cognitive tests, all study participants completed a keyboard proficiency test as a “warm-up task” to the computerized assessment.
The patient-reported cognitive function tool used was the Patient Assessment of Own Functioning Scale (PAF).[13], [14] and [15] The PAF includes eight scales that are grouped into the nature of the ability being considered. The Memory, Sensory-Perceptual, and Cognitive-Intellectual subscales of the PAF are included in this self-assessment questionnaire. Respondents are asked to rate on a six-point scale, from almost always to almost never, how often they experience a particular kind of difficulty in their everyday lives. For this study, the Memory and Cognitive-Intellectual subscales of the PAF were used, similar to other clinical research protocols investigating cognitive changes during chemotherapy treatment.15 The PAF has been shown to be directly related to the Minnesota Multiphasic Personality Inventory (MMPI)13 and to be highly correlated with other cognitive impairment indices, such as the American College of Rheumatology neuropsychology research battery of tests.16 Of note, self-reported cognitive change has not been shown to correlate formal assessments of cognitive function among individuals who have experienced cancer.[17], [18], [19], [20] and [21]
The FACT-Ntx is a validated instrument[9] and [10] that was used to evaluate neurotoxicity. This scale includes 11 items: nine to assess neurotoxicity, one to assess bodily weakness, and one to assess anemia. Neurotoxicity may affect a patient's ability to use the keyboard in the computerized neurocognitive evaluation. This complete assessment battery of tests was completed at baseline (within 5 days of initiation of chemotherapy) and again during follow-up assessments at cycle three and cycle six of chemotherapy. The medical record was reviewed and data were abstracted related to chemotherapy medications, all concomitant medications, and blood test results (eg, hemoglobin, CA-125).
Statistical Plan
This prospective study was exploratory in nature and designed to collect pilot data to determine if there is evidence of neurocognitive change in attention, processing speed, response time, or self-reported cognitive function during the course of chemotherapy among women being treated for advanced ovarian cancer. The purpose of this study was to obtain preliminary estimates of the incidence and degree of cognitive decline to aid in the planning of future studies. While prior estimates of cognitive function were not available for this population, power analyses demonstrate that with a target recruitment goal of 30 patients, a McNemar's test has 78% power at the 0.05 level of significance to detect a significant decline in impairment in a cognitive domain if 12 patients are found to have impairment prior to course six of treatment (but not at course three) and if as few as two patients demonstrate impairment prior to course three but not at course six. This study was therefore powered to detect declines in one or more of the domains that may have occurred at less than both of the study time points following the baseline assessment.
To be considered fully evaluable, patients had to have completed at least one follow-up neurocognitive evaluation and may not have received antipsychotic neuropsychological medications during the study (eg, chlorpromazine, haloperidol, clozapine). Antidepressants and antianxiety medications (eg, serotonin/norepinephrine reuptake inhibitors or benzodiazepines) were permitted and use was recorded throughout study participation. A summary score for each cognitive domain (processing speed, reaction time, and attention) was recorded at each assessment time point using the HeadMinder Web-based assessment. This summary score was assessed by time (processing speed and reaction time), measured to the hundredth of a second, and by number of errors (attention). If a cognitive domain summary score at a follow-up assessment time declined at least one standard error of measurement (SEM) from baseline, the patient was considered to have experienced a decline at that time point. For the purposes of this article, such declines are referred to as “impairments” within the cognitive domain under investigation. A cognitive index score (CIS) was calculated as the number of cognitive domains impaired for the time point. The range of a CIS is 0–3, with zero equal to no impairment on any cognitive domain and three equal to impairment on all cognitive domains. Patients with only one cognitive domain decline (CIS = 1) at any one of the follow-up assessment time points were considered as having possible cognitive function decline. Patients with more than one cognitive domain impairment (CIS >1) at any follow-up assessment time points were considered as having evidence of cognitive function decline. The incidence of cognitive function impairment was determined by the percentage of patients who experienced any cognitive domain impairment (including possible and evidence of decline) at any follow-up assessment.
A repeated-measures analyses of variance (ANOVA) was used to further explore the neurocognitive values at the various time points during the study. Many of the neurocognitive values were not normally distributed but skewed either positively or negatively, so the square roots of the values were used in the analyses. Since this is an exploratory analysis, no corrections for multiple comparisons were performed.
The patient-reported cognitive function instrument (PAF) contains items scored on a Likert-type scale from almost never to almost always (range 0–5). Patient-reported outcomes as measured with the PAF are measured as mean scale values, ranging from 0, indicating no impairment, to 5.0, indicating complete impairment. PAF score ranges indicate low (≤1.25), medium (1.26–1.92), and high (≥1.93) levels of cognitive impairment.13 A total FACT-Ntx score was obtained; lower scores represent greater neurotoxicity, ranging from 0 (extreme neurotoxicity) to 44 (no neurotoxicity). The total score was reported, with adjustments made for missing values as described elsewhere.22
Results
Thirty patients were enrolled in this study; however, two were later deemed ineligible, and one was unable to complete the baseline neurocognitive assessment prior to chemotherapy and was withdrawn from the study, resulting in 27 patients available for assessment. Five of these patients did not complete all neurocognitive assessments. The primary reason for nonadherence to the study schedule was clinical scheduling (eg, chemotherapy was administered prior to the neurocognitive assessment). The characteristics of eligible patients are provided in Table 1. The majority of patients were receiving intravenous chemotherapy (intraperitoneal therapy was at first not permitted but later was allowable following an amendment to the protocol) and taking concomitant sleep, antianxiety, and/or antidepressant medications outside of every 3- to 4-week chemotherapy regimen (primarily zolpidem, lorazepam, sertraline, and/or trazodone).
n = 27 | |
Mean age, years (range) | 59.3 (40.3–81.5) |
Education, n (%) | |
High school or less | 3 (11.1%) |
Some college | 12 (44.4%) |
College graduate | 12 (44.4%) |
Race/ethnicity, n (%) | |
White, non-Hispanic | 25 (92.6%) |
Hispanic | 1 (3.7%) |
Native American | 1 (3.7%) |
Marital status, n (%) | |
Married/cohabitating | 19 (70.4%) |
Divorced/separated | 1 (3.7%) |
Widowed | 5 (18.5%) |
Never married | 2 (7.4%) |
Mean courses of chemotherapy, n (range) | 5.9 (4–6) |
Chemotherapy route, n (%) | |
Intraperitoneal | 5 (18.5%) |
Intravenous | 22 (81.5%) |
Concurrent medication use, n (%) | |
Antidepressant | 7 (25.9%) |
Antianxiety | 16 (59.3%) |
Sleep aids | 5 (18.5%) |
Web-Assessed Cognitive Function
Keyboard proficiency remained unchanged over time (P = 0.39). As shown in Table 2, most participants demonstrated cognitive impairments in at least one of the three cognitive domains assessed during this study (92% and 86% at course 3 and course 6, respectively). Nearly half of the study participants demonstrated impairment from baseline in two or more of the three cognitive domains assessed (Table 3). Table 4 shows a detailed summary of the subscales within the Web-based cognitive tests that comprised the CIS.This table demonstrates the statistically significant increase in test subscale errors, despite the test-taking improvements over time, as shown by reduction in testing time.
CIS | COURSE 3 | COURSE 6 |
---|---|---|
No decline (CIS = 0) | 2 (8%) | 3 (14%) |
One impairment (CIS = 1) | 11 (44%) | 10 (45%) |
Two impairments (CIS = 2) | 11 (44%) | 7 (32%) |
Three impairments (CIS = 3) | 1 (4%) | 2 (9%) |
COGNITIVE IMPAIRMENT SCALE (CIS) FACTORS | BASELINE | COURSE 3 | COURSE 6 | |||||||
---|---|---|---|---|---|---|---|---|---|---|
N | MEAN | SD | N | MEAN | SD | N | MEAN | SD | P | |
Attention | ||||||||||
Number recall (number correct) | 25 | 7.08 | 1.75 | 25 | 7.16 | 2.03 | 22 | 7.45 | 1.92 | 0.887 |
Number sequencing (number correct) | 26 | 6.23 | 0.98 | 25 | 5.96 | 2.65 | 23 | 5.61 | 2.29 | 0.476 |
Processing speed | ||||||||||
Animal decoding (number of errors) | 25 | 0.4 | 0.5 | 25 | 0.72 | 0.84 | 23 | 3.26 | 0.86 | <0.0001 |
Animal decoding (number correct) | 25 | 32.48 | 6.48 | 25 | 32.96 | 8.90 | 23 | 32.22 | 8.70 | 0.678 |
Symbol scanning (number correct) | 27 | 18.59 | 1.15 | 25 | 18.76 | 1.2 | 21 | 18.67 | 1.35 | 0.883 |
Symbol scanning (response time) | 27 | 4.38 | 1.37 | 25 | 4.26 | 1.66 | 21 | 3.61 | 0.84 | 0.002 |
Reaction time | ||||||||||
Response direction 1 (number of omissions) | 27 | 0.04 | 0.19 | 26 | 0.62 | 2.35 | 23 | 0 | 0 | 0.028 |
Response direction 1 (response time, seconds) | 27 | 0.52 | 0.06 | 26 | 0.55 | 0.22 | 23 | 0.52 | 0.07 | 0.567 |
Response direction 2 (number of omissions) | 27 | 0.63 | 1.33 | 26 | 0.5 | 2.18 | 23 | 0.43 | 0.95 | 0.135 |
Response direction 2 (response time, seconds) | 27 | 0.75 | 0.13 | 26 | 0.72 | 0.20 | 23 | 0.71 | 0.17 | 0.467 |
Response direction, shift failures (number) | 27 | 4.33 | 3.13 | 26 | 2.77 | 2.29 | 23 | 3.04 | 2.58 | 0.007 |
Patient-Reported Cognitive Function
The mean values and 95% confidence intervals of the patient-reported cognitive function outcomes are presented in Figure 1. Mean values remained within the low impairment range (less than 1.25) during chemotherapy.
Blood Chemistries and Toxicity
The mean values and 95% confidence intervals of significant differences in blood chemistries and toxicities are presented in [Figure 2] and [Figure 3]. Total patient-reported neurotoxicity increased significantly during chemotherapy (ANOVA; F = 6.851, P = 0.002), while several mean blood chemistry values decreased during chemotherapy treatment (hemoglobin F = 2.465, P = 0.09; white blood cell count F = 16.95, P < 0.001; platelets F = 13.72, P < 0.001; and CA-125 F = 4.91, P = 0.01). One study participant received a blood transfusion at the final course of chemotherapy, and two and three participants received cytokines (erythropoietin or darbepoietin) at course 3 and course 6, respectively.
Discussion
This study shows preliminary evidence that cognitive decline is a significant factor experienced by women who are treated for advanced ovarian cancer. Most participants self-reported mild declines, and these were detectable by a sensitive Web-based assessment tool. There are many potential mechanisms of cognitive decline during chemotherapy, ranging from oxidative damage to reduced blood oxygenation due to anemia to stress and anxiety. While it is outside of the scope of this small pilot study to examine the causative factors of decline, it does suggest the need for further investigation of the effect and potential mechanisms of cognitive decline in this population. While most of the prior work in cognitive function has been conducted among breast cancer patients, ovarian cancer patients appear to experience cognitive decline as well. There is a need to further understand this issue so that effective preventive or treatment strategies can be developed.
The significant increase in patient-reported neurotoxicity across each study visit may be a concern for computerized assessments that require dexterity. However, the keyboard proficiency tests did not decline over time, suggesting that the neurotoxicity reported by patients in this study was not great enough to affect their ability to use the computer keyboard. Patients appear to report higher levels of difficulty with memory (eg, forgetfulness) following diagnosis than following the initiation of chemotherapy; however, higher-level cognitive processes (eg, logic, organizational abilities, calculations) reported by patients appear to decline following the initiation of chemotherapy. Although larger, adequately powered trials are needed to determine the extent of this decline, this suggests that patients experience increasing challenges that may interfere with their ability to perform necessary tasks at work and in the household. Further work is needed to examine the duration of these effects following chemotherapy. Since the cognitive impact of chemotherapy reported by patients is mild, investigators must ensure the use of appropriate patient-reported tools that are able to detect these differences. While reported decline may occur, this is likely to remain within the mild category of traditional assessment tools. It is of benefit to use patient-reported tools such as the PAF that also permit the analysis of continuous data.
This study is limited by its design as a pilot study and was challenged by several logistical issues. Four patients were unable to complete all the neurocognitive evaluations. This was due to remote study staff, who would visit various clinics in the Tucson and Phoenix metropolitan regions in Arizona (range of travel more than 120 miles). The lack of completion was entirely due to communication and travel complications. When a patient was rescheduled to a different chemotherapy date, it was not always possible for this to be communicated to the Arizona Cancer Center researchers in a timely manner, resulting in missed visits. It is recommended for future studies that require strict timelines for study assessments (such as this cognitive function study) that the assessments be conducted by staff in those practices who can identify changes in infusion dates when they occur. This will reduce the communication barriers and rate of missed visits. This study was also not designed to be a comprehensive assessment of neurocognitive function but was focused on assessing three domains: attention, processing speed, and response time. It is possible that many other domains of cognitive function could be impacted by chemotherapy that were not evaluated in this study. Many patients were also taking antidepressant medications during the study; however, these were generally not new prescriptions and were also being taken at the baseline assessment. Nevertheless, future studies should incorporate assessments of mood, depression, and anxiety to account for the potential effect of these factors on cognitive assessment scores.
Despite these limitations, the study provides preliminary data demonstrating cognitive decline during chemotherapy among ovarian cancer patients treated in the front-line setting of advanced disease. More than 90% of all patients experienced measurable impairments in cognitive function during primary chemotherapy. More than half of all patients demonstrated impairment on two or more cognitive domains. Prior work has shown that even mild cognitive impairments can influence quality of life and the ability to perform routine daily activities (eg, taking medications, returning to work, managing household finances).23 The data emphasize the critical need to further understand the impact of chemotherapy on cognitive function among ovarian cancer patients so that effective preventive and treatment strategies can be developed. Additional research is needed to understand how long these declines may persist following chemotherapy treatment.
Acknowledgments
This study was funded by an investigator-initiated grant from Ortho Biotech, Inc., to the University of Arizona Cancer Center. Any opinions, findings, and conclusions or recommendations expressed in this material are those of the authors and do not necessarily reflect those of Ortho Biotech.
References1
1 J.S. Wefel, A.E. Kayl and C.A. Meyers, Neuropsychological dysfunction associated with cancer and cancer therapies: a conceptual review of an emerging target, Br J Cancer 90 (9) (2004), pp. 1691–1696. View Record in Scopus | Cited By in Scopus (48)
2 A.J. Saykin, T.A. Ahles and B.C. McDonald, Mechanisms of chemotherapy-induced cognitive disorders: neuropsychological, pathophysiological, and neuroimaging perspectives, Semin Clin Neuropsychiatry 8 (4) (2003), pp. 201–216. View Record in Scopus | Cited By in Scopus (63)
3 L.M. Hess and K.C. Insel, Chemotherapy-related change in cognitive function: a conceptual model, Oncol Nurs Forum 34 (5) (2007), pp. 981–994. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)
4 C.A. Meyers, Neurocognitive dysfunction in cancer patients, Oncology (Williston Park) 14 (1) (2000), pp. 75–79 discussion 9, 81–82, 85. View Record in Scopus | Cited By in Scopus (68)
5 M.L. Hensley, D.D. Correa and H. Thaler et al., Phase I/II study of weekly paclitaxel plus carboplatin and gemcitabine as first-line treatment of advanced-stage ovarian cancer: pathologic complete response and longitudinal assessment of impact on cognitive functioning, Gynecol Oncol 102 (2) (2006), pp. 270–277. Article | | View Record in Scopus | Cited By in Scopus (9)
6 C.A. Meyers and J.S. Wefel, The use of the mini-mental state examination to assess cognitive functioning in cancer trials: no ifs, ands, buts, or sensitivity, J Clin Oncol 21 (19) (2003), pp. 3557–3558. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (55)
7 J. Vardy, K. Wong and Q.L. Yi et al., Assessing cognitive function in cancer patients, Support Care Cancer 14 (11) (2006), pp. 1111–1118. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (27)
8 J. Vardy, S. Rourke and I.F. Tannock, Evaluation of cognitive function associated with chemotherapy: a review of published studies and recommendations for future research, J Clin Oncol 25 (17) (2007), pp. 2455–2463. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (58)
9 H.Q. Huang, M.F. Brady, D. Cella and G. Fleming, Validation and reduction of FACT/GOG-Ntx subscale for platinum/paclitaxel-induced neurologic symptoms: a Gynecologic Oncology Group study, Int J Gynecol Cancer 17 (2) (2007), pp. 387–393. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (16)
10 E.A. Calhoun, E.E. Welshman and C.H. Chang et al., Psychometric evaluation of the Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity (Fact/GOG-Ntx) questionnaire for patients receiving systemic chemotherapy, Int J Gynecol Cancer 13 (6) (2003), pp. 741–748. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (56)
11 D.M. Erlanger, D.J. Feldman, D. Kaplan and A. Theodoracopulos, Development and validation of the cognitive stability index, a Web-based protocol for monitoring change in cognitive function, Arch Clin Neuropsychol 15 (2000), pp. 693–694. Abstract | | Full Text via CrossRef
12 D.M. Erlanger, T. Kaushik, D. Broshek, J. Freeman, D. Feldman and J. Festa, Development and validation of a Web-based screening tool for monitoring cognitive status, J Head Trauma Rehabil 17 (5) (2002), pp. 458–476. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
13 G.J. Chelune, R.K. Heaton and R.A.W. Lehman, Neuropsychological and personality correlates of patients' complaints of disability. In: G. Goldstein and R.E. Tarter, Editors, Advances in Clinical Neuropsychology, Plenum Press, New York (1986).
14 C.E. Schwartz, E. Kozora and Q. Zeng, Towards patient collaboration in cognitive assessment: specificity, sensitivity and incremental validity of self-report, Ann Behav Med 18 (3) (1996), pp. 177–184. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (16)
15 K. Paraska and C.M. Bender, Cognitive dysfunction following adjuvant chemotherapy for breast cancer: two case studies, Oncol Nurs Forum 30 (3) (2003), pp. 473–478. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (15)
16 E. Kozora, M.C. Ellison and S. West, Depression, fatigue, and pain in systemic lupus erythematosus (SLE): relationship to the American College of Rheumatology SLE neuropsychological battery, Arthritis Rheum 55 (4) (2006), pp. 628–635. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (36)
17 T.A. Ahles, A.J. Saykin and C.T. Furstenberg et al., Neuropsychologic impact of standard-dose systemic chemotherapy in long-term survivors of breast cancer and lymphoma, J Clin Oncol 20 (2) (2002), pp. 485–493. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (251)
18 P. Klepstad, P. Hilton, J. Moen, B. Fougner, P.C. Borchgrevink and S. Kaasa, Self-reports are not related to objective assessments of cognitive function and sedation in patients with cancer pain admitted to a palliative care unit, Palliat Med 16 (6) (2002), pp. 513–519. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (25)
19 S.B. Schagen, F.S. van Dam, M.J. Muller, W. Boogerd, J. Lindeboom and P.F. Bruning, Cognitive deficits after postoperative adjuvant chemotherapy for breast carcinoma, Cancer 85 (3) (1999), pp. 640–650. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (263)
20 S.B. Schagen, M.J. Muller and W. Boogerd et al., Late effects of adjuvant chemotherapy on cognitive function: a follow-up study in breast cancer patients, Ann Oncol 13 (9) (2002), pp. 1387–1397. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (99)
21 F.S. van Dam, S.B. Schagen and M.J. Muller et al., Impairment of cognitive function in women receiving adjuvant treatment for high-risk breast cancer: high-dose versus standard-dose chemotherapy, J Natl Cancer Inst 90 (3) (1998), pp. 210–218. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (322)
22 D.L. Fairclough and D.F. Cella, Functional Assessment of Cancer Therapy (FACT-G): non-response to individual questions, Qual Life Res 5 (1996), pp. 321–329. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (75)
23 C.L. Burton, E. Strauss, D.F. Hultsch and M.A. Hunter, Cognitive functioning and everyday problem solving in older adults, Clin Neuropsychol 20 (3) (2006), pp. 432–452. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (20)
Correspondence to: Lisa M. Hess, PhD, Indiana University School of Medicine, Department of Public Health, 714 N Senate Avenue, Indianapolis, IN 46202; telephone: (317) 274-3148; Fax (317) 274-3443
Original research
Lisa M. Hess PhD, a, , Setsuko K. Chambers MDa, Kenneth Hatch MDa, Alton Hallum MDa, Mike F. Janicek MDa, Joseph Buscema MDa, Matthew Borst MDa, Cynthia Johnson MAa, Lisa Slayton LPNa, Yuda Chongpison MS, MBAa and David S. Alberts MDa
Abstract
Change in cognitive function is increasingly being recognized as an adverse outcome related to chemotherapy treatment. These changes need not be severe to impact patient functional ability and quality of life. The primary goal of this study was to determine if there is evidence of changes in the cognitive function domains of attention, processing speed, and response time among women with newly diagnosed advanced ovarian cancer who receive chemotherapy. Eligible patients were women diagnosed with stage III–IV epithelial ovarian or primary peritoneal cancer who had not yet received chemotherapy but who were prescribed a minimum of six cycles (courses) of chemotherapy treatment. Cognitive function was assessed by a computerized, Web-based assessment (attention, processing speed, and reaction time) and by patient self-report. Cognitive function was assessed at three time points: prior to the first course (baseline), course three, and course six. Medical records were reviewed to abstract information on chemotherapy treatment, concomitant medications, and blood test results (eg, hemoglobin, CA-125). Of the 27 eligible participants, 92% and 86% demonstrated cognitive impairments from baseline to course three and from baseline to course six of chemotherapy, respectively. Impairment was detected in two or more cognitive domains among 48% (12 of 25) and 41% (9 of 22) of participants at course three and course six of chemotherapy, respectively. This study shows evidence of decline in cognitive function among women being treated for ovarian cancer. There is a need for additional, prospective research to better understand the impact of chemotherapy on cognitive function among ovarian cancer patients so that effective preventive and treatment strategies can be developed.
Article Outline
Although the perception of cognitive decline is a common complaint among individuals treated with chemotherapy, it is poorly understood and limited efforts have been made to identify the extent of this problem among women with ovarian cancer. To date, the few studies documenting the neuropsychological consequences of ovarian cancer and its treatment have shown that patients report cognitive problems but that these problems were not quantifiable using objective measures due to the lack of sensitivity of standard instruments to the subtle changes that occur during cancer treatment.[5], [6] and [7]
Although studies of cognitive function among oncology patients have used instruments that have been validated in their own disciplines and with a variety of diseases, the evidence is emerging that they are not comprehensive or appropriate tools for the detection and evaluation of chemotherapy-related change in cognitive function.8 Furthermore, the likelihood of having these tests conducted in a similar manner across multiple institutions, sites, and interviewers with any degree of consistency is very low. This study was designed as a pilot study of the identification of chemotherapy-related changes in cognitive function among women with advanced ovarian cancer using a Web-based assessment tool (Headminder, Inc., New York, NY).7 The primary goal of the current study was to determine if there is evidence of changes in the cognitive function domains of attention, processing speed, and reaction time as well as self-reported changes in the memory, sensory-perception, and cognitive-intellectual domains of cognitive function during chemotherapy among women with newly diagnosed advanced ovarian cancer.
Materials and Methods
All study methods and procedures were reviewed and approved by the University of Arizona Institutional Review Board. Eligible patients included women with a histologically or pathologically confirmed diagnosis of stage III–IV epithelial ovarian or primary peritoneal cancer who were prescribed at least six courses of platinum-based therapy. Patients were excluded if they had a prior history of any cancer (other than nonmelanoma skin cancer), chemotherapy, radiation therapy, erythropoietin treatment (within the last 6 months), or severe head injury. Initially, patients were excluded if they received intraperitoneal therapy, but the protocol was later amended to permit the use of any platinum-based therapy, regardless of route of administration.
Assessment Tools
After providing informed consent, patients completed a neurocognitive battery of tests and the Functional Assessment of Cancer Therapy—Neurotoxicity (FACT-Ntx, to assess patient-reported neuropathy).[9] and [10] The neurocognitive evaluation included both a computerized, Web-based and a patient-reported assessment. The Web-based assessment was provided by HeadMinders, Inc.[7] and [11] and was a modified version of the Cognitive Stability Index. The modified battery was comprised of two warm-up tasks and three empirically-derived cognitive factors: Processing Speed (Animal Decoding and Symbol Scanning subtests), Attention (Number Recall and Number Sequencing subtests), and Reaction Time (Response Direction 1 and Response Direction 2 subtests). The subtests have been validated against traditional neuropsychological tests in healthy and clinical populations, including cancer patients.12 Cognitive domain correlations in the battery's healthy normative sample range from 0.52 to 0.74, and correlations are similar or higher in clinical populations. Test–retest reliability of the factor scores between first and second administrations ranges from 0.74 to 0.82.12 This Web-based neurocognitive assessment tool is 21 CFR Part 11– and Health on the Net (HON)–compliant to ensure patient confidentiality. Prior to undergoing the Web-based cognitive tests, all study participants completed a keyboard proficiency test as a “warm-up task” to the computerized assessment.
The patient-reported cognitive function tool used was the Patient Assessment of Own Functioning Scale (PAF).[13], [14] and [15] The PAF includes eight scales that are grouped into the nature of the ability being considered. The Memory, Sensory-Perceptual, and Cognitive-Intellectual subscales of the PAF are included in this self-assessment questionnaire. Respondents are asked to rate on a six-point scale, from almost always to almost never, how often they experience a particular kind of difficulty in their everyday lives. For this study, the Memory and Cognitive-Intellectual subscales of the PAF were used, similar to other clinical research protocols investigating cognitive changes during chemotherapy treatment.15 The PAF has been shown to be directly related to the Minnesota Multiphasic Personality Inventory (MMPI)13 and to be highly correlated with other cognitive impairment indices, such as the American College of Rheumatology neuropsychology research battery of tests.16 Of note, self-reported cognitive change has not been shown to correlate formal assessments of cognitive function among individuals who have experienced cancer.[17], [18], [19], [20] and [21]
The FACT-Ntx is a validated instrument[9] and [10] that was used to evaluate neurotoxicity. This scale includes 11 items: nine to assess neurotoxicity, one to assess bodily weakness, and one to assess anemia. Neurotoxicity may affect a patient's ability to use the keyboard in the computerized neurocognitive evaluation. This complete assessment battery of tests was completed at baseline (within 5 days of initiation of chemotherapy) and again during follow-up assessments at cycle three and cycle six of chemotherapy. The medical record was reviewed and data were abstracted related to chemotherapy medications, all concomitant medications, and blood test results (eg, hemoglobin, CA-125).
Statistical Plan
This prospective study was exploratory in nature and designed to collect pilot data to determine if there is evidence of neurocognitive change in attention, processing speed, response time, or self-reported cognitive function during the course of chemotherapy among women being treated for advanced ovarian cancer. The purpose of this study was to obtain preliminary estimates of the incidence and degree of cognitive decline to aid in the planning of future studies. While prior estimates of cognitive function were not available for this population, power analyses demonstrate that with a target recruitment goal of 30 patients, a McNemar's test has 78% power at the 0.05 level of significance to detect a significant decline in impairment in a cognitive domain if 12 patients are found to have impairment prior to course six of treatment (but not at course three) and if as few as two patients demonstrate impairment prior to course three but not at course six. This study was therefore powered to detect declines in one or more of the domains that may have occurred at less than both of the study time points following the baseline assessment.
To be considered fully evaluable, patients had to have completed at least one follow-up neurocognitive evaluation and may not have received antipsychotic neuropsychological medications during the study (eg, chlorpromazine, haloperidol, clozapine). Antidepressants and antianxiety medications (eg, serotonin/norepinephrine reuptake inhibitors or benzodiazepines) were permitted and use was recorded throughout study participation. A summary score for each cognitive domain (processing speed, reaction time, and attention) was recorded at each assessment time point using the HeadMinder Web-based assessment. This summary score was assessed by time (processing speed and reaction time), measured to the hundredth of a second, and by number of errors (attention). If a cognitive domain summary score at a follow-up assessment time declined at least one standard error of measurement (SEM) from baseline, the patient was considered to have experienced a decline at that time point. For the purposes of this article, such declines are referred to as “impairments” within the cognitive domain under investigation. A cognitive index score (CIS) was calculated as the number of cognitive domains impaired for the time point. The range of a CIS is 0–3, with zero equal to no impairment on any cognitive domain and three equal to impairment on all cognitive domains. Patients with only one cognitive domain decline (CIS = 1) at any one of the follow-up assessment time points were considered as having possible cognitive function decline. Patients with more than one cognitive domain impairment (CIS >1) at any follow-up assessment time points were considered as having evidence of cognitive function decline. The incidence of cognitive function impairment was determined by the percentage of patients who experienced any cognitive domain impairment (including possible and evidence of decline) at any follow-up assessment.
A repeated-measures analyses of variance (ANOVA) was used to further explore the neurocognitive values at the various time points during the study. Many of the neurocognitive values were not normally distributed but skewed either positively or negatively, so the square roots of the values were used in the analyses. Since this is an exploratory analysis, no corrections for multiple comparisons were performed.
The patient-reported cognitive function instrument (PAF) contains items scored on a Likert-type scale from almost never to almost always (range 0–5). Patient-reported outcomes as measured with the PAF are measured as mean scale values, ranging from 0, indicating no impairment, to 5.0, indicating complete impairment. PAF score ranges indicate low (≤1.25), medium (1.26–1.92), and high (≥1.93) levels of cognitive impairment.13 A total FACT-Ntx score was obtained; lower scores represent greater neurotoxicity, ranging from 0 (extreme neurotoxicity) to 44 (no neurotoxicity). The total score was reported, with adjustments made for missing values as described elsewhere.22
Results
Thirty patients were enrolled in this study; however, two were later deemed ineligible, and one was unable to complete the baseline neurocognitive assessment prior to chemotherapy and was withdrawn from the study, resulting in 27 patients available for assessment. Five of these patients did not complete all neurocognitive assessments. The primary reason for nonadherence to the study schedule was clinical scheduling (eg, chemotherapy was administered prior to the neurocognitive assessment). The characteristics of eligible patients are provided in Table 1. The majority of patients were receiving intravenous chemotherapy (intraperitoneal therapy was at first not permitted but later was allowable following an amendment to the protocol) and taking concomitant sleep, antianxiety, and/or antidepressant medications outside of every 3- to 4-week chemotherapy regimen (primarily zolpidem, lorazepam, sertraline, and/or trazodone).
n = 27 | |
Mean age, years (range) | 59.3 (40.3–81.5) |
Education, n (%) | |
High school or less | 3 (11.1%) |
Some college | 12 (44.4%) |
College graduate | 12 (44.4%) |
Race/ethnicity, n (%) | |
White, non-Hispanic | 25 (92.6%) |
Hispanic | 1 (3.7%) |
Native American | 1 (3.7%) |
Marital status, n (%) | |
Married/cohabitating | 19 (70.4%) |
Divorced/separated | 1 (3.7%) |
Widowed | 5 (18.5%) |
Never married | 2 (7.4%) |
Mean courses of chemotherapy, n (range) | 5.9 (4–6) |
Chemotherapy route, n (%) | |
Intraperitoneal | 5 (18.5%) |
Intravenous | 22 (81.5%) |
Concurrent medication use, n (%) | |
Antidepressant | 7 (25.9%) |
Antianxiety | 16 (59.3%) |
Sleep aids | 5 (18.5%) |
Web-Assessed Cognitive Function
Keyboard proficiency remained unchanged over time (P = 0.39). As shown in Table 2, most participants demonstrated cognitive impairments in at least one of the three cognitive domains assessed during this study (92% and 86% at course 3 and course 6, respectively). Nearly half of the study participants demonstrated impairment from baseline in two or more of the three cognitive domains assessed (Table 3). Table 4 shows a detailed summary of the subscales within the Web-based cognitive tests that comprised the CIS.This table demonstrates the statistically significant increase in test subscale errors, despite the test-taking improvements over time, as shown by reduction in testing time.
CIS | COURSE 3 | COURSE 6 |
---|---|---|
No decline (CIS = 0) | 2 (8%) | 3 (14%) |
One impairment (CIS = 1) | 11 (44%) | 10 (45%) |
Two impairments (CIS = 2) | 11 (44%) | 7 (32%) |
Three impairments (CIS = 3) | 1 (4%) | 2 (9%) |
COGNITIVE IMPAIRMENT SCALE (CIS) FACTORS | BASELINE | COURSE 3 | COURSE 6 | |||||||
---|---|---|---|---|---|---|---|---|---|---|
N | MEAN | SD | N | MEAN | SD | N | MEAN | SD | P | |
Attention | ||||||||||
Number recall (number correct) | 25 | 7.08 | 1.75 | 25 | 7.16 | 2.03 | 22 | 7.45 | 1.92 | 0.887 |
Number sequencing (number correct) | 26 | 6.23 | 0.98 | 25 | 5.96 | 2.65 | 23 | 5.61 | 2.29 | 0.476 |
Processing speed | ||||||||||
Animal decoding (number of errors) | 25 | 0.4 | 0.5 | 25 | 0.72 | 0.84 | 23 | 3.26 | 0.86 | <0.0001 |
Animal decoding (number correct) | 25 | 32.48 | 6.48 | 25 | 32.96 | 8.90 | 23 | 32.22 | 8.70 | 0.678 |
Symbol scanning (number correct) | 27 | 18.59 | 1.15 | 25 | 18.76 | 1.2 | 21 | 18.67 | 1.35 | 0.883 |
Symbol scanning (response time) | 27 | 4.38 | 1.37 | 25 | 4.26 | 1.66 | 21 | 3.61 | 0.84 | 0.002 |
Reaction time | ||||||||||
Response direction 1 (number of omissions) | 27 | 0.04 | 0.19 | 26 | 0.62 | 2.35 | 23 | 0 | 0 | 0.028 |
Response direction 1 (response time, seconds) | 27 | 0.52 | 0.06 | 26 | 0.55 | 0.22 | 23 | 0.52 | 0.07 | 0.567 |
Response direction 2 (number of omissions) | 27 | 0.63 | 1.33 | 26 | 0.5 | 2.18 | 23 | 0.43 | 0.95 | 0.135 |
Response direction 2 (response time, seconds) | 27 | 0.75 | 0.13 | 26 | 0.72 | 0.20 | 23 | 0.71 | 0.17 | 0.467 |
Response direction, shift failures (number) | 27 | 4.33 | 3.13 | 26 | 2.77 | 2.29 | 23 | 3.04 | 2.58 | 0.007 |
Patient-Reported Cognitive Function
The mean values and 95% confidence intervals of the patient-reported cognitive function outcomes are presented in Figure 1. Mean values remained within the low impairment range (less than 1.25) during chemotherapy.
Blood Chemistries and Toxicity
The mean values and 95% confidence intervals of significant differences in blood chemistries and toxicities are presented in [Figure 2] and [Figure 3]. Total patient-reported neurotoxicity increased significantly during chemotherapy (ANOVA; F = 6.851, P = 0.002), while several mean blood chemistry values decreased during chemotherapy treatment (hemoglobin F = 2.465, P = 0.09; white blood cell count F = 16.95, P < 0.001; platelets F = 13.72, P < 0.001; and CA-125 F = 4.91, P = 0.01). One study participant received a blood transfusion at the final course of chemotherapy, and two and three participants received cytokines (erythropoietin or darbepoietin) at course 3 and course 6, respectively.
Discussion
This study shows preliminary evidence that cognitive decline is a significant factor experienced by women who are treated for advanced ovarian cancer. Most participants self-reported mild declines, and these were detectable by a sensitive Web-based assessment tool. There are many potential mechanisms of cognitive decline during chemotherapy, ranging from oxidative damage to reduced blood oxygenation due to anemia to stress and anxiety. While it is outside of the scope of this small pilot study to examine the causative factors of decline, it does suggest the need for further investigation of the effect and potential mechanisms of cognitive decline in this population. While most of the prior work in cognitive function has been conducted among breast cancer patients, ovarian cancer patients appear to experience cognitive decline as well. There is a need to further understand this issue so that effective preventive or treatment strategies can be developed.
The significant increase in patient-reported neurotoxicity across each study visit may be a concern for computerized assessments that require dexterity. However, the keyboard proficiency tests did not decline over time, suggesting that the neurotoxicity reported by patients in this study was not great enough to affect their ability to use the computer keyboard. Patients appear to report higher levels of difficulty with memory (eg, forgetfulness) following diagnosis than following the initiation of chemotherapy; however, higher-level cognitive processes (eg, logic, organizational abilities, calculations) reported by patients appear to decline following the initiation of chemotherapy. Although larger, adequately powered trials are needed to determine the extent of this decline, this suggests that patients experience increasing challenges that may interfere with their ability to perform necessary tasks at work and in the household. Further work is needed to examine the duration of these effects following chemotherapy. Since the cognitive impact of chemotherapy reported by patients is mild, investigators must ensure the use of appropriate patient-reported tools that are able to detect these differences. While reported decline may occur, this is likely to remain within the mild category of traditional assessment tools. It is of benefit to use patient-reported tools such as the PAF that also permit the analysis of continuous data.
This study is limited by its design as a pilot study and was challenged by several logistical issues. Four patients were unable to complete all the neurocognitive evaluations. This was due to remote study staff, who would visit various clinics in the Tucson and Phoenix metropolitan regions in Arizona (range of travel more than 120 miles). The lack of completion was entirely due to communication and travel complications. When a patient was rescheduled to a different chemotherapy date, it was not always possible for this to be communicated to the Arizona Cancer Center researchers in a timely manner, resulting in missed visits. It is recommended for future studies that require strict timelines for study assessments (such as this cognitive function study) that the assessments be conducted by staff in those practices who can identify changes in infusion dates when they occur. This will reduce the communication barriers and rate of missed visits. This study was also not designed to be a comprehensive assessment of neurocognitive function but was focused on assessing three domains: attention, processing speed, and response time. It is possible that many other domains of cognitive function could be impacted by chemotherapy that were not evaluated in this study. Many patients were also taking antidepressant medications during the study; however, these were generally not new prescriptions and were also being taken at the baseline assessment. Nevertheless, future studies should incorporate assessments of mood, depression, and anxiety to account for the potential effect of these factors on cognitive assessment scores.
Despite these limitations, the study provides preliminary data demonstrating cognitive decline during chemotherapy among ovarian cancer patients treated in the front-line setting of advanced disease. More than 90% of all patients experienced measurable impairments in cognitive function during primary chemotherapy. More than half of all patients demonstrated impairment on two or more cognitive domains. Prior work has shown that even mild cognitive impairments can influence quality of life and the ability to perform routine daily activities (eg, taking medications, returning to work, managing household finances).23 The data emphasize the critical need to further understand the impact of chemotherapy on cognitive function among ovarian cancer patients so that effective preventive and treatment strategies can be developed. Additional research is needed to understand how long these declines may persist following chemotherapy treatment.
Acknowledgments
This study was funded by an investigator-initiated grant from Ortho Biotech, Inc., to the University of Arizona Cancer Center. Any opinions, findings, and conclusions or recommendations expressed in this material are those of the authors and do not necessarily reflect those of Ortho Biotech.
References1
1 J.S. Wefel, A.E. Kayl and C.A. Meyers, Neuropsychological dysfunction associated with cancer and cancer therapies: a conceptual review of an emerging target, Br J Cancer 90 (9) (2004), pp. 1691–1696. View Record in Scopus | Cited By in Scopus (48)
2 A.J. Saykin, T.A. Ahles and B.C. McDonald, Mechanisms of chemotherapy-induced cognitive disorders: neuropsychological, pathophysiological, and neuroimaging perspectives, Semin Clin Neuropsychiatry 8 (4) (2003), pp. 201–216. View Record in Scopus | Cited By in Scopus (63)
3 L.M. Hess and K.C. Insel, Chemotherapy-related change in cognitive function: a conceptual model, Oncol Nurs Forum 34 (5) (2007), pp. 981–994. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)
4 C.A. Meyers, Neurocognitive dysfunction in cancer patients, Oncology (Williston Park) 14 (1) (2000), pp. 75–79 discussion 9, 81–82, 85. View Record in Scopus | Cited By in Scopus (68)
5 M.L. Hensley, D.D. Correa and H. Thaler et al., Phase I/II study of weekly paclitaxel plus carboplatin and gemcitabine as first-line treatment of advanced-stage ovarian cancer: pathologic complete response and longitudinal assessment of impact on cognitive functioning, Gynecol Oncol 102 (2) (2006), pp. 270–277. Article | | View Record in Scopus | Cited By in Scopus (9)
6 C.A. Meyers and J.S. Wefel, The use of the mini-mental state examination to assess cognitive functioning in cancer trials: no ifs, ands, buts, or sensitivity, J Clin Oncol 21 (19) (2003), pp. 3557–3558. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (55)
7 J. Vardy, K. Wong and Q.L. Yi et al., Assessing cognitive function in cancer patients, Support Care Cancer 14 (11) (2006), pp. 1111–1118. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (27)
8 J. Vardy, S. Rourke and I.F. Tannock, Evaluation of cognitive function associated with chemotherapy: a review of published studies and recommendations for future research, J Clin Oncol 25 (17) (2007), pp. 2455–2463. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (58)
9 H.Q. Huang, M.F. Brady, D. Cella and G. Fleming, Validation and reduction of FACT/GOG-Ntx subscale for platinum/paclitaxel-induced neurologic symptoms: a Gynecologic Oncology Group study, Int J Gynecol Cancer 17 (2) (2007), pp. 387–393. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (16)
10 E.A. Calhoun, E.E. Welshman and C.H. Chang et al., Psychometric evaluation of the Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity (Fact/GOG-Ntx) questionnaire for patients receiving systemic chemotherapy, Int J Gynecol Cancer 13 (6) (2003), pp. 741–748. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (56)
11 D.M. Erlanger, D.J. Feldman, D. Kaplan and A. Theodoracopulos, Development and validation of the cognitive stability index, a Web-based protocol for monitoring change in cognitive function, Arch Clin Neuropsychol 15 (2000), pp. 693–694. Abstract | | Full Text via CrossRef
12 D.M. Erlanger, T. Kaushik, D. Broshek, J. Freeman, D. Feldman and J. Festa, Development and validation of a Web-based screening tool for monitoring cognitive status, J Head Trauma Rehabil 17 (5) (2002), pp. 458–476. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
13 G.J. Chelune, R.K. Heaton and R.A.W. Lehman, Neuropsychological and personality correlates of patients' complaints of disability. In: G. Goldstein and R.E. Tarter, Editors, Advances in Clinical Neuropsychology, Plenum Press, New York (1986).
14 C.E. Schwartz, E. Kozora and Q. Zeng, Towards patient collaboration in cognitive assessment: specificity, sensitivity and incremental validity of self-report, Ann Behav Med 18 (3) (1996), pp. 177–184. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (16)
15 K. Paraska and C.M. Bender, Cognitive dysfunction following adjuvant chemotherapy for breast cancer: two case studies, Oncol Nurs Forum 30 (3) (2003), pp. 473–478. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (15)
16 E. Kozora, M.C. Ellison and S. West, Depression, fatigue, and pain in systemic lupus erythematosus (SLE): relationship to the American College of Rheumatology SLE neuropsychological battery, Arthritis Rheum 55 (4) (2006), pp. 628–635. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (36)
17 T.A. Ahles, A.J. Saykin and C.T. Furstenberg et al., Neuropsychologic impact of standard-dose systemic chemotherapy in long-term survivors of breast cancer and lymphoma, J Clin Oncol 20 (2) (2002), pp. 485–493. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (251)
18 P. Klepstad, P. Hilton, J. Moen, B. Fougner, P.C. Borchgrevink and S. Kaasa, Self-reports are not related to objective assessments of cognitive function and sedation in patients with cancer pain admitted to a palliative care unit, Palliat Med 16 (6) (2002), pp. 513–519. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (25)
19 S.B. Schagen, F.S. van Dam, M.J. Muller, W. Boogerd, J. Lindeboom and P.F. Bruning, Cognitive deficits after postoperative adjuvant chemotherapy for breast carcinoma, Cancer 85 (3) (1999), pp. 640–650. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (263)
20 S.B. Schagen, M.J. Muller and W. Boogerd et al., Late effects of adjuvant chemotherapy on cognitive function: a follow-up study in breast cancer patients, Ann Oncol 13 (9) (2002), pp. 1387–1397. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (99)
21 F.S. van Dam, S.B. Schagen and M.J. Muller et al., Impairment of cognitive function in women receiving adjuvant treatment for high-risk breast cancer: high-dose versus standard-dose chemotherapy, J Natl Cancer Inst 90 (3) (1998), pp. 210–218. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (322)
22 D.L. Fairclough and D.F. Cella, Functional Assessment of Cancer Therapy (FACT-G): non-response to individual questions, Qual Life Res 5 (1996), pp. 321–329. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (75)
23 C.L. Burton, E. Strauss, D.F. Hultsch and M.A. Hunter, Cognitive functioning and everyday problem solving in older adults, Clin Neuropsychol 20 (3) (2006), pp. 432–452. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (20)
Correspondence to: Lisa M. Hess, PhD, Indiana University School of Medicine, Department of Public Health, 714 N Senate Avenue, Indianapolis, IN 46202; telephone: (317) 274-3148; Fax (317) 274-3443
Cost–Utility Analysis of Palonosetron-Based Therapy in Preventing Emesis Among Breast Cancer Patients
Original research
Elenir B.C. Avritscher MD, PhD, MBA/MHA, a, , Ya-Chen T. Shih PhDa, Charlotte C. Sun DrPHa, Richard J. Gralla MDa, Steven M. Grunberg MDa, Ying Xu MD, MSa and Linda S. Elting DrPHa
Abstract
We estimated the cost-utility of palonosetron-based therapy compared with generic ondansetron-based therapy throughout four cycles of anthracycline and cyclophosphamide for treating women with breast cancer. We developed a Markov model comparing six strategies in which ondansetron and palonosetron are combined with either dexamethasone alone, dexamethasone plus aprepitant following emesis, or dexamethasone plus aprepitant up front. Data on the effectiveness of antiemetics and emesis-related utility were obtained from published sources. Relative to the ondansetron-based two-drug therapy, the incremental cost–effectiveness ratios for the palonosetron-based regimens were $115,490/quality-adjusted life years (QALY) for the two-drug strategy, $199,375/QALY for the two-drug regimen plus aprepitant after emesis, and $200,526/QALY for the three-drug strategy. In sensitivity analysis, using the $100,000/QALY benchmark, the palonosetron-based two-drug strategy and the two-drug regimen plus aprepitant following emesis were shown to be cost-effective in 39% and 26% of the Monte Carlo simulations, respectively, and with changes in values for the effectiveness of antiemetics and the rate of hospitalization. The cost-utility of palonosetron-based therapy exceeds the $100,000/QALY threshold. Future research incorporating the price structure of all antiemetics following ondansetron's recent patent expiration is needed.
Article Outline
Recent advances in emesis control have been possible due to the availability of increasingly more effective antiemetic agents. During the 1990s, the development of first-generation 5-hydroxytryptamine-3 (5-HT3) antagonists (ondansetron, granisetron, tropisetron, and dolasetron) marked a significant improvement in the control of emesis induced by chemotherapy, particularly acute emesis (ie, occurring within 24 hours following chemotherapy).
More recently, two new drugs—palonosetron, a second-generation 5-HT3 antagonist, and aprepitant, a centrally acting neurokinin-1 antagonist—were added to the armamentarium of antiemetic therapy. Compared with other single-dose 5-HT3 antagonists, palonosetron has a higher 5-HT3 binding affinity and longer plasma half-life and has shown superiority in the prevention of delayed emesis (ie, occurring more than 24 hours after chemotherapy administration) following moderately emetogenic chemotherapy with methotrexate, epirubicin, or cisplatin (MEC), including AC-based regimens.[4] and [5] In a recently published clinical trial conducted by Saito et al,6 palonosetron was also shown to be superior to granisetron in preventing delayed and overall emesis when both drugs were combined with dexamethasone following chemotherapy with either AC or cisplatin. As for aprepitant, when added to the standard of a 5-HT3 antagonist and dexamethasone therapy, it has been shown to improve emesis prevention among patients receiving AC-based chemotherapy during the acute, delayed, and overall periods.7
Such benefits have led to a recent revision in the antiemetics guidelines of both the American Society of Clinical Oncology (ASCO) and the National Comprehensive Cancer Network (NCCN), incorporating both palonosetron as one of the recommended 5-HT3 antagonists and aprepitant in combination with a 5-HT3 antagonist and dexamethasone for patients receiving AC-based chemotherapy.[8] and [9] Of note is that the revised 2010 NCCN antiemetic guidelines suggest that palonosetron may be used prior to the start of multiday chemotherapy, which is more likely to cause significant delayed emesis, instead of repeated daily doses of other first-generation 5-HT3 antagonists.9
Given the multiplicity of antiemetic strategies available for prophylaxis of nausea and vomiting associated with AC-based chemotherapy with inherent variability in effectiveness and price, it is critical for existing therapies to be analyzed in terms of both their outcomes and costs. Thus, the purpose of this study is to determine, from a third-party payer perspective, the cost-utility of palonosetron-based therapy in preventing emesis among breast cancer patients receiving four cycles of AC-based chemotherapy relative to generic ondansetron-based antiemetic therapy. Due to variations in the definition of complete emetic response found across antiemetic studies, the analysis will focus on chemotherapy-induced emesis only, rather than nausea and vomiting, as vomiting can be more objectively measured than nausea and, as such, has been more consistently reported.
Patients and Methods
We developed a Markov model to estimate the costs (in 2008 U.S. dollars) and health outcomes associated with emesis among breast cancer patients receiving multiple cycles of AC-based chemotherapy under six prophylactic strategies containing either generic ondansetron (onda) or palonosetron (palo) when each is combined with either dexamethasone (dex) alone, dex plus aprepitant in the subsequent cycles following the occurrence of emesis, or dex plus aprepitant up front (Figure 1). The time horizon for the risk of chemotherapy-induced emesis during each cycle of chemotherapy was 21 days, which is the standard duration of a cycle of AC-based chemotherapy.
Markov Model Comparing Palo-Based Therapy vs Onda-Based Therapy for Prophylaxis of Chemotherapy-Induced Emesis in Breast Cancer Patients Receiving Four Cycles of AC-Based Chemotherapy (1) Onda (32 mg intravenously) + dex (8 mg intravenously) on day 1, followed by dex (4 mg orally twice a day) on days 2−5. (2) Onda (32 mg intravenously) + dex (8 mg intravenously) on day 1, followed by dex (4 mg orally twice a day) on days 2−5 and aprepitant in the subsequent cycles following the occurrence of emesis (ie, onda 16 mg orally + aprepitant 125 mg orally + dex 12 mg orally on day 1 followed by aprepitant 80 mg orally on days 2−3). (3) Palo (0.25 mg intravenously) + dex (8 mg intravenously) on day 1, followed by dex (4 mg orally twice a day) on days 2−5. (4) Palo (0.25 mg intravenously) + dex (8 mg intravenously) on day 1, followed by dex (4 mg orally twice a day) on days 2−5 and aprepitant in the subsequent cycles following the occurrence of emesis (ie, palo 0.25 mg intravenously + aprepitant 125 mg orally + dex 12 mg orally on day 1 followed by aprepitant 80 mg orally on days 2−3). (5) Onda (16 mg orally) + aprepitant (125 mg orally) + dex (12 mg orally) on day 1 followed by aprepitant (80 mg orally) on days 2−3. (6) Palo (0.25 mg intravenously) + aprepitant (125 mg orally) + dex (12 mg orally) on day 1 followed by aprepitant (80 mg orally) on days 2−3. Palo = palonosetron; onda = ondansetron; AC = anthracycline and cyclophosphamide; dex, dexamethasone
We modeled emesis-related outcomes and direct medical costs (from a third-party payer perspective within the context of the U.S. health-care system) over a total of four cycles of chemotherapy as patients receiving AC-based regimens usually undergo at least four cycles of AC.10 We performed all analyses using TreeAge Pro 2009 Suite (Decision Analysis; TreeAge Software, Williamstown, MA). The study was submitted to our institutional review board and was determined to be exempt from review.
Probability Data
Two-drug prophylactic regimens
We estimated the effectiveness of the 5-HT3 antagonists based on secondary analysis of the raw data from the randomized clinical trial (RCT) directly comparing onda and palo when used alone for prevention of emesis associated with MEC, including 90 breast cancer patients from the palo 0.25-mg arm and 82 from the onda 32-mg arm who received AC-based chemotherapy (Table 1).5 Effectiveness estimates for palo 0.25 mg were augmented by data on 117 breast cancer patients on AC-based chemotherapy participating in a multicenter RCT comparing palo with dolasetron (Table 1).4 We assumed that dex adds the same relative benefit to either first- or second-generation 5-HT3 antagonists and obtained the expected additional benefit of dex in preventing acute emesis based on the results of an RCT comparing a single-dose of granisetron in combination with dex vs granisetron given alone to patients undergoing MEC (Table 2).11 Since in the aforementioned study dex was only given on day 1 of chemotherapy, the estimated additional benefit of adding dex to a 5-HT3 inhibitor on the delayed period was obtained from another RCT; this study, conducted by the Italian Group for Antiemetic Research, compared dex alone, dex plus onda, or placebo on days 2−5 of MEC.12
MODEL PARAMETERS | BASE-CASE VALUES (RANGES) | DATA SOURCES |
---|---|---|
Probability of acute emesis control on cycle 1 of AC: | ||
Onda-based two-drug strategyc | 0.84 (0.74−0.93) | Gralla et al,a The Italian Group[5] and [11] |
Palo-based two-drug strategyc | 0.87 (0.81−0.94) | Eisenberg et al,a Gralla et al,a The Italian Group[4], [5] and [11] |
Onda-based three-drug strategyd | 0.88 (0.85−0.91) | Warr et al7 |
Palo-based three-drug strategyd | 0.96 (0.89−0.99) | Grote et al, Grunberg et al[40] and [41] |
Probability of delayed emesis control following control of acute emesis on cycle 1 of ACc: | ||
Onda-based two-drug strategyd | 0.75 (0.62–0.85) | The Italian Group12 |
Palo-based two-drug strategyc | 0.85 (0.78–0.91) | Eisenberg et al,a Gralla et al,a The Italian Group[4], [5] and [12] |
Onda-based three-drug strategyd | 0.86 (0.82–0.90) | Warr et al7 |
Palo-based three-drug strategyc | 0.96 (0.91–0.97) | Eisenberg et al,a Gralla et al,a Warr et al[4], [5] and [7] |
Probability of delayed emesis control following acute emesis on cycle 1 of ACc: | ||
Onda-based two-drug strategyc | 0.46 (0.31–0.62) | Gralla et al,a The Italian Group[5] and [12] |
Palo-based two-drug strategyc | 0.44 (0.27–0.59) | Eisenberg et al,a Gralla et al,a The Italian Group[4], [5] and [12] |
Onda-based three-drug strategyd | 0.44 (0.29–0.57) | Warr et al7 |
Palo-based three-drug strategyc | 0.51 (0.41–0.67) | Eisenberg et al,a Gralla et al,a Warr et al[4], [5] and [7] |
Relative probability of emesis control in subsequent cycles of ACc: | ||
Two-drug therapy | 0.987 (0.970–1.0) | Herrstedt et al14e |
Three-drug therapy | 1.013 (1.0–1.030) | Herrstedt et al14e |
Probability of hospitalization (among patients who develop emesis) per cycle of ACd: | ||
Onda-based regimens | 0.0035 (0.0001−0.019) | Data from Medstat MarketScan16 |
Palo-based regimens | 0.0017 (0.00004−0.0089) | Data from Medstat MarketScan, Haislip et al[16] and [19]b |
Probability of office visit use (among patients who develop emesis) per cycle of ACd: | ||
Onda-based regimens | 0.10 (0.07−0.14) | Data from Medstat MarketScan16 |
Palo-based regimens | 0.05 (0.03−0.07) | Data from Medstat MarketScan, Haislip et al[16] and [19]b |
Probability of rescue medicine utilization use (among patients who develop emesis) per cycle of ACd: | ||
Onda-based regimens | 0.61 (0.46−0.75) | Gralla et al5a |
Palo-based regimens | 0.56 (0.45−0.66) | Eisenberg et al, Gralla et al[4] and [5]a |
Utility weights for emesis per cycle of ACf: | ||
Acute and delayed emesis | 0.15 (0.10−0.20) | Sun et al20 |
Acute emesis and no delayed emesis | 0.76 (0.70−0.83) | Sun et al20 |
No acute emesis and delayed emesis | 0.20 (0.14−0.26) | Sun et al20 |
No acute and no delayed emesis | 0.92 (0.86−0.99) | Sun et al20 |
AC = anthracycline and cyclophosphamide; onda = ondansetron; palo = palonosetron.
a Included in the analysis was the subset of women with breast cancer receiving AC-based chemotherapy.b We obtained an estimate of emesis-related hospitalization and office visit utilization based on data from Medstat MarketScan, HPM subset (Medstat Group, Inc., Ann Arbor, MI) on 707 breast cancer patients who received the first cycle of AC-based chemotherapy from 1996 to 2002 and either were admitted to the hospital or had an office visit for treatment of vomiting or dehydration. Since palo was only introduced into the U.S. market in 2003, we assumed that all these breast cancer patients received onda-based antiemetic prophylaxis. As a result, we estimated the differential rate of health-care resource utilization based on Haislip et al's19 reported differential incidence of extreme events associated with chemotherapy-induced nausea and vomiting experienced by community-based breast cancer patients who received either onda or palo for emesis prophylaxis following the first cycle of chemotherapy.c Of note is that there are two different methods for applying the benefit of adding dex and/or aprepitant to a 5-HT3 antagonist: (1) rate of emesis with 5-HT3* relative risk of emesis by adding dex and/or aprepitant and (2) rate of emesis control with 5-HT3 * relative risk of emesis control by adding dex and/or aprepitant. These produce substantially different results, with the former method skewing the results toward the least effective 5-HT3 and the latter skewing it toward the most effective one. As a result, we estimated the probability of emesis by averaging the results obtained using the two different methods. Of note is that the ranges for these effectiveness estimates were obtained by applying the two different methods to the lower and upper bounds of the 95% confidence intervals derived from the clinical trials comparing the 5-HT3 antagonists when used alone.d Ranges were obtained by constructing 95% confidence intervals for observed proportions using the normal approximation to the binomial distribution.e Ranges are based on the minimum and maximum values observed in Herrstedt et al's14 clinical trial of multicycle chemotherapy.f Ranges are based on the estimate's actual 95% confidence intervals obtained from Sun et al's20 data.
Three-drug prophylactic regimens
We estimated the rate of acute emesis for the three-drug regimens based on data from published studies in which either onda or palo was given in combination with dex and aprepitant on day 1 of MEC (Table 2).[5], [7] and [13] Because aprepitant was either used in combination with dexamethasone or not used on days 2−3 in the trials of palo-based three-drug therapy, we estimated the benefit of adding aprepitant alone to palo on days 2−3 by assuming that the added benefit in the delayed period would be the same as the benefit added to onda. Specifically, we obtained information on the relative risk of delayed emesis control when aprepitant is added on days 2−3 from a large clinical trial of aprepitant combined with onda and dex in breast cancer patients receiving either A or AC chemotherapy (Table 2).7
Effectiveness of antiemetics over multiple cycles of chemotherapy
The estimates of changes in the probability of emesis control over multiple cycles of chemotherapy were obtained from a RCT conducted by Herrstedt et al14 of ondansetron-based two- and three-drug regimens for prevention of chemotherapy-induced nausea and vomiting among breast cancer patients undergoing multiple cycles of AC-based chemotherapy. We assumed that changes in emesis control over four cycles of AC for the palo-based two- and three-drug regimens were similar to the observed changes for the onda-based two- and three-drug strategies, respectively.14
Resource Utilization and Cost Data
The cost of antiemetic prophylaxis was based on the 2008 Medicare Part B reimbursement rates for pharmaceuticals, which reflects the price of ondansetron following its recent patent expiration (Table 3).15 The costs of prophylaxis failures were estimated as follows. In the majority of prophylaxis failures, the only cost is the cost of rescue medication. In such cases, we obtained costs by multiplying the individual doses used for rescue treatment of breast cancer patients on AC participating in the clinical trials comparing palo 0.25 mg with single doses of onda or dolasetron by their unit costs based on the 2008 Medicare Part B reimbursement rates.[5] and [15] For the few patients who are seen in the office for uncontrolled emesis, we obtained estimates of the risk of such emesis-related office visits based on the MarketScan Health Productivity Management (HPM) database from Thomson Reuters on 707 breast cancer patients who received their first cycle of AC-based chemotherapy between 1997 and 2002 (Table 2) and its costs from the 2008 Medicare Physician Fee Schedule Reimbursement for a level III office visit (CPT 99213).[16] and [17]
COST COMPONENT | 2008 U.S.$ (RANGES) | DATA SOURCE |
---|---|---|
Hospitalization | $5,237.00 ($3,921−$6,112)a | HCUP charge data18 Consumer Price Index42 Medicare cost-to-charge ratio43 |
Level III office visit (CPT 99213) | $60.30 ($19.96–$122.46)d | 2008 Medicare Physician Fee Schedule Reimbursement17 |
Prophylactic antiemetics | 2008 Medicare Part B reimbursement rates for pharmaceuticals15 | |
Onda-based two-drug regimen | $49.74 | |
Palo-based two-drug regimen | $207.20 | |
Onda-based three-drug regimen | $324.51 | |
Palo-based three-drug regimen | $482.46 | |
Rescue medicinesb | $35.25 ($21.66–$48.80)c | Eisenberg et al,4 Gralla et al,5 2008 Medicare Part B reimbursement rates for pharmaceuticals15 |
AC = anthracycline and cyclophosphamide; onda = ondansetron; palo = palonosetron; HCUP = Healthcare Cost and Utilization Project
a Charges were inflated to 2008 U.S. dollars using the Consumer Price Index (CPI) for medical care and adjusted to costs using Medicare cost-to-charge ratio. The ranges were based on estimates of the 95% confidence interval.b In the randomized clinical trial directly comparing ondansetron and palonosetron, propulsives accounted for 71% of the rescue medicines used, 5-hydroxytryptamine antagonists for 20%, glucocorticoids for 7%, and aminoalkyl ethers for 2%.5c Costs for rescue medication were obtained by multiplying all drug unit costs by the individual doses used for rescue treatment of breast cancer patients on AC participating in the clinical trials comparing palo 0.25 mg with single doses of onda or dolasetron.[5] and [15] The ranges were based on estimates of the 95% confidence interval.d Ranges were based on the Medicare physician fee schedule for levels I and VI office visits.
Finally, although hospitalization for emesis is extremely rare in this population, when it occurs, it is quite expensive. For completeness, we obtained estimates of the risk of emesis-related hospitalization from the same population of breast cancer patients from whom we obtained the estimate for the risk of emesis-related office visit, whereas hospital costs were obtained from Healthcare Cost and Utilization Project (HCUP) data on 2,342 breast cancer patients who were hospitalized with a primary or admitting diagnosis of vomiting or dehydration from 1997 to 2003 ([Table 2] and [Table 3]).[16] and [18]
Of note is that since palo was only introduced into the U.S. market in 2003, we anticipated the observed risk of emesis-related office visit and hospital admission obtained from MarketScan data during the period 1997−2002 reflected the risk associated with prophylaxis with onda. As a result, given that, when compared with onda, palo has also shown superiority in reducing the severity of emetic episodes when they occur, we estimated the differential rate of health-care resource utilization for palo and onda based on Haislip et al's reported differential incidence of extreme events associated with chemotherapy-induced nausea and vomiting (CINV) experienced by community-based breast cancer patients who received either palo or onda for emesis prophylaxis following the first cycle of chemotherapy (Table 2).[5] and [19]
Utility Data
We obtained the utility weights for acute and delayed emesis from a published study of preferences elicited from ovarian cancer patients undergoing chemotherapy using a modified visual analog scale (VAS) (Table 2).20 We equally applied these emesis-related utility weights to the initial 5-day period of chemotherapy (the standard duration of follow-up in clinical trials of prophylactic antiemetics) in all six prophylactic strategies of the decision tree. Furthermore, because the risk of CINV after 5 days of chemotherapy is usually so negligible as to be unmeasured in clinical trials of antiemetics, we assumed the utility weights for the remaining 16 days of each of the chemotherapy cycles to be the same as the weight associated with complete emesis control (ie, 0.92). We subsequently converted the resulting estimates of quality-adjusted life days into quality-adjusted life years (QALY).
Analysis
We used a stepwise method to calculate the incremental cost–effectiveness ratios of the different prophylactic therapy strategies, with the generic onda-based two-drug therapy (ie, the lowest cost strategy) as the base comparator (also known as the “anchor”).21 We adopted the benchmark range of U.S. $50,000−$100,000 per QALY, which has been commonly cited for oncology-related interventions as the threshold for acceptable cost–effectiveness, and examined the robustness of the results by performing one-way sensitivity analyses of plausible ranges for the model's key parameters based on the data sources used as well as probabilistic sensitivity analysis using Monte Carlo simulation.[21] and [22]
Results
The overall rate of emesis control (on days 1−5) among breast cancer patients following a cycle of AC-based chemotherapy was estimated to be 63% (range 46%−79%) for the onda-based two-drug therapy, 74% (range 66%−85%) for the palo-based two-drug therapy, 76% (range 75%−82%) for the onda-based three-drug therapy, and 92% (range 81%−96%) for the palo-based three-drug therapy. Based on these estimates, relative to the onda-based two-drug therapy, the incremental cost–effectiveness ratios (ICERs) for the palo-based regimens were $115,490/QALY for the two-drug strategy, $199,375/QALY for the two-drug regimen plus aprepitant after emesis, and $200,526/QALY for the three-drug strategy (Table 4). The onda-based two-drug combination plus aprepitant after the onset of emesis was eliminated through extended dominance as it has a greater ICER than the next more effective therapy, the palo-based two-drug treatment strategy (Table 4). The onda-based three-drug strategy was dominated by the palo-based two-drug combination plus aprepitant after the onset of emesis as the former strategy is both less effective and more expensive than the latter (Table 4).
STRATEGY | TOTAL COST (U.S.$) | INCREMENTAL COST (U.S.$) | EFFECTIVENESS (QALY) | INCREMENTAL EFFECTIVENESS (QALY) | INCREMENTAL COST–EFFECTIVENESS (U.S.$/QALY) |
---|---|---|---|---|---|
Onda-based two-drug therapy | $269 | — | 0.1989 | — | — |
Onda-based two-drug therapy with aprepitant after emesis | $635 | $366 | 0.2010 | 0.0021 | $174, 286 Eliminated through extended dominancea |
Palo-based two-drug therapy | $858 | $589 | 0.2040 | 0.0051 | $115,490c |
Palo-based two-drug therapy plus aprepitant after emesis | $1,177 | $319 | 0.2056 | 0.0016 | 199,375 |
Onda-based three-drug therapy | $1,336 | $159 | 0.205 | (0.0006) | Dominatedb |
Palo-based three-drug therapy | $1,939 | $603 | 0.2094 | 0.0044 | $200,526d |
QALY = quality-adjusted life year; AC = anthracycline and cyclophosphamide; ICER = incremental cost–effectiveness ratio; onda = ondansetron; palo = palonosetron
a Extended dominance occurs when one of the treatment alternatives has a greater ICER than the next more effective alternative.b One intervention is said to be dominated by another when it is both less effective and more expensive than the previous less costly alternative.c Because the onda-based two-drug combination plus aprepitant after the onset of emesis was eliminated through extended dominance, the palo-based two-drug therapy was compared with the onda-based two-drug therapy.d Because the onda-based three-drug combination was dominated by the palo-based two-drug combination plus aprepitant after the onset of emesis, the palo-based three-drug therapy was compared with the latter regimen.
In sensitivity analyses using the commonly accepted cost–effectiveness benchmark range of $50,000−$100,000/QALY, the results were sensitive to changes in the overall emesis control rates for the onda-based two-drug strategy. If the probability of overall emesis control for the onda-based two-drug strategy was as low as its estimated lower bound (46%), the ICER for the palo-based two-drug treatment alternative would drop to $53,892/QALY. The results were also sensitive to changes in the effectiveness for the palo-based two-drug regimen: When its overall control rate was as high as its estimated upper bound (86%), its ICER would be $71,472. In contrast, the results were not sensitive to variations in the probability of overall emesis control for the three-drug strategies, nor were they sensitive to changes in the relative probability of emesis control in subsequent cycles of AC for either the two- or three-drug strategies.
If the probability of emesis-related hospitalization was as high as the upper limit of its 95% confidence interval (CI), the ICER for the palo-based two-drug regimen would be $97,301/QALY. However, changes in the cost of an emesis-related admission (95% CI $3,921−$6,112) did not significantly alter the results, nor did variations in office visit and rescue medicine utilization and their associated costs. The results were also not sensitive to variations in the values for the utility weights throughout their 95% CIs. We performed a threshold analysis to explore the price per dose of palo that would result in an acceptable cost–effectiveness ratio under the $100,000/QALY benchmark and found that the ICER for the palo-based two-drug treatment alternative would only fall to a $100,000/QALY threshold when the cost of palo is decreased by 11%.
Figure 2 shows the cost–effectiveness acceptability curves for each strategy, with the onda-based two-drug therapy as the base comparator. These curves show the proportion of the 100,000 simulations in which the comparing antiemetic regimen was considered more cost-effective than the base comparator at different thresholds. Using the benchmark of U.S. $100,000/QALY, the palo-based two-drug strategy and the two-drug regimen plus aprepitant following the onset of emesis were shown to be cost-effective in 39% and 26% of the simulations with the onda-based standard therapy as the baseline, respectively, whereas the palo-based and onda-based three-drug strategies and the onda-based two-drug regimen with aprepitant after emesis were cost-effective in fewer than 10% of the simulations. Of note is that the slope of the acceptability curves for the palo-based two-drug strategies are steep when willingness to pay exceeds $50,000/QALY, indicating that the greater the threshold, the greater the increase in the level of confidence that these strategies could be cost-effective. For example, the probability that the palo-based two-drug strategy is more cost-effective than the onda-based two-drug strategy rises to 51% at a threshold value of $125,000/QALY and exceeds 60% at $150,000/QALY.
Figure 3 presents the scatterplot of the results of the probabilistic sensitivity analysis for the palo-based two-drug strategy. Nearly 96% of the simulations fell within the first quadrant of the chart (ie, on the upper right quadrant), which represents the scenario where the palo-based two-drug therapy is more costly but also more effective than the onda-based standard therapy. However, only 39% of the simulations fell below the $100,000/QALY dashed threshold line, which represents the scenario where the palo-based two-drug strategy is more cost-effective than the onda-based standard therapy at the $100,000/QALY benchmark.
Discussion
Our estimates of emesis-related costs and outcomes following four cycles of AC-based chemotherapy in women with breast cancer indicate that at current antiemetic prices and utilities placed on emesis, the additional costs of palo and aprepitant are not warranted at the $100,000/QALY threshold. In probabilistic sensitivity analysis, the palo-based two-drug strategy and the two-drug regimen plus aprepitant following the onset of emesis were shown to be cost-effective at the $100,000/QALY threshold in only 39% and 26% of the simulations, respectively. The model was sensitive to changes in the values of antiemetic effectiveness for the two-drug regimens and the risk of emesis-related hospitalization.
In threshold analysis, the two-drug palo-based regimen was cost-effective at the $100,000/QALY benchmark when the cost of palo is decreased by 11%. Because the use of the $100,000/QALY threshold is uncommon in clinical practice, the cost-effectiveness of the palo-based two-drug strategy (estimated at $115,490/QALY in our study) compares favorably with other commonly used supportive care measures for women with breast cancer. Such measures include primary prophylaxis with granulocyte colony-stimulating factor in women undergoing chemotherapy with moderate to high myelosuppressive risk (ICER of $116,000/QALY, or $125,948/QALY in 2008 U.S. dollars) and the use of bisphosphonates for the prevention of skeletal complications in breast cancer patients with lytic bone metastases (ICER ranging from $108,200/QALY with chemotherapy as systemic therapy to $305,300 in conjunction with hormonal systemic therapy, or $166,381/QALY to $469,466/QALY in 2008 U.S. dollars, respectively).[23] and [24] Both interventions are considered recommended standards of supportive care for patients with breast cancer and are widely used in breast oncology practices.[25] and [26]
Decision-analytic models, such as the Markov model presented in our study, aim to reflect the reality of clinical practice in a simplified way. Therefore, modelers often need to make decisions regarding the study time frame and model parameters based on the best use of available data. In our study, we obtained estimates for the probability of chemotherapy-induced emesis from studies in which the standard duration of follow-up is 5 days. By so doing, we may have underestimated the cost-effectiveness for the palo-based and aprepitant-based regimens. Although the risk of CINV after 5 days of chemotherapy is usually negligible, anticipation of vomiting may affect a patient's quality of life throughout the cycle of chemotherapy.
In addition, our estimates of costs, which were mostly obtained from Medicare, may differ from those of other third-party payers. However, Medicare is among the largest payers for breast cancer care as 42% of the women diagnosed with cancer in the United States are older than 64 years, and many private organizations set their own reimbursement rates based on the Medicare schedule. Therefore, we believe that Medicare reimbursement data provide a suitable estimate for emesis-related medical costs for all breast cancer patients in the United States.[27] and [28]
The present results should solely be interpreted in light of the cost–effectiveness benchmark of $50,000−$100,000/QALY, which has been frequently used in the context of the U.S. health-care system.[22] and [29] Such a benchmark, however, is a historic, precedent-based threshold set by the cost of caring for patients on dialysis, which was estimated at $50,000/QALY in 1982 ($74,000−$95,000 in 1997 U.S. dollars).[30] and [31] Given the arbitrariness of such a threshold, it has been suggested that the current willingness to pay for medical interventions in the United States probably exceeds $100,000/QALY, with values as high as $300,000/QALY being cited in some oncology publications.[22], [29], [31], [32], [33] and [34] In support of that argument is the public and policy makers' strong negative reaction to the National Institutes of Health Consensus Panel not recommending mammography screening for women aged 40−49 years, a procedure reported to provide an ICER of $105,000 per life-year gained.[35] and [36] As a result, if willingness to pay goes beyond $100,000/QALY, the alternative of adding aprepitant to palo plus dex may also be deemed attractive as the slope of its acceptability curve becomes substantially steep when the willingness to pay for a QALY exceeds $125,000 (Figure 2), suggesting that its marginal gain may exceed its marginal costs at higher thresholds.
In addition, it is worth noting that the present analysis has been conducted from the perspective of a third-party payer within the context of the U.S. health-care system. The large difference in the acquisition cost of palo-based and onda-based therapy observed in the United States is mostly driven by the differential stage of product life cycles for palo and onda. Although at the time of this study palo was still under patent protection, generic onda had entered the U.S. market prior to our study. The large price discrepancy between brand and generic drugs explains the difference in drug costs in this U.S.-based analysis. As such, our results may not reflect the situation in countries with a widely different cost structure, in which the acquisition cost of palo may be substantially lower. When that is the case, the cost–effectiveness profile of the palo-based prophylactic therapy may be deemed substantially more favorable than the profile presented here. Similarly, we anticipate finding a more attractive cost–effectiveness profile for the palo-based therapies as palo reaches the end of its product life cycle in the U.S. market.37 Also of note is that the cost–effectiveness of the palo-based therapy may greatly differ when different perspectives (other than the third-party payer's perspective) are adopted.
Our study, however, has several limitations. First, the utility scores used in our model were derived with a VAS instrument, which does not incorporate patients' preferences under uncertainty. Nevertheless, the VAS approach has been shown to provide utility scores for nausea and vomiting with more variability than scores derived using other methods such as the Standard Gamble (personal communication, Grunberg SM et al, CALGB study 309801). Notwithstanding that, it remains unclear which method gives utility scores for transient health states, such as CINV, with the greatest validity.
Also of note is that due to a lack of information on emesis-related utilities among breast cancer patients in the literature, we used utilities elicited from patients with ovarian cancer. To the best of our knowledge, the utilities in Sun et al20 were the only ones available in the literature that were elicited from a homogeneous population of cancer patients (ie, solely patients with ovarian cancer) and were based on a wide range of health states combining the presence and absence of emesis during either the acute or the delayed period. In addition, the participants in the Sun et al study were treated with carboplatin, which, like the regimen used in our model, is classified as moderately emetogenic in established antiemetic guidelines.[8], [9] and [38] It is also important to emphasize that the population in that study, like our study's population, was composed exclusively of women, who are known to be at increased risk for developing CINV.39
Second, in the absence of clinical trial data, we assumed conservatively that dex and aprepitant add the same relative benefit to both onda and palo. This assumption results in an imperfect estimate of cost–effectiveness. As such, we may have overestimated or underestimated the cost–effectiveness of palo as dex and aprepitant may potentially add less value to the intrinsically more active 5-HT3 antagonist or uniquely complementary mechanisms of action could contribute to even greater activity with the palo-based therapy. However, our study's estimate of the relative effectiveness of the palo-based two-drug prophylactic therapy versus the onda-based two-drug therapy for preventing delayed emesis is consistent with that reported in a recently published clinical trial comparing palo and granisetron when both drugs are combined with dex following chemotherapy with either AC or cisplatin (1.18 vs 1.17, respectively).6
Third, our study did not include the outcomes associated with the adverse effects of antiemetics, and by so doing, we may have underestimated the costs associated with antiemetic prophylaxis. However, the incidence and duration of treatment-related adverse events occurring in the two RCTs comparing palo with either onda or dolasetron were mild and similar across treatment cohorts.[4] and [5]
Fourth, we assumed that changes in emesis control in subsequent cycles of AC for the palo-based regimens were the same as for the onda-based therapy. By so doing, we may have underestimated the cost–effectiveness of palo as the superiority of the more active 5-HT3 antagonist could be maintained in the subsequent cycles of chemotherapy (or even increased, as seen in the aprepitant-based arm of Herrstedt et al's14 study). As a result, if future prospective trials of palo-based antiemetic prophylaxis confirm its superiority in maintaining antiemetic efficacy over multiple cycles of AC, the cost–effectiveness profiles for the palo-based strategies may be more favorable than the profiles presented herein.
Last, the incremental gains in QALY observed in cost–utility analysis of interventions associated with transitory and non-life-threatening health states, such as the antiemetic regimens analyzed in our study, tend to render small denominators to be used in the incremental cost–effectiveness ratios. The issue of small denominators has led some researchers to question whether the current methodology of cost–effectiveness analysis is appropriate to determine the cost–effectiveness of treatments for terminal or supportive care.32 However, despite this shortcoming, these types of analysis benefit from having a wider scope as they allow comparisons over different types of health interventions across various diseases. In addition, by incorporating patients' utility levels over different health states (instead of merely looking into cost per additional patient controlled), cost–utility analysis makes explicit the impact of the target population's preferences for the different outcomes. Of importance is that both the Panel on Cost–Effectiveness in Health and Medicine and the Institute of Medicine (IOM) Committee on Regulatory Cost–Effectiveness Analysis recommend the use of QALY as the preferred outcome measure for economic evaluation of health-care interventions.
Conclusion
Although our base-case analysis suggests that, from a third-party payer perspective within the context of the U.S. health-care system, the cost–utility of the palo-based two-drug prophylactic therapy for breast cancer patients receiving four cycles of AC-based chemotherapy exceeds the $50,000–$100,000/QALY threshold, it is comparable to other commonly used supportive care interventions for women with breast cancer. In sensitivity analyses, such a strategy was associated with a 39% chance of being cost-effective at the $100,000/QALY threshold, and the model was sensitive to changes in the values of antiemetic effectiveness and of the probability of emesis-related hospitalization. In threshold analysis, the combination of palo and dex was shown to become cost-effective (at the $100,000/QALY benchmark) when the cost of palo is decreased by 11%. As a result, future research incorporating the price structure of all antiemetics following the recent expiration of onda's patent is needed.
References1
1 S.M. Grunberg, D. Osoba and P.J. Hesketh et al., Evaluation of new antiemetic agents and definition of antineoplastic agent emetogenicity—an update, Support Care Cancer 13 (2005), pp. 80–84 [15599601]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (68)
2 C.M. Booth, M. Clemons and G. Dranitsaris et al., Chemotherapy-induced nausea and vomiting in breast cancer patients: a prospective observational study, J Support Oncol 5 (2007), pp. 374–380 [17944146]. View Record in Scopus | Cited By in Scopus (10)
3 M. de Boer-Dennert, R. de Wit and P.I. Schmitz et al., Patient perceptions of the side-effects of chemotherapy: the influence of 5HT3 antagonists, Br J Cancer 76 (1997), pp. 1055–1061 [9376266]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (128)
4 P. Eisenberg, J. Figueroa-Vadillo, R. Zamora et al. and 99-04 Palonosetron Study Group, Improved prevention of moderately emetogenic chemotherapy-induced nausea and vomiting with palonosetron, a pharmacologically novel 5-HT3 receptor antagonist: results of a phase III, single-dose trial versus dolasetron, Cancer 98 (2003), pp. 2473–2482 [14635083]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (171)
5 R. Gralla, M. Lichinitser and S. Van Der Vegt et al., Palonosetron improves prevention of chemotherapy-induced nausea and vomiting following moderately emetogenic chemotherapy: results of a double-blind randomized phase III trial comparing single doses of palonosetron with ondansetron, Ann Oncol 14 (2003), pp. 1570–1577 [14504060]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (177)
6 M. Saito, K. Aogi and I. Sekine et al., Palonosetron plus dexamethasone versus granisetron plus dexamethasone for prevention of nausea and vomiting during chemotherapy: a double-blind, double-dummy, randomised, comparative phase III trial, Lancet Oncol 10 (2009), pp. 115–124 [19135415]. Article | | View Record in Scopus | Cited By in Scopus (43)
7 D.G. Warr, P.J. Hesketh and R.J. Gralla et al., Efficacy and tolerability of aprepitant for the prevention of chemotherapy-induced nausea and vomiting in patients with breast cancer after moderately emetogenic chemotherapy, J Clin Oncol 23 (2005), pp. 2822–2830 [15837996]. View Record in Scopus | Cited By in Scopus (139)
8 American Society of Clinical Oncology, M.G. Kris, P.J. Hesketh and M.R. Somerfield et al., American Society of Clinical Oncology guideline for antiemetics in oncology: update 2006, J Clin Oncol 24 (2006), pp. 2932–2947 [16717289]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (311)
9 D.S. Ettinger, D.K. Armstrong and S. Barbour et al., National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology—Antiemesis, version 2.2010 http://www.nccn.org/professionals/physician_gls/PDF/antiemesis.pdf Accessed September 20, 2010.
10 R.W. Carlson and B. McCormick, Update: NCCN breast cancer clinical practice guidelines, J Natl Compr Cancer Netw 3 (suppl 1) (2005), pp. S7–S11 [16280118].
11 The Italian Group for Antiemetic Research, Dexamethasone, granisetron, or both for the prevention of nausea and vomiting during chemotherapy for cancer, N Engl J Med 332 (1995), pp. 1–5 [7990859].
12 The Italian Group for Antiemetic Research, Dexamethasone alone or in combination with ondansetron for the prevention of delayed nausea and vomiting induced by chemotherapy, N Engl J Med 342 (2000), pp. 1554–1559 [10824073].
13 S.M. Grunberg, M. Dugan and H. Muss et al., Effectiveness of a single-day three-drug regimen of dexamethasone, palonosetron, and aprepitant for the prevention of acute and delayed nausea and vomiting caused by moderately emetogenic chemotherapy, Support Care Cancer 17 (2009), pp. 589–594 [19037667]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (14)
14 J. Herrstedt, H.B. Muss and D.G. Warr et al., Efficacy and tolerability of aprepitant for the prevention of chemotherapy-induced nausea and emesis over multiple cycles of moderately emetogenic chemotherapy, Cancer 104 (2005), pp. 1548–1555 [16104039]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (43)
15 Centers for Medicare and Medicaid Services, Medicare Part B Drug Average Sales Price: 2008 ASP Drug Pricing Files http://www.cms.hhs.gov/apps/ama/license.asp?file=/McrPartBDrugAvgSalesPrice/downloads/July2008ASPPricingFilebyHCPCS.zip Accessed July 18, 2008.
16 Thomson. Medstat, 1997–2002 MarketScan Health and Productivity Management Database User Guide and Data Dictionary, Thomson Medstat, Ann Arbor, MI (2003).
17 Centers for Medicare and Medicaid Services, National Physician Fee Schedule and Relative Value: 2008 Physician Fee Schedule National Payment Amount File http://www.cms.hhs.gov/PFSlookup/02_PFSSearch.asp Accessed July 18, 2008.
18 National Inpatient Sample (NIS), NIS description of data elements, Healthcare Cost and Utilization Project (HCUP) databases, Agency for Healthcare Research and Quality, Rockville, MD (2004) http://www.hcup-us.ahrq.gov/nisoverview.jsp#Data Accessed May 16, 2010.
19 S. Haislip, J. Gilmore, W.H. Lenz, T. Gondesen and B. Feinberg, Theory in practice: improving patient outcomes and practice efficiency with a simple change in 5-HT3 receptor antagonist for preventing chemotherapy-induced nausea and vomiting (CINV) In: Third Annual Meeting of the Hematology/Oncology Pharmacy Association; Abstract #PR6. June 14–16, 2007; Denver, Colorado http://www.hoparx.org/documents/2007programbook.pdf Accessed November 2, 2010.
20 C.C. Sun, D.C. Bodurka and C.B. Weaver et al., Rankings and symptom assessments of side effects from chemotherapy: insights from experienced patients with ovarian cancer, Support Care Cancer 13 (2005), pp. 219–227 [15538640]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (53)
21 M.F. Drummond, M.J. Sculpher, G.W. Torrance, B.J. O'Brien and G.L. Stoddart, Methods for the Economic Evaluation of Health Care Programmes (3rd ed.), Oxford University Press, New York (2005).
22 J. Hayman, J. Weeks and P. Mauch, Economic analyses in health care: an introduction to the methodology with an emphasis on radiation therapy, Int J Radiat Oncol Biol Phys 35 (1996), pp. 827–841 [8690653]. Article | | View Record in Scopus | Cited By in Scopus (33)
23 B.E. Hillner, J.C. Weeks, C.E. Desch and T.J. Smith, Pamidronate in prevention of bone complications in metastatic breast cancer: a cost–effectiveness analysis, J Clin Oncol 18 (2000), pp. 72–79 [10623695]. View Record in Scopus | Cited By in Scopus (90)
24 S.D. Ramsey, Z. Liu and R. Boer et al., Cost–effectiveness of primary versus secondary prophylaxis with pegfilgrastim in women with early-stage breast cancer receiving chemotherapy, Value Health 11 (2008), pp. 172–179 [18673353].
25 B.E. Hillner, J.N. Ingle, R.T. Chlebowski et al. and American Society of Clinical Oncology, American Society of Clinical Oncology 2003 update on the role of bisphosphonates and bone health issues in women with breast cancer, J Clin Oncol 21 (2003), pp. 4042–4057 [12963702]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (533)
26 T.J. Smith, J. Khatcheressian and G.H. Lyman et al., 2006 update of recommendations for the use of white blood cell growth factors: an evidence-based clinical practice guideline, J Clin Oncol 24 (2006), pp. 3187–3205 [16682719]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (485)
27 National Cancer Institute, Surveillance Epidemiology and End Results: SEER Stat Fact Sheets: Breast http://seer.cancer.gov/statfacts/html/breast.html Accessed May 16, 2010.
28 J.W. Tumeh, S.G. Moore, R. Shapiro and C.R. Flowers, Practical approach for using Medicare data to estimate costs for cost–effectiveness analysis, Expert Rev Pharmacoecon Outcomes Res 5 (2005), pp. 153–162 [19807571]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
29 P.A. Ubel, R.A. Hirth, M.E. Chernew and A.M. Fendrick, What is the price of life and why doesn't it increase at the rate of inflation?, Arch Intern Med 163 (2003), pp. 1637–1641 [12885677]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (225)
30 J.C. Hornberger, D.A. Redelmeier and J. Petersen, Variability among methods to assess patients' well-being and consequent effect on a cost–effectiveness analysis, J Clin Epidemiol 45 (1992), pp. 505–512 [1588356]. Article | | View Record in Scopus | Cited By in Scopus (138)
31 R.A. Hirth, M.E. Chernew, E. Miller, A.M. Fendrick and W.G. Weissert, Willingness to pay for a quality-adjusted life year: in search of a standard, Med Decis Making 20 (2000), pp. 332–342 [10929856]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (291)
32 Y.C. Shih and M.T. Halpern, Economic evaluations of medical care interventions for cancer patients: how, why, and what does it mean?, CA Cancer J Clin 58 (2008), pp. 231–244 [18596196]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (18)
33 E. Nadler, B. Eckert and P.J. Neumann, Do oncologists believe new cancer drugs offer good value?, Oncologist 11 (2006), pp. 90–95 [16476830]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (50)
34 R.S. Braithwaite, D.O. Meltzer, J.T. King Jr, D. Leslie and M.S. Roberts, What does the value of modern medicine say about the $50,000 per quality-adjusted life-year decision rule?, Med Care 46 (2008), pp. 349–356 [18362813]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
35 National Institutes of Health Consensus Development Panel, 1997 Consensus Statement: Breast Cancer Screening for Women Ages 40–49 http://consensus.nih.gov/1997/1997BreastCancerScreening103html.htm Accessed October 13, 2007.
36 P. Salzmann, K. Kerlikowske and K. Phillips, Cost–effectiveness of extending screening mammography guidelines to include women 40 to 49 years of age, Ann Intern Med 127 (1997), pp. 955–965 [9412300]. View Record in Scopus | Cited By in Scopus (169)
37 Y.C. Shih, S. Han and S.B. Cantor, Impact of generic drug entry on cost–effectiveness analysis, Med Decis Making 25 (2005), pp. 71–80 [15673583]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
38 F. Roila, P.J. Hesketh, J. Herrstedt and Antiemetic Subcommittee of the Multinational Association of Supportive Care in Cancer, Prevention of chemotherapy- and radiotherapy-induced emesis: results of the 2004 Perugia International Antiemetic Consensus Conference, Ann Oncol 17 (2006), pp. 20–28 [16314401]. View Record in Scopus | Cited By in Scopus (90)
39 S.M. Grunberg and A. Ireland, Epidemiology of chemotherapy-induced nausea and vomiting, Adv Studies Nurs 3 (1) (2005), pp. 9–15 http://www.jhasin.com/files/articlefiles/pdf/XASIN_3_1_p9_15.pdf Accessed September 16, 2010.
40 T. Grote, J. Hajdenberg, A. Cartmell, S. Ferguson, A. Ginkel and V. Charu, Combination therapy for chemotherapy-induced nausea and vomiting in patients receiving moderately emetogenic chemotherapy: palonosetron, dexamethasone, and aprepitant, J Support Oncol 4 (2006), pp. 403–408 [17004515]. View Record in Scopus | Cited By in Scopus (38)
41 S.M. Grunberg, M. Dugan, H.B. Muss, M. Wood, S. Burdette-Radoux and T. Weisberg, Efficacy of a 1-day 3-drug antiemetic regimen for prevention of acute and delayed nausea and vomiting induced by moderately emetogenic chemotherapy, J Clin Oncol 25 (18S) (2007), p. 9111.
42 U. S. Department of Labor. Bureau of Labor Statistics. Consumer Price Index http://www.bls.gov/cpi/home.htm Accessed May 16, 2010.
43 Department of Health and Human Services. Centers for Medicare & Medicaid Services, Medicare Program; Proposed Changes to the Hospital Inpatient Prospective Payment Systems and Fiscal Year 2008 Rates CMS-1533-P, pp 1070–1073 http://www.cms.hhs.gov/AcuteInpatientPPS/downloads/CMS-1533-P.pdf Accessed May 16, 2010.
Conflicts of interest: Dr. Sun discloses that her husband was an employee of MGI Pharma, Inc., at the time this article was being written. Dr. Gralla discloses that he is a consultant for MGI Pharma, Inc., GlaxoSmithKline, Sanofi-aventis, and Merck; he also receives honoraria from MGI Pharma, Inc., and Merck and research support from Sanofi-aventis. Dr. Grunberg discloses that he is a consultant for MGI Pharma, Inc.
Correspondence to: Elenir B. C. Avritscher, MD, PhD, MBA/MHA, Section of Health Services Research, Department of Biostatistics and Applied Mathematics, The University of Texas M. D. Anderson Cancer Center, 1400 Pressler Street, Unit 1411, Houston, TX 77230; telephone: (713) 563-8920; fax: (713) 563-4243
The Journal of Supportive Oncology
Volume 8, Issue 6, November-December 2010, Pages 242-25
Original research
Elenir B.C. Avritscher MD, PhD, MBA/MHA, a, , Ya-Chen T. Shih PhDa, Charlotte C. Sun DrPHa, Richard J. Gralla MDa, Steven M. Grunberg MDa, Ying Xu MD, MSa and Linda S. Elting DrPHa
Abstract
We estimated the cost-utility of palonosetron-based therapy compared with generic ondansetron-based therapy throughout four cycles of anthracycline and cyclophosphamide for treating women with breast cancer. We developed a Markov model comparing six strategies in which ondansetron and palonosetron are combined with either dexamethasone alone, dexamethasone plus aprepitant following emesis, or dexamethasone plus aprepitant up front. Data on the effectiveness of antiemetics and emesis-related utility were obtained from published sources. Relative to the ondansetron-based two-drug therapy, the incremental cost–effectiveness ratios for the palonosetron-based regimens were $115,490/quality-adjusted life years (QALY) for the two-drug strategy, $199,375/QALY for the two-drug regimen plus aprepitant after emesis, and $200,526/QALY for the three-drug strategy. In sensitivity analysis, using the $100,000/QALY benchmark, the palonosetron-based two-drug strategy and the two-drug regimen plus aprepitant following emesis were shown to be cost-effective in 39% and 26% of the Monte Carlo simulations, respectively, and with changes in values for the effectiveness of antiemetics and the rate of hospitalization. The cost-utility of palonosetron-based therapy exceeds the $100,000/QALY threshold. Future research incorporating the price structure of all antiemetics following ondansetron's recent patent expiration is needed.
Article Outline
Recent advances in emesis control have been possible due to the availability of increasingly more effective antiemetic agents. During the 1990s, the development of first-generation 5-hydroxytryptamine-3 (5-HT3) antagonists (ondansetron, granisetron, tropisetron, and dolasetron) marked a significant improvement in the control of emesis induced by chemotherapy, particularly acute emesis (ie, occurring within 24 hours following chemotherapy).
More recently, two new drugs—palonosetron, a second-generation 5-HT3 antagonist, and aprepitant, a centrally acting neurokinin-1 antagonist—were added to the armamentarium of antiemetic therapy. Compared with other single-dose 5-HT3 antagonists, palonosetron has a higher 5-HT3 binding affinity and longer plasma half-life and has shown superiority in the prevention of delayed emesis (ie, occurring more than 24 hours after chemotherapy administration) following moderately emetogenic chemotherapy with methotrexate, epirubicin, or cisplatin (MEC), including AC-based regimens.[4] and [5] In a recently published clinical trial conducted by Saito et al,6 palonosetron was also shown to be superior to granisetron in preventing delayed and overall emesis when both drugs were combined with dexamethasone following chemotherapy with either AC or cisplatin. As for aprepitant, when added to the standard of a 5-HT3 antagonist and dexamethasone therapy, it has been shown to improve emesis prevention among patients receiving AC-based chemotherapy during the acute, delayed, and overall periods.7
Such benefits have led to a recent revision in the antiemetics guidelines of both the American Society of Clinical Oncology (ASCO) and the National Comprehensive Cancer Network (NCCN), incorporating both palonosetron as one of the recommended 5-HT3 antagonists and aprepitant in combination with a 5-HT3 antagonist and dexamethasone for patients receiving AC-based chemotherapy.[8] and [9] Of note is that the revised 2010 NCCN antiemetic guidelines suggest that palonosetron may be used prior to the start of multiday chemotherapy, which is more likely to cause significant delayed emesis, instead of repeated daily doses of other first-generation 5-HT3 antagonists.9
Given the multiplicity of antiemetic strategies available for prophylaxis of nausea and vomiting associated with AC-based chemotherapy with inherent variability in effectiveness and price, it is critical for existing therapies to be analyzed in terms of both their outcomes and costs. Thus, the purpose of this study is to determine, from a third-party payer perspective, the cost-utility of palonosetron-based therapy in preventing emesis among breast cancer patients receiving four cycles of AC-based chemotherapy relative to generic ondansetron-based antiemetic therapy. Due to variations in the definition of complete emetic response found across antiemetic studies, the analysis will focus on chemotherapy-induced emesis only, rather than nausea and vomiting, as vomiting can be more objectively measured than nausea and, as such, has been more consistently reported.
Patients and Methods
We developed a Markov model to estimate the costs (in 2008 U.S. dollars) and health outcomes associated with emesis among breast cancer patients receiving multiple cycles of AC-based chemotherapy under six prophylactic strategies containing either generic ondansetron (onda) or palonosetron (palo) when each is combined with either dexamethasone (dex) alone, dex plus aprepitant in the subsequent cycles following the occurrence of emesis, or dex plus aprepitant up front (Figure 1). The time horizon for the risk of chemotherapy-induced emesis during each cycle of chemotherapy was 21 days, which is the standard duration of a cycle of AC-based chemotherapy.
Markov Model Comparing Palo-Based Therapy vs Onda-Based Therapy for Prophylaxis of Chemotherapy-Induced Emesis in Breast Cancer Patients Receiving Four Cycles of AC-Based Chemotherapy (1) Onda (32 mg intravenously) + dex (8 mg intravenously) on day 1, followed by dex (4 mg orally twice a day) on days 2−5. (2) Onda (32 mg intravenously) + dex (8 mg intravenously) on day 1, followed by dex (4 mg orally twice a day) on days 2−5 and aprepitant in the subsequent cycles following the occurrence of emesis (ie, onda 16 mg orally + aprepitant 125 mg orally + dex 12 mg orally on day 1 followed by aprepitant 80 mg orally on days 2−3). (3) Palo (0.25 mg intravenously) + dex (8 mg intravenously) on day 1, followed by dex (4 mg orally twice a day) on days 2−5. (4) Palo (0.25 mg intravenously) + dex (8 mg intravenously) on day 1, followed by dex (4 mg orally twice a day) on days 2−5 and aprepitant in the subsequent cycles following the occurrence of emesis (ie, palo 0.25 mg intravenously + aprepitant 125 mg orally + dex 12 mg orally on day 1 followed by aprepitant 80 mg orally on days 2−3). (5) Onda (16 mg orally) + aprepitant (125 mg orally) + dex (12 mg orally) on day 1 followed by aprepitant (80 mg orally) on days 2−3. (6) Palo (0.25 mg intravenously) + aprepitant (125 mg orally) + dex (12 mg orally) on day 1 followed by aprepitant (80 mg orally) on days 2−3. Palo = palonosetron; onda = ondansetron; AC = anthracycline and cyclophosphamide; dex, dexamethasone
We modeled emesis-related outcomes and direct medical costs (from a third-party payer perspective within the context of the U.S. health-care system) over a total of four cycles of chemotherapy as patients receiving AC-based regimens usually undergo at least four cycles of AC.10 We performed all analyses using TreeAge Pro 2009 Suite (Decision Analysis; TreeAge Software, Williamstown, MA). The study was submitted to our institutional review board and was determined to be exempt from review.
Probability Data
Two-drug prophylactic regimens
We estimated the effectiveness of the 5-HT3 antagonists based on secondary analysis of the raw data from the randomized clinical trial (RCT) directly comparing onda and palo when used alone for prevention of emesis associated with MEC, including 90 breast cancer patients from the palo 0.25-mg arm and 82 from the onda 32-mg arm who received AC-based chemotherapy (Table 1).5 Effectiveness estimates for palo 0.25 mg were augmented by data on 117 breast cancer patients on AC-based chemotherapy participating in a multicenter RCT comparing palo with dolasetron (Table 1).4 We assumed that dex adds the same relative benefit to either first- or second-generation 5-HT3 antagonists and obtained the expected additional benefit of dex in preventing acute emesis based on the results of an RCT comparing a single-dose of granisetron in combination with dex vs granisetron given alone to patients undergoing MEC (Table 2).11 Since in the aforementioned study dex was only given on day 1 of chemotherapy, the estimated additional benefit of adding dex to a 5-HT3 inhibitor on the delayed period was obtained from another RCT; this study, conducted by the Italian Group for Antiemetic Research, compared dex alone, dex plus onda, or placebo on days 2−5 of MEC.12
MODEL PARAMETERS | BASE-CASE VALUES (RANGES) | DATA SOURCES |
---|---|---|
Probability of acute emesis control on cycle 1 of AC: | ||
Onda-based two-drug strategyc | 0.84 (0.74−0.93) | Gralla et al,a The Italian Group[5] and [11] |
Palo-based two-drug strategyc | 0.87 (0.81−0.94) | Eisenberg et al,a Gralla et al,a The Italian Group[4], [5] and [11] |
Onda-based three-drug strategyd | 0.88 (0.85−0.91) | Warr et al7 |
Palo-based three-drug strategyd | 0.96 (0.89−0.99) | Grote et al, Grunberg et al[40] and [41] |
Probability of delayed emesis control following control of acute emesis on cycle 1 of ACc: | ||
Onda-based two-drug strategyd | 0.75 (0.62–0.85) | The Italian Group12 |
Palo-based two-drug strategyc | 0.85 (0.78–0.91) | Eisenberg et al,a Gralla et al,a The Italian Group[4], [5] and [12] |
Onda-based three-drug strategyd | 0.86 (0.82–0.90) | Warr et al7 |
Palo-based three-drug strategyc | 0.96 (0.91–0.97) | Eisenberg et al,a Gralla et al,a Warr et al[4], [5] and [7] |
Probability of delayed emesis control following acute emesis on cycle 1 of ACc: | ||
Onda-based two-drug strategyc | 0.46 (0.31–0.62) | Gralla et al,a The Italian Group[5] and [12] |
Palo-based two-drug strategyc | 0.44 (0.27–0.59) | Eisenberg et al,a Gralla et al,a The Italian Group[4], [5] and [12] |
Onda-based three-drug strategyd | 0.44 (0.29–0.57) | Warr et al7 |
Palo-based three-drug strategyc | 0.51 (0.41–0.67) | Eisenberg et al,a Gralla et al,a Warr et al[4], [5] and [7] |
Relative probability of emesis control in subsequent cycles of ACc: | ||
Two-drug therapy | 0.987 (0.970–1.0) | Herrstedt et al14e |
Three-drug therapy | 1.013 (1.0–1.030) | Herrstedt et al14e |
Probability of hospitalization (among patients who develop emesis) per cycle of ACd: | ||
Onda-based regimens | 0.0035 (0.0001−0.019) | Data from Medstat MarketScan16 |
Palo-based regimens | 0.0017 (0.00004−0.0089) | Data from Medstat MarketScan, Haislip et al[16] and [19]b |
Probability of office visit use (among patients who develop emesis) per cycle of ACd: | ||
Onda-based regimens | 0.10 (0.07−0.14) | Data from Medstat MarketScan16 |
Palo-based regimens | 0.05 (0.03−0.07) | Data from Medstat MarketScan, Haislip et al[16] and [19]b |
Probability of rescue medicine utilization use (among patients who develop emesis) per cycle of ACd: | ||
Onda-based regimens | 0.61 (0.46−0.75) | Gralla et al5a |
Palo-based regimens | 0.56 (0.45−0.66) | Eisenberg et al, Gralla et al[4] and [5]a |
Utility weights for emesis per cycle of ACf: | ||
Acute and delayed emesis | 0.15 (0.10−0.20) | Sun et al20 |
Acute emesis and no delayed emesis | 0.76 (0.70−0.83) | Sun et al20 |
No acute emesis and delayed emesis | 0.20 (0.14−0.26) | Sun et al20 |
No acute and no delayed emesis | 0.92 (0.86−0.99) | Sun et al20 |
AC = anthracycline and cyclophosphamide; onda = ondansetron; palo = palonosetron.
a Included in the analysis was the subset of women with breast cancer receiving AC-based chemotherapy.b We obtained an estimate of emesis-related hospitalization and office visit utilization based on data from Medstat MarketScan, HPM subset (Medstat Group, Inc., Ann Arbor, MI) on 707 breast cancer patients who received the first cycle of AC-based chemotherapy from 1996 to 2002 and either were admitted to the hospital or had an office visit for treatment of vomiting or dehydration. Since palo was only introduced into the U.S. market in 2003, we assumed that all these breast cancer patients received onda-based antiemetic prophylaxis. As a result, we estimated the differential rate of health-care resource utilization based on Haislip et al's19 reported differential incidence of extreme events associated with chemotherapy-induced nausea and vomiting experienced by community-based breast cancer patients who received either onda or palo for emesis prophylaxis following the first cycle of chemotherapy.c Of note is that there are two different methods for applying the benefit of adding dex and/or aprepitant to a 5-HT3 antagonist: (1) rate of emesis with 5-HT3* relative risk of emesis by adding dex and/or aprepitant and (2) rate of emesis control with 5-HT3 * relative risk of emesis control by adding dex and/or aprepitant. These produce substantially different results, with the former method skewing the results toward the least effective 5-HT3 and the latter skewing it toward the most effective one. As a result, we estimated the probability of emesis by averaging the results obtained using the two different methods. Of note is that the ranges for these effectiveness estimates were obtained by applying the two different methods to the lower and upper bounds of the 95% confidence intervals derived from the clinical trials comparing the 5-HT3 antagonists when used alone.d Ranges were obtained by constructing 95% confidence intervals for observed proportions using the normal approximation to the binomial distribution.e Ranges are based on the minimum and maximum values observed in Herrstedt et al's14 clinical trial of multicycle chemotherapy.f Ranges are based on the estimate's actual 95% confidence intervals obtained from Sun et al's20 data.
Three-drug prophylactic regimens
We estimated the rate of acute emesis for the three-drug regimens based on data from published studies in which either onda or palo was given in combination with dex and aprepitant on day 1 of MEC (Table 2).[5], [7] and [13] Because aprepitant was either used in combination with dexamethasone or not used on days 2−3 in the trials of palo-based three-drug therapy, we estimated the benefit of adding aprepitant alone to palo on days 2−3 by assuming that the added benefit in the delayed period would be the same as the benefit added to onda. Specifically, we obtained information on the relative risk of delayed emesis control when aprepitant is added on days 2−3 from a large clinical trial of aprepitant combined with onda and dex in breast cancer patients receiving either A or AC chemotherapy (Table 2).7
Effectiveness of antiemetics over multiple cycles of chemotherapy
The estimates of changes in the probability of emesis control over multiple cycles of chemotherapy were obtained from a RCT conducted by Herrstedt et al14 of ondansetron-based two- and three-drug regimens for prevention of chemotherapy-induced nausea and vomiting among breast cancer patients undergoing multiple cycles of AC-based chemotherapy. We assumed that changes in emesis control over four cycles of AC for the palo-based two- and three-drug regimens were similar to the observed changes for the onda-based two- and three-drug strategies, respectively.14
Resource Utilization and Cost Data
The cost of antiemetic prophylaxis was based on the 2008 Medicare Part B reimbursement rates for pharmaceuticals, which reflects the price of ondansetron following its recent patent expiration (Table 3).15 The costs of prophylaxis failures were estimated as follows. In the majority of prophylaxis failures, the only cost is the cost of rescue medication. In such cases, we obtained costs by multiplying the individual doses used for rescue treatment of breast cancer patients on AC participating in the clinical trials comparing palo 0.25 mg with single doses of onda or dolasetron by their unit costs based on the 2008 Medicare Part B reimbursement rates.[5] and [15] For the few patients who are seen in the office for uncontrolled emesis, we obtained estimates of the risk of such emesis-related office visits based on the MarketScan Health Productivity Management (HPM) database from Thomson Reuters on 707 breast cancer patients who received their first cycle of AC-based chemotherapy between 1997 and 2002 (Table 2) and its costs from the 2008 Medicare Physician Fee Schedule Reimbursement for a level III office visit (CPT 99213).[16] and [17]
COST COMPONENT | 2008 U.S.$ (RANGES) | DATA SOURCE |
---|---|---|
Hospitalization | $5,237.00 ($3,921−$6,112)a | HCUP charge data18 Consumer Price Index42 Medicare cost-to-charge ratio43 |
Level III office visit (CPT 99213) | $60.30 ($19.96–$122.46)d | 2008 Medicare Physician Fee Schedule Reimbursement17 |
Prophylactic antiemetics | 2008 Medicare Part B reimbursement rates for pharmaceuticals15 | |
Onda-based two-drug regimen | $49.74 | |
Palo-based two-drug regimen | $207.20 | |
Onda-based three-drug regimen | $324.51 | |
Palo-based three-drug regimen | $482.46 | |
Rescue medicinesb | $35.25 ($21.66–$48.80)c | Eisenberg et al,4 Gralla et al,5 2008 Medicare Part B reimbursement rates for pharmaceuticals15 |
AC = anthracycline and cyclophosphamide; onda = ondansetron; palo = palonosetron; HCUP = Healthcare Cost and Utilization Project
a Charges were inflated to 2008 U.S. dollars using the Consumer Price Index (CPI) for medical care and adjusted to costs using Medicare cost-to-charge ratio. The ranges were based on estimates of the 95% confidence interval.b In the randomized clinical trial directly comparing ondansetron and palonosetron, propulsives accounted for 71% of the rescue medicines used, 5-hydroxytryptamine antagonists for 20%, glucocorticoids for 7%, and aminoalkyl ethers for 2%.5c Costs for rescue medication were obtained by multiplying all drug unit costs by the individual doses used for rescue treatment of breast cancer patients on AC participating in the clinical trials comparing palo 0.25 mg with single doses of onda or dolasetron.[5] and [15] The ranges were based on estimates of the 95% confidence interval.d Ranges were based on the Medicare physician fee schedule for levels I and VI office visits.
Finally, although hospitalization for emesis is extremely rare in this population, when it occurs, it is quite expensive. For completeness, we obtained estimates of the risk of emesis-related hospitalization from the same population of breast cancer patients from whom we obtained the estimate for the risk of emesis-related office visit, whereas hospital costs were obtained from Healthcare Cost and Utilization Project (HCUP) data on 2,342 breast cancer patients who were hospitalized with a primary or admitting diagnosis of vomiting or dehydration from 1997 to 2003 ([Table 2] and [Table 3]).[16] and [18]
Of note is that since palo was only introduced into the U.S. market in 2003, we anticipated the observed risk of emesis-related office visit and hospital admission obtained from MarketScan data during the period 1997−2002 reflected the risk associated with prophylaxis with onda. As a result, given that, when compared with onda, palo has also shown superiority in reducing the severity of emetic episodes when they occur, we estimated the differential rate of health-care resource utilization for palo and onda based on Haislip et al's reported differential incidence of extreme events associated with chemotherapy-induced nausea and vomiting (CINV) experienced by community-based breast cancer patients who received either palo or onda for emesis prophylaxis following the first cycle of chemotherapy (Table 2).[5] and [19]
Utility Data
We obtained the utility weights for acute and delayed emesis from a published study of preferences elicited from ovarian cancer patients undergoing chemotherapy using a modified visual analog scale (VAS) (Table 2).20 We equally applied these emesis-related utility weights to the initial 5-day period of chemotherapy (the standard duration of follow-up in clinical trials of prophylactic antiemetics) in all six prophylactic strategies of the decision tree. Furthermore, because the risk of CINV after 5 days of chemotherapy is usually so negligible as to be unmeasured in clinical trials of antiemetics, we assumed the utility weights for the remaining 16 days of each of the chemotherapy cycles to be the same as the weight associated with complete emesis control (ie, 0.92). We subsequently converted the resulting estimates of quality-adjusted life days into quality-adjusted life years (QALY).
Analysis
We used a stepwise method to calculate the incremental cost–effectiveness ratios of the different prophylactic therapy strategies, with the generic onda-based two-drug therapy (ie, the lowest cost strategy) as the base comparator (also known as the “anchor”).21 We adopted the benchmark range of U.S. $50,000−$100,000 per QALY, which has been commonly cited for oncology-related interventions as the threshold for acceptable cost–effectiveness, and examined the robustness of the results by performing one-way sensitivity analyses of plausible ranges for the model's key parameters based on the data sources used as well as probabilistic sensitivity analysis using Monte Carlo simulation.[21] and [22]
Results
The overall rate of emesis control (on days 1−5) among breast cancer patients following a cycle of AC-based chemotherapy was estimated to be 63% (range 46%−79%) for the onda-based two-drug therapy, 74% (range 66%−85%) for the palo-based two-drug therapy, 76% (range 75%−82%) for the onda-based three-drug therapy, and 92% (range 81%−96%) for the palo-based three-drug therapy. Based on these estimates, relative to the onda-based two-drug therapy, the incremental cost–effectiveness ratios (ICERs) for the palo-based regimens were $115,490/QALY for the two-drug strategy, $199,375/QALY for the two-drug regimen plus aprepitant after emesis, and $200,526/QALY for the three-drug strategy (Table 4). The onda-based two-drug combination plus aprepitant after the onset of emesis was eliminated through extended dominance as it has a greater ICER than the next more effective therapy, the palo-based two-drug treatment strategy (Table 4). The onda-based three-drug strategy was dominated by the palo-based two-drug combination plus aprepitant after the onset of emesis as the former strategy is both less effective and more expensive than the latter (Table 4).
STRATEGY | TOTAL COST (U.S.$) | INCREMENTAL COST (U.S.$) | EFFECTIVENESS (QALY) | INCREMENTAL EFFECTIVENESS (QALY) | INCREMENTAL COST–EFFECTIVENESS (U.S.$/QALY) |
---|---|---|---|---|---|
Onda-based two-drug therapy | $269 | — | 0.1989 | — | — |
Onda-based two-drug therapy with aprepitant after emesis | $635 | $366 | 0.2010 | 0.0021 | $174, 286 Eliminated through extended dominancea |
Palo-based two-drug therapy | $858 | $589 | 0.2040 | 0.0051 | $115,490c |
Palo-based two-drug therapy plus aprepitant after emesis | $1,177 | $319 | 0.2056 | 0.0016 | 199,375 |
Onda-based three-drug therapy | $1,336 | $159 | 0.205 | (0.0006) | Dominatedb |
Palo-based three-drug therapy | $1,939 | $603 | 0.2094 | 0.0044 | $200,526d |
QALY = quality-adjusted life year; AC = anthracycline and cyclophosphamide; ICER = incremental cost–effectiveness ratio; onda = ondansetron; palo = palonosetron
a Extended dominance occurs when one of the treatment alternatives has a greater ICER than the next more effective alternative.b One intervention is said to be dominated by another when it is both less effective and more expensive than the previous less costly alternative.c Because the onda-based two-drug combination plus aprepitant after the onset of emesis was eliminated through extended dominance, the palo-based two-drug therapy was compared with the onda-based two-drug therapy.d Because the onda-based three-drug combination was dominated by the palo-based two-drug combination plus aprepitant after the onset of emesis, the palo-based three-drug therapy was compared with the latter regimen.
In sensitivity analyses using the commonly accepted cost–effectiveness benchmark range of $50,000−$100,000/QALY, the results were sensitive to changes in the overall emesis control rates for the onda-based two-drug strategy. If the probability of overall emesis control for the onda-based two-drug strategy was as low as its estimated lower bound (46%), the ICER for the palo-based two-drug treatment alternative would drop to $53,892/QALY. The results were also sensitive to changes in the effectiveness for the palo-based two-drug regimen: When its overall control rate was as high as its estimated upper bound (86%), its ICER would be $71,472. In contrast, the results were not sensitive to variations in the probability of overall emesis control for the three-drug strategies, nor were they sensitive to changes in the relative probability of emesis control in subsequent cycles of AC for either the two- or three-drug strategies.
If the probability of emesis-related hospitalization was as high as the upper limit of its 95% confidence interval (CI), the ICER for the palo-based two-drug regimen would be $97,301/QALY. However, changes in the cost of an emesis-related admission (95% CI $3,921−$6,112) did not significantly alter the results, nor did variations in office visit and rescue medicine utilization and their associated costs. The results were also not sensitive to variations in the values for the utility weights throughout their 95% CIs. We performed a threshold analysis to explore the price per dose of palo that would result in an acceptable cost–effectiveness ratio under the $100,000/QALY benchmark and found that the ICER for the palo-based two-drug treatment alternative would only fall to a $100,000/QALY threshold when the cost of palo is decreased by 11%.
Figure 2 shows the cost–effectiveness acceptability curves for each strategy, with the onda-based two-drug therapy as the base comparator. These curves show the proportion of the 100,000 simulations in which the comparing antiemetic regimen was considered more cost-effective than the base comparator at different thresholds. Using the benchmark of U.S. $100,000/QALY, the palo-based two-drug strategy and the two-drug regimen plus aprepitant following the onset of emesis were shown to be cost-effective in 39% and 26% of the simulations with the onda-based standard therapy as the baseline, respectively, whereas the palo-based and onda-based three-drug strategies and the onda-based two-drug regimen with aprepitant after emesis were cost-effective in fewer than 10% of the simulations. Of note is that the slope of the acceptability curves for the palo-based two-drug strategies are steep when willingness to pay exceeds $50,000/QALY, indicating that the greater the threshold, the greater the increase in the level of confidence that these strategies could be cost-effective. For example, the probability that the palo-based two-drug strategy is more cost-effective than the onda-based two-drug strategy rises to 51% at a threshold value of $125,000/QALY and exceeds 60% at $150,000/QALY.
Figure 3 presents the scatterplot of the results of the probabilistic sensitivity analysis for the palo-based two-drug strategy. Nearly 96% of the simulations fell within the first quadrant of the chart (ie, on the upper right quadrant), which represents the scenario where the palo-based two-drug therapy is more costly but also more effective than the onda-based standard therapy. However, only 39% of the simulations fell below the $100,000/QALY dashed threshold line, which represents the scenario where the palo-based two-drug strategy is more cost-effective than the onda-based standard therapy at the $100,000/QALY benchmark.
Discussion
Our estimates of emesis-related costs and outcomes following four cycles of AC-based chemotherapy in women with breast cancer indicate that at current antiemetic prices and utilities placed on emesis, the additional costs of palo and aprepitant are not warranted at the $100,000/QALY threshold. In probabilistic sensitivity analysis, the palo-based two-drug strategy and the two-drug regimen plus aprepitant following the onset of emesis were shown to be cost-effective at the $100,000/QALY threshold in only 39% and 26% of the simulations, respectively. The model was sensitive to changes in the values of antiemetic effectiveness for the two-drug regimens and the risk of emesis-related hospitalization.
In threshold analysis, the two-drug palo-based regimen was cost-effective at the $100,000/QALY benchmark when the cost of palo is decreased by 11%. Because the use of the $100,000/QALY threshold is uncommon in clinical practice, the cost-effectiveness of the palo-based two-drug strategy (estimated at $115,490/QALY in our study) compares favorably with other commonly used supportive care measures for women with breast cancer. Such measures include primary prophylaxis with granulocyte colony-stimulating factor in women undergoing chemotherapy with moderate to high myelosuppressive risk (ICER of $116,000/QALY, or $125,948/QALY in 2008 U.S. dollars) and the use of bisphosphonates for the prevention of skeletal complications in breast cancer patients with lytic bone metastases (ICER ranging from $108,200/QALY with chemotherapy as systemic therapy to $305,300 in conjunction with hormonal systemic therapy, or $166,381/QALY to $469,466/QALY in 2008 U.S. dollars, respectively).[23] and [24] Both interventions are considered recommended standards of supportive care for patients with breast cancer and are widely used in breast oncology practices.[25] and [26]
Decision-analytic models, such as the Markov model presented in our study, aim to reflect the reality of clinical practice in a simplified way. Therefore, modelers often need to make decisions regarding the study time frame and model parameters based on the best use of available data. In our study, we obtained estimates for the probability of chemotherapy-induced emesis from studies in which the standard duration of follow-up is 5 days. By so doing, we may have underestimated the cost-effectiveness for the palo-based and aprepitant-based regimens. Although the risk of CINV after 5 days of chemotherapy is usually negligible, anticipation of vomiting may affect a patient's quality of life throughout the cycle of chemotherapy.
In addition, our estimates of costs, which were mostly obtained from Medicare, may differ from those of other third-party payers. However, Medicare is among the largest payers for breast cancer care as 42% of the women diagnosed with cancer in the United States are older than 64 years, and many private organizations set their own reimbursement rates based on the Medicare schedule. Therefore, we believe that Medicare reimbursement data provide a suitable estimate for emesis-related medical costs for all breast cancer patients in the United States.[27] and [28]
The present results should solely be interpreted in light of the cost–effectiveness benchmark of $50,000−$100,000/QALY, which has been frequently used in the context of the U.S. health-care system.[22] and [29] Such a benchmark, however, is a historic, precedent-based threshold set by the cost of caring for patients on dialysis, which was estimated at $50,000/QALY in 1982 ($74,000−$95,000 in 1997 U.S. dollars).[30] and [31] Given the arbitrariness of such a threshold, it has been suggested that the current willingness to pay for medical interventions in the United States probably exceeds $100,000/QALY, with values as high as $300,000/QALY being cited in some oncology publications.[22], [29], [31], [32], [33] and [34] In support of that argument is the public and policy makers' strong negative reaction to the National Institutes of Health Consensus Panel not recommending mammography screening for women aged 40−49 years, a procedure reported to provide an ICER of $105,000 per life-year gained.[35] and [36] As a result, if willingness to pay goes beyond $100,000/QALY, the alternative of adding aprepitant to palo plus dex may also be deemed attractive as the slope of its acceptability curve becomes substantially steep when the willingness to pay for a QALY exceeds $125,000 (Figure 2), suggesting that its marginal gain may exceed its marginal costs at higher thresholds.
In addition, it is worth noting that the present analysis has been conducted from the perspective of a third-party payer within the context of the U.S. health-care system. The large difference in the acquisition cost of palo-based and onda-based therapy observed in the United States is mostly driven by the differential stage of product life cycles for palo and onda. Although at the time of this study palo was still under patent protection, generic onda had entered the U.S. market prior to our study. The large price discrepancy between brand and generic drugs explains the difference in drug costs in this U.S.-based analysis. As such, our results may not reflect the situation in countries with a widely different cost structure, in which the acquisition cost of palo may be substantially lower. When that is the case, the cost–effectiveness profile of the palo-based prophylactic therapy may be deemed substantially more favorable than the profile presented here. Similarly, we anticipate finding a more attractive cost–effectiveness profile for the palo-based therapies as palo reaches the end of its product life cycle in the U.S. market.37 Also of note is that the cost–effectiveness of the palo-based therapy may greatly differ when different perspectives (other than the third-party payer's perspective) are adopted.
Our study, however, has several limitations. First, the utility scores used in our model were derived with a VAS instrument, which does not incorporate patients' preferences under uncertainty. Nevertheless, the VAS approach has been shown to provide utility scores for nausea and vomiting with more variability than scores derived using other methods such as the Standard Gamble (personal communication, Grunberg SM et al, CALGB study 309801). Notwithstanding that, it remains unclear which method gives utility scores for transient health states, such as CINV, with the greatest validity.
Also of note is that due to a lack of information on emesis-related utilities among breast cancer patients in the literature, we used utilities elicited from patients with ovarian cancer. To the best of our knowledge, the utilities in Sun et al20 were the only ones available in the literature that were elicited from a homogeneous population of cancer patients (ie, solely patients with ovarian cancer) and were based on a wide range of health states combining the presence and absence of emesis during either the acute or the delayed period. In addition, the participants in the Sun et al study were treated with carboplatin, which, like the regimen used in our model, is classified as moderately emetogenic in established antiemetic guidelines.[8], [9] and [38] It is also important to emphasize that the population in that study, like our study's population, was composed exclusively of women, who are known to be at increased risk for developing CINV.39
Second, in the absence of clinical trial data, we assumed conservatively that dex and aprepitant add the same relative benefit to both onda and palo. This assumption results in an imperfect estimate of cost–effectiveness. As such, we may have overestimated or underestimated the cost–effectiveness of palo as dex and aprepitant may potentially add less value to the intrinsically more active 5-HT3 antagonist or uniquely complementary mechanisms of action could contribute to even greater activity with the palo-based therapy. However, our study's estimate of the relative effectiveness of the palo-based two-drug prophylactic therapy versus the onda-based two-drug therapy for preventing delayed emesis is consistent with that reported in a recently published clinical trial comparing palo and granisetron when both drugs are combined with dex following chemotherapy with either AC or cisplatin (1.18 vs 1.17, respectively).6
Third, our study did not include the outcomes associated with the adverse effects of antiemetics, and by so doing, we may have underestimated the costs associated with antiemetic prophylaxis. However, the incidence and duration of treatment-related adverse events occurring in the two RCTs comparing palo with either onda or dolasetron were mild and similar across treatment cohorts.[4] and [5]
Fourth, we assumed that changes in emesis control in subsequent cycles of AC for the palo-based regimens were the same as for the onda-based therapy. By so doing, we may have underestimated the cost–effectiveness of palo as the superiority of the more active 5-HT3 antagonist could be maintained in the subsequent cycles of chemotherapy (or even increased, as seen in the aprepitant-based arm of Herrstedt et al's14 study). As a result, if future prospective trials of palo-based antiemetic prophylaxis confirm its superiority in maintaining antiemetic efficacy over multiple cycles of AC, the cost–effectiveness profiles for the palo-based strategies may be more favorable than the profiles presented herein.
Last, the incremental gains in QALY observed in cost–utility analysis of interventions associated with transitory and non-life-threatening health states, such as the antiemetic regimens analyzed in our study, tend to render small denominators to be used in the incremental cost–effectiveness ratios. The issue of small denominators has led some researchers to question whether the current methodology of cost–effectiveness analysis is appropriate to determine the cost–effectiveness of treatments for terminal or supportive care.32 However, despite this shortcoming, these types of analysis benefit from having a wider scope as they allow comparisons over different types of health interventions across various diseases. In addition, by incorporating patients' utility levels over different health states (instead of merely looking into cost per additional patient controlled), cost–utility analysis makes explicit the impact of the target population's preferences for the different outcomes. Of importance is that both the Panel on Cost–Effectiveness in Health and Medicine and the Institute of Medicine (IOM) Committee on Regulatory Cost–Effectiveness Analysis recommend the use of QALY as the preferred outcome measure for economic evaluation of health-care interventions.
Conclusion
Although our base-case analysis suggests that, from a third-party payer perspective within the context of the U.S. health-care system, the cost–utility of the palo-based two-drug prophylactic therapy for breast cancer patients receiving four cycles of AC-based chemotherapy exceeds the $50,000–$100,000/QALY threshold, it is comparable to other commonly used supportive care interventions for women with breast cancer. In sensitivity analyses, such a strategy was associated with a 39% chance of being cost-effective at the $100,000/QALY threshold, and the model was sensitive to changes in the values of antiemetic effectiveness and of the probability of emesis-related hospitalization. In threshold analysis, the combination of palo and dex was shown to become cost-effective (at the $100,000/QALY benchmark) when the cost of palo is decreased by 11%. As a result, future research incorporating the price structure of all antiemetics following the recent expiration of onda's patent is needed.
References1
1 S.M. Grunberg, D. Osoba and P.J. Hesketh et al., Evaluation of new antiemetic agents and definition of antineoplastic agent emetogenicity—an update, Support Care Cancer 13 (2005), pp. 80–84 [15599601]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (68)
2 C.M. Booth, M. Clemons and G. Dranitsaris et al., Chemotherapy-induced nausea and vomiting in breast cancer patients: a prospective observational study, J Support Oncol 5 (2007), pp. 374–380 [17944146]. View Record in Scopus | Cited By in Scopus (10)
3 M. de Boer-Dennert, R. de Wit and P.I. Schmitz et al., Patient perceptions of the side-effects of chemotherapy: the influence of 5HT3 antagonists, Br J Cancer 76 (1997), pp. 1055–1061 [9376266]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (128)
4 P. Eisenberg, J. Figueroa-Vadillo, R. Zamora et al. and 99-04 Palonosetron Study Group, Improved prevention of moderately emetogenic chemotherapy-induced nausea and vomiting with palonosetron, a pharmacologically novel 5-HT3 receptor antagonist: results of a phase III, single-dose trial versus dolasetron, Cancer 98 (2003), pp. 2473–2482 [14635083]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (171)
5 R. Gralla, M. Lichinitser and S. Van Der Vegt et al., Palonosetron improves prevention of chemotherapy-induced nausea and vomiting following moderately emetogenic chemotherapy: results of a double-blind randomized phase III trial comparing single doses of palonosetron with ondansetron, Ann Oncol 14 (2003), pp. 1570–1577 [14504060]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (177)
6 M. Saito, K. Aogi and I. Sekine et al., Palonosetron plus dexamethasone versus granisetron plus dexamethasone for prevention of nausea and vomiting during chemotherapy: a double-blind, double-dummy, randomised, comparative phase III trial, Lancet Oncol 10 (2009), pp. 115–124 [19135415]. Article | | View Record in Scopus | Cited By in Scopus (43)
7 D.G. Warr, P.J. Hesketh and R.J. Gralla et al., Efficacy and tolerability of aprepitant for the prevention of chemotherapy-induced nausea and vomiting in patients with breast cancer after moderately emetogenic chemotherapy, J Clin Oncol 23 (2005), pp. 2822–2830 [15837996]. View Record in Scopus | Cited By in Scopus (139)
8 American Society of Clinical Oncology, M.G. Kris, P.J. Hesketh and M.R. Somerfield et al., American Society of Clinical Oncology guideline for antiemetics in oncology: update 2006, J Clin Oncol 24 (2006), pp. 2932–2947 [16717289]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (311)
9 D.S. Ettinger, D.K. Armstrong and S. Barbour et al., National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology—Antiemesis, version 2.2010 http://www.nccn.org/professionals/physician_gls/PDF/antiemesis.pdf Accessed September 20, 2010.
10 R.W. Carlson and B. McCormick, Update: NCCN breast cancer clinical practice guidelines, J Natl Compr Cancer Netw 3 (suppl 1) (2005), pp. S7–S11 [16280118].
11 The Italian Group for Antiemetic Research, Dexamethasone, granisetron, or both for the prevention of nausea and vomiting during chemotherapy for cancer, N Engl J Med 332 (1995), pp. 1–5 [7990859].
12 The Italian Group for Antiemetic Research, Dexamethasone alone or in combination with ondansetron for the prevention of delayed nausea and vomiting induced by chemotherapy, N Engl J Med 342 (2000), pp. 1554–1559 [10824073].
13 S.M. Grunberg, M. Dugan and H. Muss et al., Effectiveness of a single-day three-drug regimen of dexamethasone, palonosetron, and aprepitant for the prevention of acute and delayed nausea and vomiting caused by moderately emetogenic chemotherapy, Support Care Cancer 17 (2009), pp. 589–594 [19037667]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (14)
14 J. Herrstedt, H.B. Muss and D.G. Warr et al., Efficacy and tolerability of aprepitant for the prevention of chemotherapy-induced nausea and emesis over multiple cycles of moderately emetogenic chemotherapy, Cancer 104 (2005), pp. 1548–1555 [16104039]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (43)
15 Centers for Medicare and Medicaid Services, Medicare Part B Drug Average Sales Price: 2008 ASP Drug Pricing Files http://www.cms.hhs.gov/apps/ama/license.asp?file=/McrPartBDrugAvgSalesPrice/downloads/July2008ASPPricingFilebyHCPCS.zip Accessed July 18, 2008.
16 Thomson. Medstat, 1997–2002 MarketScan Health and Productivity Management Database User Guide and Data Dictionary, Thomson Medstat, Ann Arbor, MI (2003).
17 Centers for Medicare and Medicaid Services, National Physician Fee Schedule and Relative Value: 2008 Physician Fee Schedule National Payment Amount File http://www.cms.hhs.gov/PFSlookup/02_PFSSearch.asp Accessed July 18, 2008.
18 National Inpatient Sample (NIS), NIS description of data elements, Healthcare Cost and Utilization Project (HCUP) databases, Agency for Healthcare Research and Quality, Rockville, MD (2004) http://www.hcup-us.ahrq.gov/nisoverview.jsp#Data Accessed May 16, 2010.
19 S. Haislip, J. Gilmore, W.H. Lenz, T. Gondesen and B. Feinberg, Theory in practice: improving patient outcomes and practice efficiency with a simple change in 5-HT3 receptor antagonist for preventing chemotherapy-induced nausea and vomiting (CINV) In: Third Annual Meeting of the Hematology/Oncology Pharmacy Association; Abstract #PR6. June 14–16, 2007; Denver, Colorado http://www.hoparx.org/documents/2007programbook.pdf Accessed November 2, 2010.
20 C.C. Sun, D.C. Bodurka and C.B. Weaver et al., Rankings and symptom assessments of side effects from chemotherapy: insights from experienced patients with ovarian cancer, Support Care Cancer 13 (2005), pp. 219–227 [15538640]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (53)
21 M.F. Drummond, M.J. Sculpher, G.W. Torrance, B.J. O'Brien and G.L. Stoddart, Methods for the Economic Evaluation of Health Care Programmes (3rd ed.), Oxford University Press, New York (2005).
22 J. Hayman, J. Weeks and P. Mauch, Economic analyses in health care: an introduction to the methodology with an emphasis on radiation therapy, Int J Radiat Oncol Biol Phys 35 (1996), pp. 827–841 [8690653]. Article | | View Record in Scopus | Cited By in Scopus (33)
23 B.E. Hillner, J.C. Weeks, C.E. Desch and T.J. Smith, Pamidronate in prevention of bone complications in metastatic breast cancer: a cost–effectiveness analysis, J Clin Oncol 18 (2000), pp. 72–79 [10623695]. View Record in Scopus | Cited By in Scopus (90)
24 S.D. Ramsey, Z. Liu and R. Boer et al., Cost–effectiveness of primary versus secondary prophylaxis with pegfilgrastim in women with early-stage breast cancer receiving chemotherapy, Value Health 11 (2008), pp. 172–179 [18673353].
25 B.E. Hillner, J.N. Ingle, R.T. Chlebowski et al. and American Society of Clinical Oncology, American Society of Clinical Oncology 2003 update on the role of bisphosphonates and bone health issues in women with breast cancer, J Clin Oncol 21 (2003), pp. 4042–4057 [12963702]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (533)
26 T.J. Smith, J. Khatcheressian and G.H. Lyman et al., 2006 update of recommendations for the use of white blood cell growth factors: an evidence-based clinical practice guideline, J Clin Oncol 24 (2006), pp. 3187–3205 [16682719]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (485)
27 National Cancer Institute, Surveillance Epidemiology and End Results: SEER Stat Fact Sheets: Breast http://seer.cancer.gov/statfacts/html/breast.html Accessed May 16, 2010.
28 J.W. Tumeh, S.G. Moore, R. Shapiro and C.R. Flowers, Practical approach for using Medicare data to estimate costs for cost–effectiveness analysis, Expert Rev Pharmacoecon Outcomes Res 5 (2005), pp. 153–162 [19807571]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
29 P.A. Ubel, R.A. Hirth, M.E. Chernew and A.M. Fendrick, What is the price of life and why doesn't it increase at the rate of inflation?, Arch Intern Med 163 (2003), pp. 1637–1641 [12885677]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (225)
30 J.C. Hornberger, D.A. Redelmeier and J. Petersen, Variability among methods to assess patients' well-being and consequent effect on a cost–effectiveness analysis, J Clin Epidemiol 45 (1992), pp. 505–512 [1588356]. Article | | View Record in Scopus | Cited By in Scopus (138)
31 R.A. Hirth, M.E. Chernew, E. Miller, A.M. Fendrick and W.G. Weissert, Willingness to pay for a quality-adjusted life year: in search of a standard, Med Decis Making 20 (2000), pp. 332–342 [10929856]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (291)
32 Y.C. Shih and M.T. Halpern, Economic evaluations of medical care interventions for cancer patients: how, why, and what does it mean?, CA Cancer J Clin 58 (2008), pp. 231–244 [18596196]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (18)
33 E. Nadler, B. Eckert and P.J. Neumann, Do oncologists believe new cancer drugs offer good value?, Oncologist 11 (2006), pp. 90–95 [16476830]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (50)
34 R.S. Braithwaite, D.O. Meltzer, J.T. King Jr, D. Leslie and M.S. Roberts, What does the value of modern medicine say about the $50,000 per quality-adjusted life-year decision rule?, Med Care 46 (2008), pp. 349–356 [18362813]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
35 National Institutes of Health Consensus Development Panel, 1997 Consensus Statement: Breast Cancer Screening for Women Ages 40–49 http://consensus.nih.gov/1997/1997BreastCancerScreening103html.htm Accessed October 13, 2007.
36 P. Salzmann, K. Kerlikowske and K. Phillips, Cost–effectiveness of extending screening mammography guidelines to include women 40 to 49 years of age, Ann Intern Med 127 (1997), pp. 955–965 [9412300]. View Record in Scopus | Cited By in Scopus (169)
37 Y.C. Shih, S. Han and S.B. Cantor, Impact of generic drug entry on cost–effectiveness analysis, Med Decis Making 25 (2005), pp. 71–80 [15673583]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
38 F. Roila, P.J. Hesketh, J. Herrstedt and Antiemetic Subcommittee of the Multinational Association of Supportive Care in Cancer, Prevention of chemotherapy- and radiotherapy-induced emesis: results of the 2004 Perugia International Antiemetic Consensus Conference, Ann Oncol 17 (2006), pp. 20–28 [16314401]. View Record in Scopus | Cited By in Scopus (90)
39 S.M. Grunberg and A. Ireland, Epidemiology of chemotherapy-induced nausea and vomiting, Adv Studies Nurs 3 (1) (2005), pp. 9–15 http://www.jhasin.com/files/articlefiles/pdf/XASIN_3_1_p9_15.pdf Accessed September 16, 2010.
40 T. Grote, J. Hajdenberg, A. Cartmell, S. Ferguson, A. Ginkel and V. Charu, Combination therapy for chemotherapy-induced nausea and vomiting in patients receiving moderately emetogenic chemotherapy: palonosetron, dexamethasone, and aprepitant, J Support Oncol 4 (2006), pp. 403–408 [17004515]. View Record in Scopus | Cited By in Scopus (38)
41 S.M. Grunberg, M. Dugan, H.B. Muss, M. Wood, S. Burdette-Radoux and T. Weisberg, Efficacy of a 1-day 3-drug antiemetic regimen for prevention of acute and delayed nausea and vomiting induced by moderately emetogenic chemotherapy, J Clin Oncol 25 (18S) (2007), p. 9111.
42 U. S. Department of Labor. Bureau of Labor Statistics. Consumer Price Index http://www.bls.gov/cpi/home.htm Accessed May 16, 2010.
43 Department of Health and Human Services. Centers for Medicare & Medicaid Services, Medicare Program; Proposed Changes to the Hospital Inpatient Prospective Payment Systems and Fiscal Year 2008 Rates CMS-1533-P, pp 1070–1073 http://www.cms.hhs.gov/AcuteInpatientPPS/downloads/CMS-1533-P.pdf Accessed May 16, 2010.
Conflicts of interest: Dr. Sun discloses that her husband was an employee of MGI Pharma, Inc., at the time this article was being written. Dr. Gralla discloses that he is a consultant for MGI Pharma, Inc., GlaxoSmithKline, Sanofi-aventis, and Merck; he also receives honoraria from MGI Pharma, Inc., and Merck and research support from Sanofi-aventis. Dr. Grunberg discloses that he is a consultant for MGI Pharma, Inc.
Correspondence to: Elenir B. C. Avritscher, MD, PhD, MBA/MHA, Section of Health Services Research, Department of Biostatistics and Applied Mathematics, The University of Texas M. D. Anderson Cancer Center, 1400 Pressler Street, Unit 1411, Houston, TX 77230; telephone: (713) 563-8920; fax: (713) 563-4243
The Journal of Supportive Oncology
Volume 8, Issue 6, November-December 2010, Pages 242-25
Original research
Elenir B.C. Avritscher MD, PhD, MBA/MHA, a, , Ya-Chen T. Shih PhDa, Charlotte C. Sun DrPHa, Richard J. Gralla MDa, Steven M. Grunberg MDa, Ying Xu MD, MSa and Linda S. Elting DrPHa
Abstract
We estimated the cost-utility of palonosetron-based therapy compared with generic ondansetron-based therapy throughout four cycles of anthracycline and cyclophosphamide for treating women with breast cancer. We developed a Markov model comparing six strategies in which ondansetron and palonosetron are combined with either dexamethasone alone, dexamethasone plus aprepitant following emesis, or dexamethasone plus aprepitant up front. Data on the effectiveness of antiemetics and emesis-related utility were obtained from published sources. Relative to the ondansetron-based two-drug therapy, the incremental cost–effectiveness ratios for the palonosetron-based regimens were $115,490/quality-adjusted life years (QALY) for the two-drug strategy, $199,375/QALY for the two-drug regimen plus aprepitant after emesis, and $200,526/QALY for the three-drug strategy. In sensitivity analysis, using the $100,000/QALY benchmark, the palonosetron-based two-drug strategy and the two-drug regimen plus aprepitant following emesis were shown to be cost-effective in 39% and 26% of the Monte Carlo simulations, respectively, and with changes in values for the effectiveness of antiemetics and the rate of hospitalization. The cost-utility of palonosetron-based therapy exceeds the $100,000/QALY threshold. Future research incorporating the price structure of all antiemetics following ondansetron's recent patent expiration is needed.
Article Outline
Recent advances in emesis control have been possible due to the availability of increasingly more effective antiemetic agents. During the 1990s, the development of first-generation 5-hydroxytryptamine-3 (5-HT3) antagonists (ondansetron, granisetron, tropisetron, and dolasetron) marked a significant improvement in the control of emesis induced by chemotherapy, particularly acute emesis (ie, occurring within 24 hours following chemotherapy).
More recently, two new drugs—palonosetron, a second-generation 5-HT3 antagonist, and aprepitant, a centrally acting neurokinin-1 antagonist—were added to the armamentarium of antiemetic therapy. Compared with other single-dose 5-HT3 antagonists, palonosetron has a higher 5-HT3 binding affinity and longer plasma half-life and has shown superiority in the prevention of delayed emesis (ie, occurring more than 24 hours after chemotherapy administration) following moderately emetogenic chemotherapy with methotrexate, epirubicin, or cisplatin (MEC), including AC-based regimens.[4] and [5] In a recently published clinical trial conducted by Saito et al,6 palonosetron was also shown to be superior to granisetron in preventing delayed and overall emesis when both drugs were combined with dexamethasone following chemotherapy with either AC or cisplatin. As for aprepitant, when added to the standard of a 5-HT3 antagonist and dexamethasone therapy, it has been shown to improve emesis prevention among patients receiving AC-based chemotherapy during the acute, delayed, and overall periods.7
Such benefits have led to a recent revision in the antiemetics guidelines of both the American Society of Clinical Oncology (ASCO) and the National Comprehensive Cancer Network (NCCN), incorporating both palonosetron as one of the recommended 5-HT3 antagonists and aprepitant in combination with a 5-HT3 antagonist and dexamethasone for patients receiving AC-based chemotherapy.[8] and [9] Of note is that the revised 2010 NCCN antiemetic guidelines suggest that palonosetron may be used prior to the start of multiday chemotherapy, which is more likely to cause significant delayed emesis, instead of repeated daily doses of other first-generation 5-HT3 antagonists.9
Given the multiplicity of antiemetic strategies available for prophylaxis of nausea and vomiting associated with AC-based chemotherapy with inherent variability in effectiveness and price, it is critical for existing therapies to be analyzed in terms of both their outcomes and costs. Thus, the purpose of this study is to determine, from a third-party payer perspective, the cost-utility of palonosetron-based therapy in preventing emesis among breast cancer patients receiving four cycles of AC-based chemotherapy relative to generic ondansetron-based antiemetic therapy. Due to variations in the definition of complete emetic response found across antiemetic studies, the analysis will focus on chemotherapy-induced emesis only, rather than nausea and vomiting, as vomiting can be more objectively measured than nausea and, as such, has been more consistently reported.
Patients and Methods
We developed a Markov model to estimate the costs (in 2008 U.S. dollars) and health outcomes associated with emesis among breast cancer patients receiving multiple cycles of AC-based chemotherapy under six prophylactic strategies containing either generic ondansetron (onda) or palonosetron (palo) when each is combined with either dexamethasone (dex) alone, dex plus aprepitant in the subsequent cycles following the occurrence of emesis, or dex plus aprepitant up front (Figure 1). The time horizon for the risk of chemotherapy-induced emesis during each cycle of chemotherapy was 21 days, which is the standard duration of a cycle of AC-based chemotherapy.
Markov Model Comparing Palo-Based Therapy vs Onda-Based Therapy for Prophylaxis of Chemotherapy-Induced Emesis in Breast Cancer Patients Receiving Four Cycles of AC-Based Chemotherapy (1) Onda (32 mg intravenously) + dex (8 mg intravenously) on day 1, followed by dex (4 mg orally twice a day) on days 2−5. (2) Onda (32 mg intravenously) + dex (8 mg intravenously) on day 1, followed by dex (4 mg orally twice a day) on days 2−5 and aprepitant in the subsequent cycles following the occurrence of emesis (ie, onda 16 mg orally + aprepitant 125 mg orally + dex 12 mg orally on day 1 followed by aprepitant 80 mg orally on days 2−3). (3) Palo (0.25 mg intravenously) + dex (8 mg intravenously) on day 1, followed by dex (4 mg orally twice a day) on days 2−5. (4) Palo (0.25 mg intravenously) + dex (8 mg intravenously) on day 1, followed by dex (4 mg orally twice a day) on days 2−5 and aprepitant in the subsequent cycles following the occurrence of emesis (ie, palo 0.25 mg intravenously + aprepitant 125 mg orally + dex 12 mg orally on day 1 followed by aprepitant 80 mg orally on days 2−3). (5) Onda (16 mg orally) + aprepitant (125 mg orally) + dex (12 mg orally) on day 1 followed by aprepitant (80 mg orally) on days 2−3. (6) Palo (0.25 mg intravenously) + aprepitant (125 mg orally) + dex (12 mg orally) on day 1 followed by aprepitant (80 mg orally) on days 2−3. Palo = palonosetron; onda = ondansetron; AC = anthracycline and cyclophosphamide; dex, dexamethasone
We modeled emesis-related outcomes and direct medical costs (from a third-party payer perspective within the context of the U.S. health-care system) over a total of four cycles of chemotherapy as patients receiving AC-based regimens usually undergo at least four cycles of AC.10 We performed all analyses using TreeAge Pro 2009 Suite (Decision Analysis; TreeAge Software, Williamstown, MA). The study was submitted to our institutional review board and was determined to be exempt from review.
Probability Data
Two-drug prophylactic regimens
We estimated the effectiveness of the 5-HT3 antagonists based on secondary analysis of the raw data from the randomized clinical trial (RCT) directly comparing onda and palo when used alone for prevention of emesis associated with MEC, including 90 breast cancer patients from the palo 0.25-mg arm and 82 from the onda 32-mg arm who received AC-based chemotherapy (Table 1).5 Effectiveness estimates for palo 0.25 mg were augmented by data on 117 breast cancer patients on AC-based chemotherapy participating in a multicenter RCT comparing palo with dolasetron (Table 1).4 We assumed that dex adds the same relative benefit to either first- or second-generation 5-HT3 antagonists and obtained the expected additional benefit of dex in preventing acute emesis based on the results of an RCT comparing a single-dose of granisetron in combination with dex vs granisetron given alone to patients undergoing MEC (Table 2).11 Since in the aforementioned study dex was only given on day 1 of chemotherapy, the estimated additional benefit of adding dex to a 5-HT3 inhibitor on the delayed period was obtained from another RCT; this study, conducted by the Italian Group for Antiemetic Research, compared dex alone, dex plus onda, or placebo on days 2−5 of MEC.12
MODEL PARAMETERS | BASE-CASE VALUES (RANGES) | DATA SOURCES |
---|---|---|
Probability of acute emesis control on cycle 1 of AC: | ||
Onda-based two-drug strategyc | 0.84 (0.74−0.93) | Gralla et al,a The Italian Group[5] and [11] |
Palo-based two-drug strategyc | 0.87 (0.81−0.94) | Eisenberg et al,a Gralla et al,a The Italian Group[4], [5] and [11] |
Onda-based three-drug strategyd | 0.88 (0.85−0.91) | Warr et al7 |
Palo-based three-drug strategyd | 0.96 (0.89−0.99) | Grote et al, Grunberg et al[40] and [41] |
Probability of delayed emesis control following control of acute emesis on cycle 1 of ACc: | ||
Onda-based two-drug strategyd | 0.75 (0.62–0.85) | The Italian Group12 |
Palo-based two-drug strategyc | 0.85 (0.78–0.91) | Eisenberg et al,a Gralla et al,a The Italian Group[4], [5] and [12] |
Onda-based three-drug strategyd | 0.86 (0.82–0.90) | Warr et al7 |
Palo-based three-drug strategyc | 0.96 (0.91–0.97) | Eisenberg et al,a Gralla et al,a Warr et al[4], [5] and [7] |
Probability of delayed emesis control following acute emesis on cycle 1 of ACc: | ||
Onda-based two-drug strategyc | 0.46 (0.31–0.62) | Gralla et al,a The Italian Group[5] and [12] |
Palo-based two-drug strategyc | 0.44 (0.27–0.59) | Eisenberg et al,a Gralla et al,a The Italian Group[4], [5] and [12] |
Onda-based three-drug strategyd | 0.44 (0.29–0.57) | Warr et al7 |
Palo-based three-drug strategyc | 0.51 (0.41–0.67) | Eisenberg et al,a Gralla et al,a Warr et al[4], [5] and [7] |
Relative probability of emesis control in subsequent cycles of ACc: | ||
Two-drug therapy | 0.987 (0.970–1.0) | Herrstedt et al14e |
Three-drug therapy | 1.013 (1.0–1.030) | Herrstedt et al14e |
Probability of hospitalization (among patients who develop emesis) per cycle of ACd: | ||
Onda-based regimens | 0.0035 (0.0001−0.019) | Data from Medstat MarketScan16 |
Palo-based regimens | 0.0017 (0.00004−0.0089) | Data from Medstat MarketScan, Haislip et al[16] and [19]b |
Probability of office visit use (among patients who develop emesis) per cycle of ACd: | ||
Onda-based regimens | 0.10 (0.07−0.14) | Data from Medstat MarketScan16 |
Palo-based regimens | 0.05 (0.03−0.07) | Data from Medstat MarketScan, Haislip et al[16] and [19]b |
Probability of rescue medicine utilization use (among patients who develop emesis) per cycle of ACd: | ||
Onda-based regimens | 0.61 (0.46−0.75) | Gralla et al5a |
Palo-based regimens | 0.56 (0.45−0.66) | Eisenberg et al, Gralla et al[4] and [5]a |
Utility weights for emesis per cycle of ACf: | ||
Acute and delayed emesis | 0.15 (0.10−0.20) | Sun et al20 |
Acute emesis and no delayed emesis | 0.76 (0.70−0.83) | Sun et al20 |
No acute emesis and delayed emesis | 0.20 (0.14−0.26) | Sun et al20 |
No acute and no delayed emesis | 0.92 (0.86−0.99) | Sun et al20 |
AC = anthracycline and cyclophosphamide; onda = ondansetron; palo = palonosetron.
a Included in the analysis was the subset of women with breast cancer receiving AC-based chemotherapy.b We obtained an estimate of emesis-related hospitalization and office visit utilization based on data from Medstat MarketScan, HPM subset (Medstat Group, Inc., Ann Arbor, MI) on 707 breast cancer patients who received the first cycle of AC-based chemotherapy from 1996 to 2002 and either were admitted to the hospital or had an office visit for treatment of vomiting or dehydration. Since palo was only introduced into the U.S. market in 2003, we assumed that all these breast cancer patients received onda-based antiemetic prophylaxis. As a result, we estimated the differential rate of health-care resource utilization based on Haislip et al's19 reported differential incidence of extreme events associated with chemotherapy-induced nausea and vomiting experienced by community-based breast cancer patients who received either onda or palo for emesis prophylaxis following the first cycle of chemotherapy.c Of note is that there are two different methods for applying the benefit of adding dex and/or aprepitant to a 5-HT3 antagonist: (1) rate of emesis with 5-HT3* relative risk of emesis by adding dex and/or aprepitant and (2) rate of emesis control with 5-HT3 * relative risk of emesis control by adding dex and/or aprepitant. These produce substantially different results, with the former method skewing the results toward the least effective 5-HT3 and the latter skewing it toward the most effective one. As a result, we estimated the probability of emesis by averaging the results obtained using the two different methods. Of note is that the ranges for these effectiveness estimates were obtained by applying the two different methods to the lower and upper bounds of the 95% confidence intervals derived from the clinical trials comparing the 5-HT3 antagonists when used alone.d Ranges were obtained by constructing 95% confidence intervals for observed proportions using the normal approximation to the binomial distribution.e Ranges are based on the minimum and maximum values observed in Herrstedt et al's14 clinical trial of multicycle chemotherapy.f Ranges are based on the estimate's actual 95% confidence intervals obtained from Sun et al's20 data.
Three-drug prophylactic regimens
We estimated the rate of acute emesis for the three-drug regimens based on data from published studies in which either onda or palo was given in combination with dex and aprepitant on day 1 of MEC (Table 2).[5], [7] and [13] Because aprepitant was either used in combination with dexamethasone or not used on days 2−3 in the trials of palo-based three-drug therapy, we estimated the benefit of adding aprepitant alone to palo on days 2−3 by assuming that the added benefit in the delayed period would be the same as the benefit added to onda. Specifically, we obtained information on the relative risk of delayed emesis control when aprepitant is added on days 2−3 from a large clinical trial of aprepitant combined with onda and dex in breast cancer patients receiving either A or AC chemotherapy (Table 2).7
Effectiveness of antiemetics over multiple cycles of chemotherapy
The estimates of changes in the probability of emesis control over multiple cycles of chemotherapy were obtained from a RCT conducted by Herrstedt et al14 of ondansetron-based two- and three-drug regimens for prevention of chemotherapy-induced nausea and vomiting among breast cancer patients undergoing multiple cycles of AC-based chemotherapy. We assumed that changes in emesis control over four cycles of AC for the palo-based two- and three-drug regimens were similar to the observed changes for the onda-based two- and three-drug strategies, respectively.14
Resource Utilization and Cost Data
The cost of antiemetic prophylaxis was based on the 2008 Medicare Part B reimbursement rates for pharmaceuticals, which reflects the price of ondansetron following its recent patent expiration (Table 3).15 The costs of prophylaxis failures were estimated as follows. In the majority of prophylaxis failures, the only cost is the cost of rescue medication. In such cases, we obtained costs by multiplying the individual doses used for rescue treatment of breast cancer patients on AC participating in the clinical trials comparing palo 0.25 mg with single doses of onda or dolasetron by their unit costs based on the 2008 Medicare Part B reimbursement rates.[5] and [15] For the few patients who are seen in the office for uncontrolled emesis, we obtained estimates of the risk of such emesis-related office visits based on the MarketScan Health Productivity Management (HPM) database from Thomson Reuters on 707 breast cancer patients who received their first cycle of AC-based chemotherapy between 1997 and 2002 (Table 2) and its costs from the 2008 Medicare Physician Fee Schedule Reimbursement for a level III office visit (CPT 99213).[16] and [17]
COST COMPONENT | 2008 U.S.$ (RANGES) | DATA SOURCE |
---|---|---|
Hospitalization | $5,237.00 ($3,921−$6,112)a | HCUP charge data18 Consumer Price Index42 Medicare cost-to-charge ratio43 |
Level III office visit (CPT 99213) | $60.30 ($19.96–$122.46)d | 2008 Medicare Physician Fee Schedule Reimbursement17 |
Prophylactic antiemetics | 2008 Medicare Part B reimbursement rates for pharmaceuticals15 | |
Onda-based two-drug regimen | $49.74 | |
Palo-based two-drug regimen | $207.20 | |
Onda-based three-drug regimen | $324.51 | |
Palo-based three-drug regimen | $482.46 | |
Rescue medicinesb | $35.25 ($21.66–$48.80)c | Eisenberg et al,4 Gralla et al,5 2008 Medicare Part B reimbursement rates for pharmaceuticals15 |
AC = anthracycline and cyclophosphamide; onda = ondansetron; palo = palonosetron; HCUP = Healthcare Cost and Utilization Project
a Charges were inflated to 2008 U.S. dollars using the Consumer Price Index (CPI) for medical care and adjusted to costs using Medicare cost-to-charge ratio. The ranges were based on estimates of the 95% confidence interval.b In the randomized clinical trial directly comparing ondansetron and palonosetron, propulsives accounted for 71% of the rescue medicines used, 5-hydroxytryptamine antagonists for 20%, glucocorticoids for 7%, and aminoalkyl ethers for 2%.5c Costs for rescue medication were obtained by multiplying all drug unit costs by the individual doses used for rescue treatment of breast cancer patients on AC participating in the clinical trials comparing palo 0.25 mg with single doses of onda or dolasetron.[5] and [15] The ranges were based on estimates of the 95% confidence interval.d Ranges were based on the Medicare physician fee schedule for levels I and VI office visits.
Finally, although hospitalization for emesis is extremely rare in this population, when it occurs, it is quite expensive. For completeness, we obtained estimates of the risk of emesis-related hospitalization from the same population of breast cancer patients from whom we obtained the estimate for the risk of emesis-related office visit, whereas hospital costs were obtained from Healthcare Cost and Utilization Project (HCUP) data on 2,342 breast cancer patients who were hospitalized with a primary or admitting diagnosis of vomiting or dehydration from 1997 to 2003 ([Table 2] and [Table 3]).[16] and [18]
Of note is that since palo was only introduced into the U.S. market in 2003, we anticipated the observed risk of emesis-related office visit and hospital admission obtained from MarketScan data during the period 1997−2002 reflected the risk associated with prophylaxis with onda. As a result, given that, when compared with onda, palo has also shown superiority in reducing the severity of emetic episodes when they occur, we estimated the differential rate of health-care resource utilization for palo and onda based on Haislip et al's reported differential incidence of extreme events associated with chemotherapy-induced nausea and vomiting (CINV) experienced by community-based breast cancer patients who received either palo or onda for emesis prophylaxis following the first cycle of chemotherapy (Table 2).[5] and [19]
Utility Data
We obtained the utility weights for acute and delayed emesis from a published study of preferences elicited from ovarian cancer patients undergoing chemotherapy using a modified visual analog scale (VAS) (Table 2).20 We equally applied these emesis-related utility weights to the initial 5-day period of chemotherapy (the standard duration of follow-up in clinical trials of prophylactic antiemetics) in all six prophylactic strategies of the decision tree. Furthermore, because the risk of CINV after 5 days of chemotherapy is usually so negligible as to be unmeasured in clinical trials of antiemetics, we assumed the utility weights for the remaining 16 days of each of the chemotherapy cycles to be the same as the weight associated with complete emesis control (ie, 0.92). We subsequently converted the resulting estimates of quality-adjusted life days into quality-adjusted life years (QALY).
Analysis
We used a stepwise method to calculate the incremental cost–effectiveness ratios of the different prophylactic therapy strategies, with the generic onda-based two-drug therapy (ie, the lowest cost strategy) as the base comparator (also known as the “anchor”).21 We adopted the benchmark range of U.S. $50,000−$100,000 per QALY, which has been commonly cited for oncology-related interventions as the threshold for acceptable cost–effectiveness, and examined the robustness of the results by performing one-way sensitivity analyses of plausible ranges for the model's key parameters based on the data sources used as well as probabilistic sensitivity analysis using Monte Carlo simulation.[21] and [22]
Results
The overall rate of emesis control (on days 1−5) among breast cancer patients following a cycle of AC-based chemotherapy was estimated to be 63% (range 46%−79%) for the onda-based two-drug therapy, 74% (range 66%−85%) for the palo-based two-drug therapy, 76% (range 75%−82%) for the onda-based three-drug therapy, and 92% (range 81%−96%) for the palo-based three-drug therapy. Based on these estimates, relative to the onda-based two-drug therapy, the incremental cost–effectiveness ratios (ICERs) for the palo-based regimens were $115,490/QALY for the two-drug strategy, $199,375/QALY for the two-drug regimen plus aprepitant after emesis, and $200,526/QALY for the three-drug strategy (Table 4). The onda-based two-drug combination plus aprepitant after the onset of emesis was eliminated through extended dominance as it has a greater ICER than the next more effective therapy, the palo-based two-drug treatment strategy (Table 4). The onda-based three-drug strategy was dominated by the palo-based two-drug combination plus aprepitant after the onset of emesis as the former strategy is both less effective and more expensive than the latter (Table 4).
STRATEGY | TOTAL COST (U.S.$) | INCREMENTAL COST (U.S.$) | EFFECTIVENESS (QALY) | INCREMENTAL EFFECTIVENESS (QALY) | INCREMENTAL COST–EFFECTIVENESS (U.S.$/QALY) |
---|---|---|---|---|---|
Onda-based two-drug therapy | $269 | — | 0.1989 | — | — |
Onda-based two-drug therapy with aprepitant after emesis | $635 | $366 | 0.2010 | 0.0021 | $174, 286 Eliminated through extended dominancea |
Palo-based two-drug therapy | $858 | $589 | 0.2040 | 0.0051 | $115,490c |
Palo-based two-drug therapy plus aprepitant after emesis | $1,177 | $319 | 0.2056 | 0.0016 | 199,375 |
Onda-based three-drug therapy | $1,336 | $159 | 0.205 | (0.0006) | Dominatedb |
Palo-based three-drug therapy | $1,939 | $603 | 0.2094 | 0.0044 | $200,526d |
QALY = quality-adjusted life year; AC = anthracycline and cyclophosphamide; ICER = incremental cost–effectiveness ratio; onda = ondansetron; palo = palonosetron
a Extended dominance occurs when one of the treatment alternatives has a greater ICER than the next more effective alternative.b One intervention is said to be dominated by another when it is both less effective and more expensive than the previous less costly alternative.c Because the onda-based two-drug combination plus aprepitant after the onset of emesis was eliminated through extended dominance, the palo-based two-drug therapy was compared with the onda-based two-drug therapy.d Because the onda-based three-drug combination was dominated by the palo-based two-drug combination plus aprepitant after the onset of emesis, the palo-based three-drug therapy was compared with the latter regimen.
In sensitivity analyses using the commonly accepted cost–effectiveness benchmark range of $50,000−$100,000/QALY, the results were sensitive to changes in the overall emesis control rates for the onda-based two-drug strategy. If the probability of overall emesis control for the onda-based two-drug strategy was as low as its estimated lower bound (46%), the ICER for the palo-based two-drug treatment alternative would drop to $53,892/QALY. The results were also sensitive to changes in the effectiveness for the palo-based two-drug regimen: When its overall control rate was as high as its estimated upper bound (86%), its ICER would be $71,472. In contrast, the results were not sensitive to variations in the probability of overall emesis control for the three-drug strategies, nor were they sensitive to changes in the relative probability of emesis control in subsequent cycles of AC for either the two- or three-drug strategies.
If the probability of emesis-related hospitalization was as high as the upper limit of its 95% confidence interval (CI), the ICER for the palo-based two-drug regimen would be $97,301/QALY. However, changes in the cost of an emesis-related admission (95% CI $3,921−$6,112) did not significantly alter the results, nor did variations in office visit and rescue medicine utilization and their associated costs. The results were also not sensitive to variations in the values for the utility weights throughout their 95% CIs. We performed a threshold analysis to explore the price per dose of palo that would result in an acceptable cost–effectiveness ratio under the $100,000/QALY benchmark and found that the ICER for the palo-based two-drug treatment alternative would only fall to a $100,000/QALY threshold when the cost of palo is decreased by 11%.
Figure 2 shows the cost–effectiveness acceptability curves for each strategy, with the onda-based two-drug therapy as the base comparator. These curves show the proportion of the 100,000 simulations in which the comparing antiemetic regimen was considered more cost-effective than the base comparator at different thresholds. Using the benchmark of U.S. $100,000/QALY, the palo-based two-drug strategy and the two-drug regimen plus aprepitant following the onset of emesis were shown to be cost-effective in 39% and 26% of the simulations with the onda-based standard therapy as the baseline, respectively, whereas the palo-based and onda-based three-drug strategies and the onda-based two-drug regimen with aprepitant after emesis were cost-effective in fewer than 10% of the simulations. Of note is that the slope of the acceptability curves for the palo-based two-drug strategies are steep when willingness to pay exceeds $50,000/QALY, indicating that the greater the threshold, the greater the increase in the level of confidence that these strategies could be cost-effective. For example, the probability that the palo-based two-drug strategy is more cost-effective than the onda-based two-drug strategy rises to 51% at a threshold value of $125,000/QALY and exceeds 60% at $150,000/QALY.
Figure 3 presents the scatterplot of the results of the probabilistic sensitivity analysis for the palo-based two-drug strategy. Nearly 96% of the simulations fell within the first quadrant of the chart (ie, on the upper right quadrant), which represents the scenario where the palo-based two-drug therapy is more costly but also more effective than the onda-based standard therapy. However, only 39% of the simulations fell below the $100,000/QALY dashed threshold line, which represents the scenario where the palo-based two-drug strategy is more cost-effective than the onda-based standard therapy at the $100,000/QALY benchmark.
Discussion
Our estimates of emesis-related costs and outcomes following four cycles of AC-based chemotherapy in women with breast cancer indicate that at current antiemetic prices and utilities placed on emesis, the additional costs of palo and aprepitant are not warranted at the $100,000/QALY threshold. In probabilistic sensitivity analysis, the palo-based two-drug strategy and the two-drug regimen plus aprepitant following the onset of emesis were shown to be cost-effective at the $100,000/QALY threshold in only 39% and 26% of the simulations, respectively. The model was sensitive to changes in the values of antiemetic effectiveness for the two-drug regimens and the risk of emesis-related hospitalization.
In threshold analysis, the two-drug palo-based regimen was cost-effective at the $100,000/QALY benchmark when the cost of palo is decreased by 11%. Because the use of the $100,000/QALY threshold is uncommon in clinical practice, the cost-effectiveness of the palo-based two-drug strategy (estimated at $115,490/QALY in our study) compares favorably with other commonly used supportive care measures for women with breast cancer. Such measures include primary prophylaxis with granulocyte colony-stimulating factor in women undergoing chemotherapy with moderate to high myelosuppressive risk (ICER of $116,000/QALY, or $125,948/QALY in 2008 U.S. dollars) and the use of bisphosphonates for the prevention of skeletal complications in breast cancer patients with lytic bone metastases (ICER ranging from $108,200/QALY with chemotherapy as systemic therapy to $305,300 in conjunction with hormonal systemic therapy, or $166,381/QALY to $469,466/QALY in 2008 U.S. dollars, respectively).[23] and [24] Both interventions are considered recommended standards of supportive care for patients with breast cancer and are widely used in breast oncology practices.[25] and [26]
Decision-analytic models, such as the Markov model presented in our study, aim to reflect the reality of clinical practice in a simplified way. Therefore, modelers often need to make decisions regarding the study time frame and model parameters based on the best use of available data. In our study, we obtained estimates for the probability of chemotherapy-induced emesis from studies in which the standard duration of follow-up is 5 days. By so doing, we may have underestimated the cost-effectiveness for the palo-based and aprepitant-based regimens. Although the risk of CINV after 5 days of chemotherapy is usually negligible, anticipation of vomiting may affect a patient's quality of life throughout the cycle of chemotherapy.
In addition, our estimates of costs, which were mostly obtained from Medicare, may differ from those of other third-party payers. However, Medicare is among the largest payers for breast cancer care as 42% of the women diagnosed with cancer in the United States are older than 64 years, and many private organizations set their own reimbursement rates based on the Medicare schedule. Therefore, we believe that Medicare reimbursement data provide a suitable estimate for emesis-related medical costs for all breast cancer patients in the United States.[27] and [28]
The present results should solely be interpreted in light of the cost–effectiveness benchmark of $50,000−$100,000/QALY, which has been frequently used in the context of the U.S. health-care system.[22] and [29] Such a benchmark, however, is a historic, precedent-based threshold set by the cost of caring for patients on dialysis, which was estimated at $50,000/QALY in 1982 ($74,000−$95,000 in 1997 U.S. dollars).[30] and [31] Given the arbitrariness of such a threshold, it has been suggested that the current willingness to pay for medical interventions in the United States probably exceeds $100,000/QALY, with values as high as $300,000/QALY being cited in some oncology publications.[22], [29], [31], [32], [33] and [34] In support of that argument is the public and policy makers' strong negative reaction to the National Institutes of Health Consensus Panel not recommending mammography screening for women aged 40−49 years, a procedure reported to provide an ICER of $105,000 per life-year gained.[35] and [36] As a result, if willingness to pay goes beyond $100,000/QALY, the alternative of adding aprepitant to palo plus dex may also be deemed attractive as the slope of its acceptability curve becomes substantially steep when the willingness to pay for a QALY exceeds $125,000 (Figure 2), suggesting that its marginal gain may exceed its marginal costs at higher thresholds.
In addition, it is worth noting that the present analysis has been conducted from the perspective of a third-party payer within the context of the U.S. health-care system. The large difference in the acquisition cost of palo-based and onda-based therapy observed in the United States is mostly driven by the differential stage of product life cycles for palo and onda. Although at the time of this study palo was still under patent protection, generic onda had entered the U.S. market prior to our study. The large price discrepancy between brand and generic drugs explains the difference in drug costs in this U.S.-based analysis. As such, our results may not reflect the situation in countries with a widely different cost structure, in which the acquisition cost of palo may be substantially lower. When that is the case, the cost–effectiveness profile of the palo-based prophylactic therapy may be deemed substantially more favorable than the profile presented here. Similarly, we anticipate finding a more attractive cost–effectiveness profile for the palo-based therapies as palo reaches the end of its product life cycle in the U.S. market.37 Also of note is that the cost–effectiveness of the palo-based therapy may greatly differ when different perspectives (other than the third-party payer's perspective) are adopted.
Our study, however, has several limitations. First, the utility scores used in our model were derived with a VAS instrument, which does not incorporate patients' preferences under uncertainty. Nevertheless, the VAS approach has been shown to provide utility scores for nausea and vomiting with more variability than scores derived using other methods such as the Standard Gamble (personal communication, Grunberg SM et al, CALGB study 309801). Notwithstanding that, it remains unclear which method gives utility scores for transient health states, such as CINV, with the greatest validity.
Also of note is that due to a lack of information on emesis-related utilities among breast cancer patients in the literature, we used utilities elicited from patients with ovarian cancer. To the best of our knowledge, the utilities in Sun et al20 were the only ones available in the literature that were elicited from a homogeneous population of cancer patients (ie, solely patients with ovarian cancer) and were based on a wide range of health states combining the presence and absence of emesis during either the acute or the delayed period. In addition, the participants in the Sun et al study were treated with carboplatin, which, like the regimen used in our model, is classified as moderately emetogenic in established antiemetic guidelines.[8], [9] and [38] It is also important to emphasize that the population in that study, like our study's population, was composed exclusively of women, who are known to be at increased risk for developing CINV.39
Second, in the absence of clinical trial data, we assumed conservatively that dex and aprepitant add the same relative benefit to both onda and palo. This assumption results in an imperfect estimate of cost–effectiveness. As such, we may have overestimated or underestimated the cost–effectiveness of palo as dex and aprepitant may potentially add less value to the intrinsically more active 5-HT3 antagonist or uniquely complementary mechanisms of action could contribute to even greater activity with the palo-based therapy. However, our study's estimate of the relative effectiveness of the palo-based two-drug prophylactic therapy versus the onda-based two-drug therapy for preventing delayed emesis is consistent with that reported in a recently published clinical trial comparing palo and granisetron when both drugs are combined with dex following chemotherapy with either AC or cisplatin (1.18 vs 1.17, respectively).6
Third, our study did not include the outcomes associated with the adverse effects of antiemetics, and by so doing, we may have underestimated the costs associated with antiemetic prophylaxis. However, the incidence and duration of treatment-related adverse events occurring in the two RCTs comparing palo with either onda or dolasetron were mild and similar across treatment cohorts.[4] and [5]
Fourth, we assumed that changes in emesis control in subsequent cycles of AC for the palo-based regimens were the same as for the onda-based therapy. By so doing, we may have underestimated the cost–effectiveness of palo as the superiority of the more active 5-HT3 antagonist could be maintained in the subsequent cycles of chemotherapy (or even increased, as seen in the aprepitant-based arm of Herrstedt et al's14 study). As a result, if future prospective trials of palo-based antiemetic prophylaxis confirm its superiority in maintaining antiemetic efficacy over multiple cycles of AC, the cost–effectiveness profiles for the palo-based strategies may be more favorable than the profiles presented herein.
Last, the incremental gains in QALY observed in cost–utility analysis of interventions associated with transitory and non-life-threatening health states, such as the antiemetic regimens analyzed in our study, tend to render small denominators to be used in the incremental cost–effectiveness ratios. The issue of small denominators has led some researchers to question whether the current methodology of cost–effectiveness analysis is appropriate to determine the cost–effectiveness of treatments for terminal or supportive care.32 However, despite this shortcoming, these types of analysis benefit from having a wider scope as they allow comparisons over different types of health interventions across various diseases. In addition, by incorporating patients' utility levels over different health states (instead of merely looking into cost per additional patient controlled), cost–utility analysis makes explicit the impact of the target population's preferences for the different outcomes. Of importance is that both the Panel on Cost–Effectiveness in Health and Medicine and the Institute of Medicine (IOM) Committee on Regulatory Cost–Effectiveness Analysis recommend the use of QALY as the preferred outcome measure for economic evaluation of health-care interventions.
Conclusion
Although our base-case analysis suggests that, from a third-party payer perspective within the context of the U.S. health-care system, the cost–utility of the palo-based two-drug prophylactic therapy for breast cancer patients receiving four cycles of AC-based chemotherapy exceeds the $50,000–$100,000/QALY threshold, it is comparable to other commonly used supportive care interventions for women with breast cancer. In sensitivity analyses, such a strategy was associated with a 39% chance of being cost-effective at the $100,000/QALY threshold, and the model was sensitive to changes in the values of antiemetic effectiveness and of the probability of emesis-related hospitalization. In threshold analysis, the combination of palo and dex was shown to become cost-effective (at the $100,000/QALY benchmark) when the cost of palo is decreased by 11%. As a result, future research incorporating the price structure of all antiemetics following the recent expiration of onda's patent is needed.
References1
1 S.M. Grunberg, D. Osoba and P.J. Hesketh et al., Evaluation of new antiemetic agents and definition of antineoplastic agent emetogenicity—an update, Support Care Cancer 13 (2005), pp. 80–84 [15599601]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (68)
2 C.M. Booth, M. Clemons and G. Dranitsaris et al., Chemotherapy-induced nausea and vomiting in breast cancer patients: a prospective observational study, J Support Oncol 5 (2007), pp. 374–380 [17944146]. View Record in Scopus | Cited By in Scopus (10)
3 M. de Boer-Dennert, R. de Wit and P.I. Schmitz et al., Patient perceptions of the side-effects of chemotherapy: the influence of 5HT3 antagonists, Br J Cancer 76 (1997), pp. 1055–1061 [9376266]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (128)
4 P. Eisenberg, J. Figueroa-Vadillo, R. Zamora et al. and 99-04 Palonosetron Study Group, Improved prevention of moderately emetogenic chemotherapy-induced nausea and vomiting with palonosetron, a pharmacologically novel 5-HT3 receptor antagonist: results of a phase III, single-dose trial versus dolasetron, Cancer 98 (2003), pp. 2473–2482 [14635083]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (171)
5 R. Gralla, M. Lichinitser and S. Van Der Vegt et al., Palonosetron improves prevention of chemotherapy-induced nausea and vomiting following moderately emetogenic chemotherapy: results of a double-blind randomized phase III trial comparing single doses of palonosetron with ondansetron, Ann Oncol 14 (2003), pp. 1570–1577 [14504060]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (177)
6 M. Saito, K. Aogi and I. Sekine et al., Palonosetron plus dexamethasone versus granisetron plus dexamethasone for prevention of nausea and vomiting during chemotherapy: a double-blind, double-dummy, randomised, comparative phase III trial, Lancet Oncol 10 (2009), pp. 115–124 [19135415]. Article | | View Record in Scopus | Cited By in Scopus (43)
7 D.G. Warr, P.J. Hesketh and R.J. Gralla et al., Efficacy and tolerability of aprepitant for the prevention of chemotherapy-induced nausea and vomiting in patients with breast cancer after moderately emetogenic chemotherapy, J Clin Oncol 23 (2005), pp. 2822–2830 [15837996]. View Record in Scopus | Cited By in Scopus (139)
8 American Society of Clinical Oncology, M.G. Kris, P.J. Hesketh and M.R. Somerfield et al., American Society of Clinical Oncology guideline for antiemetics in oncology: update 2006, J Clin Oncol 24 (2006), pp. 2932–2947 [16717289]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (311)
9 D.S. Ettinger, D.K. Armstrong and S. Barbour et al., National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology—Antiemesis, version 2.2010 http://www.nccn.org/professionals/physician_gls/PDF/antiemesis.pdf Accessed September 20, 2010.
10 R.W. Carlson and B. McCormick, Update: NCCN breast cancer clinical practice guidelines, J Natl Compr Cancer Netw 3 (suppl 1) (2005), pp. S7–S11 [16280118].
11 The Italian Group for Antiemetic Research, Dexamethasone, granisetron, or both for the prevention of nausea and vomiting during chemotherapy for cancer, N Engl J Med 332 (1995), pp. 1–5 [7990859].
12 The Italian Group for Antiemetic Research, Dexamethasone alone or in combination with ondansetron for the prevention of delayed nausea and vomiting induced by chemotherapy, N Engl J Med 342 (2000), pp. 1554–1559 [10824073].
13 S.M. Grunberg, M. Dugan and H. Muss et al., Effectiveness of a single-day three-drug regimen of dexamethasone, palonosetron, and aprepitant for the prevention of acute and delayed nausea and vomiting caused by moderately emetogenic chemotherapy, Support Care Cancer 17 (2009), pp. 589–594 [19037667]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (14)
14 J. Herrstedt, H.B. Muss and D.G. Warr et al., Efficacy and tolerability of aprepitant for the prevention of chemotherapy-induced nausea and emesis over multiple cycles of moderately emetogenic chemotherapy, Cancer 104 (2005), pp. 1548–1555 [16104039]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (43)
15 Centers for Medicare and Medicaid Services, Medicare Part B Drug Average Sales Price: 2008 ASP Drug Pricing Files http://www.cms.hhs.gov/apps/ama/license.asp?file=/McrPartBDrugAvgSalesPrice/downloads/July2008ASPPricingFilebyHCPCS.zip Accessed July 18, 2008.
16 Thomson. Medstat, 1997–2002 MarketScan Health and Productivity Management Database User Guide and Data Dictionary, Thomson Medstat, Ann Arbor, MI (2003).
17 Centers for Medicare and Medicaid Services, National Physician Fee Schedule and Relative Value: 2008 Physician Fee Schedule National Payment Amount File http://www.cms.hhs.gov/PFSlookup/02_PFSSearch.asp Accessed July 18, 2008.
18 National Inpatient Sample (NIS), NIS description of data elements, Healthcare Cost and Utilization Project (HCUP) databases, Agency for Healthcare Research and Quality, Rockville, MD (2004) http://www.hcup-us.ahrq.gov/nisoverview.jsp#Data Accessed May 16, 2010.
19 S. Haislip, J. Gilmore, W.H. Lenz, T. Gondesen and B. Feinberg, Theory in practice: improving patient outcomes and practice efficiency with a simple change in 5-HT3 receptor antagonist for preventing chemotherapy-induced nausea and vomiting (CINV) In: Third Annual Meeting of the Hematology/Oncology Pharmacy Association; Abstract #PR6. June 14–16, 2007; Denver, Colorado http://www.hoparx.org/documents/2007programbook.pdf Accessed November 2, 2010.
20 C.C. Sun, D.C. Bodurka and C.B. Weaver et al., Rankings and symptom assessments of side effects from chemotherapy: insights from experienced patients with ovarian cancer, Support Care Cancer 13 (2005), pp. 219–227 [15538640]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (53)
21 M.F. Drummond, M.J. Sculpher, G.W. Torrance, B.J. O'Brien and G.L. Stoddart, Methods for the Economic Evaluation of Health Care Programmes (3rd ed.), Oxford University Press, New York (2005).
22 J. Hayman, J. Weeks and P. Mauch, Economic analyses in health care: an introduction to the methodology with an emphasis on radiation therapy, Int J Radiat Oncol Biol Phys 35 (1996), pp. 827–841 [8690653]. Article | | View Record in Scopus | Cited By in Scopus (33)
23 B.E. Hillner, J.C. Weeks, C.E. Desch and T.J. Smith, Pamidronate in prevention of bone complications in metastatic breast cancer: a cost–effectiveness analysis, J Clin Oncol 18 (2000), pp. 72–79 [10623695]. View Record in Scopus | Cited By in Scopus (90)
24 S.D. Ramsey, Z. Liu and R. Boer et al., Cost–effectiveness of primary versus secondary prophylaxis with pegfilgrastim in women with early-stage breast cancer receiving chemotherapy, Value Health 11 (2008), pp. 172–179 [18673353].
25 B.E. Hillner, J.N. Ingle, R.T. Chlebowski et al. and American Society of Clinical Oncology, American Society of Clinical Oncology 2003 update on the role of bisphosphonates and bone health issues in women with breast cancer, J Clin Oncol 21 (2003), pp. 4042–4057 [12963702]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (533)
26 T.J. Smith, J. Khatcheressian and G.H. Lyman et al., 2006 update of recommendations for the use of white blood cell growth factors: an evidence-based clinical practice guideline, J Clin Oncol 24 (2006), pp. 3187–3205 [16682719]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (485)
27 National Cancer Institute, Surveillance Epidemiology and End Results: SEER Stat Fact Sheets: Breast http://seer.cancer.gov/statfacts/html/breast.html Accessed May 16, 2010.
28 J.W. Tumeh, S.G. Moore, R. Shapiro and C.R. Flowers, Practical approach for using Medicare data to estimate costs for cost–effectiveness analysis, Expert Rev Pharmacoecon Outcomes Res 5 (2005), pp. 153–162 [19807571]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
29 P.A. Ubel, R.A. Hirth, M.E. Chernew and A.M. Fendrick, What is the price of life and why doesn't it increase at the rate of inflation?, Arch Intern Med 163 (2003), pp. 1637–1641 [12885677]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (225)
30 J.C. Hornberger, D.A. Redelmeier and J. Petersen, Variability among methods to assess patients' well-being and consequent effect on a cost–effectiveness analysis, J Clin Epidemiol 45 (1992), pp. 505–512 [1588356]. Article | | View Record in Scopus | Cited By in Scopus (138)
31 R.A. Hirth, M.E. Chernew, E. Miller, A.M. Fendrick and W.G. Weissert, Willingness to pay for a quality-adjusted life year: in search of a standard, Med Decis Making 20 (2000), pp. 332–342 [10929856]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (291)
32 Y.C. Shih and M.T. Halpern, Economic evaluations of medical care interventions for cancer patients: how, why, and what does it mean?, CA Cancer J Clin 58 (2008), pp. 231–244 [18596196]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (18)
33 E. Nadler, B. Eckert and P.J. Neumann, Do oncologists believe new cancer drugs offer good value?, Oncologist 11 (2006), pp. 90–95 [16476830]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (50)
34 R.S. Braithwaite, D.O. Meltzer, J.T. King Jr, D. Leslie and M.S. Roberts, What does the value of modern medicine say about the $50,000 per quality-adjusted life-year decision rule?, Med Care 46 (2008), pp. 349–356 [18362813]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
35 National Institutes of Health Consensus Development Panel, 1997 Consensus Statement: Breast Cancer Screening for Women Ages 40–49 http://consensus.nih.gov/1997/1997BreastCancerScreening103html.htm Accessed October 13, 2007.
36 P. Salzmann, K. Kerlikowske and K. Phillips, Cost–effectiveness of extending screening mammography guidelines to include women 40 to 49 years of age, Ann Intern Med 127 (1997), pp. 955–965 [9412300]. View Record in Scopus | Cited By in Scopus (169)
37 Y.C. Shih, S. Han and S.B. Cantor, Impact of generic drug entry on cost–effectiveness analysis, Med Decis Making 25 (2005), pp. 71–80 [15673583]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
38 F. Roila, P.J. Hesketh, J. Herrstedt and Antiemetic Subcommittee of the Multinational Association of Supportive Care in Cancer, Prevention of chemotherapy- and radiotherapy-induced emesis: results of the 2004 Perugia International Antiemetic Consensus Conference, Ann Oncol 17 (2006), pp. 20–28 [16314401]. View Record in Scopus | Cited By in Scopus (90)
39 S.M. Grunberg and A. Ireland, Epidemiology of chemotherapy-induced nausea and vomiting, Adv Studies Nurs 3 (1) (2005), pp. 9–15 http://www.jhasin.com/files/articlefiles/pdf/XASIN_3_1_p9_15.pdf Accessed September 16, 2010.
40 T. Grote, J. Hajdenberg, A. Cartmell, S. Ferguson, A. Ginkel and V. Charu, Combination therapy for chemotherapy-induced nausea and vomiting in patients receiving moderately emetogenic chemotherapy: palonosetron, dexamethasone, and aprepitant, J Support Oncol 4 (2006), pp. 403–408 [17004515]. View Record in Scopus | Cited By in Scopus (38)
41 S.M. Grunberg, M. Dugan, H.B. Muss, M. Wood, S. Burdette-Radoux and T. Weisberg, Efficacy of a 1-day 3-drug antiemetic regimen for prevention of acute and delayed nausea and vomiting induced by moderately emetogenic chemotherapy, J Clin Oncol 25 (18S) (2007), p. 9111.
42 U. S. Department of Labor. Bureau of Labor Statistics. Consumer Price Index http://www.bls.gov/cpi/home.htm Accessed May 16, 2010.
43 Department of Health and Human Services. Centers for Medicare & Medicaid Services, Medicare Program; Proposed Changes to the Hospital Inpatient Prospective Payment Systems and Fiscal Year 2008 Rates CMS-1533-P, pp 1070–1073 http://www.cms.hhs.gov/AcuteInpatientPPS/downloads/CMS-1533-P.pdf Accessed May 16, 2010.
Conflicts of interest: Dr. Sun discloses that her husband was an employee of MGI Pharma, Inc., at the time this article was being written. Dr. Gralla discloses that he is a consultant for MGI Pharma, Inc., GlaxoSmithKline, Sanofi-aventis, and Merck; he also receives honoraria from MGI Pharma, Inc., and Merck and research support from Sanofi-aventis. Dr. Grunberg discloses that he is a consultant for MGI Pharma, Inc.
Correspondence to: Elenir B. C. Avritscher, MD, PhD, MBA/MHA, Section of Health Services Research, Department of Biostatistics and Applied Mathematics, The University of Texas M. D. Anderson Cancer Center, 1400 Pressler Street, Unit 1411, Houston, TX 77230; telephone: (713) 563-8920; fax: (713) 563-4243
The Journal of Supportive Oncology
Volume 8, Issue 6, November-December 2010, Pages 242-25