Recommendations on the Use of Ultrasound Guidance for Adult Lumbar Puncture: A Position Statement of the Society of Hospital Medicine

Article Type
Changed
Sun, 10/13/2019 - 21:16

Approximately 400,000 lumbar punctures (LPs) are performed in the United States annually for either diagnostic workup or therapeutic relief.1 Lumbar punctures are increasingly being performed in the United States, with an estimated 97,000 LPs performed on Medicare fee-for-service beneficiaries in 2011 alone, which is an increase of approximately 4,000 LPs in the same population from 1991.2 Approximately 273,612 LPs were performed on hospitalized patients in the United States in 2010,1 and the inpatient hospital setting is the most common site for LPs.2,3

Many LPs are referred to radiologists who have access to imaging guidance to aid with needle insertion.2 However, referrals to radiology delay performance of LPs, and delayed diagnosis of acute bacterial meningitis, the most common yet serious condition for which LPs are performed, is associated with increased morbidity and mortality.4-8 Furthermore, although initiating empiric antibiotic treatment for suspected acute bacterial meningitis is recommended in some cases, doing so routinely can cause false-negative cerebrospinal fluid (CSF) culture results, complicating decisions about de-escalation and duration of antibiotics that could have been safely avoided by promptly performing an LP.9

Delaying the performance of LP has been associated with increased mortality.10 Demonstration of proficiency in performance of lumbar puncture is considered a core competency for hospitalists,11 and with the increasing availability of point-of-care ultrasound, hospitalists can use ultrasound to guide performance of LPs at the bedside.12 However, 30% of patients requiring LP in emergency departments have difficult-to-palpate lumbar spine landmarks,13 and lumbar puncture performed based on palpation of landmarks alone has been reported to fail or be traumatic in 28% of patients.14 Use of ultrasound guidance for lumbar puncture has been shown in randomized controlled trials to improve procedural success rates, while reducing the time to successful LP, needle passes, patient pain scores, and risk of a traumatic LP.15-17

The purpose of this position statement is to review the literature and present consensus-based recommendations on the performance of ultrasound-guided LP in adult patients. This position statement does not mandate that hospitalists use ultrasound guidance for LP, nor does it establish ultrasound guidance as the standard of care for LP. Similar to previously published Society of Hospital Medicine (SHM) position statements,12,18,19 this document presents recommendations with supporting evidence for the clinical outcomes, techniques, and training for using ultrasound guidance for LP. A manuscript describing the technique of ultrasound guidance for LPs has been previously published by some of the authors of this position statement.20

 

 

METHODS

Detailed methods are described in Appendix 1. The SHM Point-of-care Ultrasound (POCUS) Task Force was assembled to carry out this guideline development project under the direction of the SHM Board of Directors, Director of Education, and Education Committee. All expert panel members were physicians or advanced practice providers with expertise in POCUS. Expert panel members were divided into working group members, external peer reviewers, and a methodologist. All Task Force members were required to disclose any potential conflicts of interests (Appendix 2). The literature search was conducted in two independent phases. The first phase included literature searches conducted by the six working group members themselves. Key clinical questions and draft recommendations were then prepared. A systematic literature search was conducted by a medical librarian based on the findings of the initial literature search and draft recommendations. The Medline, Embase, CINAHL, and Cochrane medical databases were searched from 1975 to December 2015 initially. Google Scholar was also searched without limiters. Updated searches were conducted in November 2016, January 2018, and October 2018. The search strings are included in Appendix 3. All article abstracts were first screened for relevance by at least two members of the working group. Full-text versions of screened articles were reviewed, and articles on the use of ultrasound to guide LP were selected. In addition, the following article types were excluded: non-English language, nonhuman, age <18 years, meeting abstracts, meeting posters, narrative reviews, case reports, letters, and editorials. Moreover, studies focusing on the use of ultrasound guidance for spinal nerve root injections, regional anesthesia, and assessment of lumbar spine anatomy alone were excluded. All relevant systematic reviews, meta-analyses, randomized controlled trials, and observational studies of ultrasound-guided LP were screened and selected. Final article selection was based on working group consensus, and the selected literature was incorporated into the draft recommendations.

The Research and Development (RAND) Appropriateness Method that required panel judgment and consensus was used.21 The 27 voting members of the SHM POCUS Task Force reviewed and voted on the draft recommendations considering the following five transforming factors: (1) Problem priority and importance, (2) Level of quality of evidence, (3) Benefit/harm balance, (4) Benefit/burden balance, and (5) Certainty/concerns about PEAF (Preferences/Equity/Acceptability/Feasibility). Panel members participated in two rounds of electronic voting using an internet-based electronic data collection tool (REDCap™) in February 2018 and April 2018 (Appendix 4). Voting on appropriateness was conducted using a 9-point Likert scale. The three zones of the 9-point Likert scale were inappropriate (1-3 points), uncertain (4-6 points), and appropriate (7-9 points). The degree of consensus was assessed using the RAND algorithm (Appendix Figure 1 and Table 1). Establishing a recommendation required at least 70% agreement that a recommendation was “appropriate.” A strong recommendation required 80% of the votes within one integer of the median, following the RAND rules. Disagreement was defined as >30% of panelists voting outside of the zone of the median.

Recommendations were classified as strong or weak/conditional based on preset rules defining the panel’s level of consensus, which determined the wording of each recommendation (Table 2). The revised consensus-based recommendations underwent internal and external reviews by POCUS experts from different subspecialties. The final review of this position statement was performed by members of the SHM POCUS Task Force, SHM Education Committee, and SHM Executive Committee. The SHM Executive Committee endorsed this position statement in June 2018 before submission to the Journal of Hospital Medicine.

 

 

RESULTS

Literature Search

A total of 4,389 references were pooled from four different sources: a search by a certified medical librarian in December 2015 (3,212 citations) that was updated in November 2016 (380 citations), January 2018 (282 citations), and October 2018 (274 citations); working group members’ personal bibliographies and searches (31 citations); and a search focusing on ultrasound-guided LP training (210 citations). A total of 232 full-text articles were reviewed, and the final selection included 77 articles that were abstracted into a data table and incorporated into the draft recommendations. Details of the literature search strategy are presented in Appendix 3.

RECOMMENDATIONS

Four domains (clinical outcomes, technique, training, and knowledge gaps) with 16 draft recommendations were generated based on a review of the literature. Selected references were abstracted and assigned to each draft recommendation. Rationales for each recommendation were drafted citing supporting evidence. After two rounds of panel voting, five recommendations did not achieve agreement based on the RAND rules, one recommendation was combined with another recommendation during peer review, and 10 statements received final approval. The degree of consensus based on the median score and the dispersion of voting around the median are shown in Appendix 5. Nine statements were approved as strong recommendations, and one was approved as a conditional recommendation. Therefore, the final recommendation count was 10. The strength of the recommendation and degree of consensus for each recommendation are summarized in Table 1.

Terminology

LP is a procedure in which a spinal needle is introduced into the subarachnoid space for the purpose of collecting CSF for diagnostic evaluation and/or therapeutic relief.

Throughout this document, the phrases “ultrasound-guided” and “ultrasound guidance” refer to the use of ultrasound to mark a needle insertion site immediately before performing the procedure. This is also known as static ultrasound guidance. Real-time or dynamic ultrasound guidance refers to direct visualization of the needle tip as it traverses through the skin and soft tissues to reach the ligamentum flavum. Any reference to real-time ultrasound guidance is explicitly stated.

Clinical outcomes

1) When ultrasound equipment is available, along with providers who are appropriately trained to use it, we recommend that ultrasound guidance should be used for site selection of LPs to reduce the number of needle insertion attempts and needle redirections and increase the overall procedure success rates, especially in patients who are obese or have difficult-to-palpate landmarks.

Rationale. LPs have historically been performed by selecting a needle insertion site based on palpation of anatomical landmarks. However, an estimated 30% of patients requiring LP in emergency departments have lumbar spine landmarks that are difficult to palpate, most commonly due to obesity.13 Furthermore, lumbar puncture performed based on palpation of landmarks alone has been reported to fail in 28% of patients.14

Ultrasound can be used at the bedside to elucidate the lumbar spine anatomy to guide performance of LP or epidural catheterization. Since the early 2000s, randomized studies comparing the use of ultrasound guidance (ultrasound-guided) versus anatomical landmarks (landmark-guided) to map the lumbar spine for epidural catheterization have emerged. It is important to recognize that the exact same ultrasound technique is used for site marking of LP, epidural catheterization, and spinal anesthesia—the key difference is how deep the needle tip is inserted. Therefore, data from these three ultrasound-guided procedures are often pooled. Currently, at least 33 randomized controlled studies comparing ultrasound-guided vs landmark-guided site selection for LP, epidural catheterization, or spinal anesthesia have been published.22-49 We present three meta-analyses below that pooled data primarily from randomized controlled studies comparing ultrasound-guided vs landmark-guided site selection for LP or spinal anesthesia.

In 2013, Shaikh et al. published the first meta-analysis with 14 randomized controlled studies comparing ultrasound-guided vs landmark-guided site selection for LP (n = 5) or epidural catheterization (n = 9). The pooled data showed that use of ultrasound guidance decreased the proportion of failed procedures (risk ratio 0.21, 95% CI 0.10-0.43) with an absolute risk reduction of 6.3% (95% CI 4.1%-8.4%) and a number needed to treat of 16 (95% CI 12-25) to prevent one failed procedure. In addition, the use of ultrasound reduced the mean number of attempts by 0.44 (95% CI 0.24-0.64) and reduced the mean number of needle redirections by 1.00 (95% CI 0.75-1.24). The reduction in risk of a failed procedure was similar for LPs (risk ratio 0.19 [95% CI 0.07-0.56]) and epidural catheterizations (risk ratio 0.23 [95% CI 0.09-0.60]).16

A similar meta-analysis published by Perlas et al. in 2016 included a total of 31 studies, both randomized controlled and cohort studies, evaluating the use of ultrasound guidance for LP, spinal anesthesia, and epidural catheterization.50 The goal of this systematic review and meta-analysis was to establish clinical practice recommendations. The authors concluded (1) the data consistently suggest that ultrasound is more accurate than palpation for lumbar interspace identification, (2) ultrasound allows accurate measurement of the needle insertion depth to reach the epidural space with a mean difference of <3 mm compared with the actual needle insertion depth, and (3) ultrasound increases the efficacy of lumbar epidural or spinal anesthesia by decreasing the mean number of needle passes for success by 0.75 (95% CI 0.44-1.07) and reducing the risk of a failed procedure (risk ratio 0.51 [95% CI 0.32-0.80]), both in patients with normal surface anatomy and in those with technically difficult surface anatomy due to obesity, scoliosis, or previous spine surgery.

Compared to the two earlier meta-analyses that included studies of both LP and spinal anesthesia procedures, the meta-analysis conducted by Gottlieb et al. in 2018 pooled data from 12 randomized controlled studies of ultrasound guidance for LPs only. For the primary outcome, pooled data from both adult and pediatric studies demonstrated higher procedural success rates with ultrasound-guided vs landmark-guided LPs (90% vs 81%) with an odds ratio of 2.1 (95% CI 0.66-7.44) in favor of ultrasound; however, there were no statistically significant differences when the adult and pediatric subgroups were analyzed separately, probably due to underpowering. For the secondary outcomes, data from the adult subgroup showed that use of ultrasound guidance was associated with fewer traumatic LPs (OR 0.28, 95% CI 0.14-0.59), shorter time to procedural success (adjusted mean difference –3.03 minutes, 95% CI –3.54 to –2.52), fewer number of needle passes (adjusted mean difference –0.81 passes, 95% CI –1.57 to –0.05), and lower patient pain scores (adjusted mean difference –2.53, 95% CI –3.89 to –1.17).

At least 12 randomized controlled studies have been published comparing the use of ultrasound guidance vs landmarks for the performance of LP or spinal anesthesia in adult patients, which were not included in the abovementioned meta-analyses. These individual studies demonstrated similar benefits of using ultrasound guidance: reduced needle insertion attempts, reduced needle redirections, and increased overall procedural success rates.17,31,37,40,41,43-49

It is important to recognize that four randomized controlled studies did not demonstrate any benefits of ultrasound guidance on the number of attempts or procedural success rates,23,33,41,51 and three of these studies were included in the abovementioned meta-analyses.23,33,51 Limitations of these negative studies include potential selection bias, inadequate sample sizes, and varying levels of operator skills in procedures, ultrasound guidance, or both. One study included emergency medicine residents as operators with varying degrees of ultrasound skills, and more importantly, patient enrollment occurred by convenience sampling, which may have introduced selection bias. Furthermore, most of the patients were not obese (median BMI of 27 kg/m2), and it is unclear why 10 years lapsed from data collection until publication.33 Another study with three experienced anesthesiologists as operators performing spinal anesthesia enrolled only patients who were not obese (mean BMI of 29 kg/m2) and had easily palpable bony landmarks—two patient characteristics associated with the least benefit of using ultrasound guidance in other studies.23 Another negative study had one experienced anesthesiologist marking obstetric patients with ultrasound, but junior residents performing the actual procedure in the absence of the anesthesiologist who had marked the patient.41

In general, the greatest benefit of using ultrasound guidance for LP has been demonstrated in obese patients.24,32,34,35,52,53 Benefits have been shown in specific obese patient populations, including obstetric,31,54,55 orthopedic,24,56,57 and emergency department patients.30

By increasing the procedural success rates with the use of ultrasound at the bedside, fewer patients may be referred to interventional radiology for fluoroscopic-guided LP, decreasing the patient exposure to ionizing radiation. A randomized study (n = 112) that compared site marking with ultrasound guidance versus fluoroscopic guidance for epidural steroid injections found the two techniques to be equivalent with respect to mean procedure time, number of needle insertion attempts, or needle passes.58 Another randomized study found that the performance time of ultrasound guidance was two minutes shorter (P < .05) than fluoroscopic guidance.59

 

 

Techniques

2) We recommend that ultrasound should be used to more accurately identify the lumbar spine level than physical examination in both obese and nonobese patients.

Rationale. Traditionally, an imaginary line connecting the iliac crests (intercristal line, Tuffier’s line, or Jacoby’s line) was considered to identify the L4 vertebra or the L4-L5 interspinous space in the midline; however, studies have revealed this traditional landmark to be much less accurate than previously thought. In general, palpating the iliac crests to mark the intercristal line identifies an interspinous space that is one space cephalad (ie, the L2-L3 interspinous space) but can range from L1-L2 to L4-L5.46,60-64 If an LP is inadvertently performed in the L1-L2 interspinous space, the risk of spinal cord injury is higher than that when performed in a more distal interspinous space.

A study by Margarido et al. with 45 patients with a mean BMI of 30 kg/m2 found that the intercristal line was located above the L4-L5 interspinous space in 100% of patients. More importantly, the intercristal line was above L2-L3 in 36% of patients and above L1-L2 in 4% of patients. It is important to note that patients with scoliosis or previous spine surgery were excluded from this study, and all examinations were performed by two experienced anesthesiologists with patients in a sitting position—all factors that would favor accurate palpation and marking of the iliac crests.60

In a study of nonobese patients (mean BMI 28 kg/m2) undergoing spinal anesthesia, Duniec et al. compared the lumbar level identified by palpation versus ultrasound and found discordance between the two techniques in 36% of patients; 18% were one space too cephalad, 16% were one space too caudal, and 2% were off by two interspinous spaces.61 Another study found discordance in 64% of patients (mean BMI 28 kg/m2) when comparing the interspinous level where spinal anesthesia had been performed by palpation versus a post-procedural ultrasound examination. This study revealed that the interspinous space was more cephalad in 50% of patients with 6% of punctures performed in the L1-L2 interspace.62 A similar study compared the accuracy of palpation vs ultrasound to identify the L3-L4 interspinous space in obese (mean BMI 34 kg/m2) versus nonobese (mean BMI 27 kg/m2) patients. This study found marking a space above L3-L4 in 51% of obese and 40% of nonobese patients and marking of the L1-L2 interspace in 7% of obese and 4% of nonobese patients.64

A study comparing palpation vs ultrasound found that 68% of obese patients with a BMI of >30 kg/m2 had difficult-to-palpate lumbar spine landmarks, but with the use of ultrasound, landmarks were identified in 76% of all patients, including obese and nonobese, with difficult-to-palpate landmarks.65

3) We suggest using ultrasound for selecting and marking a needle insertion site just before performing LPs in either a lateral decubitus or sitting position. The patient should remain in the same position after marking the needle insertion site.

Rationale. Ultrasound mapping of the lumbar spine can be performed in either a lateral decubitus or sitting position. Selecting and marking a needle insertion site should be performed at the bedside just before performing the procedure. The patient must remain in the same position in the interim between marking and inserting the needle, as a slight change in position can alter the needle trajectory, lowering the LP success rate. Although performing LPs in a lateral decubitus position has the advantage of accurately measuring the opening pressure, misalignment of the shoulder and pelvic girdles and bowing of the bed in a lateral decubitus position may lower LP success rates.

 

 

One randomized study comparing ultrasound-guided spinal anesthesia in a lateral decubitus versus sitting position found no difference in the number of needle insertion attempts or measurement of the skin-dura distance; however, the needle insertion depth was 0.73 cm greater in a lateral decubitus vs sitting position (P = .002).66 Procedural success rates of LP with ultrasound guidance have not been directly compared in a sitting versus lateral decubitus position, although the overall procedural success rates were higher in one study that allowed the operator to choose either sitting or lateral decubitus position when ultrasound was used.32

4) We recommend that a low-frequency transducer, preferably a curvilinear array transducer, should be used to evaluate the lumbar spine and mark a needle insertion site in most patients. A high-frequency linear array transducer may be used in nonobese patients.

Rationale. Low-frequency transducers emit sound waves that penetrate deep tissues, allowing visualization of bones and ligaments of the lumbar spine. A high-frequency linear transducer offers better resolution but shallower penetration to approximately 6-9 cm, limiting its use for site marking in overweight and obese patients. In obese patients, the ligamentum flavum is often deeper than 6 cm, which requires a low-frequency transducer to be visualized.

Most of the randomized controlled studies demonstrating benefits of using ultrasound guidance compared with landmark guidance for performance of LP, epidural anesthesia, or spinal anesthesia have used a low-frequency, curvilinear transducer.22,24,26-28,31,34-36,39,43-45,67 Two randomized controlled trials used a high-frequency linear transducer for site marking of lumbar procedures.30,32,37 Using a high-frequency linear transducer has been described in real-time, ultrasound-guided LPs, the advantage being better needle visualization with a linear transducer.29 Detection of blood vessels by color flow Doppler may be another advantage of using a high-frequency linear transducer, although a study by Grau et al. showed that use of color flow Doppler with a low-frequency curvilinear transducer permitted visualization of interspinous vessels as small as 0.5 mm in size.68

5) We recommend that ultrasound should be used to map the lumbar spine, starting at the level of the sacrum and sliding the transducer cephalad, sequentially identifying the lumbar spine interspaces.Rationale. Although no studies have directly compared different ultrasound scanning protocols to map the lumbar spine, starting at the level of the sacrum and sliding the transducer cephalad to sequentially identify the lumbar interspinous spaces is the most commonly described technique in studies demonstrating improved clinical outcomes with the use of ultrasound.24,31,34,37,39,40,45,56,57,67 Because the sacrum can be easily recognized, identifying it first is most beneficial in patients with few or no palpable landmarks.

All five lumbar spinous processes and interspinous spaces can be mapped from the sacrum using either a midline or a paramedian approach, and the widest interspinous space can be selected. In a midline approach, either a transverse or a longitudinal view is obtained. The transducer is centered on the sacrum and slid cephalad from L5 to L1 to identify each spinous process and interspinous space. In a paramedian approach, longitudinal paramedian views are obtained from the L5–sacrum interspace to the L1–L2 interspace, and each interspinous space is identified as the transducer is slid cephalad. Both these approaches are effective for mapping the lumbar spine. Whether the entire lumbar spine is mapped, and whether a midline or a paramedian approach is utilized, will depend on the operator’s preference.

 

 

6) We recommend that ultrasound should be used in a transverse plane to mark the midline of the lumbar spine and a longitudinal plane to mark the interspinous spaces. The intersection of these two lines marks the needle insertion site.

Rationale. The most common technique described in comparative studies of ultrasound vs landmarks includes visualization of the lumbar spine in two planes, a transverse plane to identify the midline and a longitudinal plane to identify the interspinous spaces. The majority of randomized controlled studies that demonstrated a reduction in the number of needle insertion attempts and an increase in the procedural success rates have used this technique (see Clinical Outcomes).22,24,28,32,35-37,43,44 Marking the midline and interspinous space(s) for LP may be performed in any order, starting with either the transverse or longitudinal plane first.

The midline of the spine is marked by placing the transducer in a transverse plane over the lumbar spine, centering over the spinous processes that have a distinct hyperechoic tip and a prominent acoustic shadow deep to the bone, and drawing a line perpendicular to the center of the transducer delineating the midline. The midline should be marked over a minimum of two or three spinous processes.

To identify the interspinous spaces, the transducer is aligned longitudinally over the midline. The transducer is slid along the midline to identify the widest interspinous space. Once the transducer is centered over the widest interspinous space, a line perpendicular to the center of the transducer is drawn to mark the interspinous space. The intersection of the lines marking the spinal midline and the selected interspinous space identifies the needle entry point.

To visualize the ligamentum flavum from a paramedian view, the transducer is oriented longitudinally over the midline, slid approximately 1 cm laterally, and tilted approximately 15 degrees aiming the ultrasound beam toward the midline. The skin–ligamentum flavum distance is most reliably measured from a paramedian view. Alternatively, in some patients, the ligamentum flavum may be visualized in the midline and the depth can be measured.

7) We recommend that ultrasound should be used during a preprocedural evaluation to measure the distance from the skin surface to the ligamentum flavum from a longitudinal paramedian view to estimate the needle insertion depth and ensure that a spinal needle of adequate length is used.

Rationale. The distance from the skin to the ligamentum flavum can be measured using ultrasound during preprocedural planning. Knowing the depth to the ligamentum flavum preprocedurally allows the operator to procure a spinal needle of adequate length, anticipate the insertion depth before CSF can be obtained, determine the depth to which a local anesthetic will need to be injected, and decide whether the anticipated difficulty of the procedure warrants referral to or consultation with another specialist.

The skin–ligamentum flavum distance can be measured from a transverse midline view or a longitudinal paramedian view. A longitudinal paramedian view provides an unobstructed view of the ligamentum flavum due to less shadowing from bony structures compared with a midline view. Several studies have demonstrated a strong correlation between the skin–ligamentum flavum distance measured by ultrasound and the actual needle insertion depth in both midline and paramedian views.28,34,36,53,54,57,69,70

A meta-analysis that included 13 comparative studies evaluating the correlation between ultrasound-measured depth and actual needle insertion depth to reach the epidural or intrathecal space consistently demonstrated a strong correlation between the measured and actual depth.50 A few studies have reported near-perfect Pearson correlation coefficients of 0.98.55,71,72 The pooled correlation was 0.91 (95% CI 0.87-0.94). All studies measured the depth from the skin to the ventral side of the ligamentum flavum or the intrathecal space from either a longitudinal paramedian view (n = 4) or a transverse midline view (n = 9). Eight of the more recent studies evaluated the accuracy of the ultrasound measurements and found the depth measurements by ultrasound to be accurate within 1-13 mm of the actual needle insertion depth, with seven of the eight studies reporting a mean difference of ≤3 mm.50

Measurement of the distance between the skin and the ligamentum flavum generally underestimates the needle insertion depth. One study reported that measurement of the skin–ligamentum flavum distance underestimates the needle insertion depth by 7.6 mm to obtain CSF, whereas measurement of the skin–posterior longitudinal ligament distance overestimates the needle insertion depth by 2.5 mm.57 A well-accepted contributor to underestimation of the depth measurements using ultrasound is compression of the skin and soft tissues by the transducer, and therefore, pressure on the skin must be released before freezing an image and measuring the depth to the subarachnoid space.

 

 

Training

8) We recommend that novices should undergo simulation-based training, where available, before attempting ultrasound-guided LPs on actual patients.

Rationale. Similar to training for other bedside procedures, dedicated training sessions, including didactics, supervised practice on patients, and simulation-based practice, should be considered when teaching novices to perform ultrasound-guided LP. Simulation-based training facilitates acquisition of knowledge and skills to perform invasive bedside procedures, including LP.73 Simulation-based training has been commonly incorporated into procedure training for trainees using an immersive experience, such as a “boot camp,”74-77 or a standardized curriculum,78,79 and has demonstrated improvements in post-course procedural knowledge, technical skills, and operator confidence. Two of these studies included training in the use of ultrasound guidance for LP. These studies showed that simulation-based practice improved skill acquisition and confidence.80,81 Simulation using novel computer software may improve skill acquisition in the use of ultrasound guidance for LP.82

9) We recommend that training in ultrasound-guided LPs should be adapted based on prior ultrasound experience, as learning curves will vary.Rationale. The learning curve to achieve competency in the use of ultrasound guidance for LP has not been well studied. The rate of attaining competency in identifying lumbar spine structures using ultrasound will vary by provider based on prior skills in ultrasound-guided procedures.83 Thus, providers with prior ultrasound experience may require less training than those without such experience to achieve competency. However, extensive experience in performing landmark-guided LPs does not necessarily translate into rapid acquisition of skills to perform the procedure with ultrasound guidance. A study of practicing anesthesiologists with no prior ultrasound experience demonstrated that 20 supervised trials of ultrasound-guided spinal anesthesia were insufficient to achieve competency.84 Although minimums may be a necessary step to gain competence, using them as a sole means to define competence does not account for variable learning curves.12 Based on a national survey of 21 hospitalist procedure experts, the mean current vs suggested minimums for initial and ongoing hospital privileging for LPs were 1.8 vs 6.9 and 2.2 vs 4.6 annually in one report.85

A fundamental question that needs to be answered is how to define competency in the use of ultrasound guidance for LP, including the specific skills and knowledge that must be mastered. At a minimum, providers must be able to identify lumbar spinous processes and distinguish them from the sacrum, identify the lumbar interspinous spaces and their corresponding levels, and estimate the depth from the skin to the ligamentum flavum from the midline and paramedian planes. Novice operators may benefit from practicing lumbar spine mapping of nonobese patients using a high-frequency linear transducer that generates high-resolution images and facilitates recognition of lumbar spine structures.

10) We recommend that novice providers should be supervised when performing ultrasound-guided LPs before performing the procedure independently on patients.

Rationale: Demonstration of competency in the use of ultrasound to identify lumbar spine anatomy should be achieved before routinely performing the procedure independently on patients.18 All providers will require a variable period of supervised practice to demonstrate the appropriate technique, followed by a period of unsupervised practice before competency is achieved. Supervised practice with guidance and feedback has been shown to significantly improve providers’ ability to delineate lumbar spine anatomy.86

 

 

KNOWLEDGE GAPS

The process of producing these guidelines revealed areas of uncertainty and important gaps in the literature regarding the use of ultrasound guidance for LP.

First, it is unclear whether the use of ultrasound guidance for LP reduces postprocedural back pain and whether it improves patient satisfaction. Several studies have evaluated postprocedural back pain28,30,32,33,52 and patient satisfaction28,29,33,51 with the use of ultrasound guidance, but these studies have found inconsistent results. Some of these results were probably due to insufficient statistical power or confounding variables. Furthermore, benefits have been demonstrated in certain subgroups, such as overweight patients or those with anatomical abnormalities, as was found in two studies.52,87 Use of ultrasound guidance for spinal anesthesia has been shown to reduce postprocedural headache28 and improve patient satisfaction51, although similar benefit has not been demonstrated in patients undergoing LP.

Second, the effect of using ultrasound guidance on the frequency of traumatic LPs is an area of uncertainty. A “traumatic tap” is defined as an inadvertent puncture of an epidural vein during passage of the spinal needle through the dura. It remains difficult to discern in these studies whether red blood cells detected in the CSF resulted from puncture of an epidural vein or from needle trauma of the skin and soft tissues. Despite this uncertainty, at least seven randomized controlled studies have assessed the effect of ultrasound guidance on traumatic LPs. The meta-analysis by Shaikh et al. included five randomized controlled studies that assessed the effect of ultrasound guidance on the reporting of traumatic taps. The study found a reduced risk of traumatic taps (risk ratio 0.27 [95% CI 0.11-0.67]), an absolute risk reduction of 5.9% (95% CI 2.3%-9.5%), and a number needed to treat of 17 (95% CI 11-44) to prevent one traumatic tap.16 Similarly, the meta-analysis by Gottlieb et al. showed a lower risk of traumatic taps among adults undergoing LP with ultrasound guidance in five randomized controlled studies with an odds ratio of 0.28 (95% CI 0.14-0.59). The meta-analysis by Gottlieb et al. included two adult studies that were not included by Shaikh et al.

Third, several important questions about the technique of ultrasound-guided LP remain unanswered. In addition to the static technique, a dynamic technique with real-time needle tracking has been described to perform ultrasound-guided LP, epidural catheterization, and spinal anesthesia. A pilot study by Grau et al. found that ultrasound used either statically or dynamically had fewer insertion attempts and needle redirections than use of landmarks alone.29 Three other pilot studies showed successful spinal anesthesia in almost all patients88-90 and one large study demonstrated successful spinal anesthesia with real-time ultrasound guidance in 97 of 100 patients with a median of three needle passes.91 Furthermore, a few industry-sponsored studies with small numbers of patients have described the use of novel needle tracking systems that facilitate needle visualization during real-time ultrasound-guided LP.92,93 However, to our knowledge, no comparative studies of static versus dynamic guidance using novel needle tracking systems in human subjects have been published, and any potential role for these novel needle tracking systems has not yet been defined.

Finally, the effects of using ultrasound guidance on clinical decision-making, timeliness, and cost-effectiveness of LP have not yet been explored but could have important clinical practice implications.

 

 

CONCLUSION

Randomized controlled trials have demonstrated that using ultrasound guidance for LPs can reduce the number of needle insertion attempts and needle redirections and increase the overall procedural success rates. Ultrasound can more accurately identify the lumbar spine level than physical examination in both obese and nonobese patients, although the greatest benefit of using ultrasound guidance for LPs has been shown in obese patients.

Ultrasound permits assessment of the interspinous space width and measurement of the ligamentum flavum depth to select an optimal needle insertion site and adequate length spinal needle. Although the use of real-time ultrasound guidance has been described, the use of static ultrasound guidance for LP site marking remains the standard technique.

Acknowledgments

The authors thank all the members of the Society of Hospital Medicine Point-of-care Ultrasound Task Force and the Education Committee members for their time and dedication to develop these guidelines.

Collaborators from Society of Hospital Medicine Point-of-care Ultrasound Task Force: Saaid Abdel-Ghani, Robert Arntfield, Jeffrey Bates, Anjali Bhagra, Michael Blaivas, Daniel Brotman, Carolina Candotti, Richard Hoppmann, Susan Hunt, Trevor P. Jensen, Paul Mayo, Benji Mathews, Satyen Nichani, Vicki Noble, Martin Perez, Nitin Puri, Aliaksei Pustavoitau, Kreegan Reierson, Sophia Rodgers, Kirk Spencer, Vivek Tayal, David Tierney

SHM Point-of-care Ultrasound Task Force: CHAIRS: Nilam Soni, Ricardo Franco-Sadud, Jeff Bates. WORKING GROUPS: Thoracentesis Working Group: Ria Dancel (chair), Daniel Schnobrich, Nitin Puri. Vascular Access Working Group: Ricardo Franco (chair), Benji Matthews, Saaid Abdel-Ghani, Sophia Rodgers, Martin Perez, Daniel Schnobrich. Paracentesis Working Group: Joel Cho (chair), Benji Matthews, Kreegan Reierson, Anjali Bhagra, Trevor P. Jensen Lumbar Puncture Working Group: Nilam J. Soni (chair), Ricardo Franco, Gerard Salame, Josh Lenchus, Venkat Kalidindi, Ketino Kobaidze. Credentialing Working Group: Brian P Lucas (chair), David Tierney, Trevor P. Jensen PEER REVIEWERS: Robert Arntfield, Michael Blaivas, Richard Hoppmann, Paul Mayo, Vicki Noble, Aliaksei Pustavoitau, Kirk Spencer, Vivek Tayal. METHODOLOGIST: Mahmoud El Barbary. LIBRARIAN: Loretta Grikis. SOCIETY OF HOSPITAL MEDICINE EDUCATION COMMITTEE: Daniel Brotman (past chair), Satyen Nichani (current chair), Susan Hunt. SOCIETY OF HOSPITAL MEDICINE STAFF: Nick Marzano.

Disclosures

The authors have nothing to disclose.

Funding

Brian P Lucas: Department of Veterans Affairs, Veterans Health Administration, Office of Research and Development and Dartmouth SYNERGY, National Institutes of Health, National Center for Translational Science (UL1TR001086). Nilam Soni: Department of Veterans Affairs, Quality Enhancement Research Initiative (QUERI) Partnered Evaluation Initiative Grant (HX002263-01A1).

Disclaimer

The contents of this publication do not represent the views of the U.S. Department of Veterans Affairs or the United States Government.

 

Files
References

1. Wolfe KS, Kress JP. Risk of procedural hemorrhage. Chest. 2016;150(1):237-246. https://doi.org/10.1016/j.chest.2016.01.023.
2. Kroll H, Duszak R, Jr, Nsiah E, et al. Trends in lumbar puncture over 2 decades: a dramatic shift to radiology. AJR Am J Roentgenol. 2015;204(1):15-19. https://doi.org/10.2214/AJR.14.12622.
3. Vickers A, Donnelly JP, Moore JX, Wang HE. 263EMF epidemiology of lumbar punctures in hospitalized patients in United States. Ann Emerg Med. 2017;70(4):S104. https://doi.org/10.1016/j.annemergmed.2017.07.241.
4. Køster-Rasmussen R, Korshin A, Meyer CN. Antibiotic treatment delay and outcome in acute bacterial meningitis. J Infect. 2008;57(6):449-454. https://doi.org/10.1016/j.jinf.2008.09.033.
5. Aronin SI, Peduzzi P, Quagliarello VJ. Community-acquired bacterial meningitis: risk stratification for adverse clinical outcome and effect of antibiotic timing. Ann Intern Med. 1998;129(11):862-869. https://doi.org/10.7326/0003-4819-129-11_Part_1-199812010-00004.
6. Lepur D, Barsić B. Community-acquired bacterial meningitis in adults: antibiotic timing in disease course and outcome. Infection. 2007;35(4):225-231. https://doi.org/10.1007/s15010-007-6202-0.
7. Proulx N, Fréchette D, Toye B, Chan J, Kravcik S. Delays in the administration of antibiotics are associated with mortality from adult acute bacterial meningitis. QJM. 2005;98(4):291-298. https://doi.org/10.1093/qjmed/hci047.
8. Auburtin M, Wolff M, Charpentier J, et al. Detrimental role of delayed antibiotic administration and penicillin-nonsusceptible strains in adult intensive care unit patients with pneumococcal meningitis: the PNEUMOREA prospective multicenter study. Crit Care Med. 2006;34(11):2758-2765. https://doi.org/10.1097/01.CCM.0000239434.26669.65.
9. Michael B, Menezes BF, Cunniffe J, et al. Effect of delayed lumbar punctures on the diagnosis of acute bacterial meningitis in adults. Emerg Med J. 2010;27(6):433-438. https://doi.org/10.1136/emj.2009.075598.
10. Glimåker M, Johansson B, Grindborg Ö, et al. Adult bacterial meningitis: earlier treatment and improved outcome following guideline revision promoting prompt lumbar puncture. Clin Infect Dis. 2015;60(8):1162-1169. https://doi.org/10.1093/cid/civ011.
11. Nichani S, Crocker J, Fitterman N, Lukela M. Updating the core competencies in hospital medicine--2017 Revision: introduction and methodology. J Hosp Med. 2017;12(4):283-287. https://doi.org/10.12788/jhm.2715.
12. Soni NJ, Schnobrich D, Matthews BK, et al. Point-of-care ultrasound for hospitalists: a position statement of the Society of Hospital Medicine. J Hosp Med. 2019;14:E1-E6. https://doi.org/10.12788/jhm.3079.
13. Shah KH, McGillicuddy D, Spear J, Edlow JA. Predicting difficult and traumatic lumbar punctures. Am J Emerg Med. 2007;25(6):608-611. https://doi.org/10.1016/j.ajem.2006.11.025.
14. Williams P, Tait G, Wijeratne T. Success rate of elective lumbar puncture at a major Melbourne neurology unit. Surg Neurol Int. 2018;9:12. https://doi.org/10.4103/sni.sni_426_17.
15. Gottlieb M, Holladay D, Peksa GD. Ultrasound-assisted lumbar punctures: a systematic review and meta-analysis. Acad Emerg Med. 2018;26(1). https://doi.org/10.1111/acem.13558.
16. Shaikh F, Brzezinski J, Alexander S, et al. Ultrasound imaging for lumbar punctures and epidural catheterisations: systematic review and meta-analysis. BMJ. 2013;346:f1720. https://doi.org/10.1136/bmj.f1720.
17. Perlas A, Chaparro LE, Chin KJ. Lumbar neuraxial ultrasound for spinal and epidural anesthesia: a systematic review and meta-analysis. Reg Anesth Pain Med. 2016;41(2):251-260. https://doi.org/10.1097/AAP.0000000000000184.
18. Lucas BP, Tierney DM, Jensen TP, et al. Credentialing of hospitalists in ultrasound-guided bedside procedures: a position statement of the Society of Hospital Medicine. J Hosp Med. 2018;13(2):117-125. https://doi.org/10.12788/jhm.2917.
19. Dancel R, Schnobrich D, Puri N, et al. Recommendations on the use of ultrasound guidance for adult thoracentesis: a position statement of the Society of Hospital Medicine. J Hosp Med. 2018;13(2):126-135. https://doi.org/10.12788/jhm.2940.
20. Soni NJ, Franco-Sadud R, Schnobrich D, et al. Ultrasound guidance for lumbar puncture. Neurol Clin Pract. 2016;6(4):358-368. https://doi.org/10.1212/CPJ.0000000000000265.
21. Fitch K, Bernstein SJ, Aguilar MD, Burnand B, LaCalle JR. The Rand/UCLA Appropriateness Method User’s Manual. Santa Monica, CA: Rand Corp; 2001.
22. Abdelhamid SA, Mansour MA. Ultrasound-guided intrathecal anesthesia: does scanning help? Egypt J Anaesth. 2013;29(4):389-394. https://doi.org/10.1016/j.egja.2013.06.003.
23. Ansari T, Yousef A, El Gamassy A, Fayez M. Ultrasound-guided spinal anaesthesia in obstetrics: is there an advantage over the landmark technique in patients with easily palpable spines? Int J Obstet Anesth. 2014;23(3):213-216. https://doi.org/10.1016/j.ijoa.2014.03.001.
24. Chin KJ, Perlas A, Chan V, et al. Ultrasound imaging facilitates spinal anesthesia in adults with difficult surface anatomic landmarks. Anesthesiology. 2011;115(1):94-101. https://doi.org/10.1097/ALN.0b013e31821a8ad4.
25. Cho YC, Koo DH, Oh SK, et al. Comparison of ultrasound-assisted lumbar puncture with lumbar puncture using palpation of landmarks in aged patients in an emergency center. J Korean Soc Emerg Med. 2009;20(3):304.
26. Grau T, Leipold RW, Conradi R, Martin E. Ultrasound control for presumed difficult epidural puncture. Acta Anaesthesiol Scand. 2001;45(6):766-771. https://doi.org/10.1034/j.1399-6576.2001.045006766.x.
27. Grau T, Leipold RW, Conradi R, Martin E, Motsch J. Ultrasound imaging facilitates localization of the epidural space during combined spinal and epidural anesthesia. Reg Anesth Pain Med. 2001;26(1):64-67. https://doi.org/10.1053/rapm.2001.19633.
28. Grau T, Leipold RW, Conradi R, Martin E, Motsch J. Efficacy of ultrasound imaging in obstetric epidural anesthesia. J Clin Anesth. 2002;14(3):169-175. https://doi.org/10.1016/S0952-8180(01)00378-6.
29. Grau T, Leipold RW, Fatehi S, Martin E, Motsch J. Real-time ultrasonic observation of combined spinal-epidural anaesthesia. Eur J Anaesthesiol. 2004;21(1):25-31. https://doi.org/10.1017/S026502150400105X.
30. Mofidi M, Mohammadi M, Saidi H, et al. Ultrasound guided lumbar puncture in emergency department: time saving and less complications. J Res Med Sci. 2013;18(4):303-307. PubMed
31. Nassar M, Abdelazim IA. Pre-puncture ultrasound guided epidural insertion before vaginal delivery. J Clin Monit Comput. 2015;29(5):573-577. https://doi.org/10.1007/s10877-014-9634-y.

32. Nomura JT, Leech SJ, Shenbagamurthi S, et al. A randomized controlled trial of ultrasound-assisted lumbar puncture. J Ultrasound Med. 2007;26(10):1341-1348. https://doi.org/10.7863/jum.2007.26.10.1341.
33. Peterson MA, Pisupati D, Heyming TW, Abele JA, Lewis RJ. Ultrasound for routine lumbar puncture. Acad Emerg Med. 2014;21(2):130-136. https://doi.org/10.1111/acem.12305.
34. Sahin T, Balaban O, Sahin L, Solak M, Toker K. A randomized controlled trial of preinsertion ultrasound guidance for spinal anaesthesia in pregnancy: outcomes among obese and lean parturients: ultrasound for spinal anesthesia in pregnancy. J Anesth. 2014;28(3):413-419. https://doi.org/10.1007/s00540-013-1726-1.
35. Wang Q, Yin C, Wang TL. Ultrasound facilitates identification of combined spinal-epidural puncture in obese parturients. Chin Med J (Engl). 2012;125(21):3840-3843. PubMed
36. Vallejo MC, Phelps AL, Singh S, Orebaugh SL, Sah N. Ultrasound decreases the failed labor epidural rate in resident trainees. Int J Obstet Anesth. 2010;19(4):373-378. https://doi.org/10.1016/j.ijoa.2010.04.002.
37. Darrieutort-Laffite C, Bart G, Planche L, et al. Usefulness of a pre-procedure ultrasound scanning of the lumbar spine before epidural injection in patients with a presumed difficult puncture: a randomized controlled trial. Joint Bone Spine. 2015;82(5):356-361. https://doi.org/10.1016/j.jbspin.2015.02.001.
38. Vosko MR, Brunner C, Schreiber S. Lumbar puncture with ultrasound study (lupus study)-international prospective randomized multicentre trial. Int J Stroke. 2017;12(1):22. https://doi.org/10.1055/s-0037-1606991.
39. Urfalioğlu A, Bilal B, Öksüz G, et al. Comparison of the landmark and ultrasound methods in cesarean sections performed under spinal anesthesia on obese pregnants. J Matern Fetal Neonatal Med. 2017;30(9):1051-1056. https://doi.org/10.1080/14767058.2016.1199677.
40. Tawfik MM, Atallah MM, Elkharboutly WS, Allakkany NS, Abdelkhalek M. Does preprocedural ultrasound increase the first-pass success rate of epidural catheterization before cesarean delivery? A randomized controlled trial. Anesth Analg. 2017;124(3):851-856. https://doi.org/10.1213/ANE.0000000000001325.
41. Turkstra TP, Marmai KL, Armstrong KP, Kumar K, Singh SI. Preprocedural ultrasound assessment does not improve trainee performance of spinal anesthesia for obstetrical patients: a randomized controlled trial. J Clin Anesth. 2017;37:21-24. https://doi.org/10.1016/j.jclinane.2016.10.034.
42. Chong SE, Mohd Nikman A, Saedah A, et al. Real-time ultrasound-guided paramedian spinal anaesthesia: evaluation of the efficacy and the success rate of single needle pass. Br J Anaesth. 2017;118(5):799-801. https://doi.org/10.1093/bja/aex108.
43. Creaney M, Mullane D, Casby C, Tan T. Ultrasound to identify the lumbar space in women with impalpable bony landmarks presenting for elective caesarean delivery under spinal anaesthesia: a randomised trial. Int J Obstet Anesth. 2016;28:12-16. https://doi.org/10.1016/j.ijoa.2016.07.007.
44. Ekinci M, Alici HA, Ahiskalioglu A, et al. The use of ultrasound in planned cesarean delivery under spinal anesthesia for patients having nonprominent anatomic landmarks. J Clin Anesth. 2017;37:82-85. https://doi.org/10.1016/j.jclinane.2016.10.014.
45. Perna P, Gioia A, Ragazzi R, Volta CA, Innamorato M. Can pre-procedure neuroaxial ultrasound improve the identification of the potential epidural space when compared with anatomical landmarks? A prospective randomized study. Minerva Anestesiol. 2017;83(1):41-49. https://doi.org/10.23736/S0375-9393.16.11399-9.
46. Chin A, Crooke B, Heywood L, et al. A randomised controlled trial comparing needle movements during combined spinal-epidural anaesthesia with and without ultrasound assistance. Anaesthesia. 2018;73(4):466-473. https://doi.org/10.1111/anae.14206.
47. Dhanger S, Vinayagam S, Vaidhyanathan B, Rajesh IJ, Tripathy DK. Comparison of landmark versus pre-procedural ultrasonography-assisted midline approach for identification of subarachnoid space in elective caesarean section: a randomised controlled trial. Indian J Anaesth. 2018;62(4):280-284. https://doi.org/10.4103/ija.IJA_488_17.
48. Evans DP, Tozer J, Joyce M, Vitto MJ. Comparison of ultrasound-guided and landmark-based lumbar punctures in inexperienced resident physicians. J Ultrasound Med. 2019;38(3):613-620. https://doi.org/10.1002/jum.14728.
49. Srinivasan KK, Leo AM, Iohom G, Loughnane F, Lee PJ. Pre-procedure ultrasound-guided paramedian spinal anaesthesia at L5-S1: is this better than landmark-guided midline approach? A randomised controlled trial. Indian J Anaesth. 2018;62(1):53-60. https://doi.org/10.4103/ija.IJA_448_17.
50. Perlas A, Chaparro LE, Chin KJ. Lumbar neuraxial ultrasound for spinal and epidural anesthesia: a systematic review and meta-analysis. Reg Anesth Pain Med. 2016;41(2):251-260. https://doi.org/10.1097/AAP.0000000000000184.
51. Lim YC, Choo CY, Tan KT. A randomised controlled trial of ultrasound-assisted spinal anaesthesia. Anaesth Intensive Care. 2014;42(2):191-198. https://doi.org/10.1177/0310057X1404200205.

52. Honarbakhsh S, Osman C, Teo JTH, Gabriel C. Ultrasound-guided lumbar puncture as a diagnostic aid to reduce number of attempts and complication rates. Ultrasound. 2013;21(4):170-175. https://doi.org/10.1177/1742271X13504332.
53. Sahota JS, Carvalho JC, Balki M, Fanning N, Arzola C. Ultrasound estimates for midline epidural punctures in the obese parturient: paramedian sagittal oblique is comparable to transverse median plane. Anesth Analg. 2013;116(4):829-835. https://doi.org/10.1213/ANE.0b013e31827f55f0.
54. Balki M, Lee Y, Halpern S, Carvalho JC. Ultrasound imaging of the lumbar spine in the transverse plane: the correlation between estimated and actual depth to the epidural space in obese parturients. Anesth Analg. 2009;108(6):1876-1881. https://doi.org/10.1213/ane.0b013e3181a323f6.
55. Wallace DH, Currie JM, Gilstrap LC, Santos R. Indirect sonographic guidance for epidural anesthesia in obese pregnant patients. Reg Anesth. 1992;17(4):233-236. PubMed
56. Srinivasan KK, Iohom G, Loughnane F, Lee PJ. Conventional landmark-guided midline versus preprocedure ultrasound-guided paramedian techniques in spinal anesthesia. Anesth Analg. 2015;21(4):1089-1096. https://doi.org/10.1213/ANE.0000000000000911.
57. Chin KJ, Perlas A, Singh M, et al. An ultrasound-assisted approach facilitates spinal anesthesia for total joint arthroplasty. Can J Anaesth. 2009;56(9):643-650. https://doi.org/10.1007/s12630-009-9132-8.
58. Evansa I, Logina I, Vanags I, Borgeat A. Ultrasound versus fluoroscopic-guided epidural steroid injections in patients with degenerative spinal diseases: a randomised study. Eur J Anaesthesiol. 2015;32(4):262-268. https://doi.org/10.1097/EJA.0000000000000103.
59. Park Y, Lee JH, Park KD, et al. Ultrasound-guided vs fluoroscopy-guided caudal epidural steroid injection for the treatment of unilateral lower lumbar radicular pain: a prospective, randomized, single-blind clinical study. Am J Phys Med Rehabil. 2013;92(7):575-586. https://doi.org/10.1097/PHM.0b013e318292356b.
60. Margarido CB, Mikhael R, Arzola C, Balki M, Carvalho JC. The intercristal line determined by palpation is not a reliable anatomical landmark for neuraxial anesthesia. Can J Anaesth. 2011;58(3):262-266. https://doi.org/10.1007/s12630-010-9432-z.
61. Duniec L, Nowakowski P, Kosson D, Łazowski T. Anatomical landmarks based assessment of intravertebral space level for lumbar puncture is misleading in more than 30%. Anaesthesiol Intensive Ther. 2013;45(1):1-6. https://doi.org/10.5603/AIT.2013.0001.
62. Schlotterbeck H, Schaeffer R, Dow WA, et al. Ultrasonographic control of the puncture level for lumbar neuraxial block in obstetric anaesthesia. Br J Anaesth. 2008;100(2):230-234. https://doi.org/10.1093/bja/aem371.
63. Whitty R, Moore M, Macarthur A. Identification of the lumbar interspinous spaces: palpation versus ultrasound. Anesth Analg. 2008;106(2):538-540, table of contents. https://doi.org/10.1213/ane.0b013e31816069d9.
64. Locks Gde F, Almeida MC, Pereira AA. Use of the ultrasound to determine the level of lumbar puncture in pregnant women. Rev Bras Anestesiol. 2010;60(1):13-19. https://doi.org/10.1016/S0034-7094(10)70002-7.
65. Stiffler KA, Jwayyed S, Wilber ST, Robinson A. The use of ultrasound to identify pertinent landmarks for lumbar puncture. Am J Emerg Med. 2007;25(3):331-334. https://doi.org/10.1016/j.ajem.2006.07.010.

66. Gulay U, Meltem T, Nadir SS, Aysin A. Ultrasound-guided evaluation of the lumbar subarachnoid space in lateral and sitting positions in pregnant patients to receive elective cesarean operation. Pak J Med Sci. 2015;31(1):76-81. https://doi.org/10.12669/pjms.311.5647.
67. Kawaguchi R, Yamauchi M, Sugino S, Yamakage M. Ultrasound-aided ipsilateral-dominant epidural block for total hip arthroplasty: a randomised controlled single-blind study. Eur J Anaesthesiol. 2011;28(2):137-140. https://doi.org/10.1097/EJA.0b013e3283423457.
68. Grau T, Leipold RW, Horter J, Martin E, Motsch J. Colour Doppler imaging of the interspinous and epidural space. Eur J Anaesthesiol. 2001;18(11):706-712. https://doi.org/10.1097/00003643-200111000-00002.
69. Arzola C, Davies S, Rofaeel A, Carvalho JC. Ultrasound using the transverse approach to the lumbar spine provides reliable landmarks for labor epidurals. Anesth Analg. 2007;104(5):1188-92, tables of contents. https://doi.org/10.1213/01.ane.0000250912.66057.41.
70. Chauhan AK, Bhatia R, Agrawal S. Lumbar epidural depth using transverse ultrasound scan and its correlation with loss of resistance technique: a prospective observational study in Indian population. Saudi J Anaesth. 2018;12(2):279-282. https://doi.org/10.4103/sja.SJA_679_17.
71. Gnaho A, Nguyen V, Villevielle T, et al. Assessing the depth of the subarachnoid space by ultrasound. Rev Bras Anestesiol. 2012;62(4):520-530. https://doi.org/10.1016/S0034-7094(12)70150-2.
72. Cork RC, Kryc JJ, Vaughan RW. Ultrasonic localization of the lumbar epidural space. Anesthesiology. 1980;52(6):513-516. https://doi.org/10.1097/00000542-198006000-00013.
73. Barsuk JH, Cohen ER, Caprio T, et al. Simulation-based education with mastery learning improves residents’ lumbar puncture skills. Neurology. 2012;79(2):132-137. https://doi.org/10.1212/WNL.0b013e31825dd39d.
74. Lenchus J, Issenberg SB, Murphy D, et al. A blended approach to invasive bedside procedural instruction. Med Teach. 2011;33(2):116-123. https://doi.org/10.3109/0142159X.2010.509412.
75. Wayne DB, Cohen ER, Singer BD, et al. Progress toward improving medical school graduates’ skills via a “boot camp” curriculum. Simul Healthc. 2014;9(1):33-39. https://doi.org/10.1097/SIH.0000000000000001.
76. Cohen ER, Barsuk JH, Moazed F, et al. Making July safer: simulation-based mastery learning during intern boot camp. Acad Med. 2013;88(2):233-239. https://doi.org/10.1097/ACM.0b013e31827bfc0a.
77. Martin R, Gannon D, Riggle J, et al. A comprehensive workshop using simulation to train internal medicine residents in bedside procedures performed by internists. Chest. 2012;142(4):545A. https://doi.org/10.1378/chest.1390093.
78. Lenchus JD. End of the “see one, do one, teach one” era: the next generation of invasive bedside procedural instruction. J Am Osteopath Assoc. 2010;110(6):340-346. PubMed
79. Mourad M, Ranji S, Sliwka D. A randomized controlled trial of the impact of a teaching procedure service on the training of internal medicine residents. J Grad Med Educ. 2012;4(2):170-175. https://doi.org/10.4300/JGME-D-11-00136.1.
80. Restrepo CG, Baker MD, Pruitt CM, Gullett JP, Pigott DC. Ability of pediatric emergency medicine physicians to identify anatomic landmarks with the assistance of ultrasound prior to lumbar puncture in a simulated obese model. Pediatr Emerg Care. 2015;31(1):15-19. https://doi.org/10.1097/PEC.0000000000000330.
81. VanderWielen BA, Harris R, Galgon RE, VanderWielen LM, Schroeder KM. Teaching sonoanatomy to anesthesia faculty and residents: utility of hands-on gel phantom and instructional video training models. J Clin Anesth. 2015;27(3):188-194. https://doi.org/10.1016/j.jclinane.2014.07.007.
82. Keri Z, Sydor D, Ungi T, et al. Computerized training system for ultrasound-guided lumbar puncture on abnormal spine models: a randomized controlled trial. Can J Anaesth. 2015;62(7):777-784. https://doi.org/10.1007/s12630-015-0367-2.
83. Deacon AJ, Melhuishi NS, Terblanche NC. CUSUM method for construction of trainee spinal ultrasound learning curves following standardised teaching. Anaesth Intensive Care. 2014;42(4):480-486. https://doi.org/10.1177/0310057X1404200409.
84. Margarido CB, Arzola C, Balki M, Carvalho JC. Anesthesiologists’ learning curves for ultrasound assessment of the lumbar spine. Can J Anaesth. 2010;57(2):120-126. https://doi.org/10.1007/s12630-009-9219-2.
85. Jensen TP, Soni NJ, Tierney DM, Lucas BP. Hospital privileging practices for bedside procedures: a survey of hospitalist experts. J Hosp Med. 2017;12(10):836-839. https://doi.org/10.12788/jhm.2837.
86. Terblanche NC, Arzola C, Wills KE, et al. Standardised training program in spinal ultrasound for epidural insertion: protocol driven versus non-protocol driven teaching approach. Anaesth Intensive Care. 2014;42(4):460-466. https://doi.org/10.1177/0310057X1404200406.
87. Mofidi M, Mohammadi M, Saidi H, et al. Ultrasound guided lumbar puncture in emergency department: time saving and less complications. J Res Med Sci. 2013;18(4):303-307. PubMed
88. Karmakar MK, Li X, Ho AM, Kwok WH, Chui PT. Real-time ultrasound-guided paramedian epidural access: evaluation of a novel in-plane technique. Br J Anaesth. 2009;102(6):845-854. https://doi.org/10.1093/bja/aep079.
89. Tran D, Kamani AA, Al-Attas E, et al. Single-operator real-time ultrasound-guidance to aim and insert a lumbar epidural needle. Can J Anaesth. 2010;57(4):313-321. https://doi.org/10.1007/s12630-009-9252-1.
90. Liu Y, Qian W, Ke XJ, Mei W. Real-time ultrasound-guided spinal anesthesia using a new paramedian transverse approach. Curr Med Sci. 2018;38(5):910-913. https://doi.org/10.1007/s11596-018-1961-7.
91. Conroy PH, Luyet C, McCartney CJ, McHardy PG. Real-time ultrasound-guided spinal anaesthesia: a prospective observational study of a new approach. Anesthesiol Res Pract. 2013;2013:525818. https://doi.org/10.1155/2013/525818.
92. Brinkmann S, Tang R, Sawka A, Vaghadia H. Single-operator real-time ultrasound-guided spinal injection using SonixGPS™: a case series. Can J Anaesth. 2013;60(9):896-901. https://doi.org/10.1007/s12630-013-9984-9.
93. Niazi AU, Chin KJ, Jin R, Chan VW. Real-time ultrasound-guided spinal anesthesia using the SonixGPS ultrasound guidance system: a feasibility study. Acta Anaesthesiol Scand. 2014;58(7):875-881. https://doi.org/10.1111/aas.12353.

Article PDF
Issue
Journal of Hospital Medicine 14(10)
Publications
Topics
Page Number
591-601. Published online first June 10, 2019
Sections
Files
Files
Article PDF
Article PDF
Related Articles

Approximately 400,000 lumbar punctures (LPs) are performed in the United States annually for either diagnostic workup or therapeutic relief.1 Lumbar punctures are increasingly being performed in the United States, with an estimated 97,000 LPs performed on Medicare fee-for-service beneficiaries in 2011 alone, which is an increase of approximately 4,000 LPs in the same population from 1991.2 Approximately 273,612 LPs were performed on hospitalized patients in the United States in 2010,1 and the inpatient hospital setting is the most common site for LPs.2,3

Many LPs are referred to radiologists who have access to imaging guidance to aid with needle insertion.2 However, referrals to radiology delay performance of LPs, and delayed diagnosis of acute bacterial meningitis, the most common yet serious condition for which LPs are performed, is associated with increased morbidity and mortality.4-8 Furthermore, although initiating empiric antibiotic treatment for suspected acute bacterial meningitis is recommended in some cases, doing so routinely can cause false-negative cerebrospinal fluid (CSF) culture results, complicating decisions about de-escalation and duration of antibiotics that could have been safely avoided by promptly performing an LP.9

Delaying the performance of LP has been associated with increased mortality.10 Demonstration of proficiency in performance of lumbar puncture is considered a core competency for hospitalists,11 and with the increasing availability of point-of-care ultrasound, hospitalists can use ultrasound to guide performance of LPs at the bedside.12 However, 30% of patients requiring LP in emergency departments have difficult-to-palpate lumbar spine landmarks,13 and lumbar puncture performed based on palpation of landmarks alone has been reported to fail or be traumatic in 28% of patients.14 Use of ultrasound guidance for lumbar puncture has been shown in randomized controlled trials to improve procedural success rates, while reducing the time to successful LP, needle passes, patient pain scores, and risk of a traumatic LP.15-17

The purpose of this position statement is to review the literature and present consensus-based recommendations on the performance of ultrasound-guided LP in adult patients. This position statement does not mandate that hospitalists use ultrasound guidance for LP, nor does it establish ultrasound guidance as the standard of care for LP. Similar to previously published Society of Hospital Medicine (SHM) position statements,12,18,19 this document presents recommendations with supporting evidence for the clinical outcomes, techniques, and training for using ultrasound guidance for LP. A manuscript describing the technique of ultrasound guidance for LPs has been previously published by some of the authors of this position statement.20

 

 

METHODS

Detailed methods are described in Appendix 1. The SHM Point-of-care Ultrasound (POCUS) Task Force was assembled to carry out this guideline development project under the direction of the SHM Board of Directors, Director of Education, and Education Committee. All expert panel members were physicians or advanced practice providers with expertise in POCUS. Expert panel members were divided into working group members, external peer reviewers, and a methodologist. All Task Force members were required to disclose any potential conflicts of interests (Appendix 2). The literature search was conducted in two independent phases. The first phase included literature searches conducted by the six working group members themselves. Key clinical questions and draft recommendations were then prepared. A systematic literature search was conducted by a medical librarian based on the findings of the initial literature search and draft recommendations. The Medline, Embase, CINAHL, and Cochrane medical databases were searched from 1975 to December 2015 initially. Google Scholar was also searched without limiters. Updated searches were conducted in November 2016, January 2018, and October 2018. The search strings are included in Appendix 3. All article abstracts were first screened for relevance by at least two members of the working group. Full-text versions of screened articles were reviewed, and articles on the use of ultrasound to guide LP were selected. In addition, the following article types were excluded: non-English language, nonhuman, age <18 years, meeting abstracts, meeting posters, narrative reviews, case reports, letters, and editorials. Moreover, studies focusing on the use of ultrasound guidance for spinal nerve root injections, regional anesthesia, and assessment of lumbar spine anatomy alone were excluded. All relevant systematic reviews, meta-analyses, randomized controlled trials, and observational studies of ultrasound-guided LP were screened and selected. Final article selection was based on working group consensus, and the selected literature was incorporated into the draft recommendations.

The Research and Development (RAND) Appropriateness Method that required panel judgment and consensus was used.21 The 27 voting members of the SHM POCUS Task Force reviewed and voted on the draft recommendations considering the following five transforming factors: (1) Problem priority and importance, (2) Level of quality of evidence, (3) Benefit/harm balance, (4) Benefit/burden balance, and (5) Certainty/concerns about PEAF (Preferences/Equity/Acceptability/Feasibility). Panel members participated in two rounds of electronic voting using an internet-based electronic data collection tool (REDCap™) in February 2018 and April 2018 (Appendix 4). Voting on appropriateness was conducted using a 9-point Likert scale. The three zones of the 9-point Likert scale were inappropriate (1-3 points), uncertain (4-6 points), and appropriate (7-9 points). The degree of consensus was assessed using the RAND algorithm (Appendix Figure 1 and Table 1). Establishing a recommendation required at least 70% agreement that a recommendation was “appropriate.” A strong recommendation required 80% of the votes within one integer of the median, following the RAND rules. Disagreement was defined as >30% of panelists voting outside of the zone of the median.

Recommendations were classified as strong or weak/conditional based on preset rules defining the panel’s level of consensus, which determined the wording of each recommendation (Table 2). The revised consensus-based recommendations underwent internal and external reviews by POCUS experts from different subspecialties. The final review of this position statement was performed by members of the SHM POCUS Task Force, SHM Education Committee, and SHM Executive Committee. The SHM Executive Committee endorsed this position statement in June 2018 before submission to the Journal of Hospital Medicine.

 

 

RESULTS

Literature Search

A total of 4,389 references were pooled from four different sources: a search by a certified medical librarian in December 2015 (3,212 citations) that was updated in November 2016 (380 citations), January 2018 (282 citations), and October 2018 (274 citations); working group members’ personal bibliographies and searches (31 citations); and a search focusing on ultrasound-guided LP training (210 citations). A total of 232 full-text articles were reviewed, and the final selection included 77 articles that were abstracted into a data table and incorporated into the draft recommendations. Details of the literature search strategy are presented in Appendix 3.

RECOMMENDATIONS

Four domains (clinical outcomes, technique, training, and knowledge gaps) with 16 draft recommendations were generated based on a review of the literature. Selected references were abstracted and assigned to each draft recommendation. Rationales for each recommendation were drafted citing supporting evidence. After two rounds of panel voting, five recommendations did not achieve agreement based on the RAND rules, one recommendation was combined with another recommendation during peer review, and 10 statements received final approval. The degree of consensus based on the median score and the dispersion of voting around the median are shown in Appendix 5. Nine statements were approved as strong recommendations, and one was approved as a conditional recommendation. Therefore, the final recommendation count was 10. The strength of the recommendation and degree of consensus for each recommendation are summarized in Table 1.

Terminology

LP is a procedure in which a spinal needle is introduced into the subarachnoid space for the purpose of collecting CSF for diagnostic evaluation and/or therapeutic relief.

Throughout this document, the phrases “ultrasound-guided” and “ultrasound guidance” refer to the use of ultrasound to mark a needle insertion site immediately before performing the procedure. This is also known as static ultrasound guidance. Real-time or dynamic ultrasound guidance refers to direct visualization of the needle tip as it traverses through the skin and soft tissues to reach the ligamentum flavum. Any reference to real-time ultrasound guidance is explicitly stated.

Clinical outcomes

1) When ultrasound equipment is available, along with providers who are appropriately trained to use it, we recommend that ultrasound guidance should be used for site selection of LPs to reduce the number of needle insertion attempts and needle redirections and increase the overall procedure success rates, especially in patients who are obese or have difficult-to-palpate landmarks.

Rationale. LPs have historically been performed by selecting a needle insertion site based on palpation of anatomical landmarks. However, an estimated 30% of patients requiring LP in emergency departments have lumbar spine landmarks that are difficult to palpate, most commonly due to obesity.13 Furthermore, lumbar puncture performed based on palpation of landmarks alone has been reported to fail in 28% of patients.14

Ultrasound can be used at the bedside to elucidate the lumbar spine anatomy to guide performance of LP or epidural catheterization. Since the early 2000s, randomized studies comparing the use of ultrasound guidance (ultrasound-guided) versus anatomical landmarks (landmark-guided) to map the lumbar spine for epidural catheterization have emerged. It is important to recognize that the exact same ultrasound technique is used for site marking of LP, epidural catheterization, and spinal anesthesia—the key difference is how deep the needle tip is inserted. Therefore, data from these three ultrasound-guided procedures are often pooled. Currently, at least 33 randomized controlled studies comparing ultrasound-guided vs landmark-guided site selection for LP, epidural catheterization, or spinal anesthesia have been published.22-49 We present three meta-analyses below that pooled data primarily from randomized controlled studies comparing ultrasound-guided vs landmark-guided site selection for LP or spinal anesthesia.

In 2013, Shaikh et al. published the first meta-analysis with 14 randomized controlled studies comparing ultrasound-guided vs landmark-guided site selection for LP (n = 5) or epidural catheterization (n = 9). The pooled data showed that use of ultrasound guidance decreased the proportion of failed procedures (risk ratio 0.21, 95% CI 0.10-0.43) with an absolute risk reduction of 6.3% (95% CI 4.1%-8.4%) and a number needed to treat of 16 (95% CI 12-25) to prevent one failed procedure. In addition, the use of ultrasound reduced the mean number of attempts by 0.44 (95% CI 0.24-0.64) and reduced the mean number of needle redirections by 1.00 (95% CI 0.75-1.24). The reduction in risk of a failed procedure was similar for LPs (risk ratio 0.19 [95% CI 0.07-0.56]) and epidural catheterizations (risk ratio 0.23 [95% CI 0.09-0.60]).16

A similar meta-analysis published by Perlas et al. in 2016 included a total of 31 studies, both randomized controlled and cohort studies, evaluating the use of ultrasound guidance for LP, spinal anesthesia, and epidural catheterization.50 The goal of this systematic review and meta-analysis was to establish clinical practice recommendations. The authors concluded (1) the data consistently suggest that ultrasound is more accurate than palpation for lumbar interspace identification, (2) ultrasound allows accurate measurement of the needle insertion depth to reach the epidural space with a mean difference of <3 mm compared with the actual needle insertion depth, and (3) ultrasound increases the efficacy of lumbar epidural or spinal anesthesia by decreasing the mean number of needle passes for success by 0.75 (95% CI 0.44-1.07) and reducing the risk of a failed procedure (risk ratio 0.51 [95% CI 0.32-0.80]), both in patients with normal surface anatomy and in those with technically difficult surface anatomy due to obesity, scoliosis, or previous spine surgery.

Compared to the two earlier meta-analyses that included studies of both LP and spinal anesthesia procedures, the meta-analysis conducted by Gottlieb et al. in 2018 pooled data from 12 randomized controlled studies of ultrasound guidance for LPs only. For the primary outcome, pooled data from both adult and pediatric studies demonstrated higher procedural success rates with ultrasound-guided vs landmark-guided LPs (90% vs 81%) with an odds ratio of 2.1 (95% CI 0.66-7.44) in favor of ultrasound; however, there were no statistically significant differences when the adult and pediatric subgroups were analyzed separately, probably due to underpowering. For the secondary outcomes, data from the adult subgroup showed that use of ultrasound guidance was associated with fewer traumatic LPs (OR 0.28, 95% CI 0.14-0.59), shorter time to procedural success (adjusted mean difference –3.03 minutes, 95% CI –3.54 to –2.52), fewer number of needle passes (adjusted mean difference –0.81 passes, 95% CI –1.57 to –0.05), and lower patient pain scores (adjusted mean difference –2.53, 95% CI –3.89 to –1.17).

At least 12 randomized controlled studies have been published comparing the use of ultrasound guidance vs landmarks for the performance of LP or spinal anesthesia in adult patients, which were not included in the abovementioned meta-analyses. These individual studies demonstrated similar benefits of using ultrasound guidance: reduced needle insertion attempts, reduced needle redirections, and increased overall procedural success rates.17,31,37,40,41,43-49

It is important to recognize that four randomized controlled studies did not demonstrate any benefits of ultrasound guidance on the number of attempts or procedural success rates,23,33,41,51 and three of these studies were included in the abovementioned meta-analyses.23,33,51 Limitations of these negative studies include potential selection bias, inadequate sample sizes, and varying levels of operator skills in procedures, ultrasound guidance, or both. One study included emergency medicine residents as operators with varying degrees of ultrasound skills, and more importantly, patient enrollment occurred by convenience sampling, which may have introduced selection bias. Furthermore, most of the patients were not obese (median BMI of 27 kg/m2), and it is unclear why 10 years lapsed from data collection until publication.33 Another study with three experienced anesthesiologists as operators performing spinal anesthesia enrolled only patients who were not obese (mean BMI of 29 kg/m2) and had easily palpable bony landmarks—two patient characteristics associated with the least benefit of using ultrasound guidance in other studies.23 Another negative study had one experienced anesthesiologist marking obstetric patients with ultrasound, but junior residents performing the actual procedure in the absence of the anesthesiologist who had marked the patient.41

In general, the greatest benefit of using ultrasound guidance for LP has been demonstrated in obese patients.24,32,34,35,52,53 Benefits have been shown in specific obese patient populations, including obstetric,31,54,55 orthopedic,24,56,57 and emergency department patients.30

By increasing the procedural success rates with the use of ultrasound at the bedside, fewer patients may be referred to interventional radiology for fluoroscopic-guided LP, decreasing the patient exposure to ionizing radiation. A randomized study (n = 112) that compared site marking with ultrasound guidance versus fluoroscopic guidance for epidural steroid injections found the two techniques to be equivalent with respect to mean procedure time, number of needle insertion attempts, or needle passes.58 Another randomized study found that the performance time of ultrasound guidance was two minutes shorter (P < .05) than fluoroscopic guidance.59

 

 

Techniques

2) We recommend that ultrasound should be used to more accurately identify the lumbar spine level than physical examination in both obese and nonobese patients.

Rationale. Traditionally, an imaginary line connecting the iliac crests (intercristal line, Tuffier’s line, or Jacoby’s line) was considered to identify the L4 vertebra or the L4-L5 interspinous space in the midline; however, studies have revealed this traditional landmark to be much less accurate than previously thought. In general, palpating the iliac crests to mark the intercristal line identifies an interspinous space that is one space cephalad (ie, the L2-L3 interspinous space) but can range from L1-L2 to L4-L5.46,60-64 If an LP is inadvertently performed in the L1-L2 interspinous space, the risk of spinal cord injury is higher than that when performed in a more distal interspinous space.

A study by Margarido et al. with 45 patients with a mean BMI of 30 kg/m2 found that the intercristal line was located above the L4-L5 interspinous space in 100% of patients. More importantly, the intercristal line was above L2-L3 in 36% of patients and above L1-L2 in 4% of patients. It is important to note that patients with scoliosis or previous spine surgery were excluded from this study, and all examinations were performed by two experienced anesthesiologists with patients in a sitting position—all factors that would favor accurate palpation and marking of the iliac crests.60

In a study of nonobese patients (mean BMI 28 kg/m2) undergoing spinal anesthesia, Duniec et al. compared the lumbar level identified by palpation versus ultrasound and found discordance between the two techniques in 36% of patients; 18% were one space too cephalad, 16% were one space too caudal, and 2% were off by two interspinous spaces.61 Another study found discordance in 64% of patients (mean BMI 28 kg/m2) when comparing the interspinous level where spinal anesthesia had been performed by palpation versus a post-procedural ultrasound examination. This study revealed that the interspinous space was more cephalad in 50% of patients with 6% of punctures performed in the L1-L2 interspace.62 A similar study compared the accuracy of palpation vs ultrasound to identify the L3-L4 interspinous space in obese (mean BMI 34 kg/m2) versus nonobese (mean BMI 27 kg/m2) patients. This study found marking a space above L3-L4 in 51% of obese and 40% of nonobese patients and marking of the L1-L2 interspace in 7% of obese and 4% of nonobese patients.64

A study comparing palpation vs ultrasound found that 68% of obese patients with a BMI of >30 kg/m2 had difficult-to-palpate lumbar spine landmarks, but with the use of ultrasound, landmarks were identified in 76% of all patients, including obese and nonobese, with difficult-to-palpate landmarks.65

3) We suggest using ultrasound for selecting and marking a needle insertion site just before performing LPs in either a lateral decubitus or sitting position. The patient should remain in the same position after marking the needle insertion site.

Rationale. Ultrasound mapping of the lumbar spine can be performed in either a lateral decubitus or sitting position. Selecting and marking a needle insertion site should be performed at the bedside just before performing the procedure. The patient must remain in the same position in the interim between marking and inserting the needle, as a slight change in position can alter the needle trajectory, lowering the LP success rate. Although performing LPs in a lateral decubitus position has the advantage of accurately measuring the opening pressure, misalignment of the shoulder and pelvic girdles and bowing of the bed in a lateral decubitus position may lower LP success rates.

 

 

One randomized study comparing ultrasound-guided spinal anesthesia in a lateral decubitus versus sitting position found no difference in the number of needle insertion attempts or measurement of the skin-dura distance; however, the needle insertion depth was 0.73 cm greater in a lateral decubitus vs sitting position (P = .002).66 Procedural success rates of LP with ultrasound guidance have not been directly compared in a sitting versus lateral decubitus position, although the overall procedural success rates were higher in one study that allowed the operator to choose either sitting or lateral decubitus position when ultrasound was used.32

4) We recommend that a low-frequency transducer, preferably a curvilinear array transducer, should be used to evaluate the lumbar spine and mark a needle insertion site in most patients. A high-frequency linear array transducer may be used in nonobese patients.

Rationale. Low-frequency transducers emit sound waves that penetrate deep tissues, allowing visualization of bones and ligaments of the lumbar spine. A high-frequency linear transducer offers better resolution but shallower penetration to approximately 6-9 cm, limiting its use for site marking in overweight and obese patients. In obese patients, the ligamentum flavum is often deeper than 6 cm, which requires a low-frequency transducer to be visualized.

Most of the randomized controlled studies demonstrating benefits of using ultrasound guidance compared with landmark guidance for performance of LP, epidural anesthesia, or spinal anesthesia have used a low-frequency, curvilinear transducer.22,24,26-28,31,34-36,39,43-45,67 Two randomized controlled trials used a high-frequency linear transducer for site marking of lumbar procedures.30,32,37 Using a high-frequency linear transducer has been described in real-time, ultrasound-guided LPs, the advantage being better needle visualization with a linear transducer.29 Detection of blood vessels by color flow Doppler may be another advantage of using a high-frequency linear transducer, although a study by Grau et al. showed that use of color flow Doppler with a low-frequency curvilinear transducer permitted visualization of interspinous vessels as small as 0.5 mm in size.68

5) We recommend that ultrasound should be used to map the lumbar spine, starting at the level of the sacrum and sliding the transducer cephalad, sequentially identifying the lumbar spine interspaces.Rationale. Although no studies have directly compared different ultrasound scanning protocols to map the lumbar spine, starting at the level of the sacrum and sliding the transducer cephalad to sequentially identify the lumbar interspinous spaces is the most commonly described technique in studies demonstrating improved clinical outcomes with the use of ultrasound.24,31,34,37,39,40,45,56,57,67 Because the sacrum can be easily recognized, identifying it first is most beneficial in patients with few or no palpable landmarks.

All five lumbar spinous processes and interspinous spaces can be mapped from the sacrum using either a midline or a paramedian approach, and the widest interspinous space can be selected. In a midline approach, either a transverse or a longitudinal view is obtained. The transducer is centered on the sacrum and slid cephalad from L5 to L1 to identify each spinous process and interspinous space. In a paramedian approach, longitudinal paramedian views are obtained from the L5–sacrum interspace to the L1–L2 interspace, and each interspinous space is identified as the transducer is slid cephalad. Both these approaches are effective for mapping the lumbar spine. Whether the entire lumbar spine is mapped, and whether a midline or a paramedian approach is utilized, will depend on the operator’s preference.

 

 

6) We recommend that ultrasound should be used in a transverse plane to mark the midline of the lumbar spine and a longitudinal plane to mark the interspinous spaces. The intersection of these two lines marks the needle insertion site.

Rationale. The most common technique described in comparative studies of ultrasound vs landmarks includes visualization of the lumbar spine in two planes, a transverse plane to identify the midline and a longitudinal plane to identify the interspinous spaces. The majority of randomized controlled studies that demonstrated a reduction in the number of needle insertion attempts and an increase in the procedural success rates have used this technique (see Clinical Outcomes).22,24,28,32,35-37,43,44 Marking the midline and interspinous space(s) for LP may be performed in any order, starting with either the transverse or longitudinal plane first.

The midline of the spine is marked by placing the transducer in a transverse plane over the lumbar spine, centering over the spinous processes that have a distinct hyperechoic tip and a prominent acoustic shadow deep to the bone, and drawing a line perpendicular to the center of the transducer delineating the midline. The midline should be marked over a minimum of two or three spinous processes.

To identify the interspinous spaces, the transducer is aligned longitudinally over the midline. The transducer is slid along the midline to identify the widest interspinous space. Once the transducer is centered over the widest interspinous space, a line perpendicular to the center of the transducer is drawn to mark the interspinous space. The intersection of the lines marking the spinal midline and the selected interspinous space identifies the needle entry point.

To visualize the ligamentum flavum from a paramedian view, the transducer is oriented longitudinally over the midline, slid approximately 1 cm laterally, and tilted approximately 15 degrees aiming the ultrasound beam toward the midline. The skin–ligamentum flavum distance is most reliably measured from a paramedian view. Alternatively, in some patients, the ligamentum flavum may be visualized in the midline and the depth can be measured.

7) We recommend that ultrasound should be used during a preprocedural evaluation to measure the distance from the skin surface to the ligamentum flavum from a longitudinal paramedian view to estimate the needle insertion depth and ensure that a spinal needle of adequate length is used.

Rationale. The distance from the skin to the ligamentum flavum can be measured using ultrasound during preprocedural planning. Knowing the depth to the ligamentum flavum preprocedurally allows the operator to procure a spinal needle of adequate length, anticipate the insertion depth before CSF can be obtained, determine the depth to which a local anesthetic will need to be injected, and decide whether the anticipated difficulty of the procedure warrants referral to or consultation with another specialist.

The skin–ligamentum flavum distance can be measured from a transverse midline view or a longitudinal paramedian view. A longitudinal paramedian view provides an unobstructed view of the ligamentum flavum due to less shadowing from bony structures compared with a midline view. Several studies have demonstrated a strong correlation between the skin–ligamentum flavum distance measured by ultrasound and the actual needle insertion depth in both midline and paramedian views.28,34,36,53,54,57,69,70

A meta-analysis that included 13 comparative studies evaluating the correlation between ultrasound-measured depth and actual needle insertion depth to reach the epidural or intrathecal space consistently demonstrated a strong correlation between the measured and actual depth.50 A few studies have reported near-perfect Pearson correlation coefficients of 0.98.55,71,72 The pooled correlation was 0.91 (95% CI 0.87-0.94). All studies measured the depth from the skin to the ventral side of the ligamentum flavum or the intrathecal space from either a longitudinal paramedian view (n = 4) or a transverse midline view (n = 9). Eight of the more recent studies evaluated the accuracy of the ultrasound measurements and found the depth measurements by ultrasound to be accurate within 1-13 mm of the actual needle insertion depth, with seven of the eight studies reporting a mean difference of ≤3 mm.50

Measurement of the distance between the skin and the ligamentum flavum generally underestimates the needle insertion depth. One study reported that measurement of the skin–ligamentum flavum distance underestimates the needle insertion depth by 7.6 mm to obtain CSF, whereas measurement of the skin–posterior longitudinal ligament distance overestimates the needle insertion depth by 2.5 mm.57 A well-accepted contributor to underestimation of the depth measurements using ultrasound is compression of the skin and soft tissues by the transducer, and therefore, pressure on the skin must be released before freezing an image and measuring the depth to the subarachnoid space.

 

 

Training

8) We recommend that novices should undergo simulation-based training, where available, before attempting ultrasound-guided LPs on actual patients.

Rationale. Similar to training for other bedside procedures, dedicated training sessions, including didactics, supervised practice on patients, and simulation-based practice, should be considered when teaching novices to perform ultrasound-guided LP. Simulation-based training facilitates acquisition of knowledge and skills to perform invasive bedside procedures, including LP.73 Simulation-based training has been commonly incorporated into procedure training for trainees using an immersive experience, such as a “boot camp,”74-77 or a standardized curriculum,78,79 and has demonstrated improvements in post-course procedural knowledge, technical skills, and operator confidence. Two of these studies included training in the use of ultrasound guidance for LP. These studies showed that simulation-based practice improved skill acquisition and confidence.80,81 Simulation using novel computer software may improve skill acquisition in the use of ultrasound guidance for LP.82

9) We recommend that training in ultrasound-guided LPs should be adapted based on prior ultrasound experience, as learning curves will vary.Rationale. The learning curve to achieve competency in the use of ultrasound guidance for LP has not been well studied. The rate of attaining competency in identifying lumbar spine structures using ultrasound will vary by provider based on prior skills in ultrasound-guided procedures.83 Thus, providers with prior ultrasound experience may require less training than those without such experience to achieve competency. However, extensive experience in performing landmark-guided LPs does not necessarily translate into rapid acquisition of skills to perform the procedure with ultrasound guidance. A study of practicing anesthesiologists with no prior ultrasound experience demonstrated that 20 supervised trials of ultrasound-guided spinal anesthesia were insufficient to achieve competency.84 Although minimums may be a necessary step to gain competence, using them as a sole means to define competence does not account for variable learning curves.12 Based on a national survey of 21 hospitalist procedure experts, the mean current vs suggested minimums for initial and ongoing hospital privileging for LPs were 1.8 vs 6.9 and 2.2 vs 4.6 annually in one report.85

A fundamental question that needs to be answered is how to define competency in the use of ultrasound guidance for LP, including the specific skills and knowledge that must be mastered. At a minimum, providers must be able to identify lumbar spinous processes and distinguish them from the sacrum, identify the lumbar interspinous spaces and their corresponding levels, and estimate the depth from the skin to the ligamentum flavum from the midline and paramedian planes. Novice operators may benefit from practicing lumbar spine mapping of nonobese patients using a high-frequency linear transducer that generates high-resolution images and facilitates recognition of lumbar spine structures.

10) We recommend that novice providers should be supervised when performing ultrasound-guided LPs before performing the procedure independently on patients.

Rationale: Demonstration of competency in the use of ultrasound to identify lumbar spine anatomy should be achieved before routinely performing the procedure independently on patients.18 All providers will require a variable period of supervised practice to demonstrate the appropriate technique, followed by a period of unsupervised practice before competency is achieved. Supervised practice with guidance and feedback has been shown to significantly improve providers’ ability to delineate lumbar spine anatomy.86

 

 

KNOWLEDGE GAPS

The process of producing these guidelines revealed areas of uncertainty and important gaps in the literature regarding the use of ultrasound guidance for LP.

First, it is unclear whether the use of ultrasound guidance for LP reduces postprocedural back pain and whether it improves patient satisfaction. Several studies have evaluated postprocedural back pain28,30,32,33,52 and patient satisfaction28,29,33,51 with the use of ultrasound guidance, but these studies have found inconsistent results. Some of these results were probably due to insufficient statistical power or confounding variables. Furthermore, benefits have been demonstrated in certain subgroups, such as overweight patients or those with anatomical abnormalities, as was found in two studies.52,87 Use of ultrasound guidance for spinal anesthesia has been shown to reduce postprocedural headache28 and improve patient satisfaction51, although similar benefit has not been demonstrated in patients undergoing LP.

Second, the effect of using ultrasound guidance on the frequency of traumatic LPs is an area of uncertainty. A “traumatic tap” is defined as an inadvertent puncture of an epidural vein during passage of the spinal needle through the dura. It remains difficult to discern in these studies whether red blood cells detected in the CSF resulted from puncture of an epidural vein or from needle trauma of the skin and soft tissues. Despite this uncertainty, at least seven randomized controlled studies have assessed the effect of ultrasound guidance on traumatic LPs. The meta-analysis by Shaikh et al. included five randomized controlled studies that assessed the effect of ultrasound guidance on the reporting of traumatic taps. The study found a reduced risk of traumatic taps (risk ratio 0.27 [95% CI 0.11-0.67]), an absolute risk reduction of 5.9% (95% CI 2.3%-9.5%), and a number needed to treat of 17 (95% CI 11-44) to prevent one traumatic tap.16 Similarly, the meta-analysis by Gottlieb et al. showed a lower risk of traumatic taps among adults undergoing LP with ultrasound guidance in five randomized controlled studies with an odds ratio of 0.28 (95% CI 0.14-0.59). The meta-analysis by Gottlieb et al. included two adult studies that were not included by Shaikh et al.

Third, several important questions about the technique of ultrasound-guided LP remain unanswered. In addition to the static technique, a dynamic technique with real-time needle tracking has been described to perform ultrasound-guided LP, epidural catheterization, and spinal anesthesia. A pilot study by Grau et al. found that ultrasound used either statically or dynamically had fewer insertion attempts and needle redirections than use of landmarks alone.29 Three other pilot studies showed successful spinal anesthesia in almost all patients88-90 and one large study demonstrated successful spinal anesthesia with real-time ultrasound guidance in 97 of 100 patients with a median of three needle passes.91 Furthermore, a few industry-sponsored studies with small numbers of patients have described the use of novel needle tracking systems that facilitate needle visualization during real-time ultrasound-guided LP.92,93 However, to our knowledge, no comparative studies of static versus dynamic guidance using novel needle tracking systems in human subjects have been published, and any potential role for these novel needle tracking systems has not yet been defined.

Finally, the effects of using ultrasound guidance on clinical decision-making, timeliness, and cost-effectiveness of LP have not yet been explored but could have important clinical practice implications.

 

 

CONCLUSION

Randomized controlled trials have demonstrated that using ultrasound guidance for LPs can reduce the number of needle insertion attempts and needle redirections and increase the overall procedural success rates. Ultrasound can more accurately identify the lumbar spine level than physical examination in both obese and nonobese patients, although the greatest benefit of using ultrasound guidance for LPs has been shown in obese patients.

Ultrasound permits assessment of the interspinous space width and measurement of the ligamentum flavum depth to select an optimal needle insertion site and adequate length spinal needle. Although the use of real-time ultrasound guidance has been described, the use of static ultrasound guidance for LP site marking remains the standard technique.

Acknowledgments

The authors thank all the members of the Society of Hospital Medicine Point-of-care Ultrasound Task Force and the Education Committee members for their time and dedication to develop these guidelines.

Collaborators from Society of Hospital Medicine Point-of-care Ultrasound Task Force: Saaid Abdel-Ghani, Robert Arntfield, Jeffrey Bates, Anjali Bhagra, Michael Blaivas, Daniel Brotman, Carolina Candotti, Richard Hoppmann, Susan Hunt, Trevor P. Jensen, Paul Mayo, Benji Mathews, Satyen Nichani, Vicki Noble, Martin Perez, Nitin Puri, Aliaksei Pustavoitau, Kreegan Reierson, Sophia Rodgers, Kirk Spencer, Vivek Tayal, David Tierney

SHM Point-of-care Ultrasound Task Force: CHAIRS: Nilam Soni, Ricardo Franco-Sadud, Jeff Bates. WORKING GROUPS: Thoracentesis Working Group: Ria Dancel (chair), Daniel Schnobrich, Nitin Puri. Vascular Access Working Group: Ricardo Franco (chair), Benji Matthews, Saaid Abdel-Ghani, Sophia Rodgers, Martin Perez, Daniel Schnobrich. Paracentesis Working Group: Joel Cho (chair), Benji Matthews, Kreegan Reierson, Anjali Bhagra, Trevor P. Jensen Lumbar Puncture Working Group: Nilam J. Soni (chair), Ricardo Franco, Gerard Salame, Josh Lenchus, Venkat Kalidindi, Ketino Kobaidze. Credentialing Working Group: Brian P Lucas (chair), David Tierney, Trevor P. Jensen PEER REVIEWERS: Robert Arntfield, Michael Blaivas, Richard Hoppmann, Paul Mayo, Vicki Noble, Aliaksei Pustavoitau, Kirk Spencer, Vivek Tayal. METHODOLOGIST: Mahmoud El Barbary. LIBRARIAN: Loretta Grikis. SOCIETY OF HOSPITAL MEDICINE EDUCATION COMMITTEE: Daniel Brotman (past chair), Satyen Nichani (current chair), Susan Hunt. SOCIETY OF HOSPITAL MEDICINE STAFF: Nick Marzano.

Disclosures

The authors have nothing to disclose.

Funding

Brian P Lucas: Department of Veterans Affairs, Veterans Health Administration, Office of Research and Development and Dartmouth SYNERGY, National Institutes of Health, National Center for Translational Science (UL1TR001086). Nilam Soni: Department of Veterans Affairs, Quality Enhancement Research Initiative (QUERI) Partnered Evaluation Initiative Grant (HX002263-01A1).

Disclaimer

The contents of this publication do not represent the views of the U.S. Department of Veterans Affairs or the United States Government.

 

Approximately 400,000 lumbar punctures (LPs) are performed in the United States annually for either diagnostic workup or therapeutic relief.1 Lumbar punctures are increasingly being performed in the United States, with an estimated 97,000 LPs performed on Medicare fee-for-service beneficiaries in 2011 alone, which is an increase of approximately 4,000 LPs in the same population from 1991.2 Approximately 273,612 LPs were performed on hospitalized patients in the United States in 2010,1 and the inpatient hospital setting is the most common site for LPs.2,3

Many LPs are referred to radiologists who have access to imaging guidance to aid with needle insertion.2 However, referrals to radiology delay performance of LPs, and delayed diagnosis of acute bacterial meningitis, the most common yet serious condition for which LPs are performed, is associated with increased morbidity and mortality.4-8 Furthermore, although initiating empiric antibiotic treatment for suspected acute bacterial meningitis is recommended in some cases, doing so routinely can cause false-negative cerebrospinal fluid (CSF) culture results, complicating decisions about de-escalation and duration of antibiotics that could have been safely avoided by promptly performing an LP.9

Delaying the performance of LP has been associated with increased mortality.10 Demonstration of proficiency in performance of lumbar puncture is considered a core competency for hospitalists,11 and with the increasing availability of point-of-care ultrasound, hospitalists can use ultrasound to guide performance of LPs at the bedside.12 However, 30% of patients requiring LP in emergency departments have difficult-to-palpate lumbar spine landmarks,13 and lumbar puncture performed based on palpation of landmarks alone has been reported to fail or be traumatic in 28% of patients.14 Use of ultrasound guidance for lumbar puncture has been shown in randomized controlled trials to improve procedural success rates, while reducing the time to successful LP, needle passes, patient pain scores, and risk of a traumatic LP.15-17

The purpose of this position statement is to review the literature and present consensus-based recommendations on the performance of ultrasound-guided LP in adult patients. This position statement does not mandate that hospitalists use ultrasound guidance for LP, nor does it establish ultrasound guidance as the standard of care for LP. Similar to previously published Society of Hospital Medicine (SHM) position statements,12,18,19 this document presents recommendations with supporting evidence for the clinical outcomes, techniques, and training for using ultrasound guidance for LP. A manuscript describing the technique of ultrasound guidance for LPs has been previously published by some of the authors of this position statement.20

 

 

METHODS

Detailed methods are described in Appendix 1. The SHM Point-of-care Ultrasound (POCUS) Task Force was assembled to carry out this guideline development project under the direction of the SHM Board of Directors, Director of Education, and Education Committee. All expert panel members were physicians or advanced practice providers with expertise in POCUS. Expert panel members were divided into working group members, external peer reviewers, and a methodologist. All Task Force members were required to disclose any potential conflicts of interests (Appendix 2). The literature search was conducted in two independent phases. The first phase included literature searches conducted by the six working group members themselves. Key clinical questions and draft recommendations were then prepared. A systematic literature search was conducted by a medical librarian based on the findings of the initial literature search and draft recommendations. The Medline, Embase, CINAHL, and Cochrane medical databases were searched from 1975 to December 2015 initially. Google Scholar was also searched without limiters. Updated searches were conducted in November 2016, January 2018, and October 2018. The search strings are included in Appendix 3. All article abstracts were first screened for relevance by at least two members of the working group. Full-text versions of screened articles were reviewed, and articles on the use of ultrasound to guide LP were selected. In addition, the following article types were excluded: non-English language, nonhuman, age <18 years, meeting abstracts, meeting posters, narrative reviews, case reports, letters, and editorials. Moreover, studies focusing on the use of ultrasound guidance for spinal nerve root injections, regional anesthesia, and assessment of lumbar spine anatomy alone were excluded. All relevant systematic reviews, meta-analyses, randomized controlled trials, and observational studies of ultrasound-guided LP were screened and selected. Final article selection was based on working group consensus, and the selected literature was incorporated into the draft recommendations.

The Research and Development (RAND) Appropriateness Method that required panel judgment and consensus was used.21 The 27 voting members of the SHM POCUS Task Force reviewed and voted on the draft recommendations considering the following five transforming factors: (1) Problem priority and importance, (2) Level of quality of evidence, (3) Benefit/harm balance, (4) Benefit/burden balance, and (5) Certainty/concerns about PEAF (Preferences/Equity/Acceptability/Feasibility). Panel members participated in two rounds of electronic voting using an internet-based electronic data collection tool (REDCap™) in February 2018 and April 2018 (Appendix 4). Voting on appropriateness was conducted using a 9-point Likert scale. The three zones of the 9-point Likert scale were inappropriate (1-3 points), uncertain (4-6 points), and appropriate (7-9 points). The degree of consensus was assessed using the RAND algorithm (Appendix Figure 1 and Table 1). Establishing a recommendation required at least 70% agreement that a recommendation was “appropriate.” A strong recommendation required 80% of the votes within one integer of the median, following the RAND rules. Disagreement was defined as >30% of panelists voting outside of the zone of the median.

Recommendations were classified as strong or weak/conditional based on preset rules defining the panel’s level of consensus, which determined the wording of each recommendation (Table 2). The revised consensus-based recommendations underwent internal and external reviews by POCUS experts from different subspecialties. The final review of this position statement was performed by members of the SHM POCUS Task Force, SHM Education Committee, and SHM Executive Committee. The SHM Executive Committee endorsed this position statement in June 2018 before submission to the Journal of Hospital Medicine.

 

 

RESULTS

Literature Search

A total of 4,389 references were pooled from four different sources: a search by a certified medical librarian in December 2015 (3,212 citations) that was updated in November 2016 (380 citations), January 2018 (282 citations), and October 2018 (274 citations); working group members’ personal bibliographies and searches (31 citations); and a search focusing on ultrasound-guided LP training (210 citations). A total of 232 full-text articles were reviewed, and the final selection included 77 articles that were abstracted into a data table and incorporated into the draft recommendations. Details of the literature search strategy are presented in Appendix 3.

RECOMMENDATIONS

Four domains (clinical outcomes, technique, training, and knowledge gaps) with 16 draft recommendations were generated based on a review of the literature. Selected references were abstracted and assigned to each draft recommendation. Rationales for each recommendation were drafted citing supporting evidence. After two rounds of panel voting, five recommendations did not achieve agreement based on the RAND rules, one recommendation was combined with another recommendation during peer review, and 10 statements received final approval. The degree of consensus based on the median score and the dispersion of voting around the median are shown in Appendix 5. Nine statements were approved as strong recommendations, and one was approved as a conditional recommendation. Therefore, the final recommendation count was 10. The strength of the recommendation and degree of consensus for each recommendation are summarized in Table 1.

Terminology

LP is a procedure in which a spinal needle is introduced into the subarachnoid space for the purpose of collecting CSF for diagnostic evaluation and/or therapeutic relief.

Throughout this document, the phrases “ultrasound-guided” and “ultrasound guidance” refer to the use of ultrasound to mark a needle insertion site immediately before performing the procedure. This is also known as static ultrasound guidance. Real-time or dynamic ultrasound guidance refers to direct visualization of the needle tip as it traverses through the skin and soft tissues to reach the ligamentum flavum. Any reference to real-time ultrasound guidance is explicitly stated.

Clinical outcomes

1) When ultrasound equipment is available, along with providers who are appropriately trained to use it, we recommend that ultrasound guidance should be used for site selection of LPs to reduce the number of needle insertion attempts and needle redirections and increase the overall procedure success rates, especially in patients who are obese or have difficult-to-palpate landmarks.

Rationale. LPs have historically been performed by selecting a needle insertion site based on palpation of anatomical landmarks. However, an estimated 30% of patients requiring LP in emergency departments have lumbar spine landmarks that are difficult to palpate, most commonly due to obesity.13 Furthermore, lumbar puncture performed based on palpation of landmarks alone has been reported to fail in 28% of patients.14

Ultrasound can be used at the bedside to elucidate the lumbar spine anatomy to guide performance of LP or epidural catheterization. Since the early 2000s, randomized studies comparing the use of ultrasound guidance (ultrasound-guided) versus anatomical landmarks (landmark-guided) to map the lumbar spine for epidural catheterization have emerged. It is important to recognize that the exact same ultrasound technique is used for site marking of LP, epidural catheterization, and spinal anesthesia—the key difference is how deep the needle tip is inserted. Therefore, data from these three ultrasound-guided procedures are often pooled. Currently, at least 33 randomized controlled studies comparing ultrasound-guided vs landmark-guided site selection for LP, epidural catheterization, or spinal anesthesia have been published.22-49 We present three meta-analyses below that pooled data primarily from randomized controlled studies comparing ultrasound-guided vs landmark-guided site selection for LP or spinal anesthesia.

In 2013, Shaikh et al. published the first meta-analysis with 14 randomized controlled studies comparing ultrasound-guided vs landmark-guided site selection for LP (n = 5) or epidural catheterization (n = 9). The pooled data showed that use of ultrasound guidance decreased the proportion of failed procedures (risk ratio 0.21, 95% CI 0.10-0.43) with an absolute risk reduction of 6.3% (95% CI 4.1%-8.4%) and a number needed to treat of 16 (95% CI 12-25) to prevent one failed procedure. In addition, the use of ultrasound reduced the mean number of attempts by 0.44 (95% CI 0.24-0.64) and reduced the mean number of needle redirections by 1.00 (95% CI 0.75-1.24). The reduction in risk of a failed procedure was similar for LPs (risk ratio 0.19 [95% CI 0.07-0.56]) and epidural catheterizations (risk ratio 0.23 [95% CI 0.09-0.60]).16

A similar meta-analysis published by Perlas et al. in 2016 included a total of 31 studies, both randomized controlled and cohort studies, evaluating the use of ultrasound guidance for LP, spinal anesthesia, and epidural catheterization.50 The goal of this systematic review and meta-analysis was to establish clinical practice recommendations. The authors concluded (1) the data consistently suggest that ultrasound is more accurate than palpation for lumbar interspace identification, (2) ultrasound allows accurate measurement of the needle insertion depth to reach the epidural space with a mean difference of <3 mm compared with the actual needle insertion depth, and (3) ultrasound increases the efficacy of lumbar epidural or spinal anesthesia by decreasing the mean number of needle passes for success by 0.75 (95% CI 0.44-1.07) and reducing the risk of a failed procedure (risk ratio 0.51 [95% CI 0.32-0.80]), both in patients with normal surface anatomy and in those with technically difficult surface anatomy due to obesity, scoliosis, or previous spine surgery.

Compared to the two earlier meta-analyses that included studies of both LP and spinal anesthesia procedures, the meta-analysis conducted by Gottlieb et al. in 2018 pooled data from 12 randomized controlled studies of ultrasound guidance for LPs only. For the primary outcome, pooled data from both adult and pediatric studies demonstrated higher procedural success rates with ultrasound-guided vs landmark-guided LPs (90% vs 81%) with an odds ratio of 2.1 (95% CI 0.66-7.44) in favor of ultrasound; however, there were no statistically significant differences when the adult and pediatric subgroups were analyzed separately, probably due to underpowering. For the secondary outcomes, data from the adult subgroup showed that use of ultrasound guidance was associated with fewer traumatic LPs (OR 0.28, 95% CI 0.14-0.59), shorter time to procedural success (adjusted mean difference –3.03 minutes, 95% CI –3.54 to –2.52), fewer number of needle passes (adjusted mean difference –0.81 passes, 95% CI –1.57 to –0.05), and lower patient pain scores (adjusted mean difference –2.53, 95% CI –3.89 to –1.17).

At least 12 randomized controlled studies have been published comparing the use of ultrasound guidance vs landmarks for the performance of LP or spinal anesthesia in adult patients, which were not included in the abovementioned meta-analyses. These individual studies demonstrated similar benefits of using ultrasound guidance: reduced needle insertion attempts, reduced needle redirections, and increased overall procedural success rates.17,31,37,40,41,43-49

It is important to recognize that four randomized controlled studies did not demonstrate any benefits of ultrasound guidance on the number of attempts or procedural success rates,23,33,41,51 and three of these studies were included in the abovementioned meta-analyses.23,33,51 Limitations of these negative studies include potential selection bias, inadequate sample sizes, and varying levels of operator skills in procedures, ultrasound guidance, or both. One study included emergency medicine residents as operators with varying degrees of ultrasound skills, and more importantly, patient enrollment occurred by convenience sampling, which may have introduced selection bias. Furthermore, most of the patients were not obese (median BMI of 27 kg/m2), and it is unclear why 10 years lapsed from data collection until publication.33 Another study with three experienced anesthesiologists as operators performing spinal anesthesia enrolled only patients who were not obese (mean BMI of 29 kg/m2) and had easily palpable bony landmarks—two patient characteristics associated with the least benefit of using ultrasound guidance in other studies.23 Another negative study had one experienced anesthesiologist marking obstetric patients with ultrasound, but junior residents performing the actual procedure in the absence of the anesthesiologist who had marked the patient.41

In general, the greatest benefit of using ultrasound guidance for LP has been demonstrated in obese patients.24,32,34,35,52,53 Benefits have been shown in specific obese patient populations, including obstetric,31,54,55 orthopedic,24,56,57 and emergency department patients.30

By increasing the procedural success rates with the use of ultrasound at the bedside, fewer patients may be referred to interventional radiology for fluoroscopic-guided LP, decreasing the patient exposure to ionizing radiation. A randomized study (n = 112) that compared site marking with ultrasound guidance versus fluoroscopic guidance for epidural steroid injections found the two techniques to be equivalent with respect to mean procedure time, number of needle insertion attempts, or needle passes.58 Another randomized study found that the performance time of ultrasound guidance was two minutes shorter (P < .05) than fluoroscopic guidance.59

 

 

Techniques

2) We recommend that ultrasound should be used to more accurately identify the lumbar spine level than physical examination in both obese and nonobese patients.

Rationale. Traditionally, an imaginary line connecting the iliac crests (intercristal line, Tuffier’s line, or Jacoby’s line) was considered to identify the L4 vertebra or the L4-L5 interspinous space in the midline; however, studies have revealed this traditional landmark to be much less accurate than previously thought. In general, palpating the iliac crests to mark the intercristal line identifies an interspinous space that is one space cephalad (ie, the L2-L3 interspinous space) but can range from L1-L2 to L4-L5.46,60-64 If an LP is inadvertently performed in the L1-L2 interspinous space, the risk of spinal cord injury is higher than that when performed in a more distal interspinous space.

A study by Margarido et al. with 45 patients with a mean BMI of 30 kg/m2 found that the intercristal line was located above the L4-L5 interspinous space in 100% of patients. More importantly, the intercristal line was above L2-L3 in 36% of patients and above L1-L2 in 4% of patients. It is important to note that patients with scoliosis or previous spine surgery were excluded from this study, and all examinations were performed by two experienced anesthesiologists with patients in a sitting position—all factors that would favor accurate palpation and marking of the iliac crests.60

In a study of nonobese patients (mean BMI 28 kg/m2) undergoing spinal anesthesia, Duniec et al. compared the lumbar level identified by palpation versus ultrasound and found discordance between the two techniques in 36% of patients; 18% were one space too cephalad, 16% were one space too caudal, and 2% were off by two interspinous spaces.61 Another study found discordance in 64% of patients (mean BMI 28 kg/m2) when comparing the interspinous level where spinal anesthesia had been performed by palpation versus a post-procedural ultrasound examination. This study revealed that the interspinous space was more cephalad in 50% of patients with 6% of punctures performed in the L1-L2 interspace.62 A similar study compared the accuracy of palpation vs ultrasound to identify the L3-L4 interspinous space in obese (mean BMI 34 kg/m2) versus nonobese (mean BMI 27 kg/m2) patients. This study found marking a space above L3-L4 in 51% of obese and 40% of nonobese patients and marking of the L1-L2 interspace in 7% of obese and 4% of nonobese patients.64

A study comparing palpation vs ultrasound found that 68% of obese patients with a BMI of >30 kg/m2 had difficult-to-palpate lumbar spine landmarks, but with the use of ultrasound, landmarks were identified in 76% of all patients, including obese and nonobese, with difficult-to-palpate landmarks.65

3) We suggest using ultrasound for selecting and marking a needle insertion site just before performing LPs in either a lateral decubitus or sitting position. The patient should remain in the same position after marking the needle insertion site.

Rationale. Ultrasound mapping of the lumbar spine can be performed in either a lateral decubitus or sitting position. Selecting and marking a needle insertion site should be performed at the bedside just before performing the procedure. The patient must remain in the same position in the interim between marking and inserting the needle, as a slight change in position can alter the needle trajectory, lowering the LP success rate. Although performing LPs in a lateral decubitus position has the advantage of accurately measuring the opening pressure, misalignment of the shoulder and pelvic girdles and bowing of the bed in a lateral decubitus position may lower LP success rates.

 

 

One randomized study comparing ultrasound-guided spinal anesthesia in a lateral decubitus versus sitting position found no difference in the number of needle insertion attempts or measurement of the skin-dura distance; however, the needle insertion depth was 0.73 cm greater in a lateral decubitus vs sitting position (P = .002).66 Procedural success rates of LP with ultrasound guidance have not been directly compared in a sitting versus lateral decubitus position, although the overall procedural success rates were higher in one study that allowed the operator to choose either sitting or lateral decubitus position when ultrasound was used.32

4) We recommend that a low-frequency transducer, preferably a curvilinear array transducer, should be used to evaluate the lumbar spine and mark a needle insertion site in most patients. A high-frequency linear array transducer may be used in nonobese patients.

Rationale. Low-frequency transducers emit sound waves that penetrate deep tissues, allowing visualization of bones and ligaments of the lumbar spine. A high-frequency linear transducer offers better resolution but shallower penetration to approximately 6-9 cm, limiting its use for site marking in overweight and obese patients. In obese patients, the ligamentum flavum is often deeper than 6 cm, which requires a low-frequency transducer to be visualized.

Most of the randomized controlled studies demonstrating benefits of using ultrasound guidance compared with landmark guidance for performance of LP, epidural anesthesia, or spinal anesthesia have used a low-frequency, curvilinear transducer.22,24,26-28,31,34-36,39,43-45,67 Two randomized controlled trials used a high-frequency linear transducer for site marking of lumbar procedures.30,32,37 Using a high-frequency linear transducer has been described in real-time, ultrasound-guided LPs, the advantage being better needle visualization with a linear transducer.29 Detection of blood vessels by color flow Doppler may be another advantage of using a high-frequency linear transducer, although a study by Grau et al. showed that use of color flow Doppler with a low-frequency curvilinear transducer permitted visualization of interspinous vessels as small as 0.5 mm in size.68

5) We recommend that ultrasound should be used to map the lumbar spine, starting at the level of the sacrum and sliding the transducer cephalad, sequentially identifying the lumbar spine interspaces.Rationale. Although no studies have directly compared different ultrasound scanning protocols to map the lumbar spine, starting at the level of the sacrum and sliding the transducer cephalad to sequentially identify the lumbar interspinous spaces is the most commonly described technique in studies demonstrating improved clinical outcomes with the use of ultrasound.24,31,34,37,39,40,45,56,57,67 Because the sacrum can be easily recognized, identifying it first is most beneficial in patients with few or no palpable landmarks.

All five lumbar spinous processes and interspinous spaces can be mapped from the sacrum using either a midline or a paramedian approach, and the widest interspinous space can be selected. In a midline approach, either a transverse or a longitudinal view is obtained. The transducer is centered on the sacrum and slid cephalad from L5 to L1 to identify each spinous process and interspinous space. In a paramedian approach, longitudinal paramedian views are obtained from the L5–sacrum interspace to the L1–L2 interspace, and each interspinous space is identified as the transducer is slid cephalad. Both these approaches are effective for mapping the lumbar spine. Whether the entire lumbar spine is mapped, and whether a midline or a paramedian approach is utilized, will depend on the operator’s preference.

 

 

6) We recommend that ultrasound should be used in a transverse plane to mark the midline of the lumbar spine and a longitudinal plane to mark the interspinous spaces. The intersection of these two lines marks the needle insertion site.

Rationale. The most common technique described in comparative studies of ultrasound vs landmarks includes visualization of the lumbar spine in two planes, a transverse plane to identify the midline and a longitudinal plane to identify the interspinous spaces. The majority of randomized controlled studies that demonstrated a reduction in the number of needle insertion attempts and an increase in the procedural success rates have used this technique (see Clinical Outcomes).22,24,28,32,35-37,43,44 Marking the midline and interspinous space(s) for LP may be performed in any order, starting with either the transverse or longitudinal plane first.

The midline of the spine is marked by placing the transducer in a transverse plane over the lumbar spine, centering over the spinous processes that have a distinct hyperechoic tip and a prominent acoustic shadow deep to the bone, and drawing a line perpendicular to the center of the transducer delineating the midline. The midline should be marked over a minimum of two or three spinous processes.

To identify the interspinous spaces, the transducer is aligned longitudinally over the midline. The transducer is slid along the midline to identify the widest interspinous space. Once the transducer is centered over the widest interspinous space, a line perpendicular to the center of the transducer is drawn to mark the interspinous space. The intersection of the lines marking the spinal midline and the selected interspinous space identifies the needle entry point.

To visualize the ligamentum flavum from a paramedian view, the transducer is oriented longitudinally over the midline, slid approximately 1 cm laterally, and tilted approximately 15 degrees aiming the ultrasound beam toward the midline. The skin–ligamentum flavum distance is most reliably measured from a paramedian view. Alternatively, in some patients, the ligamentum flavum may be visualized in the midline and the depth can be measured.

7) We recommend that ultrasound should be used during a preprocedural evaluation to measure the distance from the skin surface to the ligamentum flavum from a longitudinal paramedian view to estimate the needle insertion depth and ensure that a spinal needle of adequate length is used.

Rationale. The distance from the skin to the ligamentum flavum can be measured using ultrasound during preprocedural planning. Knowing the depth to the ligamentum flavum preprocedurally allows the operator to procure a spinal needle of adequate length, anticipate the insertion depth before CSF can be obtained, determine the depth to which a local anesthetic will need to be injected, and decide whether the anticipated difficulty of the procedure warrants referral to or consultation with another specialist.

The skin–ligamentum flavum distance can be measured from a transverse midline view or a longitudinal paramedian view. A longitudinal paramedian view provides an unobstructed view of the ligamentum flavum due to less shadowing from bony structures compared with a midline view. Several studies have demonstrated a strong correlation between the skin–ligamentum flavum distance measured by ultrasound and the actual needle insertion depth in both midline and paramedian views.28,34,36,53,54,57,69,70

A meta-analysis that included 13 comparative studies evaluating the correlation between ultrasound-measured depth and actual needle insertion depth to reach the epidural or intrathecal space consistently demonstrated a strong correlation between the measured and actual depth.50 A few studies have reported near-perfect Pearson correlation coefficients of 0.98.55,71,72 The pooled correlation was 0.91 (95% CI 0.87-0.94). All studies measured the depth from the skin to the ventral side of the ligamentum flavum or the intrathecal space from either a longitudinal paramedian view (n = 4) or a transverse midline view (n = 9). Eight of the more recent studies evaluated the accuracy of the ultrasound measurements and found the depth measurements by ultrasound to be accurate within 1-13 mm of the actual needle insertion depth, with seven of the eight studies reporting a mean difference of ≤3 mm.50

Measurement of the distance between the skin and the ligamentum flavum generally underestimates the needle insertion depth. One study reported that measurement of the skin–ligamentum flavum distance underestimates the needle insertion depth by 7.6 mm to obtain CSF, whereas measurement of the skin–posterior longitudinal ligament distance overestimates the needle insertion depth by 2.5 mm.57 A well-accepted contributor to underestimation of the depth measurements using ultrasound is compression of the skin and soft tissues by the transducer, and therefore, pressure on the skin must be released before freezing an image and measuring the depth to the subarachnoid space.

 

 

Training

8) We recommend that novices should undergo simulation-based training, where available, before attempting ultrasound-guided LPs on actual patients.

Rationale. Similar to training for other bedside procedures, dedicated training sessions, including didactics, supervised practice on patients, and simulation-based practice, should be considered when teaching novices to perform ultrasound-guided LP. Simulation-based training facilitates acquisition of knowledge and skills to perform invasive bedside procedures, including LP.73 Simulation-based training has been commonly incorporated into procedure training for trainees using an immersive experience, such as a “boot camp,”74-77 or a standardized curriculum,78,79 and has demonstrated improvements in post-course procedural knowledge, technical skills, and operator confidence. Two of these studies included training in the use of ultrasound guidance for LP. These studies showed that simulation-based practice improved skill acquisition and confidence.80,81 Simulation using novel computer software may improve skill acquisition in the use of ultrasound guidance for LP.82

9) We recommend that training in ultrasound-guided LPs should be adapted based on prior ultrasound experience, as learning curves will vary.Rationale. The learning curve to achieve competency in the use of ultrasound guidance for LP has not been well studied. The rate of attaining competency in identifying lumbar spine structures using ultrasound will vary by provider based on prior skills in ultrasound-guided procedures.83 Thus, providers with prior ultrasound experience may require less training than those without such experience to achieve competency. However, extensive experience in performing landmark-guided LPs does not necessarily translate into rapid acquisition of skills to perform the procedure with ultrasound guidance. A study of practicing anesthesiologists with no prior ultrasound experience demonstrated that 20 supervised trials of ultrasound-guided spinal anesthesia were insufficient to achieve competency.84 Although minimums may be a necessary step to gain competence, using them as a sole means to define competence does not account for variable learning curves.12 Based on a national survey of 21 hospitalist procedure experts, the mean current vs suggested minimums for initial and ongoing hospital privileging for LPs were 1.8 vs 6.9 and 2.2 vs 4.6 annually in one report.85

A fundamental question that needs to be answered is how to define competency in the use of ultrasound guidance for LP, including the specific skills and knowledge that must be mastered. At a minimum, providers must be able to identify lumbar spinous processes and distinguish them from the sacrum, identify the lumbar interspinous spaces and their corresponding levels, and estimate the depth from the skin to the ligamentum flavum from the midline and paramedian planes. Novice operators may benefit from practicing lumbar spine mapping of nonobese patients using a high-frequency linear transducer that generates high-resolution images and facilitates recognition of lumbar spine structures.

10) We recommend that novice providers should be supervised when performing ultrasound-guided LPs before performing the procedure independently on patients.

Rationale: Demonstration of competency in the use of ultrasound to identify lumbar spine anatomy should be achieved before routinely performing the procedure independently on patients.18 All providers will require a variable period of supervised practice to demonstrate the appropriate technique, followed by a period of unsupervised practice before competency is achieved. Supervised practice with guidance and feedback has been shown to significantly improve providers’ ability to delineate lumbar spine anatomy.86

 

 

KNOWLEDGE GAPS

The process of producing these guidelines revealed areas of uncertainty and important gaps in the literature regarding the use of ultrasound guidance for LP.

First, it is unclear whether the use of ultrasound guidance for LP reduces postprocedural back pain and whether it improves patient satisfaction. Several studies have evaluated postprocedural back pain28,30,32,33,52 and patient satisfaction28,29,33,51 with the use of ultrasound guidance, but these studies have found inconsistent results. Some of these results were probably due to insufficient statistical power or confounding variables. Furthermore, benefits have been demonstrated in certain subgroups, such as overweight patients or those with anatomical abnormalities, as was found in two studies.52,87 Use of ultrasound guidance for spinal anesthesia has been shown to reduce postprocedural headache28 and improve patient satisfaction51, although similar benefit has not been demonstrated in patients undergoing LP.

Second, the effect of using ultrasound guidance on the frequency of traumatic LPs is an area of uncertainty. A “traumatic tap” is defined as an inadvertent puncture of an epidural vein during passage of the spinal needle through the dura. It remains difficult to discern in these studies whether red blood cells detected in the CSF resulted from puncture of an epidural vein or from needle trauma of the skin and soft tissues. Despite this uncertainty, at least seven randomized controlled studies have assessed the effect of ultrasound guidance on traumatic LPs. The meta-analysis by Shaikh et al. included five randomized controlled studies that assessed the effect of ultrasound guidance on the reporting of traumatic taps. The study found a reduced risk of traumatic taps (risk ratio 0.27 [95% CI 0.11-0.67]), an absolute risk reduction of 5.9% (95% CI 2.3%-9.5%), and a number needed to treat of 17 (95% CI 11-44) to prevent one traumatic tap.16 Similarly, the meta-analysis by Gottlieb et al. showed a lower risk of traumatic taps among adults undergoing LP with ultrasound guidance in five randomized controlled studies with an odds ratio of 0.28 (95% CI 0.14-0.59). The meta-analysis by Gottlieb et al. included two adult studies that were not included by Shaikh et al.

Third, several important questions about the technique of ultrasound-guided LP remain unanswered. In addition to the static technique, a dynamic technique with real-time needle tracking has been described to perform ultrasound-guided LP, epidural catheterization, and spinal anesthesia. A pilot study by Grau et al. found that ultrasound used either statically or dynamically had fewer insertion attempts and needle redirections than use of landmarks alone.29 Three other pilot studies showed successful spinal anesthesia in almost all patients88-90 and one large study demonstrated successful spinal anesthesia with real-time ultrasound guidance in 97 of 100 patients with a median of three needle passes.91 Furthermore, a few industry-sponsored studies with small numbers of patients have described the use of novel needle tracking systems that facilitate needle visualization during real-time ultrasound-guided LP.92,93 However, to our knowledge, no comparative studies of static versus dynamic guidance using novel needle tracking systems in human subjects have been published, and any potential role for these novel needle tracking systems has not yet been defined.

Finally, the effects of using ultrasound guidance on clinical decision-making, timeliness, and cost-effectiveness of LP have not yet been explored but could have important clinical practice implications.

 

 

CONCLUSION

Randomized controlled trials have demonstrated that using ultrasound guidance for LPs can reduce the number of needle insertion attempts and needle redirections and increase the overall procedural success rates. Ultrasound can more accurately identify the lumbar spine level than physical examination in both obese and nonobese patients, although the greatest benefit of using ultrasound guidance for LPs has been shown in obese patients.

Ultrasound permits assessment of the interspinous space width and measurement of the ligamentum flavum depth to select an optimal needle insertion site and adequate length spinal needle. Although the use of real-time ultrasound guidance has been described, the use of static ultrasound guidance for LP site marking remains the standard technique.

Acknowledgments

The authors thank all the members of the Society of Hospital Medicine Point-of-care Ultrasound Task Force and the Education Committee members for their time and dedication to develop these guidelines.

Collaborators from Society of Hospital Medicine Point-of-care Ultrasound Task Force: Saaid Abdel-Ghani, Robert Arntfield, Jeffrey Bates, Anjali Bhagra, Michael Blaivas, Daniel Brotman, Carolina Candotti, Richard Hoppmann, Susan Hunt, Trevor P. Jensen, Paul Mayo, Benji Mathews, Satyen Nichani, Vicki Noble, Martin Perez, Nitin Puri, Aliaksei Pustavoitau, Kreegan Reierson, Sophia Rodgers, Kirk Spencer, Vivek Tayal, David Tierney

SHM Point-of-care Ultrasound Task Force: CHAIRS: Nilam Soni, Ricardo Franco-Sadud, Jeff Bates. WORKING GROUPS: Thoracentesis Working Group: Ria Dancel (chair), Daniel Schnobrich, Nitin Puri. Vascular Access Working Group: Ricardo Franco (chair), Benji Matthews, Saaid Abdel-Ghani, Sophia Rodgers, Martin Perez, Daniel Schnobrich. Paracentesis Working Group: Joel Cho (chair), Benji Matthews, Kreegan Reierson, Anjali Bhagra, Trevor P. Jensen Lumbar Puncture Working Group: Nilam J. Soni (chair), Ricardo Franco, Gerard Salame, Josh Lenchus, Venkat Kalidindi, Ketino Kobaidze. Credentialing Working Group: Brian P Lucas (chair), David Tierney, Trevor P. Jensen PEER REVIEWERS: Robert Arntfield, Michael Blaivas, Richard Hoppmann, Paul Mayo, Vicki Noble, Aliaksei Pustavoitau, Kirk Spencer, Vivek Tayal. METHODOLOGIST: Mahmoud El Barbary. LIBRARIAN: Loretta Grikis. SOCIETY OF HOSPITAL MEDICINE EDUCATION COMMITTEE: Daniel Brotman (past chair), Satyen Nichani (current chair), Susan Hunt. SOCIETY OF HOSPITAL MEDICINE STAFF: Nick Marzano.

Disclosures

The authors have nothing to disclose.

Funding

Brian P Lucas: Department of Veterans Affairs, Veterans Health Administration, Office of Research and Development and Dartmouth SYNERGY, National Institutes of Health, National Center for Translational Science (UL1TR001086). Nilam Soni: Department of Veterans Affairs, Quality Enhancement Research Initiative (QUERI) Partnered Evaluation Initiative Grant (HX002263-01A1).

Disclaimer

The contents of this publication do not represent the views of the U.S. Department of Veterans Affairs or the United States Government.

 

References

1. Wolfe KS, Kress JP. Risk of procedural hemorrhage. Chest. 2016;150(1):237-246. https://doi.org/10.1016/j.chest.2016.01.023.
2. Kroll H, Duszak R, Jr, Nsiah E, et al. Trends in lumbar puncture over 2 decades: a dramatic shift to radiology. AJR Am J Roentgenol. 2015;204(1):15-19. https://doi.org/10.2214/AJR.14.12622.
3. Vickers A, Donnelly JP, Moore JX, Wang HE. 263EMF epidemiology of lumbar punctures in hospitalized patients in United States. Ann Emerg Med. 2017;70(4):S104. https://doi.org/10.1016/j.annemergmed.2017.07.241.
4. Køster-Rasmussen R, Korshin A, Meyer CN. Antibiotic treatment delay and outcome in acute bacterial meningitis. J Infect. 2008;57(6):449-454. https://doi.org/10.1016/j.jinf.2008.09.033.
5. Aronin SI, Peduzzi P, Quagliarello VJ. Community-acquired bacterial meningitis: risk stratification for adverse clinical outcome and effect of antibiotic timing. Ann Intern Med. 1998;129(11):862-869. https://doi.org/10.7326/0003-4819-129-11_Part_1-199812010-00004.
6. Lepur D, Barsić B. Community-acquired bacterial meningitis in adults: antibiotic timing in disease course and outcome. Infection. 2007;35(4):225-231. https://doi.org/10.1007/s15010-007-6202-0.
7. Proulx N, Fréchette D, Toye B, Chan J, Kravcik S. Delays in the administration of antibiotics are associated with mortality from adult acute bacterial meningitis. QJM. 2005;98(4):291-298. https://doi.org/10.1093/qjmed/hci047.
8. Auburtin M, Wolff M, Charpentier J, et al. Detrimental role of delayed antibiotic administration and penicillin-nonsusceptible strains in adult intensive care unit patients with pneumococcal meningitis: the PNEUMOREA prospective multicenter study. Crit Care Med. 2006;34(11):2758-2765. https://doi.org/10.1097/01.CCM.0000239434.26669.65.
9. Michael B, Menezes BF, Cunniffe J, et al. Effect of delayed lumbar punctures on the diagnosis of acute bacterial meningitis in adults. Emerg Med J. 2010;27(6):433-438. https://doi.org/10.1136/emj.2009.075598.
10. Glimåker M, Johansson B, Grindborg Ö, et al. Adult bacterial meningitis: earlier treatment and improved outcome following guideline revision promoting prompt lumbar puncture. Clin Infect Dis. 2015;60(8):1162-1169. https://doi.org/10.1093/cid/civ011.
11. Nichani S, Crocker J, Fitterman N, Lukela M. Updating the core competencies in hospital medicine--2017 Revision: introduction and methodology. J Hosp Med. 2017;12(4):283-287. https://doi.org/10.12788/jhm.2715.
12. Soni NJ, Schnobrich D, Matthews BK, et al. Point-of-care ultrasound for hospitalists: a position statement of the Society of Hospital Medicine. J Hosp Med. 2019;14:E1-E6. https://doi.org/10.12788/jhm.3079.
13. Shah KH, McGillicuddy D, Spear J, Edlow JA. Predicting difficult and traumatic lumbar punctures. Am J Emerg Med. 2007;25(6):608-611. https://doi.org/10.1016/j.ajem.2006.11.025.
14. Williams P, Tait G, Wijeratne T. Success rate of elective lumbar puncture at a major Melbourne neurology unit. Surg Neurol Int. 2018;9:12. https://doi.org/10.4103/sni.sni_426_17.
15. Gottlieb M, Holladay D, Peksa GD. Ultrasound-assisted lumbar punctures: a systematic review and meta-analysis. Acad Emerg Med. 2018;26(1). https://doi.org/10.1111/acem.13558.
16. Shaikh F, Brzezinski J, Alexander S, et al. Ultrasound imaging for lumbar punctures and epidural catheterisations: systematic review and meta-analysis. BMJ. 2013;346:f1720. https://doi.org/10.1136/bmj.f1720.
17. Perlas A, Chaparro LE, Chin KJ. Lumbar neuraxial ultrasound for spinal and epidural anesthesia: a systematic review and meta-analysis. Reg Anesth Pain Med. 2016;41(2):251-260. https://doi.org/10.1097/AAP.0000000000000184.
18. Lucas BP, Tierney DM, Jensen TP, et al. Credentialing of hospitalists in ultrasound-guided bedside procedures: a position statement of the Society of Hospital Medicine. J Hosp Med. 2018;13(2):117-125. https://doi.org/10.12788/jhm.2917.
19. Dancel R, Schnobrich D, Puri N, et al. Recommendations on the use of ultrasound guidance for adult thoracentesis: a position statement of the Society of Hospital Medicine. J Hosp Med. 2018;13(2):126-135. https://doi.org/10.12788/jhm.2940.
20. Soni NJ, Franco-Sadud R, Schnobrich D, et al. Ultrasound guidance for lumbar puncture. Neurol Clin Pract. 2016;6(4):358-368. https://doi.org/10.1212/CPJ.0000000000000265.
21. Fitch K, Bernstein SJ, Aguilar MD, Burnand B, LaCalle JR. The Rand/UCLA Appropriateness Method User’s Manual. Santa Monica, CA: Rand Corp; 2001.
22. Abdelhamid SA, Mansour MA. Ultrasound-guided intrathecal anesthesia: does scanning help? Egypt J Anaesth. 2013;29(4):389-394. https://doi.org/10.1016/j.egja.2013.06.003.
23. Ansari T, Yousef A, El Gamassy A, Fayez M. Ultrasound-guided spinal anaesthesia in obstetrics: is there an advantage over the landmark technique in patients with easily palpable spines? Int J Obstet Anesth. 2014;23(3):213-216. https://doi.org/10.1016/j.ijoa.2014.03.001.
24. Chin KJ, Perlas A, Chan V, et al. Ultrasound imaging facilitates spinal anesthesia in adults with difficult surface anatomic landmarks. Anesthesiology. 2011;115(1):94-101. https://doi.org/10.1097/ALN.0b013e31821a8ad4.
25. Cho YC, Koo DH, Oh SK, et al. Comparison of ultrasound-assisted lumbar puncture with lumbar puncture using palpation of landmarks in aged patients in an emergency center. J Korean Soc Emerg Med. 2009;20(3):304.
26. Grau T, Leipold RW, Conradi R, Martin E. Ultrasound control for presumed difficult epidural puncture. Acta Anaesthesiol Scand. 2001;45(6):766-771. https://doi.org/10.1034/j.1399-6576.2001.045006766.x.
27. Grau T, Leipold RW, Conradi R, Martin E, Motsch J. Ultrasound imaging facilitates localization of the epidural space during combined spinal and epidural anesthesia. Reg Anesth Pain Med. 2001;26(1):64-67. https://doi.org/10.1053/rapm.2001.19633.
28. Grau T, Leipold RW, Conradi R, Martin E, Motsch J. Efficacy of ultrasound imaging in obstetric epidural anesthesia. J Clin Anesth. 2002;14(3):169-175. https://doi.org/10.1016/S0952-8180(01)00378-6.
29. Grau T, Leipold RW, Fatehi S, Martin E, Motsch J. Real-time ultrasonic observation of combined spinal-epidural anaesthesia. Eur J Anaesthesiol. 2004;21(1):25-31. https://doi.org/10.1017/S026502150400105X.
30. Mofidi M, Mohammadi M, Saidi H, et al. Ultrasound guided lumbar puncture in emergency department: time saving and less complications. J Res Med Sci. 2013;18(4):303-307. PubMed
31. Nassar M, Abdelazim IA. Pre-puncture ultrasound guided epidural insertion before vaginal delivery. J Clin Monit Comput. 2015;29(5):573-577. https://doi.org/10.1007/s10877-014-9634-y.

32. Nomura JT, Leech SJ, Shenbagamurthi S, et al. A randomized controlled trial of ultrasound-assisted lumbar puncture. J Ultrasound Med. 2007;26(10):1341-1348. https://doi.org/10.7863/jum.2007.26.10.1341.
33. Peterson MA, Pisupati D, Heyming TW, Abele JA, Lewis RJ. Ultrasound for routine lumbar puncture. Acad Emerg Med. 2014;21(2):130-136. https://doi.org/10.1111/acem.12305.
34. Sahin T, Balaban O, Sahin L, Solak M, Toker K. A randomized controlled trial of preinsertion ultrasound guidance for spinal anaesthesia in pregnancy: outcomes among obese and lean parturients: ultrasound for spinal anesthesia in pregnancy. J Anesth. 2014;28(3):413-419. https://doi.org/10.1007/s00540-013-1726-1.
35. Wang Q, Yin C, Wang TL. Ultrasound facilitates identification of combined spinal-epidural puncture in obese parturients. Chin Med J (Engl). 2012;125(21):3840-3843. PubMed
36. Vallejo MC, Phelps AL, Singh S, Orebaugh SL, Sah N. Ultrasound decreases the failed labor epidural rate in resident trainees. Int J Obstet Anesth. 2010;19(4):373-378. https://doi.org/10.1016/j.ijoa.2010.04.002.
37. Darrieutort-Laffite C, Bart G, Planche L, et al. Usefulness of a pre-procedure ultrasound scanning of the lumbar spine before epidural injection in patients with a presumed difficult puncture: a randomized controlled trial. Joint Bone Spine. 2015;82(5):356-361. https://doi.org/10.1016/j.jbspin.2015.02.001.
38. Vosko MR, Brunner C, Schreiber S. Lumbar puncture with ultrasound study (lupus study)-international prospective randomized multicentre trial. Int J Stroke. 2017;12(1):22. https://doi.org/10.1055/s-0037-1606991.
39. Urfalioğlu A, Bilal B, Öksüz G, et al. Comparison of the landmark and ultrasound methods in cesarean sections performed under spinal anesthesia on obese pregnants. J Matern Fetal Neonatal Med. 2017;30(9):1051-1056. https://doi.org/10.1080/14767058.2016.1199677.
40. Tawfik MM, Atallah MM, Elkharboutly WS, Allakkany NS, Abdelkhalek M. Does preprocedural ultrasound increase the first-pass success rate of epidural catheterization before cesarean delivery? A randomized controlled trial. Anesth Analg. 2017;124(3):851-856. https://doi.org/10.1213/ANE.0000000000001325.
41. Turkstra TP, Marmai KL, Armstrong KP, Kumar K, Singh SI. Preprocedural ultrasound assessment does not improve trainee performance of spinal anesthesia for obstetrical patients: a randomized controlled trial. J Clin Anesth. 2017;37:21-24. https://doi.org/10.1016/j.jclinane.2016.10.034.
42. Chong SE, Mohd Nikman A, Saedah A, et al. Real-time ultrasound-guided paramedian spinal anaesthesia: evaluation of the efficacy and the success rate of single needle pass. Br J Anaesth. 2017;118(5):799-801. https://doi.org/10.1093/bja/aex108.
43. Creaney M, Mullane D, Casby C, Tan T. Ultrasound to identify the lumbar space in women with impalpable bony landmarks presenting for elective caesarean delivery under spinal anaesthesia: a randomised trial. Int J Obstet Anesth. 2016;28:12-16. https://doi.org/10.1016/j.ijoa.2016.07.007.
44. Ekinci M, Alici HA, Ahiskalioglu A, et al. The use of ultrasound in planned cesarean delivery under spinal anesthesia for patients having nonprominent anatomic landmarks. J Clin Anesth. 2017;37:82-85. https://doi.org/10.1016/j.jclinane.2016.10.014.
45. Perna P, Gioia A, Ragazzi R, Volta CA, Innamorato M. Can pre-procedure neuroaxial ultrasound improve the identification of the potential epidural space when compared with anatomical landmarks? A prospective randomized study. Minerva Anestesiol. 2017;83(1):41-49. https://doi.org/10.23736/S0375-9393.16.11399-9.
46. Chin A, Crooke B, Heywood L, et al. A randomised controlled trial comparing needle movements during combined spinal-epidural anaesthesia with and without ultrasound assistance. Anaesthesia. 2018;73(4):466-473. https://doi.org/10.1111/anae.14206.
47. Dhanger S, Vinayagam S, Vaidhyanathan B, Rajesh IJ, Tripathy DK. Comparison of landmark versus pre-procedural ultrasonography-assisted midline approach for identification of subarachnoid space in elective caesarean section: a randomised controlled trial. Indian J Anaesth. 2018;62(4):280-284. https://doi.org/10.4103/ija.IJA_488_17.
48. Evans DP, Tozer J, Joyce M, Vitto MJ. Comparison of ultrasound-guided and landmark-based lumbar punctures in inexperienced resident physicians. J Ultrasound Med. 2019;38(3):613-620. https://doi.org/10.1002/jum.14728.
49. Srinivasan KK, Leo AM, Iohom G, Loughnane F, Lee PJ. Pre-procedure ultrasound-guided paramedian spinal anaesthesia at L5-S1: is this better than landmark-guided midline approach? A randomised controlled trial. Indian J Anaesth. 2018;62(1):53-60. https://doi.org/10.4103/ija.IJA_448_17.
50. Perlas A, Chaparro LE, Chin KJ. Lumbar neuraxial ultrasound for spinal and epidural anesthesia: a systematic review and meta-analysis. Reg Anesth Pain Med. 2016;41(2):251-260. https://doi.org/10.1097/AAP.0000000000000184.
51. Lim YC, Choo CY, Tan KT. A randomised controlled trial of ultrasound-assisted spinal anaesthesia. Anaesth Intensive Care. 2014;42(2):191-198. https://doi.org/10.1177/0310057X1404200205.

52. Honarbakhsh S, Osman C, Teo JTH, Gabriel C. Ultrasound-guided lumbar puncture as a diagnostic aid to reduce number of attempts and complication rates. Ultrasound. 2013;21(4):170-175. https://doi.org/10.1177/1742271X13504332.
53. Sahota JS, Carvalho JC, Balki M, Fanning N, Arzola C. Ultrasound estimates for midline epidural punctures in the obese parturient: paramedian sagittal oblique is comparable to transverse median plane. Anesth Analg. 2013;116(4):829-835. https://doi.org/10.1213/ANE.0b013e31827f55f0.
54. Balki M, Lee Y, Halpern S, Carvalho JC. Ultrasound imaging of the lumbar spine in the transverse plane: the correlation between estimated and actual depth to the epidural space in obese parturients. Anesth Analg. 2009;108(6):1876-1881. https://doi.org/10.1213/ane.0b013e3181a323f6.
55. Wallace DH, Currie JM, Gilstrap LC, Santos R. Indirect sonographic guidance for epidural anesthesia in obese pregnant patients. Reg Anesth. 1992;17(4):233-236. PubMed
56. Srinivasan KK, Iohom G, Loughnane F, Lee PJ. Conventional landmark-guided midline versus preprocedure ultrasound-guided paramedian techniques in spinal anesthesia. Anesth Analg. 2015;21(4):1089-1096. https://doi.org/10.1213/ANE.0000000000000911.
57. Chin KJ, Perlas A, Singh M, et al. An ultrasound-assisted approach facilitates spinal anesthesia for total joint arthroplasty. Can J Anaesth. 2009;56(9):643-650. https://doi.org/10.1007/s12630-009-9132-8.
58. Evansa I, Logina I, Vanags I, Borgeat A. Ultrasound versus fluoroscopic-guided epidural steroid injections in patients with degenerative spinal diseases: a randomised study. Eur J Anaesthesiol. 2015;32(4):262-268. https://doi.org/10.1097/EJA.0000000000000103.
59. Park Y, Lee JH, Park KD, et al. Ultrasound-guided vs fluoroscopy-guided caudal epidural steroid injection for the treatment of unilateral lower lumbar radicular pain: a prospective, randomized, single-blind clinical study. Am J Phys Med Rehabil. 2013;92(7):575-586. https://doi.org/10.1097/PHM.0b013e318292356b.
60. Margarido CB, Mikhael R, Arzola C, Balki M, Carvalho JC. The intercristal line determined by palpation is not a reliable anatomical landmark for neuraxial anesthesia. Can J Anaesth. 2011;58(3):262-266. https://doi.org/10.1007/s12630-010-9432-z.
61. Duniec L, Nowakowski P, Kosson D, Łazowski T. Anatomical landmarks based assessment of intravertebral space level for lumbar puncture is misleading in more than 30%. Anaesthesiol Intensive Ther. 2013;45(1):1-6. https://doi.org/10.5603/AIT.2013.0001.
62. Schlotterbeck H, Schaeffer R, Dow WA, et al. Ultrasonographic control of the puncture level for lumbar neuraxial block in obstetric anaesthesia. Br J Anaesth. 2008;100(2):230-234. https://doi.org/10.1093/bja/aem371.
63. Whitty R, Moore M, Macarthur A. Identification of the lumbar interspinous spaces: palpation versus ultrasound. Anesth Analg. 2008;106(2):538-540, table of contents. https://doi.org/10.1213/ane.0b013e31816069d9.
64. Locks Gde F, Almeida MC, Pereira AA. Use of the ultrasound to determine the level of lumbar puncture in pregnant women. Rev Bras Anestesiol. 2010;60(1):13-19. https://doi.org/10.1016/S0034-7094(10)70002-7.
65. Stiffler KA, Jwayyed S, Wilber ST, Robinson A. The use of ultrasound to identify pertinent landmarks for lumbar puncture. Am J Emerg Med. 2007;25(3):331-334. https://doi.org/10.1016/j.ajem.2006.07.010.

66. Gulay U, Meltem T, Nadir SS, Aysin A. Ultrasound-guided evaluation of the lumbar subarachnoid space in lateral and sitting positions in pregnant patients to receive elective cesarean operation. Pak J Med Sci. 2015;31(1):76-81. https://doi.org/10.12669/pjms.311.5647.
67. Kawaguchi R, Yamauchi M, Sugino S, Yamakage M. Ultrasound-aided ipsilateral-dominant epidural block for total hip arthroplasty: a randomised controlled single-blind study. Eur J Anaesthesiol. 2011;28(2):137-140. https://doi.org/10.1097/EJA.0b013e3283423457.
68. Grau T, Leipold RW, Horter J, Martin E, Motsch J. Colour Doppler imaging of the interspinous and epidural space. Eur J Anaesthesiol. 2001;18(11):706-712. https://doi.org/10.1097/00003643-200111000-00002.
69. Arzola C, Davies S, Rofaeel A, Carvalho JC. Ultrasound using the transverse approach to the lumbar spine provides reliable landmarks for labor epidurals. Anesth Analg. 2007;104(5):1188-92, tables of contents. https://doi.org/10.1213/01.ane.0000250912.66057.41.
70. Chauhan AK, Bhatia R, Agrawal S. Lumbar epidural depth using transverse ultrasound scan and its correlation with loss of resistance technique: a prospective observational study in Indian population. Saudi J Anaesth. 2018;12(2):279-282. https://doi.org/10.4103/sja.SJA_679_17.
71. Gnaho A, Nguyen V, Villevielle T, et al. Assessing the depth of the subarachnoid space by ultrasound. Rev Bras Anestesiol. 2012;62(4):520-530. https://doi.org/10.1016/S0034-7094(12)70150-2.
72. Cork RC, Kryc JJ, Vaughan RW. Ultrasonic localization of the lumbar epidural space. Anesthesiology. 1980;52(6):513-516. https://doi.org/10.1097/00000542-198006000-00013.
73. Barsuk JH, Cohen ER, Caprio T, et al. Simulation-based education with mastery learning improves residents’ lumbar puncture skills. Neurology. 2012;79(2):132-137. https://doi.org/10.1212/WNL.0b013e31825dd39d.
74. Lenchus J, Issenberg SB, Murphy D, et al. A blended approach to invasive bedside procedural instruction. Med Teach. 2011;33(2):116-123. https://doi.org/10.3109/0142159X.2010.509412.
75. Wayne DB, Cohen ER, Singer BD, et al. Progress toward improving medical school graduates’ skills via a “boot camp” curriculum. Simul Healthc. 2014;9(1):33-39. https://doi.org/10.1097/SIH.0000000000000001.
76. Cohen ER, Barsuk JH, Moazed F, et al. Making July safer: simulation-based mastery learning during intern boot camp. Acad Med. 2013;88(2):233-239. https://doi.org/10.1097/ACM.0b013e31827bfc0a.
77. Martin R, Gannon D, Riggle J, et al. A comprehensive workshop using simulation to train internal medicine residents in bedside procedures performed by internists. Chest. 2012;142(4):545A. https://doi.org/10.1378/chest.1390093.
78. Lenchus JD. End of the “see one, do one, teach one” era: the next generation of invasive bedside procedural instruction. J Am Osteopath Assoc. 2010;110(6):340-346. PubMed
79. Mourad M, Ranji S, Sliwka D. A randomized controlled trial of the impact of a teaching procedure service on the training of internal medicine residents. J Grad Med Educ. 2012;4(2):170-175. https://doi.org/10.4300/JGME-D-11-00136.1.
80. Restrepo CG, Baker MD, Pruitt CM, Gullett JP, Pigott DC. Ability of pediatric emergency medicine physicians to identify anatomic landmarks with the assistance of ultrasound prior to lumbar puncture in a simulated obese model. Pediatr Emerg Care. 2015;31(1):15-19. https://doi.org/10.1097/PEC.0000000000000330.
81. VanderWielen BA, Harris R, Galgon RE, VanderWielen LM, Schroeder KM. Teaching sonoanatomy to anesthesia faculty and residents: utility of hands-on gel phantom and instructional video training models. J Clin Anesth. 2015;27(3):188-194. https://doi.org/10.1016/j.jclinane.2014.07.007.
82. Keri Z, Sydor D, Ungi T, et al. Computerized training system for ultrasound-guided lumbar puncture on abnormal spine models: a randomized controlled trial. Can J Anaesth. 2015;62(7):777-784. https://doi.org/10.1007/s12630-015-0367-2.
83. Deacon AJ, Melhuishi NS, Terblanche NC. CUSUM method for construction of trainee spinal ultrasound learning curves following standardised teaching. Anaesth Intensive Care. 2014;42(4):480-486. https://doi.org/10.1177/0310057X1404200409.
84. Margarido CB, Arzola C, Balki M, Carvalho JC. Anesthesiologists’ learning curves for ultrasound assessment of the lumbar spine. Can J Anaesth. 2010;57(2):120-126. https://doi.org/10.1007/s12630-009-9219-2.
85. Jensen TP, Soni NJ, Tierney DM, Lucas BP. Hospital privileging practices for bedside procedures: a survey of hospitalist experts. J Hosp Med. 2017;12(10):836-839. https://doi.org/10.12788/jhm.2837.
86. Terblanche NC, Arzola C, Wills KE, et al. Standardised training program in spinal ultrasound for epidural insertion: protocol driven versus non-protocol driven teaching approach. Anaesth Intensive Care. 2014;42(4):460-466. https://doi.org/10.1177/0310057X1404200406.
87. Mofidi M, Mohammadi M, Saidi H, et al. Ultrasound guided lumbar puncture in emergency department: time saving and less complications. J Res Med Sci. 2013;18(4):303-307. PubMed
88. Karmakar MK, Li X, Ho AM, Kwok WH, Chui PT. Real-time ultrasound-guided paramedian epidural access: evaluation of a novel in-plane technique. Br J Anaesth. 2009;102(6):845-854. https://doi.org/10.1093/bja/aep079.
89. Tran D, Kamani AA, Al-Attas E, et al. Single-operator real-time ultrasound-guidance to aim and insert a lumbar epidural needle. Can J Anaesth. 2010;57(4):313-321. https://doi.org/10.1007/s12630-009-9252-1.
90. Liu Y, Qian W, Ke XJ, Mei W. Real-time ultrasound-guided spinal anesthesia using a new paramedian transverse approach. Curr Med Sci. 2018;38(5):910-913. https://doi.org/10.1007/s11596-018-1961-7.
91. Conroy PH, Luyet C, McCartney CJ, McHardy PG. Real-time ultrasound-guided spinal anaesthesia: a prospective observational study of a new approach. Anesthesiol Res Pract. 2013;2013:525818. https://doi.org/10.1155/2013/525818.
92. Brinkmann S, Tang R, Sawka A, Vaghadia H. Single-operator real-time ultrasound-guided spinal injection using SonixGPS™: a case series. Can J Anaesth. 2013;60(9):896-901. https://doi.org/10.1007/s12630-013-9984-9.
93. Niazi AU, Chin KJ, Jin R, Chan VW. Real-time ultrasound-guided spinal anesthesia using the SonixGPS ultrasound guidance system: a feasibility study. Acta Anaesthesiol Scand. 2014;58(7):875-881. https://doi.org/10.1111/aas.12353.

References

1. Wolfe KS, Kress JP. Risk of procedural hemorrhage. Chest. 2016;150(1):237-246. https://doi.org/10.1016/j.chest.2016.01.023.
2. Kroll H, Duszak R, Jr, Nsiah E, et al. Trends in lumbar puncture over 2 decades: a dramatic shift to radiology. AJR Am J Roentgenol. 2015;204(1):15-19. https://doi.org/10.2214/AJR.14.12622.
3. Vickers A, Donnelly JP, Moore JX, Wang HE. 263EMF epidemiology of lumbar punctures in hospitalized patients in United States. Ann Emerg Med. 2017;70(4):S104. https://doi.org/10.1016/j.annemergmed.2017.07.241.
4. Køster-Rasmussen R, Korshin A, Meyer CN. Antibiotic treatment delay and outcome in acute bacterial meningitis. J Infect. 2008;57(6):449-454. https://doi.org/10.1016/j.jinf.2008.09.033.
5. Aronin SI, Peduzzi P, Quagliarello VJ. Community-acquired bacterial meningitis: risk stratification for adverse clinical outcome and effect of antibiotic timing. Ann Intern Med. 1998;129(11):862-869. https://doi.org/10.7326/0003-4819-129-11_Part_1-199812010-00004.
6. Lepur D, Barsić B. Community-acquired bacterial meningitis in adults: antibiotic timing in disease course and outcome. Infection. 2007;35(4):225-231. https://doi.org/10.1007/s15010-007-6202-0.
7. Proulx N, Fréchette D, Toye B, Chan J, Kravcik S. Delays in the administration of antibiotics are associated with mortality from adult acute bacterial meningitis. QJM. 2005;98(4):291-298. https://doi.org/10.1093/qjmed/hci047.
8. Auburtin M, Wolff M, Charpentier J, et al. Detrimental role of delayed antibiotic administration and penicillin-nonsusceptible strains in adult intensive care unit patients with pneumococcal meningitis: the PNEUMOREA prospective multicenter study. Crit Care Med. 2006;34(11):2758-2765. https://doi.org/10.1097/01.CCM.0000239434.26669.65.
9. Michael B, Menezes BF, Cunniffe J, et al. Effect of delayed lumbar punctures on the diagnosis of acute bacterial meningitis in adults. Emerg Med J. 2010;27(6):433-438. https://doi.org/10.1136/emj.2009.075598.
10. Glimåker M, Johansson B, Grindborg Ö, et al. Adult bacterial meningitis: earlier treatment and improved outcome following guideline revision promoting prompt lumbar puncture. Clin Infect Dis. 2015;60(8):1162-1169. https://doi.org/10.1093/cid/civ011.
11. Nichani S, Crocker J, Fitterman N, Lukela M. Updating the core competencies in hospital medicine--2017 Revision: introduction and methodology. J Hosp Med. 2017;12(4):283-287. https://doi.org/10.12788/jhm.2715.
12. Soni NJ, Schnobrich D, Matthews BK, et al. Point-of-care ultrasound for hospitalists: a position statement of the Society of Hospital Medicine. J Hosp Med. 2019;14:E1-E6. https://doi.org/10.12788/jhm.3079.
13. Shah KH, McGillicuddy D, Spear J, Edlow JA. Predicting difficult and traumatic lumbar punctures. Am J Emerg Med. 2007;25(6):608-611. https://doi.org/10.1016/j.ajem.2006.11.025.
14. Williams P, Tait G, Wijeratne T. Success rate of elective lumbar puncture at a major Melbourne neurology unit. Surg Neurol Int. 2018;9:12. https://doi.org/10.4103/sni.sni_426_17.
15. Gottlieb M, Holladay D, Peksa GD. Ultrasound-assisted lumbar punctures: a systematic review and meta-analysis. Acad Emerg Med. 2018;26(1). https://doi.org/10.1111/acem.13558.
16. Shaikh F, Brzezinski J, Alexander S, et al. Ultrasound imaging for lumbar punctures and epidural catheterisations: systematic review and meta-analysis. BMJ. 2013;346:f1720. https://doi.org/10.1136/bmj.f1720.
17. Perlas A, Chaparro LE, Chin KJ. Lumbar neuraxial ultrasound for spinal and epidural anesthesia: a systematic review and meta-analysis. Reg Anesth Pain Med. 2016;41(2):251-260. https://doi.org/10.1097/AAP.0000000000000184.
18. Lucas BP, Tierney DM, Jensen TP, et al. Credentialing of hospitalists in ultrasound-guided bedside procedures: a position statement of the Society of Hospital Medicine. J Hosp Med. 2018;13(2):117-125. https://doi.org/10.12788/jhm.2917.
19. Dancel R, Schnobrich D, Puri N, et al. Recommendations on the use of ultrasound guidance for adult thoracentesis: a position statement of the Society of Hospital Medicine. J Hosp Med. 2018;13(2):126-135. https://doi.org/10.12788/jhm.2940.
20. Soni NJ, Franco-Sadud R, Schnobrich D, et al. Ultrasound guidance for lumbar puncture. Neurol Clin Pract. 2016;6(4):358-368. https://doi.org/10.1212/CPJ.0000000000000265.
21. Fitch K, Bernstein SJ, Aguilar MD, Burnand B, LaCalle JR. The Rand/UCLA Appropriateness Method User’s Manual. Santa Monica, CA: Rand Corp; 2001.
22. Abdelhamid SA, Mansour MA. Ultrasound-guided intrathecal anesthesia: does scanning help? Egypt J Anaesth. 2013;29(4):389-394. https://doi.org/10.1016/j.egja.2013.06.003.
23. Ansari T, Yousef A, El Gamassy A, Fayez M. Ultrasound-guided spinal anaesthesia in obstetrics: is there an advantage over the landmark technique in patients with easily palpable spines? Int J Obstet Anesth. 2014;23(3):213-216. https://doi.org/10.1016/j.ijoa.2014.03.001.
24. Chin KJ, Perlas A, Chan V, et al. Ultrasound imaging facilitates spinal anesthesia in adults with difficult surface anatomic landmarks. Anesthesiology. 2011;115(1):94-101. https://doi.org/10.1097/ALN.0b013e31821a8ad4.
25. Cho YC, Koo DH, Oh SK, et al. Comparison of ultrasound-assisted lumbar puncture with lumbar puncture using palpation of landmarks in aged patients in an emergency center. J Korean Soc Emerg Med. 2009;20(3):304.
26. Grau T, Leipold RW, Conradi R, Martin E. Ultrasound control for presumed difficult epidural puncture. Acta Anaesthesiol Scand. 2001;45(6):766-771. https://doi.org/10.1034/j.1399-6576.2001.045006766.x.
27. Grau T, Leipold RW, Conradi R, Martin E, Motsch J. Ultrasound imaging facilitates localization of the epidural space during combined spinal and epidural anesthesia. Reg Anesth Pain Med. 2001;26(1):64-67. https://doi.org/10.1053/rapm.2001.19633.
28. Grau T, Leipold RW, Conradi R, Martin E, Motsch J. Efficacy of ultrasound imaging in obstetric epidural anesthesia. J Clin Anesth. 2002;14(3):169-175. https://doi.org/10.1016/S0952-8180(01)00378-6.
29. Grau T, Leipold RW, Fatehi S, Martin E, Motsch J. Real-time ultrasonic observation of combined spinal-epidural anaesthesia. Eur J Anaesthesiol. 2004;21(1):25-31. https://doi.org/10.1017/S026502150400105X.
30. Mofidi M, Mohammadi M, Saidi H, et al. Ultrasound guided lumbar puncture in emergency department: time saving and less complications. J Res Med Sci. 2013;18(4):303-307. PubMed
31. Nassar M, Abdelazim IA. Pre-puncture ultrasound guided epidural insertion before vaginal delivery. J Clin Monit Comput. 2015;29(5):573-577. https://doi.org/10.1007/s10877-014-9634-y.

32. Nomura JT, Leech SJ, Shenbagamurthi S, et al. A randomized controlled trial of ultrasound-assisted lumbar puncture. J Ultrasound Med. 2007;26(10):1341-1348. https://doi.org/10.7863/jum.2007.26.10.1341.
33. Peterson MA, Pisupati D, Heyming TW, Abele JA, Lewis RJ. Ultrasound for routine lumbar puncture. Acad Emerg Med. 2014;21(2):130-136. https://doi.org/10.1111/acem.12305.
34. Sahin T, Balaban O, Sahin L, Solak M, Toker K. A randomized controlled trial of preinsertion ultrasound guidance for spinal anaesthesia in pregnancy: outcomes among obese and lean parturients: ultrasound for spinal anesthesia in pregnancy. J Anesth. 2014;28(3):413-419. https://doi.org/10.1007/s00540-013-1726-1.
35. Wang Q, Yin C, Wang TL. Ultrasound facilitates identification of combined spinal-epidural puncture in obese parturients. Chin Med J (Engl). 2012;125(21):3840-3843. PubMed
36. Vallejo MC, Phelps AL, Singh S, Orebaugh SL, Sah N. Ultrasound decreases the failed labor epidural rate in resident trainees. Int J Obstet Anesth. 2010;19(4):373-378. https://doi.org/10.1016/j.ijoa.2010.04.002.
37. Darrieutort-Laffite C, Bart G, Planche L, et al. Usefulness of a pre-procedure ultrasound scanning of the lumbar spine before epidural injection in patients with a presumed difficult puncture: a randomized controlled trial. Joint Bone Spine. 2015;82(5):356-361. https://doi.org/10.1016/j.jbspin.2015.02.001.
38. Vosko MR, Brunner C, Schreiber S. Lumbar puncture with ultrasound study (lupus study)-international prospective randomized multicentre trial. Int J Stroke. 2017;12(1):22. https://doi.org/10.1055/s-0037-1606991.
39. Urfalioğlu A, Bilal B, Öksüz G, et al. Comparison of the landmark and ultrasound methods in cesarean sections performed under spinal anesthesia on obese pregnants. J Matern Fetal Neonatal Med. 2017;30(9):1051-1056. https://doi.org/10.1080/14767058.2016.1199677.
40. Tawfik MM, Atallah MM, Elkharboutly WS, Allakkany NS, Abdelkhalek M. Does preprocedural ultrasound increase the first-pass success rate of epidural catheterization before cesarean delivery? A randomized controlled trial. Anesth Analg. 2017;124(3):851-856. https://doi.org/10.1213/ANE.0000000000001325.
41. Turkstra TP, Marmai KL, Armstrong KP, Kumar K, Singh SI. Preprocedural ultrasound assessment does not improve trainee performance of spinal anesthesia for obstetrical patients: a randomized controlled trial. J Clin Anesth. 2017;37:21-24. https://doi.org/10.1016/j.jclinane.2016.10.034.
42. Chong SE, Mohd Nikman A, Saedah A, et al. Real-time ultrasound-guided paramedian spinal anaesthesia: evaluation of the efficacy and the success rate of single needle pass. Br J Anaesth. 2017;118(5):799-801. https://doi.org/10.1093/bja/aex108.
43. Creaney M, Mullane D, Casby C, Tan T. Ultrasound to identify the lumbar space in women with impalpable bony landmarks presenting for elective caesarean delivery under spinal anaesthesia: a randomised trial. Int J Obstet Anesth. 2016;28:12-16. https://doi.org/10.1016/j.ijoa.2016.07.007.
44. Ekinci M, Alici HA, Ahiskalioglu A, et al. The use of ultrasound in planned cesarean delivery under spinal anesthesia for patients having nonprominent anatomic landmarks. J Clin Anesth. 2017;37:82-85. https://doi.org/10.1016/j.jclinane.2016.10.014.
45. Perna P, Gioia A, Ragazzi R, Volta CA, Innamorato M. Can pre-procedure neuroaxial ultrasound improve the identification of the potential epidural space when compared with anatomical landmarks? A prospective randomized study. Minerva Anestesiol. 2017;83(1):41-49. https://doi.org/10.23736/S0375-9393.16.11399-9.
46. Chin A, Crooke B, Heywood L, et al. A randomised controlled trial comparing needle movements during combined spinal-epidural anaesthesia with and without ultrasound assistance. Anaesthesia. 2018;73(4):466-473. https://doi.org/10.1111/anae.14206.
47. Dhanger S, Vinayagam S, Vaidhyanathan B, Rajesh IJ, Tripathy DK. Comparison of landmark versus pre-procedural ultrasonography-assisted midline approach for identification of subarachnoid space in elective caesarean section: a randomised controlled trial. Indian J Anaesth. 2018;62(4):280-284. https://doi.org/10.4103/ija.IJA_488_17.
48. Evans DP, Tozer J, Joyce M, Vitto MJ. Comparison of ultrasound-guided and landmark-based lumbar punctures in inexperienced resident physicians. J Ultrasound Med. 2019;38(3):613-620. https://doi.org/10.1002/jum.14728.
49. Srinivasan KK, Leo AM, Iohom G, Loughnane F, Lee PJ. Pre-procedure ultrasound-guided paramedian spinal anaesthesia at L5-S1: is this better than landmark-guided midline approach? A randomised controlled trial. Indian J Anaesth. 2018;62(1):53-60. https://doi.org/10.4103/ija.IJA_448_17.
50. Perlas A, Chaparro LE, Chin KJ. Lumbar neuraxial ultrasound for spinal and epidural anesthesia: a systematic review and meta-analysis. Reg Anesth Pain Med. 2016;41(2):251-260. https://doi.org/10.1097/AAP.0000000000000184.
51. Lim YC, Choo CY, Tan KT. A randomised controlled trial of ultrasound-assisted spinal anaesthesia. Anaesth Intensive Care. 2014;42(2):191-198. https://doi.org/10.1177/0310057X1404200205.

52. Honarbakhsh S, Osman C, Teo JTH, Gabriel C. Ultrasound-guided lumbar puncture as a diagnostic aid to reduce number of attempts and complication rates. Ultrasound. 2013;21(4):170-175. https://doi.org/10.1177/1742271X13504332.
53. Sahota JS, Carvalho JC, Balki M, Fanning N, Arzola C. Ultrasound estimates for midline epidural punctures in the obese parturient: paramedian sagittal oblique is comparable to transverse median plane. Anesth Analg. 2013;116(4):829-835. https://doi.org/10.1213/ANE.0b013e31827f55f0.
54. Balki M, Lee Y, Halpern S, Carvalho JC. Ultrasound imaging of the lumbar spine in the transverse plane: the correlation between estimated and actual depth to the epidural space in obese parturients. Anesth Analg. 2009;108(6):1876-1881. https://doi.org/10.1213/ane.0b013e3181a323f6.
55. Wallace DH, Currie JM, Gilstrap LC, Santos R. Indirect sonographic guidance for epidural anesthesia in obese pregnant patients. Reg Anesth. 1992;17(4):233-236. PubMed
56. Srinivasan KK, Iohom G, Loughnane F, Lee PJ. Conventional landmark-guided midline versus preprocedure ultrasound-guided paramedian techniques in spinal anesthesia. Anesth Analg. 2015;21(4):1089-1096. https://doi.org/10.1213/ANE.0000000000000911.
57. Chin KJ, Perlas A, Singh M, et al. An ultrasound-assisted approach facilitates spinal anesthesia for total joint arthroplasty. Can J Anaesth. 2009;56(9):643-650. https://doi.org/10.1007/s12630-009-9132-8.
58. Evansa I, Logina I, Vanags I, Borgeat A. Ultrasound versus fluoroscopic-guided epidural steroid injections in patients with degenerative spinal diseases: a randomised study. Eur J Anaesthesiol. 2015;32(4):262-268. https://doi.org/10.1097/EJA.0000000000000103.
59. Park Y, Lee JH, Park KD, et al. Ultrasound-guided vs fluoroscopy-guided caudal epidural steroid injection for the treatment of unilateral lower lumbar radicular pain: a prospective, randomized, single-blind clinical study. Am J Phys Med Rehabil. 2013;92(7):575-586. https://doi.org/10.1097/PHM.0b013e318292356b.
60. Margarido CB, Mikhael R, Arzola C, Balki M, Carvalho JC. The intercristal line determined by palpation is not a reliable anatomical landmark for neuraxial anesthesia. Can J Anaesth. 2011;58(3):262-266. https://doi.org/10.1007/s12630-010-9432-z.
61. Duniec L, Nowakowski P, Kosson D, Łazowski T. Anatomical landmarks based assessment of intravertebral space level for lumbar puncture is misleading in more than 30%. Anaesthesiol Intensive Ther. 2013;45(1):1-6. https://doi.org/10.5603/AIT.2013.0001.
62. Schlotterbeck H, Schaeffer R, Dow WA, et al. Ultrasonographic control of the puncture level for lumbar neuraxial block in obstetric anaesthesia. Br J Anaesth. 2008;100(2):230-234. https://doi.org/10.1093/bja/aem371.
63. Whitty R, Moore M, Macarthur A. Identification of the lumbar interspinous spaces: palpation versus ultrasound. Anesth Analg. 2008;106(2):538-540, table of contents. https://doi.org/10.1213/ane.0b013e31816069d9.
64. Locks Gde F, Almeida MC, Pereira AA. Use of the ultrasound to determine the level of lumbar puncture in pregnant women. Rev Bras Anestesiol. 2010;60(1):13-19. https://doi.org/10.1016/S0034-7094(10)70002-7.
65. Stiffler KA, Jwayyed S, Wilber ST, Robinson A. The use of ultrasound to identify pertinent landmarks for lumbar puncture. Am J Emerg Med. 2007;25(3):331-334. https://doi.org/10.1016/j.ajem.2006.07.010.

66. Gulay U, Meltem T, Nadir SS, Aysin A. Ultrasound-guided evaluation of the lumbar subarachnoid space in lateral and sitting positions in pregnant patients to receive elective cesarean operation. Pak J Med Sci. 2015;31(1):76-81. https://doi.org/10.12669/pjms.311.5647.
67. Kawaguchi R, Yamauchi M, Sugino S, Yamakage M. Ultrasound-aided ipsilateral-dominant epidural block for total hip arthroplasty: a randomised controlled single-blind study. Eur J Anaesthesiol. 2011;28(2):137-140. https://doi.org/10.1097/EJA.0b013e3283423457.
68. Grau T, Leipold RW, Horter J, Martin E, Motsch J. Colour Doppler imaging of the interspinous and epidural space. Eur J Anaesthesiol. 2001;18(11):706-712. https://doi.org/10.1097/00003643-200111000-00002.
69. Arzola C, Davies S, Rofaeel A, Carvalho JC. Ultrasound using the transverse approach to the lumbar spine provides reliable landmarks for labor epidurals. Anesth Analg. 2007;104(5):1188-92, tables of contents. https://doi.org/10.1213/01.ane.0000250912.66057.41.
70. Chauhan AK, Bhatia R, Agrawal S. Lumbar epidural depth using transverse ultrasound scan and its correlation with loss of resistance technique: a prospective observational study in Indian population. Saudi J Anaesth. 2018;12(2):279-282. https://doi.org/10.4103/sja.SJA_679_17.
71. Gnaho A, Nguyen V, Villevielle T, et al. Assessing the depth of the subarachnoid space by ultrasound. Rev Bras Anestesiol. 2012;62(4):520-530. https://doi.org/10.1016/S0034-7094(12)70150-2.
72. Cork RC, Kryc JJ, Vaughan RW. Ultrasonic localization of the lumbar epidural space. Anesthesiology. 1980;52(6):513-516. https://doi.org/10.1097/00000542-198006000-00013.
73. Barsuk JH, Cohen ER, Caprio T, et al. Simulation-based education with mastery learning improves residents’ lumbar puncture skills. Neurology. 2012;79(2):132-137. https://doi.org/10.1212/WNL.0b013e31825dd39d.
74. Lenchus J, Issenberg SB, Murphy D, et al. A blended approach to invasive bedside procedural instruction. Med Teach. 2011;33(2):116-123. https://doi.org/10.3109/0142159X.2010.509412.
75. Wayne DB, Cohen ER, Singer BD, et al. Progress toward improving medical school graduates’ skills via a “boot camp” curriculum. Simul Healthc. 2014;9(1):33-39. https://doi.org/10.1097/SIH.0000000000000001.
76. Cohen ER, Barsuk JH, Moazed F, et al. Making July safer: simulation-based mastery learning during intern boot camp. Acad Med. 2013;88(2):233-239. https://doi.org/10.1097/ACM.0b013e31827bfc0a.
77. Martin R, Gannon D, Riggle J, et al. A comprehensive workshop using simulation to train internal medicine residents in bedside procedures performed by internists. Chest. 2012;142(4):545A. https://doi.org/10.1378/chest.1390093.
78. Lenchus JD. End of the “see one, do one, teach one” era: the next generation of invasive bedside procedural instruction. J Am Osteopath Assoc. 2010;110(6):340-346. PubMed
79. Mourad M, Ranji S, Sliwka D. A randomized controlled trial of the impact of a teaching procedure service on the training of internal medicine residents. J Grad Med Educ. 2012;4(2):170-175. https://doi.org/10.4300/JGME-D-11-00136.1.
80. Restrepo CG, Baker MD, Pruitt CM, Gullett JP, Pigott DC. Ability of pediatric emergency medicine physicians to identify anatomic landmarks with the assistance of ultrasound prior to lumbar puncture in a simulated obese model. Pediatr Emerg Care. 2015;31(1):15-19. https://doi.org/10.1097/PEC.0000000000000330.
81. VanderWielen BA, Harris R, Galgon RE, VanderWielen LM, Schroeder KM. Teaching sonoanatomy to anesthesia faculty and residents: utility of hands-on gel phantom and instructional video training models. J Clin Anesth. 2015;27(3):188-194. https://doi.org/10.1016/j.jclinane.2014.07.007.
82. Keri Z, Sydor D, Ungi T, et al. Computerized training system for ultrasound-guided lumbar puncture on abnormal spine models: a randomized controlled trial. Can J Anaesth. 2015;62(7):777-784. https://doi.org/10.1007/s12630-015-0367-2.
83. Deacon AJ, Melhuishi NS, Terblanche NC. CUSUM method for construction of trainee spinal ultrasound learning curves following standardised teaching. Anaesth Intensive Care. 2014;42(4):480-486. https://doi.org/10.1177/0310057X1404200409.
84. Margarido CB, Arzola C, Balki M, Carvalho JC. Anesthesiologists’ learning curves for ultrasound assessment of the lumbar spine. Can J Anaesth. 2010;57(2):120-126. https://doi.org/10.1007/s12630-009-9219-2.
85. Jensen TP, Soni NJ, Tierney DM, Lucas BP. Hospital privileging practices for bedside procedures: a survey of hospitalist experts. J Hosp Med. 2017;12(10):836-839. https://doi.org/10.12788/jhm.2837.
86. Terblanche NC, Arzola C, Wills KE, et al. Standardised training program in spinal ultrasound for epidural insertion: protocol driven versus non-protocol driven teaching approach. Anaesth Intensive Care. 2014;42(4):460-466. https://doi.org/10.1177/0310057X1404200406.
87. Mofidi M, Mohammadi M, Saidi H, et al. Ultrasound guided lumbar puncture in emergency department: time saving and less complications. J Res Med Sci. 2013;18(4):303-307. PubMed
88. Karmakar MK, Li X, Ho AM, Kwok WH, Chui PT. Real-time ultrasound-guided paramedian epidural access: evaluation of a novel in-plane technique. Br J Anaesth. 2009;102(6):845-854. https://doi.org/10.1093/bja/aep079.
89. Tran D, Kamani AA, Al-Attas E, et al. Single-operator real-time ultrasound-guidance to aim and insert a lumbar epidural needle. Can J Anaesth. 2010;57(4):313-321. https://doi.org/10.1007/s12630-009-9252-1.
90. Liu Y, Qian W, Ke XJ, Mei W. Real-time ultrasound-guided spinal anesthesia using a new paramedian transverse approach. Curr Med Sci. 2018;38(5):910-913. https://doi.org/10.1007/s11596-018-1961-7.
91. Conroy PH, Luyet C, McCartney CJ, McHardy PG. Real-time ultrasound-guided spinal anaesthesia: a prospective observational study of a new approach. Anesthesiol Res Pract. 2013;2013:525818. https://doi.org/10.1155/2013/525818.
92. Brinkmann S, Tang R, Sawka A, Vaghadia H. Single-operator real-time ultrasound-guided spinal injection using SonixGPS™: a case series. Can J Anaesth. 2013;60(9):896-901. https://doi.org/10.1007/s12630-013-9984-9.
93. Niazi AU, Chin KJ, Jin R, Chan VW. Real-time ultrasound-guided spinal anesthesia using the SonixGPS ultrasound guidance system: a feasibility study. Acta Anaesthesiol Scand. 2014;58(7):875-881. https://doi.org/10.1111/aas.12353.

Issue
Journal of Hospital Medicine 14(10)
Issue
Journal of Hospital Medicine 14(10)
Page Number
591-601. Published online first June 10, 2019
Page Number
591-601. Published online first June 10, 2019
Publications
Publications
Topics
Article Type
Sections
Article Source

© 2019 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Nilam J Soni, MD, MSc; E-mail: sonin@uthscsa.edu; Telephone: 210-743-6030.
Content Gating
Open Access (article Unlocked/Open Access)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media
Media Files

Point-of-Care Ultrasound for Hospitalists: A Position Statement of the Society of Hospital Medicine

Article Type
Changed
Wed, 03/17/2021 - 08:24

Many hospitalists incorporate point-of-care ultrasound (POCUS) into their daily practice because it adds value to their bedside evaluation of patients. However, standards for training and assessing hospitalists in POCUS have not yet been established. Other acute care specialties, including emergency medicine and critical care medicine, have already incorporated POCUS into their graduate medical education training programs, but most internal medicine residency programs are only beginning to provide POCUS training.1

Several features distinguish POCUS from comprehensive ultrasound examinations. First, POCUS is designed to answer focused questions, whereas comprehensive ultrasound examinations evaluate all organs in an anatomical region; for example, an abdominal POCUS exam may evaluate only for presence or absence of intraperitoneal free fluid, whereas a comprehensive examination of the right upper quadrant will evaluate the liver, gallbladder, and biliary ducts. Second, POCUS examinations are generally performed by the same clinician who generates the relevant clinical question to answer with POCUS and ultimately integrates the findings into the patient’s care.2 By contrast, comprehensive ultrasound examinations involve multiple providers and steps: a clinician generates a relevant clinical question and requests an ultrasound examination that is acquired by a sonographer, interpreted by a radiologist, and reported back to the requesting clinician. Third, POCUS is often used to evaluate multiple body systems. For example, to evaluate a patient with undifferentiated hypotension, a multisystem POCUS examination of the heart, inferior vena cava, lungs, abdomen, and lower extremity veins is typically performed. Finally, POCUS examinations can be performed serially to investigate changes in clinical status or evaluate response to therapy, such as monitoring the heart, lungs, and inferior vena cava during fluid resuscitation.

The purpose of this position statement is to inform a broad audience about how hospitalists are using diagnostic and procedural applications of POCUS. This position statement does not mandate that hospitalists use POCUS. Rather, it is intended to provide guidance on the safe and effective use of POCUS by the hospitalists who use it and the administrators who oversee its use. We discuss POCUS (1) applications, (2) training, (3) assessments, and (4) program management. This position statement was reviewed and approved by the Society of Hospital Medicine (SHM) Executive Committee in March 2018.

 

 

APPLICATIONS

Common diagnostic and procedural applications of POCUS used by hospitalists are listed in Table 1. Selected evidence supporting the use of these applications is described in the supplementary online content (Appendices 1–8 available at http://journalofhospitalmedicine.com) and SHM position statements on specific ultrasound-guided bedside procedures.3,4 Additional applications not listed in Table 1 that may be performed by some hospitalists include assessment of the eyes, stomach, bowels, ovaries, pregnancy, and testicles, as well as performance of regional anesthesia. Moreover, hospitalists caring for pediatric and adolescent patients may use additional applications besides those listed here. Currently, many hospitalists already perform more complex and sophisticated POCUS examinations than those listed in Table 1. The scope of POCUS use by hospitalists continues to expand, and this position statement should not restrict that expansion.

As outlined in our earlier position statements,3,4 ultrasound guidance lowers complication rates and increases success rates of invasive bedside procedures. Diagnostic POCUS can guide clinical decision making prior to bedside procedures. For instance, hospitalists may use POCUS to assess the size and character of a pleural effusion to help determine the most appropriate management strategy: observation, medical treatment, thoracentesis, chest tube placement, or surgical therapy. Furthermore, diagnostic POCUS can be used to rapidly assess for immediate postprocedural complications, such as pneumothorax, or if the patient develops new symptoms.

TRAINING

Basic Knowledge

Basic knowledge includes fundamentals of ultrasound physics; safety;4 anatomy; physiology; and device operation, including maintenance and cleaning. Basic knowledge can be taught by multiple methods, including live or recorded lectures, online modules, or directed readings.

Image Acquisition

Training should occur across multiple types of patients (eg, obese, cachectic, postsurgical) and clinical settings (eg, intensive care unit, general medicine wards, emergency department) when available. Training is largely hands-on because the relevant skills involve integration of 3D anatomy with spatial manipulation, hand-eye coordination, and fine motor movements. Virtual reality ultrasound simulators may accelerate mastery, particularly for cardiac image acquisition, and expose learners to standardized sets of pathologic findings. Real-time bedside feedback on image acquisition is ideal because understanding how ultrasound probe manipulation affects the images acquired is essential to learning.

Image Interpretation

Training in image interpretation relies on visual pattern recognition of normal and abnormal findings. Therefore, the normal to abnormal spectrum should be broad, and learners should maintain a log of what abnormalities have been identified. Giving real-time feedback at the bedside is ideal because of the connection between image acquisition and interpretation. Image interpretation can be taught through didactic sessions, image review sessions, or review of teaching files with annotated images.

Clinical Integration

Learners must interpret and integrate image findings with other clinical data considering the image quality, patient characteristics, and changing physiology. Clinical integration should be taught by instructors that share similar clinical knowledge as learners. Although sonographers are well suited to teach image acquisition, they should not be the sole instructors to teach hospitalists how to integrate ultrasound findings in clinical decision making. Likewise, emphasis should be placed on the appropriate use of POCUS within a provider’s skill set. Learners must appreciate the clinical significance of POCUS findings, including recognition of incidental findings that may require further workup. Supplemental training in clinical integration can occur through didactics that include complex patient scenarios.

 

 

Pathways

Clinical competency can be achieved with training adherent to five criteria. First, the training environment should be similar to where the trainee will practice. Second, training and feedback should occur in real time. Third, specific applications should be taught rather than broad training in “hospitalist POCUS.” Each application requires unique skills and knowledge, including image acquisition pitfalls and artifacts. Fourth, clinical competence must be achieved and demonstrated; it is not necessarily gained through experience. Fifth, once competency is achieved, continued education and feedback are necessary to ensure it is maintained.

Residency-based POCUS training pathways can best fulfill these criteria. They may eventually become commonplace, but until then alternative pathways must exist for hospitalist providers who are already in practice. There are three important attributes of such pathways. First, administrators’ expectations about learners’ clinical productivity must be realistically, but only temporarily, relaxed; otherwise, competing demands on time will likely overwhelm learners and subvert training. Second, training should begin through a local or national hands-on training program. The SHM POCUS certificate program consolidates training for common diagnostic POCUS applications for hospitalists.6 Other medical societies offer training for their respective clinical specialties.7 Third, once basic POCUS training has begun, longitudinal training should continue ideally with a local hospitalist POCUS expert.

In some settings, a subgroup of hospitalists may not desire, or be able to achieve, competency in the manual skills of POCUS image acquisition. Nevertheless, hospitalists may still find value in understanding POCUS nomenclature, image pattern recognition, and the evidence and pitfalls behind clinical integration of specific POCUS findings. This subset of POCUS skills allows hospitalists to communicate effectively with and understand the clinical decisions made by their colleagues who are competent in POCUS use.

The minimal skills a hospitalist should possess to serve as a POCUS trainer include proficiency of basic knowledge, image acquisition, image interpretation, and clinical integration of the POCUS applications being taught; effectiveness as a hands-on instructor to teach image acquisition skills; and an in-depth understanding of common POCUS pitfalls and limitations.

ASSESSMENTS

Assessment methods for POCUS can include the following: knowledge-based questions, image acquisition using task-specific checklists on human or simulation models, image interpretation using a series of videos or still images with normal and abnormal findings, clinical integration using “next best step” in a multiple choice format with POCUS images, and simulation-based clinical scenarios. Assessment methods should be aligned with local availability of resources and trainers.

Basic Knowledge

Basic knowledge can be assessed via multiple choice questions assessing knowledge of ultrasound physics, image optimization, relevant anatomy, and limitations of POCUS imaging. Basic knowledge lies primarily in the cognitive domain and does not assess manual skills.

Image Acquisition

Image acquisition can be assessed by observation and rating of image quality. Where resources allow, assessment of image acquisition is likely best done through a combination of developing an image portfolio with a minimum number of high quality images, plus direct observation of image acquisition by an expert. Various programs have utilized minimum numbers of images acquired to help define competence with image acquisition skills.6–8 Although minimums may be a necessary step to gain competence, using them as a sole means to determine competence does not account for variable learning curves.9 As with other manual skills in hospital medicine, such as ultrasound-guided bedside procedures, minimum numbers are best used as a starting point for assessments.3,10 In this regard, portfolio development with meticulous attention to the gain, depth, and proper tomographic plane of images can monitor a hospitalist’s progress toward competence by providing objective assessments and feedback. Simulation may also be used as it allows assessment of image acquisition skills and an opportunity to provide real-time feedback, similar to direct observation but without actual patients.

 

 

Image Interpretation

Image interpretation is best assessed by an expert observing the learner at bedside; however, when bedside assessment is not possible, image interpretation skills may be assessed using multiple choice or free text interpretation of archived ultrasound images with normal and abnormal findings. This is often incorporated into the portfolio development portion of a training program, as learners can submit their image interpretation along with the video clip. Both normal and abnormal images can be used to assess anatomic recognition and interpretation. Emphasis should be placed on determining when an image is suboptimal for diagnosis (eg, incomplete exam or poor-quality images). Quality assurance programs should incorporate structured feedback sessions.

Clinical Integration

Assessment of clinical integration can be completed through case scenarios that assess knowledge, interpretation of images, and integration of findings into clinical decision making, which is often delivered via a computer-based assessment. Assessments should combine specific POCUS applications to evaluate common clinical problems in hospital medicine, such as undifferentiated hypotension and dyspnea. High-fidelity simulators can be used to blend clinical case scenarios with image acquisition, image interpretation, and clinical integration. When feasible, comprehensive feedback on how providers acquire, interpret, and apply ultrasound at the bedside is likely the best mechanism to assess clinical integration. This process can be done with a hospitalist’s own patients.

General Assessment

A general assessment that includes a summative knowledge and hands-on skills assessment using task-specific checklists can be performed upon completion of training. A high-fidelity simulator with dynamic or virtual anatomy can provide reproducible standardized assessments with variation in the type and difficulty of cases. When available, we encourage the use of dynamic assessments on actual patients that have both normal and abnormal ultrasound findings because simulated patient scenarios have limitations, even with the use of high-fidelity simulators. Programs are recommended to use formative and summative assessments for evaluation. Quantitative scoring systems using checklists are likely the best framework.11,12

CERTIFICATES AND CERTIFICATION

A certificate of completion is proof of a provider’s participation in an educational activity; it does not equate with competency, though it may be a step toward it. Most POCUS training workshops and short courses provide certificates of completion. Certification of competency is an attestation of a hospitalist’s basic competence within a defined scope of practice (Table 2).13 However, without longitudinal supervision and feedback, skills can decay; therefore, we recommend a longitudinal training program that provides mentored feedback and incorporates periodic competency assessments. At present, no national board certification in POCUS is available to grant external certification of competency for hospitalists.

External Certificate

Certificates of completion can be external through a national organization. An external certificate of completion designed for hospitalists includes the POCUS Certificate of Completion offered by SHM in collaboration with CHEST.6 This certificate program provides regional training options and longitudinal portfolio development. Other external certificates are also available to hospitalists.7,14,15

Most hospitalists are boarded by the American Board of Internal Medicine or the American Board of Family Medicine. These boards do not yet include certification of competency in POCUS. Other specialty boards, such as emergency medicine, include competency in POCUS. For emergency medicine, completion of an accredited residency training program and certification by the national board includes POCUS competency.

 

 

Internal Certificate

There are a few examples of successful local institutional programs that have provided internal certificates of competency.12,14 Competency assessments require significant resources including investment by both faculty and learners. Ongoing evaluation of competency should be based on quality assurance processes.

Credentialing and Privileging

The American Medical Association (AMA) House of Delegates in 1999 passed a resolution (AMA HR. 802) recommending hospitals follow specialty-specific guidelines for privileging decisions related to POCUS use.17 The resolution included a statement that, “ultrasound imaging is within the scope of practice of appropriately trained physicians.”

Some institutions have begun to rely on a combination of internal and external certificate programs to grant privileges to hospitalists.10 Although specific privileges for POCUS may not be required in some hospitals, some institutions may require certification of training and assessments prior to granting permission to use POCUS.

Hospitalist programs are encouraged to evaluate ongoing POCUS use by their providers after granting initial permission. If privileging is instituted by a hospital, hospitalists must play a significant role in determining the requirements for privileging and ongoing maintenance of skills.

Maintenance of Skills

All medical skills can decay with disuse, including those associated with POCUS.12,18 Thus, POCUS users should continue using POCUS regularly in clinical practice and participate in POCUS continuing medical education activities, ideally with ongoing assessments. Maintenance of skills may be confirmed through routine participation in a quality assurance program.

PROGRAM MANAGEMENT

Use of POCUS in hospital medicine has unique considerations, and hospitalists should be integrally involved in decision making surrounding institutional POCUS program management. Appointing a dedicated POCUS director can help a program succeed.8

Equipment and Image Archiving

Several factors are important to consider when selecting an ultrasound machine: portability, screen size, and ease of use; integration with the electronic medical record and options for image archiving; manufacturer’s service plan, including technical and clinical support; and compliance with local infection control policies. The ability to easily archive and retrieve images is essential for quality assurance, continuing education, institutional quality improvement, documentation, and reimbursement. In certain scenarios, image archiving may not be possible (such as with personal handheld devices or in emergency situations) or necessary (such as with frequent serial examinations during fluid resuscitation). An image archive is ideally linked to reports, orders, and billing software.10,19 If such linkages are not feasible, parallel external storage that complies with regulatory standards (ie, HIPAA compliance) may be suitable.20

Documentation and Billing

Components of documentation include the indication and type of ultrasound examination performed, date and time of the examination, patient identifying information, name of provider(s) acquiring and interpreting the images, specific scanning protocols used, patient position, probe used, and findings. Documentation can occur through a standalone note or as part of another note, such as a progress note. Whenever possible, documentation should be timely to facilitate communication with other providers.

Billing is supported through the AMA Current Procedural Terminology codes for “focused” or “limited” ultrasound examinations (Appendix 9). The following three criteria must be satisfied for billing. First, images must be permanently stored. Specific requirements vary by insurance policy, though current practice suggests a minimum of one image demonstrating relevant anatomy and pathology for the ultrasound examination coded. For ultrasound-guided procedures that require needle insertion, images should be captured at the point of interest, and a procedure note should reflect that the needle was guided and visualized under ultrasound.21 Second, proper documentation must be entered in the medical record. Third, local institutional privileges for POCUS must be considered. Although privileges are not required to bill, some hospitals or payers may require them.

 

 

Quality Assurance

Published guidelines on quality assurance in POCUS are available from different specialty organizations, including emergency medicine, pediatric emergency medicine, critical care, anesthesiology, obstetrics, and cardiology.8,22–28 Quality assurance is aimed at ensuring that physicians maintain basic competency in using POCUS to influence bedside decisions.

Quality assurance should be carried out by an individual or committee with expertise in POCUS. Multidisciplinary QA programs in which hospital medicine providers are working collaboratively with other POCUS providers have been demonstrated to be highly effective.10 Oversight includes ensuring that providers using POCUS are appropriately trained,10,22,28 using the equipment correctly,8,26,28 and documenting properly. Some programs have implemented mechanisms to review and provide feedback on image acquisition, interpretation, and clinical integration.8,10 Other programs have compared POCUS findings with referral studies, such as comprehensive ultrasound examinations.

CONCLUSIONS

Practicing hospitalists must continue to collaborate with their institutions to build POCUS capabilities. In particular, they must work with their local privileging body to determine what credentials are required. The distinction between certificates of completion and certificates of competency, including whether those certificates are internal or external, is important in the credentialing process.

External certificates of competency are currently unavailable for most practicing hospitalists because ABIM certification does not include POCUS-related competencies. As internal medicine residency training programs begin to adopt POCUS training and certification into their educational curricula, we foresee a need to update the ABIM Policies and Procedures for Certification. Until then, we recommend that certificates of competency be defined and granted internally by local hospitalist groups.

Given the many advantages of POCUS over traditional tools, we anticipate its increasing implementation among hospitalists in the future. As with all medical technology, its role in clinical care should be continuously reexamined and redefined through health services research. Such information will be useful in developing practice guidelines, educational curricula, and training standards.

Acknowledgments

The authors would like to thank all members that participated in the discussion and finalization of this position statement during the Point-of-care Ultrasound Faculty Retreat at the 2018 Society of Hospital Medicine Annual Conference: Saaid Abdel-Ghani, Brandon Boesch, Joel Cho, Ria Dancel, Renee Dversdal, Ricardo Franco-Sadud, Benjamin Galen, Trevor P. Jensen, Mohit Jindal, Gordon Johnson, Linda M. Kurian, Gigi Liu, Charles M. LoPresti, Brian P. Lucas, Venkat Kalidindi, Benji Matthews, Anna Maw, Gregory Mints, Kreegan Reierson, Gerard Salame, Richard Schildhouse, Daniel Schnobrich, Nilam Soni, Kirk Spencer, Hiromizu Takahashi, David M. Tierney, Tanping Wong, and Toru Yamada.

Files
References

1. Schnobrich DJ, Mathews BK, Trappey BE, Muthyala BK, Olson APJ. Entrusting internal medicine residents to use point of care ultrasound: Towards improved assessment and supervision. Med Teach. 2018:1-6. doi:10.1080/0142159X.2018.1457210.
2. Soni NJ, Lucas BP. Diagnostic point-of-care ultrasound for hospitalists. J Hosp Med. 2015;10(2):120-124. doi:10.1002/jhm.2285.
3. Lucas BP, Tierney DM, Jensen TP, et al. Credentialing of hospitalists in ultrasound-guided bedside procedures: a position statement of the society of hospital medicine. J Hosp Med. 2018;13(2):117-125. doi:10.12788/jhm.2917.
4. Dancel R, Schnobrich D, Puri N, et al. Recommendations on the use of ultrasound guidance for adult thoracentesis: a position statement of the society of hospital medicine. J Hosp Med. 2018;13(2):126-135. doi:10.12788/jhm.2940.
5. National Council on Radiation Protection and Measurements, The Council. Implementation of the Principle of as Low as Reasonably Achievable (ALARA) for Medical and Dental Personnel.; 1990.
6. Society of Hospital Medicine. Point of Care Ultrasound course: https://www.hospitalmedicine.org/clinical-topics/ultrasonography-cert/. Accessed February 6, 2018.
7. Critical Care Ultrasonography Certificate of Completion Program. CHEST. American College of Chest Physicians. http://www.chestnet.org/Education/Advanced-Clinical-Training/Certificate-of-Completion-Program/Critical-Care-Ultrasonography. Accessed February 6, 2018.
8. American College of Emergency Physicians Policy Statement: Emergency Ultrasound Guidelines. 2016. https://www.acep.org/Clinical---Practice-Management/ACEP-Ultrasound-Guidelines/. Accessed February 6, 2018.
9. Blehar DJ, Barton B, Gaspari RJ. Learning curves in emergency ultrasound education. Acad Emerg Med. 2015;22(5):574-582. doi:10.1111/acem.12653.
10. Mathews BK, Zwank M. Hospital medicine point of care ultrasound credentialing: an example protocol. J Hosp Med. 2017;12(9):767-772. doi:10.12788/jhm.2809.
11. Barsuk JH, McGaghie WC, Cohen ER, Balachandran JS, Wayne DB. Use of simulation-based mastery learning to improve the quality of central venous catheter placement in a medical intensive care unit. J Hosp Med. 2009;4(7):397-403. doi:10.1002/jhm.468.
12. Mathews BK, Reierson K, Vuong K, et al. The design and evaluation of the Comprehensive Hospitalist Assessment and Mentorship with Portfolios (CHAMP) ultrasound program. J Hosp Med. 2018;13(8):544-550. doi:10.12788/jhm.2938.
13. Soni NJ, Tierney DM, Jensen TP, Lucas BP. Certification of point-of-care ultrasound competency. J Hosp Med. 2017;12(9):775-776. doi:10.12788/jhm.2812.
14. Ultrasound Certification for Physicians. Alliance for Physician Certification and Advancement. APCA. https://apca.org/. Accessed February 6, 2018.
15. National Board of Echocardiography, Inc. https://www.echoboards.org/EchoBoards/News/2019_Adult_Critical_Care_Echocardiography_Exam.aspx. Accessed June 18, 2018.
16. Tierney DM. Internal Medicine Bedside Ultrasound Program (IMBUS). Abbott Northwestern. http://imbus.anwresidency.com/index.html. Accessed February 6, 2018.
17. American Medical Association House of Delegates Resolution H-230.960: Privileging for Ultrasound Imaging. Resolution 802. Policy Finder Website. http://search0.ama-assn.org/search/pfonline. Published 1999. Accessed February 18, 2018.
18. Kelm D, Ratelle J, Azeem N, et al. Longitudinal ultrasound curriculum improves long-term retention among internal medicine residents. J Grad Med Educ. 2015;7(3):454-457. doi:10.4300/JGME-14-00284.1.
19. Flannigan MJ, Adhikari S. Point-of-care ultrasound work flow innovation: impact on documentation and billing. J Ultrasound Med. 2017;36(12):2467-2474. doi:10.1002/jum.14284.
20. Emergency Ultrasound: Workflow White Paper. https://www.acep.org/uploadedFiles/ACEP/memberCenter/SectionsofMembership/ultra/Workflow%20White%20Paper.pdf. Published 2013. Accessed February 18, 2018.
21. Ultrasound Coding and Reimbursement Document 2009. Emergency Ultrasound Section. American College of Emergency Physicians. http://emergencyultrasoundteaching.com/assets/2009_coding_update.pdf. Published 2009. Accessed February 18, 2018.
22. Mayo PH, Beaulieu Y, Doelken P, et al. American College of Chest Physicians/La Societe de Reanimation de Langue Francaise statement on competence in critical care ultrasonography. Chest. 2009;135(4):1050-1060. doi:10.1378/chest.08-2305.
23. Frankel HL, Kirkpatrick AW, Elbarbary M, et al. Guidelines for the appropriate use of bedside general and cardiac ultrasonography in the evaluation of critically ill patients-part I: general ultrasonography. Crit Care Med. 2015;43(11):2479-2502. doi:10.1097/ccm.0000000000001216.
24. Levitov A, Frankel HL, Blaivas M, et al. Guidelines for the appropriate use of bedside general and cardiac ultrasonography in the evaluation of critically ill patients-part ii: cardiac ultrasonography. Crit Care Med. 2016;44(6):1206-1227. doi:10.1097/ccm.0000000000001847.
25. ACR–ACOG–AIUM–SRU Practice Parameter for the Performance of Obstetrical Ultrasound. https://www.acr.org/-/media/ACR/Files/Practice-Parameters/us-ob.pdf. Published 2013. Accessed February 18, 2018.
26. AIUM practice guideline for documentation of an ultrasound examination. J Ultrasound Med. 2014;33(6):1098-1102. doi:10.7863/ultra.33.6.1098.
27. Marin JR, Lewiss RE. Point-of-care ultrasonography by pediatric emergency medicine physicians. Pediatrics. 2015;135(4):e1113-e1122. doi:10.1542/peds.2015-0343.
28. Spencer KT, Kimura BJ, Korcarz CE, Pellikka PA, Rahko PS, Siegel RJ. Focused cardiac ultrasound: recommendations from the American Society of Echocardiography. J Am Soc Echocardiogr. 2013;26(6):567-581. doi:10.1016/j.echo.2013.04.001.

Article PDF
Author and Disclosure Information

1Division of General & Hospital Medicine, The University of Texas Health San Antonio, San Antonio, Texas; 2Section of Hospital Medicine, South Texas Veterans Health Care System, San Antonio, Texas; 3Divisions of General Internal Medicine and Hospital Pediatrics, University of Minnesota, Minneapolis, Minnesota; 4Department of Hospital Medicine, HealthPartners Medical Group, Regions Hospital, St. Paul, Minnesota; 5Department of Medical Education, Abbott Northwestern Hospital, Minneapolis, Minnesota; 6Division of Hospital Medicine, Department of Medicine, University of California San Francisco, San Francisco, California; 7Division of Hospital Medicine, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina; 8Division of General Pediatrics and Adolescent Medicine, Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina; 9Department of Hospital Medicine, Kaiser Permanente San Francisco Medical Center, San Francisco, California; 10Division of Hospital Medicine, Oregon Health & Science University, Portland, Oregon; 11Division of Hospital Medicine, Weill Cornell Medicine, New York, New York; 12Division of General Internal Medicine, Mayo Clinic, Rochester, Minnesota; 13Division of Hospital Medicine, Zucker School of Medicine at Hofstra Northwell, New Hyde Park, New York; 14Hospitalist Program, Division of General Internal Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; 15Division of Hospital Medicine, University of California Davis, Davis, California; 16Division of Hospital Medicine, Alameda Health System-Highland Hospital, Oakland, California; 17Louis Stokes Cleveland Veterans Affairs Hospital, Cleveland, Ohio; 18Case Western Reserve University School of Medicine, Cleveland, Ohio; 19Division of Hospital Medicine, University of Miami, Miami, Florida; 20Division of Hospital Medicine, Legacy Healthcare System, Portland, Oregon; 21Division of Hospital Medicine, University of Colorado, Aurora, Colorado; 22Department of Medicine, University of Central Florida, Naples, Florida; 23White River Junction VA Medical Center, White River Junction, Vermont; 24Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire.

Funding

Nilam Soni: Department of Veterans Affairs, Quality Enhancement Research Initiative (QUERI) Partnered Evaluation Initiative Grant (HX002263-01A1). Brian P Lucas: Department of Veterans Affairs, Veterans Health Administration, Office of Research and Development and Dartmouth SYNERGY, National Institutes of Health, National Center for Translational Science (UL1TR001086)

Disclaimer

The contents of this publication do not represent the views of the US Department of Veterans Affairs or the United States Government.

Publications
Topics
Sections
Files
Files
Author and Disclosure Information

1Division of General & Hospital Medicine, The University of Texas Health San Antonio, San Antonio, Texas; 2Section of Hospital Medicine, South Texas Veterans Health Care System, San Antonio, Texas; 3Divisions of General Internal Medicine and Hospital Pediatrics, University of Minnesota, Minneapolis, Minnesota; 4Department of Hospital Medicine, HealthPartners Medical Group, Regions Hospital, St. Paul, Minnesota; 5Department of Medical Education, Abbott Northwestern Hospital, Minneapolis, Minnesota; 6Division of Hospital Medicine, Department of Medicine, University of California San Francisco, San Francisco, California; 7Division of Hospital Medicine, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina; 8Division of General Pediatrics and Adolescent Medicine, Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina; 9Department of Hospital Medicine, Kaiser Permanente San Francisco Medical Center, San Francisco, California; 10Division of Hospital Medicine, Oregon Health & Science University, Portland, Oregon; 11Division of Hospital Medicine, Weill Cornell Medicine, New York, New York; 12Division of General Internal Medicine, Mayo Clinic, Rochester, Minnesota; 13Division of Hospital Medicine, Zucker School of Medicine at Hofstra Northwell, New Hyde Park, New York; 14Hospitalist Program, Division of General Internal Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; 15Division of Hospital Medicine, University of California Davis, Davis, California; 16Division of Hospital Medicine, Alameda Health System-Highland Hospital, Oakland, California; 17Louis Stokes Cleveland Veterans Affairs Hospital, Cleveland, Ohio; 18Case Western Reserve University School of Medicine, Cleveland, Ohio; 19Division of Hospital Medicine, University of Miami, Miami, Florida; 20Division of Hospital Medicine, Legacy Healthcare System, Portland, Oregon; 21Division of Hospital Medicine, University of Colorado, Aurora, Colorado; 22Department of Medicine, University of Central Florida, Naples, Florida; 23White River Junction VA Medical Center, White River Junction, Vermont; 24Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire.

Funding

Nilam Soni: Department of Veterans Affairs, Quality Enhancement Research Initiative (QUERI) Partnered Evaluation Initiative Grant (HX002263-01A1). Brian P Lucas: Department of Veterans Affairs, Veterans Health Administration, Office of Research and Development and Dartmouth SYNERGY, National Institutes of Health, National Center for Translational Science (UL1TR001086)

Disclaimer

The contents of this publication do not represent the views of the US Department of Veterans Affairs or the United States Government.

Author and Disclosure Information

1Division of General & Hospital Medicine, The University of Texas Health San Antonio, San Antonio, Texas; 2Section of Hospital Medicine, South Texas Veterans Health Care System, San Antonio, Texas; 3Divisions of General Internal Medicine and Hospital Pediatrics, University of Minnesota, Minneapolis, Minnesota; 4Department of Hospital Medicine, HealthPartners Medical Group, Regions Hospital, St. Paul, Minnesota; 5Department of Medical Education, Abbott Northwestern Hospital, Minneapolis, Minnesota; 6Division of Hospital Medicine, Department of Medicine, University of California San Francisco, San Francisco, California; 7Division of Hospital Medicine, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina; 8Division of General Pediatrics and Adolescent Medicine, Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina; 9Department of Hospital Medicine, Kaiser Permanente San Francisco Medical Center, San Francisco, California; 10Division of Hospital Medicine, Oregon Health & Science University, Portland, Oregon; 11Division of Hospital Medicine, Weill Cornell Medicine, New York, New York; 12Division of General Internal Medicine, Mayo Clinic, Rochester, Minnesota; 13Division of Hospital Medicine, Zucker School of Medicine at Hofstra Northwell, New Hyde Park, New York; 14Hospitalist Program, Division of General Internal Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; 15Division of Hospital Medicine, University of California Davis, Davis, California; 16Division of Hospital Medicine, Alameda Health System-Highland Hospital, Oakland, California; 17Louis Stokes Cleveland Veterans Affairs Hospital, Cleveland, Ohio; 18Case Western Reserve University School of Medicine, Cleveland, Ohio; 19Division of Hospital Medicine, University of Miami, Miami, Florida; 20Division of Hospital Medicine, Legacy Healthcare System, Portland, Oregon; 21Division of Hospital Medicine, University of Colorado, Aurora, Colorado; 22Department of Medicine, University of Central Florida, Naples, Florida; 23White River Junction VA Medical Center, White River Junction, Vermont; 24Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire.

Funding

Nilam Soni: Department of Veterans Affairs, Quality Enhancement Research Initiative (QUERI) Partnered Evaluation Initiative Grant (HX002263-01A1). Brian P Lucas: Department of Veterans Affairs, Veterans Health Administration, Office of Research and Development and Dartmouth SYNERGY, National Institutes of Health, National Center for Translational Science (UL1TR001086)

Disclaimer

The contents of this publication do not represent the views of the US Department of Veterans Affairs or the United States Government.

Article PDF
Article PDF
Related Articles

Many hospitalists incorporate point-of-care ultrasound (POCUS) into their daily practice because it adds value to their bedside evaluation of patients. However, standards for training and assessing hospitalists in POCUS have not yet been established. Other acute care specialties, including emergency medicine and critical care medicine, have already incorporated POCUS into their graduate medical education training programs, but most internal medicine residency programs are only beginning to provide POCUS training.1

Several features distinguish POCUS from comprehensive ultrasound examinations. First, POCUS is designed to answer focused questions, whereas comprehensive ultrasound examinations evaluate all organs in an anatomical region; for example, an abdominal POCUS exam may evaluate only for presence or absence of intraperitoneal free fluid, whereas a comprehensive examination of the right upper quadrant will evaluate the liver, gallbladder, and biliary ducts. Second, POCUS examinations are generally performed by the same clinician who generates the relevant clinical question to answer with POCUS and ultimately integrates the findings into the patient’s care.2 By contrast, comprehensive ultrasound examinations involve multiple providers and steps: a clinician generates a relevant clinical question and requests an ultrasound examination that is acquired by a sonographer, interpreted by a radiologist, and reported back to the requesting clinician. Third, POCUS is often used to evaluate multiple body systems. For example, to evaluate a patient with undifferentiated hypotension, a multisystem POCUS examination of the heart, inferior vena cava, lungs, abdomen, and lower extremity veins is typically performed. Finally, POCUS examinations can be performed serially to investigate changes in clinical status or evaluate response to therapy, such as monitoring the heart, lungs, and inferior vena cava during fluid resuscitation.

The purpose of this position statement is to inform a broad audience about how hospitalists are using diagnostic and procedural applications of POCUS. This position statement does not mandate that hospitalists use POCUS. Rather, it is intended to provide guidance on the safe and effective use of POCUS by the hospitalists who use it and the administrators who oversee its use. We discuss POCUS (1) applications, (2) training, (3) assessments, and (4) program management. This position statement was reviewed and approved by the Society of Hospital Medicine (SHM) Executive Committee in March 2018.

 

 

APPLICATIONS

Common diagnostic and procedural applications of POCUS used by hospitalists are listed in Table 1. Selected evidence supporting the use of these applications is described in the supplementary online content (Appendices 1–8 available at http://journalofhospitalmedicine.com) and SHM position statements on specific ultrasound-guided bedside procedures.3,4 Additional applications not listed in Table 1 that may be performed by some hospitalists include assessment of the eyes, stomach, bowels, ovaries, pregnancy, and testicles, as well as performance of regional anesthesia. Moreover, hospitalists caring for pediatric and adolescent patients may use additional applications besides those listed here. Currently, many hospitalists already perform more complex and sophisticated POCUS examinations than those listed in Table 1. The scope of POCUS use by hospitalists continues to expand, and this position statement should not restrict that expansion.

As outlined in our earlier position statements,3,4 ultrasound guidance lowers complication rates and increases success rates of invasive bedside procedures. Diagnostic POCUS can guide clinical decision making prior to bedside procedures. For instance, hospitalists may use POCUS to assess the size and character of a pleural effusion to help determine the most appropriate management strategy: observation, medical treatment, thoracentesis, chest tube placement, or surgical therapy. Furthermore, diagnostic POCUS can be used to rapidly assess for immediate postprocedural complications, such as pneumothorax, or if the patient develops new symptoms.

TRAINING

Basic Knowledge

Basic knowledge includes fundamentals of ultrasound physics; safety;4 anatomy; physiology; and device operation, including maintenance and cleaning. Basic knowledge can be taught by multiple methods, including live or recorded lectures, online modules, or directed readings.

Image Acquisition

Training should occur across multiple types of patients (eg, obese, cachectic, postsurgical) and clinical settings (eg, intensive care unit, general medicine wards, emergency department) when available. Training is largely hands-on because the relevant skills involve integration of 3D anatomy with spatial manipulation, hand-eye coordination, and fine motor movements. Virtual reality ultrasound simulators may accelerate mastery, particularly for cardiac image acquisition, and expose learners to standardized sets of pathologic findings. Real-time bedside feedback on image acquisition is ideal because understanding how ultrasound probe manipulation affects the images acquired is essential to learning.

Image Interpretation

Training in image interpretation relies on visual pattern recognition of normal and abnormal findings. Therefore, the normal to abnormal spectrum should be broad, and learners should maintain a log of what abnormalities have been identified. Giving real-time feedback at the bedside is ideal because of the connection between image acquisition and interpretation. Image interpretation can be taught through didactic sessions, image review sessions, or review of teaching files with annotated images.

Clinical Integration

Learners must interpret and integrate image findings with other clinical data considering the image quality, patient characteristics, and changing physiology. Clinical integration should be taught by instructors that share similar clinical knowledge as learners. Although sonographers are well suited to teach image acquisition, they should not be the sole instructors to teach hospitalists how to integrate ultrasound findings in clinical decision making. Likewise, emphasis should be placed on the appropriate use of POCUS within a provider’s skill set. Learners must appreciate the clinical significance of POCUS findings, including recognition of incidental findings that may require further workup. Supplemental training in clinical integration can occur through didactics that include complex patient scenarios.

 

 

Pathways

Clinical competency can be achieved with training adherent to five criteria. First, the training environment should be similar to where the trainee will practice. Second, training and feedback should occur in real time. Third, specific applications should be taught rather than broad training in “hospitalist POCUS.” Each application requires unique skills and knowledge, including image acquisition pitfalls and artifacts. Fourth, clinical competence must be achieved and demonstrated; it is not necessarily gained through experience. Fifth, once competency is achieved, continued education and feedback are necessary to ensure it is maintained.

Residency-based POCUS training pathways can best fulfill these criteria. They may eventually become commonplace, but until then alternative pathways must exist for hospitalist providers who are already in practice. There are three important attributes of such pathways. First, administrators’ expectations about learners’ clinical productivity must be realistically, but only temporarily, relaxed; otherwise, competing demands on time will likely overwhelm learners and subvert training. Second, training should begin through a local or national hands-on training program. The SHM POCUS certificate program consolidates training for common diagnostic POCUS applications for hospitalists.6 Other medical societies offer training for their respective clinical specialties.7 Third, once basic POCUS training has begun, longitudinal training should continue ideally with a local hospitalist POCUS expert.

In some settings, a subgroup of hospitalists may not desire, or be able to achieve, competency in the manual skills of POCUS image acquisition. Nevertheless, hospitalists may still find value in understanding POCUS nomenclature, image pattern recognition, and the evidence and pitfalls behind clinical integration of specific POCUS findings. This subset of POCUS skills allows hospitalists to communicate effectively with and understand the clinical decisions made by their colleagues who are competent in POCUS use.

The minimal skills a hospitalist should possess to serve as a POCUS trainer include proficiency of basic knowledge, image acquisition, image interpretation, and clinical integration of the POCUS applications being taught; effectiveness as a hands-on instructor to teach image acquisition skills; and an in-depth understanding of common POCUS pitfalls and limitations.

ASSESSMENTS

Assessment methods for POCUS can include the following: knowledge-based questions, image acquisition using task-specific checklists on human or simulation models, image interpretation using a series of videos or still images with normal and abnormal findings, clinical integration using “next best step” in a multiple choice format with POCUS images, and simulation-based clinical scenarios. Assessment methods should be aligned with local availability of resources and trainers.

Basic Knowledge

Basic knowledge can be assessed via multiple choice questions assessing knowledge of ultrasound physics, image optimization, relevant anatomy, and limitations of POCUS imaging. Basic knowledge lies primarily in the cognitive domain and does not assess manual skills.

Image Acquisition

Image acquisition can be assessed by observation and rating of image quality. Where resources allow, assessment of image acquisition is likely best done through a combination of developing an image portfolio with a minimum number of high quality images, plus direct observation of image acquisition by an expert. Various programs have utilized minimum numbers of images acquired to help define competence with image acquisition skills.6–8 Although minimums may be a necessary step to gain competence, using them as a sole means to determine competence does not account for variable learning curves.9 As with other manual skills in hospital medicine, such as ultrasound-guided bedside procedures, minimum numbers are best used as a starting point for assessments.3,10 In this regard, portfolio development with meticulous attention to the gain, depth, and proper tomographic plane of images can monitor a hospitalist’s progress toward competence by providing objective assessments and feedback. Simulation may also be used as it allows assessment of image acquisition skills and an opportunity to provide real-time feedback, similar to direct observation but without actual patients.

 

 

Image Interpretation

Image interpretation is best assessed by an expert observing the learner at bedside; however, when bedside assessment is not possible, image interpretation skills may be assessed using multiple choice or free text interpretation of archived ultrasound images with normal and abnormal findings. This is often incorporated into the portfolio development portion of a training program, as learners can submit their image interpretation along with the video clip. Both normal and abnormal images can be used to assess anatomic recognition and interpretation. Emphasis should be placed on determining when an image is suboptimal for diagnosis (eg, incomplete exam or poor-quality images). Quality assurance programs should incorporate structured feedback sessions.

Clinical Integration

Assessment of clinical integration can be completed through case scenarios that assess knowledge, interpretation of images, and integration of findings into clinical decision making, which is often delivered via a computer-based assessment. Assessments should combine specific POCUS applications to evaluate common clinical problems in hospital medicine, such as undifferentiated hypotension and dyspnea. High-fidelity simulators can be used to blend clinical case scenarios with image acquisition, image interpretation, and clinical integration. When feasible, comprehensive feedback on how providers acquire, interpret, and apply ultrasound at the bedside is likely the best mechanism to assess clinical integration. This process can be done with a hospitalist’s own patients.

General Assessment

A general assessment that includes a summative knowledge and hands-on skills assessment using task-specific checklists can be performed upon completion of training. A high-fidelity simulator with dynamic or virtual anatomy can provide reproducible standardized assessments with variation in the type and difficulty of cases. When available, we encourage the use of dynamic assessments on actual patients that have both normal and abnormal ultrasound findings because simulated patient scenarios have limitations, even with the use of high-fidelity simulators. Programs are recommended to use formative and summative assessments for evaluation. Quantitative scoring systems using checklists are likely the best framework.11,12

CERTIFICATES AND CERTIFICATION

A certificate of completion is proof of a provider’s participation in an educational activity; it does not equate with competency, though it may be a step toward it. Most POCUS training workshops and short courses provide certificates of completion. Certification of competency is an attestation of a hospitalist’s basic competence within a defined scope of practice (Table 2).13 However, without longitudinal supervision and feedback, skills can decay; therefore, we recommend a longitudinal training program that provides mentored feedback and incorporates periodic competency assessments. At present, no national board certification in POCUS is available to grant external certification of competency for hospitalists.

External Certificate

Certificates of completion can be external through a national organization. An external certificate of completion designed for hospitalists includes the POCUS Certificate of Completion offered by SHM in collaboration with CHEST.6 This certificate program provides regional training options and longitudinal portfolio development. Other external certificates are also available to hospitalists.7,14,15

Most hospitalists are boarded by the American Board of Internal Medicine or the American Board of Family Medicine. These boards do not yet include certification of competency in POCUS. Other specialty boards, such as emergency medicine, include competency in POCUS. For emergency medicine, completion of an accredited residency training program and certification by the national board includes POCUS competency.

 

 

Internal Certificate

There are a few examples of successful local institutional programs that have provided internal certificates of competency.12,14 Competency assessments require significant resources including investment by both faculty and learners. Ongoing evaluation of competency should be based on quality assurance processes.

Credentialing and Privileging

The American Medical Association (AMA) House of Delegates in 1999 passed a resolution (AMA HR. 802) recommending hospitals follow specialty-specific guidelines for privileging decisions related to POCUS use.17 The resolution included a statement that, “ultrasound imaging is within the scope of practice of appropriately trained physicians.”

Some institutions have begun to rely on a combination of internal and external certificate programs to grant privileges to hospitalists.10 Although specific privileges for POCUS may not be required in some hospitals, some institutions may require certification of training and assessments prior to granting permission to use POCUS.

Hospitalist programs are encouraged to evaluate ongoing POCUS use by their providers after granting initial permission. If privileging is instituted by a hospital, hospitalists must play a significant role in determining the requirements for privileging and ongoing maintenance of skills.

Maintenance of Skills

All medical skills can decay with disuse, including those associated with POCUS.12,18 Thus, POCUS users should continue using POCUS regularly in clinical practice and participate in POCUS continuing medical education activities, ideally with ongoing assessments. Maintenance of skills may be confirmed through routine participation in a quality assurance program.

PROGRAM MANAGEMENT

Use of POCUS in hospital medicine has unique considerations, and hospitalists should be integrally involved in decision making surrounding institutional POCUS program management. Appointing a dedicated POCUS director can help a program succeed.8

Equipment and Image Archiving

Several factors are important to consider when selecting an ultrasound machine: portability, screen size, and ease of use; integration with the electronic medical record and options for image archiving; manufacturer’s service plan, including technical and clinical support; and compliance with local infection control policies. The ability to easily archive and retrieve images is essential for quality assurance, continuing education, institutional quality improvement, documentation, and reimbursement. In certain scenarios, image archiving may not be possible (such as with personal handheld devices or in emergency situations) or necessary (such as with frequent serial examinations during fluid resuscitation). An image archive is ideally linked to reports, orders, and billing software.10,19 If such linkages are not feasible, parallel external storage that complies with regulatory standards (ie, HIPAA compliance) may be suitable.20

Documentation and Billing

Components of documentation include the indication and type of ultrasound examination performed, date and time of the examination, patient identifying information, name of provider(s) acquiring and interpreting the images, specific scanning protocols used, patient position, probe used, and findings. Documentation can occur through a standalone note or as part of another note, such as a progress note. Whenever possible, documentation should be timely to facilitate communication with other providers.

Billing is supported through the AMA Current Procedural Terminology codes for “focused” or “limited” ultrasound examinations (Appendix 9). The following three criteria must be satisfied for billing. First, images must be permanently stored. Specific requirements vary by insurance policy, though current practice suggests a minimum of one image demonstrating relevant anatomy and pathology for the ultrasound examination coded. For ultrasound-guided procedures that require needle insertion, images should be captured at the point of interest, and a procedure note should reflect that the needle was guided and visualized under ultrasound.21 Second, proper documentation must be entered in the medical record. Third, local institutional privileges for POCUS must be considered. Although privileges are not required to bill, some hospitals or payers may require them.

 

 

Quality Assurance

Published guidelines on quality assurance in POCUS are available from different specialty organizations, including emergency medicine, pediatric emergency medicine, critical care, anesthesiology, obstetrics, and cardiology.8,22–28 Quality assurance is aimed at ensuring that physicians maintain basic competency in using POCUS to influence bedside decisions.

Quality assurance should be carried out by an individual or committee with expertise in POCUS. Multidisciplinary QA programs in which hospital medicine providers are working collaboratively with other POCUS providers have been demonstrated to be highly effective.10 Oversight includes ensuring that providers using POCUS are appropriately trained,10,22,28 using the equipment correctly,8,26,28 and documenting properly. Some programs have implemented mechanisms to review and provide feedback on image acquisition, interpretation, and clinical integration.8,10 Other programs have compared POCUS findings with referral studies, such as comprehensive ultrasound examinations.

CONCLUSIONS

Practicing hospitalists must continue to collaborate with their institutions to build POCUS capabilities. In particular, they must work with their local privileging body to determine what credentials are required. The distinction between certificates of completion and certificates of competency, including whether those certificates are internal or external, is important in the credentialing process.

External certificates of competency are currently unavailable for most practicing hospitalists because ABIM certification does not include POCUS-related competencies. As internal medicine residency training programs begin to adopt POCUS training and certification into their educational curricula, we foresee a need to update the ABIM Policies and Procedures for Certification. Until then, we recommend that certificates of competency be defined and granted internally by local hospitalist groups.

Given the many advantages of POCUS over traditional tools, we anticipate its increasing implementation among hospitalists in the future. As with all medical technology, its role in clinical care should be continuously reexamined and redefined through health services research. Such information will be useful in developing practice guidelines, educational curricula, and training standards.

Acknowledgments

The authors would like to thank all members that participated in the discussion and finalization of this position statement during the Point-of-care Ultrasound Faculty Retreat at the 2018 Society of Hospital Medicine Annual Conference: Saaid Abdel-Ghani, Brandon Boesch, Joel Cho, Ria Dancel, Renee Dversdal, Ricardo Franco-Sadud, Benjamin Galen, Trevor P. Jensen, Mohit Jindal, Gordon Johnson, Linda M. Kurian, Gigi Liu, Charles M. LoPresti, Brian P. Lucas, Venkat Kalidindi, Benji Matthews, Anna Maw, Gregory Mints, Kreegan Reierson, Gerard Salame, Richard Schildhouse, Daniel Schnobrich, Nilam Soni, Kirk Spencer, Hiromizu Takahashi, David M. Tierney, Tanping Wong, and Toru Yamada.

Many hospitalists incorporate point-of-care ultrasound (POCUS) into their daily practice because it adds value to their bedside evaluation of patients. However, standards for training and assessing hospitalists in POCUS have not yet been established. Other acute care specialties, including emergency medicine and critical care medicine, have already incorporated POCUS into their graduate medical education training programs, but most internal medicine residency programs are only beginning to provide POCUS training.1

Several features distinguish POCUS from comprehensive ultrasound examinations. First, POCUS is designed to answer focused questions, whereas comprehensive ultrasound examinations evaluate all organs in an anatomical region; for example, an abdominal POCUS exam may evaluate only for presence or absence of intraperitoneal free fluid, whereas a comprehensive examination of the right upper quadrant will evaluate the liver, gallbladder, and biliary ducts. Second, POCUS examinations are generally performed by the same clinician who generates the relevant clinical question to answer with POCUS and ultimately integrates the findings into the patient’s care.2 By contrast, comprehensive ultrasound examinations involve multiple providers and steps: a clinician generates a relevant clinical question and requests an ultrasound examination that is acquired by a sonographer, interpreted by a radiologist, and reported back to the requesting clinician. Third, POCUS is often used to evaluate multiple body systems. For example, to evaluate a patient with undifferentiated hypotension, a multisystem POCUS examination of the heart, inferior vena cava, lungs, abdomen, and lower extremity veins is typically performed. Finally, POCUS examinations can be performed serially to investigate changes in clinical status or evaluate response to therapy, such as monitoring the heart, lungs, and inferior vena cava during fluid resuscitation.

The purpose of this position statement is to inform a broad audience about how hospitalists are using diagnostic and procedural applications of POCUS. This position statement does not mandate that hospitalists use POCUS. Rather, it is intended to provide guidance on the safe and effective use of POCUS by the hospitalists who use it and the administrators who oversee its use. We discuss POCUS (1) applications, (2) training, (3) assessments, and (4) program management. This position statement was reviewed and approved by the Society of Hospital Medicine (SHM) Executive Committee in March 2018.

 

 

APPLICATIONS

Common diagnostic and procedural applications of POCUS used by hospitalists are listed in Table 1. Selected evidence supporting the use of these applications is described in the supplementary online content (Appendices 1–8 available at http://journalofhospitalmedicine.com) and SHM position statements on specific ultrasound-guided bedside procedures.3,4 Additional applications not listed in Table 1 that may be performed by some hospitalists include assessment of the eyes, stomach, bowels, ovaries, pregnancy, and testicles, as well as performance of regional anesthesia. Moreover, hospitalists caring for pediatric and adolescent patients may use additional applications besides those listed here. Currently, many hospitalists already perform more complex and sophisticated POCUS examinations than those listed in Table 1. The scope of POCUS use by hospitalists continues to expand, and this position statement should not restrict that expansion.

As outlined in our earlier position statements,3,4 ultrasound guidance lowers complication rates and increases success rates of invasive bedside procedures. Diagnostic POCUS can guide clinical decision making prior to bedside procedures. For instance, hospitalists may use POCUS to assess the size and character of a pleural effusion to help determine the most appropriate management strategy: observation, medical treatment, thoracentesis, chest tube placement, or surgical therapy. Furthermore, diagnostic POCUS can be used to rapidly assess for immediate postprocedural complications, such as pneumothorax, or if the patient develops new symptoms.

TRAINING

Basic Knowledge

Basic knowledge includes fundamentals of ultrasound physics; safety;4 anatomy; physiology; and device operation, including maintenance and cleaning. Basic knowledge can be taught by multiple methods, including live or recorded lectures, online modules, or directed readings.

Image Acquisition

Training should occur across multiple types of patients (eg, obese, cachectic, postsurgical) and clinical settings (eg, intensive care unit, general medicine wards, emergency department) when available. Training is largely hands-on because the relevant skills involve integration of 3D anatomy with spatial manipulation, hand-eye coordination, and fine motor movements. Virtual reality ultrasound simulators may accelerate mastery, particularly for cardiac image acquisition, and expose learners to standardized sets of pathologic findings. Real-time bedside feedback on image acquisition is ideal because understanding how ultrasound probe manipulation affects the images acquired is essential to learning.

Image Interpretation

Training in image interpretation relies on visual pattern recognition of normal and abnormal findings. Therefore, the normal to abnormal spectrum should be broad, and learners should maintain a log of what abnormalities have been identified. Giving real-time feedback at the bedside is ideal because of the connection between image acquisition and interpretation. Image interpretation can be taught through didactic sessions, image review sessions, or review of teaching files with annotated images.

Clinical Integration

Learners must interpret and integrate image findings with other clinical data considering the image quality, patient characteristics, and changing physiology. Clinical integration should be taught by instructors that share similar clinical knowledge as learners. Although sonographers are well suited to teach image acquisition, they should not be the sole instructors to teach hospitalists how to integrate ultrasound findings in clinical decision making. Likewise, emphasis should be placed on the appropriate use of POCUS within a provider’s skill set. Learners must appreciate the clinical significance of POCUS findings, including recognition of incidental findings that may require further workup. Supplemental training in clinical integration can occur through didactics that include complex patient scenarios.

 

 

Pathways

Clinical competency can be achieved with training adherent to five criteria. First, the training environment should be similar to where the trainee will practice. Second, training and feedback should occur in real time. Third, specific applications should be taught rather than broad training in “hospitalist POCUS.” Each application requires unique skills and knowledge, including image acquisition pitfalls and artifacts. Fourth, clinical competence must be achieved and demonstrated; it is not necessarily gained through experience. Fifth, once competency is achieved, continued education and feedback are necessary to ensure it is maintained.

Residency-based POCUS training pathways can best fulfill these criteria. They may eventually become commonplace, but until then alternative pathways must exist for hospitalist providers who are already in practice. There are three important attributes of such pathways. First, administrators’ expectations about learners’ clinical productivity must be realistically, but only temporarily, relaxed; otherwise, competing demands on time will likely overwhelm learners and subvert training. Second, training should begin through a local or national hands-on training program. The SHM POCUS certificate program consolidates training for common diagnostic POCUS applications for hospitalists.6 Other medical societies offer training for their respective clinical specialties.7 Third, once basic POCUS training has begun, longitudinal training should continue ideally with a local hospitalist POCUS expert.

In some settings, a subgroup of hospitalists may not desire, or be able to achieve, competency in the manual skills of POCUS image acquisition. Nevertheless, hospitalists may still find value in understanding POCUS nomenclature, image pattern recognition, and the evidence and pitfalls behind clinical integration of specific POCUS findings. This subset of POCUS skills allows hospitalists to communicate effectively with and understand the clinical decisions made by their colleagues who are competent in POCUS use.

The minimal skills a hospitalist should possess to serve as a POCUS trainer include proficiency of basic knowledge, image acquisition, image interpretation, and clinical integration of the POCUS applications being taught; effectiveness as a hands-on instructor to teach image acquisition skills; and an in-depth understanding of common POCUS pitfalls and limitations.

ASSESSMENTS

Assessment methods for POCUS can include the following: knowledge-based questions, image acquisition using task-specific checklists on human or simulation models, image interpretation using a series of videos or still images with normal and abnormal findings, clinical integration using “next best step” in a multiple choice format with POCUS images, and simulation-based clinical scenarios. Assessment methods should be aligned with local availability of resources and trainers.

Basic Knowledge

Basic knowledge can be assessed via multiple choice questions assessing knowledge of ultrasound physics, image optimization, relevant anatomy, and limitations of POCUS imaging. Basic knowledge lies primarily in the cognitive domain and does not assess manual skills.

Image Acquisition

Image acquisition can be assessed by observation and rating of image quality. Where resources allow, assessment of image acquisition is likely best done through a combination of developing an image portfolio with a minimum number of high quality images, plus direct observation of image acquisition by an expert. Various programs have utilized minimum numbers of images acquired to help define competence with image acquisition skills.6–8 Although minimums may be a necessary step to gain competence, using them as a sole means to determine competence does not account for variable learning curves.9 As with other manual skills in hospital medicine, such as ultrasound-guided bedside procedures, minimum numbers are best used as a starting point for assessments.3,10 In this regard, portfolio development with meticulous attention to the gain, depth, and proper tomographic plane of images can monitor a hospitalist’s progress toward competence by providing objective assessments and feedback. Simulation may also be used as it allows assessment of image acquisition skills and an opportunity to provide real-time feedback, similar to direct observation but without actual patients.

 

 

Image Interpretation

Image interpretation is best assessed by an expert observing the learner at bedside; however, when bedside assessment is not possible, image interpretation skills may be assessed using multiple choice or free text interpretation of archived ultrasound images with normal and abnormal findings. This is often incorporated into the portfolio development portion of a training program, as learners can submit their image interpretation along with the video clip. Both normal and abnormal images can be used to assess anatomic recognition and interpretation. Emphasis should be placed on determining when an image is suboptimal for diagnosis (eg, incomplete exam or poor-quality images). Quality assurance programs should incorporate structured feedback sessions.

Clinical Integration

Assessment of clinical integration can be completed through case scenarios that assess knowledge, interpretation of images, and integration of findings into clinical decision making, which is often delivered via a computer-based assessment. Assessments should combine specific POCUS applications to evaluate common clinical problems in hospital medicine, such as undifferentiated hypotension and dyspnea. High-fidelity simulators can be used to blend clinical case scenarios with image acquisition, image interpretation, and clinical integration. When feasible, comprehensive feedback on how providers acquire, interpret, and apply ultrasound at the bedside is likely the best mechanism to assess clinical integration. This process can be done with a hospitalist’s own patients.

General Assessment

A general assessment that includes a summative knowledge and hands-on skills assessment using task-specific checklists can be performed upon completion of training. A high-fidelity simulator with dynamic or virtual anatomy can provide reproducible standardized assessments with variation in the type and difficulty of cases. When available, we encourage the use of dynamic assessments on actual patients that have both normal and abnormal ultrasound findings because simulated patient scenarios have limitations, even with the use of high-fidelity simulators. Programs are recommended to use formative and summative assessments for evaluation. Quantitative scoring systems using checklists are likely the best framework.11,12

CERTIFICATES AND CERTIFICATION

A certificate of completion is proof of a provider’s participation in an educational activity; it does not equate with competency, though it may be a step toward it. Most POCUS training workshops and short courses provide certificates of completion. Certification of competency is an attestation of a hospitalist’s basic competence within a defined scope of practice (Table 2).13 However, without longitudinal supervision and feedback, skills can decay; therefore, we recommend a longitudinal training program that provides mentored feedback and incorporates periodic competency assessments. At present, no national board certification in POCUS is available to grant external certification of competency for hospitalists.

External Certificate

Certificates of completion can be external through a national organization. An external certificate of completion designed for hospitalists includes the POCUS Certificate of Completion offered by SHM in collaboration with CHEST.6 This certificate program provides regional training options and longitudinal portfolio development. Other external certificates are also available to hospitalists.7,14,15

Most hospitalists are boarded by the American Board of Internal Medicine or the American Board of Family Medicine. These boards do not yet include certification of competency in POCUS. Other specialty boards, such as emergency medicine, include competency in POCUS. For emergency medicine, completion of an accredited residency training program and certification by the national board includes POCUS competency.

 

 

Internal Certificate

There are a few examples of successful local institutional programs that have provided internal certificates of competency.12,14 Competency assessments require significant resources including investment by both faculty and learners. Ongoing evaluation of competency should be based on quality assurance processes.

Credentialing and Privileging

The American Medical Association (AMA) House of Delegates in 1999 passed a resolution (AMA HR. 802) recommending hospitals follow specialty-specific guidelines for privileging decisions related to POCUS use.17 The resolution included a statement that, “ultrasound imaging is within the scope of practice of appropriately trained physicians.”

Some institutions have begun to rely on a combination of internal and external certificate programs to grant privileges to hospitalists.10 Although specific privileges for POCUS may not be required in some hospitals, some institutions may require certification of training and assessments prior to granting permission to use POCUS.

Hospitalist programs are encouraged to evaluate ongoing POCUS use by their providers after granting initial permission. If privileging is instituted by a hospital, hospitalists must play a significant role in determining the requirements for privileging and ongoing maintenance of skills.

Maintenance of Skills

All medical skills can decay with disuse, including those associated with POCUS.12,18 Thus, POCUS users should continue using POCUS regularly in clinical practice and participate in POCUS continuing medical education activities, ideally with ongoing assessments. Maintenance of skills may be confirmed through routine participation in a quality assurance program.

PROGRAM MANAGEMENT

Use of POCUS in hospital medicine has unique considerations, and hospitalists should be integrally involved in decision making surrounding institutional POCUS program management. Appointing a dedicated POCUS director can help a program succeed.8

Equipment and Image Archiving

Several factors are important to consider when selecting an ultrasound machine: portability, screen size, and ease of use; integration with the electronic medical record and options for image archiving; manufacturer’s service plan, including technical and clinical support; and compliance with local infection control policies. The ability to easily archive and retrieve images is essential for quality assurance, continuing education, institutional quality improvement, documentation, and reimbursement. In certain scenarios, image archiving may not be possible (such as with personal handheld devices or in emergency situations) or necessary (such as with frequent serial examinations during fluid resuscitation). An image archive is ideally linked to reports, orders, and billing software.10,19 If such linkages are not feasible, parallel external storage that complies with regulatory standards (ie, HIPAA compliance) may be suitable.20

Documentation and Billing

Components of documentation include the indication and type of ultrasound examination performed, date and time of the examination, patient identifying information, name of provider(s) acquiring and interpreting the images, specific scanning protocols used, patient position, probe used, and findings. Documentation can occur through a standalone note or as part of another note, such as a progress note. Whenever possible, documentation should be timely to facilitate communication with other providers.

Billing is supported through the AMA Current Procedural Terminology codes for “focused” or “limited” ultrasound examinations (Appendix 9). The following three criteria must be satisfied for billing. First, images must be permanently stored. Specific requirements vary by insurance policy, though current practice suggests a minimum of one image demonstrating relevant anatomy and pathology for the ultrasound examination coded. For ultrasound-guided procedures that require needle insertion, images should be captured at the point of interest, and a procedure note should reflect that the needle was guided and visualized under ultrasound.21 Second, proper documentation must be entered in the medical record. Third, local institutional privileges for POCUS must be considered. Although privileges are not required to bill, some hospitals or payers may require them.

 

 

Quality Assurance

Published guidelines on quality assurance in POCUS are available from different specialty organizations, including emergency medicine, pediatric emergency medicine, critical care, anesthesiology, obstetrics, and cardiology.8,22–28 Quality assurance is aimed at ensuring that physicians maintain basic competency in using POCUS to influence bedside decisions.

Quality assurance should be carried out by an individual or committee with expertise in POCUS. Multidisciplinary QA programs in which hospital medicine providers are working collaboratively with other POCUS providers have been demonstrated to be highly effective.10 Oversight includes ensuring that providers using POCUS are appropriately trained,10,22,28 using the equipment correctly,8,26,28 and documenting properly. Some programs have implemented mechanisms to review and provide feedback on image acquisition, interpretation, and clinical integration.8,10 Other programs have compared POCUS findings with referral studies, such as comprehensive ultrasound examinations.

CONCLUSIONS

Practicing hospitalists must continue to collaborate with their institutions to build POCUS capabilities. In particular, they must work with their local privileging body to determine what credentials are required. The distinction between certificates of completion and certificates of competency, including whether those certificates are internal or external, is important in the credentialing process.

External certificates of competency are currently unavailable for most practicing hospitalists because ABIM certification does not include POCUS-related competencies. As internal medicine residency training programs begin to adopt POCUS training and certification into their educational curricula, we foresee a need to update the ABIM Policies and Procedures for Certification. Until then, we recommend that certificates of competency be defined and granted internally by local hospitalist groups.

Given the many advantages of POCUS over traditional tools, we anticipate its increasing implementation among hospitalists in the future. As with all medical technology, its role in clinical care should be continuously reexamined and redefined through health services research. Such information will be useful in developing practice guidelines, educational curricula, and training standards.

Acknowledgments

The authors would like to thank all members that participated in the discussion and finalization of this position statement during the Point-of-care Ultrasound Faculty Retreat at the 2018 Society of Hospital Medicine Annual Conference: Saaid Abdel-Ghani, Brandon Boesch, Joel Cho, Ria Dancel, Renee Dversdal, Ricardo Franco-Sadud, Benjamin Galen, Trevor P. Jensen, Mohit Jindal, Gordon Johnson, Linda M. Kurian, Gigi Liu, Charles M. LoPresti, Brian P. Lucas, Venkat Kalidindi, Benji Matthews, Anna Maw, Gregory Mints, Kreegan Reierson, Gerard Salame, Richard Schildhouse, Daniel Schnobrich, Nilam Soni, Kirk Spencer, Hiromizu Takahashi, David M. Tierney, Tanping Wong, and Toru Yamada.

References

1. Schnobrich DJ, Mathews BK, Trappey BE, Muthyala BK, Olson APJ. Entrusting internal medicine residents to use point of care ultrasound: Towards improved assessment and supervision. Med Teach. 2018:1-6. doi:10.1080/0142159X.2018.1457210.
2. Soni NJ, Lucas BP. Diagnostic point-of-care ultrasound for hospitalists. J Hosp Med. 2015;10(2):120-124. doi:10.1002/jhm.2285.
3. Lucas BP, Tierney DM, Jensen TP, et al. Credentialing of hospitalists in ultrasound-guided bedside procedures: a position statement of the society of hospital medicine. J Hosp Med. 2018;13(2):117-125. doi:10.12788/jhm.2917.
4. Dancel R, Schnobrich D, Puri N, et al. Recommendations on the use of ultrasound guidance for adult thoracentesis: a position statement of the society of hospital medicine. J Hosp Med. 2018;13(2):126-135. doi:10.12788/jhm.2940.
5. National Council on Radiation Protection and Measurements, The Council. Implementation of the Principle of as Low as Reasonably Achievable (ALARA) for Medical and Dental Personnel.; 1990.
6. Society of Hospital Medicine. Point of Care Ultrasound course: https://www.hospitalmedicine.org/clinical-topics/ultrasonography-cert/. Accessed February 6, 2018.
7. Critical Care Ultrasonography Certificate of Completion Program. CHEST. American College of Chest Physicians. http://www.chestnet.org/Education/Advanced-Clinical-Training/Certificate-of-Completion-Program/Critical-Care-Ultrasonography. Accessed February 6, 2018.
8. American College of Emergency Physicians Policy Statement: Emergency Ultrasound Guidelines. 2016. https://www.acep.org/Clinical---Practice-Management/ACEP-Ultrasound-Guidelines/. Accessed February 6, 2018.
9. Blehar DJ, Barton B, Gaspari RJ. Learning curves in emergency ultrasound education. Acad Emerg Med. 2015;22(5):574-582. doi:10.1111/acem.12653.
10. Mathews BK, Zwank M. Hospital medicine point of care ultrasound credentialing: an example protocol. J Hosp Med. 2017;12(9):767-772. doi:10.12788/jhm.2809.
11. Barsuk JH, McGaghie WC, Cohen ER, Balachandran JS, Wayne DB. Use of simulation-based mastery learning to improve the quality of central venous catheter placement in a medical intensive care unit. J Hosp Med. 2009;4(7):397-403. doi:10.1002/jhm.468.
12. Mathews BK, Reierson K, Vuong K, et al. The design and evaluation of the Comprehensive Hospitalist Assessment and Mentorship with Portfolios (CHAMP) ultrasound program. J Hosp Med. 2018;13(8):544-550. doi:10.12788/jhm.2938.
13. Soni NJ, Tierney DM, Jensen TP, Lucas BP. Certification of point-of-care ultrasound competency. J Hosp Med. 2017;12(9):775-776. doi:10.12788/jhm.2812.
14. Ultrasound Certification for Physicians. Alliance for Physician Certification and Advancement. APCA. https://apca.org/. Accessed February 6, 2018.
15. National Board of Echocardiography, Inc. https://www.echoboards.org/EchoBoards/News/2019_Adult_Critical_Care_Echocardiography_Exam.aspx. Accessed June 18, 2018.
16. Tierney DM. Internal Medicine Bedside Ultrasound Program (IMBUS). Abbott Northwestern. http://imbus.anwresidency.com/index.html. Accessed February 6, 2018.
17. American Medical Association House of Delegates Resolution H-230.960: Privileging for Ultrasound Imaging. Resolution 802. Policy Finder Website. http://search0.ama-assn.org/search/pfonline. Published 1999. Accessed February 18, 2018.
18. Kelm D, Ratelle J, Azeem N, et al. Longitudinal ultrasound curriculum improves long-term retention among internal medicine residents. J Grad Med Educ. 2015;7(3):454-457. doi:10.4300/JGME-14-00284.1.
19. Flannigan MJ, Adhikari S. Point-of-care ultrasound work flow innovation: impact on documentation and billing. J Ultrasound Med. 2017;36(12):2467-2474. doi:10.1002/jum.14284.
20. Emergency Ultrasound: Workflow White Paper. https://www.acep.org/uploadedFiles/ACEP/memberCenter/SectionsofMembership/ultra/Workflow%20White%20Paper.pdf. Published 2013. Accessed February 18, 2018.
21. Ultrasound Coding and Reimbursement Document 2009. Emergency Ultrasound Section. American College of Emergency Physicians. http://emergencyultrasoundteaching.com/assets/2009_coding_update.pdf. Published 2009. Accessed February 18, 2018.
22. Mayo PH, Beaulieu Y, Doelken P, et al. American College of Chest Physicians/La Societe de Reanimation de Langue Francaise statement on competence in critical care ultrasonography. Chest. 2009;135(4):1050-1060. doi:10.1378/chest.08-2305.
23. Frankel HL, Kirkpatrick AW, Elbarbary M, et al. Guidelines for the appropriate use of bedside general and cardiac ultrasonography in the evaluation of critically ill patients-part I: general ultrasonography. Crit Care Med. 2015;43(11):2479-2502. doi:10.1097/ccm.0000000000001216.
24. Levitov A, Frankel HL, Blaivas M, et al. Guidelines for the appropriate use of bedside general and cardiac ultrasonography in the evaluation of critically ill patients-part ii: cardiac ultrasonography. Crit Care Med. 2016;44(6):1206-1227. doi:10.1097/ccm.0000000000001847.
25. ACR–ACOG–AIUM–SRU Practice Parameter for the Performance of Obstetrical Ultrasound. https://www.acr.org/-/media/ACR/Files/Practice-Parameters/us-ob.pdf. Published 2013. Accessed February 18, 2018.
26. AIUM practice guideline for documentation of an ultrasound examination. J Ultrasound Med. 2014;33(6):1098-1102. doi:10.7863/ultra.33.6.1098.
27. Marin JR, Lewiss RE. Point-of-care ultrasonography by pediatric emergency medicine physicians. Pediatrics. 2015;135(4):e1113-e1122. doi:10.1542/peds.2015-0343.
28. Spencer KT, Kimura BJ, Korcarz CE, Pellikka PA, Rahko PS, Siegel RJ. Focused cardiac ultrasound: recommendations from the American Society of Echocardiography. J Am Soc Echocardiogr. 2013;26(6):567-581. doi:10.1016/j.echo.2013.04.001.

References

1. Schnobrich DJ, Mathews BK, Trappey BE, Muthyala BK, Olson APJ. Entrusting internal medicine residents to use point of care ultrasound: Towards improved assessment and supervision. Med Teach. 2018:1-6. doi:10.1080/0142159X.2018.1457210.
2. Soni NJ, Lucas BP. Diagnostic point-of-care ultrasound for hospitalists. J Hosp Med. 2015;10(2):120-124. doi:10.1002/jhm.2285.
3. Lucas BP, Tierney DM, Jensen TP, et al. Credentialing of hospitalists in ultrasound-guided bedside procedures: a position statement of the society of hospital medicine. J Hosp Med. 2018;13(2):117-125. doi:10.12788/jhm.2917.
4. Dancel R, Schnobrich D, Puri N, et al. Recommendations on the use of ultrasound guidance for adult thoracentesis: a position statement of the society of hospital medicine. J Hosp Med. 2018;13(2):126-135. doi:10.12788/jhm.2940.
5. National Council on Radiation Protection and Measurements, The Council. Implementation of the Principle of as Low as Reasonably Achievable (ALARA) for Medical and Dental Personnel.; 1990.
6. Society of Hospital Medicine. Point of Care Ultrasound course: https://www.hospitalmedicine.org/clinical-topics/ultrasonography-cert/. Accessed February 6, 2018.
7. Critical Care Ultrasonography Certificate of Completion Program. CHEST. American College of Chest Physicians. http://www.chestnet.org/Education/Advanced-Clinical-Training/Certificate-of-Completion-Program/Critical-Care-Ultrasonography. Accessed February 6, 2018.
8. American College of Emergency Physicians Policy Statement: Emergency Ultrasound Guidelines. 2016. https://www.acep.org/Clinical---Practice-Management/ACEP-Ultrasound-Guidelines/. Accessed February 6, 2018.
9. Blehar DJ, Barton B, Gaspari RJ. Learning curves in emergency ultrasound education. Acad Emerg Med. 2015;22(5):574-582. doi:10.1111/acem.12653.
10. Mathews BK, Zwank M. Hospital medicine point of care ultrasound credentialing: an example protocol. J Hosp Med. 2017;12(9):767-772. doi:10.12788/jhm.2809.
11. Barsuk JH, McGaghie WC, Cohen ER, Balachandran JS, Wayne DB. Use of simulation-based mastery learning to improve the quality of central venous catheter placement in a medical intensive care unit. J Hosp Med. 2009;4(7):397-403. doi:10.1002/jhm.468.
12. Mathews BK, Reierson K, Vuong K, et al. The design and evaluation of the Comprehensive Hospitalist Assessment and Mentorship with Portfolios (CHAMP) ultrasound program. J Hosp Med. 2018;13(8):544-550. doi:10.12788/jhm.2938.
13. Soni NJ, Tierney DM, Jensen TP, Lucas BP. Certification of point-of-care ultrasound competency. J Hosp Med. 2017;12(9):775-776. doi:10.12788/jhm.2812.
14. Ultrasound Certification for Physicians. Alliance for Physician Certification and Advancement. APCA. https://apca.org/. Accessed February 6, 2018.
15. National Board of Echocardiography, Inc. https://www.echoboards.org/EchoBoards/News/2019_Adult_Critical_Care_Echocardiography_Exam.aspx. Accessed June 18, 2018.
16. Tierney DM. Internal Medicine Bedside Ultrasound Program (IMBUS). Abbott Northwestern. http://imbus.anwresidency.com/index.html. Accessed February 6, 2018.
17. American Medical Association House of Delegates Resolution H-230.960: Privileging for Ultrasound Imaging. Resolution 802. Policy Finder Website. http://search0.ama-assn.org/search/pfonline. Published 1999. Accessed February 18, 2018.
18. Kelm D, Ratelle J, Azeem N, et al. Longitudinal ultrasound curriculum improves long-term retention among internal medicine residents. J Grad Med Educ. 2015;7(3):454-457. doi:10.4300/JGME-14-00284.1.
19. Flannigan MJ, Adhikari S. Point-of-care ultrasound work flow innovation: impact on documentation and billing. J Ultrasound Med. 2017;36(12):2467-2474. doi:10.1002/jum.14284.
20. Emergency Ultrasound: Workflow White Paper. https://www.acep.org/uploadedFiles/ACEP/memberCenter/SectionsofMembership/ultra/Workflow%20White%20Paper.pdf. Published 2013. Accessed February 18, 2018.
21. Ultrasound Coding and Reimbursement Document 2009. Emergency Ultrasound Section. American College of Emergency Physicians. http://emergencyultrasoundteaching.com/assets/2009_coding_update.pdf. Published 2009. Accessed February 18, 2018.
22. Mayo PH, Beaulieu Y, Doelken P, et al. American College of Chest Physicians/La Societe de Reanimation de Langue Francaise statement on competence in critical care ultrasonography. Chest. 2009;135(4):1050-1060. doi:10.1378/chest.08-2305.
23. Frankel HL, Kirkpatrick AW, Elbarbary M, et al. Guidelines for the appropriate use of bedside general and cardiac ultrasonography in the evaluation of critically ill patients-part I: general ultrasonography. Crit Care Med. 2015;43(11):2479-2502. doi:10.1097/ccm.0000000000001216.
24. Levitov A, Frankel HL, Blaivas M, et al. Guidelines for the appropriate use of bedside general and cardiac ultrasonography in the evaluation of critically ill patients-part ii: cardiac ultrasonography. Crit Care Med. 2016;44(6):1206-1227. doi:10.1097/ccm.0000000000001847.
25. ACR–ACOG–AIUM–SRU Practice Parameter for the Performance of Obstetrical Ultrasound. https://www.acr.org/-/media/ACR/Files/Practice-Parameters/us-ob.pdf. Published 2013. Accessed February 18, 2018.
26. AIUM practice guideline for documentation of an ultrasound examination. J Ultrasound Med. 2014;33(6):1098-1102. doi:10.7863/ultra.33.6.1098.
27. Marin JR, Lewiss RE. Point-of-care ultrasonography by pediatric emergency medicine physicians. Pediatrics. 2015;135(4):e1113-e1122. doi:10.1542/peds.2015-0343.
28. Spencer KT, Kimura BJ, Korcarz CE, Pellikka PA, Rahko PS, Siegel RJ. Focused cardiac ultrasound: recommendations from the American Society of Echocardiography. J Am Soc Echocardiogr. 2013;26(6):567-581. doi:10.1016/j.echo.2013.04.001.

Publications
Publications
Topics
Article Type
Sections
Article Source

© 2019 Society of Hospital Medicine

Citation Override
Published Online Only January 2, 2019. doi: 10.12788/jhm.3079
Disallow All Ads
Correspondence Location
Corresponding Author: Nilam J. Soni, MD MS; E-mail: sonin@uthscsa.edu; Telephone: 210-743-6030.
Content Gating
Open Access (article Unlocked/Open Access)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Article PDF Media
Media Files

An innovative perioperative/consultative curriculum for third-year internal medicine residents

Article Type
Changed
Tue, 10/02/2018 - 14:20
Display Headline
An innovative perioperative/consultative curriculum for third-year internal medicine residents
Article PDF
Author and Disclosure Information

Alex Rico, MD
University of Miami, Miami, FL

Joshua Lenchus, DO
University of Miami, Miami, FL

Amir Jaffer, MD
University of Miami, Miami, FL

Publications
Page Number
eS28
Author and Disclosure Information

Alex Rico, MD
University of Miami, Miami, FL

Joshua Lenchus, DO
University of Miami, Miami, FL

Amir Jaffer, MD
University of Miami, Miami, FL

Author and Disclosure Information

Alex Rico, MD
University of Miami, Miami, FL

Joshua Lenchus, DO
University of Miami, Miami, FL

Amir Jaffer, MD
University of Miami, Miami, FL

Article PDF
Article PDF
Page Number
eS28
Page Number
eS28
Publications
Publications
Article Type
Display Headline
An innovative perioperative/consultative curriculum for third-year internal medicine residents
Display Headline
An innovative perioperative/consultative curriculum for third-year internal medicine residents
Citation Override
Cleveland Clinic Journal of Medicine 2009 February;76(e-suppl 1):eS28
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media