Genetic alterations affect everolimus benefit in breast cancer

Mutation profile needs further study
Article Type
Changed
Thu, 12/15/2022 - 18:16
Display Headline
Genetic alterations affect everolimus benefit in breast cancer

CHICAGO – The benefit of everolimus in hormone receptor–positive, HER2-negative advanced breast cancer appears to be greater for women who have little or no genetic alteration in this cancer’s most commonly altered molecular signaling pathways, based on data presented at the annual meeting of the American Society of Clinical Oncology.

A team led by Dr. Gabriel N. Hortobagyi performed next-generation sequencing on archival tumor tissue from patients enrolled in the randomized BOLERO-2 trial, which tested the addition to exemestane of everolimus, an inhibitor of the mammalian target of rapamycin (mTOR).

Dr. Gabriel N. Hortobagyi

Among all 227 patients whose tumors were tested, adding everolimus reduced the risk of progression or death by 60% as compared with adding placebo.

Patients having alterations in any of the four genes that were most commonly altered in the tumors or that were part of their respective signaling pathways derived similar benefit from everolimus as patients who had wild-type (normal) genes.

Thus, "no predictive marker of everolimus efficacy was identified in individual analysis of the four pathways," commented Dr. Hortobagyi, who chairs the department of breast medical oncology at the University of Texas MD Anderson Cancer Center in Houston.

However, there was differential benefit when patients were broadly grouped according to the extent of alterations – either minimal or multiple.

Three-fourths had minimal (none or one) alteration of these genes or another gene lying on the same pathway: the PIK3CA gene or the PTEN gene (PI3 kinase pathway), the CCND1 gene (cell cycle pathway), or the FGFR1 gene (FGFR1/2 pathway).

This group had a 73% reduction in the risk of progression or death if given everolimus as compared with placebo. In contrast, the group having multiple (two or more) alterations of these genes had a much smaller 22% reduction in risk.

"Further delineation of the interaction of these genetic events might help us to understand how the oncogenic signal cascades work and find better options to attenuate disease progression," Dr. Hortobagyi proposed. "These results are hypothesis generating only and should be validated in independent patient cohorts. But if confirmed, they could lead to new hypotheses for development of novel and more effective combination targeted therapies in hormone receptor–positive, HER2-negative breast cancer."

Session attendee Dr. Larry Norton of the Memorial Sloan-Kettering Cancer Center in New York, asked, "How many of these biopsies came from metastatic lesions and how many came from the primary? And if you just looked at the ones from the metastatic sites, do you see any differences there, because as we know, there is clonal evolution in the metastatic sites."

"About 20% came from metastatic disease, and about 80% from the primary. There are a number of individual differences between the two groups, but unfortunately, the numbers in the metastatic group are so small that you can’t really derive patterns," Dr. Hortobagyi replied. "Now all of the samples, both the primary and the metastatic ones, were from before the study, so it would not really directly reflect on the study itself. But that’s a very important question that we eventually have to address."

Attendee Dr. Ian Krop of the Dana-Farber Cancer Institute in Boston, commented, "It was interesting that those tumors that had multiple alterations in one of those four genes had what appeared to be decreased benefit from everolimus. Could that represent just genomic instability? In other words, if you look at random alterations in your data set, did that also correlate with lack of benefit?"

"We looked at that, and the genetic background for the two groups is very similar," Dr. Hortobagyi replied. "So my a priori thinking was exactly as you mentioned, that those would be associated with instability and therefore multiple driver pathways. But we did not see that."

Women in the BOLERO-2 trial were postmenopausal and had locally advanced or metastatic breast cancer that was refractory to a nonsteroidal aromatase inhibitor. They were assigned 2:1 to exemestane (brand name Aromasin) plus either everolimus (Afinitor) or placebo.

Everolimus is currently approved by the Food and Drug Administration for use in combination with exemestane to treat advanced hormone receptor–positive breast cancer in postmenopausal women. It is also approved for selected indications in other cancers.

In the new retrospective exploratory analysis, Dr. Hortobagyi’s team collected formalin-fixed, paraffin-embedded archival tumor tissue in a subset of patients and performed sequencing for 3,230 exons in 182 oncogenes and tumor suppressor genes.

Results showed that the four most commonly altered genes were PIK3CA (altered in 48% of patients), CCND1 (31%), TP53 (23%), and FGFR1 (18%).

 

 

In the patient subset as a whole, addition of everolimus was associated with a significant reduction in the risk of progression or death (hazard ratio, 0.40) similar to that previously reported in the entire trial population (HR, 0.45).

Neither alteration of any one of these four genes nor alteration of any one gene in their respective pathways predicted everolimus benefit. "Basically, the treatment effect was independent of the genetic status of these genes and pathways," Dr. Hortobagyi commented.

But when the pathways were considered simultaneously, patients having minimal genetic alteration derived a progression-free survival benefit from everolimus that exceeded that in the whole subset (HR, 0.27), whereas their counterparts having multiple alterations derived a benefit much less than that in the whole subset (HR, 0.78).

Dr. Hortobagyi disclosed that he is a consultant to Allergan, AstraZeneca, Galena Biopharma, Genentech, Myriad Genetics, Novartis, and Sanofi; receives research funding from Novartis; and has other affiliations with Genentech, Novartis, and Sanofi. The BOLERO-2 trial was sponsored by Novartis.

Body

Failure of single genetic alterations, such as PIK3CA-activating mutations, to predict everolimus benefit likely reflects the fact that there are other, independent ways of activating the mTOR pathway. So maybe pathway activation is what we should be looking at, not necessarily gene mutations.

Are we going to do anything different in clinic tomorrow as a result of these findings when it comes to using everolimus? No, we are still going to select therapy based on clinical criteria of acquired resistance to previous nonsteroidal aromatase inhibitors. Perhaps in the future, we will continue to do more work because we may be still able to develop that mutation profile that perhaps will spare some patients the treatment because there are other drugs that they can benefit from.

Dr. Stephen R.D. Johnston is a professor of breast cancer medicine at the Royal Marsden Hospital in London. He was the invited discussant of the research. Dr. Johnston disclosed that he is a consultant to AstraZeneca, GlaxoSmithKline, and Roche/Genentech, and that he receives honoraria from GlaxoSmithKline and Novartis.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
everolimus, advanced breast cancer, American Society of Clinical Oncology, Dr. Gabriel N. Hortobagyi, BOLERO-2 trial, everolimus, rapamycin, mTOR)
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event
Body

Failure of single genetic alterations, such as PIK3CA-activating mutations, to predict everolimus benefit likely reflects the fact that there are other, independent ways of activating the mTOR pathway. So maybe pathway activation is what we should be looking at, not necessarily gene mutations.

Are we going to do anything different in clinic tomorrow as a result of these findings when it comes to using everolimus? No, we are still going to select therapy based on clinical criteria of acquired resistance to previous nonsteroidal aromatase inhibitors. Perhaps in the future, we will continue to do more work because we may be still able to develop that mutation profile that perhaps will spare some patients the treatment because there are other drugs that they can benefit from.

Dr. Stephen R.D. Johnston is a professor of breast cancer medicine at the Royal Marsden Hospital in London. He was the invited discussant of the research. Dr. Johnston disclosed that he is a consultant to AstraZeneca, GlaxoSmithKline, and Roche/Genentech, and that he receives honoraria from GlaxoSmithKline and Novartis.

Body

Failure of single genetic alterations, such as PIK3CA-activating mutations, to predict everolimus benefit likely reflects the fact that there are other, independent ways of activating the mTOR pathway. So maybe pathway activation is what we should be looking at, not necessarily gene mutations.

Are we going to do anything different in clinic tomorrow as a result of these findings when it comes to using everolimus? No, we are still going to select therapy based on clinical criteria of acquired resistance to previous nonsteroidal aromatase inhibitors. Perhaps in the future, we will continue to do more work because we may be still able to develop that mutation profile that perhaps will spare some patients the treatment because there are other drugs that they can benefit from.

Dr. Stephen R.D. Johnston is a professor of breast cancer medicine at the Royal Marsden Hospital in London. He was the invited discussant of the research. Dr. Johnston disclosed that he is a consultant to AstraZeneca, GlaxoSmithKline, and Roche/Genentech, and that he receives honoraria from GlaxoSmithKline and Novartis.

Title
Mutation profile needs further study
Mutation profile needs further study

CHICAGO – The benefit of everolimus in hormone receptor–positive, HER2-negative advanced breast cancer appears to be greater for women who have little or no genetic alteration in this cancer’s most commonly altered molecular signaling pathways, based on data presented at the annual meeting of the American Society of Clinical Oncology.

A team led by Dr. Gabriel N. Hortobagyi performed next-generation sequencing on archival tumor tissue from patients enrolled in the randomized BOLERO-2 trial, which tested the addition to exemestane of everolimus, an inhibitor of the mammalian target of rapamycin (mTOR).

Dr. Gabriel N. Hortobagyi

Among all 227 patients whose tumors were tested, adding everolimus reduced the risk of progression or death by 60% as compared with adding placebo.

Patients having alterations in any of the four genes that were most commonly altered in the tumors or that were part of their respective signaling pathways derived similar benefit from everolimus as patients who had wild-type (normal) genes.

Thus, "no predictive marker of everolimus efficacy was identified in individual analysis of the four pathways," commented Dr. Hortobagyi, who chairs the department of breast medical oncology at the University of Texas MD Anderson Cancer Center in Houston.

However, there was differential benefit when patients were broadly grouped according to the extent of alterations – either minimal or multiple.

Three-fourths had minimal (none or one) alteration of these genes or another gene lying on the same pathway: the PIK3CA gene or the PTEN gene (PI3 kinase pathway), the CCND1 gene (cell cycle pathway), or the FGFR1 gene (FGFR1/2 pathway).

This group had a 73% reduction in the risk of progression or death if given everolimus as compared with placebo. In contrast, the group having multiple (two or more) alterations of these genes had a much smaller 22% reduction in risk.

"Further delineation of the interaction of these genetic events might help us to understand how the oncogenic signal cascades work and find better options to attenuate disease progression," Dr. Hortobagyi proposed. "These results are hypothesis generating only and should be validated in independent patient cohorts. But if confirmed, they could lead to new hypotheses for development of novel and more effective combination targeted therapies in hormone receptor–positive, HER2-negative breast cancer."

Session attendee Dr. Larry Norton of the Memorial Sloan-Kettering Cancer Center in New York, asked, "How many of these biopsies came from metastatic lesions and how many came from the primary? And if you just looked at the ones from the metastatic sites, do you see any differences there, because as we know, there is clonal evolution in the metastatic sites."

"About 20% came from metastatic disease, and about 80% from the primary. There are a number of individual differences between the two groups, but unfortunately, the numbers in the metastatic group are so small that you can’t really derive patterns," Dr. Hortobagyi replied. "Now all of the samples, both the primary and the metastatic ones, were from before the study, so it would not really directly reflect on the study itself. But that’s a very important question that we eventually have to address."

Attendee Dr. Ian Krop of the Dana-Farber Cancer Institute in Boston, commented, "It was interesting that those tumors that had multiple alterations in one of those four genes had what appeared to be decreased benefit from everolimus. Could that represent just genomic instability? In other words, if you look at random alterations in your data set, did that also correlate with lack of benefit?"

"We looked at that, and the genetic background for the two groups is very similar," Dr. Hortobagyi replied. "So my a priori thinking was exactly as you mentioned, that those would be associated with instability and therefore multiple driver pathways. But we did not see that."

Women in the BOLERO-2 trial were postmenopausal and had locally advanced or metastatic breast cancer that was refractory to a nonsteroidal aromatase inhibitor. They were assigned 2:1 to exemestane (brand name Aromasin) plus either everolimus (Afinitor) or placebo.

Everolimus is currently approved by the Food and Drug Administration for use in combination with exemestane to treat advanced hormone receptor–positive breast cancer in postmenopausal women. It is also approved for selected indications in other cancers.

In the new retrospective exploratory analysis, Dr. Hortobagyi’s team collected formalin-fixed, paraffin-embedded archival tumor tissue in a subset of patients and performed sequencing for 3,230 exons in 182 oncogenes and tumor suppressor genes.

Results showed that the four most commonly altered genes were PIK3CA (altered in 48% of patients), CCND1 (31%), TP53 (23%), and FGFR1 (18%).

 

 

In the patient subset as a whole, addition of everolimus was associated with a significant reduction in the risk of progression or death (hazard ratio, 0.40) similar to that previously reported in the entire trial population (HR, 0.45).

Neither alteration of any one of these four genes nor alteration of any one gene in their respective pathways predicted everolimus benefit. "Basically, the treatment effect was independent of the genetic status of these genes and pathways," Dr. Hortobagyi commented.

But when the pathways were considered simultaneously, patients having minimal genetic alteration derived a progression-free survival benefit from everolimus that exceeded that in the whole subset (HR, 0.27), whereas their counterparts having multiple alterations derived a benefit much less than that in the whole subset (HR, 0.78).

Dr. Hortobagyi disclosed that he is a consultant to Allergan, AstraZeneca, Galena Biopharma, Genentech, Myriad Genetics, Novartis, and Sanofi; receives research funding from Novartis; and has other affiliations with Genentech, Novartis, and Sanofi. The BOLERO-2 trial was sponsored by Novartis.

CHICAGO – The benefit of everolimus in hormone receptor–positive, HER2-negative advanced breast cancer appears to be greater for women who have little or no genetic alteration in this cancer’s most commonly altered molecular signaling pathways, based on data presented at the annual meeting of the American Society of Clinical Oncology.

A team led by Dr. Gabriel N. Hortobagyi performed next-generation sequencing on archival tumor tissue from patients enrolled in the randomized BOLERO-2 trial, which tested the addition to exemestane of everolimus, an inhibitor of the mammalian target of rapamycin (mTOR).

Dr. Gabriel N. Hortobagyi

Among all 227 patients whose tumors were tested, adding everolimus reduced the risk of progression or death by 60% as compared with adding placebo.

Patients having alterations in any of the four genes that were most commonly altered in the tumors or that were part of their respective signaling pathways derived similar benefit from everolimus as patients who had wild-type (normal) genes.

Thus, "no predictive marker of everolimus efficacy was identified in individual analysis of the four pathways," commented Dr. Hortobagyi, who chairs the department of breast medical oncology at the University of Texas MD Anderson Cancer Center in Houston.

However, there was differential benefit when patients were broadly grouped according to the extent of alterations – either minimal or multiple.

Three-fourths had minimal (none or one) alteration of these genes or another gene lying on the same pathway: the PIK3CA gene or the PTEN gene (PI3 kinase pathway), the CCND1 gene (cell cycle pathway), or the FGFR1 gene (FGFR1/2 pathway).

This group had a 73% reduction in the risk of progression or death if given everolimus as compared with placebo. In contrast, the group having multiple (two or more) alterations of these genes had a much smaller 22% reduction in risk.

"Further delineation of the interaction of these genetic events might help us to understand how the oncogenic signal cascades work and find better options to attenuate disease progression," Dr. Hortobagyi proposed. "These results are hypothesis generating only and should be validated in independent patient cohorts. But if confirmed, they could lead to new hypotheses for development of novel and more effective combination targeted therapies in hormone receptor–positive, HER2-negative breast cancer."

Session attendee Dr. Larry Norton of the Memorial Sloan-Kettering Cancer Center in New York, asked, "How many of these biopsies came from metastatic lesions and how many came from the primary? And if you just looked at the ones from the metastatic sites, do you see any differences there, because as we know, there is clonal evolution in the metastatic sites."

"About 20% came from metastatic disease, and about 80% from the primary. There are a number of individual differences between the two groups, but unfortunately, the numbers in the metastatic group are so small that you can’t really derive patterns," Dr. Hortobagyi replied. "Now all of the samples, both the primary and the metastatic ones, were from before the study, so it would not really directly reflect on the study itself. But that’s a very important question that we eventually have to address."

Attendee Dr. Ian Krop of the Dana-Farber Cancer Institute in Boston, commented, "It was interesting that those tumors that had multiple alterations in one of those four genes had what appeared to be decreased benefit from everolimus. Could that represent just genomic instability? In other words, if you look at random alterations in your data set, did that also correlate with lack of benefit?"

"We looked at that, and the genetic background for the two groups is very similar," Dr. Hortobagyi replied. "So my a priori thinking was exactly as you mentioned, that those would be associated with instability and therefore multiple driver pathways. But we did not see that."

Women in the BOLERO-2 trial were postmenopausal and had locally advanced or metastatic breast cancer that was refractory to a nonsteroidal aromatase inhibitor. They were assigned 2:1 to exemestane (brand name Aromasin) plus either everolimus (Afinitor) or placebo.

Everolimus is currently approved by the Food and Drug Administration for use in combination with exemestane to treat advanced hormone receptor–positive breast cancer in postmenopausal women. It is also approved for selected indications in other cancers.

In the new retrospective exploratory analysis, Dr. Hortobagyi’s team collected formalin-fixed, paraffin-embedded archival tumor tissue in a subset of patients and performed sequencing for 3,230 exons in 182 oncogenes and tumor suppressor genes.

Results showed that the four most commonly altered genes were PIK3CA (altered in 48% of patients), CCND1 (31%), TP53 (23%), and FGFR1 (18%).

 

 

In the patient subset as a whole, addition of everolimus was associated with a significant reduction in the risk of progression or death (hazard ratio, 0.40) similar to that previously reported in the entire trial population (HR, 0.45).

Neither alteration of any one of these four genes nor alteration of any one gene in their respective pathways predicted everolimus benefit. "Basically, the treatment effect was independent of the genetic status of these genes and pathways," Dr. Hortobagyi commented.

But when the pathways were considered simultaneously, patients having minimal genetic alteration derived a progression-free survival benefit from everolimus that exceeded that in the whole subset (HR, 0.27), whereas their counterparts having multiple alterations derived a benefit much less than that in the whole subset (HR, 0.78).

Dr. Hortobagyi disclosed that he is a consultant to Allergan, AstraZeneca, Galena Biopharma, Genentech, Myriad Genetics, Novartis, and Sanofi; receives research funding from Novartis; and has other affiliations with Genentech, Novartis, and Sanofi. The BOLERO-2 trial was sponsored by Novartis.

Publications
Publications
Topics
Article Type
Display Headline
Genetic alterations affect everolimus benefit in breast cancer
Display Headline
Genetic alterations affect everolimus benefit in breast cancer
Legacy Keywords
everolimus, advanced breast cancer, American Society of Clinical Oncology, Dr. Gabriel N. Hortobagyi, BOLERO-2 trial, everolimus, rapamycin, mTOR)
Legacy Keywords
everolimus, advanced breast cancer, American Society of Clinical Oncology, Dr. Gabriel N. Hortobagyi, BOLERO-2 trial, everolimus, rapamycin, mTOR)
Article Source

AT THE ASCO ANNUAL MEETING 2013

PURLs Copyright

Inside the Article

Vitals

Major Finding: Everolimus decreased the risk of progression or death by 73% in patients whose tumors had none or one genetic alteration in the most commonly altered signaling pathways and by 22% in patients whose tumors had multiple alterations.

Data Source: An exploratory analysis among 227 patients with hormone receptor–positive, HER2-negative advanced breast cancer (BOLERO-2 trial)

Disclosures: Dr. Hortobagyi disclosed that he is a consultant to Allergan, AstraZeneca, Galena Biopharma, Genentech, Myriad Genetics, Novartis, and Sanofi; receives research funding from Novartis; and has other affiliations with Genentech, Novartis, and Sanofi. The BOLERO-2 trial was sponsored by Novartis.

Upamostat falls short in HER2-negative metastatic breast cancer

Testing targeted agents remains challenging
Article Type
Changed
Thu, 12/15/2022 - 18:16
Display Headline
Upamostat falls short in HER2-negative metastatic breast cancer

CHICAGO – Upamostat, an investigational oral inhibitor of urokinase-type plasminogen activator, did not add to the efficacy of capecitabine monotherapy when given as first-line therapy to unselected patients with HER2-negative metastatic breast cancer, based on the results of a phase II trial.

The 132 women studied were assigned evenly to capecitabine monotherapy or to capecitabine plus upamostat. Median progression-free survival, the trial’s primary endpoint, was statistically indistinguishable at about 8 months in each group.

Courtesy Fox Chase Cancer Center
Dr. Lori Goldstein

However, upamostat was associated with a 4-month gain in this outcome in the subset of patients who had previously received chemotherapy in the adjuvant or neoadjuvant setting, an unplanned exploratory analysis found.

"Treatment with prior chemotherapy potentially defines a more homogeneous subpopulation in this study. Also, prior chemotherapy could possibly identify a population with more aggressive disease due to higher expression of uPA [urokinase-type plasminogen activator]," said lead author Dr. Lori J. Goldstein of the Fox Chase Cancer Center in Philadelphia. "This would be in line with the treatment effects seen in the group who progressed early after initial diagnosis."

"Future studies with upamostat should target a more homogeneous patient population and incorporate biomarker-specific strategies for patient characterization and selection," Dr. Goldstein recommended.

Adding upamostat to chemotherapy, for nearly 3 years in some cases, was generally safe and well tolerated. The only increase in adverse events was in the incidence of hand-foot syndrome.

"Upamostat can safely be given over extended periods of time. [The study] is also the first proof of activity of an anti-uPA inhibitor in breast cancer therapy," Dr. Goldstein said at the annual meeting of the American Society of Clinical Oncology.

Part of the plasminogen activation system, uPA promotes tumor cell invasion, migration, and proliferation, Dr. Goldstein explained, giving some background to the research.

"As the uPA system is quantitatively more expressed in tumor cells and tumor-associated stromal cells, it is an interesting target for cancer therapy. uPA and PAI-1 [plasminogen activator inhibitor-1] are both ASCO-recommended prognostic and predictive biomarkers for early-stage breast cancer," she said.

Among all the patients enrolled in the trial, median progression-free survival was 8.3 months with the upamostat-capecitabine combination and 7.5 months with capecitabine alone, a nonsignificant difference. The estimated 6-month rates of progression-free survival were 56% and 50%.

Archival tumor specimens were not available for uPA and PAI-1 testing, according to Dr. Goldstein. And testing of serial plasma samples for a variety of biomarkers – uPA, PAI-1, CAIX (a marker associated with poorer prognostic features), and the fibrin degradation product D-dimer – failed to show any differences between groups.

In unplanned exploratory analyses, median progression-free survival among patients who had received prior adjuvant or neoadjuvant chemotherapy was 8.3 months with upamostat-capecitabine combination therapy and 4.3 months with capecitabine monotherapy.

The combination therapy also seemed to have an edge among patients who had experienced their recurrence within 3 years from initial diagnosis (5.6 vs. 2.7 months), Dr. Goldstein reported.

The upamostat-capecitabine group had a higher rate of any-grade hand-foot syndrome (77% vs. 46%), possibly related to a longer duration of treatment, whereas rates of other toxicities were similar.

"The pharmacokinetic analyses demonstrated no drug-drug interactions between upamostat and capecitabine," she noted.

Dr. Goldstein disclosed that she receives research funding from the U.S. Department of Defense and Wilex. The trial was sponsored by Wilex, the maker of upamostat.

Body

Overall, I think we’d have to conclude that this is a negative trial. But it actually begs the question as to how certain we are that this drug has no role in breast cancer.

It remains unclear as to whether upamostat hit its target and whether activation of uPA or its naturally occurring inhibitor PAI-1 is even necessary for benefit, as archival tissue was not obtained and, even if it had been, the relevant assays require fresh tissue. So the correlative questions that we all want to answer simply cannot be answered at this time.

The study illustrates the challenges in designing trials of targeted agents. We have hundreds of agents in development. The prevailing wisdom is that many of these agents will only benefit a small minority and that combinations will often be needed. There is a substantial risk of falsely concluding that an agent is inactive depending on the design of the study. And given all of these agents and the expense of developing agents, many drugs will simply not be given a second chance.

Thus, these trials must be carefully designed with consideration of factors such as whether the drug target can be reliably measured, whether power is adequate to assess benefit in relevant subgroups, and what is the best endpoint.

Dr. Eric P. Winer, of the Dana-Farber Cancer Institute in Boston, was the invited discussant of the study. Dr. Farber disclosed that he is a consultant to AstraZeneca, Merrimack, and other companies, and receives research funding from Genentech.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
Upamostat, urokinase-type plasminogen activator, capecitabine monotherapy, metastatic breast cancer
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event
Body

Overall, I think we’d have to conclude that this is a negative trial. But it actually begs the question as to how certain we are that this drug has no role in breast cancer.

It remains unclear as to whether upamostat hit its target and whether activation of uPA or its naturally occurring inhibitor PAI-1 is even necessary for benefit, as archival tissue was not obtained and, even if it had been, the relevant assays require fresh tissue. So the correlative questions that we all want to answer simply cannot be answered at this time.

The study illustrates the challenges in designing trials of targeted agents. We have hundreds of agents in development. The prevailing wisdom is that many of these agents will only benefit a small minority and that combinations will often be needed. There is a substantial risk of falsely concluding that an agent is inactive depending on the design of the study. And given all of these agents and the expense of developing agents, many drugs will simply not be given a second chance.

Thus, these trials must be carefully designed with consideration of factors such as whether the drug target can be reliably measured, whether power is adequate to assess benefit in relevant subgroups, and what is the best endpoint.

Dr. Eric P. Winer, of the Dana-Farber Cancer Institute in Boston, was the invited discussant of the study. Dr. Farber disclosed that he is a consultant to AstraZeneca, Merrimack, and other companies, and receives research funding from Genentech.

Body

Overall, I think we’d have to conclude that this is a negative trial. But it actually begs the question as to how certain we are that this drug has no role in breast cancer.

It remains unclear as to whether upamostat hit its target and whether activation of uPA or its naturally occurring inhibitor PAI-1 is even necessary for benefit, as archival tissue was not obtained and, even if it had been, the relevant assays require fresh tissue. So the correlative questions that we all want to answer simply cannot be answered at this time.

The study illustrates the challenges in designing trials of targeted agents. We have hundreds of agents in development. The prevailing wisdom is that many of these agents will only benefit a small minority and that combinations will often be needed. There is a substantial risk of falsely concluding that an agent is inactive depending on the design of the study. And given all of these agents and the expense of developing agents, many drugs will simply not be given a second chance.

Thus, these trials must be carefully designed with consideration of factors such as whether the drug target can be reliably measured, whether power is adequate to assess benefit in relevant subgroups, and what is the best endpoint.

Dr. Eric P. Winer, of the Dana-Farber Cancer Institute in Boston, was the invited discussant of the study. Dr. Farber disclosed that he is a consultant to AstraZeneca, Merrimack, and other companies, and receives research funding from Genentech.

Title
Testing targeted agents remains challenging
Testing targeted agents remains challenging

CHICAGO – Upamostat, an investigational oral inhibitor of urokinase-type plasminogen activator, did not add to the efficacy of capecitabine monotherapy when given as first-line therapy to unselected patients with HER2-negative metastatic breast cancer, based on the results of a phase II trial.

The 132 women studied were assigned evenly to capecitabine monotherapy or to capecitabine plus upamostat. Median progression-free survival, the trial’s primary endpoint, was statistically indistinguishable at about 8 months in each group.

Courtesy Fox Chase Cancer Center
Dr. Lori Goldstein

However, upamostat was associated with a 4-month gain in this outcome in the subset of patients who had previously received chemotherapy in the adjuvant or neoadjuvant setting, an unplanned exploratory analysis found.

"Treatment with prior chemotherapy potentially defines a more homogeneous subpopulation in this study. Also, prior chemotherapy could possibly identify a population with more aggressive disease due to higher expression of uPA [urokinase-type plasminogen activator]," said lead author Dr. Lori J. Goldstein of the Fox Chase Cancer Center in Philadelphia. "This would be in line with the treatment effects seen in the group who progressed early after initial diagnosis."

"Future studies with upamostat should target a more homogeneous patient population and incorporate biomarker-specific strategies for patient characterization and selection," Dr. Goldstein recommended.

Adding upamostat to chemotherapy, for nearly 3 years in some cases, was generally safe and well tolerated. The only increase in adverse events was in the incidence of hand-foot syndrome.

"Upamostat can safely be given over extended periods of time. [The study] is also the first proof of activity of an anti-uPA inhibitor in breast cancer therapy," Dr. Goldstein said at the annual meeting of the American Society of Clinical Oncology.

Part of the plasminogen activation system, uPA promotes tumor cell invasion, migration, and proliferation, Dr. Goldstein explained, giving some background to the research.

"As the uPA system is quantitatively more expressed in tumor cells and tumor-associated stromal cells, it is an interesting target for cancer therapy. uPA and PAI-1 [plasminogen activator inhibitor-1] are both ASCO-recommended prognostic and predictive biomarkers for early-stage breast cancer," she said.

Among all the patients enrolled in the trial, median progression-free survival was 8.3 months with the upamostat-capecitabine combination and 7.5 months with capecitabine alone, a nonsignificant difference. The estimated 6-month rates of progression-free survival were 56% and 50%.

Archival tumor specimens were not available for uPA and PAI-1 testing, according to Dr. Goldstein. And testing of serial plasma samples for a variety of biomarkers – uPA, PAI-1, CAIX (a marker associated with poorer prognostic features), and the fibrin degradation product D-dimer – failed to show any differences between groups.

In unplanned exploratory analyses, median progression-free survival among patients who had received prior adjuvant or neoadjuvant chemotherapy was 8.3 months with upamostat-capecitabine combination therapy and 4.3 months with capecitabine monotherapy.

The combination therapy also seemed to have an edge among patients who had experienced their recurrence within 3 years from initial diagnosis (5.6 vs. 2.7 months), Dr. Goldstein reported.

The upamostat-capecitabine group had a higher rate of any-grade hand-foot syndrome (77% vs. 46%), possibly related to a longer duration of treatment, whereas rates of other toxicities were similar.

"The pharmacokinetic analyses demonstrated no drug-drug interactions between upamostat and capecitabine," she noted.

Dr. Goldstein disclosed that she receives research funding from the U.S. Department of Defense and Wilex. The trial was sponsored by Wilex, the maker of upamostat.

CHICAGO – Upamostat, an investigational oral inhibitor of urokinase-type plasminogen activator, did not add to the efficacy of capecitabine monotherapy when given as first-line therapy to unselected patients with HER2-negative metastatic breast cancer, based on the results of a phase II trial.

The 132 women studied were assigned evenly to capecitabine monotherapy or to capecitabine plus upamostat. Median progression-free survival, the trial’s primary endpoint, was statistically indistinguishable at about 8 months in each group.

Courtesy Fox Chase Cancer Center
Dr. Lori Goldstein

However, upamostat was associated with a 4-month gain in this outcome in the subset of patients who had previously received chemotherapy in the adjuvant or neoadjuvant setting, an unplanned exploratory analysis found.

"Treatment with prior chemotherapy potentially defines a more homogeneous subpopulation in this study. Also, prior chemotherapy could possibly identify a population with more aggressive disease due to higher expression of uPA [urokinase-type plasminogen activator]," said lead author Dr. Lori J. Goldstein of the Fox Chase Cancer Center in Philadelphia. "This would be in line with the treatment effects seen in the group who progressed early after initial diagnosis."

"Future studies with upamostat should target a more homogeneous patient population and incorporate biomarker-specific strategies for patient characterization and selection," Dr. Goldstein recommended.

Adding upamostat to chemotherapy, for nearly 3 years in some cases, was generally safe and well tolerated. The only increase in adverse events was in the incidence of hand-foot syndrome.

"Upamostat can safely be given over extended periods of time. [The study] is also the first proof of activity of an anti-uPA inhibitor in breast cancer therapy," Dr. Goldstein said at the annual meeting of the American Society of Clinical Oncology.

Part of the plasminogen activation system, uPA promotes tumor cell invasion, migration, and proliferation, Dr. Goldstein explained, giving some background to the research.

"As the uPA system is quantitatively more expressed in tumor cells and tumor-associated stromal cells, it is an interesting target for cancer therapy. uPA and PAI-1 [plasminogen activator inhibitor-1] are both ASCO-recommended prognostic and predictive biomarkers for early-stage breast cancer," she said.

Among all the patients enrolled in the trial, median progression-free survival was 8.3 months with the upamostat-capecitabine combination and 7.5 months with capecitabine alone, a nonsignificant difference. The estimated 6-month rates of progression-free survival were 56% and 50%.

Archival tumor specimens were not available for uPA and PAI-1 testing, according to Dr. Goldstein. And testing of serial plasma samples for a variety of biomarkers – uPA, PAI-1, CAIX (a marker associated with poorer prognostic features), and the fibrin degradation product D-dimer – failed to show any differences between groups.

In unplanned exploratory analyses, median progression-free survival among patients who had received prior adjuvant or neoadjuvant chemotherapy was 8.3 months with upamostat-capecitabine combination therapy and 4.3 months with capecitabine monotherapy.

The combination therapy also seemed to have an edge among patients who had experienced their recurrence within 3 years from initial diagnosis (5.6 vs. 2.7 months), Dr. Goldstein reported.

The upamostat-capecitabine group had a higher rate of any-grade hand-foot syndrome (77% vs. 46%), possibly related to a longer duration of treatment, whereas rates of other toxicities were similar.

"The pharmacokinetic analyses demonstrated no drug-drug interactions between upamostat and capecitabine," she noted.

Dr. Goldstein disclosed that she receives research funding from the U.S. Department of Defense and Wilex. The trial was sponsored by Wilex, the maker of upamostat.

Publications
Publications
Topics
Article Type
Display Headline
Upamostat falls short in HER2-negative metastatic breast cancer
Display Headline
Upamostat falls short in HER2-negative metastatic breast cancer
Legacy Keywords
Upamostat, urokinase-type plasminogen activator, capecitabine monotherapy, metastatic breast cancer
Legacy Keywords
Upamostat, urokinase-type plasminogen activator, capecitabine monotherapy, metastatic breast cancer
Article Source

AT THE ASCO ANNUAL MEETING 2013

PURLs Copyright

Inside the Article

Vitals

Major finding: Adding upamostat vs. placebo did not significantly prolong progression-free survival in the entire trial population (8.3 vs. 7.5 months), but there was a suggestion of benefit in the subset previously given chemotherapy (8.3 vs. 4.3 months).

Data source: A randomized double-blind phase II trial comparing upamostat plus capecitabine vs. capecitabine alone as first-line therapy among 132 patients with HER2-negative metastatic breast cancer

Disclosures: Dr. Goldstein disclosed that she receives research funding from the U.S. Department of Defense and Wilex. The trial was sponsored by Wilex, the maker of upamostat.

Prolaris test eyed as predictor of prostate cancer outcomes

Outcome prediction doesn’t reflect clinical utility
Article Type
Changed
Fri, 01/04/2019 - 12:14
Display Headline
Prolaris test eyed as predictor of prostate cancer outcomes

CHICAGO – Prostate cancer outcomes were predicted by a test that measures the expression of cell cycle progression genes, according to results from a retrospective analysis of prostate tissue samples from five patient cohorts.

The Prolaris test gives each tissue sample a cell cycle progression (CCP) score based on measures of 31 CCP genes, normalized to 15 "housekeeper" genes. A unit change in the test is defined as a doubling in CCP genes. For each unit increase in the test’s score, there was a two- to threefold increase in the risk of disease progression, Dr. Jack M. Cuzick reported at the annual meeting of the American Society of Clinical Oncology.

The CCP signature of Myriad Genetics’ Prolaris test was a highly significant predictor of outcome, said Dr. Cuzick of the Wolfson Institute of Preventive Medicine, London. In all five studies, the hazard ratio per unit change in the CCP score was similar, ranging from 1.89 to 2.92. The findings indicate that the effect size for the CCP score is robust in multiple patient cohorts and diverse clinical settings.

The test provides information for differentiating aggressive and indolent disease beyond that available from clinicopathologic variables, he said. As the natural history of prostate cancer can be variable and difficult to predict, the Prolaris test could help to match treatment more appropriately to each individual’s risk of progression.

In the study that examined the test’s predictive value, five patient groups were evaluated. Formalin-fixed tissue samples were obtained from two English patient cohorts that were conservatively managed (n = 337 and 349), two U.S. patient cohorts that underwent radical prostatectomy (366 men treated at Scott & White Hospital, Temple, Tex.; and 413 men treated at the University of California, San Francisco), and one U.S. cohort that underwent external beam radiation therapy (141 men treated at the Durham, N.C., VA Medical Center).

The cohort of conservatively managed English patients was from the late 1990s and had more than 15 years of follow up. In the 337-patient cohort diagnosed via transurethral resection of the prostate (TURP) and conservatively managed, there were 57 deaths from prostate cancer. In the 349-patient cohort diagnosed via needle biopsy and conservatively managed, there were 90 deaths from prostate cancer.

For each unit increase in the CCP score, the hazard ratio for the cohort diagnosed via TURP was 2.9 and the hazard ratio for those diagnosed via needle biopsy was 2. The CCP score was the dominant variable for predicting death from prostate cancer in univariate analysis (P = 6.1 x 10–22 after diagnosis via TURP, and P = 8.6 x 10–10 after diagnosis via needle biopsy). In both studies, the CCP score remained highly significant in multivariate analysis and was a stronger predictor of disease-specific mortality than other prognostic variables, he said.

In the U.S. prostatectomy cohorts, there were 132 biochemical recurrences (BCRs) in the first cohort and 83 BCRs in the second cohort. With each unit increase in the CCP score, there was a doubling of risk for recurrence. After prostatectomy, the CCP score predicted BCR in univariate analysis (Scott & White: P = 5.6 x 10–9; University of California: P = 2.23 x 10–6) and provided additional prognostic information in multivariate analysis (Scott & White: P = 3.3 x 10–6; University of California: P = 9.5 x10–5).

After radiation therapy, the CCP score predicted BCR in univariate (P = .0017) and multivariate (P = .034) analysis. In the 141-patient cohort that was diagnosed by needle biopsy and underwent external beam radiation, there were 19 prostate cancer deaths and more than a doubling of risk with each unit increase in CCP score.

CCP scores only modestly correlated with the Gleason score and prostate-specific antigen (PSA) value. The test adds value beyond those measures, Dr. Cuzick said.

CCP scores predict patient outcome in multiple clinical settings, provide independent information beyond clinicopathological variables, and help to further differentiate aggressive from indolent prostate cancer. With low-grade Gleason 6 cancers, the results can aid in telling who is at low risk and who needs aggressive therapy, he concluded.

The study was funded by Myriad Genetics, the maker of the Prolaris test. Dr. Cuzick received honoraria and research support from Myriad.

Body

The ability to improve clinical management by finding prostate cancer patients who would benefit from more – or less – therapy is much needed. Clinicians are concerned that many prostate cancer patients are now overtreated, but they lack reliable prognostic guides.

Cell cycle progression (CCP) scores are interesting retrospectively, but how much are they able to improve on CAPRA (Cancer of the Prostate Risk Assessment) scores for predicting prognosis?

Prostate cancer is uniquely multifocal, with most men having multiple independent foci of cancer. In the example of the conservatively managed patients, if one is looking at men with indolent disease who have low-volume disease as a single core of one or two foci, is one really going to be able to predict the biologic outcome of the cancer? Those who fail after a surveillance approach often do so early and had undersampling of their disease. So it hasn’t been proven yet that this test can predict the behavior of cancer that hasn’t been sampled.

The CCP results proved to be statistically significant, but that finding does not indicate clinical utility. It’s not known whether the novel biomarkers in this test improve on existing markers. You find yourself asking what you would do differently in a patient whose risk of progression goes from 7% to 12%.

Even if a test independently predicts outcome, that doesn’t necessarily indicate it has clinical utility. The ability to improve clinical management is key to the adoption of new prognostic tests. The real question is whether CCP results improve on the existing model. Does the test improve on CAPRA for prognosis?

Dr. Scott Tomlins is with the department of urology at the University of Michigan Health System, Ann Arbor. He was the invited discussant of the paper at the meeting. Dr. Tomlins disclosed that he is a consultant to and receives honoraria from Ventana Medical Systems/Roche. He has patents via the University of Michigan on several diagnostic genetic tests.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
Prostate cancer, cell cycle progression genes, Prolaris, CCP, Dr. Jack M. Cuzick, American Society of Clinical Oncology
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event
Body

The ability to improve clinical management by finding prostate cancer patients who would benefit from more – or less – therapy is much needed. Clinicians are concerned that many prostate cancer patients are now overtreated, but they lack reliable prognostic guides.

Cell cycle progression (CCP) scores are interesting retrospectively, but how much are they able to improve on CAPRA (Cancer of the Prostate Risk Assessment) scores for predicting prognosis?

Prostate cancer is uniquely multifocal, with most men having multiple independent foci of cancer. In the example of the conservatively managed patients, if one is looking at men with indolent disease who have low-volume disease as a single core of one or two foci, is one really going to be able to predict the biologic outcome of the cancer? Those who fail after a surveillance approach often do so early and had undersampling of their disease. So it hasn’t been proven yet that this test can predict the behavior of cancer that hasn’t been sampled.

The CCP results proved to be statistically significant, but that finding does not indicate clinical utility. It’s not known whether the novel biomarkers in this test improve on existing markers. You find yourself asking what you would do differently in a patient whose risk of progression goes from 7% to 12%.

Even if a test independently predicts outcome, that doesn’t necessarily indicate it has clinical utility. The ability to improve clinical management is key to the adoption of new prognostic tests. The real question is whether CCP results improve on the existing model. Does the test improve on CAPRA for prognosis?

Dr. Scott Tomlins is with the department of urology at the University of Michigan Health System, Ann Arbor. He was the invited discussant of the paper at the meeting. Dr. Tomlins disclosed that he is a consultant to and receives honoraria from Ventana Medical Systems/Roche. He has patents via the University of Michigan on several diagnostic genetic tests.

Body

The ability to improve clinical management by finding prostate cancer patients who would benefit from more – or less – therapy is much needed. Clinicians are concerned that many prostate cancer patients are now overtreated, but they lack reliable prognostic guides.

Cell cycle progression (CCP) scores are interesting retrospectively, but how much are they able to improve on CAPRA (Cancer of the Prostate Risk Assessment) scores for predicting prognosis?

Prostate cancer is uniquely multifocal, with most men having multiple independent foci of cancer. In the example of the conservatively managed patients, if one is looking at men with indolent disease who have low-volume disease as a single core of one or two foci, is one really going to be able to predict the biologic outcome of the cancer? Those who fail after a surveillance approach often do so early and had undersampling of their disease. So it hasn’t been proven yet that this test can predict the behavior of cancer that hasn’t been sampled.

The CCP results proved to be statistically significant, but that finding does not indicate clinical utility. It’s not known whether the novel biomarkers in this test improve on existing markers. You find yourself asking what you would do differently in a patient whose risk of progression goes from 7% to 12%.

Even if a test independently predicts outcome, that doesn’t necessarily indicate it has clinical utility. The ability to improve clinical management is key to the adoption of new prognostic tests. The real question is whether CCP results improve on the existing model. Does the test improve on CAPRA for prognosis?

Dr. Scott Tomlins is with the department of urology at the University of Michigan Health System, Ann Arbor. He was the invited discussant of the paper at the meeting. Dr. Tomlins disclosed that he is a consultant to and receives honoraria from Ventana Medical Systems/Roche. He has patents via the University of Michigan on several diagnostic genetic tests.

Title
Outcome prediction doesn’t reflect clinical utility
Outcome prediction doesn’t reflect clinical utility

CHICAGO – Prostate cancer outcomes were predicted by a test that measures the expression of cell cycle progression genes, according to results from a retrospective analysis of prostate tissue samples from five patient cohorts.

The Prolaris test gives each tissue sample a cell cycle progression (CCP) score based on measures of 31 CCP genes, normalized to 15 "housekeeper" genes. A unit change in the test is defined as a doubling in CCP genes. For each unit increase in the test’s score, there was a two- to threefold increase in the risk of disease progression, Dr. Jack M. Cuzick reported at the annual meeting of the American Society of Clinical Oncology.

The CCP signature of Myriad Genetics’ Prolaris test was a highly significant predictor of outcome, said Dr. Cuzick of the Wolfson Institute of Preventive Medicine, London. In all five studies, the hazard ratio per unit change in the CCP score was similar, ranging from 1.89 to 2.92. The findings indicate that the effect size for the CCP score is robust in multiple patient cohorts and diverse clinical settings.

The test provides information for differentiating aggressive and indolent disease beyond that available from clinicopathologic variables, he said. As the natural history of prostate cancer can be variable and difficult to predict, the Prolaris test could help to match treatment more appropriately to each individual’s risk of progression.

In the study that examined the test’s predictive value, five patient groups were evaluated. Formalin-fixed tissue samples were obtained from two English patient cohorts that were conservatively managed (n = 337 and 349), two U.S. patient cohorts that underwent radical prostatectomy (366 men treated at Scott & White Hospital, Temple, Tex.; and 413 men treated at the University of California, San Francisco), and one U.S. cohort that underwent external beam radiation therapy (141 men treated at the Durham, N.C., VA Medical Center).

The cohort of conservatively managed English patients was from the late 1990s and had more than 15 years of follow up. In the 337-patient cohort diagnosed via transurethral resection of the prostate (TURP) and conservatively managed, there were 57 deaths from prostate cancer. In the 349-patient cohort diagnosed via needle biopsy and conservatively managed, there were 90 deaths from prostate cancer.

For each unit increase in the CCP score, the hazard ratio for the cohort diagnosed via TURP was 2.9 and the hazard ratio for those diagnosed via needle biopsy was 2. The CCP score was the dominant variable for predicting death from prostate cancer in univariate analysis (P = 6.1 x 10–22 after diagnosis via TURP, and P = 8.6 x 10–10 after diagnosis via needle biopsy). In both studies, the CCP score remained highly significant in multivariate analysis and was a stronger predictor of disease-specific mortality than other prognostic variables, he said.

In the U.S. prostatectomy cohorts, there were 132 biochemical recurrences (BCRs) in the first cohort and 83 BCRs in the second cohort. With each unit increase in the CCP score, there was a doubling of risk for recurrence. After prostatectomy, the CCP score predicted BCR in univariate analysis (Scott & White: P = 5.6 x 10–9; University of California: P = 2.23 x 10–6) and provided additional prognostic information in multivariate analysis (Scott & White: P = 3.3 x 10–6; University of California: P = 9.5 x10–5).

After radiation therapy, the CCP score predicted BCR in univariate (P = .0017) and multivariate (P = .034) analysis. In the 141-patient cohort that was diagnosed by needle biopsy and underwent external beam radiation, there were 19 prostate cancer deaths and more than a doubling of risk with each unit increase in CCP score.

CCP scores only modestly correlated with the Gleason score and prostate-specific antigen (PSA) value. The test adds value beyond those measures, Dr. Cuzick said.

CCP scores predict patient outcome in multiple clinical settings, provide independent information beyond clinicopathological variables, and help to further differentiate aggressive from indolent prostate cancer. With low-grade Gleason 6 cancers, the results can aid in telling who is at low risk and who needs aggressive therapy, he concluded.

The study was funded by Myriad Genetics, the maker of the Prolaris test. Dr. Cuzick received honoraria and research support from Myriad.

CHICAGO – Prostate cancer outcomes were predicted by a test that measures the expression of cell cycle progression genes, according to results from a retrospective analysis of prostate tissue samples from five patient cohorts.

The Prolaris test gives each tissue sample a cell cycle progression (CCP) score based on measures of 31 CCP genes, normalized to 15 "housekeeper" genes. A unit change in the test is defined as a doubling in CCP genes. For each unit increase in the test’s score, there was a two- to threefold increase in the risk of disease progression, Dr. Jack M. Cuzick reported at the annual meeting of the American Society of Clinical Oncology.

The CCP signature of Myriad Genetics’ Prolaris test was a highly significant predictor of outcome, said Dr. Cuzick of the Wolfson Institute of Preventive Medicine, London. In all five studies, the hazard ratio per unit change in the CCP score was similar, ranging from 1.89 to 2.92. The findings indicate that the effect size for the CCP score is robust in multiple patient cohorts and diverse clinical settings.

The test provides information for differentiating aggressive and indolent disease beyond that available from clinicopathologic variables, he said. As the natural history of prostate cancer can be variable and difficult to predict, the Prolaris test could help to match treatment more appropriately to each individual’s risk of progression.

In the study that examined the test’s predictive value, five patient groups were evaluated. Formalin-fixed tissue samples were obtained from two English patient cohorts that were conservatively managed (n = 337 and 349), two U.S. patient cohorts that underwent radical prostatectomy (366 men treated at Scott & White Hospital, Temple, Tex.; and 413 men treated at the University of California, San Francisco), and one U.S. cohort that underwent external beam radiation therapy (141 men treated at the Durham, N.C., VA Medical Center).

The cohort of conservatively managed English patients was from the late 1990s and had more than 15 years of follow up. In the 337-patient cohort diagnosed via transurethral resection of the prostate (TURP) and conservatively managed, there were 57 deaths from prostate cancer. In the 349-patient cohort diagnosed via needle biopsy and conservatively managed, there were 90 deaths from prostate cancer.

For each unit increase in the CCP score, the hazard ratio for the cohort diagnosed via TURP was 2.9 and the hazard ratio for those diagnosed via needle biopsy was 2. The CCP score was the dominant variable for predicting death from prostate cancer in univariate analysis (P = 6.1 x 10–22 after diagnosis via TURP, and P = 8.6 x 10–10 after diagnosis via needle biopsy). In both studies, the CCP score remained highly significant in multivariate analysis and was a stronger predictor of disease-specific mortality than other prognostic variables, he said.

In the U.S. prostatectomy cohorts, there were 132 biochemical recurrences (BCRs) in the first cohort and 83 BCRs in the second cohort. With each unit increase in the CCP score, there was a doubling of risk for recurrence. After prostatectomy, the CCP score predicted BCR in univariate analysis (Scott & White: P = 5.6 x 10–9; University of California: P = 2.23 x 10–6) and provided additional prognostic information in multivariate analysis (Scott & White: P = 3.3 x 10–6; University of California: P = 9.5 x10–5).

After radiation therapy, the CCP score predicted BCR in univariate (P = .0017) and multivariate (P = .034) analysis. In the 141-patient cohort that was diagnosed by needle biopsy and underwent external beam radiation, there were 19 prostate cancer deaths and more than a doubling of risk with each unit increase in CCP score.

CCP scores only modestly correlated with the Gleason score and prostate-specific antigen (PSA) value. The test adds value beyond those measures, Dr. Cuzick said.

CCP scores predict patient outcome in multiple clinical settings, provide independent information beyond clinicopathological variables, and help to further differentiate aggressive from indolent prostate cancer. With low-grade Gleason 6 cancers, the results can aid in telling who is at low risk and who needs aggressive therapy, he concluded.

The study was funded by Myriad Genetics, the maker of the Prolaris test. Dr. Cuzick received honoraria and research support from Myriad.

Publications
Publications
Topics
Article Type
Display Headline
Prolaris test eyed as predictor of prostate cancer outcomes
Display Headline
Prolaris test eyed as predictor of prostate cancer outcomes
Legacy Keywords
Prostate cancer, cell cycle progression genes, Prolaris, CCP, Dr. Jack M. Cuzick, American Society of Clinical Oncology
Legacy Keywords
Prostate cancer, cell cycle progression genes, Prolaris, CCP, Dr. Jack M. Cuzick, American Society of Clinical Oncology
Sections
Article Source

AT THE ASCO ANNUAL MEETING 2013

PURLs Copyright

Inside the Article

Vitals

Major finding: In conservatively managed prostate cancer patients, the cell cycle progression score in tissue samples was the dominant variable for predicting death from prostate cancer in univariate analysis (P = 6.1 x 10–22 after diagnosis via TURP, and P = 8.6 x 10–10 after diagnosis via needle biopsy).

Data source: A retrospective study of tissue samples from more than 1,600 patients in five patient cohorts who were either managed conservatively, underwent prostatectomy, or received external beam radiotherapy.

Disclosures: The study was funded by Myriad Genetics, the maker of the Prolaris test. Dr. Cuzick received honoraria and research support from Myriad.

BOLERO 3: Everolimus may overcome trastuzumab resistance in HER2-positive breast cancer

An option after pertuzumab and TDM-1
Article Type
Changed
Thu, 12/15/2022 - 18:16
Display Headline
BOLERO 3: Everolimus may overcome trastuzumab resistance in HER2-positive breast cancer

CHICAGO – The mTOR inhibitor everolimus appears to be effective at overcoming trastuzumab resistance in patients with advanced HER2-positive breast cancer, based on findings from the BOLERO-3 trial.

The 572 patients studied all had locally advanced or metastatic HER2-positive breast cancer that had progressed despite trastuzumab therapy.

The main trial results, reported at the annual meeting of the American Society of Clinical Oncology, showed that when everolimus (Afinitor) was added to trastuzumab and vinorelbine, it reduced the risk of progression or death by 22% compared with placebo, a difference amounting to about a 1-month benefit.

As expected, patients receiving everolimus had higher rates of certain grade 3/4 adverse events, such as stomatitis, but the two groups did not differ with respect to quality of life. Overall survival results are not yet mature.

"This is the first phase III study showing the benefit of mTOR [mammalian target of rapamycin] pathway inhibition in HER2-positive breast cancer. Targeting mTOR is a viable approach to maximize the benefit of trastuzumab-based therapy," commented first author Dr. Ruth O’Regan, director of translational breast cancer research at the Winship Cancer Institute of Emory University in Atlanta. "The combination of everolimus with vinorelbine and trastuzumab can be considered an appropriate option for some patients with trastuzumab-resistant HER2-positive breast cancer."

"The ongoing BOLERO-1 trial will evaluate the addition of everolimus to chemotherapy and trastuzumab in the first-line setting," she added.

Session attendee Dr. Jose Baselga, physician-in-chief of Memorial Hospital at the Memorial Sloan-Kettering Cancer Center in New York, said, "In looking at the data, the first reaction I have is that you have two groups: the ER [estrogen receptor]-positives and the ER-negatives. And I’m just wondering whether this theme of mTOR and ER, this cross-talk, is so important, that whether even in the HER2-positives, we need to address the issue of ER blockade at the same time, because if the study had been only in the ER-negatives, this would have been a massively positive trial."

"It is possible that when you inhibit mTOR, in that situation, that perhaps we do need to also inhibit ER," Dr. O’Regan agreed. "It’s a fairly large trial, and the [progression-free survival] lines cross completely in the hormone receptor–positive group. So that’s a very good question. I think maybe it’s worthwhile looking at HER2, mTOR, estrogen receptor blockade in another trial."

Dr. Steven Vogl, a medical oncologist in the Bronx, N.Y., asked whether BOLERO-1 is powered to look at ER-positive and ER-negative patients individually. "Your benefit was small and probably confined to only the ER-negative patients. It would be nice to know how big the benefit is in those patients and to have a second trial showing us that we need to give everolimus to these patients only if they are ER negative."

Dr. O’Regan said she did not know whether BOLERO-1 was adequately powered to answer that question. "I doubt it is, in reality, but I think we will get some interesting data. If we find the same thing in that study, that will be something certainly we can take forward," she said.

Dr. Alan Astrow, of the Maimonides Medical Center in Brooklyn, N.Y., who also attended the session, noted that the new data suggest that everolimus now presents a third treatment option in this patient population, in addition to pertuzumab (Perjeta) and TDM-1 (Kadcyla). "So what are your thoughts about further development of this drug now that we have other drugs available?" he asked.

"At this point, it would probably be third-line after those agents in the metastatic setting," Dr. O’Regan replied. "The question is, where do you put it in regard to lapatinib? And are we going to have to start looking at ER-negative versus ER-positive differences – would you put this combination up higher in the ER-negative, HER2-positive group? There are a lot of questions that we just don’t know the answer to right now."

Session comoderator Dr. Rebecca Alexander Dent, an oncologist with Duke-NUS in Singapore, the University of Toronto, and the Sunnybrook Odette Cancer Center in Toronto, said, "Obviously, the doses [of everolimus] that have been used in trials are somewhat different. Can you comment on what kind of impact that might make or how we should move forward with that?"

Dr. O’Regan said the 5-mg dose used was based on phase I data, whereas other trials, such as BOLERO-1, are using a 10-mg dose. "Obviously, we don’t know if we had used a higher dose if we would have had a greater progression-free survival difference, but it would definitely increase the toxicity," she said; the optimal dose may depend on whether it is given with chemotherapy.

 

 

The patients in the BOLERO-3 trial were randomized evenly to daily everolimus versus daily placebo, each added to weekly trastuzumab (Herceptin) and vinorelbine.

Everolimus is currently approved by the Food and Drug Administration for use in combination with exemestane to treat advanced hormone receptor–positive, HER2-positive breast cancer in postmenopausal women. It is also approved for selected indications in other cancers.

A fourth of patients had previously received lapatinib (Tykerb). The large majority had received one or two prior lines of therapy for metastatic disease.

Median progression-free survival was 1.22 months longer with everolimus (7.00 vs 5.78 months; hazard ratio, 0.78; P = .0067).

The overall rate of response did not differ significantly between the everolimus and placebo groups (41% vs. 37%).

The everolimus arm had higher rates than the placebo group for grade 3/4 stomatitis (13% vs. 1%), fatigue (12% vs. 4%), and hematologic adverse events. But the groups were statistically indistinguishable with respect to the time to definitive deterioration of global health status.

The rate of death was 36% with everolimus and 41% with placebo, a nonsignificant difference, although longer follow-up is needed for definitive overall survival data.

The trial was sponsored by Novartis, the maker of Afinitor. Dr. O’Regan disclosed that she is a consultant to Novartis, and receives research funding from Genentech, the maker of Kadcyla and Perjeta, and Novartis.

Body

BOLERO-3 has landed in a crowded field of trastuzumab-resistant clinical trials. The unique aspect of BOLERO-3 was that it did allow for prior lapatinib use, and patients were allowed to go on to study even in the fourth-line metastatic setting.

Clinically, there was no benefit for the addition of everolimus in terms of the objective response rate or clinical benefit rate. In addition – and I think this is one of the most interesting aspects of this study – there were subgroups of patients who appeared to derive less benefit from the addition of everolimus, including the estrogen receptor–positive subgroup, patients with liver involvement, and probably most intriguing, those patients who had never seen trastuzumab in the adjuvant setting.

Is there a role for mTOR inhibition in HER2-positive metastatic breast cancer? My answer is ... yes, no, maybe so.

Yes, everolimus did improve progression-free survival. The study met its primary endpoint.

No, it had little impact on other clinically meaningful outcomes, some subgroups did not derive significant benefit, and it really is too early to see an overall survival advantage.

And finally, maybe so, because it did appear to have activity in a unique study design in patients having received prior lapatinib and relapsing having had a year of adjuvant trastuzumab, which makes this study unique.

In sum, the BOLERO-3 regimen offers a potential treatment option for patients after pertuzumab and TDM-1 therapy.

Dr. Kimberly L. Blackwell is a professor at the Duke Cancer Institute in Durham, N.C. She was the invited discussant of the study. Dr. Blackwell disclosed that she is a consultant to Genentech, the maker of Perjeta and Kadcyla, and GlaxoSmithKline; she receives research funding from Novartis, the maker of Afinitor, and Roche/Genentech.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
mTOR inhibitor, everolimus, trastuzumab resistance, HER2-positive breast cancer, breast cancer, American Society of Clinical Oncology, Afinitor, vinorelbine
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event
Body

BOLERO-3 has landed in a crowded field of trastuzumab-resistant clinical trials. The unique aspect of BOLERO-3 was that it did allow for prior lapatinib use, and patients were allowed to go on to study even in the fourth-line metastatic setting.

Clinically, there was no benefit for the addition of everolimus in terms of the objective response rate or clinical benefit rate. In addition – and I think this is one of the most interesting aspects of this study – there were subgroups of patients who appeared to derive less benefit from the addition of everolimus, including the estrogen receptor–positive subgroup, patients with liver involvement, and probably most intriguing, those patients who had never seen trastuzumab in the adjuvant setting.

Is there a role for mTOR inhibition in HER2-positive metastatic breast cancer? My answer is ... yes, no, maybe so.

Yes, everolimus did improve progression-free survival. The study met its primary endpoint.

No, it had little impact on other clinically meaningful outcomes, some subgroups did not derive significant benefit, and it really is too early to see an overall survival advantage.

And finally, maybe so, because it did appear to have activity in a unique study design in patients having received prior lapatinib and relapsing having had a year of adjuvant trastuzumab, which makes this study unique.

In sum, the BOLERO-3 regimen offers a potential treatment option for patients after pertuzumab and TDM-1 therapy.

Dr. Kimberly L. Blackwell is a professor at the Duke Cancer Institute in Durham, N.C. She was the invited discussant of the study. Dr. Blackwell disclosed that she is a consultant to Genentech, the maker of Perjeta and Kadcyla, and GlaxoSmithKline; she receives research funding from Novartis, the maker of Afinitor, and Roche/Genentech.

Body

BOLERO-3 has landed in a crowded field of trastuzumab-resistant clinical trials. The unique aspect of BOLERO-3 was that it did allow for prior lapatinib use, and patients were allowed to go on to study even in the fourth-line metastatic setting.

Clinically, there was no benefit for the addition of everolimus in terms of the objective response rate or clinical benefit rate. In addition – and I think this is one of the most interesting aspects of this study – there were subgroups of patients who appeared to derive less benefit from the addition of everolimus, including the estrogen receptor–positive subgroup, patients with liver involvement, and probably most intriguing, those patients who had never seen trastuzumab in the adjuvant setting.

Is there a role for mTOR inhibition in HER2-positive metastatic breast cancer? My answer is ... yes, no, maybe so.

Yes, everolimus did improve progression-free survival. The study met its primary endpoint.

No, it had little impact on other clinically meaningful outcomes, some subgroups did not derive significant benefit, and it really is too early to see an overall survival advantage.

And finally, maybe so, because it did appear to have activity in a unique study design in patients having received prior lapatinib and relapsing having had a year of adjuvant trastuzumab, which makes this study unique.

In sum, the BOLERO-3 regimen offers a potential treatment option for patients after pertuzumab and TDM-1 therapy.

Dr. Kimberly L. Blackwell is a professor at the Duke Cancer Institute in Durham, N.C. She was the invited discussant of the study. Dr. Blackwell disclosed that she is a consultant to Genentech, the maker of Perjeta and Kadcyla, and GlaxoSmithKline; she receives research funding from Novartis, the maker of Afinitor, and Roche/Genentech.

Title
An option after pertuzumab and TDM-1
An option after pertuzumab and TDM-1

CHICAGO – The mTOR inhibitor everolimus appears to be effective at overcoming trastuzumab resistance in patients with advanced HER2-positive breast cancer, based on findings from the BOLERO-3 trial.

The 572 patients studied all had locally advanced or metastatic HER2-positive breast cancer that had progressed despite trastuzumab therapy.

The main trial results, reported at the annual meeting of the American Society of Clinical Oncology, showed that when everolimus (Afinitor) was added to trastuzumab and vinorelbine, it reduced the risk of progression or death by 22% compared with placebo, a difference amounting to about a 1-month benefit.

As expected, patients receiving everolimus had higher rates of certain grade 3/4 adverse events, such as stomatitis, but the two groups did not differ with respect to quality of life. Overall survival results are not yet mature.

"This is the first phase III study showing the benefit of mTOR [mammalian target of rapamycin] pathway inhibition in HER2-positive breast cancer. Targeting mTOR is a viable approach to maximize the benefit of trastuzumab-based therapy," commented first author Dr. Ruth O’Regan, director of translational breast cancer research at the Winship Cancer Institute of Emory University in Atlanta. "The combination of everolimus with vinorelbine and trastuzumab can be considered an appropriate option for some patients with trastuzumab-resistant HER2-positive breast cancer."

"The ongoing BOLERO-1 trial will evaluate the addition of everolimus to chemotherapy and trastuzumab in the first-line setting," she added.

Session attendee Dr. Jose Baselga, physician-in-chief of Memorial Hospital at the Memorial Sloan-Kettering Cancer Center in New York, said, "In looking at the data, the first reaction I have is that you have two groups: the ER [estrogen receptor]-positives and the ER-negatives. And I’m just wondering whether this theme of mTOR and ER, this cross-talk, is so important, that whether even in the HER2-positives, we need to address the issue of ER blockade at the same time, because if the study had been only in the ER-negatives, this would have been a massively positive trial."

"It is possible that when you inhibit mTOR, in that situation, that perhaps we do need to also inhibit ER," Dr. O’Regan agreed. "It’s a fairly large trial, and the [progression-free survival] lines cross completely in the hormone receptor–positive group. So that’s a very good question. I think maybe it’s worthwhile looking at HER2, mTOR, estrogen receptor blockade in another trial."

Dr. Steven Vogl, a medical oncologist in the Bronx, N.Y., asked whether BOLERO-1 is powered to look at ER-positive and ER-negative patients individually. "Your benefit was small and probably confined to only the ER-negative patients. It would be nice to know how big the benefit is in those patients and to have a second trial showing us that we need to give everolimus to these patients only if they are ER negative."

Dr. O’Regan said she did not know whether BOLERO-1 was adequately powered to answer that question. "I doubt it is, in reality, but I think we will get some interesting data. If we find the same thing in that study, that will be something certainly we can take forward," she said.

Dr. Alan Astrow, of the Maimonides Medical Center in Brooklyn, N.Y., who also attended the session, noted that the new data suggest that everolimus now presents a third treatment option in this patient population, in addition to pertuzumab (Perjeta) and TDM-1 (Kadcyla). "So what are your thoughts about further development of this drug now that we have other drugs available?" he asked.

"At this point, it would probably be third-line after those agents in the metastatic setting," Dr. O’Regan replied. "The question is, where do you put it in regard to lapatinib? And are we going to have to start looking at ER-negative versus ER-positive differences – would you put this combination up higher in the ER-negative, HER2-positive group? There are a lot of questions that we just don’t know the answer to right now."

Session comoderator Dr. Rebecca Alexander Dent, an oncologist with Duke-NUS in Singapore, the University of Toronto, and the Sunnybrook Odette Cancer Center in Toronto, said, "Obviously, the doses [of everolimus] that have been used in trials are somewhat different. Can you comment on what kind of impact that might make or how we should move forward with that?"

Dr. O’Regan said the 5-mg dose used was based on phase I data, whereas other trials, such as BOLERO-1, are using a 10-mg dose. "Obviously, we don’t know if we had used a higher dose if we would have had a greater progression-free survival difference, but it would definitely increase the toxicity," she said; the optimal dose may depend on whether it is given with chemotherapy.

 

 

The patients in the BOLERO-3 trial were randomized evenly to daily everolimus versus daily placebo, each added to weekly trastuzumab (Herceptin) and vinorelbine.

Everolimus is currently approved by the Food and Drug Administration for use in combination with exemestane to treat advanced hormone receptor–positive, HER2-positive breast cancer in postmenopausal women. It is also approved for selected indications in other cancers.

A fourth of patients had previously received lapatinib (Tykerb). The large majority had received one or two prior lines of therapy for metastatic disease.

Median progression-free survival was 1.22 months longer with everolimus (7.00 vs 5.78 months; hazard ratio, 0.78; P = .0067).

The overall rate of response did not differ significantly between the everolimus and placebo groups (41% vs. 37%).

The everolimus arm had higher rates than the placebo group for grade 3/4 stomatitis (13% vs. 1%), fatigue (12% vs. 4%), and hematologic adverse events. But the groups were statistically indistinguishable with respect to the time to definitive deterioration of global health status.

The rate of death was 36% with everolimus and 41% with placebo, a nonsignificant difference, although longer follow-up is needed for definitive overall survival data.

The trial was sponsored by Novartis, the maker of Afinitor. Dr. O’Regan disclosed that she is a consultant to Novartis, and receives research funding from Genentech, the maker of Kadcyla and Perjeta, and Novartis.

CHICAGO – The mTOR inhibitor everolimus appears to be effective at overcoming trastuzumab resistance in patients with advanced HER2-positive breast cancer, based on findings from the BOLERO-3 trial.

The 572 patients studied all had locally advanced or metastatic HER2-positive breast cancer that had progressed despite trastuzumab therapy.

The main trial results, reported at the annual meeting of the American Society of Clinical Oncology, showed that when everolimus (Afinitor) was added to trastuzumab and vinorelbine, it reduced the risk of progression or death by 22% compared with placebo, a difference amounting to about a 1-month benefit.

As expected, patients receiving everolimus had higher rates of certain grade 3/4 adverse events, such as stomatitis, but the two groups did not differ with respect to quality of life. Overall survival results are not yet mature.

"This is the first phase III study showing the benefit of mTOR [mammalian target of rapamycin] pathway inhibition in HER2-positive breast cancer. Targeting mTOR is a viable approach to maximize the benefit of trastuzumab-based therapy," commented first author Dr. Ruth O’Regan, director of translational breast cancer research at the Winship Cancer Institute of Emory University in Atlanta. "The combination of everolimus with vinorelbine and trastuzumab can be considered an appropriate option for some patients with trastuzumab-resistant HER2-positive breast cancer."

"The ongoing BOLERO-1 trial will evaluate the addition of everolimus to chemotherapy and trastuzumab in the first-line setting," she added.

Session attendee Dr. Jose Baselga, physician-in-chief of Memorial Hospital at the Memorial Sloan-Kettering Cancer Center in New York, said, "In looking at the data, the first reaction I have is that you have two groups: the ER [estrogen receptor]-positives and the ER-negatives. And I’m just wondering whether this theme of mTOR and ER, this cross-talk, is so important, that whether even in the HER2-positives, we need to address the issue of ER blockade at the same time, because if the study had been only in the ER-negatives, this would have been a massively positive trial."

"It is possible that when you inhibit mTOR, in that situation, that perhaps we do need to also inhibit ER," Dr. O’Regan agreed. "It’s a fairly large trial, and the [progression-free survival] lines cross completely in the hormone receptor–positive group. So that’s a very good question. I think maybe it’s worthwhile looking at HER2, mTOR, estrogen receptor blockade in another trial."

Dr. Steven Vogl, a medical oncologist in the Bronx, N.Y., asked whether BOLERO-1 is powered to look at ER-positive and ER-negative patients individually. "Your benefit was small and probably confined to only the ER-negative patients. It would be nice to know how big the benefit is in those patients and to have a second trial showing us that we need to give everolimus to these patients only if they are ER negative."

Dr. O’Regan said she did not know whether BOLERO-1 was adequately powered to answer that question. "I doubt it is, in reality, but I think we will get some interesting data. If we find the same thing in that study, that will be something certainly we can take forward," she said.

Dr. Alan Astrow, of the Maimonides Medical Center in Brooklyn, N.Y., who also attended the session, noted that the new data suggest that everolimus now presents a third treatment option in this patient population, in addition to pertuzumab (Perjeta) and TDM-1 (Kadcyla). "So what are your thoughts about further development of this drug now that we have other drugs available?" he asked.

"At this point, it would probably be third-line after those agents in the metastatic setting," Dr. O’Regan replied. "The question is, where do you put it in regard to lapatinib? And are we going to have to start looking at ER-negative versus ER-positive differences – would you put this combination up higher in the ER-negative, HER2-positive group? There are a lot of questions that we just don’t know the answer to right now."

Session comoderator Dr. Rebecca Alexander Dent, an oncologist with Duke-NUS in Singapore, the University of Toronto, and the Sunnybrook Odette Cancer Center in Toronto, said, "Obviously, the doses [of everolimus] that have been used in trials are somewhat different. Can you comment on what kind of impact that might make or how we should move forward with that?"

Dr. O’Regan said the 5-mg dose used was based on phase I data, whereas other trials, such as BOLERO-1, are using a 10-mg dose. "Obviously, we don’t know if we had used a higher dose if we would have had a greater progression-free survival difference, but it would definitely increase the toxicity," she said; the optimal dose may depend on whether it is given with chemotherapy.

 

 

The patients in the BOLERO-3 trial were randomized evenly to daily everolimus versus daily placebo, each added to weekly trastuzumab (Herceptin) and vinorelbine.

Everolimus is currently approved by the Food and Drug Administration for use in combination with exemestane to treat advanced hormone receptor–positive, HER2-positive breast cancer in postmenopausal women. It is also approved for selected indications in other cancers.

A fourth of patients had previously received lapatinib (Tykerb). The large majority had received one or two prior lines of therapy for metastatic disease.

Median progression-free survival was 1.22 months longer with everolimus (7.00 vs 5.78 months; hazard ratio, 0.78; P = .0067).

The overall rate of response did not differ significantly between the everolimus and placebo groups (41% vs. 37%).

The everolimus arm had higher rates than the placebo group for grade 3/4 stomatitis (13% vs. 1%), fatigue (12% vs. 4%), and hematologic adverse events. But the groups were statistically indistinguishable with respect to the time to definitive deterioration of global health status.

The rate of death was 36% with everolimus and 41% with placebo, a nonsignificant difference, although longer follow-up is needed for definitive overall survival data.

The trial was sponsored by Novartis, the maker of Afinitor. Dr. O’Regan disclosed that she is a consultant to Novartis, and receives research funding from Genentech, the maker of Kadcyla and Perjeta, and Novartis.

Publications
Publications
Topics
Article Type
Display Headline
BOLERO 3: Everolimus may overcome trastuzumab resistance in HER2-positive breast cancer
Display Headline
BOLERO 3: Everolimus may overcome trastuzumab resistance in HER2-positive breast cancer
Legacy Keywords
mTOR inhibitor, everolimus, trastuzumab resistance, HER2-positive breast cancer, breast cancer, American Society of Clinical Oncology, Afinitor, vinorelbine
Legacy Keywords
mTOR inhibitor, everolimus, trastuzumab resistance, HER2-positive breast cancer, breast cancer, American Society of Clinical Oncology, Afinitor, vinorelbine
Article Source

AT THE ASCO ANNUAL MEETING 2013

PURLs Copyright

Inside the Article

Vitals

Major finding: Compared with placebo, everolimus led to better progression-free survival when added to trastuzumab and vinorelbine (hazard ratio, 0.78).

Data source: A phase III, randomized, double-blind trial among 572 patients with trastuzumab-resistant HER2-positive advanced breast cancer (BOLERO-3 trial)

Disclosures: The trial was sponsored by Novartis, the maker of Afinitor. Dr. O’Regan disclosed that she has ties to Novartis, and receives research funding from Genentech, the maker of Kadcyla and Perjeta.

PAM50 assay aids prediction of metastasis in early node-positive breast cancer

Confirmatory analysis would be welcomed
Article Type
Changed
Thu, 12/15/2022 - 18:16
Display Headline
PAM50 assay aids prediction of metastasis in early node-positive breast cancer

CHICAGO – The multigene PAM50 assay identifies postmenopausal women with node-positive early breast cancer who have a low risk of distant metastases and may be able to skip adjuvant chemotherapy.

The 543 women studied had one to three positive nodes and low- or intermediate-grade tumors; all received adjuvant endocrine therapy but no chemotherapy in a pair of randomized trials: the ABCSG-8 (Austrian Breast Cancer Study Group 8) trial and the ATAC (Arimidex, Tamoxifen, Alone or in Combination) trial.

Results showed that three PAM50 parameters – the risk of recurrence (ROR) score, ROR risk group, and intrinsic subtype – significantly added to clinicopathologic factors in predicting the 10-year risk of metastases.

Dr. Michael Gnant

"A significant proportion of early breast cancer patients with only one positive node has a limited long-term risk of metastases, as well as at least some patients with two or three positive nodes," first author Dr. Michael Gnant commented in presenting the findings at the annual meeting of the American Society of Clinical Oncology.

In particular, among patients with only one positive node, those falling into the ROR low-risk group (41% of the one-node subset) and those having the luminal A intrinsic subtype (71% of the one-node subset) had 10-year risks of metastases that were less than 9%.

"Based on their low risk of metastases, these patients could – and I would personally add should be spared the side effects of adjuvant chemotherapy despite having node-positive early breast cancer," maintained Dr. Gnant, who is a professor of surgery at the Medical University of Vienna, Austria.

"In the adjuvant treatment of hormone receptor–positive breast cancer, we every day face the dilemma of undertreatment versus overtreatment. Assessing an individual patient’s risk of metastasis as accurately as possible is an important goal in early breast cancer, and it directly impacts on treatment decisions," he added. "Still, risk assessment is mainly based on clinicopathological variables, of which nodal status is considered to be most important. As a consequence, most node-positive patients with hormone receptor–positive breast cancer globally still receive adjuvant chemotherapy."

In the study, the researchers assessed the added prognostic value derived from NanoString Technologies’ PAM50, a 50-gene assay designed to identify intrinsic subtypes of breast cancer. The subtypes are combined with a proliferation score and tumor size to yield the ROR score, and scores are used to classify patients into ROR-based risk groups.

The clinical usefulness of the PAM50 assay and other similar molecular assays, such as Genomic Health’s Oncotype DX and Agendia’s MammaPrint, has not been defined in node-positive patients, he noted.

The node-positive patients studied were drawn from the ABCSG-8 and ATAC trials. The former trial assigned women either to tamoxifen for 5 years or to 2 years of tamoxifen followed by 3 years of the aromatase inhibitor anastrozole (Arimidex); the latter assigned patients to 5 years of adjuvant anastrozole alone, tamoxifen alone, or the (subsequently discontinued) combination.

For each patient, the investigators analyzed tumor tissue to determine the PAM50 ROR score, ROR risk group, and intrinsic subtype. In addition, they derived the clinical treatment score, which is an overall integrated measure of clinicopathologic factors.

The median follow-up was 11 years, according to Dr. Gnant. As the ROR score increased from 0 to 100, so did the 10-year risk of metastases.

When added to the clinical treatment score alone, the ROR score significantly improved on the prediction of metastasis (chi-square for the difference in likelihood ratios, 32.45; P less than .0001).

The ROR risk groups also stratified patients well. The risk of metastases was about 30% in the high-risk group, 14% in the intermediate-risk group, and 8% in the low-risk group.

When added to the clinical treatment score, the luminal subtype significantly improved on prediction of metastasis (chi-square for the difference in likelihood ratios, 20.48; P less than .0001).

The estimated 10-year risk of metastasis was 11% in the luminal A group and 31% in the luminal B group.

In a combined analysis, the risk of metastases was less than 10% in several subsets of patients: those with one or two positive nodes in the ROR low-risk group (6.6% and 7.2% risks, respectively) and those with one positive node having the luminal A subtype (8.4% risk).

Finally, each factor – ROR score, ROR risk group, and intrinsic subtype – significantly improved on the prediction of risk above and beyond clinicopathologic factors in patients with one positive node, two positive nodes, and two or three positive nodes individually.

One session attendee asked, "Was there any opportunity to look at the interaction [of PAM50 parameters] with therapy?"

 

 

"Given the rather limited overall difference between anastrozole and tamoxifen both in the ATAC trial and in the sequencing study, ABCSG-8 – tamoxifen versus tamoxifen followed by anastrozole – I don’t think that we can reasonably comment on the interaction between these scores and treatment," Dr. Gnant replied.

Another attendee, Dr. Carlos Arteaga of the Vanderbilt-Ingram Cancer Center in Nashville, Tenn., asked, "Until we all start doing these genomic tests, can you tell us whether the Ki-67 had equal power in your study? It’s a simple test, and we are all using 14% as a cutoff for luminal A and B [intrinsic subtypes]. How did that pan out in this cohort?

"We are trying to have central review for Ki-67 and bring it into the multivariate model. Hopefully, later this year we will be able to answer that question," Dr. Gnant replied.

Dr. Gnant disclosed ties to several companies including Sanofi-Aventis, Novartis, and Roche, as well as NanoString Technologies, the maker of the PAM50 assay.

Click for Credit Link
Body

The ATAC population was used in this study for training and validation of PAM50, which could lead to overestimation of the test’s prognostic value. That said, the data looked pretty impressive.

Is the assay ready for prime time in node-positive patients? In my mind, probably not quite yet. The present analysis involved the testing of multiple comparisons, and in the end, even with 543 specimens it was limited by a relatively small number of events.

PAM50 classification approaches are probably useful, but which approaches are best – the ROR score, the intrinsic subtype, or the ROR group – remains uncertain, and a confirmatory analysis would certainly be welcomed.

Dr. Eric P. Winer is with the Dana-Farber Cancer Institute in Boston. He was the invited discussant of the study. Dr. Winer disclosed ties to AstraZeneca, Merrimack, Pfizer, and other companies.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
multigene PAM50, postmenopausal women, breast cancer, adjuvant chemotherapy, adjuvant endocrine therapy, risk of recurrence,
Click for Credit Link
Click for Credit Link
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event
Body

The ATAC population was used in this study for training and validation of PAM50, which could lead to overestimation of the test’s prognostic value. That said, the data looked pretty impressive.

Is the assay ready for prime time in node-positive patients? In my mind, probably not quite yet. The present analysis involved the testing of multiple comparisons, and in the end, even with 543 specimens it was limited by a relatively small number of events.

PAM50 classification approaches are probably useful, but which approaches are best – the ROR score, the intrinsic subtype, or the ROR group – remains uncertain, and a confirmatory analysis would certainly be welcomed.

Dr. Eric P. Winer is with the Dana-Farber Cancer Institute in Boston. He was the invited discussant of the study. Dr. Winer disclosed ties to AstraZeneca, Merrimack, Pfizer, and other companies.

Body

The ATAC population was used in this study for training and validation of PAM50, which could lead to overestimation of the test’s prognostic value. That said, the data looked pretty impressive.

Is the assay ready for prime time in node-positive patients? In my mind, probably not quite yet. The present analysis involved the testing of multiple comparisons, and in the end, even with 543 specimens it was limited by a relatively small number of events.

PAM50 classification approaches are probably useful, but which approaches are best – the ROR score, the intrinsic subtype, or the ROR group – remains uncertain, and a confirmatory analysis would certainly be welcomed.

Dr. Eric P. Winer is with the Dana-Farber Cancer Institute in Boston. He was the invited discussant of the study. Dr. Winer disclosed ties to AstraZeneca, Merrimack, Pfizer, and other companies.

Title
Confirmatory analysis would be welcomed
Confirmatory analysis would be welcomed

CHICAGO – The multigene PAM50 assay identifies postmenopausal women with node-positive early breast cancer who have a low risk of distant metastases and may be able to skip adjuvant chemotherapy.

The 543 women studied had one to three positive nodes and low- or intermediate-grade tumors; all received adjuvant endocrine therapy but no chemotherapy in a pair of randomized trials: the ABCSG-8 (Austrian Breast Cancer Study Group 8) trial and the ATAC (Arimidex, Tamoxifen, Alone or in Combination) trial.

Results showed that three PAM50 parameters – the risk of recurrence (ROR) score, ROR risk group, and intrinsic subtype – significantly added to clinicopathologic factors in predicting the 10-year risk of metastases.

Dr. Michael Gnant

"A significant proportion of early breast cancer patients with only one positive node has a limited long-term risk of metastases, as well as at least some patients with two or three positive nodes," first author Dr. Michael Gnant commented in presenting the findings at the annual meeting of the American Society of Clinical Oncology.

In particular, among patients with only one positive node, those falling into the ROR low-risk group (41% of the one-node subset) and those having the luminal A intrinsic subtype (71% of the one-node subset) had 10-year risks of metastases that were less than 9%.

"Based on their low risk of metastases, these patients could – and I would personally add should be spared the side effects of adjuvant chemotherapy despite having node-positive early breast cancer," maintained Dr. Gnant, who is a professor of surgery at the Medical University of Vienna, Austria.

"In the adjuvant treatment of hormone receptor–positive breast cancer, we every day face the dilemma of undertreatment versus overtreatment. Assessing an individual patient’s risk of metastasis as accurately as possible is an important goal in early breast cancer, and it directly impacts on treatment decisions," he added. "Still, risk assessment is mainly based on clinicopathological variables, of which nodal status is considered to be most important. As a consequence, most node-positive patients with hormone receptor–positive breast cancer globally still receive adjuvant chemotherapy."

In the study, the researchers assessed the added prognostic value derived from NanoString Technologies’ PAM50, a 50-gene assay designed to identify intrinsic subtypes of breast cancer. The subtypes are combined with a proliferation score and tumor size to yield the ROR score, and scores are used to classify patients into ROR-based risk groups.

The clinical usefulness of the PAM50 assay and other similar molecular assays, such as Genomic Health’s Oncotype DX and Agendia’s MammaPrint, has not been defined in node-positive patients, he noted.

The node-positive patients studied were drawn from the ABCSG-8 and ATAC trials. The former trial assigned women either to tamoxifen for 5 years or to 2 years of tamoxifen followed by 3 years of the aromatase inhibitor anastrozole (Arimidex); the latter assigned patients to 5 years of adjuvant anastrozole alone, tamoxifen alone, or the (subsequently discontinued) combination.

For each patient, the investigators analyzed tumor tissue to determine the PAM50 ROR score, ROR risk group, and intrinsic subtype. In addition, they derived the clinical treatment score, which is an overall integrated measure of clinicopathologic factors.

The median follow-up was 11 years, according to Dr. Gnant. As the ROR score increased from 0 to 100, so did the 10-year risk of metastases.

When added to the clinical treatment score alone, the ROR score significantly improved on the prediction of metastasis (chi-square for the difference in likelihood ratios, 32.45; P less than .0001).

The ROR risk groups also stratified patients well. The risk of metastases was about 30% in the high-risk group, 14% in the intermediate-risk group, and 8% in the low-risk group.

When added to the clinical treatment score, the luminal subtype significantly improved on prediction of metastasis (chi-square for the difference in likelihood ratios, 20.48; P less than .0001).

The estimated 10-year risk of metastasis was 11% in the luminal A group and 31% in the luminal B group.

In a combined analysis, the risk of metastases was less than 10% in several subsets of patients: those with one or two positive nodes in the ROR low-risk group (6.6% and 7.2% risks, respectively) and those with one positive node having the luminal A subtype (8.4% risk).

Finally, each factor – ROR score, ROR risk group, and intrinsic subtype – significantly improved on the prediction of risk above and beyond clinicopathologic factors in patients with one positive node, two positive nodes, and two or three positive nodes individually.

One session attendee asked, "Was there any opportunity to look at the interaction [of PAM50 parameters] with therapy?"

 

 

"Given the rather limited overall difference between anastrozole and tamoxifen both in the ATAC trial and in the sequencing study, ABCSG-8 – tamoxifen versus tamoxifen followed by anastrozole – I don’t think that we can reasonably comment on the interaction between these scores and treatment," Dr. Gnant replied.

Another attendee, Dr. Carlos Arteaga of the Vanderbilt-Ingram Cancer Center in Nashville, Tenn., asked, "Until we all start doing these genomic tests, can you tell us whether the Ki-67 had equal power in your study? It’s a simple test, and we are all using 14% as a cutoff for luminal A and B [intrinsic subtypes]. How did that pan out in this cohort?

"We are trying to have central review for Ki-67 and bring it into the multivariate model. Hopefully, later this year we will be able to answer that question," Dr. Gnant replied.

Dr. Gnant disclosed ties to several companies including Sanofi-Aventis, Novartis, and Roche, as well as NanoString Technologies, the maker of the PAM50 assay.

CHICAGO – The multigene PAM50 assay identifies postmenopausal women with node-positive early breast cancer who have a low risk of distant metastases and may be able to skip adjuvant chemotherapy.

The 543 women studied had one to three positive nodes and low- or intermediate-grade tumors; all received adjuvant endocrine therapy but no chemotherapy in a pair of randomized trials: the ABCSG-8 (Austrian Breast Cancer Study Group 8) trial and the ATAC (Arimidex, Tamoxifen, Alone or in Combination) trial.

Results showed that three PAM50 parameters – the risk of recurrence (ROR) score, ROR risk group, and intrinsic subtype – significantly added to clinicopathologic factors in predicting the 10-year risk of metastases.

Dr. Michael Gnant

"A significant proportion of early breast cancer patients with only one positive node has a limited long-term risk of metastases, as well as at least some patients with two or three positive nodes," first author Dr. Michael Gnant commented in presenting the findings at the annual meeting of the American Society of Clinical Oncology.

In particular, among patients with only one positive node, those falling into the ROR low-risk group (41% of the one-node subset) and those having the luminal A intrinsic subtype (71% of the one-node subset) had 10-year risks of metastases that were less than 9%.

"Based on their low risk of metastases, these patients could – and I would personally add should be spared the side effects of adjuvant chemotherapy despite having node-positive early breast cancer," maintained Dr. Gnant, who is a professor of surgery at the Medical University of Vienna, Austria.

"In the adjuvant treatment of hormone receptor–positive breast cancer, we every day face the dilemma of undertreatment versus overtreatment. Assessing an individual patient’s risk of metastasis as accurately as possible is an important goal in early breast cancer, and it directly impacts on treatment decisions," he added. "Still, risk assessment is mainly based on clinicopathological variables, of which nodal status is considered to be most important. As a consequence, most node-positive patients with hormone receptor–positive breast cancer globally still receive adjuvant chemotherapy."

In the study, the researchers assessed the added prognostic value derived from NanoString Technologies’ PAM50, a 50-gene assay designed to identify intrinsic subtypes of breast cancer. The subtypes are combined with a proliferation score and tumor size to yield the ROR score, and scores are used to classify patients into ROR-based risk groups.

The clinical usefulness of the PAM50 assay and other similar molecular assays, such as Genomic Health’s Oncotype DX and Agendia’s MammaPrint, has not been defined in node-positive patients, he noted.

The node-positive patients studied were drawn from the ABCSG-8 and ATAC trials. The former trial assigned women either to tamoxifen for 5 years or to 2 years of tamoxifen followed by 3 years of the aromatase inhibitor anastrozole (Arimidex); the latter assigned patients to 5 years of adjuvant anastrozole alone, tamoxifen alone, or the (subsequently discontinued) combination.

For each patient, the investigators analyzed tumor tissue to determine the PAM50 ROR score, ROR risk group, and intrinsic subtype. In addition, they derived the clinical treatment score, which is an overall integrated measure of clinicopathologic factors.

The median follow-up was 11 years, according to Dr. Gnant. As the ROR score increased from 0 to 100, so did the 10-year risk of metastases.

When added to the clinical treatment score alone, the ROR score significantly improved on the prediction of metastasis (chi-square for the difference in likelihood ratios, 32.45; P less than .0001).

The ROR risk groups also stratified patients well. The risk of metastases was about 30% in the high-risk group, 14% in the intermediate-risk group, and 8% in the low-risk group.

When added to the clinical treatment score, the luminal subtype significantly improved on prediction of metastasis (chi-square for the difference in likelihood ratios, 20.48; P less than .0001).

The estimated 10-year risk of metastasis was 11% in the luminal A group and 31% in the luminal B group.

In a combined analysis, the risk of metastases was less than 10% in several subsets of patients: those with one or two positive nodes in the ROR low-risk group (6.6% and 7.2% risks, respectively) and those with one positive node having the luminal A subtype (8.4% risk).

Finally, each factor – ROR score, ROR risk group, and intrinsic subtype – significantly improved on the prediction of risk above and beyond clinicopathologic factors in patients with one positive node, two positive nodes, and two or three positive nodes individually.

One session attendee asked, "Was there any opportunity to look at the interaction [of PAM50 parameters] with therapy?"

 

 

"Given the rather limited overall difference between anastrozole and tamoxifen both in the ATAC trial and in the sequencing study, ABCSG-8 – tamoxifen versus tamoxifen followed by anastrozole – I don’t think that we can reasonably comment on the interaction between these scores and treatment," Dr. Gnant replied.

Another attendee, Dr. Carlos Arteaga of the Vanderbilt-Ingram Cancer Center in Nashville, Tenn., asked, "Until we all start doing these genomic tests, can you tell us whether the Ki-67 had equal power in your study? It’s a simple test, and we are all using 14% as a cutoff for luminal A and B [intrinsic subtypes]. How did that pan out in this cohort?

"We are trying to have central review for Ki-67 and bring it into the multivariate model. Hopefully, later this year we will be able to answer that question," Dr. Gnant replied.

Dr. Gnant disclosed ties to several companies including Sanofi-Aventis, Novartis, and Roche, as well as NanoString Technologies, the maker of the PAM50 assay.

Publications
Publications
Topics
Article Type
Display Headline
PAM50 assay aids prediction of metastasis in early node-positive breast cancer
Display Headline
PAM50 assay aids prediction of metastasis in early node-positive breast cancer
Legacy Keywords
multigene PAM50, postmenopausal women, breast cancer, adjuvant chemotherapy, adjuvant endocrine therapy, risk of recurrence,
Legacy Keywords
multigene PAM50, postmenopausal women, breast cancer, adjuvant chemotherapy, adjuvant endocrine therapy, risk of recurrence,
Article Source

AT THE ASCO ANNUAL MEETING 2013

PURLs Copyright

Inside the Article

Vitals

Major finding: Among patients with one positive node, those falling into the ROR low-risk group (41% of the one-node subset) and those having the luminal A intrinsic subtype (71% of the one-node subset) had 10-year risks of metastases that were less than 9%.

Data source: A combined analysis of 543 postmenopausal patients with node-positive, hormone receptor–positive, early-stage breast cancer who received adjuvant endocrine therapy in the ABCSG-8 and ATAC trials.

Disclosures: Dr. Gnant disclosed ties to several companies including Sanofi-Aventis, Novartis, and Roche, as well as NanoString Technologies, the maker of the PAM50 assay.

Pomalidomide plus low-dose dexamethasone may be new standard for MM

Minor benefits may translate into long-term benefits
Article Type
Changed
Fri, 01/04/2019 - 12:13
Display Headline
Pomalidomide plus low-dose dexamethasone may be new standard for MM

CHICAGO – The combination of pomalidomide and low-dose dexamethasone is superior to high-dose dexamethasone monotherapy for treating patients with relapsed and refractory multiple myeloma, based on updated results from the multicenter, randomized MM-003 trial.

Among the 455 patients studied in the trial, those assigned to the combination therapy had a 52% lower risk of progression or death and a 26% lower risk of death alone when compared with peers assigned to single-agent high-dose dexamethasone.

Dr. Katja C. Weisel

The two regimens had much the same toxicity profile, although the combination was associated with a higher rate of grade 3/4 hematologic toxicity.

"Pomalidomide in combination with low-dose dexamethasone should be considered as a new standard of care for treatment of relapsed and refractory multiple myeloma patients after treatment with lenalidomide and bortezomib," presenting author Dr. Katja C. Weisel commented at the annual meeting of the American Society of Clinical Oncology.

All of the patients enrolled in the MM-003 trial had received at least two prior therapies and had disease refractory to their last therapy, according to Dr. Weisel, a hematologist-oncologist with the University Hospital Tübingen, Germany. All had experienced a failure of both Millennium’s bortezomib (Velcade) and Celgene’s lenalidomide (Revlimid).

The patients were randomized 2:1 to receive low-dose dexamethasone plus Celgene’s pomalidomide (Pomalyst), an antiangiogenic and immune-modulating agent, or high-dose dexamethasone alone. Patients given pomalidomide or who had a history of deep vein thrombosis were given thromboprophylaxis.

Patients who experienced progression on high-dose dexamethasone entered the companion MM-003C trial, in which they were given pomalidomide.

Initial trial results after a median follow-up of 4 months, which were previously reported, showed there were significantly better progression-free survival and overall survival with the combination. These results led to a recommendation by the trial’s monitoring committee that all patients in the high-dose dexamethasone group be given access to pomalidomide regardless of whether they had progression. In all, half of the patients in that group received pomalidomide after high-dose dexamethasone due to either this recommendation or entry into the companion trial.

In the updated analysis, now with a median follow-up of 10 months, progression-free survival was still significantly better with pomalidomide plus low-dose dexamethasone than with high-dose dexamethasone (4.0 vs. 1.9 months; hazard ratio, 0.48; P less than .001).

Overall survival was also still significantly better with the combination (12.7 vs. 8.1 months; HR, 0.74; P = .028).

"This overall survival benefit was maintained despite a high crossover rate, in 50% of the patients. ... In addition, all patients who were still alive at this time in the high-dose dexamethasone group had received pomalidomide as a salvage treatment," Dr. Weisel noted.

The progression-free survival and overall survival benefits were generally similar across subgroups of patients whose disease was refractory to both lenalidomide and bortezomib, who had received lenalidomide as their last prior therapy, and who had received bortezomib as their last prior therapy.

"The safety profile of pomalidomide is predictable and manageable, and the drug with its oral application is generally well tolerated in this heavily pretreated patient group," Dr. Weisel commented.

The main toxicity with the combination was hematologic toxicity: The rate of grade 3/4 neutropenia was 48% with the combination, compared with 16% with high-dose dexamethasone. The combination group and the high-dose dexamethasone group had essentially the same rates of grade 3/4 deep vein thrombosis and pulmonary embolism (1% and 0%, respectively), peripheral neuropathy (1% and 1%), and discontinuation due to adverse events (9% and 10%).

Dr. Weisel disclosed that she is a consultant to and receives honoraria from Celgene and Janssen.

Body

The median progression-free survival in patients who achieved a minimal response (MR) of 8 months was about the same as the 7 months seen in patients who achieved a partial response or better.

When you don’t have many options left, even something like an MR can carry significant clinical benefit. And the important message in my mind here is that if you don’t get a PR (partial response) or you don’t get a CR (complete response), don’t throw the therapy away. These minor benefits may actually translate into significant long-term clinical benefits, and it’s a different situation than we are discussing in the context of a newly diagnosed, treatment-naive patient, where our goal ultimately should be a CR.

Also, despite the clear survival benefit of the combination, there was a late crossing of the overall survival curves in favor of high-dose dexamethasone. If you look at the number of patients who stayed on high-dose dexamethasone, it was vanishingly small. So I think that late improvement in survival was a consequence of getting the better therapy as part of the crossover design.

Dr. Sagar Lonial, of the Winship Cancer Institute, Emory University, Atlanta, was the invited discussant of the study. Dr. Lonial disclosed that he is a consultant to and receives research funding from Bristol-Myers Squibb, Celgene, Millennium, and Novartis, and also is a consultant to Onyx.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
pomalidomide, low-dose dexamethasone, high-dose dexamethasone monotherapy, relapsed and refractory multiple myeloma, multicenter, randomized MM-003 trial, multiple myeloma, lenalidomide and bortezomib, Dr. Katja C. Weisel, annual meeting of the American Society of Clinical Oncology, MM-003 trial, bortezomib, Velcade, Celgene’s lenalidomide, Revlimid, low-dose dexamethasone plus Celgene’s pomalidomide, Pomalyst, antiangiogenic and immune-modulating agent, deep vein thrombosis,
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event
Body

The median progression-free survival in patients who achieved a minimal response (MR) of 8 months was about the same as the 7 months seen in patients who achieved a partial response or better.

When you don’t have many options left, even something like an MR can carry significant clinical benefit. And the important message in my mind here is that if you don’t get a PR (partial response) or you don’t get a CR (complete response), don’t throw the therapy away. These minor benefits may actually translate into significant long-term clinical benefits, and it’s a different situation than we are discussing in the context of a newly diagnosed, treatment-naive patient, where our goal ultimately should be a CR.

Also, despite the clear survival benefit of the combination, there was a late crossing of the overall survival curves in favor of high-dose dexamethasone. If you look at the number of patients who stayed on high-dose dexamethasone, it was vanishingly small. So I think that late improvement in survival was a consequence of getting the better therapy as part of the crossover design.

Dr. Sagar Lonial, of the Winship Cancer Institute, Emory University, Atlanta, was the invited discussant of the study. Dr. Lonial disclosed that he is a consultant to and receives research funding from Bristol-Myers Squibb, Celgene, Millennium, and Novartis, and also is a consultant to Onyx.

Body

The median progression-free survival in patients who achieved a minimal response (MR) of 8 months was about the same as the 7 months seen in patients who achieved a partial response or better.

When you don’t have many options left, even something like an MR can carry significant clinical benefit. And the important message in my mind here is that if you don’t get a PR (partial response) or you don’t get a CR (complete response), don’t throw the therapy away. These minor benefits may actually translate into significant long-term clinical benefits, and it’s a different situation than we are discussing in the context of a newly diagnosed, treatment-naive patient, where our goal ultimately should be a CR.

Also, despite the clear survival benefit of the combination, there was a late crossing of the overall survival curves in favor of high-dose dexamethasone. If you look at the number of patients who stayed on high-dose dexamethasone, it was vanishingly small. So I think that late improvement in survival was a consequence of getting the better therapy as part of the crossover design.

Dr. Sagar Lonial, of the Winship Cancer Institute, Emory University, Atlanta, was the invited discussant of the study. Dr. Lonial disclosed that he is a consultant to and receives research funding from Bristol-Myers Squibb, Celgene, Millennium, and Novartis, and also is a consultant to Onyx.

Title
Minor benefits may translate into long-term benefits
Minor benefits may translate into long-term benefits

CHICAGO – The combination of pomalidomide and low-dose dexamethasone is superior to high-dose dexamethasone monotherapy for treating patients with relapsed and refractory multiple myeloma, based on updated results from the multicenter, randomized MM-003 trial.

Among the 455 patients studied in the trial, those assigned to the combination therapy had a 52% lower risk of progression or death and a 26% lower risk of death alone when compared with peers assigned to single-agent high-dose dexamethasone.

Dr. Katja C. Weisel

The two regimens had much the same toxicity profile, although the combination was associated with a higher rate of grade 3/4 hematologic toxicity.

"Pomalidomide in combination with low-dose dexamethasone should be considered as a new standard of care for treatment of relapsed and refractory multiple myeloma patients after treatment with lenalidomide and bortezomib," presenting author Dr. Katja C. Weisel commented at the annual meeting of the American Society of Clinical Oncology.

All of the patients enrolled in the MM-003 trial had received at least two prior therapies and had disease refractory to their last therapy, according to Dr. Weisel, a hematologist-oncologist with the University Hospital Tübingen, Germany. All had experienced a failure of both Millennium’s bortezomib (Velcade) and Celgene’s lenalidomide (Revlimid).

The patients were randomized 2:1 to receive low-dose dexamethasone plus Celgene’s pomalidomide (Pomalyst), an antiangiogenic and immune-modulating agent, or high-dose dexamethasone alone. Patients given pomalidomide or who had a history of deep vein thrombosis were given thromboprophylaxis.

Patients who experienced progression on high-dose dexamethasone entered the companion MM-003C trial, in which they were given pomalidomide.

Initial trial results after a median follow-up of 4 months, which were previously reported, showed there were significantly better progression-free survival and overall survival with the combination. These results led to a recommendation by the trial’s monitoring committee that all patients in the high-dose dexamethasone group be given access to pomalidomide regardless of whether they had progression. In all, half of the patients in that group received pomalidomide after high-dose dexamethasone due to either this recommendation or entry into the companion trial.

In the updated analysis, now with a median follow-up of 10 months, progression-free survival was still significantly better with pomalidomide plus low-dose dexamethasone than with high-dose dexamethasone (4.0 vs. 1.9 months; hazard ratio, 0.48; P less than .001).

Overall survival was also still significantly better with the combination (12.7 vs. 8.1 months; HR, 0.74; P = .028).

"This overall survival benefit was maintained despite a high crossover rate, in 50% of the patients. ... In addition, all patients who were still alive at this time in the high-dose dexamethasone group had received pomalidomide as a salvage treatment," Dr. Weisel noted.

The progression-free survival and overall survival benefits were generally similar across subgroups of patients whose disease was refractory to both lenalidomide and bortezomib, who had received lenalidomide as their last prior therapy, and who had received bortezomib as their last prior therapy.

"The safety profile of pomalidomide is predictable and manageable, and the drug with its oral application is generally well tolerated in this heavily pretreated patient group," Dr. Weisel commented.

The main toxicity with the combination was hematologic toxicity: The rate of grade 3/4 neutropenia was 48% with the combination, compared with 16% with high-dose dexamethasone. The combination group and the high-dose dexamethasone group had essentially the same rates of grade 3/4 deep vein thrombosis and pulmonary embolism (1% and 0%, respectively), peripheral neuropathy (1% and 1%), and discontinuation due to adverse events (9% and 10%).

Dr. Weisel disclosed that she is a consultant to and receives honoraria from Celgene and Janssen.

CHICAGO – The combination of pomalidomide and low-dose dexamethasone is superior to high-dose dexamethasone monotherapy for treating patients with relapsed and refractory multiple myeloma, based on updated results from the multicenter, randomized MM-003 trial.

Among the 455 patients studied in the trial, those assigned to the combination therapy had a 52% lower risk of progression or death and a 26% lower risk of death alone when compared with peers assigned to single-agent high-dose dexamethasone.

Dr. Katja C. Weisel

The two regimens had much the same toxicity profile, although the combination was associated with a higher rate of grade 3/4 hematologic toxicity.

"Pomalidomide in combination with low-dose dexamethasone should be considered as a new standard of care for treatment of relapsed and refractory multiple myeloma patients after treatment with lenalidomide and bortezomib," presenting author Dr. Katja C. Weisel commented at the annual meeting of the American Society of Clinical Oncology.

All of the patients enrolled in the MM-003 trial had received at least two prior therapies and had disease refractory to their last therapy, according to Dr. Weisel, a hematologist-oncologist with the University Hospital Tübingen, Germany. All had experienced a failure of both Millennium’s bortezomib (Velcade) and Celgene’s lenalidomide (Revlimid).

The patients were randomized 2:1 to receive low-dose dexamethasone plus Celgene’s pomalidomide (Pomalyst), an antiangiogenic and immune-modulating agent, or high-dose dexamethasone alone. Patients given pomalidomide or who had a history of deep vein thrombosis were given thromboprophylaxis.

Patients who experienced progression on high-dose dexamethasone entered the companion MM-003C trial, in which they were given pomalidomide.

Initial trial results after a median follow-up of 4 months, which were previously reported, showed there were significantly better progression-free survival and overall survival with the combination. These results led to a recommendation by the trial’s monitoring committee that all patients in the high-dose dexamethasone group be given access to pomalidomide regardless of whether they had progression. In all, half of the patients in that group received pomalidomide after high-dose dexamethasone due to either this recommendation or entry into the companion trial.

In the updated analysis, now with a median follow-up of 10 months, progression-free survival was still significantly better with pomalidomide plus low-dose dexamethasone than with high-dose dexamethasone (4.0 vs. 1.9 months; hazard ratio, 0.48; P less than .001).

Overall survival was also still significantly better with the combination (12.7 vs. 8.1 months; HR, 0.74; P = .028).

"This overall survival benefit was maintained despite a high crossover rate, in 50% of the patients. ... In addition, all patients who were still alive at this time in the high-dose dexamethasone group had received pomalidomide as a salvage treatment," Dr. Weisel noted.

The progression-free survival and overall survival benefits were generally similar across subgroups of patients whose disease was refractory to both lenalidomide and bortezomib, who had received lenalidomide as their last prior therapy, and who had received bortezomib as their last prior therapy.

"The safety profile of pomalidomide is predictable and manageable, and the drug with its oral application is generally well tolerated in this heavily pretreated patient group," Dr. Weisel commented.

The main toxicity with the combination was hematologic toxicity: The rate of grade 3/4 neutropenia was 48% with the combination, compared with 16% with high-dose dexamethasone. The combination group and the high-dose dexamethasone group had essentially the same rates of grade 3/4 deep vein thrombosis and pulmonary embolism (1% and 0%, respectively), peripheral neuropathy (1% and 1%), and discontinuation due to adverse events (9% and 10%).

Dr. Weisel disclosed that she is a consultant to and receives honoraria from Celgene and Janssen.

Publications
Publications
Topics
Article Type
Display Headline
Pomalidomide plus low-dose dexamethasone may be new standard for MM
Display Headline
Pomalidomide plus low-dose dexamethasone may be new standard for MM
Legacy Keywords
pomalidomide, low-dose dexamethasone, high-dose dexamethasone monotherapy, relapsed and refractory multiple myeloma, multicenter, randomized MM-003 trial, multiple myeloma, lenalidomide and bortezomib, Dr. Katja C. Weisel, annual meeting of the American Society of Clinical Oncology, MM-003 trial, bortezomib, Velcade, Celgene’s lenalidomide, Revlimid, low-dose dexamethasone plus Celgene’s pomalidomide, Pomalyst, antiangiogenic and immune-modulating agent, deep vein thrombosis,
Legacy Keywords
pomalidomide, low-dose dexamethasone, high-dose dexamethasone monotherapy, relapsed and refractory multiple myeloma, multicenter, randomized MM-003 trial, multiple myeloma, lenalidomide and bortezomib, Dr. Katja C. Weisel, annual meeting of the American Society of Clinical Oncology, MM-003 trial, bortezomib, Velcade, Celgene’s lenalidomide, Revlimid, low-dose dexamethasone plus Celgene’s pomalidomide, Pomalyst, antiangiogenic and immune-modulating agent, deep vein thrombosis,
Article Source

AT THE ASCO ANNUAL MEETING 2013

PURLs Copyright

Inside the Article

Vitals

Major finding: Compared with high-dose dexamethasone, pomalidomide plus low-dose dexamethasone yielded better median progression-free survival (4.0 vs. 1.9 months) and overall survival (12.7 vs. 8.1 months).

Data source: A phase III, multicenter, randomized open-label trial of 455 patients with relapsed and refractory multiple myeloma (MM-003 trial).

Disclosures: Dr. Weisel disclosed that she is a consultant to and receives honoraria from Janssen and Celgene, the maker of pomalidomide.

No survival benefit for routine surveillance scans in classical Hodgkin disease

Article Type
Changed
Fri, 01/04/2019 - 09:37
Display Headline
No survival benefit for routine surveillance scans in classical Hodgkin disease

CHICAGO – Routine surveillance imaging does not improve clinical outcomes in patients with classic Hodgkin disease who are in first complete remission and it sharply increases costs, researchers reported at the annual meeting of the American Society of Clinical Oncology.

The team, led by Dr. Sai Ravi Pingali, retrospectively reviewed the charts of 241 adult patients who achieved a complete remission after first-line therapy.

In 68%, the treating physicians’ planned approach was routine surveillance imaging, which consisted of radiologic imaging with scans every few months, plus clinical exams and laboratory testing. In the other 32%, the planned approach was clinical surveillance, meaning that radiologic imaging was performed only if concerning signs or symptoms occurred.

The two groups had statistically indistinguishable overall survival, and in both groups, all patients experiencing relapse successfully achieved a second complete remission with salvage therapy.

In the routinely imaged group, scanning increased costs by nearly $20,000 per patient and by almost $600,000 per each relapse detected. In addition, patients were exposed to the associated radiation.

"We were unable to detect an overall survival benefit associated with routine surveillance imaging, although I have to acknowledge that our study was limited in power given the small number of deaths and relapses," commented Dr. Pingali, an oncologist with the Medical College of Wisconsin Affiliated Hospitals in Milwaukee.

"Relapses in both ... groups were effectively salvaged with autologous stem cell transplantation, arguing against a critical advantage of detection of asymptomatic relapse. Also, we need to keep in mind that the costs associated with routine surveillance imaging are significant, and it is also associated with potential risks, both in terms of radiation exposure and unnecessary work-up," he added.

"We do not feel that potential risks and costs without overall survival benefit or any other clinical benefit justify the practice of routine surveillance imaging in classical Hodgkin lymphoma patients who have achieved a complete remission after first-line therapy. We recommend that such patients be followed clinically," Dr. Pingali concluded.

Invited discussant Dr. Leo Gordon of Northwestern University in Chicago, agreed that accumulating data argue against routine imaging for surveillance in this context and noted that insurers will likely not continue to cover scans having no proven benefit. The data should prompt a revision of guidelines and reeducation of clinicians and patients, he said.

"For translational researchers and investigators and academics, I think we need to convince journal reviewers that a manuscript is acceptable if scans are not so frequent. And for industry trials, I think we need to discuss with the Food and Drug Administration the endpoint of progression-free survival and that those endpoints may not only be driven by scans but by more mundane parameters," he said—namely, the history and physical examination.

But session comoderator Dr. Gilles A. Salles of Hospices Civils de Lyon, Université Claude Bernard, France, expressed reservations, noting that the study did not provide information on how patients were allocated to groups and the time frame of relapse.

"It may be different whether relapses occur early, in the first year, or they occur later, and that may have some implications for the surveillance," he said. "I understand that you and many others jumped over the idea that we should immediately stop. A few people may think that we need more solid data, despite the provocative and quality data that were presented, to really make this jump in clinical practice. That’s my personal opinion."

Dr. Pingali and his team retrospectively reviewed the charts of adult patients who received a new diagnosis of classical Hodgkin lymphoma between 2000 and 2010 at three institutions, achieved complete remission after first-line therapy and had at least 2 years of follow-up.

The routine surveillance imaging and clinical surveillance groups had similar demographic and disease characteristics, Dr. Pingali reported. The former were significantly more likely to have received ABVD (doxorubicin [Adriamycin], bleomycin, vinblastine, and dacarbazine) and less likely to have received the Stanford V regimen as first-line therapy, and they were significantly less likely to have received radiation therapy.

With a median duration of follow-up of about 4 years, the groups did not differ significantly with respect to overall survival. "When we look at the 5-year time point, when typically the surveillance CT scans are discontinued, the curves are pretty much superimposable," he pointed out.

There were five deaths in the routine surveillance imaging group, one of which was from relapsed disease; the other deaths were from cancer, heart failure, hip fracture, and myelodysplastic syndrome. There were four deaths in the clinical surveillance group: two were from non-Hodgkin lymphoma and two from unknown causes while the patient was in confirmed remission.

 

 

All of the six patients in the routinely imaged group and all of the five patients in the clinically followed group experiencing a relapse achieved another complete remission with second-line therapy.

The mean number of scans received was 1.14 in the clinical surveillance group – usually the scan performed after first-line treatment to confirm remission, according to Dr. Pingali – and 4.25 in the routine surveillance imaging group. The ratio of scans to detected relapses was 18 vs. 124.

The extra charges incurred from scans using the routine surveillance imaging approach were $18,896/patient and $593,698/relapse.

"It is important to note that this does not include additional costs from the work-up of the false-positive scans and also the wages lost," he noted.

Dr. Pingali disclosed no relevant conflicts of interest. Dr. Gordon disclosed that he receives honoraria from Genentech and research funding from Millennium and Pharmacyclics. Dr. Salles disclosed serving as an advisor or consultant for Calistoga Pharmaceuticals, Celgene; Genentech, Janssen Pharmaceutica, and Roche. He has served as a speaker or a member of a speakers bureau and has received grants for clinical research from Celgene and Roche.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
surveillance imaging, Hodgkin disease, Dr. Sai Ravi Pingali, radiologic imaging,
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

CHICAGO – Routine surveillance imaging does not improve clinical outcomes in patients with classic Hodgkin disease who are in first complete remission and it sharply increases costs, researchers reported at the annual meeting of the American Society of Clinical Oncology.

The team, led by Dr. Sai Ravi Pingali, retrospectively reviewed the charts of 241 adult patients who achieved a complete remission after first-line therapy.

In 68%, the treating physicians’ planned approach was routine surveillance imaging, which consisted of radiologic imaging with scans every few months, plus clinical exams and laboratory testing. In the other 32%, the planned approach was clinical surveillance, meaning that radiologic imaging was performed only if concerning signs or symptoms occurred.

The two groups had statistically indistinguishable overall survival, and in both groups, all patients experiencing relapse successfully achieved a second complete remission with salvage therapy.

In the routinely imaged group, scanning increased costs by nearly $20,000 per patient and by almost $600,000 per each relapse detected. In addition, patients were exposed to the associated radiation.

"We were unable to detect an overall survival benefit associated with routine surveillance imaging, although I have to acknowledge that our study was limited in power given the small number of deaths and relapses," commented Dr. Pingali, an oncologist with the Medical College of Wisconsin Affiliated Hospitals in Milwaukee.

"Relapses in both ... groups were effectively salvaged with autologous stem cell transplantation, arguing against a critical advantage of detection of asymptomatic relapse. Also, we need to keep in mind that the costs associated with routine surveillance imaging are significant, and it is also associated with potential risks, both in terms of radiation exposure and unnecessary work-up," he added.

"We do not feel that potential risks and costs without overall survival benefit or any other clinical benefit justify the practice of routine surveillance imaging in classical Hodgkin lymphoma patients who have achieved a complete remission after first-line therapy. We recommend that such patients be followed clinically," Dr. Pingali concluded.

Invited discussant Dr. Leo Gordon of Northwestern University in Chicago, agreed that accumulating data argue against routine imaging for surveillance in this context and noted that insurers will likely not continue to cover scans having no proven benefit. The data should prompt a revision of guidelines and reeducation of clinicians and patients, he said.

"For translational researchers and investigators and academics, I think we need to convince journal reviewers that a manuscript is acceptable if scans are not so frequent. And for industry trials, I think we need to discuss with the Food and Drug Administration the endpoint of progression-free survival and that those endpoints may not only be driven by scans but by more mundane parameters," he said—namely, the history and physical examination.

But session comoderator Dr. Gilles A. Salles of Hospices Civils de Lyon, Université Claude Bernard, France, expressed reservations, noting that the study did not provide information on how patients were allocated to groups and the time frame of relapse.

"It may be different whether relapses occur early, in the first year, or they occur later, and that may have some implications for the surveillance," he said. "I understand that you and many others jumped over the idea that we should immediately stop. A few people may think that we need more solid data, despite the provocative and quality data that were presented, to really make this jump in clinical practice. That’s my personal opinion."

Dr. Pingali and his team retrospectively reviewed the charts of adult patients who received a new diagnosis of classical Hodgkin lymphoma between 2000 and 2010 at three institutions, achieved complete remission after first-line therapy and had at least 2 years of follow-up.

The routine surveillance imaging and clinical surveillance groups had similar demographic and disease characteristics, Dr. Pingali reported. The former were significantly more likely to have received ABVD (doxorubicin [Adriamycin], bleomycin, vinblastine, and dacarbazine) and less likely to have received the Stanford V regimen as first-line therapy, and they were significantly less likely to have received radiation therapy.

With a median duration of follow-up of about 4 years, the groups did not differ significantly with respect to overall survival. "When we look at the 5-year time point, when typically the surveillance CT scans are discontinued, the curves are pretty much superimposable," he pointed out.

There were five deaths in the routine surveillance imaging group, one of which was from relapsed disease; the other deaths were from cancer, heart failure, hip fracture, and myelodysplastic syndrome. There were four deaths in the clinical surveillance group: two were from non-Hodgkin lymphoma and two from unknown causes while the patient was in confirmed remission.

 

 

All of the six patients in the routinely imaged group and all of the five patients in the clinically followed group experiencing a relapse achieved another complete remission with second-line therapy.

The mean number of scans received was 1.14 in the clinical surveillance group – usually the scan performed after first-line treatment to confirm remission, according to Dr. Pingali – and 4.25 in the routine surveillance imaging group. The ratio of scans to detected relapses was 18 vs. 124.

The extra charges incurred from scans using the routine surveillance imaging approach were $18,896/patient and $593,698/relapse.

"It is important to note that this does not include additional costs from the work-up of the false-positive scans and also the wages lost," he noted.

Dr. Pingali disclosed no relevant conflicts of interest. Dr. Gordon disclosed that he receives honoraria from Genentech and research funding from Millennium and Pharmacyclics. Dr. Salles disclosed serving as an advisor or consultant for Calistoga Pharmaceuticals, Celgene; Genentech, Janssen Pharmaceutica, and Roche. He has served as a speaker or a member of a speakers bureau and has received grants for clinical research from Celgene and Roche.

CHICAGO – Routine surveillance imaging does not improve clinical outcomes in patients with classic Hodgkin disease who are in first complete remission and it sharply increases costs, researchers reported at the annual meeting of the American Society of Clinical Oncology.

The team, led by Dr. Sai Ravi Pingali, retrospectively reviewed the charts of 241 adult patients who achieved a complete remission after first-line therapy.

In 68%, the treating physicians’ planned approach was routine surveillance imaging, which consisted of radiologic imaging with scans every few months, plus clinical exams and laboratory testing. In the other 32%, the planned approach was clinical surveillance, meaning that radiologic imaging was performed only if concerning signs or symptoms occurred.

The two groups had statistically indistinguishable overall survival, and in both groups, all patients experiencing relapse successfully achieved a second complete remission with salvage therapy.

In the routinely imaged group, scanning increased costs by nearly $20,000 per patient and by almost $600,000 per each relapse detected. In addition, patients were exposed to the associated radiation.

"We were unable to detect an overall survival benefit associated with routine surveillance imaging, although I have to acknowledge that our study was limited in power given the small number of deaths and relapses," commented Dr. Pingali, an oncologist with the Medical College of Wisconsin Affiliated Hospitals in Milwaukee.

"Relapses in both ... groups were effectively salvaged with autologous stem cell transplantation, arguing against a critical advantage of detection of asymptomatic relapse. Also, we need to keep in mind that the costs associated with routine surveillance imaging are significant, and it is also associated with potential risks, both in terms of radiation exposure and unnecessary work-up," he added.

"We do not feel that potential risks and costs without overall survival benefit or any other clinical benefit justify the practice of routine surveillance imaging in classical Hodgkin lymphoma patients who have achieved a complete remission after first-line therapy. We recommend that such patients be followed clinically," Dr. Pingali concluded.

Invited discussant Dr. Leo Gordon of Northwestern University in Chicago, agreed that accumulating data argue against routine imaging for surveillance in this context and noted that insurers will likely not continue to cover scans having no proven benefit. The data should prompt a revision of guidelines and reeducation of clinicians and patients, he said.

"For translational researchers and investigators and academics, I think we need to convince journal reviewers that a manuscript is acceptable if scans are not so frequent. And for industry trials, I think we need to discuss with the Food and Drug Administration the endpoint of progression-free survival and that those endpoints may not only be driven by scans but by more mundane parameters," he said—namely, the history and physical examination.

But session comoderator Dr. Gilles A. Salles of Hospices Civils de Lyon, Université Claude Bernard, France, expressed reservations, noting that the study did not provide information on how patients were allocated to groups and the time frame of relapse.

"It may be different whether relapses occur early, in the first year, or they occur later, and that may have some implications for the surveillance," he said. "I understand that you and many others jumped over the idea that we should immediately stop. A few people may think that we need more solid data, despite the provocative and quality data that were presented, to really make this jump in clinical practice. That’s my personal opinion."

Dr. Pingali and his team retrospectively reviewed the charts of adult patients who received a new diagnosis of classical Hodgkin lymphoma between 2000 and 2010 at three institutions, achieved complete remission after first-line therapy and had at least 2 years of follow-up.

The routine surveillance imaging and clinical surveillance groups had similar demographic and disease characteristics, Dr. Pingali reported. The former were significantly more likely to have received ABVD (doxorubicin [Adriamycin], bleomycin, vinblastine, and dacarbazine) and less likely to have received the Stanford V regimen as first-line therapy, and they were significantly less likely to have received radiation therapy.

With a median duration of follow-up of about 4 years, the groups did not differ significantly with respect to overall survival. "When we look at the 5-year time point, when typically the surveillance CT scans are discontinued, the curves are pretty much superimposable," he pointed out.

There were five deaths in the routine surveillance imaging group, one of which was from relapsed disease; the other deaths were from cancer, heart failure, hip fracture, and myelodysplastic syndrome. There were four deaths in the clinical surveillance group: two were from non-Hodgkin lymphoma and two from unknown causes while the patient was in confirmed remission.

 

 

All of the six patients in the routinely imaged group and all of the five patients in the clinically followed group experiencing a relapse achieved another complete remission with second-line therapy.

The mean number of scans received was 1.14 in the clinical surveillance group – usually the scan performed after first-line treatment to confirm remission, according to Dr. Pingali – and 4.25 in the routine surveillance imaging group. The ratio of scans to detected relapses was 18 vs. 124.

The extra charges incurred from scans using the routine surveillance imaging approach were $18,896/patient and $593,698/relapse.

"It is important to note that this does not include additional costs from the work-up of the false-positive scans and also the wages lost," he noted.

Dr. Pingali disclosed no relevant conflicts of interest. Dr. Gordon disclosed that he receives honoraria from Genentech and research funding from Millennium and Pharmacyclics. Dr. Salles disclosed serving as an advisor or consultant for Calistoga Pharmaceuticals, Celgene; Genentech, Janssen Pharmaceutica, and Roche. He has served as a speaker or a member of a speakers bureau and has received grants for clinical research from Celgene and Roche.

Publications
Publications
Topics
Article Type
Display Headline
No survival benefit for routine surveillance scans in classical Hodgkin disease
Display Headline
No survival benefit for routine surveillance scans in classical Hodgkin disease
Legacy Keywords
surveillance imaging, Hodgkin disease, Dr. Sai Ravi Pingali, radiologic imaging,
Legacy Keywords
surveillance imaging, Hodgkin disease, Dr. Sai Ravi Pingali, radiologic imaging,
Sections
Article Source

AT THE ASCO ANNUAL MEETING 2013

PURLs Copyright

Inside the Article

Vitals

Major finding: The ratio of scans to detected relapses was 18 for the clinical surveillance group and 124 for the routine scan group. The extra charges incurred from scans using the routine surveillance imaging approach were $18,896/patient and $593,698/relapse.

Data source: A retrospective analysis of 241 patients with classical Hodgkin lymphoma in first complete remission

Disclosures: Dr. Pingali disclosed no relevant conflicts of interest. Dr. Gordon disclosed that he receives honoraria from Genentech and research funding from Millennium and Pharmacyclics. Dr. Salles disclosed serving as an advisor or consultant for Calistoga Pharmaceuticals, Celgene; Genentech, Janssen Pharmaceutica, and Roche. He has served as a speaker or a member of a speakers bureau and has received grants for clinical research from Celgene and Roche.

Obinutuzumab plus chlorambucil packs 1-2 punch against CLL in elderly

Time to rethink chlorambucil?
Article Type
Changed
Fri, 01/04/2019 - 12:13
Display Headline
Obinutuzumab plus chlorambucil packs 1-2 punch against CLL in elderly

CHICAGO – Adding an anti-CD20 antibody to chlorambucil significantly improved the progression-free survival of older patients with chronic lymphocytic leukemia and comorbidities, but it’s still unknown whether the novel agent obinutuzumab has added benefit over tried-and-true rituximab, researchers reported at the annual meeting of the American Society of Clinical Oncology.

In the phase III CLL11 study, median progression-free survival (PFS) was 23 months for patients treated with a combination of obinutuzumab (GA101) and chlorambucil (Leukeran), 15.7 months for those treated with rituximab (MabThera) and chlorambucil, and 10.9 months for chlorambucil alone. The respective PFS rates at 1 year were 84%, 63%, and 27%, reported principal investigator Dr. Valentin Goede, an oncologist at the University of Cologne, Germany.

Dr. Valentin Goede

"At this stage, my conclusion is that if an older, unfit CLL patient is put on chlorambucil treatment, then he or she in addition should receive an anti-CD20 monoclonal antibody," Dr. Goede said.

Anti-CD20 agents increased the risk of infusion reactions and neutropenias, but these side effects are manageable and should not outweigh the PFS benefits, he added.

Obinutuzumab is a type II anti-CD20 antibody with a mechanism of action that differs from that of rituximab. In preclinical models, obinutuzumab showed comparatively more direct killing action of B cells and an enhanced affinity for immunoglobulin receptors, with lower complement-dependent cytotoxicity.

The CLL11 study is a 2-stage trial asking whether chlorambucil and an anti-CD20 antibody can be superior to chlorambucil monotherapy in CLL patients with coexisting medical conditions (stage 1), and whether obinutuzumab plus chlorambucil could surpass rituximab plus chlorambucil (stage 2).

Dr. Goede reported final data from stage 1 of the trial, in which the investigators enrolled 581 adults aged 18 years and older with untreated CLL and comorbidities, defined as either a total Cumulative Illness Rating Scale score higher than 6 and/or a creatinine clearance rate lower than 70 mL/minute. An additional 190 patients are planned for stage 2 of the trial, which will be a head-to-head anti-CD20 comparison, Dr. Goede said.

The patients were randomized on a 1:2:2 ratio to either six cycles of chlorambucil 0.5 mg/kg on day 1 and day 15 of every 28-day cycle (110 patients), or to the same regimen plus either obinutuzumab 1,000 mg on days 1, 8 and 15 of the first cycle, and on day 1 of cycles two through 6 (238 patients), or to rituximab 375 mg/m2 on day 1 of cycle one, and 500 mg/m2 on day 1 of every subsequent cycle, for a total of six 28-day cycles (233 patients). Patients in the chlorambucil-only arm who experienced disease progression on treatment were allowed to cross over to the obinutuzumab combination.

The median patient age was about 73 years in each study arm and about three-fourths of patients were aged 65 years or older.

Overall response rates were 30.2% for patients assigned to chlorambucil alone, compared with 75.5% for patients assigned to obinutuzumab/chlorambucil, and 65.9% in the rituximab/chlorambucil arm. There were no complete responses (CR) in the monotherapy arm, 22.2% CR with obinutuzumab/chlorambucil, and 8.3% with rituximab/chlorambucil. Partial responses occurred in 30.2%, 53.3%, and 57.6%, respectively.

No patients on chlorambucil were found to be negative for minimal residual disease (MRD) in peripheral blood or bone marrow as assessed by a central laboratory at study end. In contrast, 31% of patients on obinutuzumab/chlorambucil and 2% of those on rituximab/chlorambucil were MRD-negative in peripheral blood; 17% and 2.8%, respectively, were MRD-negative in bone marrow.

The stratified hazard ratios for PFS with the anti-CD20 agent combinations, compared with chlorambucil monotherapy were 0.14 (P less than .0001) in the obinutuzumab/chlorambucil arm, and 0.32 (P less than .0001) in the rituximab/chlorambucil arm.

In a PFS subgroup analysis stratified by age, sex, comorbidity scores, mutational status, and other factors, both anti-CD20 agents showed a significant benefit over chlorambucil alone with the exceptions of patients with the 17p chromosomal deletion (both combination groups) and those with baseline chromosomal abnormalities (rituximab arm).

There were no significant differences in overall survival at a median follow-up of 14.2 months in the obinutuzumab group and 15.2 months in the rituximab group, but overall survival data are not yet mature, Dr. Goede noted.

Grade 3 or greater adverse events of any type occurred in 41% of patients on chlorambucil monotherapy, compared with 67% of patients on obinutuzumab, and 46% of those on rituximab.

Infusion-related reactions, neutropenia, and thrombocytopenia were more frequent with obinutuzumab than with the other two drugs, although adverse events leading to death were higher with chlorambucil monotherapy. The incidence of any new malignancy was 0.9% for patients on chlorambucil only, compared with 2.5% for obinutuzumab and 2.7% for rituximab.

 

 

The study was supported by Roche. Dr. Goede disclosed consulting to and receiving honoraria and research support from the company.

Click for Credit Link
Body

This trial was an extremely interesting, large sample size, multinational planned and executed trial.

Patients with comorbidities, the population selected for this study, are the people with chronic lymphocytic leukemia who need to be addressed. They form the largest majority of the CLL population. This study confirms that chlorambucil, which we in the United States have virtually thrown out of the window as totally ineffective, may not be so if you use it in combination with an anti-CD20 monoclonal antibody. These are excellent results that we have to keep in mind.

Dr. Kanti Rai is chief of hematology-oncology at Long Island Jewish Medical Center in New Hyde Park, N.Y. He was the invited discussant of the study and disclosed serving as a consultant or advisor to Celgene, Genentech, and Teva and receiving honoraria from Celgene, Cephalon, and Genentech.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
anti-CD20 antibody, chlorambucil, chronic lymphocytic leukemia, obinutuzumab, rituximab, Leukeran, MabThera, Dr. Valentin Goede,
Click for Credit Link
Click for Credit Link
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event
Body

This trial was an extremely interesting, large sample size, multinational planned and executed trial.

Patients with comorbidities, the population selected for this study, are the people with chronic lymphocytic leukemia who need to be addressed. They form the largest majority of the CLL population. This study confirms that chlorambucil, which we in the United States have virtually thrown out of the window as totally ineffective, may not be so if you use it in combination with an anti-CD20 monoclonal antibody. These are excellent results that we have to keep in mind.

Dr. Kanti Rai is chief of hematology-oncology at Long Island Jewish Medical Center in New Hyde Park, N.Y. He was the invited discussant of the study and disclosed serving as a consultant or advisor to Celgene, Genentech, and Teva and receiving honoraria from Celgene, Cephalon, and Genentech.

Body

This trial was an extremely interesting, large sample size, multinational planned and executed trial.

Patients with comorbidities, the population selected for this study, are the people with chronic lymphocytic leukemia who need to be addressed. They form the largest majority of the CLL population. This study confirms that chlorambucil, which we in the United States have virtually thrown out of the window as totally ineffective, may not be so if you use it in combination with an anti-CD20 monoclonal antibody. These are excellent results that we have to keep in mind.

Dr. Kanti Rai is chief of hematology-oncology at Long Island Jewish Medical Center in New Hyde Park, N.Y. He was the invited discussant of the study and disclosed serving as a consultant or advisor to Celgene, Genentech, and Teva and receiving honoraria from Celgene, Cephalon, and Genentech.

Title
Time to rethink chlorambucil?
Time to rethink chlorambucil?

CHICAGO – Adding an anti-CD20 antibody to chlorambucil significantly improved the progression-free survival of older patients with chronic lymphocytic leukemia and comorbidities, but it’s still unknown whether the novel agent obinutuzumab has added benefit over tried-and-true rituximab, researchers reported at the annual meeting of the American Society of Clinical Oncology.

In the phase III CLL11 study, median progression-free survival (PFS) was 23 months for patients treated with a combination of obinutuzumab (GA101) and chlorambucil (Leukeran), 15.7 months for those treated with rituximab (MabThera) and chlorambucil, and 10.9 months for chlorambucil alone. The respective PFS rates at 1 year were 84%, 63%, and 27%, reported principal investigator Dr. Valentin Goede, an oncologist at the University of Cologne, Germany.

Dr. Valentin Goede

"At this stage, my conclusion is that if an older, unfit CLL patient is put on chlorambucil treatment, then he or she in addition should receive an anti-CD20 monoclonal antibody," Dr. Goede said.

Anti-CD20 agents increased the risk of infusion reactions and neutropenias, but these side effects are manageable and should not outweigh the PFS benefits, he added.

Obinutuzumab is a type II anti-CD20 antibody with a mechanism of action that differs from that of rituximab. In preclinical models, obinutuzumab showed comparatively more direct killing action of B cells and an enhanced affinity for immunoglobulin receptors, with lower complement-dependent cytotoxicity.

The CLL11 study is a 2-stage trial asking whether chlorambucil and an anti-CD20 antibody can be superior to chlorambucil monotherapy in CLL patients with coexisting medical conditions (stage 1), and whether obinutuzumab plus chlorambucil could surpass rituximab plus chlorambucil (stage 2).

Dr. Goede reported final data from stage 1 of the trial, in which the investigators enrolled 581 adults aged 18 years and older with untreated CLL and comorbidities, defined as either a total Cumulative Illness Rating Scale score higher than 6 and/or a creatinine clearance rate lower than 70 mL/minute. An additional 190 patients are planned for stage 2 of the trial, which will be a head-to-head anti-CD20 comparison, Dr. Goede said.

The patients were randomized on a 1:2:2 ratio to either six cycles of chlorambucil 0.5 mg/kg on day 1 and day 15 of every 28-day cycle (110 patients), or to the same regimen plus either obinutuzumab 1,000 mg on days 1, 8 and 15 of the first cycle, and on day 1 of cycles two through 6 (238 patients), or to rituximab 375 mg/m2 on day 1 of cycle one, and 500 mg/m2 on day 1 of every subsequent cycle, for a total of six 28-day cycles (233 patients). Patients in the chlorambucil-only arm who experienced disease progression on treatment were allowed to cross over to the obinutuzumab combination.

The median patient age was about 73 years in each study arm and about three-fourths of patients were aged 65 years or older.

Overall response rates were 30.2% for patients assigned to chlorambucil alone, compared with 75.5% for patients assigned to obinutuzumab/chlorambucil, and 65.9% in the rituximab/chlorambucil arm. There were no complete responses (CR) in the monotherapy arm, 22.2% CR with obinutuzumab/chlorambucil, and 8.3% with rituximab/chlorambucil. Partial responses occurred in 30.2%, 53.3%, and 57.6%, respectively.

No patients on chlorambucil were found to be negative for minimal residual disease (MRD) in peripheral blood or bone marrow as assessed by a central laboratory at study end. In contrast, 31% of patients on obinutuzumab/chlorambucil and 2% of those on rituximab/chlorambucil were MRD-negative in peripheral blood; 17% and 2.8%, respectively, were MRD-negative in bone marrow.

The stratified hazard ratios for PFS with the anti-CD20 agent combinations, compared with chlorambucil monotherapy were 0.14 (P less than .0001) in the obinutuzumab/chlorambucil arm, and 0.32 (P less than .0001) in the rituximab/chlorambucil arm.

In a PFS subgroup analysis stratified by age, sex, comorbidity scores, mutational status, and other factors, both anti-CD20 agents showed a significant benefit over chlorambucil alone with the exceptions of patients with the 17p chromosomal deletion (both combination groups) and those with baseline chromosomal abnormalities (rituximab arm).

There were no significant differences in overall survival at a median follow-up of 14.2 months in the obinutuzumab group and 15.2 months in the rituximab group, but overall survival data are not yet mature, Dr. Goede noted.

Grade 3 or greater adverse events of any type occurred in 41% of patients on chlorambucil monotherapy, compared with 67% of patients on obinutuzumab, and 46% of those on rituximab.

Infusion-related reactions, neutropenia, and thrombocytopenia were more frequent with obinutuzumab than with the other two drugs, although adverse events leading to death were higher with chlorambucil monotherapy. The incidence of any new malignancy was 0.9% for patients on chlorambucil only, compared with 2.5% for obinutuzumab and 2.7% for rituximab.

 

 

The study was supported by Roche. Dr. Goede disclosed consulting to and receiving honoraria and research support from the company.

CHICAGO – Adding an anti-CD20 antibody to chlorambucil significantly improved the progression-free survival of older patients with chronic lymphocytic leukemia and comorbidities, but it’s still unknown whether the novel agent obinutuzumab has added benefit over tried-and-true rituximab, researchers reported at the annual meeting of the American Society of Clinical Oncology.

In the phase III CLL11 study, median progression-free survival (PFS) was 23 months for patients treated with a combination of obinutuzumab (GA101) and chlorambucil (Leukeran), 15.7 months for those treated with rituximab (MabThera) and chlorambucil, and 10.9 months for chlorambucil alone. The respective PFS rates at 1 year were 84%, 63%, and 27%, reported principal investigator Dr. Valentin Goede, an oncologist at the University of Cologne, Germany.

Dr. Valentin Goede

"At this stage, my conclusion is that if an older, unfit CLL patient is put on chlorambucil treatment, then he or she in addition should receive an anti-CD20 monoclonal antibody," Dr. Goede said.

Anti-CD20 agents increased the risk of infusion reactions and neutropenias, but these side effects are manageable and should not outweigh the PFS benefits, he added.

Obinutuzumab is a type II anti-CD20 antibody with a mechanism of action that differs from that of rituximab. In preclinical models, obinutuzumab showed comparatively more direct killing action of B cells and an enhanced affinity for immunoglobulin receptors, with lower complement-dependent cytotoxicity.

The CLL11 study is a 2-stage trial asking whether chlorambucil and an anti-CD20 antibody can be superior to chlorambucil monotherapy in CLL patients with coexisting medical conditions (stage 1), and whether obinutuzumab plus chlorambucil could surpass rituximab plus chlorambucil (stage 2).

Dr. Goede reported final data from stage 1 of the trial, in which the investigators enrolled 581 adults aged 18 years and older with untreated CLL and comorbidities, defined as either a total Cumulative Illness Rating Scale score higher than 6 and/or a creatinine clearance rate lower than 70 mL/minute. An additional 190 patients are planned for stage 2 of the trial, which will be a head-to-head anti-CD20 comparison, Dr. Goede said.

The patients were randomized on a 1:2:2 ratio to either six cycles of chlorambucil 0.5 mg/kg on day 1 and day 15 of every 28-day cycle (110 patients), or to the same regimen plus either obinutuzumab 1,000 mg on days 1, 8 and 15 of the first cycle, and on day 1 of cycles two through 6 (238 patients), or to rituximab 375 mg/m2 on day 1 of cycle one, and 500 mg/m2 on day 1 of every subsequent cycle, for a total of six 28-day cycles (233 patients). Patients in the chlorambucil-only arm who experienced disease progression on treatment were allowed to cross over to the obinutuzumab combination.

The median patient age was about 73 years in each study arm and about three-fourths of patients were aged 65 years or older.

Overall response rates were 30.2% for patients assigned to chlorambucil alone, compared with 75.5% for patients assigned to obinutuzumab/chlorambucil, and 65.9% in the rituximab/chlorambucil arm. There were no complete responses (CR) in the monotherapy arm, 22.2% CR with obinutuzumab/chlorambucil, and 8.3% with rituximab/chlorambucil. Partial responses occurred in 30.2%, 53.3%, and 57.6%, respectively.

No patients on chlorambucil were found to be negative for minimal residual disease (MRD) in peripheral blood or bone marrow as assessed by a central laboratory at study end. In contrast, 31% of patients on obinutuzumab/chlorambucil and 2% of those on rituximab/chlorambucil were MRD-negative in peripheral blood; 17% and 2.8%, respectively, were MRD-negative in bone marrow.

The stratified hazard ratios for PFS with the anti-CD20 agent combinations, compared with chlorambucil monotherapy were 0.14 (P less than .0001) in the obinutuzumab/chlorambucil arm, and 0.32 (P less than .0001) in the rituximab/chlorambucil arm.

In a PFS subgroup analysis stratified by age, sex, comorbidity scores, mutational status, and other factors, both anti-CD20 agents showed a significant benefit over chlorambucil alone with the exceptions of patients with the 17p chromosomal deletion (both combination groups) and those with baseline chromosomal abnormalities (rituximab arm).

There were no significant differences in overall survival at a median follow-up of 14.2 months in the obinutuzumab group and 15.2 months in the rituximab group, but overall survival data are not yet mature, Dr. Goede noted.

Grade 3 or greater adverse events of any type occurred in 41% of patients on chlorambucil monotherapy, compared with 67% of patients on obinutuzumab, and 46% of those on rituximab.

Infusion-related reactions, neutropenia, and thrombocytopenia were more frequent with obinutuzumab than with the other two drugs, although adverse events leading to death were higher with chlorambucil monotherapy. The incidence of any new malignancy was 0.9% for patients on chlorambucil only, compared with 2.5% for obinutuzumab and 2.7% for rituximab.

 

 

The study was supported by Roche. Dr. Goede disclosed consulting to and receiving honoraria and research support from the company.

Publications
Publications
Topics
Article Type
Display Headline
Obinutuzumab plus chlorambucil packs 1-2 punch against CLL in elderly
Display Headline
Obinutuzumab plus chlorambucil packs 1-2 punch against CLL in elderly
Legacy Keywords
anti-CD20 antibody, chlorambucil, chronic lymphocytic leukemia, obinutuzumab, rituximab, Leukeran, MabThera, Dr. Valentin Goede,
Legacy Keywords
anti-CD20 antibody, chlorambucil, chronic lymphocytic leukemia, obinutuzumab, rituximab, Leukeran, MabThera, Dr. Valentin Goede,
Article Source

AT THE ASCO ANNUAL MEETING 2013

PURLs Copyright

Inside the Article

Vitals

Major finding: One-year progression-free survival in elderly patients with chronic lymphocytic leukemia and comorbidities was 84% with obinutuzumab and chlorambucil and 27% with chlorambucil alone.

Data source: Randomized, controlled, two-stage, phase III trial with a planned enrollment of 780 patients.

Disclosures: The study was supported by Roche. Dr. Goede disclosed consulting to and receiving honoraria and research support from the company. Dr. Rai disclosed serving as a consultant or advisor to Celgene, Genentech, and Teva and receiving honoraria from Celgene, Cephalon, and Genentech.

Nintedanib and docetaxel provide small PFS advantage in NSCLC

Data lacking for 14% of study group
Article Type
Changed
Fri, 01/04/2019 - 12:13
Display Headline
Nintedanib and docetaxel provide small PFS advantage in NSCLC

CHICAGO – A combination of docetaxel and nintedanib, a novel inhibitor of multiple tumor growth factors, was associated with a small but significant improvement in progression-free survival in patients with advanced non–small cell lung cancer as compared with docetaxel alone.

In the phase III LUME-Lung I trial in patients with previously treated stage IIIB/IV or recurrent non–-small cell lung cancer (NSCLC), median progression-free survival (PFS) was 3.4 months for patients randomized to nintedanib and docetaxel, compared with 2.7 months for patients assigned to placebo and docetaxel (hazard ratio, 0.79; P = .0019), reported Dr. Martin Reck of the department of thoracic oncology at Hospital Grosshansdorf, Germany.

In addition, "we have seen a significant improvement of overall survival in patients with adenocarcinoma," Dr. Reck said at the annual meeting of the American Society of Clinical Oncology.

However, when patients with squamous cell NSCLC were included in the analysis, there was no significant difference in overall survival between the treatment groups, he said.

Nintedanib is an oral kinase inhibitor that targets vascular endothelial growth factor receptors 1-3, fibroblast growth factor receptors 1-3, platelet-derived growth factor receptors alpha and beta, and the RET tyrosine kinase.

In clinical trials, nintedanib has been delivered safely in combination with various chemotherapy agents, including docetaxel, paclitaxel, platinum comp2ounds, and gemcitabine, and as a single agent had clinical activity in a phase II trial against recurrent NSCLC.

Dr. Reck and his colleagues enrolled 1,314 patients with stage IIIB or IV or recurrent NSCLC after first-line chemotherapy and randomly assigned them in a double-blinded fashion to receive intravenous docetaxel 75 mg/m2 on day 1 of a 21-day cycle plus either oral nintedanib 200 mg or placebo twice daily on days 2 through 21 of each cycle. The number of potential docetaxel cycles was unlimited, and docetaxel monotherapy could continue after four cycles of the combination at the investigator’s discretion.

Patients were stratified by performance status, prior bevacizumab (Avastin) use, squamous or nonsquamous histology, and presence or absence of brain metastases.

As of the February 2013 data cutoff, 6 of the 652 patients assigned to the combination and 6 of the 655 assigned to docetaxel/placebo remained on study.

In the intention-to-treat analysis, the trial met its primary endpoint of a centrally reviewed PFS advantage for the combination.

PFS was significantly better both in patients with adenocarcinoma histologies treated with the combination (median 4.0 vs. 2.8 months; HR, 0.77; P = .0193) and with squamous cell carcinomas (2.9 vs. 2.6 months; HR, 0.77; P = .02).

Overall survival did not differ, at a median of 10.1 vs. 9.1 month. Among patients with adenocarcinomas, however, survival was significantly better, at a median of 12.6 vs. 10.3 months, respectively (HR, 0.82; P = .0359). There was also a hint of benefit in patients with adenocarcinomas refractory to prior chemotherapy with a platinum compound (median overall survival, 9.8 vs. 6.3 months; HR, 0.62; P = .0246), although this subanalysis included only 107 patients.

The combination was no better than docetaxel and placebo among patients with squamous cell cancers.

Grade 3 or greater adverse events occurred in 71% of patients on nintedanib/docetaxel, and 64% of controls. In each group, about 22% of patients had adverse events leading to discontinuation of therapy. The most common adverse events were bleeding, thromboembolic events, and hypertension, each of which occurred more frequently with the combination than with docetaxel alone.

The LUME-Lung 1 trial was sponsored by Boehringer Ingelheim. Dr. Reck disclosed serving as an advisor and lecturer for Hoffman-La Roche, Lilly, AstraZeneca, Pfizer, Bristol-Myers Squibb, and Daiichi-Sankyo.

Body

The progression-free survival analysis was performed in 1,134 (86%) of the 1,314 patients randomized, because the PFS data cutoff occurred 3 months before the actual completion of trial accrual. Also, there was a 2-year gap between the final PFS analysis and the overall survival analysis.

Although the study was reported as being positive, the lack of PFS data on 14% of the population precluded a full intention-to-treat analysis. It would be interesting to see whether the survival benefit with nintedanib in patients with adenocarcinomas would remain if this subgroup of patients were further stratified by prior bevacizumab exposure. Further, the apparent advantage for the combination in patients with platinum-refractory adenocarcinomas needs to be validated in larger studies.

Dr. Benjamin Besse is a thoracic oncologist at the Institut Gustave-Roussy in Villejuif, France. Dr. Besse, who was the invited discussant of the study, reported serving as an uncompensated advisor to Boehringer-Ingelheim.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
docetaxel, nintedanib, multiple tumor growth, non small cell lung cancer, progression-free survival, Dr. Martin Reck
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event
Body

The progression-free survival analysis was performed in 1,134 (86%) of the 1,314 patients randomized, because the PFS data cutoff occurred 3 months before the actual completion of trial accrual. Also, there was a 2-year gap between the final PFS analysis and the overall survival analysis.

Although the study was reported as being positive, the lack of PFS data on 14% of the population precluded a full intention-to-treat analysis. It would be interesting to see whether the survival benefit with nintedanib in patients with adenocarcinomas would remain if this subgroup of patients were further stratified by prior bevacizumab exposure. Further, the apparent advantage for the combination in patients with platinum-refractory adenocarcinomas needs to be validated in larger studies.

Dr. Benjamin Besse is a thoracic oncologist at the Institut Gustave-Roussy in Villejuif, France. Dr. Besse, who was the invited discussant of the study, reported serving as an uncompensated advisor to Boehringer-Ingelheim.

Body

The progression-free survival analysis was performed in 1,134 (86%) of the 1,314 patients randomized, because the PFS data cutoff occurred 3 months before the actual completion of trial accrual. Also, there was a 2-year gap between the final PFS analysis and the overall survival analysis.

Although the study was reported as being positive, the lack of PFS data on 14% of the population precluded a full intention-to-treat analysis. It would be interesting to see whether the survival benefit with nintedanib in patients with adenocarcinomas would remain if this subgroup of patients were further stratified by prior bevacizumab exposure. Further, the apparent advantage for the combination in patients with platinum-refractory adenocarcinomas needs to be validated in larger studies.

Dr. Benjamin Besse is a thoracic oncologist at the Institut Gustave-Roussy in Villejuif, France. Dr. Besse, who was the invited discussant of the study, reported serving as an uncompensated advisor to Boehringer-Ingelheim.

Title
Data lacking for 14% of study group
Data lacking for 14% of study group

CHICAGO – A combination of docetaxel and nintedanib, a novel inhibitor of multiple tumor growth factors, was associated with a small but significant improvement in progression-free survival in patients with advanced non–small cell lung cancer as compared with docetaxel alone.

In the phase III LUME-Lung I trial in patients with previously treated stage IIIB/IV or recurrent non–-small cell lung cancer (NSCLC), median progression-free survival (PFS) was 3.4 months for patients randomized to nintedanib and docetaxel, compared with 2.7 months for patients assigned to placebo and docetaxel (hazard ratio, 0.79; P = .0019), reported Dr. Martin Reck of the department of thoracic oncology at Hospital Grosshansdorf, Germany.

In addition, "we have seen a significant improvement of overall survival in patients with adenocarcinoma," Dr. Reck said at the annual meeting of the American Society of Clinical Oncology.

However, when patients with squamous cell NSCLC were included in the analysis, there was no significant difference in overall survival between the treatment groups, he said.

Nintedanib is an oral kinase inhibitor that targets vascular endothelial growth factor receptors 1-3, fibroblast growth factor receptors 1-3, platelet-derived growth factor receptors alpha and beta, and the RET tyrosine kinase.

In clinical trials, nintedanib has been delivered safely in combination with various chemotherapy agents, including docetaxel, paclitaxel, platinum comp2ounds, and gemcitabine, and as a single agent had clinical activity in a phase II trial against recurrent NSCLC.

Dr. Reck and his colleagues enrolled 1,314 patients with stage IIIB or IV or recurrent NSCLC after first-line chemotherapy and randomly assigned them in a double-blinded fashion to receive intravenous docetaxel 75 mg/m2 on day 1 of a 21-day cycle plus either oral nintedanib 200 mg or placebo twice daily on days 2 through 21 of each cycle. The number of potential docetaxel cycles was unlimited, and docetaxel monotherapy could continue after four cycles of the combination at the investigator’s discretion.

Patients were stratified by performance status, prior bevacizumab (Avastin) use, squamous or nonsquamous histology, and presence or absence of brain metastases.

As of the February 2013 data cutoff, 6 of the 652 patients assigned to the combination and 6 of the 655 assigned to docetaxel/placebo remained on study.

In the intention-to-treat analysis, the trial met its primary endpoint of a centrally reviewed PFS advantage for the combination.

PFS was significantly better both in patients with adenocarcinoma histologies treated with the combination (median 4.0 vs. 2.8 months; HR, 0.77; P = .0193) and with squamous cell carcinomas (2.9 vs. 2.6 months; HR, 0.77; P = .02).

Overall survival did not differ, at a median of 10.1 vs. 9.1 month. Among patients with adenocarcinomas, however, survival was significantly better, at a median of 12.6 vs. 10.3 months, respectively (HR, 0.82; P = .0359). There was also a hint of benefit in patients with adenocarcinomas refractory to prior chemotherapy with a platinum compound (median overall survival, 9.8 vs. 6.3 months; HR, 0.62; P = .0246), although this subanalysis included only 107 patients.

The combination was no better than docetaxel and placebo among patients with squamous cell cancers.

Grade 3 or greater adverse events occurred in 71% of patients on nintedanib/docetaxel, and 64% of controls. In each group, about 22% of patients had adverse events leading to discontinuation of therapy. The most common adverse events were bleeding, thromboembolic events, and hypertension, each of which occurred more frequently with the combination than with docetaxel alone.

The LUME-Lung 1 trial was sponsored by Boehringer Ingelheim. Dr. Reck disclosed serving as an advisor and lecturer for Hoffman-La Roche, Lilly, AstraZeneca, Pfizer, Bristol-Myers Squibb, and Daiichi-Sankyo.

CHICAGO – A combination of docetaxel and nintedanib, a novel inhibitor of multiple tumor growth factors, was associated with a small but significant improvement in progression-free survival in patients with advanced non–small cell lung cancer as compared with docetaxel alone.

In the phase III LUME-Lung I trial in patients with previously treated stage IIIB/IV or recurrent non–-small cell lung cancer (NSCLC), median progression-free survival (PFS) was 3.4 months for patients randomized to nintedanib and docetaxel, compared with 2.7 months for patients assigned to placebo and docetaxel (hazard ratio, 0.79; P = .0019), reported Dr. Martin Reck of the department of thoracic oncology at Hospital Grosshansdorf, Germany.

In addition, "we have seen a significant improvement of overall survival in patients with adenocarcinoma," Dr. Reck said at the annual meeting of the American Society of Clinical Oncology.

However, when patients with squamous cell NSCLC were included in the analysis, there was no significant difference in overall survival between the treatment groups, he said.

Nintedanib is an oral kinase inhibitor that targets vascular endothelial growth factor receptors 1-3, fibroblast growth factor receptors 1-3, platelet-derived growth factor receptors alpha and beta, and the RET tyrosine kinase.

In clinical trials, nintedanib has been delivered safely in combination with various chemotherapy agents, including docetaxel, paclitaxel, platinum comp2ounds, and gemcitabine, and as a single agent had clinical activity in a phase II trial against recurrent NSCLC.

Dr. Reck and his colleagues enrolled 1,314 patients with stage IIIB or IV or recurrent NSCLC after first-line chemotherapy and randomly assigned them in a double-blinded fashion to receive intravenous docetaxel 75 mg/m2 on day 1 of a 21-day cycle plus either oral nintedanib 200 mg or placebo twice daily on days 2 through 21 of each cycle. The number of potential docetaxel cycles was unlimited, and docetaxel monotherapy could continue after four cycles of the combination at the investigator’s discretion.

Patients were stratified by performance status, prior bevacizumab (Avastin) use, squamous or nonsquamous histology, and presence or absence of brain metastases.

As of the February 2013 data cutoff, 6 of the 652 patients assigned to the combination and 6 of the 655 assigned to docetaxel/placebo remained on study.

In the intention-to-treat analysis, the trial met its primary endpoint of a centrally reviewed PFS advantage for the combination.

PFS was significantly better both in patients with adenocarcinoma histologies treated with the combination (median 4.0 vs. 2.8 months; HR, 0.77; P = .0193) and with squamous cell carcinomas (2.9 vs. 2.6 months; HR, 0.77; P = .02).

Overall survival did not differ, at a median of 10.1 vs. 9.1 month. Among patients with adenocarcinomas, however, survival was significantly better, at a median of 12.6 vs. 10.3 months, respectively (HR, 0.82; P = .0359). There was also a hint of benefit in patients with adenocarcinomas refractory to prior chemotherapy with a platinum compound (median overall survival, 9.8 vs. 6.3 months; HR, 0.62; P = .0246), although this subanalysis included only 107 patients.

The combination was no better than docetaxel and placebo among patients with squamous cell cancers.

Grade 3 or greater adverse events occurred in 71% of patients on nintedanib/docetaxel, and 64% of controls. In each group, about 22% of patients had adverse events leading to discontinuation of therapy. The most common adverse events were bleeding, thromboembolic events, and hypertension, each of which occurred more frequently with the combination than with docetaxel alone.

The LUME-Lung 1 trial was sponsored by Boehringer Ingelheim. Dr. Reck disclosed serving as an advisor and lecturer for Hoffman-La Roche, Lilly, AstraZeneca, Pfizer, Bristol-Myers Squibb, and Daiichi-Sankyo.

Publications
Publications
Topics
Article Type
Display Headline
Nintedanib and docetaxel provide small PFS advantage in NSCLC
Display Headline
Nintedanib and docetaxel provide small PFS advantage in NSCLC
Legacy Keywords
docetaxel, nintedanib, multiple tumor growth, non small cell lung cancer, progression-free survival, Dr. Martin Reck
Legacy Keywords
docetaxel, nintedanib, multiple tumor growth, non small cell lung cancer, progression-free survival, Dr. Martin Reck
Article Source

AT THE ASCO ANNUAL MEETING 2013

PURLs Copyright

Inside the Article

Vitals

Major finding: Median progression-free survival was 3.4 months for patients randomized to nintedanib and docetaxel, compared with 2.7 months for patients assigned to placebo and docetaxel (P = .019)

Data source: Randomized, placebo controlled trial in 1,314 patients, 1,134 of whom were included in the primary endpoint analysis.

Disclosures: The LUME-Lung 1 trial was sponsored by Boehringer Ingelheim. Dr. Reck disclosed serving as an advisor and lecturer for Hoffman-La Roche, Lilly, AstraZeneca, Pfizer, Bristol-Myers Squibb, and Daiichi-Sankyo. Dr. Besse reported serving as an uncompensated advisor to Boehringer Ingelheim.

Good survival rates with temozolomide CRT for high-risk, low-grade gliomas

Article Type
Changed
Mon, 01/07/2019 - 11:43
Display Headline
Good survival rates with temozolomide CRT for high-risk, low-grade gliomas

CHICAGO – Patients with high-risk, low-grade gliomas had 3-year survival rates significantly higher than those of historical controls when they were treated with a temozolomide-based chemoradiotherapy regimen, investigators reported at the annual meeting of the American Society of Clinical Oncology.

Preliminary results from the phase II RTOG 0424 trial showed a 73% overall survival rate for patients treated with temozolomide chemoradiotherapy, compared with 54% overall survival for historical controls (P less than .001), and exceeding the study’s hypothesized 65% overall survival, reported Dr. Barbara Fisher, a radiation oncologist at the University of Western Ontario in London.

Dr. Barbara Fisher

At a median follow-up of 4 years, with a minimum potential follow-up of 3 years, median survival of patients has not been reached, and the median follow-up time for surviving patients is 5 years, Dr. Fisher noted.

Median progression-free survival was 4.5 years, and 3-year PFS was 59%. In contrast to historical controls, patients with four or five risk factors appeared to have an overall survival rate similar to that of patients with three risk factors, Dr. Fisher reported.

Although the trial was originally proposed in 2000 as a randomized controlled study, "there wasn’t much information about temozolomide and radiation at that point," Dr. Fisher said.

The control population comes from a 2002 study by Francesco Pignatti and his colleagues (J. Clin. Oncol. 2002;20:2076-84), which identified prognostic factors for survival in adults with low-grade glioma, based on data from two European Organization for Research and Treatment of Cancer (EORTC) trials conducted in the 1990s.

The risk factors are age 40 years and older; astrocytoma subtype; tumor crossing the midline; largest tumor diameter more than 6 cm preoperatively; and preoperative neurological function status (neurocognitive function greater than 1: moderate impairment).

A total of 136 patients were accrued, and 129 patients were eligible for the protocol, which consisted of temozolomide 75 mg/m2 per day for 6 weeks with concurrent conformal radiotherapy, consisting of 54 Gy divided into 30 fractions delivered 5 days each week for 6 weeks, followed by temozolomide 150-200 mg/m2 per day for days 1-5 of each 28-day cycle for a total of 12 cycles.

The patients had previously untreated, histologically proven supratentorial World Health Organization grade II astrocytoma (71 patients); oligodendroglioma (29) or oligoastrocytoma (29) confirmed by central pathology; and at least three of the aforementioned risk factors. The majority of patients (80%) were aged 40 years or older, 79% had tumors greater than 6 cm in the largest diameter, 53% had tumors that crossed the midline, 66% had a dominant astrocytoma subtype, and 50% had preoperative neurological function status scores greater than 1. In all, 89 patients had three risk factors, 32 had four, and 8 had five factors.

The investigators hypothesized that they would see a 43% relative increase in median survival, from 40.5 months in controls to 57.9 months, and a 20% improvement in 3-year survival, from 54% to 65%. As noted before, the survival rates exceeded their expectations.

Dr. Helen Shih

In an analysis of survival by risk factors 5 years after trial registration, the authors found that 35 of the 89 patients with three risk factors (39%) had died, compared with 17 of the 40 patients with four or five risk factors (43%).

In all, 55 patients had grade 3 adverse events as their worst overall events, 13 had grade 4 toxicities, and 1 patient died of herpes simplex encephalitis, possibly related to treatment.

Grade 3 adverse events were seen in 43% of patients, and grade 4 toxicities in 10%. The most common toxicities were hematologic, constitutional, or gastrointestinal, including nausea and anorexia.

In her commentary, Dr. Helen Shih, the invited discussant, said that a randomized trial would have been preferable, but she acknowledged the difficulties the investigators had in designing and implementing the trial. Dr. Shih, of the radiation oncology department at Massachusetts General Hospital in Boston, also noted that the study used for control purposes "itself was conducted in a prior decade, when the management of radiation as well as surgery were slightly different, and perhaps those advancements also have affected overall survival of patients who are treated today."

In addition, the incidences of grade 3 and 4 toxicities in the study by Dr. Fisher and her colleagues were "not trivial, and should be kept in mind," Dr. Shih said.

The study was supported by the National Cancer Institute. Dr. Fisher and Dr. Shih reported having no relevant financial disclosures.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
gliomas, temozolomide-based chemoradiotherapy, American Society of Clinical Oncology, RTOG 0424 trial, temozolomide chemoradiotherapy, Dr. Barbara Fisher
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

CHICAGO – Patients with high-risk, low-grade gliomas had 3-year survival rates significantly higher than those of historical controls when they were treated with a temozolomide-based chemoradiotherapy regimen, investigators reported at the annual meeting of the American Society of Clinical Oncology.

Preliminary results from the phase II RTOG 0424 trial showed a 73% overall survival rate for patients treated with temozolomide chemoradiotherapy, compared with 54% overall survival for historical controls (P less than .001), and exceeding the study’s hypothesized 65% overall survival, reported Dr. Barbara Fisher, a radiation oncologist at the University of Western Ontario in London.

Dr. Barbara Fisher

At a median follow-up of 4 years, with a minimum potential follow-up of 3 years, median survival of patients has not been reached, and the median follow-up time for surviving patients is 5 years, Dr. Fisher noted.

Median progression-free survival was 4.5 years, and 3-year PFS was 59%. In contrast to historical controls, patients with four or five risk factors appeared to have an overall survival rate similar to that of patients with three risk factors, Dr. Fisher reported.

Although the trial was originally proposed in 2000 as a randomized controlled study, "there wasn’t much information about temozolomide and radiation at that point," Dr. Fisher said.

The control population comes from a 2002 study by Francesco Pignatti and his colleagues (J. Clin. Oncol. 2002;20:2076-84), which identified prognostic factors for survival in adults with low-grade glioma, based on data from two European Organization for Research and Treatment of Cancer (EORTC) trials conducted in the 1990s.

The risk factors are age 40 years and older; astrocytoma subtype; tumor crossing the midline; largest tumor diameter more than 6 cm preoperatively; and preoperative neurological function status (neurocognitive function greater than 1: moderate impairment).

A total of 136 patients were accrued, and 129 patients were eligible for the protocol, which consisted of temozolomide 75 mg/m2 per day for 6 weeks with concurrent conformal radiotherapy, consisting of 54 Gy divided into 30 fractions delivered 5 days each week for 6 weeks, followed by temozolomide 150-200 mg/m2 per day for days 1-5 of each 28-day cycle for a total of 12 cycles.

The patients had previously untreated, histologically proven supratentorial World Health Organization grade II astrocytoma (71 patients); oligodendroglioma (29) or oligoastrocytoma (29) confirmed by central pathology; and at least three of the aforementioned risk factors. The majority of patients (80%) were aged 40 years or older, 79% had tumors greater than 6 cm in the largest diameter, 53% had tumors that crossed the midline, 66% had a dominant astrocytoma subtype, and 50% had preoperative neurological function status scores greater than 1. In all, 89 patients had three risk factors, 32 had four, and 8 had five factors.

The investigators hypothesized that they would see a 43% relative increase in median survival, from 40.5 months in controls to 57.9 months, and a 20% improvement in 3-year survival, from 54% to 65%. As noted before, the survival rates exceeded their expectations.

Dr. Helen Shih

In an analysis of survival by risk factors 5 years after trial registration, the authors found that 35 of the 89 patients with three risk factors (39%) had died, compared with 17 of the 40 patients with four or five risk factors (43%).

In all, 55 patients had grade 3 adverse events as their worst overall events, 13 had grade 4 toxicities, and 1 patient died of herpes simplex encephalitis, possibly related to treatment.

Grade 3 adverse events were seen in 43% of patients, and grade 4 toxicities in 10%. The most common toxicities were hematologic, constitutional, or gastrointestinal, including nausea and anorexia.

In her commentary, Dr. Helen Shih, the invited discussant, said that a randomized trial would have been preferable, but she acknowledged the difficulties the investigators had in designing and implementing the trial. Dr. Shih, of the radiation oncology department at Massachusetts General Hospital in Boston, also noted that the study used for control purposes "itself was conducted in a prior decade, when the management of radiation as well as surgery were slightly different, and perhaps those advancements also have affected overall survival of patients who are treated today."

In addition, the incidences of grade 3 and 4 toxicities in the study by Dr. Fisher and her colleagues were "not trivial, and should be kept in mind," Dr. Shih said.

The study was supported by the National Cancer Institute. Dr. Fisher and Dr. Shih reported having no relevant financial disclosures.

CHICAGO – Patients with high-risk, low-grade gliomas had 3-year survival rates significantly higher than those of historical controls when they were treated with a temozolomide-based chemoradiotherapy regimen, investigators reported at the annual meeting of the American Society of Clinical Oncology.

Preliminary results from the phase II RTOG 0424 trial showed a 73% overall survival rate for patients treated with temozolomide chemoradiotherapy, compared with 54% overall survival for historical controls (P less than .001), and exceeding the study’s hypothesized 65% overall survival, reported Dr. Barbara Fisher, a radiation oncologist at the University of Western Ontario in London.

Dr. Barbara Fisher

At a median follow-up of 4 years, with a minimum potential follow-up of 3 years, median survival of patients has not been reached, and the median follow-up time for surviving patients is 5 years, Dr. Fisher noted.

Median progression-free survival was 4.5 years, and 3-year PFS was 59%. In contrast to historical controls, patients with four or five risk factors appeared to have an overall survival rate similar to that of patients with three risk factors, Dr. Fisher reported.

Although the trial was originally proposed in 2000 as a randomized controlled study, "there wasn’t much information about temozolomide and radiation at that point," Dr. Fisher said.

The control population comes from a 2002 study by Francesco Pignatti and his colleagues (J. Clin. Oncol. 2002;20:2076-84), which identified prognostic factors for survival in adults with low-grade glioma, based on data from two European Organization for Research and Treatment of Cancer (EORTC) trials conducted in the 1990s.

The risk factors are age 40 years and older; astrocytoma subtype; tumor crossing the midline; largest tumor diameter more than 6 cm preoperatively; and preoperative neurological function status (neurocognitive function greater than 1: moderate impairment).

A total of 136 patients were accrued, and 129 patients were eligible for the protocol, which consisted of temozolomide 75 mg/m2 per day for 6 weeks with concurrent conformal radiotherapy, consisting of 54 Gy divided into 30 fractions delivered 5 days each week for 6 weeks, followed by temozolomide 150-200 mg/m2 per day for days 1-5 of each 28-day cycle for a total of 12 cycles.

The patients had previously untreated, histologically proven supratentorial World Health Organization grade II astrocytoma (71 patients); oligodendroglioma (29) or oligoastrocytoma (29) confirmed by central pathology; and at least three of the aforementioned risk factors. The majority of patients (80%) were aged 40 years or older, 79% had tumors greater than 6 cm in the largest diameter, 53% had tumors that crossed the midline, 66% had a dominant astrocytoma subtype, and 50% had preoperative neurological function status scores greater than 1. In all, 89 patients had three risk factors, 32 had four, and 8 had five factors.

The investigators hypothesized that they would see a 43% relative increase in median survival, from 40.5 months in controls to 57.9 months, and a 20% improvement in 3-year survival, from 54% to 65%. As noted before, the survival rates exceeded their expectations.

Dr. Helen Shih

In an analysis of survival by risk factors 5 years after trial registration, the authors found that 35 of the 89 patients with three risk factors (39%) had died, compared with 17 of the 40 patients with four or five risk factors (43%).

In all, 55 patients had grade 3 adverse events as their worst overall events, 13 had grade 4 toxicities, and 1 patient died of herpes simplex encephalitis, possibly related to treatment.

Grade 3 adverse events were seen in 43% of patients, and grade 4 toxicities in 10%. The most common toxicities were hematologic, constitutional, or gastrointestinal, including nausea and anorexia.

In her commentary, Dr. Helen Shih, the invited discussant, said that a randomized trial would have been preferable, but she acknowledged the difficulties the investigators had in designing and implementing the trial. Dr. Shih, of the radiation oncology department at Massachusetts General Hospital in Boston, also noted that the study used for control purposes "itself was conducted in a prior decade, when the management of radiation as well as surgery were slightly different, and perhaps those advancements also have affected overall survival of patients who are treated today."

In addition, the incidences of grade 3 and 4 toxicities in the study by Dr. Fisher and her colleagues were "not trivial, and should be kept in mind," Dr. Shih said.

The study was supported by the National Cancer Institute. Dr. Fisher and Dr. Shih reported having no relevant financial disclosures.

Publications
Publications
Topics
Article Type
Display Headline
Good survival rates with temozolomide CRT for high-risk, low-grade gliomas
Display Headline
Good survival rates with temozolomide CRT for high-risk, low-grade gliomas
Legacy Keywords
gliomas, temozolomide-based chemoradiotherapy, American Society of Clinical Oncology, RTOG 0424 trial, temozolomide chemoradiotherapy, Dr. Barbara Fisher
Legacy Keywords
gliomas, temozolomide-based chemoradiotherapy, American Society of Clinical Oncology, RTOG 0424 trial, temozolomide chemoradiotherapy, Dr. Barbara Fisher
Article Source

AT THE ASCO ANNUAL MEETING 2013

PURLs Copyright

Inside the Article

Vitals

Major finding: With temozolomide chemoradiotherapy, 3-year overall survival was 73% for patients with low-grade gliomas and three or more risk factors for worse prognosis, compared with 54% for historical controls (P less than .001).

Data source: Single-arm, prospective phase II study in 129 patients compared with historical controls.

Disclosures: The study was supported by the National Cancer Institute. Dr. Fisher and Dr. Shih reported having no relevant financial disclosures.