LayerRx Mapping ID
184
Slot System
Featured Buckets
Featured Buckets Admin

Genetic Signatures May Predict CAR T Responders

Article Type
Changed
Wed, 04/24/2024 - 13:52

 

Key novel genetic signatures in patients with relapsed/refractory large B-cell lymphoma (r/r LBCL) strongly correlate with improved survival outcomes in treatment with the anti-CD19 CAR T-cell therapy axicabtagene ciloleucel (axi-cel).

“Our transcriptomic analysis of ZUMA-7 dataset identified novel gene expression signatures predictive of outcome with axi-cel,” the authors reported in research presented at the annual meeting of the American Association for Cancer Research earlier in April. “These gene expression signatures could support risk-stratification of LBCL patients.”

The results are from a subanalysis of the phase 3 ZUMA-7 trial in which patients with early relapsed or primary refractory LBCL were treated with axi-cel, administered as a one-time dose in the second-line setting.

Long-term results from the trial showed a 4-year overall survival of 54.6% with axi-cel versus 46.0% with the standard of care (P = .03), with a median rate of progression-free survival of 14.7 months with axi-cel versus 3.7 months in the standard-second-line treatment.

In the study, the authors noted that, “although the use of axi-cel resulted in long-term survival in more than half of treated patients, it is important to continue to strive to improve patient outcomes.”

Following up on that, senior author Simone Filosto, of Kite, a Gilead Company, of Santa Monica, California, and colleagues launched their analysis of the genetic profiles of those who did and did not have favorable responses, using data from the ZUMA-7 trial.

Using gene expression profiling with the IO-360 Nanostring gene expression panel of 769 genes, they evaluated pretreated LBCL tumor samples from 134 of the patients treated with axi-cel.

After multivariate adjustment, the results showed that those with a distinctive 6-transcript genetic expression signature, consisting of CD19, CD45RA, CCL22, KLRK1, SOX11, and SIGLEC5, had a significantly higher rate of event-free survival (hazard ratio [HR], 0.27; P = 1.82 x 10-8), as well as progression-free survival (HR, 0.27; P = 1.35 x 10-7) after treatment with axi-cel, compared with those who did not have the signature.

The authors speculated that “the 6-gene expression signature may capture lymphomas with abundant adhesion molecules, a relatively low inflammation, and abundant expression of the targeted antigen (CD19).”

Conversely, the analysis showed that increased levels of an unfavorable 17-transcript gene expression signature had a strong negative correlation with event-free survival (HR, 6.19; P = 1.51 x 10-13) and progression-free survival (HR, 7.58; P = 2.70 x 10-14).

The 17-transcript signature included CD45RO, BCL2, IL-18R1, TNFSF4 [OX40L], KLRB1 [CD161], KIR3DL2, ITGB8, DUSP5, GPC4, PSMB5, RPS6KB1, SERPINA9, NBN, GLUD1, ESR1, ARID1A, and SLC16A1.

“The 17-gene expression signature is consistent with a high level of immune infiltration and inflammation paralleled by the activation of immune-escape mechanisms, such as the upregulation of anti-apoptotic genes,” the authors explain.

Of note, the 17-gene expression signature was elevated among 18 patients who progressed after axi-cel treatment.

Importantly, the gene expression signatures were not associated with outcomes observed among patients receiving second-line standard of care in the ZUMA-7 trial. And the signatures also did not correspond with outcomes following first-line R-CHOP chemotherapy reported in two online datasets, indicating their predictive rather than prognostic value.

Commenting on the findings, Marco Ruella, MD, noted that “stratifying the [CAR T-treated] patients is extremely important given that only a subset of them, 30%-40%, will experience long-term remission.”

“In an ideal scenario, we would want to treat only the patients who would benefit from such a complex and expensive therapy,” underscored Dr. Ruella, assistant professor in the Division of Hematology/Oncology and the Center for Cellular Immunotherapies and Scientific Director of the Lymphoma Program at the Hospital of the University of Pennsylvania in Philadelphia.

A key caveat is that the results need more validation before they true gain clinical value, he noted.

“We need more data before we can use such a score in the clinic as we would need to be absolutely confident on the predictive value of such a score in additional confirmatory cohorts.”

Furthermore, caution is warranted in avoiding excluding any patients unnecessarily, he added.

“Only if there are approximately zero chances of response would we be able to exclude a patient from a treatment,” Dr. Ruella noted. “If the chance of long-term cure are minimal but still present, it might still make sense for the patient.” 

Nevertheless, such findings advance the understanding of the therapy’s implication in a meaningful way, he said.

“I think this study [and similar others] are important studies that help us better understand the mechanisms of relapse,” he said.

“Translationally, we are getting closer to reaching a point where we can precisely predict outcomes and, perhaps in the future, select the patients that would benefit the most from these treatments.”

Dr. Filosto and other authors are employees of Kite, which manufactures axi-cel. Dr. Ruella treats patients with CAR T products that have been licensed to Novartis, Kite, and Vittoria Bio.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

Key novel genetic signatures in patients with relapsed/refractory large B-cell lymphoma (r/r LBCL) strongly correlate with improved survival outcomes in treatment with the anti-CD19 CAR T-cell therapy axicabtagene ciloleucel (axi-cel).

“Our transcriptomic analysis of ZUMA-7 dataset identified novel gene expression signatures predictive of outcome with axi-cel,” the authors reported in research presented at the annual meeting of the American Association for Cancer Research earlier in April. “These gene expression signatures could support risk-stratification of LBCL patients.”

The results are from a subanalysis of the phase 3 ZUMA-7 trial in which patients with early relapsed or primary refractory LBCL were treated with axi-cel, administered as a one-time dose in the second-line setting.

Long-term results from the trial showed a 4-year overall survival of 54.6% with axi-cel versus 46.0% with the standard of care (P = .03), with a median rate of progression-free survival of 14.7 months with axi-cel versus 3.7 months in the standard-second-line treatment.

In the study, the authors noted that, “although the use of axi-cel resulted in long-term survival in more than half of treated patients, it is important to continue to strive to improve patient outcomes.”

Following up on that, senior author Simone Filosto, of Kite, a Gilead Company, of Santa Monica, California, and colleagues launched their analysis of the genetic profiles of those who did and did not have favorable responses, using data from the ZUMA-7 trial.

Using gene expression profiling with the IO-360 Nanostring gene expression panel of 769 genes, they evaluated pretreated LBCL tumor samples from 134 of the patients treated with axi-cel.

After multivariate adjustment, the results showed that those with a distinctive 6-transcript genetic expression signature, consisting of CD19, CD45RA, CCL22, KLRK1, SOX11, and SIGLEC5, had a significantly higher rate of event-free survival (hazard ratio [HR], 0.27; P = 1.82 x 10-8), as well as progression-free survival (HR, 0.27; P = 1.35 x 10-7) after treatment with axi-cel, compared with those who did not have the signature.

The authors speculated that “the 6-gene expression signature may capture lymphomas with abundant adhesion molecules, a relatively low inflammation, and abundant expression of the targeted antigen (CD19).”

Conversely, the analysis showed that increased levels of an unfavorable 17-transcript gene expression signature had a strong negative correlation with event-free survival (HR, 6.19; P = 1.51 x 10-13) and progression-free survival (HR, 7.58; P = 2.70 x 10-14).

The 17-transcript signature included CD45RO, BCL2, IL-18R1, TNFSF4 [OX40L], KLRB1 [CD161], KIR3DL2, ITGB8, DUSP5, GPC4, PSMB5, RPS6KB1, SERPINA9, NBN, GLUD1, ESR1, ARID1A, and SLC16A1.

“The 17-gene expression signature is consistent with a high level of immune infiltration and inflammation paralleled by the activation of immune-escape mechanisms, such as the upregulation of anti-apoptotic genes,” the authors explain.

Of note, the 17-gene expression signature was elevated among 18 patients who progressed after axi-cel treatment.

Importantly, the gene expression signatures were not associated with outcomes observed among patients receiving second-line standard of care in the ZUMA-7 trial. And the signatures also did not correspond with outcomes following first-line R-CHOP chemotherapy reported in two online datasets, indicating their predictive rather than prognostic value.

Commenting on the findings, Marco Ruella, MD, noted that “stratifying the [CAR T-treated] patients is extremely important given that only a subset of them, 30%-40%, will experience long-term remission.”

“In an ideal scenario, we would want to treat only the patients who would benefit from such a complex and expensive therapy,” underscored Dr. Ruella, assistant professor in the Division of Hematology/Oncology and the Center for Cellular Immunotherapies and Scientific Director of the Lymphoma Program at the Hospital of the University of Pennsylvania in Philadelphia.

A key caveat is that the results need more validation before they true gain clinical value, he noted.

“We need more data before we can use such a score in the clinic as we would need to be absolutely confident on the predictive value of such a score in additional confirmatory cohorts.”

Furthermore, caution is warranted in avoiding excluding any patients unnecessarily, he added.

“Only if there are approximately zero chances of response would we be able to exclude a patient from a treatment,” Dr. Ruella noted. “If the chance of long-term cure are minimal but still present, it might still make sense for the patient.” 

Nevertheless, such findings advance the understanding of the therapy’s implication in a meaningful way, he said.

“I think this study [and similar others] are important studies that help us better understand the mechanisms of relapse,” he said.

“Translationally, we are getting closer to reaching a point where we can precisely predict outcomes and, perhaps in the future, select the patients that would benefit the most from these treatments.”

Dr. Filosto and other authors are employees of Kite, which manufactures axi-cel. Dr. Ruella treats patients with CAR T products that have been licensed to Novartis, Kite, and Vittoria Bio.

 

Key novel genetic signatures in patients with relapsed/refractory large B-cell lymphoma (r/r LBCL) strongly correlate with improved survival outcomes in treatment with the anti-CD19 CAR T-cell therapy axicabtagene ciloleucel (axi-cel).

“Our transcriptomic analysis of ZUMA-7 dataset identified novel gene expression signatures predictive of outcome with axi-cel,” the authors reported in research presented at the annual meeting of the American Association for Cancer Research earlier in April. “These gene expression signatures could support risk-stratification of LBCL patients.”

The results are from a subanalysis of the phase 3 ZUMA-7 trial in which patients with early relapsed or primary refractory LBCL were treated with axi-cel, administered as a one-time dose in the second-line setting.

Long-term results from the trial showed a 4-year overall survival of 54.6% with axi-cel versus 46.0% with the standard of care (P = .03), with a median rate of progression-free survival of 14.7 months with axi-cel versus 3.7 months in the standard-second-line treatment.

In the study, the authors noted that, “although the use of axi-cel resulted in long-term survival in more than half of treated patients, it is important to continue to strive to improve patient outcomes.”

Following up on that, senior author Simone Filosto, of Kite, a Gilead Company, of Santa Monica, California, and colleagues launched their analysis of the genetic profiles of those who did and did not have favorable responses, using data from the ZUMA-7 trial.

Using gene expression profiling with the IO-360 Nanostring gene expression panel of 769 genes, they evaluated pretreated LBCL tumor samples from 134 of the patients treated with axi-cel.

After multivariate adjustment, the results showed that those with a distinctive 6-transcript genetic expression signature, consisting of CD19, CD45RA, CCL22, KLRK1, SOX11, and SIGLEC5, had a significantly higher rate of event-free survival (hazard ratio [HR], 0.27; P = 1.82 x 10-8), as well as progression-free survival (HR, 0.27; P = 1.35 x 10-7) after treatment with axi-cel, compared with those who did not have the signature.

The authors speculated that “the 6-gene expression signature may capture lymphomas with abundant adhesion molecules, a relatively low inflammation, and abundant expression of the targeted antigen (CD19).”

Conversely, the analysis showed that increased levels of an unfavorable 17-transcript gene expression signature had a strong negative correlation with event-free survival (HR, 6.19; P = 1.51 x 10-13) and progression-free survival (HR, 7.58; P = 2.70 x 10-14).

The 17-transcript signature included CD45RO, BCL2, IL-18R1, TNFSF4 [OX40L], KLRB1 [CD161], KIR3DL2, ITGB8, DUSP5, GPC4, PSMB5, RPS6KB1, SERPINA9, NBN, GLUD1, ESR1, ARID1A, and SLC16A1.

“The 17-gene expression signature is consistent with a high level of immune infiltration and inflammation paralleled by the activation of immune-escape mechanisms, such as the upregulation of anti-apoptotic genes,” the authors explain.

Of note, the 17-gene expression signature was elevated among 18 patients who progressed after axi-cel treatment.

Importantly, the gene expression signatures were not associated with outcomes observed among patients receiving second-line standard of care in the ZUMA-7 trial. And the signatures also did not correspond with outcomes following first-line R-CHOP chemotherapy reported in two online datasets, indicating their predictive rather than prognostic value.

Commenting on the findings, Marco Ruella, MD, noted that “stratifying the [CAR T-treated] patients is extremely important given that only a subset of them, 30%-40%, will experience long-term remission.”

“In an ideal scenario, we would want to treat only the patients who would benefit from such a complex and expensive therapy,” underscored Dr. Ruella, assistant professor in the Division of Hematology/Oncology and the Center for Cellular Immunotherapies and Scientific Director of the Lymphoma Program at the Hospital of the University of Pennsylvania in Philadelphia.

A key caveat is that the results need more validation before they true gain clinical value, he noted.

“We need more data before we can use such a score in the clinic as we would need to be absolutely confident on the predictive value of such a score in additional confirmatory cohorts.”

Furthermore, caution is warranted in avoiding excluding any patients unnecessarily, he added.

“Only if there are approximately zero chances of response would we be able to exclude a patient from a treatment,” Dr. Ruella noted. “If the chance of long-term cure are minimal but still present, it might still make sense for the patient.” 

Nevertheless, such findings advance the understanding of the therapy’s implication in a meaningful way, he said.

“I think this study [and similar others] are important studies that help us better understand the mechanisms of relapse,” he said.

“Translationally, we are getting closer to reaching a point where we can precisely predict outcomes and, perhaps in the future, select the patients that would benefit the most from these treatments.”

Dr. Filosto and other authors are employees of Kite, which manufactures axi-cel. Dr. Ruella treats patients with CAR T products that have been licensed to Novartis, Kite, and Vittoria Bio.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167836</fileName> <TBEID>0C04FC84.SIG</TBEID> <TBUniqueIdentifier>MD_0C04FC84</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>AACR_CARTGenetic</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240424T134557</QCDate> <firstPublished>20240424T134711</firstPublished> <LastPublished>20240424T134711</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240424T134711</CMSDate> <articleSource>FROM AACR 2024</articleSource> <facebookInfo/> <meetingNumber>2976-24</meetingNumber> <byline>Nancy A. Melville</byline> <bylineText>NANCY A. MELVILLE</bylineText> <bylineFull>NANCY A. MELVILLE</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>Key novel genetic signatures in patients with relapsed/refractory large B-cell lymphoma (r/r LBCL) strongly correlate with improved survival outcomes in treatme</metaDescription> <articlePDF/> <teaserImage/> <teaser>Research data suggest that key genetic clues of patients with LBCL could show which are more — or less — likely to respond to axi-cel CAR T.</teaser> <title>Genetic Signatures May Predict CAR T Responders</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>hemn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">18</term> </publications> <sections> <term canonical="true">53</term> <term>39313</term> </sections> <topics> <term>61821</term> <term canonical="true">195</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Genetic Signatures May Predict CAR T Responders</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">Key novel genetic signatures in patients with relapsed/refractory large B-cell lymphoma (r/r LBCL) strongly correlate with improved survival outcomes in treatment with the anti-CD19 CAR T-cell therapy axicabtagene ciloleucel (axi-cel).</span><br/><br/> “Our transcriptomic analysis of ZUMA-7 dataset identified novel gene expression signatures predictive of outcome with axi-cel,” the authors reported in <span class="Hyperlink"><a href="https://www.abstractsonline.com/pp8/#!/20272/presentation/10384">research presented </a></span>at the annual meeting of the American Association for Cancer Research earlier in April. “These gene expression signatures could support risk-stratification of LBCL patients.”<br/><br/>The results are from a subanalysis of the phase 3 ZUMA-7 trial in which patients with early relapsed or primary refractory LBCL were treated with axi-cel, administered as a one-time dose in the second-line setting. <br/><br/><span class="Hyperlink"><a href="ttps://www.nejm.org/doi/full/10.1056/NEJMoa2301665">Long-term results</a></span> from the trial showed a 4-year overall survival of 54.6% with axi-cel versus 46.0% with the standard of care (<em>P</em> = .03), with a median rate of progression-free survival of 14.7 months with axi-cel versus 3.7 months in the standard-second-line treatment. <br/><br/>In the study, the authors noted that, “although the use of axi-cel resulted in long-term survival in more than half of treated patients, it is important to continue to strive to improve patient outcomes.”<br/><br/>Following up on that, senior author Simone Filosto, of Kite, a Gilead Company, of Santa Monica, California, and colleagues launched their analysis of the genetic profiles of those who did and did not have favorable responses, using data from the ZUMA-7 trial.<br/><br/>Using gene expression profiling with the IO-360 Nanostring gene expression panel of 769 genes, they evaluated pretreated LBCL tumor samples from 134 of the patients treated with axi-cel. <br/><br/>After multivariate adjustment, the results showed that those with a distinctive 6-transcript genetic expression signature, consisting of CD19, CD45RA, CCL22, KLRK1, SOX11, and SIGLEC5, had a significantly higher rate of event-free survival (hazard ratio [HR], 0.27; <em>P</em> = 1.82 x 10<sup>-8</sup>), as well as progression-free survival (HR, 0.27; <em>P</em> = 1.35 x 10<sup>-7</sup>) after treatment with axi-cel, compared with those who did not have the signature. <br/><br/>The authors speculated that “the 6-gene expression signature may capture lymphomas with abundant adhesion molecules, a relatively low inflammation, and abundant expression of the targeted antigen (CD19).”<br/><br/>Conversely, the analysis showed that increased levels of an unfavorable 17-transcript gene expression signature had a strong negative correlation with event-free survival (HR, 6.19; <em>P</em> = 1.51 x 10<sup>-13</sup>) and progression-free survival (HR, 7.58; <em>P</em> = 2.70 x 10<sup>-14</sup>). <br/><br/>The 17-transcript signature included CD45RO, BCL2, IL-18R1, TNFSF4 [OX40L], KLRB1 [CD161], KIR3DL2, ITGB8, DUSP5, GPC4, PSMB5, RPS6KB1, SERPINA9, NBN, GLUD1, ESR1, ARID1A, and SLC16A1.<br/><br/>“The 17-gene expression signature is consistent with a high level of immune infiltration and inflammation paralleled by the activation of immune-escape mechanisms, such as the upregulation of anti-apoptotic genes,” the authors explain. <br/><br/>Of note, the 17-gene expression signature was elevated among 18 patients who progressed after axi-cel treatment. <br/><br/>Importantly, the gene expression signatures were not associated with outcomes observed among patients receiving second-line standard of care in the ZUMA-7 trial. And the signatures also did not correspond with outcomes following first-line R-CHOP chemotherapy reported in two online datasets, indicating their predictive rather than prognostic value.<br/><br/>Commenting on the findings, Marco Ruella, MD, noted that “stratifying the [CAR T-treated] patients is extremely important given that only a subset of them, 30%-40%, will experience long-term remission.” <br/><br/>“In an ideal scenario, we would want to treat only the patients who would benefit from such a complex and expensive therapy,” underscored Dr. Ruella, assistant professor in the Division of Hematology/Oncology and the Center for Cellular Immunotherapies and Scientific Director of the Lymphoma Program at the Hospital of the University of Pennsylvania in Philadelphia.<br/><br/>A key caveat is that the results need more validation before they true gain clinical value, he noted.<br/><br/>“We need more data before we can use such a score in the clinic as we would need to be absolutely confident on the predictive value of such a score in additional confirmatory cohorts.”<br/><br/>Furthermore, caution is warranted in avoiding excluding any patients unnecessarily, he added.<br/><br/>“Only if there are approximately zero chances of response would we be able to exclude a patient from a treatment,” Dr. Ruella noted. “If the chance of long-term cure are minimal but still present, it might still make sense for the patient.” <br/><br/>Nevertheless, such findings advance the understanding of the therapy’s implication in a meaningful way, he said. <br/><br/>“I think this study [and similar others] are important studies that help us better understand the mechanisms of relapse,” he said. <br/><br/>“Translationally, we are getting closer to reaching a point where we can precisely predict outcomes and, perhaps in the future, select the patients that would benefit the most from these treatments.”<br/><br/>Dr. Filosto and other authors are employees of Kite, which manufactures axi-cel. Dr. Ruella treats patients with CAR T products that have been licensed to Novartis, Kite, and Vittoria Bio.<span class="end"/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM AACR 2024

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Tiny Doses of Metabolically Armed CAR T Show Benefits

Article Type
Changed
Tue, 04/23/2024 - 10:21

 

A novel CD19 chimeric antigen responder (CAR) T-cell therapy administered in exceptionally low doses and designed to reinvigorate exhausted T-cells shows impressive early results in the treatment of patients with relapsed or refractory (r/r) diffuse large B-cell lymphoma (DLBCL) and B-cell acute lymphoblastic leukemia (B-ALL).

“Our study showed a manageable safety profile in r/r DLBCL/B-ALL, with promising breakthrough efficacy of a 100% complete remission in all dose groups,” said first author Jingjing Ren, MD, PhD, associate director of research and development with Leman Biotech in Shenzhen, China. Dr. Ren presented these findings at the American Association for Cancer Research annual meeting held in San Diego.

While CD19 CAR T-cell therapy has been transformative in the treatment of relapsed B -cell hematological malignancies in recent years, more than half of patients relapse within a year because of inadequate CAR T persistence.

To address the problem, Dr. Ren and her colleagues developed a metabolically armed, interleukin (IL)-10-expressing CAR T-cell product called Meta10-19 for the treatment patients with r/r DLBCL or r/r B-ALL.

According to the authors, the IL-10-expressing CAR T-cells trigger “stem-like memory responses” in various lymphoid organs, which prompt a “robust tumor eradication and durable protection,” and hence, better persistence.

Preclinical studies in mice showed the Meta10-19 CAR T-cells exhibited substantially higher expansion of approximately 100-fold compared with a control CD19 CAR-T product.

Therefore, “we dramatically reduced the dose to approximately 1% to 5% of commercial products for the IL-10-expressing CD19 CAR-T for patients,” coauthor Yugang Guo, PhD, cofounder and president of Leman Biotech said in an interview.

For the ongoing, open-label clinical trial, 12 adult patients with r/r DLBCL or r/r B-ALL and confirmed CD19 expression at a hospital center in China were enrolled between December 2022 and November 2023 and treated in three cohorts, receiving doses that corresponded to 1%, 2.5%, or 5% of the doses of other commercialized CAR-T infusion products.

All patients also underwent lympho-depleting chemotherapy with cyclophosphamide and fludarabine prior to the CAR T-cell infusion.

Six patients had r/r DLBCL and the other six had r/r B-ALL; their median age was 47 and their median time since diagnosis was 1 year.

In the single-arm, intent-to-treat analysis, the treatment induced a complete remission in all 12 patients, as evaluated by PET-CT scan, nuclear magnetic resonance (NMR) spectroscopy, or minimal residual disease assessment of bone marrow.

The median time to best response was 1 month (range 0.5 to 2.2 months).

There were no cases of severe cytokine storm syndrome or neurotoxicity, which are among key limitations with current commercial CAR-T products.

All of the patients continued to have a complete remission at 3 months. Two of the 12 patients, both with B-ALL, experienced relapses, one after 4.7 months and the other at 8 months.

The authors reported that the first treated patient had maintained continuous remission as of 9 months.

In comparison with the much higher full doses of commercial CD19 CAR-T products, only about 50% of patients with DLBCL and 70% of B-ALL patients have been shown to achieve CR at 3 months, the authors reported.

“Our IL-10 expressing CAR-T sustains CR at 3 months post infusion in the context of not following allogeneic hematopoietic stem cell transplant, which suggests IL-10 expressing CAR-T is more resistant to relapse,” Dr. Guo said.

In terms of safety, six patients with DLBCL and four with B-ALL experienced grade 1 cytokine release syndrome (CRS), and two patients with B-ALL developed grade 2 CRS. There were no grade 3 or 4 CRS cases.

One patient with B-ALL developed grade 3 ICANS.

Grade 3-4 cytopenias occurred in most patients, but all were limited to no later than 90 days.

“We observed reduced CRS, with no level 3 or 4, or ICANS,” Dr. Guo said. “There was increased cytopenia, but still manageable, compared with commercial products.”

Of note, the Meta10-19 cells showed efficacy in the extremely low infusion doses even among patients with bulky mass (≥ 7.5 cm) of DLBCL, which is associated with an increased risk of relapse.

One patient had primary central nervous system lymphoma (PCNSL), a rare form of DLBCL that is known to have the worst prognosis of all non-Hodgkin lymphomas.

Due to the unique nature of CNS primary tumors, the CAR T-cell infusion dose was further reduced to 1% of the standard dose for the patient.

The patient maintained complete remission for more than 8 months before relapsing in periphery blood, but not in the CNS, Dr. Guo noted.

“Luckily, this relapse has been easily controlled by chemotherapy, and the patient is maintaining complete remission again now,” Dr. Guo said.
 

 

 

Mechanisms?

Dr. Guo noted that the mechanism believed to explain the improvements despite such low doses is that “IL-10-expressing CAR-T exhibits enhanced proliferation, cytotoxicity, and stem-like antitumor memory due to enhanced metabolic activities of oxidative phosphorylation.”

The authors noted that a key major factor limiting accessibility to CAR-T therapies is the lengthy production cycle and high costs; however, the “extremely low doses of 1% to 5% can significantly reduce the production cycle and cost of CAR T-cell therapies, increasing accessibility,” they wrote in a press statement.

Currently, more than 20 patients have achieved a CR overall, and studies with a larger cohort and longer follow-up are ongoing, Dr. Guo reported.

The research team plans to launch further clinical investigation this year into patients with solid tumors.

Commenting on the study, Hongbo Chi, PhD, the Robert G. Webster Endowed Chair in Immunology at St. Jude Children’s Research Hospital in Memphis, Tennessee, noted that, based on the abstract, “the effects are quite remarkable, considering the therapeutic efficacy observed even at the low dose.

“Results from more patients are needed to fully validate these findings, but the results to date are very encouraging,” he said.

The study was sponsored by Leman Biotech. Dr. Chi had no disclosures to report.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

A novel CD19 chimeric antigen responder (CAR) T-cell therapy administered in exceptionally low doses and designed to reinvigorate exhausted T-cells shows impressive early results in the treatment of patients with relapsed or refractory (r/r) diffuse large B-cell lymphoma (DLBCL) and B-cell acute lymphoblastic leukemia (B-ALL).

“Our study showed a manageable safety profile in r/r DLBCL/B-ALL, with promising breakthrough efficacy of a 100% complete remission in all dose groups,” said first author Jingjing Ren, MD, PhD, associate director of research and development with Leman Biotech in Shenzhen, China. Dr. Ren presented these findings at the American Association for Cancer Research annual meeting held in San Diego.

While CD19 CAR T-cell therapy has been transformative in the treatment of relapsed B -cell hematological malignancies in recent years, more than half of patients relapse within a year because of inadequate CAR T persistence.

To address the problem, Dr. Ren and her colleagues developed a metabolically armed, interleukin (IL)-10-expressing CAR T-cell product called Meta10-19 for the treatment patients with r/r DLBCL or r/r B-ALL.

According to the authors, the IL-10-expressing CAR T-cells trigger “stem-like memory responses” in various lymphoid organs, which prompt a “robust tumor eradication and durable protection,” and hence, better persistence.

Preclinical studies in mice showed the Meta10-19 CAR T-cells exhibited substantially higher expansion of approximately 100-fold compared with a control CD19 CAR-T product.

Therefore, “we dramatically reduced the dose to approximately 1% to 5% of commercial products for the IL-10-expressing CD19 CAR-T for patients,” coauthor Yugang Guo, PhD, cofounder and president of Leman Biotech said in an interview.

For the ongoing, open-label clinical trial, 12 adult patients with r/r DLBCL or r/r B-ALL and confirmed CD19 expression at a hospital center in China were enrolled between December 2022 and November 2023 and treated in three cohorts, receiving doses that corresponded to 1%, 2.5%, or 5% of the doses of other commercialized CAR-T infusion products.

All patients also underwent lympho-depleting chemotherapy with cyclophosphamide and fludarabine prior to the CAR T-cell infusion.

Six patients had r/r DLBCL and the other six had r/r B-ALL; their median age was 47 and their median time since diagnosis was 1 year.

In the single-arm, intent-to-treat analysis, the treatment induced a complete remission in all 12 patients, as evaluated by PET-CT scan, nuclear magnetic resonance (NMR) spectroscopy, or minimal residual disease assessment of bone marrow.

The median time to best response was 1 month (range 0.5 to 2.2 months).

There were no cases of severe cytokine storm syndrome or neurotoxicity, which are among key limitations with current commercial CAR-T products.

All of the patients continued to have a complete remission at 3 months. Two of the 12 patients, both with B-ALL, experienced relapses, one after 4.7 months and the other at 8 months.

The authors reported that the first treated patient had maintained continuous remission as of 9 months.

In comparison with the much higher full doses of commercial CD19 CAR-T products, only about 50% of patients with DLBCL and 70% of B-ALL patients have been shown to achieve CR at 3 months, the authors reported.

“Our IL-10 expressing CAR-T sustains CR at 3 months post infusion in the context of not following allogeneic hematopoietic stem cell transplant, which suggests IL-10 expressing CAR-T is more resistant to relapse,” Dr. Guo said.

In terms of safety, six patients with DLBCL and four with B-ALL experienced grade 1 cytokine release syndrome (CRS), and two patients with B-ALL developed grade 2 CRS. There were no grade 3 or 4 CRS cases.

One patient with B-ALL developed grade 3 ICANS.

Grade 3-4 cytopenias occurred in most patients, but all were limited to no later than 90 days.

“We observed reduced CRS, with no level 3 or 4, or ICANS,” Dr. Guo said. “There was increased cytopenia, but still manageable, compared with commercial products.”

Of note, the Meta10-19 cells showed efficacy in the extremely low infusion doses even among patients with bulky mass (≥ 7.5 cm) of DLBCL, which is associated with an increased risk of relapse.

One patient had primary central nervous system lymphoma (PCNSL), a rare form of DLBCL that is known to have the worst prognosis of all non-Hodgkin lymphomas.

Due to the unique nature of CNS primary tumors, the CAR T-cell infusion dose was further reduced to 1% of the standard dose for the patient.

The patient maintained complete remission for more than 8 months before relapsing in periphery blood, but not in the CNS, Dr. Guo noted.

“Luckily, this relapse has been easily controlled by chemotherapy, and the patient is maintaining complete remission again now,” Dr. Guo said.
 

 

 

Mechanisms?

Dr. Guo noted that the mechanism believed to explain the improvements despite such low doses is that “IL-10-expressing CAR-T exhibits enhanced proliferation, cytotoxicity, and stem-like antitumor memory due to enhanced metabolic activities of oxidative phosphorylation.”

The authors noted that a key major factor limiting accessibility to CAR-T therapies is the lengthy production cycle and high costs; however, the “extremely low doses of 1% to 5% can significantly reduce the production cycle and cost of CAR T-cell therapies, increasing accessibility,” they wrote in a press statement.

Currently, more than 20 patients have achieved a CR overall, and studies with a larger cohort and longer follow-up are ongoing, Dr. Guo reported.

The research team plans to launch further clinical investigation this year into patients with solid tumors.

Commenting on the study, Hongbo Chi, PhD, the Robert G. Webster Endowed Chair in Immunology at St. Jude Children’s Research Hospital in Memphis, Tennessee, noted that, based on the abstract, “the effects are quite remarkable, considering the therapeutic efficacy observed even at the low dose.

“Results from more patients are needed to fully validate these findings, but the results to date are very encouraging,” he said.

The study was sponsored by Leman Biotech. Dr. Chi had no disclosures to report.

 

A novel CD19 chimeric antigen responder (CAR) T-cell therapy administered in exceptionally low doses and designed to reinvigorate exhausted T-cells shows impressive early results in the treatment of patients with relapsed or refractory (r/r) diffuse large B-cell lymphoma (DLBCL) and B-cell acute lymphoblastic leukemia (B-ALL).

“Our study showed a manageable safety profile in r/r DLBCL/B-ALL, with promising breakthrough efficacy of a 100% complete remission in all dose groups,” said first author Jingjing Ren, MD, PhD, associate director of research and development with Leman Biotech in Shenzhen, China. Dr. Ren presented these findings at the American Association for Cancer Research annual meeting held in San Diego.

While CD19 CAR T-cell therapy has been transformative in the treatment of relapsed B -cell hematological malignancies in recent years, more than half of patients relapse within a year because of inadequate CAR T persistence.

To address the problem, Dr. Ren and her colleagues developed a metabolically armed, interleukin (IL)-10-expressing CAR T-cell product called Meta10-19 for the treatment patients with r/r DLBCL or r/r B-ALL.

According to the authors, the IL-10-expressing CAR T-cells trigger “stem-like memory responses” in various lymphoid organs, which prompt a “robust tumor eradication and durable protection,” and hence, better persistence.

Preclinical studies in mice showed the Meta10-19 CAR T-cells exhibited substantially higher expansion of approximately 100-fold compared with a control CD19 CAR-T product.

Therefore, “we dramatically reduced the dose to approximately 1% to 5% of commercial products for the IL-10-expressing CD19 CAR-T for patients,” coauthor Yugang Guo, PhD, cofounder and president of Leman Biotech said in an interview.

For the ongoing, open-label clinical trial, 12 adult patients with r/r DLBCL or r/r B-ALL and confirmed CD19 expression at a hospital center in China were enrolled between December 2022 and November 2023 and treated in three cohorts, receiving doses that corresponded to 1%, 2.5%, or 5% of the doses of other commercialized CAR-T infusion products.

All patients also underwent lympho-depleting chemotherapy with cyclophosphamide and fludarabine prior to the CAR T-cell infusion.

Six patients had r/r DLBCL and the other six had r/r B-ALL; their median age was 47 and their median time since diagnosis was 1 year.

In the single-arm, intent-to-treat analysis, the treatment induced a complete remission in all 12 patients, as evaluated by PET-CT scan, nuclear magnetic resonance (NMR) spectroscopy, or minimal residual disease assessment of bone marrow.

The median time to best response was 1 month (range 0.5 to 2.2 months).

There were no cases of severe cytokine storm syndrome or neurotoxicity, which are among key limitations with current commercial CAR-T products.

All of the patients continued to have a complete remission at 3 months. Two of the 12 patients, both with B-ALL, experienced relapses, one after 4.7 months and the other at 8 months.

The authors reported that the first treated patient had maintained continuous remission as of 9 months.

In comparison with the much higher full doses of commercial CD19 CAR-T products, only about 50% of patients with DLBCL and 70% of B-ALL patients have been shown to achieve CR at 3 months, the authors reported.

“Our IL-10 expressing CAR-T sustains CR at 3 months post infusion in the context of not following allogeneic hematopoietic stem cell transplant, which suggests IL-10 expressing CAR-T is more resistant to relapse,” Dr. Guo said.

In terms of safety, six patients with DLBCL and four with B-ALL experienced grade 1 cytokine release syndrome (CRS), and two patients with B-ALL developed grade 2 CRS. There were no grade 3 or 4 CRS cases.

One patient with B-ALL developed grade 3 ICANS.

Grade 3-4 cytopenias occurred in most patients, but all were limited to no later than 90 days.

“We observed reduced CRS, with no level 3 or 4, or ICANS,” Dr. Guo said. “There was increased cytopenia, but still manageable, compared with commercial products.”

Of note, the Meta10-19 cells showed efficacy in the extremely low infusion doses even among patients with bulky mass (≥ 7.5 cm) of DLBCL, which is associated with an increased risk of relapse.

One patient had primary central nervous system lymphoma (PCNSL), a rare form of DLBCL that is known to have the worst prognosis of all non-Hodgkin lymphomas.

Due to the unique nature of CNS primary tumors, the CAR T-cell infusion dose was further reduced to 1% of the standard dose for the patient.

The patient maintained complete remission for more than 8 months before relapsing in periphery blood, but not in the CNS, Dr. Guo noted.

“Luckily, this relapse has been easily controlled by chemotherapy, and the patient is maintaining complete remission again now,” Dr. Guo said.
 

 

 

Mechanisms?

Dr. Guo noted that the mechanism believed to explain the improvements despite such low doses is that “IL-10-expressing CAR-T exhibits enhanced proliferation, cytotoxicity, and stem-like antitumor memory due to enhanced metabolic activities of oxidative phosphorylation.”

The authors noted that a key major factor limiting accessibility to CAR-T therapies is the lengthy production cycle and high costs; however, the “extremely low doses of 1% to 5% can significantly reduce the production cycle and cost of CAR T-cell therapies, increasing accessibility,” they wrote in a press statement.

Currently, more than 20 patients have achieved a CR overall, and studies with a larger cohort and longer follow-up are ongoing, Dr. Guo reported.

The research team plans to launch further clinical investigation this year into patients with solid tumors.

Commenting on the study, Hongbo Chi, PhD, the Robert G. Webster Endowed Chair in Immunology at St. Jude Children’s Research Hospital in Memphis, Tennessee, noted that, based on the abstract, “the effects are quite remarkable, considering the therapeutic efficacy observed even at the low dose.

“Results from more patients are needed to fully validate these findings, but the results to date are very encouraging,” he said.

The study was sponsored by Leman Biotech. Dr. Chi had no disclosures to report.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167804</fileName> <TBEID>0C04FB94.SIG</TBEID> <TBUniqueIdentifier>MD_0C04FB94</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>AACR_Metabolic CAR-T Therapy</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240423T101429</QCDate> <firstPublished>20240423T101437</firstPublished> <LastPublished>20240423T101437</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240423T101436</CMSDate> <articleSource>FROM AACR 2024 </articleSource> <facebookInfo/> <meetingNumber>2976-24</meetingNumber> <byline>Nancy A. Melville</byline> <bylineText>NANCY A. MELVILLE</bylineText> <bylineFull>NANCY A. MELVILLE</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>A novel CD19 chimeric antigen responder (CAR) T-cell therapy administered in exceptionally low doses and designed to reinvigorate exhausted T-cells shows impres</metaDescription> <articlePDF/> <teaserImage/> <teaser>The IL-10-expressing Meta10-19 CAR T-cell therapy improved outcomes in B-ALL and DLBCL at doses 5% and lower of standard CAR T products.</teaser> <title>Tiny Doses of Metabolically Armed CAR T Show Benefits</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>hemn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">18</term> </publications> <sections> <term canonical="true">53</term> <term>39313</term> </sections> <topics> <term canonical="true">195</term> <term>179</term> <term>61821</term> <term>49434</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Tiny Doses of Metabolically Armed CAR T Show Benefits</title> <deck/> </itemMeta> <itemContent> <p> <span class="tag metaDescription">A novel CD19 chimeric antigen responder (CAR) T-cell therapy administered in exceptionally low doses and designed to reinvigorate exhausted T-cells shows impressive early results in the treatment of patients with relapsed or refractory (r/r) diffuse large B-cell lymphoma (DLBCL) and B-cell acute lymphoblastic leukemia (B-ALL).</span> </p> <p>“Our study showed a manageable safety profile in r/r DLBCL/B-ALL, with promising breakthrough efficacy of a 100% complete remission in all dose groups,” said first author Jingjing Ren, MD, PhD, associate director of research and development with Leman Biotech in Shenzhen, China. Dr. Ren <span class="Hyperlink"><a href="https://www.abstractsonline.com/pp8/#!/20272/presentation/11336">presented these findings</a></span> at the American Association for Cancer Research annual meeting held in San Diego.<br/><br/>While CD19 CAR T-cell therapy has been transformative in the treatment of relapsed B -cell hematological malignancies in recent years, more than half of patients relapse within a year because of inadequate CAR T persistence.<br/><br/>To address the problem, Dr. Ren and her colleagues developed a metabolically armed, interleukin (IL)-10-expressing CAR T-cell product called Meta10-19 for the treatment patients with r/r DLBCL or r/r B-ALL.<br/><br/>According to the authors, the IL-10-expressing CAR T-cells trigger “stem-like memory responses” in various lymphoid organs, which prompt a “robust tumor eradication and durable protection,” and hence, better persistence. <br/><br/>Preclinical studies in mice showed the Meta10-19 CAR T-cells exhibited substantially higher expansion of approximately 100-fold compared with a control CD19 CAR-T product.<br/><br/>Therefore, “we dramatically reduced the dose to approximately 1% to 5% of commercial products for the IL-10-expressing CD19 CAR-T for patients,” coauthor Yugang Guo, PhD, cofounder and president of Leman Biotech said in an interview.<br/><br/>For the ongoing, open-label clinical trial, 12 adult patients with r/r DLBCL or r/r B-ALL and confirmed CD19 expression at a hospital center in China were enrolled between December 2022 and November 2023 and treated in three cohorts, receiving doses that corresponded to 1%, 2.5%, or 5% of the doses of other commercialized CAR-T infusion products.<br/><br/>All patients also underwent lympho-depleting chemotherapy with cyclophosphamide and fludarabine prior to the CAR T-cell infusion. <br/><br/>Six patients had r/r DLBCL and the other six had r/r B-ALL; their median age was 47 and their median time since diagnosis was 1 year. <br/><br/>In the single-arm, intent-to-treat analysis, the treatment induced a complete remission in all 12 patients, as evaluated by PET-CT scan, nuclear magnetic resonance (NMR) spectroscopy, or minimal residual disease assessment of bone marrow. <br/><br/>The median time to best response was 1 month (range 0.5 to 2.2 months).<br/><br/>There were no cases of severe cytokine storm syndrome or neurotoxicity, which are among key limitations with current commercial CAR-T products.<br/><br/>All of the patients continued to have a complete remission at 3 months. Two of the 12 patients, both with B-ALL, experienced relapses, one after 4.7 months and the other at 8 months.<br/><br/>The authors reported that the first treated patient had maintained continuous remission as of 9 months.<br/><br/>In comparison with the much higher full doses of commercial CD19 CAR-T products, only about 50% of patients with DLBCL and 70% of B-ALL patients have been shown to achieve CR at 3 months, the authors reported.<br/><br/>“Our IL-10 expressing CAR-T sustains CR at 3 months post infusion in the context of not following allogeneic hematopoietic stem cell transplant, which suggests IL-10 expressing CAR-T is more resistant to relapse,” Dr. Guo said.<br/><br/>In terms of safety, six patients with DLBCL and four with B-ALL experienced grade 1 cytokine release syndrome (CRS), and two patients with B-ALL developed grade 2 CRS. There were no grade 3 or 4 CRS cases.<br/><br/>One patient with B-ALL developed grade 3 ICANS.<br/><br/>Grade 3-4 cytopenias occurred in most patients, but all were limited to no later than 90 days.<br/><br/>“We observed reduced CRS, with no level 3 or 4, or ICANS,” Dr. Guo said. “There was increased cytopenia, but still manageable, compared with commercial products.”<br/><br/>Of note, the Meta10-19 cells showed efficacy in the extremely low infusion doses even among patients with bulky mass (≥ 7.5 cm) of DLBCL, which is associated with an increased risk of relapse.<br/><br/>One patient had primary central nervous system lymphoma (PCNSL), a rare form of DLBCL that is known to have the worst prognosis of all non-Hodgkin lymphomas. <br/><br/>Due to the unique nature of CNS primary tumors, the CAR T-cell infusion dose was further reduced to 1% of the standard dose for the patient. <br/><br/>The patient maintained complete remission for more than 8 months before relapsing in periphery blood, but not in the CNS, Dr. Guo noted. <br/><br/>“Luckily, this relapse has been easily controlled by chemotherapy, and the patient is maintaining complete remission again now,” Dr. Guo said.<br/><br/></p> <h2>Mechanisms?</h2> <p>Dr. Guo noted that the mechanism believed to explain the improvements despite such low doses is that “IL-10-expressing CAR-T exhibits enhanced proliferation, cytotoxicity, and stem-like antitumor memory due to enhanced metabolic activities of oxidative phosphorylation.” <br/><br/>The authors noted that a key major factor limiting accessibility to CAR-T therapies is the lengthy production cycle and high costs; however, the “extremely low doses of 1% to 5% can significantly reduce the production cycle and cost of CAR T-cell therapies, increasing accessibility,” they wrote in a press statement.<br/><br/>Currently, more than 20 patients have achieved a CR overall, and studies with a larger cohort and longer follow-up are ongoing, Dr. Guo reported. <br/><br/>The research team plans to launch further clinical investigation this year into patients with solid tumors.<br/><br/>Commenting on the study, Hongbo Chi, PhD, the Robert G. Webster Endowed Chair in Immunology at St. Jude Children’s Research Hospital in Memphis, Tennessee, noted that, based on the abstract, “the effects are quite remarkable, considering the therapeutic efficacy observed even at the low dose.<br/><br/>“Results from more patients are needed to fully validate these findings, but the results to date are very encouraging,” he said. <br/><br/>The study was sponsored by Leman Biotech. Dr. Chi had no disclosures to report.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM AACR 2024

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Timing Is Everything: CAR T for Follicular Lymphoma

Article Type
Changed
Thu, 04/25/2024 - 09:37

 

For patients with relapsed/refractory follicular lymphoma (R/R FL), chimeric antigen responder (CAR) T-cell therapy offers the best chance of survival, despite its high rate of serious side effects and the need to travel to infusion centers. Alternatively, updated chemo+immunotherapy regimens and bispecific antibodies provide less rigorous but effective treatment options for low-risk patients who cannot tolerate or access CAR T.

“CAR T-cells offer patients with relapsed or refractory follicular lymphoma the most durable responses and improved chance of survival beyond all other available therapies. This holds true for a broad range of high-risk disease features in patients with relapsed or refractory FL. Furthermore, it accomplishes this with a single infusion, and a discrete toxicity that is predictable, reversible and manageable,” said Caron Jacobson, MD, MMSc, of the Dana-Farber Cancer Institute in Boston.

Presenting at the Great Debates & Updates Hematologic Malignancies conference, held April 5-6 in New York City, Dr. Jacobson argued that more patients with R/R FL should be treated with CAR T.

Jacobson_Caron_MA_web.jpg
Dr. Caron Jacobson


She cited follow-up results from the ZUMA-5 study indicating that patients with R/R FL treated with the CAR T axicabtagene ciloleucel (YESCARTA; Kite Pharma) have a median progression free survival (PFS) of 57.3 months and a complete response rate (CR) of 80%. Furthermore, the lymphoma-specific four-year PFS appears to be reaching a plateau, suggesting that some patients treated with the agent may be cured.

The most significant drawback of treatment with axicabtagene ciloleucel is cytokine release syndrome (CRS) and neurotoxicity, which occurred at grade three and higher in 6% and 15%, of ZUMA-5 participants, respectively.

Two newer studies of anti-CD-19 CAR T-cell therapy in R/R FL, tisagenlecleucel in ELARA and lisocabtagene maraleucel in TRANSCEND FL, suggest that other CAR T-cell treatments can be as effective as axicabtagene ciloleucel, but with fewer side effects.

At a median follow up of 29 months, CR among patients in the ELARA study was 68.1%, and the overall response rate (ORR) was 86.2%. Fewer than half of patients had any CRS, and none had grade three or higher. Only 10% of patients had serious neurologic events, with only 1% of these events rated as grade three or higher.

At a median of 18.1 months, patients in the TRANSCEND FL study had a CR of 94% and an ORR of 97%. Over 58% of patients had CRS but it was grade three or higher only 1% of the time (one patient); 15% of patients had neurologic toxicity, but it was grade three or higher only 2% of the time (three patients).

Dr. Jacobson’s opponent in the debate, Peter Martin, MD, of NewYork–Presbyterian Hospital, Weill Cornell Medicine in New York City, acknowledged the strong performance of CAR T in R/R FL patients but argued that they should be used only in a small subset of patients.

“About 20% of patients will experience an early recurrence or progression of diseases within 24 months (PoD-24) which is associated with worse outcomes. About half of those patients experienced transformation, so they have diffuse large B-cell lymphoma, and they’re getting CAR T-cells. In the end, only 10% of patients with follicular lymphoma are relapsed or refractory and should consider getting Car T-cell therapy,” said Dr. Martin, who focused the rest of his presentation on the best options for treating patients with indolent R/R FL who did not have PoD-24.

Martin_Peter_NY_web.jpg
Dr. Peter Martin


He said these patients may be able to avoid the side effects of CAR T and perform well when treated with lenalidomide rituximab (R2) or bispecific antibodies. Data from the MAGNIFY trial of patients with R/R FL indicate that patients treated with R2 who did not experience relapse less than 24 months after starting treatment and were not heavily refractory to rituximab achieved a median PFS of over 4 years, with grade 3 or higher adverse events occurring in 5% of patients or less.

Treatment with bispecific antibodies, although inferior in performance to CAR T-cell therapy, may offer durable responses in some R/R CL patients without the risk of side effects associated with CAR T.

Mosunetuzumab, a bispecific antibody that is currently approved for follicular lymphoma, is designed with step-up dosing to reduce cytokine release syndrome and “achieved a complete response rate of 60% and a median PFS that looks like it’s probably about two years,” said Dr. Martin, noting that some patients continue to do well after the 3-year mark and speculated that “there will be some really long-term responders.”

In addition to the possibly durable nature of bispecific antibodies, they induce cytokine release syndrome at a much lower rate than CAR T, and most side effects are manageable in an outpatient setting, “usually just with Tylenol occasionally with a dose of steroids,” said Dr. Martin.

He contrasted this response with CAR T-cell therapy, which requires referral and travel to a specialized center for at least 1 month around the time of therapy.

Despite the differences of opinion between the presenters about whether CAR T should be used more or less in R/R FL, essentially the two specialists were making recommendations for different patient groups.

Dr. Jacobson observed that “Dr. Martin is looking at the 80% of people who do really well with follicular lymphoma." Those are the people who don’t require a third line of therapy. They are the people who don’t have PoD-24. I’m looking at the 20% of people who either do require a third line of therapy or who do have PoD-24, and we’re not treating nearly enough of those patients with follicular lymphoma.

“We’re actually arguing about treatment strategies for different populations of patients. And I think ultimately, we agree more than we disagree in the end,” she concluded.

The notion that CAR T, chemotherapy, and bispecific antibodies all have a place in treating R/R FL patients is supported by Charalambos (Babis) Andreadis, MD, hematologist at the University of California San Francisco’s Helen Diller Family Comprehensive Care Center. “If I had a patient with follicular who relapsed 24 months or later after primary therapy and had active disease that needed treatment, most providers would do a lenalidomide-based or chemo-based regimen. Down the line either bispecific or CAR T would be appropriate in third line,” said Dr. Andreadis.

However, he noted,“for someone who is an early progressor, I would similarly not be able to use either [chemotherapy or bispecific antibodies] in second line [therapy] but would definitely think that early CART would be a good option to consider given the longevity of the observed responses so far.”

Dr. Martin disclosed ties with AbbVie, AstraZeneca, BeiGene, Daiichi Sankyo, Epizyme, Genentech, Janssen, Merck, and PeproMene. Dr. Jacobson reported relationships with AbbVie, Abintus Bio, ADC Therapeutics, Appia Bio, AstraZeneca, BMS/Celgene, Caribou Bio, Daiichi Sankyo, ImmPACT Bio, Ipsen, Janssen, Kite/Gilead, MorphoSys, Novartis, Sana, Synthekine, Kite/Gilead, and Pfizer. Dr. Andreadis had no disclosures.

Publications
Topics
Sections

 

For patients with relapsed/refractory follicular lymphoma (R/R FL), chimeric antigen responder (CAR) T-cell therapy offers the best chance of survival, despite its high rate of serious side effects and the need to travel to infusion centers. Alternatively, updated chemo+immunotherapy regimens and bispecific antibodies provide less rigorous but effective treatment options for low-risk patients who cannot tolerate or access CAR T.

“CAR T-cells offer patients with relapsed or refractory follicular lymphoma the most durable responses and improved chance of survival beyond all other available therapies. This holds true for a broad range of high-risk disease features in patients with relapsed or refractory FL. Furthermore, it accomplishes this with a single infusion, and a discrete toxicity that is predictable, reversible and manageable,” said Caron Jacobson, MD, MMSc, of the Dana-Farber Cancer Institute in Boston.

Presenting at the Great Debates & Updates Hematologic Malignancies conference, held April 5-6 in New York City, Dr. Jacobson argued that more patients with R/R FL should be treated with CAR T.

Jacobson_Caron_MA_web.jpg
Dr. Caron Jacobson


She cited follow-up results from the ZUMA-5 study indicating that patients with R/R FL treated with the CAR T axicabtagene ciloleucel (YESCARTA; Kite Pharma) have a median progression free survival (PFS) of 57.3 months and a complete response rate (CR) of 80%. Furthermore, the lymphoma-specific four-year PFS appears to be reaching a plateau, suggesting that some patients treated with the agent may be cured.

The most significant drawback of treatment with axicabtagene ciloleucel is cytokine release syndrome (CRS) and neurotoxicity, which occurred at grade three and higher in 6% and 15%, of ZUMA-5 participants, respectively.

Two newer studies of anti-CD-19 CAR T-cell therapy in R/R FL, tisagenlecleucel in ELARA and lisocabtagene maraleucel in TRANSCEND FL, suggest that other CAR T-cell treatments can be as effective as axicabtagene ciloleucel, but with fewer side effects.

At a median follow up of 29 months, CR among patients in the ELARA study was 68.1%, and the overall response rate (ORR) was 86.2%. Fewer than half of patients had any CRS, and none had grade three or higher. Only 10% of patients had serious neurologic events, with only 1% of these events rated as grade three or higher.

At a median of 18.1 months, patients in the TRANSCEND FL study had a CR of 94% and an ORR of 97%. Over 58% of patients had CRS but it was grade three or higher only 1% of the time (one patient); 15% of patients had neurologic toxicity, but it was grade three or higher only 2% of the time (three patients).

Dr. Jacobson’s opponent in the debate, Peter Martin, MD, of NewYork–Presbyterian Hospital, Weill Cornell Medicine in New York City, acknowledged the strong performance of CAR T in R/R FL patients but argued that they should be used only in a small subset of patients.

“About 20% of patients will experience an early recurrence or progression of diseases within 24 months (PoD-24) which is associated with worse outcomes. About half of those patients experienced transformation, so they have diffuse large B-cell lymphoma, and they’re getting CAR T-cells. In the end, only 10% of patients with follicular lymphoma are relapsed or refractory and should consider getting Car T-cell therapy,” said Dr. Martin, who focused the rest of his presentation on the best options for treating patients with indolent R/R FL who did not have PoD-24.

Martin_Peter_NY_web.jpg
Dr. Peter Martin


He said these patients may be able to avoid the side effects of CAR T and perform well when treated with lenalidomide rituximab (R2) or bispecific antibodies. Data from the MAGNIFY trial of patients with R/R FL indicate that patients treated with R2 who did not experience relapse less than 24 months after starting treatment and were not heavily refractory to rituximab achieved a median PFS of over 4 years, with grade 3 or higher adverse events occurring in 5% of patients or less.

Treatment with bispecific antibodies, although inferior in performance to CAR T-cell therapy, may offer durable responses in some R/R CL patients without the risk of side effects associated with CAR T.

Mosunetuzumab, a bispecific antibody that is currently approved for follicular lymphoma, is designed with step-up dosing to reduce cytokine release syndrome and “achieved a complete response rate of 60% and a median PFS that looks like it’s probably about two years,” said Dr. Martin, noting that some patients continue to do well after the 3-year mark and speculated that “there will be some really long-term responders.”

In addition to the possibly durable nature of bispecific antibodies, they induce cytokine release syndrome at a much lower rate than CAR T, and most side effects are manageable in an outpatient setting, “usually just with Tylenol occasionally with a dose of steroids,” said Dr. Martin.

He contrasted this response with CAR T-cell therapy, which requires referral and travel to a specialized center for at least 1 month around the time of therapy.

Despite the differences of opinion between the presenters about whether CAR T should be used more or less in R/R FL, essentially the two specialists were making recommendations for different patient groups.

Dr. Jacobson observed that “Dr. Martin is looking at the 80% of people who do really well with follicular lymphoma." Those are the people who don’t require a third line of therapy. They are the people who don’t have PoD-24. I’m looking at the 20% of people who either do require a third line of therapy or who do have PoD-24, and we’re not treating nearly enough of those patients with follicular lymphoma.

“We’re actually arguing about treatment strategies for different populations of patients. And I think ultimately, we agree more than we disagree in the end,” she concluded.

The notion that CAR T, chemotherapy, and bispecific antibodies all have a place in treating R/R FL patients is supported by Charalambos (Babis) Andreadis, MD, hematologist at the University of California San Francisco’s Helen Diller Family Comprehensive Care Center. “If I had a patient with follicular who relapsed 24 months or later after primary therapy and had active disease that needed treatment, most providers would do a lenalidomide-based or chemo-based regimen. Down the line either bispecific or CAR T would be appropriate in third line,” said Dr. Andreadis.

However, he noted,“for someone who is an early progressor, I would similarly not be able to use either [chemotherapy or bispecific antibodies] in second line [therapy] but would definitely think that early CART would be a good option to consider given the longevity of the observed responses so far.”

Dr. Martin disclosed ties with AbbVie, AstraZeneca, BeiGene, Daiichi Sankyo, Epizyme, Genentech, Janssen, Merck, and PeproMene. Dr. Jacobson reported relationships with AbbVie, Abintus Bio, ADC Therapeutics, Appia Bio, AstraZeneca, BMS/Celgene, Caribou Bio, Daiichi Sankyo, ImmPACT Bio, Ipsen, Janssen, Kite/Gilead, MorphoSys, Novartis, Sana, Synthekine, Kite/Gilead, and Pfizer. Dr. Andreadis had no disclosures.

 

For patients with relapsed/refractory follicular lymphoma (R/R FL), chimeric antigen responder (CAR) T-cell therapy offers the best chance of survival, despite its high rate of serious side effects and the need to travel to infusion centers. Alternatively, updated chemo+immunotherapy regimens and bispecific antibodies provide less rigorous but effective treatment options for low-risk patients who cannot tolerate or access CAR T.

“CAR T-cells offer patients with relapsed or refractory follicular lymphoma the most durable responses and improved chance of survival beyond all other available therapies. This holds true for a broad range of high-risk disease features in patients with relapsed or refractory FL. Furthermore, it accomplishes this with a single infusion, and a discrete toxicity that is predictable, reversible and manageable,” said Caron Jacobson, MD, MMSc, of the Dana-Farber Cancer Institute in Boston.

Presenting at the Great Debates & Updates Hematologic Malignancies conference, held April 5-6 in New York City, Dr. Jacobson argued that more patients with R/R FL should be treated with CAR T.

Jacobson_Caron_MA_web.jpg
Dr. Caron Jacobson


She cited follow-up results from the ZUMA-5 study indicating that patients with R/R FL treated with the CAR T axicabtagene ciloleucel (YESCARTA; Kite Pharma) have a median progression free survival (PFS) of 57.3 months and a complete response rate (CR) of 80%. Furthermore, the lymphoma-specific four-year PFS appears to be reaching a plateau, suggesting that some patients treated with the agent may be cured.

The most significant drawback of treatment with axicabtagene ciloleucel is cytokine release syndrome (CRS) and neurotoxicity, which occurred at grade three and higher in 6% and 15%, of ZUMA-5 participants, respectively.

Two newer studies of anti-CD-19 CAR T-cell therapy in R/R FL, tisagenlecleucel in ELARA and lisocabtagene maraleucel in TRANSCEND FL, suggest that other CAR T-cell treatments can be as effective as axicabtagene ciloleucel, but with fewer side effects.

At a median follow up of 29 months, CR among patients in the ELARA study was 68.1%, and the overall response rate (ORR) was 86.2%. Fewer than half of patients had any CRS, and none had grade three or higher. Only 10% of patients had serious neurologic events, with only 1% of these events rated as grade three or higher.

At a median of 18.1 months, patients in the TRANSCEND FL study had a CR of 94% and an ORR of 97%. Over 58% of patients had CRS but it was grade three or higher only 1% of the time (one patient); 15% of patients had neurologic toxicity, but it was grade three or higher only 2% of the time (three patients).

Dr. Jacobson’s opponent in the debate, Peter Martin, MD, of NewYork–Presbyterian Hospital, Weill Cornell Medicine in New York City, acknowledged the strong performance of CAR T in R/R FL patients but argued that they should be used only in a small subset of patients.

“About 20% of patients will experience an early recurrence or progression of diseases within 24 months (PoD-24) which is associated with worse outcomes. About half of those patients experienced transformation, so they have diffuse large B-cell lymphoma, and they’re getting CAR T-cells. In the end, only 10% of patients with follicular lymphoma are relapsed or refractory and should consider getting Car T-cell therapy,” said Dr. Martin, who focused the rest of his presentation on the best options for treating patients with indolent R/R FL who did not have PoD-24.

Martin_Peter_NY_web.jpg
Dr. Peter Martin


He said these patients may be able to avoid the side effects of CAR T and perform well when treated with lenalidomide rituximab (R2) or bispecific antibodies. Data from the MAGNIFY trial of patients with R/R FL indicate that patients treated with R2 who did not experience relapse less than 24 months after starting treatment and were not heavily refractory to rituximab achieved a median PFS of over 4 years, with grade 3 or higher adverse events occurring in 5% of patients or less.

Treatment with bispecific antibodies, although inferior in performance to CAR T-cell therapy, may offer durable responses in some R/R CL patients without the risk of side effects associated with CAR T.

Mosunetuzumab, a bispecific antibody that is currently approved for follicular lymphoma, is designed with step-up dosing to reduce cytokine release syndrome and “achieved a complete response rate of 60% and a median PFS that looks like it’s probably about two years,” said Dr. Martin, noting that some patients continue to do well after the 3-year mark and speculated that “there will be some really long-term responders.”

In addition to the possibly durable nature of bispecific antibodies, they induce cytokine release syndrome at a much lower rate than CAR T, and most side effects are manageable in an outpatient setting, “usually just with Tylenol occasionally with a dose of steroids,” said Dr. Martin.

He contrasted this response with CAR T-cell therapy, which requires referral and travel to a specialized center for at least 1 month around the time of therapy.

Despite the differences of opinion between the presenters about whether CAR T should be used more or less in R/R FL, essentially the two specialists were making recommendations for different patient groups.

Dr. Jacobson observed that “Dr. Martin is looking at the 80% of people who do really well with follicular lymphoma." Those are the people who don’t require a third line of therapy. They are the people who don’t have PoD-24. I’m looking at the 20% of people who either do require a third line of therapy or who do have PoD-24, and we’re not treating nearly enough of those patients with follicular lymphoma.

“We’re actually arguing about treatment strategies for different populations of patients. And I think ultimately, we agree more than we disagree in the end,” she concluded.

The notion that CAR T, chemotherapy, and bispecific antibodies all have a place in treating R/R FL patients is supported by Charalambos (Babis) Andreadis, MD, hematologist at the University of California San Francisco’s Helen Diller Family Comprehensive Care Center. “If I had a patient with follicular who relapsed 24 months or later after primary therapy and had active disease that needed treatment, most providers would do a lenalidomide-based or chemo-based regimen. Down the line either bispecific or CAR T would be appropriate in third line,” said Dr. Andreadis.

However, he noted,“for someone who is an early progressor, I would similarly not be able to use either [chemotherapy or bispecific antibodies] in second line [therapy] but would definitely think that early CART would be a good option to consider given the longevity of the observed responses so far.”

Dr. Martin disclosed ties with AbbVie, AstraZeneca, BeiGene, Daiichi Sankyo, Epizyme, Genentech, Janssen, Merck, and PeproMene. Dr. Jacobson reported relationships with AbbVie, Abintus Bio, ADC Therapeutics, Appia Bio, AstraZeneca, BMS/Celgene, Caribou Bio, Daiichi Sankyo, ImmPACT Bio, Ipsen, Janssen, Kite/Gilead, MorphoSys, Novartis, Sana, Synthekine, Kite/Gilead, and Pfizer. Dr. Andreadis had no disclosures.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167746</fileName> <TBEID>0C04FA4C.SIG</TBEID> <TBUniqueIdentifier>MD_0C04FA4C</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>CAR T RR FL</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240418T112914</QCDate> <firstPublished>20240418T114215</firstPublished> <LastPublished>20240418T114215</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240418T114215</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Myles Starr</byline> <bylineText>MYLES STARR</bylineText> <bylineFull>MYLES STARR</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>For patients with relapsed/refractory follicular lymphoma (R/R FL), chimeric antigen responder (CAR) T-cell therapy offers the best chance of survival, despite </metaDescription> <articlePDF/> <teaserImage>301136</teaserImage> <teaser>Experts debated about whether and when patients with relapsed/refractory follicular lymphoma should receive CAR T-cell therapy.</teaser> <title>Timing Is Everything: CAR T for Follicular Lymphoma</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>hemn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">18</term> </publications> <sections> <term canonical="true">53</term> <term>39313</term> </sections> <topics> <term canonical="true">49434</term> <term>195</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/2401284b.jpg</altRep> <description role="drol:caption">Dr. Caron Jacobson</description> <description role="drol:credit">Dana-Farber Cancer Institute</description> </link> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/2401284c.jpg</altRep> <description role="drol:caption">Dr. Peter Martin</description> <description role="drol:credit">NY Presbyterian Hospital, Weill Cornell Medicine</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Timing Is Everything: CAR T for Follicular Lymphoma</title> <deck/> </itemMeta> <itemContent> <p> <span class="tag metaDescription">For patients with relapsed/refractory follicular lymphoma (R/R FL), chimeric antigen responder (CAR) T-cell therapy offers the best chance of survival, despite its high rate of serious side effects and the need to travel to infusion centers. Alternatively, updated chemo+immunotherapy regimens and bispecific antibodies provide less rigorous but effective treatment options for low-risk patients who cannot tolerate or access CAR T.</span> </p> <p>“CAR T-cells offer patients with relapsed or refractory follicular lymphoma the most durable responses and improved chance of survival beyond all other available therapies. This holds true for a broad range of high-risk disease features in patients with relapsed or refractory FL. Furthermore, it accomplishes this with a single infusion, and a discrete toxicity that is predictable, reversible and manageable,” said Caron Jacobson, MD, MMSc, of the Dana-Farber Cancer Institute in Boston. <br/><br/>Presenting at the Great Debates &amp; Updates Hematologic Malignancies conference, held April 5-6 in New York City, Dr. Jacobson argued that more patients with R/R FL should be treated with CAR T.[[{"fid":"301136","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Caron Jacobson, MD, MMSc, Medical Director, Cell Therapy Program, Dana-Farber Cancer Institute, Boston, MA","field_file_image_credit[und][0][value]":"Dana-Farber Cancer Institute","field_file_image_caption[und][0][value]":"Dr. Caron Jacobson"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_left"}}]]<br/><br/>She cited follow-up results from the <span class="Hyperlink"><a href="https://ashpublications.org/blood/article/142/Supplement%201/4869/501128/An-Updated-Comparison-of-Clinical-Outcomes-from-4">ZUMA-5</a></span> study indicating that patients with R/R FL treated with the CAR T axicabtagene ciloleucel (YESCARTA; Kite Pharma) have a median progression free survival (PFS) of 57.3 months and a complete response rate (CR) of 80%. Furthermore, the lymphoma-specific four-year PFS appears to be reaching a plateau, suggesting that some patients treated with the agent may be cured.<br/><br/>The most significant drawback of treatment with axicabtagene ciloleucel is cytokine release syndrome (CRS) and neurotoxicity, which occurred at grade three and higher in 6% and 15%, of ZUMA-5 participants, respectively.<br/><br/>Two newer studies of anti-CD-19 CAR T-cell therapy in R/R FL, tisagenlecleucel in <span class="Hyperlink"><a href="https://ashpublications.org/blood/article/doi/10.1182/blood.2023021567/507096/Durable-Response-After-Tisagenlecleucel-in-Adults">ELARA</a></span> and lisocabtagene maraleucel in <span class="Hyperlink"><a href="https://ash.confex.com/ash/2023/webprogram/Paper179474.html">TRANSCEND FL</a></span>, suggest that other CAR T-cell treatments can be as effective as axicabtagene ciloleucel, but with fewer side effects. <br/><br/>At a median follow up of 29 months, CR among patients in the ELARA study was 68.1%, and the overall response rate (ORR) was 86.2%. Fewer than half of patients had any CRS, and none had grade three or higher. Only 10% of patients had serious neurologic events, with only 1% of these events rated as grade three or higher. <br/><br/>At a median of 18.1 months, patients in the TRANSCEND FL study had a CR of 94% and an ORR or 97%. Over 58% of patients had CRS but it was grade three or higher only 1% of the time (one patient); ,15% of patients had neurologic toxicity, but it was grade three or higher only 2% of the time (three patients). <br/><br/>Dr. Jacobson’s opponent in the debate, Peter Martin, MD, of New York Presbyterian Hospital, Weill Cornell Medicine in New York City, acknowledged the strong performance of CAR T in R/R FL patients but argued that they should be used only in a small subset of patients. <br/><br/>“About 20% of patients will experience an early recurrence or progression of diseases within 24 months (PoD-24) which is associated with worse outcomes. About half of those patients experienced transformation, so they have diffuse large B-cell lymphoma, and they’re getting CAR T-cells. In the end, only 10% of patients with follicular lymphoma are relapsed or refractory and should consider getting Car T-cell therapy,” said Dr. Martin, who focused the rest of his presentation on the best options for treating patients with indolent R/R FL who did not have PoD-24.[[{"fid":"301137","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Peter Martin, MD, lymphoma specialist, New York Presbyterian/Weill Cornell Medidine","field_file_image_credit[und][0][value]":"NY Presbyterian Hospital, Weill Cornell Medicine","field_file_image_caption[und][0][value]":"Dr. Peter Martin"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]<br/><br/>He said these patients may be able to avoid the side effects of CAR T and perform well when treated with lenalidomide rituximab (R2) or bispecific antibodies. Data from the <span class="Hyperlink"><a href="https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9430939/">MAGNIFY</a></span> trial of patients with R/R FL indicate that patients treated with R2 who did not experience relapse less than 24 months after starting treatment and were not heavily refractory to rituximab achieved a median PFS of over 4 years, with grade 3 or higher adverse events occurring in 5% of patients or less. <br/><br/>Treatment with bispecific antibodies, although inferior in performance to CAR T-cell therapy, may offer durable responses in some R/R CL patients without the risk of side effects associated with CAR T. <br/><br/><span class="Hyperlink"><a href="https://www.thelancet.com/journals/lanonc/article/PIIS1470-2045(22)00335-7/abstract">Mosunetuzumab</a></span> a bispecific antibody that is currently approved for follicular lymphoma, is designed with step-up dosing to reduce cytokine release syndrome and “achieved a complete response rate of 60% and a median PFS that looks like it’s probably about two years,” said Dr. Martin, noting that some patients continue to do well after the 3-year mark and speculated that “there will be some really long-term responders.”<br/><br/>In addition to the possibly durable nature of bispecific antibodies, they induce cytokine release syndrome at a much lower rate than CAR T, and most side effects are manageable in an outpatient setting, “usually just with Tylenol occasionally with a dose of steroids,” said Dr. Martin. <br/><br/>He contrasted this response with CAR T-cell therapy, which requires referral and travel to a specialized center for at least 1 month around the time of therapy. <br/><br/>Despite the differences of opinion between the presenters about whether CAR T should be used more or less in R/R FL, essentially the two specialists were making recommendations for different patient groups. <br/><br/>Dr. Jacobson observed that “Dr. Martin is looking at the 80% of people who do really well with follicular lymphoma. Those are the people who don’t require a third line of therapy. They are the people who don’t have PoD-24. I’m looking at the 20% of people who either do require a third line of therapy or who do have PoD-24, and we’re not treating nearly enough of those patients with follicular lymphoma.<br/><br/>“We’re actually arguing about treatment strategies for different populations of patients. And I think ultimately, we agree more than we disagree in the end,” she concluded.<br/><br/>The notion that CAR T, chemotherapy, and bispecific antibodies all have a place in treating R/R FL patients is supported by Charalambos (Babis) Andreadis MD, hematologist at the University of California San Francisco’s Helen Diller Family Comprehensive Care Center. “If I had a patient with follicular who relapsed 24 months or later after primary therapy and had active disease that needed treatment, most providers would do a lenalidomide-based or chemo-based regimen. Down the line either bispecific or CAR T would be appropriate in third line,” said Dr. Andreadis. <br/><br/>However, he noted,“for someone who is an early progressor, I would similarly not be able to use either [chemotherapy or bispecific antibodies] in second line [therapy] but would definitely think that early CART would be a good option to consider given the longevity of the observed responses so far.”<br/><br/>Dr. Martin disclosed ties with Abbvie, AstraZeneca, Beigene, Daiichi Sankyo, Epizyme, Genentech, Janssen, Merck, and Pepromene. Dr. Jacobson reported relationships with Abbvie, Abintus Bio, ADC Therapeutics, Appia Bio, AstraZeneca, BMS/Celgene, Caribou Bio, Daiichi Sankyo, ImmPACT Bio, Ipsen, Janssen, Kite/Gilead, Morphosys, Novartis, Sana, Synthekine, Kite/Gilead, and Pfizer. Dr. Andreadis had no disclosures.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Most Targeted Cancer Drugs Lack Substantial Clinical Benefit

Article Type
Changed
Tue, 04/23/2024 - 17:03

 

TOPLINE:

An analysis of molecular-targeted cancer drug therapies recently approved in the United States found that fewer than one-third demonstrated substantial clinical benefits at the time of approval.

METHODOLOGY:

  • The strength and quality of evidence supporting genome-targeted cancer drug approvals vary. A big reason is the growing number of cancer drug approvals based on surrogate endpoints, such as disease-free and progression-free survival, instead of clinical endpoints, such as overall survival or quality of life. The US Food and Drug Administration (FDA) has also approved genome-targeted cancer drugs based on phase 1 or single-arm trials.
  • Given these less rigorous considerations for approval, “the validity and value of the targets and surrogate measures underlying FDA genome-targeted cancer drug approvals are uncertain,” the researchers explained.
  • In the current analysis, researchers assessed the validity of the molecular targets as well as the clinical benefits of genome-targeted cancer drugs approved in the United States from 2015 to 2022 based on results from pivotal trials.
  • The researchers evaluated the strength of evidence supporting molecular targetability using the European Society for Medical Oncology (ESMO) Scale for Clinical Actionability of Molecular Targets (ESCAT) and the clinical benefit using the ESMO–Magnitude of Clinical Benefit Scale (ESMO-MCBS).
  • The authors defined a substantial clinical benefit as an A or B grade for curative intent and a 4 or 5 for noncurative intent. High-benefit genomic-based cancer treatments were defined as those associated with a substantial clinical benefit (ESMO-MCBS) and that qualified as ESCAT category level I-A (a clinical benefit based on prospective randomized data) or I-B (prospective nonrandomized data).

TAKEAWAY:

  • The analyses focused on 50 molecular-targeted cancer drugs covering 84 indications. Of which, 45 indications (54%) were approved based on phase 1 or 2 pivotal trials, 45 (54%) were supported by single-arm pivotal trials and the remaining 39 (46%) by randomized trial, and 48 (57%) were approved based on subgroup analyses.
  • Among the 84 indications, more than half (55%) of the pivotal trials supporting approval used overall response rate as a primary endpoint, 31% used progression-free survival, and 6% used disease-free survival. Only seven indications (8%) were supported by pivotal trials demonstrating an improvement in overall survival.
  • Among the 84 trials, 24 (29%) met the ESMO-MCBS threshold for substantial clinical benefit.
  • Overall, when combining all ratings, only 24 of the 84 indications (29%) were considered high-benefit genomic-based cancer treatments.

IN PRACTICE:

“We applied the ESMO-MCBS and ESCAT value frameworks to identify therapies and molecular targets providing high clinical value that should be widely available to patients” and “found that drug indications supported by these characteristics represent a minority of cancer drug approvals in recent years,” the authors said. Using these value frameworks could help payers, governments, and individual patients “prioritize the availability of high-value molecular-targeted therapies.”

SOURCE:

The study, with first author Ariadna Tibau, MD, PhD, Brigham and Women’s Hospital and Harvard Medical School, Boston, was published online in JAMA Oncology.

LIMITATIONS:

The study evaluated only trials that supported regulatory approval and did not include outcomes of postapproval clinical studies, which could lead to changes in ESMO-MCBS grades and ESCAT levels of evidence over time.

DISCLOSURES:

The study was funded by the Kaiser Permanente Institute for Health Policy, Arnold Ventures, and the Commonwealth Fund. The authors had no relevant disclosures.

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

 

TOPLINE:

An analysis of molecular-targeted cancer drug therapies recently approved in the United States found that fewer than one-third demonstrated substantial clinical benefits at the time of approval.

METHODOLOGY:

  • The strength and quality of evidence supporting genome-targeted cancer drug approvals vary. A big reason is the growing number of cancer drug approvals based on surrogate endpoints, such as disease-free and progression-free survival, instead of clinical endpoints, such as overall survival or quality of life. The US Food and Drug Administration (FDA) has also approved genome-targeted cancer drugs based on phase 1 or single-arm trials.
  • Given these less rigorous considerations for approval, “the validity and value of the targets and surrogate measures underlying FDA genome-targeted cancer drug approvals are uncertain,” the researchers explained.
  • In the current analysis, researchers assessed the validity of the molecular targets as well as the clinical benefits of genome-targeted cancer drugs approved in the United States from 2015 to 2022 based on results from pivotal trials.
  • The researchers evaluated the strength of evidence supporting molecular targetability using the European Society for Medical Oncology (ESMO) Scale for Clinical Actionability of Molecular Targets (ESCAT) and the clinical benefit using the ESMO–Magnitude of Clinical Benefit Scale (ESMO-MCBS).
  • The authors defined a substantial clinical benefit as an A or B grade for curative intent and a 4 or 5 for noncurative intent. High-benefit genomic-based cancer treatments were defined as those associated with a substantial clinical benefit (ESMO-MCBS) and that qualified as ESCAT category level I-A (a clinical benefit based on prospective randomized data) or I-B (prospective nonrandomized data).

TAKEAWAY:

  • The analyses focused on 50 molecular-targeted cancer drugs covering 84 indications. Of which, 45 indications (54%) were approved based on phase 1 or 2 pivotal trials, 45 (54%) were supported by single-arm pivotal trials and the remaining 39 (46%) by randomized trial, and 48 (57%) were approved based on subgroup analyses.
  • Among the 84 indications, more than half (55%) of the pivotal trials supporting approval used overall response rate as a primary endpoint, 31% used progression-free survival, and 6% used disease-free survival. Only seven indications (8%) were supported by pivotal trials demonstrating an improvement in overall survival.
  • Among the 84 trials, 24 (29%) met the ESMO-MCBS threshold for substantial clinical benefit.
  • Overall, when combining all ratings, only 24 of the 84 indications (29%) were considered high-benefit genomic-based cancer treatments.

IN PRACTICE:

“We applied the ESMO-MCBS and ESCAT value frameworks to identify therapies and molecular targets providing high clinical value that should be widely available to patients” and “found that drug indications supported by these characteristics represent a minority of cancer drug approvals in recent years,” the authors said. Using these value frameworks could help payers, governments, and individual patients “prioritize the availability of high-value molecular-targeted therapies.”

SOURCE:

The study, with first author Ariadna Tibau, MD, PhD, Brigham and Women’s Hospital and Harvard Medical School, Boston, was published online in JAMA Oncology.

LIMITATIONS:

The study evaluated only trials that supported regulatory approval and did not include outcomes of postapproval clinical studies, which could lead to changes in ESMO-MCBS grades and ESCAT levels of evidence over time.

DISCLOSURES:

The study was funded by the Kaiser Permanente Institute for Health Policy, Arnold Ventures, and the Commonwealth Fund. The authors had no relevant disclosures.

A version of this article appeared on Medscape.com.

 

TOPLINE:

An analysis of molecular-targeted cancer drug therapies recently approved in the United States found that fewer than one-third demonstrated substantial clinical benefits at the time of approval.

METHODOLOGY:

  • The strength and quality of evidence supporting genome-targeted cancer drug approvals vary. A big reason is the growing number of cancer drug approvals based on surrogate endpoints, such as disease-free and progression-free survival, instead of clinical endpoints, such as overall survival or quality of life. The US Food and Drug Administration (FDA) has also approved genome-targeted cancer drugs based on phase 1 or single-arm trials.
  • Given these less rigorous considerations for approval, “the validity and value of the targets and surrogate measures underlying FDA genome-targeted cancer drug approvals are uncertain,” the researchers explained.
  • In the current analysis, researchers assessed the validity of the molecular targets as well as the clinical benefits of genome-targeted cancer drugs approved in the United States from 2015 to 2022 based on results from pivotal trials.
  • The researchers evaluated the strength of evidence supporting molecular targetability using the European Society for Medical Oncology (ESMO) Scale for Clinical Actionability of Molecular Targets (ESCAT) and the clinical benefit using the ESMO–Magnitude of Clinical Benefit Scale (ESMO-MCBS).
  • The authors defined a substantial clinical benefit as an A or B grade for curative intent and a 4 or 5 for noncurative intent. High-benefit genomic-based cancer treatments were defined as those associated with a substantial clinical benefit (ESMO-MCBS) and that qualified as ESCAT category level I-A (a clinical benefit based on prospective randomized data) or I-B (prospective nonrandomized data).

TAKEAWAY:

  • The analyses focused on 50 molecular-targeted cancer drugs covering 84 indications. Of which, 45 indications (54%) were approved based on phase 1 or 2 pivotal trials, 45 (54%) were supported by single-arm pivotal trials and the remaining 39 (46%) by randomized trial, and 48 (57%) were approved based on subgroup analyses.
  • Among the 84 indications, more than half (55%) of the pivotal trials supporting approval used overall response rate as a primary endpoint, 31% used progression-free survival, and 6% used disease-free survival. Only seven indications (8%) were supported by pivotal trials demonstrating an improvement in overall survival.
  • Among the 84 trials, 24 (29%) met the ESMO-MCBS threshold for substantial clinical benefit.
  • Overall, when combining all ratings, only 24 of the 84 indications (29%) were considered high-benefit genomic-based cancer treatments.

IN PRACTICE:

“We applied the ESMO-MCBS and ESCAT value frameworks to identify therapies and molecular targets providing high clinical value that should be widely available to patients” and “found that drug indications supported by these characteristics represent a minority of cancer drug approvals in recent years,” the authors said. Using these value frameworks could help payers, governments, and individual patients “prioritize the availability of high-value molecular-targeted therapies.”

SOURCE:

The study, with first author Ariadna Tibau, MD, PhD, Brigham and Women’s Hospital and Harvard Medical School, Boston, was published online in JAMA Oncology.

LIMITATIONS:

The study evaluated only trials that supported regulatory approval and did not include outcomes of postapproval clinical studies, which could lead to changes in ESMO-MCBS grades and ESCAT levels of evidence over time.

DISCLOSURES:

The study was funded by the Kaiser Permanente Institute for Health Policy, Arnold Ventures, and the Commonwealth Fund. The authors had no relevant disclosures.

A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167758</fileName> <TBEID>0C04FA8F.SIG</TBEID> <TBUniqueIdentifier>MD_0C04FA8F</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240417T163556</QCDate> <firstPublished>20240417T163834</firstPublished> <LastPublished>20240417T163835</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240417T163834</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Megan Brooks</byline> <bylineText>MEGAN BROOKS</bylineText> <bylineFull>MEGAN BROOKS</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>An analysis of molecular-targeted cancer drug therapies recently approved in the United States found that fewer than one-third demonstrated substantial clinical</metaDescription> <articlePDF/> <teaserImage/> <teaser>Researchers assess validity of the molecular targets and clinical benefits of genome-targeted cancer drugs approved in the United States from 2015 to 2022.</teaser> <title>Most Targeted Cancer Drugs Lack Substantial Clinical Benefit</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>oncr</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>ob</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>chph</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>skin</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>nr</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Neurology Reviews</journalTitle> <journalFullTitle>Neurology Reviews</journalFullTitle> <copyrightStatement>2018 Frontline Medical Communications Inc.,</copyrightStatement> </publicationData> <publicationData> <publicationCode>hemn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>GIHOLD</publicationCode> <pubIssueName>January 2014</pubIssueName> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle/> <journalFullTitle/> <copyrightStatement/> </publicationData> </publications_g> <publications> <term canonical="true">31</term> <term>23</term> <term>6</term> <term>13</term> <term>22</term> <term>18</term> </publications> <sections> <term>37225</term> <term>39313</term> <term canonical="true">27970</term> </sections> <topics> <term>192</term> <term>198</term> <term>61821</term> <term>59244</term> <term>67020</term> <term>214</term> <term>217</term> <term>61642</term> <term>221</term> <term>232</term> <term>238</term> <term>240</term> <term>242</term> <term>39570</term> <term>244</term> <term>256</term> <term>245</term> <term>270</term> <term canonical="true">278</term> <term>280</term> <term>292</term> <term>31848</term> <term>271</term> <term>27442</term> <term>38029</term> <term>179</term> <term>178</term> <term>181</term> <term>59374</term> <term>195</term> <term>196</term> <term>197</term> <term>37637</term> <term>233</term> <term>243</term> <term>49434</term> <term>303</term> <term>250</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Most Targeted Cancer Drugs Lack Substantial Clinical Benefit</title> <deck/> </itemMeta> <itemContent> <h2>TOPLINE:</h2> <p> <span class="tag metaDescription">An analysis of molecular-targeted cancer drug therapies recently approved in the United States found that fewer than one-third demonstrated substantial clinical benefits at the time of approval.</span> </p> <h2>METHODOLOGY:</h2> <ul class="body"> <li>The strength and quality of evidence supporting genome-targeted cancer drug approvals vary. A big reason is the growing number of cancer drug approvals based on surrogate endpoints, such as disease-free and progression-free survival, instead of clinical endpoints, such as overall survival or quality of life. The US Food and Drug Administration (FDA) has also approved genome-targeted cancer drugs based on phase 1 or single-arm trials.</li> <li>Given these less rigorous considerations for approval, “the validity and value of the targets and surrogate measures underlying FDA genome-targeted cancer drug approvals are uncertain,” the researchers explained.</li> <li>In the current analysis, researchers assessed the validity of the molecular targets as well as the clinical benefits of genome-targeted cancer drugs approved in the United States from 2015 to 2022 based on results from pivotal trials.</li> <li>The researchers evaluated the strength of evidence supporting molecular targetability using the European Society for Medical Oncology (ESMO) Scale for Clinical Actionability of Molecular Targets (ESCAT) and the clinical benefit using the ESMO–Magnitude of Clinical Benefit Scale (ESMO-MCBS).</li> <li>The authors defined a substantial clinical benefit as an A or B grade for curative intent and a 4 or 5 for noncurative intent. High-benefit genomic-based cancer treatments were defined as those associated with a substantial clinical benefit (ESMO-MCBS) and that qualified as ESCAT category level I-A (a clinical benefit based on prospective randomized data) or I-B (prospective nonrandomized data).</li> </ul> <h2>TAKEAWAY:</h2> <ul class="body"> <li>The analyses focused on 50 molecular-targeted cancer drugs covering 84 indications. Of which, 45 indications (54%) were approved based on phase 1 or 2 pivotal trials, 45 (54%) were supported by single-arm pivotal trials and the remaining 39 (46%) by randomized trial, and 48 (57%) were approved based on subgroup analyses.</li> <li>Among the 84 indications, more than half (55%) of the pivotal trials supporting approval used overall response rate as a primary endpoint, 31% used progression-free survival, and 6% used disease-free survival. Only seven indications (8%) were supported by pivotal trials demonstrating an improvement in overall survival.</li> <li>Among the 84 trials, 24 (29%) met the ESMO-MCBS threshold for substantial clinical benefit.</li> <li>Overall, when combining all ratings, only 24 of the 84 indications (29%) were considered high-benefit genomic-based cancer treatments.</li> </ul> <h2>IN PRACTICE:</h2> <p>“We applied the ESMO-MCBS and ESCAT value frameworks to identify therapies and molecular targets providing high clinical value that should be widely available to patients” and “found that drug indications supported by these characteristics represent a minority of cancer drug approvals in recent years,” the authors said. Using these value frameworks could help payers, governments, and individual patients “prioritize the availability of high-value molecular-targeted therapies.”</p> <h2>SOURCE:</h2> <p>The study, with first author Ariadna Tibau, MD, PhD, Brigham and Women’s Hospital and Harvard Medical School, Boston, was <a href="https://jamanetwork.com/journals/jamaoncology/article-abstract/2817121">published online</a> in <em>JAMA Oncology</em>.</p> <h2>LIMITATIONS:</h2> <p>The study evaluated only trials that supported regulatory approval and did not include outcomes of postapproval clinical studies, which could lead to changes in ESMO-MCBS grades and ESCAT levels of evidence over time.</p> <h2>DISCLOSURES:</h2> <p>The study was funded by the Kaiser Permanente Institute for Health Policy, Arnold Ventures, and the Commonwealth Fund. The authors had no relevant disclosures.</p> <p> <em>A version of this article appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/most-targeted-cancer-drugs-lack-substantial-clinical-benefit-2024a10007bm">Medscape.com</a></span>.</em> </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

ALL: Which Life-Saving Tx Is Best?

Article Type
Changed
Mon, 04/15/2024 - 17:51

In recent years, innovative use of bispecific antibodies and CAR T-cell therapy has ushered in an era when many patients with relapsed/refractory acute lymphoblastic leukemia (ALL) — who once had prognoses of 6 months or less — now survive for multiple years with the malignancy, and some are cured.

The comparative benefits and limitations of these two treatments for r/r ALL were a topic for discussion at the Great Debates & Updates Hematological Malignancies conference, held April 5-6 in New York City.

“Every single patient with ALL should benefit from bispecific antibodies before getting CAR-T cells, and I want to make the case that everybody should get CAR T as well. But they should get blinatumomab before they get CAR T,” said Elias Jabbour, MD, of the MD Anderson Cancer Center at The University of Texas in Houston, whose presentation focused on the merits of bispecific antibodies.

Jabbour_Elias_TX_web.jpg
Dr. Elias Jabbour

His argument was based on data indicating that patients have better chances of long-term remission with the use of bispecific antibodies when they are administered in an earlier round of salvage treatment — and the fact that patients who are not cured with these drugs can still achieve a lower disease burden and perform better on CAR T-cell therapy than those who don’t receive the drugs.

“When blinatumomab is used as a consolidation during the first salvage treatment and spaces out transplantation, 3-year overall survival increases in the relapse setting, deepening responses and reducing the rate of VOD (veno-occlusive disease). The safety and efficacy of CAR T depends on a disease burden. If you have a minimal residual disease (MRD), you have a safer outcome and a better outcome in the long run,” Dr. Jabbour explained.

This point of view is supported by data from the treatment of patients r/r ALL with low intensity chemotherapy + inotuzumab ozogamicin (Besponsa; Pfizer) +/- blinatumomab (Blincyto; Amgen), knows as Mini-HCVD + Ino +/-Blina. Trial members achieved a median overall survival (OS) rate of 17 months, a 3-year survival rate of 42%, and an overall MRD negativity rate of 85%.

Dr. Jabbour noted that blinatumomab has its limitations. Generally, this treatment is administered intravenously every few weeks and can be cumbersome for patients who must travel to an infusion center. However, data from a phase 1b trial of single agent subcutaneous blinatumomab for advanced ALL has demonstrated that this formulation can be effective and can lead to MRD negativity, possibly paving the way for easier administration of the drug.

Aditi Shastri, MD, a leukemia specialist at New York’s Montefiore Medical Center who attended the debate, agreed that the data presented did support Dr. Jabbour’s contention that subcutaneous blinatumomab could make treatment available to even more people with r/r ALL. “It’s easier to administer than the blina pump and could act as a bridge to curative therapies like AlloHSCT,” she said.

Jae Park, MD, a leukemia and cellular therapy specialist at Memorial Sloan Kettering Cancer Center in New York City, argued that CAR T is the most potent therapy for r/r ALL. Dr. Park agreed that inotuzumab and blinatumomab have yielded tremendous progress in the treatment of patients with r/r ALL, but he noted that bispecific antibodies lack some of the advantages of CAR T.

Park_Jae_NY_web.jpeg
Dr. Jae Park


Dr. Park said that the biggest difference between the two therapies is that CAR T requires but a single infusion of a living drug. Patients do need to stay close to treatment centers to receive treatment for toxicities, but after about 28 days, they can go home and be monitored from a distance. Furthermore, patients may start by receiving 1 million T-cells, but those cells exponentially expand 100,000- to 1,000,000-fold, meaning that the T-cells to treat cancer have the potential to persist for months and sometimes years.

Furthermore, results from ZUMA-3 Trial of the CD19-targeting CAR T-Cell therapy brexucabtagene autoleucel (Tecartus; Kite Pharma) suggest that CAR T outperforms Mini-HCVD + Ino +/-Blina in patients with r/r ALL. Participants in the trial showed an overall response rate around 80%, a 71% complete response rate, and a median OS of 25.4 months. Patients who achieved a complete response had an even better median OS of 47 months. Although this was not a head-to-head trial with Mini-HCVD + Ino +/-Blina, if the plateau of long-term survivors continues, “this drug could be set apart from treatment with monoclonal antibodies,” Dr. Park said.

However, brexucabtagene autoleucel is not a cure or even an option for all patients. Some patients are too frail to get the drug, and they risk experiencing cytokine release syndrome (CRS). Data from the FELIX study suggest that the CAR T-cell treatment Obe-cel could offer a safety profile that reduces the risk of serious side effects while remaining effective at treating r/r ALL. Obe-cel showed efficacy very similar to that of brexucabtagene autoleucel, with a 70%-80% response rate, and only 2% of patients experienced CRS.

Dr. Park noted that the next frontier in CAR T-cell therapy is figuring out which patients will respond well to CAR T and which are going to need more treatment after CAR T. However, he noted that evidence suggests patients with low MRD are likely to do best on CAR T and that bispecific antibodies can help patients get to what might be the best chance at a cure for r/r ALL, namely CAR-T.

The moderator of the debate, Jessica Altman, MD, professor of medicine, hematology oncology division, Feinberg School of Medicine at Northwestern University in Chicago, noted: “My take home is that antibody therapy and CAR-T will be sequenced and used together.” She noted that blinatumomab is moving into the front line of therapy, as in the E1910 trials, and how this treatment allows for study and use of CAR T earlier in the care of patients “when there may be less toxicity and higher response.”

Jabbour concluded on a similar note, adding that the “cure for this disease will happen in our lifetime. We will shorten therapy by doing immunotherapy upfront followed by CAR T consolidation and no more transplantation. I don’t think antibodies immunotherapies or CAR T need be competitive, they can be used in a complimentary fashion.”

Jabbour reported no financial disclosures. Park disclosed ties with Allogene, Artiva Biotherapeutics, Amgen, Affyimmune, BeBiopharma, Beigene, Bright Pharmaceuticals, Autolus, Caribou Biosciences, Galapagos, Kite, Medpace, Minerva Biotechnologies, Pfizer, Servier, Sobi, and Takeda. Neither Altman nor Shastri reported any disclosures.
 

Publications
Topics
Sections

In recent years, innovative use of bispecific antibodies and CAR T-cell therapy has ushered in an era when many patients with relapsed/refractory acute lymphoblastic leukemia (ALL) — who once had prognoses of 6 months or less — now survive for multiple years with the malignancy, and some are cured.

The comparative benefits and limitations of these two treatments for r/r ALL were a topic for discussion at the Great Debates & Updates Hematological Malignancies conference, held April 5-6 in New York City.

“Every single patient with ALL should benefit from bispecific antibodies before getting CAR-T cells, and I want to make the case that everybody should get CAR T as well. But they should get blinatumomab before they get CAR T,” said Elias Jabbour, MD, of the MD Anderson Cancer Center at The University of Texas in Houston, whose presentation focused on the merits of bispecific antibodies.

Jabbour_Elias_TX_web.jpg
Dr. Elias Jabbour

His argument was based on data indicating that patients have better chances of long-term remission with the use of bispecific antibodies when they are administered in an earlier round of salvage treatment — and the fact that patients who are not cured with these drugs can still achieve a lower disease burden and perform better on CAR T-cell therapy than those who don’t receive the drugs.

“When blinatumomab is used as a consolidation during the first salvage treatment and spaces out transplantation, 3-year overall survival increases in the relapse setting, deepening responses and reducing the rate of VOD (veno-occlusive disease). The safety and efficacy of CAR T depends on a disease burden. If you have a minimal residual disease (MRD), you have a safer outcome and a better outcome in the long run,” Dr. Jabbour explained.

This point of view is supported by data from the treatment of patients r/r ALL with low intensity chemotherapy + inotuzumab ozogamicin (Besponsa; Pfizer) +/- blinatumomab (Blincyto; Amgen), knows as Mini-HCVD + Ino +/-Blina. Trial members achieved a median overall survival (OS) rate of 17 months, a 3-year survival rate of 42%, and an overall MRD negativity rate of 85%.

Dr. Jabbour noted that blinatumomab has its limitations. Generally, this treatment is administered intravenously every few weeks and can be cumbersome for patients who must travel to an infusion center. However, data from a phase 1b trial of single agent subcutaneous blinatumomab for advanced ALL has demonstrated that this formulation can be effective and can lead to MRD negativity, possibly paving the way for easier administration of the drug.

Aditi Shastri, MD, a leukemia specialist at New York’s Montefiore Medical Center who attended the debate, agreed that the data presented did support Dr. Jabbour’s contention that subcutaneous blinatumomab could make treatment available to even more people with r/r ALL. “It’s easier to administer than the blina pump and could act as a bridge to curative therapies like AlloHSCT,” she said.

Jae Park, MD, a leukemia and cellular therapy specialist at Memorial Sloan Kettering Cancer Center in New York City, argued that CAR T is the most potent therapy for r/r ALL. Dr. Park agreed that inotuzumab and blinatumomab have yielded tremendous progress in the treatment of patients with r/r ALL, but he noted that bispecific antibodies lack some of the advantages of CAR T.

Park_Jae_NY_web.jpeg
Dr. Jae Park


Dr. Park said that the biggest difference between the two therapies is that CAR T requires but a single infusion of a living drug. Patients do need to stay close to treatment centers to receive treatment for toxicities, but after about 28 days, they can go home and be monitored from a distance. Furthermore, patients may start by receiving 1 million T-cells, but those cells exponentially expand 100,000- to 1,000,000-fold, meaning that the T-cells to treat cancer have the potential to persist for months and sometimes years.

Furthermore, results from ZUMA-3 Trial of the CD19-targeting CAR T-Cell therapy brexucabtagene autoleucel (Tecartus; Kite Pharma) suggest that CAR T outperforms Mini-HCVD + Ino +/-Blina in patients with r/r ALL. Participants in the trial showed an overall response rate around 80%, a 71% complete response rate, and a median OS of 25.4 months. Patients who achieved a complete response had an even better median OS of 47 months. Although this was not a head-to-head trial with Mini-HCVD + Ino +/-Blina, if the plateau of long-term survivors continues, “this drug could be set apart from treatment with monoclonal antibodies,” Dr. Park said.

However, brexucabtagene autoleucel is not a cure or even an option for all patients. Some patients are too frail to get the drug, and they risk experiencing cytokine release syndrome (CRS). Data from the FELIX study suggest that the CAR T-cell treatment Obe-cel could offer a safety profile that reduces the risk of serious side effects while remaining effective at treating r/r ALL. Obe-cel showed efficacy very similar to that of brexucabtagene autoleucel, with a 70%-80% response rate, and only 2% of patients experienced CRS.

Dr. Park noted that the next frontier in CAR T-cell therapy is figuring out which patients will respond well to CAR T and which are going to need more treatment after CAR T. However, he noted that evidence suggests patients with low MRD are likely to do best on CAR T and that bispecific antibodies can help patients get to what might be the best chance at a cure for r/r ALL, namely CAR-T.

The moderator of the debate, Jessica Altman, MD, professor of medicine, hematology oncology division, Feinberg School of Medicine at Northwestern University in Chicago, noted: “My take home is that antibody therapy and CAR-T will be sequenced and used together.” She noted that blinatumomab is moving into the front line of therapy, as in the E1910 trials, and how this treatment allows for study and use of CAR T earlier in the care of patients “when there may be less toxicity and higher response.”

Jabbour concluded on a similar note, adding that the “cure for this disease will happen in our lifetime. We will shorten therapy by doing immunotherapy upfront followed by CAR T consolidation and no more transplantation. I don’t think antibodies immunotherapies or CAR T need be competitive, they can be used in a complimentary fashion.”

Jabbour reported no financial disclosures. Park disclosed ties with Allogene, Artiva Biotherapeutics, Amgen, Affyimmune, BeBiopharma, Beigene, Bright Pharmaceuticals, Autolus, Caribou Biosciences, Galapagos, Kite, Medpace, Minerva Biotechnologies, Pfizer, Servier, Sobi, and Takeda. Neither Altman nor Shastri reported any disclosures.
 

In recent years, innovative use of bispecific antibodies and CAR T-cell therapy has ushered in an era when many patients with relapsed/refractory acute lymphoblastic leukemia (ALL) — who once had prognoses of 6 months or less — now survive for multiple years with the malignancy, and some are cured.

The comparative benefits and limitations of these two treatments for r/r ALL were a topic for discussion at the Great Debates & Updates Hematological Malignancies conference, held April 5-6 in New York City.

“Every single patient with ALL should benefit from bispecific antibodies before getting CAR-T cells, and I want to make the case that everybody should get CAR T as well. But they should get blinatumomab before they get CAR T,” said Elias Jabbour, MD, of the MD Anderson Cancer Center at The University of Texas in Houston, whose presentation focused on the merits of bispecific antibodies.

Jabbour_Elias_TX_web.jpg
Dr. Elias Jabbour

His argument was based on data indicating that patients have better chances of long-term remission with the use of bispecific antibodies when they are administered in an earlier round of salvage treatment — and the fact that patients who are not cured with these drugs can still achieve a lower disease burden and perform better on CAR T-cell therapy than those who don’t receive the drugs.

“When blinatumomab is used as a consolidation during the first salvage treatment and spaces out transplantation, 3-year overall survival increases in the relapse setting, deepening responses and reducing the rate of VOD (veno-occlusive disease). The safety and efficacy of CAR T depends on a disease burden. If you have a minimal residual disease (MRD), you have a safer outcome and a better outcome in the long run,” Dr. Jabbour explained.

This point of view is supported by data from the treatment of patients r/r ALL with low intensity chemotherapy + inotuzumab ozogamicin (Besponsa; Pfizer) +/- blinatumomab (Blincyto; Amgen), knows as Mini-HCVD + Ino +/-Blina. Trial members achieved a median overall survival (OS) rate of 17 months, a 3-year survival rate of 42%, and an overall MRD negativity rate of 85%.

Dr. Jabbour noted that blinatumomab has its limitations. Generally, this treatment is administered intravenously every few weeks and can be cumbersome for patients who must travel to an infusion center. However, data from a phase 1b trial of single agent subcutaneous blinatumomab for advanced ALL has demonstrated that this formulation can be effective and can lead to MRD negativity, possibly paving the way for easier administration of the drug.

Aditi Shastri, MD, a leukemia specialist at New York’s Montefiore Medical Center who attended the debate, agreed that the data presented did support Dr. Jabbour’s contention that subcutaneous blinatumomab could make treatment available to even more people with r/r ALL. “It’s easier to administer than the blina pump and could act as a bridge to curative therapies like AlloHSCT,” she said.

Jae Park, MD, a leukemia and cellular therapy specialist at Memorial Sloan Kettering Cancer Center in New York City, argued that CAR T is the most potent therapy for r/r ALL. Dr. Park agreed that inotuzumab and blinatumomab have yielded tremendous progress in the treatment of patients with r/r ALL, but he noted that bispecific antibodies lack some of the advantages of CAR T.

Park_Jae_NY_web.jpeg
Dr. Jae Park


Dr. Park said that the biggest difference between the two therapies is that CAR T requires but a single infusion of a living drug. Patients do need to stay close to treatment centers to receive treatment for toxicities, but after about 28 days, they can go home and be monitored from a distance. Furthermore, patients may start by receiving 1 million T-cells, but those cells exponentially expand 100,000- to 1,000,000-fold, meaning that the T-cells to treat cancer have the potential to persist for months and sometimes years.

Furthermore, results from ZUMA-3 Trial of the CD19-targeting CAR T-Cell therapy brexucabtagene autoleucel (Tecartus; Kite Pharma) suggest that CAR T outperforms Mini-HCVD + Ino +/-Blina in patients with r/r ALL. Participants in the trial showed an overall response rate around 80%, a 71% complete response rate, and a median OS of 25.4 months. Patients who achieved a complete response had an even better median OS of 47 months. Although this was not a head-to-head trial with Mini-HCVD + Ino +/-Blina, if the plateau of long-term survivors continues, “this drug could be set apart from treatment with monoclonal antibodies,” Dr. Park said.

However, brexucabtagene autoleucel is not a cure or even an option for all patients. Some patients are too frail to get the drug, and they risk experiencing cytokine release syndrome (CRS). Data from the FELIX study suggest that the CAR T-cell treatment Obe-cel could offer a safety profile that reduces the risk of serious side effects while remaining effective at treating r/r ALL. Obe-cel showed efficacy very similar to that of brexucabtagene autoleucel, with a 70%-80% response rate, and only 2% of patients experienced CRS.

Dr. Park noted that the next frontier in CAR T-cell therapy is figuring out which patients will respond well to CAR T and which are going to need more treatment after CAR T. However, he noted that evidence suggests patients with low MRD are likely to do best on CAR T and that bispecific antibodies can help patients get to what might be the best chance at a cure for r/r ALL, namely CAR-T.

The moderator of the debate, Jessica Altman, MD, professor of medicine, hematology oncology division, Feinberg School of Medicine at Northwestern University in Chicago, noted: “My take home is that antibody therapy and CAR-T will be sequenced and used together.” She noted that blinatumomab is moving into the front line of therapy, as in the E1910 trials, and how this treatment allows for study and use of CAR T earlier in the care of patients “when there may be less toxicity and higher response.”

Jabbour concluded on a similar note, adding that the “cure for this disease will happen in our lifetime. We will shorten therapy by doing immunotherapy upfront followed by CAR T consolidation and no more transplantation. I don’t think antibodies immunotherapies or CAR T need be competitive, they can be used in a complimentary fashion.”

Jabbour reported no financial disclosures. Park disclosed ties with Allogene, Artiva Biotherapeutics, Amgen, Affyimmune, BeBiopharma, Beigene, Bright Pharmaceuticals, Autolus, Caribou Biosciences, Galapagos, Kite, Medpace, Minerva Biotechnologies, Pfizer, Servier, Sobi, and Takeda. Neither Altman nor Shastri reported any disclosures.
 

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167674</fileName> <TBEID>0C04F870.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F870</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>CAR T vs Blina</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240412T130859</QCDate> <firstPublished>20240412T134754</firstPublished> <LastPublished>20240412T134754</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240412T134754</CMSDate> <articleSource>FROM GREAT DEBATES &amp; UPDATES HEMATOLOGIC MALIGNANCIES</articleSource> <facebookInfo/> <meetingNumber>NA</meetingNumber> <byline>Mylesstarr@gmail.com</byline> <bylineText>MYLES STARR</bylineText> <bylineFull>MYLES STARR</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>In recent years, innovative use of bispecific antibodies and CAR T-cell therapy has ushered in an era when many patients with relapsed/refractory acute lymphobl</metaDescription> <articlePDF/> <teaserImage>301116</teaserImage> <teaser>Both CAR T-cell therapy and bispecific antibodies are helping patients with patients with R/R ALL live longer, but which treatment is better?</teaser> <title>ALL: Which Life-Saving Tx Is Best?</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>hemn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">18</term> </publications> <sections> <term canonical="true">53</term> <term>39313</term> </sections> <topics> <term canonical="true">179</term> <term>195</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/24012822.jpg</altRep> <description role="drol:caption">Dr. Elias Jabbour</description> <description role="drol:credit">MD Anderson Cancer Center</description> </link> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/24012823.jpg</altRep> <description role="drol:caption">Dr. Jae Park</description> <description role="drol:credit">Memorial Sloan Kettering Cancer Center</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>ALL: Which Life-Saving Tx Is Best?</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">In recent years, innovative use of bispecific antibodies and CAR T-cell therapy has ushered in an era when many patients with relapsed/refractory acute lymphoblastic leukemia (ALL) — who once had <span class="Hyperlink"><a href="https://pubmed.ncbi.nlm.nih.gov/27587380/">prognoses </a></span>of 6 months or less — now survive for multiple years with the malignancy, and some are cured. </span><br/><br/>The comparative benefits and limitations of these two treatments for r/r ALL were a topic for discussion at the Great Debates &amp; Updates Hematological Malignancies conference, held April 5-6 in New York City. <br/><br/>“Every single patient with ALL should benefit from bispecific antibodies before getting CAR-T cells, and I want to make the case that everybody should get CAR T as well. But they should get blinatumomab before they get CAR T,” said Elias Jabbour, MD, of the MD Anderson Cancer Center at The University of Texas in Houston, whose presentation focused on the merits of bispecific antibodies.<br/><br/>[[{"fid":"301116","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Elias Jabbour, professor, leukemia department, MD Anderson Cancer Center, Houston, TX","field_file_image_credit[und][0][value]":"MD Anderson Cancer Center","field_file_image_caption[und][0][value]":"Dr. Elias Jabbour"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]His argument was based on data indicating that patients have better chances of long-term remission with the use of bispecific antibodies when they are administered in an earlier round of salvage treatment — and the fact that patients who are not cured with these drugs can still achieve a lower disease burden and perform better on CAR T-cell therapy than those who don’t receive the drugs. <br/><br/>“When blinatumomab is used as a consolidation during the first salvage treatment and spaces out transplantation, 3-year overall survival increases in the relapse setting, deepening responses and reducing the rate of VOD (veno-occlusive disease). The safety and efficacy of CAR T depends on a disease burden. If you have a minimal residual disease (MRD), you have a safer outcome and a better outcome in the long run,” Dr. Jabbour explained. <br/><br/>This point of view is supported by <span class="Hyperlink"><a href="https://library.ehaweb.org/eha/2023/eha2023-congress/386202/">data</a></span> from the treatment of patients r/r ALL with low intensity chemotherapy + inotuzumab ozogamicin (Besponsa; Pfizer) +/- blinatumomab (Blincyto; Amgen), knows as Mini-HCVD + Ino +/-Blina. Trial members achieved a median overall survival (OS) rate of 17 months, a 3-year survival rate of 42%, and an overall MRD negativity rate of 85%.<br/><br/>Dr. Jabbour noted that blinatumomab has its limitations. Generally, this treatment is administered intravenously every few weeks and can be cumbersome for patients who must travel to an infusion center. However, data from a phase 1b <span class="Hyperlink"><a href="https://pubmed.ncbi.nlm.nih.gov/38317420/">trial</a></span> of single agent subcutaneous blinatumomab for advanced ALL has demonstrated that this formulation can be effective and can lead to MRD negativity, possibly paving the way for easier administration of the drug. <br/><br/>Aditi Shastri, MD, a leukemia specialist at New York’s Montefiore Medical Center who attended the debate, agreed that the data presented did support Dr. Jabbour’s contention that subcutaneous blinatumomab could make treatment available to even more people with r/r ALL. “It’s easier to administer than the blina pump and could act as a bridge to curative therapies like AlloHSCT,” she said.<br/><br/>Jae Park, MD, a leukemia and cellular therapy specialist at Memorial Sloan Kettering Cancer Center in New York City, argued that CAR T is the most potent therapy for r/r ALL. Dr. Park agreed that inotuzumab and blinatumomab have yielded tremendous progress in the treatment of patients with r/r ALL, but he noted that bispecific antibodies lack some of the advantages of CAR T. [[{"fid":"301117","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Jae Park, leukemia &amp; cellular therapy specialist, Memorial Sloan Kettering Cancer Center, New York, NY","field_file_image_credit[und][0][value]":"Memorial Sloan Kettering Cancer Center","field_file_image_caption[und][0][value]":"Dr. Jae Park"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_left"}}]]<br/><br/>Dr. Park said that the biggest difference between the two therapies is that CAR T requires but a single infusion of a living drug. Patients do need to stay close to treatment centers to receive treatment for toxicities, but after about 28 days, they can go home and be monitored from a distance. Furthermore, patients may start by receiving 1 million T-cells, but those cells exponentially expand 100,000- to 1,000,000-fold, meaning that the T-cells to treat cancer have the potential to persist for months and sometimes years.<br/><br/>Furthermore, results from ZUMA-3 Trial of the CD19-targeting CAR T-Cell therapy brexucabtagene autoleucel (Tecartus; Kite Pharma) suggest that CAR T outperforms Mini-HCVD + Ino +/-Blina in patients with r/r ALL. Participants in the trial showed an overall response rate around 80%, a 71% complete response rate, and a median OS of 25.4 months. Patients who achieved a complete response had an even better median OS of 47 months. Although this was not a head-to-head trial with Mini-HCVD + Ino +/-Blina, if the plateau of long-term survivors continues, “this drug could be set apart from treatment with monoclonal antibodies,” Dr. Park said.<br/><br/>However, brexucabtagene autoleucel is not a cure or even an option for all patients. Some patients are too frail to get the drug, and they risk experiencing cytokine release syndrome (CRS). Data from the FELIX study suggest that the CAR T-cell treatment Obe-cel could offer a safety profile that reduces the risk of serious side effects while remaining effective at treating r/r ALL. Obe-cel showed efficacy very similar to that of brexucabtagene autoleucel, with a 70%-80% response rate, and only 2% of patients experienced CRS. <br/><br/>Dr. Park noted that the next frontier in CAR T-cell therapy is figuring out which patients will respond well to CAR T and which are going to need more treatment after CAR T. However, he noted that evidence suggests patients with low MRD are likely to do best on CAR T and that bispecific antibodies can help patients get to what might be the best chance at a cure for r/r ALL, namely CAR-T.<br/><br/>The moderator of the debate, Jessica Altman, MD, professor of medicine, hematology oncology division, Feinberg School of Medicine at Northwestern University in Chicago, noted: “My take home is that antibody therapy and CAR-T will be sequenced and used together.” She noted that blinatumomab is moving into the front line of therapy, as in the <span class="Hyperlink"><a href="https://ashpublications.org/blood/article/140/Supplement%202/LBA-1/493429/Consolidation-Therapy-with-Blinatumomab-Improves">E1910</a></span> trials, and how this treatment allows for study and use of CAR T earlier in the care of patients “when there may be less toxicity and higher response.” <br/><br/>Jabbour concluded on a similar note, adding that the “cure for this disease will happen in our lifetime. We will shorten therapy by doing immunotherapy upfront followed by CAR T consolidation and no more transplantation. I don’t think antibodies immunotherapies or CAR T need be competitive, they can be used in a complimentary fashion.” <br/><br/>Jabbour reported no financial disclosures. Park disclosed ties with Allogene, Artiva Biotherapeutics, Amgen, Affyimmune, BeBiopharma, Beigene, Bright Pharmaceuticals, Autolus, Caribou Biosciences, Galapagos, Kite, Medpace, Minerva Biotechnologies, Pfizer, Servier, Sobi, and Takeda. Neither Altman nor Shastri reported any disclosures. <br/><br/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM GREAT DEBATES & UPDATES HEMATOLOGIC MALIGNANCIES

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Less Than 50% of Accelerated Approvals Show Clinical Benefit

Article Type
Changed
Tue, 04/09/2024 - 23:03

— Fewer than half of the cancer drugs approved under the US Food and Drug Administration’s (FDA’s) accelerated approval pathway between 2013 and 2017 have been shown to improve overall survival or quality of life, despite being on the US market for more than 5 years, according to a new study. 

Under the program, drugs are approved for marketing if they show benefit in surrogate markers thought to indicate efficacy. Progression-free survival, tumor response, and duration of response are the most used surrogate markers for accelerated approvals of cancer drugs. These are based largely on imaging studies that show either a stop in growth in the case of progression-free survival or tumor shrinkage in the case of tumor response. 

Following accelerated approvals, companies are then supposed to show actual clinical benefit in confirmatory trials.

The problem with relying on surrogate markers for drug approvals is that they don’t always correlate with longer survival or improved quality of life, said Edward Cliff, MBBS, who presented the findings at the American Association for Cancer Research 2024 annual meeting (abstract 918). The study was also published in JAMA to coincide with the meeting presentation.

In some cancers, these markers work well, but in others they don’t, said Dr. Cliff, a hematology trainee at Brigham and Women’s Hospital, Boston, when the work was conducted, and now a hematology fellow at the Peter MacCallum Cancer Centre in Melbourne, Australia.

To determine whether cancer drugs granted accelerated approval ultimately show an overall survival or quality of life benefit, researchers reviewed 46 cancer drugs granted accelerated approvals between 2013 and 2017. Twenty (43%) were granted full approval after demonstrating survival or quality-of-life benefits. 

Nine, however, were converted to full approvals on the basis of surrogate markers. These include a full approval for pembrolizumab in previously treated recurrent or refractory head and neck squamous cell carcinoma and a full approval for nivolumab for refractory locally advanced or metastatic urothelial carcinoma, both based on tumor response rate and duration of response.

Of the remaining 17 drugs evaluated in the trial, 10 have been withdrawn and seven do not yet have confirmatory trial results. 

The reliance on surrogate markers means that these drugs are used for treatment, covered by insurance, and added to guidelines — all without solid evidence of real-world clinical benefit, said Dr. Cliff. 

However, the goal should not be to do away with the accelerated approval process, because it sometimes does deliver powerful agents to patients quickly. Instead, Dr. Cliff told this news organization, the system needs to be improved so that “we keep the speed while getting certainty around clinical benefits” with robust and timely confirmatory trials. 

In the meantime, “clinicians should communicate with patients about any residual uncertainty of clinical benefit when they offer novel therapies,” Dr. Cliff explained. “It’s important for them to have the information.”

There has been some progress on the issue. In December 2022, the US Congress passed the Food and Drug Administration Omnibus Reform Act. Among other things, the Act requires companies to have confirmation trials underway as a condition for accelerated approval, and to provide regular reports on their progress. The Act also expedites the withdrawal process for drugs that don’t show a benefit. 

The Act has been put to the test twice recently. In February, FDA used the expedited process to remove the multiple myeloma drug melphalan flufenamide from the market. Melphalan flufenamide hadn’t been sold in the US for quite some time, so the process wasn’t contentious. 

In March, Regeneron announced that accelerated approval for the follicular and diffuse B cell lymphoma drug odronextamab has been delayed pending enrollment in a confirmatory trial. 

“There have been some promising steps,” Dr. Cliff said, but much work needs to be done. 

Study moderator Shivaani Kummar, MD, agreed, noting that “the data is showing that the confirmatory trials aren’t happening at the pace which they should.” 

But the solution is not to curtail approvals; it’s to make sure that accelerated approval commitments are met, said Dr. Kummar.

Still, “as a practicing oncologist, I welcome the accelerated pathway,” Dr. Kummar, a medical oncologist/hematologist at Oregon Health & Science University, Portland, told this news organization. “I want the availability to my patients.” 

Having drugs approved on the basis of surrogate markers doesn’t necessarily mean patients are getting ineffective therapies, Dr. Kummar noted. For instance, if an agent just shrinks the tumor, it can sometimes still be “a huge clinical benefit because it can take the symptoms away.” 

As for prescribing drugs based on accelerated approvals, she said she tells her patients that trials have been promising, but we don’t know what the long-term effects are. She and her patient then make a decision together. 

The study was funded by Arnold Ventures. Dr. Kummar reported support from several companies, including Bayer, Gilead, and others. Dr. Cliff had no disclosures. 
 

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

— Fewer than half of the cancer drugs approved under the US Food and Drug Administration’s (FDA’s) accelerated approval pathway between 2013 and 2017 have been shown to improve overall survival or quality of life, despite being on the US market for more than 5 years, according to a new study. 

Under the program, drugs are approved for marketing if they show benefit in surrogate markers thought to indicate efficacy. Progression-free survival, tumor response, and duration of response are the most used surrogate markers for accelerated approvals of cancer drugs. These are based largely on imaging studies that show either a stop in growth in the case of progression-free survival or tumor shrinkage in the case of tumor response. 

Following accelerated approvals, companies are then supposed to show actual clinical benefit in confirmatory trials.

The problem with relying on surrogate markers for drug approvals is that they don’t always correlate with longer survival or improved quality of life, said Edward Cliff, MBBS, who presented the findings at the American Association for Cancer Research 2024 annual meeting (abstract 918). The study was also published in JAMA to coincide with the meeting presentation.

In some cancers, these markers work well, but in others they don’t, said Dr. Cliff, a hematology trainee at Brigham and Women’s Hospital, Boston, when the work was conducted, and now a hematology fellow at the Peter MacCallum Cancer Centre in Melbourne, Australia.

To determine whether cancer drugs granted accelerated approval ultimately show an overall survival or quality of life benefit, researchers reviewed 46 cancer drugs granted accelerated approvals between 2013 and 2017. Twenty (43%) were granted full approval after demonstrating survival or quality-of-life benefits. 

Nine, however, were converted to full approvals on the basis of surrogate markers. These include a full approval for pembrolizumab in previously treated recurrent or refractory head and neck squamous cell carcinoma and a full approval for nivolumab for refractory locally advanced or metastatic urothelial carcinoma, both based on tumor response rate and duration of response.

Of the remaining 17 drugs evaluated in the trial, 10 have been withdrawn and seven do not yet have confirmatory trial results. 

The reliance on surrogate markers means that these drugs are used for treatment, covered by insurance, and added to guidelines — all without solid evidence of real-world clinical benefit, said Dr. Cliff. 

However, the goal should not be to do away with the accelerated approval process, because it sometimes does deliver powerful agents to patients quickly. Instead, Dr. Cliff told this news organization, the system needs to be improved so that “we keep the speed while getting certainty around clinical benefits” with robust and timely confirmatory trials. 

In the meantime, “clinicians should communicate with patients about any residual uncertainty of clinical benefit when they offer novel therapies,” Dr. Cliff explained. “It’s important for them to have the information.”

There has been some progress on the issue. In December 2022, the US Congress passed the Food and Drug Administration Omnibus Reform Act. Among other things, the Act requires companies to have confirmation trials underway as a condition for accelerated approval, and to provide regular reports on their progress. The Act also expedites the withdrawal process for drugs that don’t show a benefit. 

The Act has been put to the test twice recently. In February, FDA used the expedited process to remove the multiple myeloma drug melphalan flufenamide from the market. Melphalan flufenamide hadn’t been sold in the US for quite some time, so the process wasn’t contentious. 

In March, Regeneron announced that accelerated approval for the follicular and diffuse B cell lymphoma drug odronextamab has been delayed pending enrollment in a confirmatory trial. 

“There have been some promising steps,” Dr. Cliff said, but much work needs to be done. 

Study moderator Shivaani Kummar, MD, agreed, noting that “the data is showing that the confirmatory trials aren’t happening at the pace which they should.” 

But the solution is not to curtail approvals; it’s to make sure that accelerated approval commitments are met, said Dr. Kummar.

Still, “as a practicing oncologist, I welcome the accelerated pathway,” Dr. Kummar, a medical oncologist/hematologist at Oregon Health & Science University, Portland, told this news organization. “I want the availability to my patients.” 

Having drugs approved on the basis of surrogate markers doesn’t necessarily mean patients are getting ineffective therapies, Dr. Kummar noted. For instance, if an agent just shrinks the tumor, it can sometimes still be “a huge clinical benefit because it can take the symptoms away.” 

As for prescribing drugs based on accelerated approvals, she said she tells her patients that trials have been promising, but we don’t know what the long-term effects are. She and her patient then make a decision together. 

The study was funded by Arnold Ventures. Dr. Kummar reported support from several companies, including Bayer, Gilead, and others. Dr. Cliff had no disclosures. 
 

A version of this article appeared on Medscape.com.

— Fewer than half of the cancer drugs approved under the US Food and Drug Administration’s (FDA’s) accelerated approval pathway between 2013 and 2017 have been shown to improve overall survival or quality of life, despite being on the US market for more than 5 years, according to a new study. 

Under the program, drugs are approved for marketing if they show benefit in surrogate markers thought to indicate efficacy. Progression-free survival, tumor response, and duration of response are the most used surrogate markers for accelerated approvals of cancer drugs. These are based largely on imaging studies that show either a stop in growth in the case of progression-free survival or tumor shrinkage in the case of tumor response. 

Following accelerated approvals, companies are then supposed to show actual clinical benefit in confirmatory trials.

The problem with relying on surrogate markers for drug approvals is that they don’t always correlate with longer survival or improved quality of life, said Edward Cliff, MBBS, who presented the findings at the American Association for Cancer Research 2024 annual meeting (abstract 918). The study was also published in JAMA to coincide with the meeting presentation.

In some cancers, these markers work well, but in others they don’t, said Dr. Cliff, a hematology trainee at Brigham and Women’s Hospital, Boston, when the work was conducted, and now a hematology fellow at the Peter MacCallum Cancer Centre in Melbourne, Australia.

To determine whether cancer drugs granted accelerated approval ultimately show an overall survival or quality of life benefit, researchers reviewed 46 cancer drugs granted accelerated approvals between 2013 and 2017. Twenty (43%) were granted full approval after demonstrating survival or quality-of-life benefits. 

Nine, however, were converted to full approvals on the basis of surrogate markers. These include a full approval for pembrolizumab in previously treated recurrent or refractory head and neck squamous cell carcinoma and a full approval for nivolumab for refractory locally advanced or metastatic urothelial carcinoma, both based on tumor response rate and duration of response.

Of the remaining 17 drugs evaluated in the trial, 10 have been withdrawn and seven do not yet have confirmatory trial results. 

The reliance on surrogate markers means that these drugs are used for treatment, covered by insurance, and added to guidelines — all without solid evidence of real-world clinical benefit, said Dr. Cliff. 

However, the goal should not be to do away with the accelerated approval process, because it sometimes does deliver powerful agents to patients quickly. Instead, Dr. Cliff told this news organization, the system needs to be improved so that “we keep the speed while getting certainty around clinical benefits” with robust and timely confirmatory trials. 

In the meantime, “clinicians should communicate with patients about any residual uncertainty of clinical benefit when they offer novel therapies,” Dr. Cliff explained. “It’s important for them to have the information.”

There has been some progress on the issue. In December 2022, the US Congress passed the Food and Drug Administration Omnibus Reform Act. Among other things, the Act requires companies to have confirmation trials underway as a condition for accelerated approval, and to provide regular reports on their progress. The Act also expedites the withdrawal process for drugs that don’t show a benefit. 

The Act has been put to the test twice recently. In February, FDA used the expedited process to remove the multiple myeloma drug melphalan flufenamide from the market. Melphalan flufenamide hadn’t been sold in the US for quite some time, so the process wasn’t contentious. 

In March, Regeneron announced that accelerated approval for the follicular and diffuse B cell lymphoma drug odronextamab has been delayed pending enrollment in a confirmatory trial. 

“There have been some promising steps,” Dr. Cliff said, but much work needs to be done. 

Study moderator Shivaani Kummar, MD, agreed, noting that “the data is showing that the confirmatory trials aren’t happening at the pace which they should.” 

But the solution is not to curtail approvals; it’s to make sure that accelerated approval commitments are met, said Dr. Kummar.

Still, “as a practicing oncologist, I welcome the accelerated pathway,” Dr. Kummar, a medical oncologist/hematologist at Oregon Health & Science University, Portland, told this news organization. “I want the availability to my patients.” 

Having drugs approved on the basis of surrogate markers doesn’t necessarily mean patients are getting ineffective therapies, Dr. Kummar noted. For instance, if an agent just shrinks the tumor, it can sometimes still be “a huge clinical benefit because it can take the symptoms away.” 

As for prescribing drugs based on accelerated approvals, she said she tells her patients that trials have been promising, but we don’t know what the long-term effects are. She and her patient then make a decision together. 

The study was funded by Arnold Ventures. Dr. Kummar reported support from several companies, including Bayer, Gilead, and others. Dr. Cliff had no disclosures. 
 

A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167634</fileName> <TBEID>0C04F7D5.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F7D5</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240409T145931</QCDate> <firstPublished>20240409T150541</firstPublished> <LastPublished>20240409T150541</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240409T150541</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>M. Alexander Otto, PA</byline> <bylineText>M. ALEXANDER OTTO, PA, MMS</bylineText> <bylineFull>M. ALEXANDER OTTO, PA, MMS</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>SAN DIEGO — Fewer than half of the cancer drugs approved under the US Food and Drug Administration’s (FDA’s) accelerated approval pathway between 2013 and 2017 </metaDescription> <articlePDF/> <teaserImage/> <title>Less Than 50% of Accelerated Approvals Show Clinical Benefit</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>oncr</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>hemn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>chph</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>skin</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>pn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>GIHOLD</publicationCode> <pubIssueName>January 2014</pubIssueName> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle/> <journalFullTitle/> <copyrightStatement/> </publicationData> <publicationData> <publicationCode>ob</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>nr</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Neurology Reviews</journalTitle> <journalFullTitle>Neurology Reviews</journalFullTitle> <copyrightStatement>2018 Frontline Medical Communications Inc.,</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">31</term> <term>18</term> <term>6</term> <term>13</term> <term>25</term> <term>23</term> <term>22</term> </publications> <sections> <term canonical="true">27970</term> <term>37225</term> <term>39313</term> </sections> <topics> <term>192</term> <term>198</term> <term>61821</term> <term>59244</term> <term>67020</term> <term>214</term> <term>217</term> <term>221</term> <term>232</term> <term>238</term> <term>242</term> <term>244</term> <term>39570</term> <term>27442</term> <term>256</term> <term>245</term> <term>270</term> <term canonical="true">278</term> <term>31848</term> <term>292</term> <term>178</term> <term>179</term> <term>181</term> <term>59374</term> <term>196</term> <term>195</term> <term>197</term> <term>61642</term> <term>37637</term> <term>233</term> <term>243</term> <term>250</term> <term>303</term> <term>271</term> <term>49434</term> <term>263</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Less Than 50% of Accelerated Approvals Show Clinical Benefit</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription"><span class="dateline">SAN DIEGO</span> — Fewer than half of the cancer drugs approved under the US Food and Drug Administration’s (FDA’s) accelerated approval pathway between 2013 and 2017 have been shown to improve overall survival or quality of life,</span> despite being on the US market for more than 5 years, according to a new study. </p> <p>Under the program, drugs are approved for marketing if they show benefit in surrogate markers thought to indicate efficacy. Progression-free survival, tumor response, and duration of response are the most used surrogate markers for accelerated approvals of cancer drugs. These are based largely on imaging studies that show either a stop in growth in the case of progression-free survival or tumor shrinkage in the case of tumor response. <br/><br/>Following accelerated approvals, companies are then supposed to show actual clinical benefit in confirmatory trials.<br/><br/>The problem with relying on surrogate markers for drug approvals is that they don’t always correlate with longer survival or improved quality of life, said Edward Cliff, MBBS, who presented the findings at the American Association for Cancer Research 2024 annual meeting (abstract 918). The study was also <span class="Hyperlink"><a href="https://jamanetwork.com/journals/jama/fullarticle/2817324">published</a></span> in <span class="Emphasis">JAMA</span> to coincide with the meeting presentation.<br/><br/>In some cancers, these markers work well, but in others they don’t, said Dr. Cliff, a hematology trainee at Brigham and Women’s Hospital, Boston, when the work was conducted, and now a hematology fellow at the Peter MacCallum Cancer Centre in Melbourne, Australia.<br/><br/>To determine whether cancer drugs granted accelerated approval ultimately show an overall survival or quality of life benefit, researchers reviewed 46 cancer drugs granted accelerated approvals between 2013 and 2017. Twenty (43%) were granted full approval after demonstrating survival or quality-of-life benefits. <br/><br/>Nine, however, were converted to full approvals on the basis of surrogate markers. These include a full approval for <span class="Hyperlink"><a href="https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/125514s160lbl.pdf">pembrolizumab</a></span> in previously treated recurrent or refractory head and <span class="Hyperlink">neck squamous cell carcinoma</span> and a full approval for <span class="Hyperlink"><a href="https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/125554s128lbl.pdf">nivolumab</a></span> for refractory locally advanced or metastatic urothelial carcinoma, both based on tumor response rate and duration of response.<br/><br/>Of the remaining 17 drugs evaluated in the trial, 10 have been withdrawn and seven do not yet have confirmatory trial results. <br/><br/>The reliance on surrogate markers means that these drugs are used for treatment, covered by insurance, and added to guidelines — all without solid evidence of real-world clinical benefit, said Dr. Cliff. <br/><br/>However, the goal should not be to do away with the accelerated approval process, because it sometimes does deliver powerful agents to patients quickly. Instead, Dr. Cliff told this news organization, the system needs to be improved so that “we keep the speed while getting certainty around clinical benefits” with robust and timely confirmatory trials. <br/><br/>In the meantime, “clinicians should communicate with patients about any residual uncertainty of clinical benefit when they offer novel therapies,” Dr. Cliff explained. “It’s important for them to have the information.”<br/><br/>There has been some progress on the issue. In December 2022, the US Congress passed the <span class="Hyperlink"><a href="https://www.thefdalawblog.com/wp-content/uploads/2023/01/HPM-FDORA-Summary-and-Analysis.pdf">Food and Drug Administration Omnibus Reform Act</a></span>. Among other things, the Act requires companies to have confirmation trials underway as a condition for accelerated approval, and to provide regular reports on their progress. The Act also expedites the withdrawal process for drugs that don’t show a benefit. <br/><br/>The Act has been put to the test twice recently. In February, FDA <span class="Hyperlink"><a href="https://www.mdedge.com/hematology-oncology/article/268038/multiple-myeloma/fda-withdraws-melflufen-approval-ema-still">used the expedited process</a></span> to remove the <span class="Hyperlink">multiple myeloma</span> drug <span class="Hyperlink">melphalan flufenamide</span> from the market. Melphalan flufenamide hadn’t been sold in the US for quite some time, so the process wasn’t contentious. <br/><br/>In March, Regeneron announced that accelerated approval for the follicular and diffuse <span class="Hyperlink">B cell lymphoma</span> drug odronextamab has been <span class="Hyperlink"><a href="https://investor.regeneron.com/news-releases/news-release-details/regeneron-provides-update-biologics-license-application">delayed</a></span> pending enrollment in a confirmatory trial. <br/><br/>“There have been some promising steps,” Dr. Cliff said, but much work needs to be done. <br/><br/>Study moderator <span class="Hyperlink"><a href="https://www.ohsu.edu/providers/shivaani-kummar-md-facp">Shivaani Kummar</a></span>, MD, agreed, noting that “the data is showing that the confirmatory trials aren’t happening at the pace which they should.” <br/><br/>But the solution is not to curtail approvals; it’s to make sure that accelerated approval commitments are met, said Dr. Kummar.<br/><br/>Still, “as a practicing oncologist, I welcome the accelerated pathway,” Dr. Kummar, a medical oncologist/hematologist at Oregon Health &amp; Science University, Portland, told this news organization. “I want the availability to my patients.” <br/><br/>Having drugs approved on the basis of surrogate markers doesn’t necessarily mean patients are getting ineffective therapies, Dr. Kummar noted. For instance, if an agent just shrinks the tumor, it can sometimes still be “a huge clinical benefit because it can take the symptoms away.” <br/><br/>As for prescribing drugs based on accelerated approvals, she said she tells her patients that trials have been promising, but we don’t know what the long-term effects are. She and her patient then make a decision together. <br/><br/>The study was funded by Arnold Ventures. Dr. Kummar reported support from several companies, including Bayer, Gilead, and others. Dr. Cliff had no disclosures.<span class="Emphasis"> <br/><br/></span></p> <p> <em>A version of this article appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/less-than-50-accelerated-approvals-show-clinical-benefit-2024a10006nm">Medscape.com</a></span>.</em> </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p>Researchers review 46 cancer drugs granted accelerated approvals between 2013 and 2017.</p> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Abecma Approved for Earlier Lines in Relapsed/Refractory Multiple Myeloma

Article Type
Changed
Tue, 04/09/2024 - 09:47

The US Food and Drug Administration (FDA) has approved idecabtagene vicleucel (ide-cel) (Abecma, Bristol-Myers Squibb/2seventy bio) for adults with relapsed or refractory multiple myeloma after two or more prior lines of therapy, including an immunomodulatory agent, a proteasome inhibitor, and an anti-CD38 monoclonal antibody. 

The approval expands the chimeric antigen receptor (CAR) T-cell therapy’s indications to earlier lines of treatment after exposure to these other main therapy classes, Bristol Myers Squibb said in a press release

Approval was based on the KarMMa-3 trial, in which 254 patients were randomly assigned to ide-cel and 132 to investigators’ choice of standard regimens, consisting of combinations of daratumumabdexamethasone, and other agents. 

After a median follow-up of 15.9 months, median progression-free survival was three times higher in the ide-cel arm: 13.3 months with the CAR T-cell therapy vs 4.4 months with standard treatment. Overall, 39% of patients on ide-cel had a complete response vs 5% on standard regimens. 

The approval includes a new recommended dose range of 300-510 x 106 CAR-positive T cells. 

Ide-cel carries a boxed warning for cytokine release syndrome, neurologic toxicities, hemophagocytic lymphohistiocytosis/macrophage activation syndrome, prolonged cytopenia, and secondary hematologic cancers. 

In trials, cytokine release syndrome occurred in 89% (310 of 349) of patients in the KarMMa-3 and KarMMa studies, which included grade 3 syndrome in 7% (23 of 349) and fatal cases in 0.9% (3 of 349) of patients.

A one-time treatment is over $500,000, according to drugs.com.

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

The US Food and Drug Administration (FDA) has approved idecabtagene vicleucel (ide-cel) (Abecma, Bristol-Myers Squibb/2seventy bio) for adults with relapsed or refractory multiple myeloma after two or more prior lines of therapy, including an immunomodulatory agent, a proteasome inhibitor, and an anti-CD38 monoclonal antibody. 

The approval expands the chimeric antigen receptor (CAR) T-cell therapy’s indications to earlier lines of treatment after exposure to these other main therapy classes, Bristol Myers Squibb said in a press release

Approval was based on the KarMMa-3 trial, in which 254 patients were randomly assigned to ide-cel and 132 to investigators’ choice of standard regimens, consisting of combinations of daratumumabdexamethasone, and other agents. 

After a median follow-up of 15.9 months, median progression-free survival was three times higher in the ide-cel arm: 13.3 months with the CAR T-cell therapy vs 4.4 months with standard treatment. Overall, 39% of patients on ide-cel had a complete response vs 5% on standard regimens. 

The approval includes a new recommended dose range of 300-510 x 106 CAR-positive T cells. 

Ide-cel carries a boxed warning for cytokine release syndrome, neurologic toxicities, hemophagocytic lymphohistiocytosis/macrophage activation syndrome, prolonged cytopenia, and secondary hematologic cancers. 

In trials, cytokine release syndrome occurred in 89% (310 of 349) of patients in the KarMMa-3 and KarMMa studies, which included grade 3 syndrome in 7% (23 of 349) and fatal cases in 0.9% (3 of 349) of patients.

A one-time treatment is over $500,000, according to drugs.com.

A version of this article appeared on Medscape.com.

The US Food and Drug Administration (FDA) has approved idecabtagene vicleucel (ide-cel) (Abecma, Bristol-Myers Squibb/2seventy bio) for adults with relapsed or refractory multiple myeloma after two or more prior lines of therapy, including an immunomodulatory agent, a proteasome inhibitor, and an anti-CD38 monoclonal antibody. 

The approval expands the chimeric antigen receptor (CAR) T-cell therapy’s indications to earlier lines of treatment after exposure to these other main therapy classes, Bristol Myers Squibb said in a press release

Approval was based on the KarMMa-3 trial, in which 254 patients were randomly assigned to ide-cel and 132 to investigators’ choice of standard regimens, consisting of combinations of daratumumabdexamethasone, and other agents. 

After a median follow-up of 15.9 months, median progression-free survival was three times higher in the ide-cel arm: 13.3 months with the CAR T-cell therapy vs 4.4 months with standard treatment. Overall, 39% of patients on ide-cel had a complete response vs 5% on standard regimens. 

The approval includes a new recommended dose range of 300-510 x 106 CAR-positive T cells. 

Ide-cel carries a boxed warning for cytokine release syndrome, neurologic toxicities, hemophagocytic lymphohistiocytosis/macrophage activation syndrome, prolonged cytopenia, and secondary hematologic cancers. 

In trials, cytokine release syndrome occurred in 89% (310 of 349) of patients in the KarMMa-3 and KarMMa studies, which included grade 3 syndrome in 7% (23 of 349) and fatal cases in 0.9% (3 of 349) of patients.

A one-time treatment is over $500,000, according to drugs.com.

A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167619</fileName> <TBEID>0C04F799.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F799</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240409T082644</QCDate> <firstPublished>20240409T094510</firstPublished> <LastPublished>20240409T094510</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240409T094510</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Alex Otto</byline> <bylineText>M. ALEXANDER OTTO, PA, MMS</bylineText> <bylineFull>M. ALEXANDER OTTO, PA, MMS</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>The US Food and Drug Administration (FDA) has approved idecabtagene vicleucel (ide-cel) (Abecma, Bristol-Myers Squibb/2seventy bio) for adults with relapsed or </metaDescription> <articlePDF/> <teaserImage/> <teaser>The FDA greenlighted a costly CAR T-cell therapy for earlier lines in the treatment of adults with relapsed/refractory myeloma. </teaser> <title>Abecma Approved for Earlier Lines in Relapsed/Refractory Multiple Myeloma</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>hemn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">18</term> </publications> <sections> <term canonical="true">37225</term> <term>39313</term> </sections> <topics> <term>195</term> <term canonical="true">250</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Abecma Approved for Earlier Lines in Relapsed/Refractory Multiple Myeloma</title> <deck/> </itemMeta> <itemContent> <p><br/><br/><span class="tag metaDescription">The US Food and Drug Administration (FDA) has approved <span class="Hyperlink"><a href="https://reference.medscape.com/drug/abecma-idecabtagene-vicleucel-4000133">idecabtagene vicleucel</a></span> (ide-cel) (Abecma, Bristol-Myers Squibb/2seventy bio) for adults with relapsed or refractory multiple myeloma after two or more prior lines of therapy, including an immunomodulatory agent, a proteasome inhibitor, and an anti-CD38 monoclonal antibody. </span><br/><br/>The approval expands the chimeric antigen receptor (CAR) T-cell therapy’s indications to earlier lines of treatment after exposure to these other main therapy classes, Bristol Myers Squibb said in a <span class="Hyperlink"><a href="https://news.bms.com/news/corporate-financial/2024/U.S.-FDA-Approves-Bristol-Myers-Squibb-and-2seventy-bios-Abecma-for-Triple-Class-Exposed-Relapsed-or-Refractory-Multiple-Myeloma-After-Two-Prior-Lines-of-Therapy/default.aspx">press release</a></span>. <br/><br/>Approval was based on the KarMMa-3 trial, in which 254 patients were randomly assigned to ide-cel and 132 to investigators’ choice of standard regimens, consisting of combinations of <span class="Hyperlink">daratumumab</span>, <span class="Hyperlink">dexamethasone</span>, and other agents. <br/><br/>After a median follow-up of 15.9 months, median progression-free survival was three times higher in the ide-cel arm: 13.3 months with the CAR T-cell therapy vs 4.4 months with standard treatment. Overall, 39% of patients on ide-cel had a complete response vs 5% on standard regimens. <br/><br/>The approval includes a new recommended dose range of 300-510 x 106 CAR-positive T cells. <br/><br/>Ide-cel carries <span class="Hyperlink"><a href="https://www.fda.gov/media/147055/download">a boxed warning</a></span> for <span class="Hyperlink">cytokine release syndrome</span>, neurologic toxicities, hemophagocytic <span class="Hyperlink">lymphohistiocytosis</span>/macrophage activation syndrome, prolonged cytopenia, and secondary hematologic cancers. <br/><br/>In trials, cytokine release syndrome occurred in 89% (310 of 349) of patients in the KarMMa-3 and KarMMa studies, which included grade 3 syndrome in 7% (23 of 349) and fatal cases in 0.9% (3 of 349) of patients.<br/><br/>A one-time treatment is over $500,000, according to <span class="Hyperlink"><a href="https://www.drugs.com/price-guide/abecma">drugs.com</a></span>.</p> <p> <em>A version of this article appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/abecma-approved-earlier-lines-relapsed-refractory-multiple-2024a10006no?src=">Medscape.com</a></span>.</em> </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Managing CAR-T Neurotoxicity: EEG Bests the Rest

Article Type
Changed
Wed, 04/03/2024 - 12:12

Among diagnostic interventions recommended to manage the common CAR-T cell therapy–associated side effect of immune effector cell–associated neurotoxicity syndrome (ICANS), only electroencephalogram (EEG) shows significant therapeutic benefit — while magnetic resonance imaging (MRI) and lumbar puncture appear to have limited value, new research shows.

“Our results emphasize for the first time the role of EEG in the current guidelines [for ICANS] but question the need for systematic MRI and lumbar puncture,” reported the authors of the study, published in Blood Advances.

The study underscores that “EEG does more that depict insignificant anomalies and plays a key role in patient management in daily practice,” first author Mattéo Mauget, said in an interview. He is a resident in the intensive care unit at the University Hospital of Rennes in France.

ICANS is among the most common of acute neurotoxicities occurring after CAR T-cell therapy, and international guidelines recommend MRI, lumbar puncture, and EEG in the management of the toxicity, which is typically treated with anti-cytokine therapy and steroids.

However, the guidelines widely vary. All recommend the use of MRI for ICANS grade 3 or higher, but fewer recommend the approach for grade 2. Meanwhile, only some recommend the use of lumbar puncture, and even fewer guidelines recommend the use of EEG.

While these measures are expensive — and in the case of lumbar puncture, invasive and burdensome for patients — the recommendations on these measures “rely on empirical practices and are only based on expert opinions with low scientific evidence,” the authors wrote.

To evaluate the interventions in a cohort of real-life patients treated with CAR T-cell therapy, the authors identified 190 consecutive patients receiving the therapy at the University Hospital of Rennes, France, between August 2018 and January 2023.

Of the patients, 62% were male and their median age was 64. Overall, 91 (48%) developed ICANS.

The majority of patients (73%) received CAR-T cell therapy for a refractory/relapsed (R/R) DLBCL (73%), and most (60%) had received the CAR-T product axicabtagene-ciloleucel (axi-cel) after two or more prior therapies.

While MRI was performed in 78% of patients with ICANS, the measure was determined to have had a therapeutic impact in just 4% of patients, despite common observations of abnormal findings.

Lumbar puncture was meanwhile performed in 47% of patients, resulting in preemptive antimicrobial agents in 7% of patients, with no infection detected.

While systematic EEG was performed in 56% of patients, the intervention led to therapeutic modifications among 16% of those patients.

“Our findings highlight some divergences between guidelines and daily practice regarding diagnostic investigations,” the authors noted.

The study “shows that EEG is the diagnostic investigation with the greatest therapeutic impact, while MRI and lumbar puncture appear to have a limited therapeutic impact,” they concluded.
 

EEG Findings

Of note, only 18% of EEGs in the cohort were normal, ranging from 50% of those with ICANS grade 1 to 6% among those with ICANS grade 4.

Encephalopathy was the most common EEG finding, observed in 45% of patients, while 6 EEGs (12%) showed seizures or status epilepticus.

Two patients with ICANS grade 2 and 3 (6% of EEG) developed seizure or status epilepticus on their EEGs, despite the absence of clinical symptoms of epilepsy, while the rate was 4 (33%) among patients with ICANS grade 4.

Among the eight (16%) patients who received therapeutic modification as the result of the EEG, seven were in the severe and life-threatening ICANS (grade 3+) group (24%).

In addition, all EEGs detecting seizure or status epilepticus resulted in an increase in antiepileptic prophylaxis with levetiracetam or the introduction of a new antiepileptics, mainly phenytoin.

Surprisingly, there were no cases of diffuse edema in the entire cohort, even among those with grade 4 ICANS, which is one of the key concerns of treating physicians managing severe ICANS, the authors noted.

A notable caveat is that EEG can be a time- and physician-consuming examination not easily accessed on a 24/7 daily practice level.

With such challenges, “[we] advocate for a close partnership between hematologists and electrophysiologists to make EEG access as easy as possible for this kind of patient, as EEG is a key game changer in patient course,” Mr. Mauget said.

Commenting on the findings, Marcela V. Maus, MD, PhD, director of the Cellular Immunotherapy Program at the Massachusetts General Hospital Cancer Center in Boston, agreed that the study adds importantly to a topic in need of more data.

“This is a very interesting study that starts to provide data behind the consensus recommendations that were initially made based purely on expert opinion and collective practices,” she said in an interview.

“I think [the EEG findings] are interesting, because EEG is often the most non-specific of these tests, and I would not have predicted this result. I also think that monitoring of cerebral spinal fluid [through lumbar puncture] could have potentially higher impact if there was a way to routinely quantify and detect the CAR-T cells,” Dr. Maus said.

“Although admittedly I think this may be of greater benefit when patients present with neurologic findings outside the typical window of ICANS, such as what can occur with delayed neurologic toxicities such as Parkinsonism after BCMA-directed CAR T cells,” she added.

Senior author Guillaume Manson, MD, a hematologist also with the University Hospital of Rennes, underscored that the results shouldn’t be construed to suggest that MRI or LP should not be used in such cases, but may often not be necessary.

“Every patient’s case is different, and these findings certainly do not say that certain tests should or should not be performed,” he said in a press statement.

“We did this research to generate clinical evidence to inform guidelines that support physicians in making clinical decisions when treating patients with these complex, and sometimes severe conditions,” he added.

Dr. Manson reported relationships with BMS-Celgene, Gilead-Kite, and Takeda. Dr. Maus disclosed ties with Century Therapeutics, TCR2, Kite/Gilead, Novartis, and several other companies in the field of cellular therapies.

Publications
Topics
Sections

Among diagnostic interventions recommended to manage the common CAR-T cell therapy–associated side effect of immune effector cell–associated neurotoxicity syndrome (ICANS), only electroencephalogram (EEG) shows significant therapeutic benefit — while magnetic resonance imaging (MRI) and lumbar puncture appear to have limited value, new research shows.

“Our results emphasize for the first time the role of EEG in the current guidelines [for ICANS] but question the need for systematic MRI and lumbar puncture,” reported the authors of the study, published in Blood Advances.

The study underscores that “EEG does more that depict insignificant anomalies and plays a key role in patient management in daily practice,” first author Mattéo Mauget, said in an interview. He is a resident in the intensive care unit at the University Hospital of Rennes in France.

ICANS is among the most common of acute neurotoxicities occurring after CAR T-cell therapy, and international guidelines recommend MRI, lumbar puncture, and EEG in the management of the toxicity, which is typically treated with anti-cytokine therapy and steroids.

However, the guidelines widely vary. All recommend the use of MRI for ICANS grade 3 or higher, but fewer recommend the approach for grade 2. Meanwhile, only some recommend the use of lumbar puncture, and even fewer guidelines recommend the use of EEG.

While these measures are expensive — and in the case of lumbar puncture, invasive and burdensome for patients — the recommendations on these measures “rely on empirical practices and are only based on expert opinions with low scientific evidence,” the authors wrote.

To evaluate the interventions in a cohort of real-life patients treated with CAR T-cell therapy, the authors identified 190 consecutive patients receiving the therapy at the University Hospital of Rennes, France, between August 2018 and January 2023.

Of the patients, 62% were male and their median age was 64. Overall, 91 (48%) developed ICANS.

The majority of patients (73%) received CAR-T cell therapy for a refractory/relapsed (R/R) DLBCL (73%), and most (60%) had received the CAR-T product axicabtagene-ciloleucel (axi-cel) after two or more prior therapies.

While MRI was performed in 78% of patients with ICANS, the measure was determined to have had a therapeutic impact in just 4% of patients, despite common observations of abnormal findings.

Lumbar puncture was meanwhile performed in 47% of patients, resulting in preemptive antimicrobial agents in 7% of patients, with no infection detected.

While systematic EEG was performed in 56% of patients, the intervention led to therapeutic modifications among 16% of those patients.

“Our findings highlight some divergences between guidelines and daily practice regarding diagnostic investigations,” the authors noted.

The study “shows that EEG is the diagnostic investigation with the greatest therapeutic impact, while MRI and lumbar puncture appear to have a limited therapeutic impact,” they concluded.
 

EEG Findings

Of note, only 18% of EEGs in the cohort were normal, ranging from 50% of those with ICANS grade 1 to 6% among those with ICANS grade 4.

Encephalopathy was the most common EEG finding, observed in 45% of patients, while 6 EEGs (12%) showed seizures or status epilepticus.

Two patients with ICANS grade 2 and 3 (6% of EEG) developed seizure or status epilepticus on their EEGs, despite the absence of clinical symptoms of epilepsy, while the rate was 4 (33%) among patients with ICANS grade 4.

Among the eight (16%) patients who received therapeutic modification as the result of the EEG, seven were in the severe and life-threatening ICANS (grade 3+) group (24%).

In addition, all EEGs detecting seizure or status epilepticus resulted in an increase in antiepileptic prophylaxis with levetiracetam or the introduction of a new antiepileptics, mainly phenytoin.

Surprisingly, there were no cases of diffuse edema in the entire cohort, even among those with grade 4 ICANS, which is one of the key concerns of treating physicians managing severe ICANS, the authors noted.

A notable caveat is that EEG can be a time- and physician-consuming examination not easily accessed on a 24/7 daily practice level.

With such challenges, “[we] advocate for a close partnership between hematologists and electrophysiologists to make EEG access as easy as possible for this kind of patient, as EEG is a key game changer in patient course,” Mr. Mauget said.

Commenting on the findings, Marcela V. Maus, MD, PhD, director of the Cellular Immunotherapy Program at the Massachusetts General Hospital Cancer Center in Boston, agreed that the study adds importantly to a topic in need of more data.

“This is a very interesting study that starts to provide data behind the consensus recommendations that were initially made based purely on expert opinion and collective practices,” she said in an interview.

“I think [the EEG findings] are interesting, because EEG is often the most non-specific of these tests, and I would not have predicted this result. I also think that monitoring of cerebral spinal fluid [through lumbar puncture] could have potentially higher impact if there was a way to routinely quantify and detect the CAR-T cells,” Dr. Maus said.

“Although admittedly I think this may be of greater benefit when patients present with neurologic findings outside the typical window of ICANS, such as what can occur with delayed neurologic toxicities such as Parkinsonism after BCMA-directed CAR T cells,” she added.

Senior author Guillaume Manson, MD, a hematologist also with the University Hospital of Rennes, underscored that the results shouldn’t be construed to suggest that MRI or LP should not be used in such cases, but may often not be necessary.

“Every patient’s case is different, and these findings certainly do not say that certain tests should or should not be performed,” he said in a press statement.

“We did this research to generate clinical evidence to inform guidelines that support physicians in making clinical decisions when treating patients with these complex, and sometimes severe conditions,” he added.

Dr. Manson reported relationships with BMS-Celgene, Gilead-Kite, and Takeda. Dr. Maus disclosed ties with Century Therapeutics, TCR2, Kite/Gilead, Novartis, and several other companies in the field of cellular therapies.

Among diagnostic interventions recommended to manage the common CAR-T cell therapy–associated side effect of immune effector cell–associated neurotoxicity syndrome (ICANS), only electroencephalogram (EEG) shows significant therapeutic benefit — while magnetic resonance imaging (MRI) and lumbar puncture appear to have limited value, new research shows.

“Our results emphasize for the first time the role of EEG in the current guidelines [for ICANS] but question the need for systematic MRI and lumbar puncture,” reported the authors of the study, published in Blood Advances.

The study underscores that “EEG does more that depict insignificant anomalies and plays a key role in patient management in daily practice,” first author Mattéo Mauget, said in an interview. He is a resident in the intensive care unit at the University Hospital of Rennes in France.

ICANS is among the most common of acute neurotoxicities occurring after CAR T-cell therapy, and international guidelines recommend MRI, lumbar puncture, and EEG in the management of the toxicity, which is typically treated with anti-cytokine therapy and steroids.

However, the guidelines widely vary. All recommend the use of MRI for ICANS grade 3 or higher, but fewer recommend the approach for grade 2. Meanwhile, only some recommend the use of lumbar puncture, and even fewer guidelines recommend the use of EEG.

While these measures are expensive — and in the case of lumbar puncture, invasive and burdensome for patients — the recommendations on these measures “rely on empirical practices and are only based on expert opinions with low scientific evidence,” the authors wrote.

To evaluate the interventions in a cohort of real-life patients treated with CAR T-cell therapy, the authors identified 190 consecutive patients receiving the therapy at the University Hospital of Rennes, France, between August 2018 and January 2023.

Of the patients, 62% were male and their median age was 64. Overall, 91 (48%) developed ICANS.

The majority of patients (73%) received CAR-T cell therapy for a refractory/relapsed (R/R) DLBCL (73%), and most (60%) had received the CAR-T product axicabtagene-ciloleucel (axi-cel) after two or more prior therapies.

While MRI was performed in 78% of patients with ICANS, the measure was determined to have had a therapeutic impact in just 4% of patients, despite common observations of abnormal findings.

Lumbar puncture was meanwhile performed in 47% of patients, resulting in preemptive antimicrobial agents in 7% of patients, with no infection detected.

While systematic EEG was performed in 56% of patients, the intervention led to therapeutic modifications among 16% of those patients.

“Our findings highlight some divergences between guidelines and daily practice regarding diagnostic investigations,” the authors noted.

The study “shows that EEG is the diagnostic investigation with the greatest therapeutic impact, while MRI and lumbar puncture appear to have a limited therapeutic impact,” they concluded.
 

EEG Findings

Of note, only 18% of EEGs in the cohort were normal, ranging from 50% of those with ICANS grade 1 to 6% among those with ICANS grade 4.

Encephalopathy was the most common EEG finding, observed in 45% of patients, while 6 EEGs (12%) showed seizures or status epilepticus.

Two patients with ICANS grade 2 and 3 (6% of EEG) developed seizure or status epilepticus on their EEGs, despite the absence of clinical symptoms of epilepsy, while the rate was 4 (33%) among patients with ICANS grade 4.

Among the eight (16%) patients who received therapeutic modification as the result of the EEG, seven were in the severe and life-threatening ICANS (grade 3+) group (24%).

In addition, all EEGs detecting seizure or status epilepticus resulted in an increase in antiepileptic prophylaxis with levetiracetam or the introduction of a new antiepileptics, mainly phenytoin.

Surprisingly, there were no cases of diffuse edema in the entire cohort, even among those with grade 4 ICANS, which is one of the key concerns of treating physicians managing severe ICANS, the authors noted.

A notable caveat is that EEG can be a time- and physician-consuming examination not easily accessed on a 24/7 daily practice level.

With such challenges, “[we] advocate for a close partnership between hematologists and electrophysiologists to make EEG access as easy as possible for this kind of patient, as EEG is a key game changer in patient course,” Mr. Mauget said.

Commenting on the findings, Marcela V. Maus, MD, PhD, director of the Cellular Immunotherapy Program at the Massachusetts General Hospital Cancer Center in Boston, agreed that the study adds importantly to a topic in need of more data.

“This is a very interesting study that starts to provide data behind the consensus recommendations that were initially made based purely on expert opinion and collective practices,” she said in an interview.

“I think [the EEG findings] are interesting, because EEG is often the most non-specific of these tests, and I would not have predicted this result. I also think that monitoring of cerebral spinal fluid [through lumbar puncture] could have potentially higher impact if there was a way to routinely quantify and detect the CAR-T cells,” Dr. Maus said.

“Although admittedly I think this may be of greater benefit when patients present with neurologic findings outside the typical window of ICANS, such as what can occur with delayed neurologic toxicities such as Parkinsonism after BCMA-directed CAR T cells,” she added.

Senior author Guillaume Manson, MD, a hematologist also with the University Hospital of Rennes, underscored that the results shouldn’t be construed to suggest that MRI or LP should not be used in such cases, but may often not be necessary.

“Every patient’s case is different, and these findings certainly do not say that certain tests should or should not be performed,” he said in a press statement.

“We did this research to generate clinical evidence to inform guidelines that support physicians in making clinical decisions when treating patients with these complex, and sometimes severe conditions,” he added.

Dr. Manson reported relationships with BMS-Celgene, Gilead-Kite, and Takeda. Dr. Maus disclosed ties with Century Therapeutics, TCR2, Kite/Gilead, Novartis, and several other companies in the field of cellular therapies.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167447</fileName> <TBEID>0C04F39E.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F39E</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>EEG in ICANS CAR-T</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240326T115705</QCDate> <firstPublished>20240326T121108</firstPublished> <LastPublished>20240326T121108</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240326T121108</CMSDate> <articleSource>FROM BLOOD ADVANCES</articleSource> <facebookInfo/> <meetingNumber/> <byline>Nancy A. Melville</byline> <bylineText>NANCY A. MELVILLE</bylineText> <bylineFull>NANCY A. MELVILLE</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>Among diagnostic interventions recommended to manage the common CAR-T cell therapy–associated side effect of immune effector cell–associated neurotoxicity syndr</metaDescription> <articlePDF/> <teaserImage/> <teaser>In managing the ICANS side effect, electroencephalogram is associated with greater therapeutic intervention versus MRI and lumbar puncture. </teaser> <title>Managing CAR-T Neurotoxicity: EEG Bests the Rest</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>hemn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">18</term> </publications> <sections> <term canonical="true">27970</term> <term>39313</term> </sections> <topics> <term canonical="true">195</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Managing CAR-T Neurotoxicity: EEG Bests the Rest</title> <deck/> </itemMeta> <itemContent> <p> <span class="tag metaDescription">Among diagnostic interventions recommended to manage the common CAR-T cell therapy–associated side effect of immune effector cell–associated neurotoxicity syndrome (ICANS), only electroencephalogram (EEG) shows significant therapeutic benefit — while magnetic resonance imaging (MRI) and lumbar puncture appear to have limited value, new research shows. </span> </p> <p>“Our results emphasize for the first time the role of EEG in the current guidelines [for ICANS] but question the need for systematic MRI and lumbar puncture,” reported the authors of the <span class="Hyperlink"><a href="https://ashpublications.org/bloodadvances/article/doi/10.1182/bloodadvances.2023011669/515343/Impact-of-diagnostic-investigations-in-the">study, published </a></span>in <em>Blood Advances</em>.<br/><br/>The study underscores that “EEG does more that depict insignificant anomalies and plays a key role in patient management in daily practice,” first author Mattéo Mauget, said in an interview. He is a resident in the intensive care unit at the University Hospital of Rennes in France.<br/><br/>ICANS is among the most common of acute neurotoxicities occurring after CAR T-cell therapy, and international guidelines recommend MRI, lumbar puncture, and EEG in the management of the toxicity, which is typically treated with anti-cytokine therapy and steroids.<br/><br/>However, the guidelines widely vary. All recommend the use of MRI for ICANS grade 3 or higher, but fewer recommend the approach for grade 2. Meanwhile, only some recommend the use of lumbar puncture, and even fewer guidelines recommend the use of EEG.<br/><br/>While these measures are expensive — and in the case of lumbar puncture, invasive and burdensome for patients — the recommendations on these measures “rely on empirical practices and are only based on expert opinions with low scientific evidence,” the authors wrote.<br/><br/>To evaluate the interventions in a cohort of real-life patients treated with CAR T-cell therapy, the authors identified 190 consecutive patients receiving the therapy at the University Hospital of Rennes, France, between August 2018 and January 2023.<br/><br/>Of the patients, 62% were male and their median age was 64. Overall, 91 (48%) developed ICANS. <br/><br/>The majority of patients (73%) received CAR-T cell therapy for a refractory/relapsed (R/R) DLBCL (73%), and most (60%) had received the CAR-T product axicabtagene-ciloleucel (axi-cel) after two or more prior therapies. <br/><br/>While MRI was performed in 78% of patients with ICANS, the measure was determined to have had a therapeutic impact in just 4% of patients, despite common observations of abnormal findings. <br/><br/>Lumbar puncture was meanwhile performed in 47% of patients, resulting in preemptive antimicrobial agents in 7% of patients, with no infection detected. <br/><br/>While systematic EEG was performed in 56% of patients, the intervention led to therapeutic modifications among 16% of those patients. <br/><br/>“Our findings highlight some divergences between guidelines and daily practice regarding diagnostic investigations,” the authors noted.<br/><br/>The study “shows that EEG is the diagnostic investigation with the greatest therapeutic impact, while MRI and lumbar puncture appear to have a limited therapeutic impact,” they concluded.<br/><br/></p> <h2>EEG Findings</h2> <p>Of note, only 18% of EEGs in the cohort were normal, ranging from 50% of those with ICANS grade 1 to 6% among those with ICANS grade 4.</p> <p>Encephalopathy was the most common EEG finding, observed in 45% of patients, while 6 EEGs (12%) showed seizures or status epilepticus. <br/><br/>Two patients with ICANS grade 2 and 3 (6% of EEG) developed seizure or status epilepticus on their EEGs, despite the absence of clinical symptoms of epilepsy, while the rate was 4 (33%) among patients with ICANS grade 4. <br/><br/>Among the eight (16%) patients who received therapeutic modification as the result of the EEG, seven were in the severe and life-threatening ICANS (grade 3+) group (24%). <br/><br/>In addition, all EEGs detecting seizure or status epilepticus resulted in an increase in antiepileptic prophylaxis with levetiracetam or the introduction of a new antiepileptics, mainly phenytoin. <br/><br/>Surprisingly, there were no cases of diffuse edema in the entire cohort, even among those with grade 4 ICANS, which is one of the key concerns of treating physicians managing severe ICANS, the authors noted.<br/><br/>A notable caveat is that EEG can be a time- and physician-consuming examination not easily accessed on a 24/7 daily practice level. <br/><br/>With such challenges, “[we] advocate for a close partnership between hematologists and electrophysiologists to make EEG access as easy as possible for this kind of patient, as EEG is a key game changer in patient course,” Mr. Mauget said.<br/><br/>Commenting on the findings, Marcela V. Maus, MD, PhD, director of the Cellular Immunotherapy Program at the Massachusetts General Hospital Cancer Center in Boston, agreed that the study adds importantly to a topic in need of more data.<br/><br/>“This is a very interesting study that starts to provide data behind the consensus recommendations that were initially made based purely on expert opinion and collective practices,” she said in an interview.<br/><br/>“I think [the EEG findings] are interesting, because EEG is often the most non-specific of these tests, and I would not have predicted this result. I also think that monitoring of cerebral spinal fluid [through lumbar puncture] could have potentially higher impact if there was a way to routinely quantify and detect the CAR-T cells,” Dr. Maus said.<br/><br/>“Although admittedly I think this may be of greater benefit when patients present with neurologic findings outside the typical window of ICANS, such as what can occur with delayed neurologic toxicities such as Parkinsonism after BCMA-directed CAR T cells,” she added.<br/><br/>Senior author Guillaume Manson, MD, a hematologist also with the University Hospital of Rennes, underscored that the results shouldn’t be construed to suggest that MRI or LP should not be used in such cases, but may often not be necessary.<br/><br/>“Every patient’s case is different, and these findings certainly do not say that certain tests should or should not be performed,” he said in a press statement.<br/><br/>“We did this research to generate clinical evidence to inform guidelines that support physicians in making clinical decisions when treating patients with these complex, and sometimes severe conditions,” he added.<br/><br/>Dr. Manson reported relationships with BMS-Celgene, Gilead-Kite, and Takeda. Dr. Maus disclosed ties with Century Therapeutics, TCR2, Kite/Gilead, Novartis, and several other companies in the field of cellular therapies.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM BLOOD ADVANCES

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Secondary Cancers Post CAR T Therapy: A Concern?

Article Type
Changed
Wed, 03/20/2024 - 15:23

 

TOPLINE:

Secondary cancers were flagged in 4.3% of all adverse event reports among patients who received CAR T therapy, with T-cell malignancies comprising only 0.15% of the total reports and 3.54% of all second primary malignancies, according to an analysis of adverse event reports submitted to the US Food and Drug Administration (FDA).

METHODOLOGY:

  • In November 2023, the FDA announced its investigation into whether chimeric antigen receptor (CAR) T-cell immunotherapies can cause secondary blood cancers, specifically T-cell malignancies. At the time, the agency said: “Although the overall benefits of these products continue to outweigh their potential risks for their approved uses, FDA is investigating the identified risk of T-cell malignancy with serious outcomes.”
  • In January 2024, the FDA issued boxed warnings on the six approved CART cell therapies, citing the possibility of second primary malignancies, including CAR-positive lymphomas, in patients who had received a CAR T agent.
  • To evaluate the extent of these secondary cancers, researchers analyzed the FDA Adverse Event Reporting System database for CAR T-cell reports citing second primary malignancies.

TAKEAWAY:

  • Overall, the authors identified 12,394 unique adverse events associated with CAR T therapy; of these, 536 adverse events (4.3%) were second primary malignancies.
  • Axicabtagene ciloleucel (axi-cel) and tisagenlecleucel (tis-cel) accounted for most of the second primary malignancies reports — 51.7% (277 of 536 patients) for axi-cel and 33% (177 of 536 patients) for tis-cel.
  • The researchers identified 19 cases of T-cell malignancies, representing only 0.15% of all unique adverse events and 3.54% of all second primary malignancies (19 of 536 patients); 17 of these cases were T-cell non-Hodgkin lymphomas, and two were T-cell large granular lymphocytic leukemia.
  • Among the reported 536 second primary malignancies, the most frequent cancers were leukemias (333 reports, or 62%), followed by skin neoplasms (54 reports, or 10.1%), hematopoietic neoplasms excluding leukemias and lymphomas (26 reports, 4.85%), nervous system tumors (21 reports, 3.92%), and respiratory neoplasms (20 reports, 3.73%).

IN PRACTICE:

“We will continue to monitor the data released by the FDA to learn more about CAR T-associated risks. However, it’s crucial to stress that the benefits of CAR T-cell therapies still outweigh the risks for the approved indications,” Magdi Elsallab, MD, the study’s co-lead author, said in a news release.

SOURCE:

This work, led by Dr. Elsallab from Harvard Medical School in Boston, was published online on March 14 in Blood.

LIMITATIONS:

The limitations of the analysis include the presence of duplicate report submissions, incomplete data, difficulty establishing causal relationships, and the potential for both underreporting and overreporting based on the severity of adverse events. Furthermore, without the total number of prescribed products, it was difficult to determine the adverse event frequency.

DISCLOSURES:

The study funding source was not disclosed. Some of the authors reported financial ties with various organizations outside this work, including Bristol Myers Squibb, Janssen Biotech, Johnson & Johnson, Kite Pharma, and Novartis.
 

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

 

TOPLINE:

Secondary cancers were flagged in 4.3% of all adverse event reports among patients who received CAR T therapy, with T-cell malignancies comprising only 0.15% of the total reports and 3.54% of all second primary malignancies, according to an analysis of adverse event reports submitted to the US Food and Drug Administration (FDA).

METHODOLOGY:

  • In November 2023, the FDA announced its investigation into whether chimeric antigen receptor (CAR) T-cell immunotherapies can cause secondary blood cancers, specifically T-cell malignancies. At the time, the agency said: “Although the overall benefits of these products continue to outweigh their potential risks for their approved uses, FDA is investigating the identified risk of T-cell malignancy with serious outcomes.”
  • In January 2024, the FDA issued boxed warnings on the six approved CART cell therapies, citing the possibility of second primary malignancies, including CAR-positive lymphomas, in patients who had received a CAR T agent.
  • To evaluate the extent of these secondary cancers, researchers analyzed the FDA Adverse Event Reporting System database for CAR T-cell reports citing second primary malignancies.

TAKEAWAY:

  • Overall, the authors identified 12,394 unique adverse events associated with CAR T therapy; of these, 536 adverse events (4.3%) were second primary malignancies.
  • Axicabtagene ciloleucel (axi-cel) and tisagenlecleucel (tis-cel) accounted for most of the second primary malignancies reports — 51.7% (277 of 536 patients) for axi-cel and 33% (177 of 536 patients) for tis-cel.
  • The researchers identified 19 cases of T-cell malignancies, representing only 0.15% of all unique adverse events and 3.54% of all second primary malignancies (19 of 536 patients); 17 of these cases were T-cell non-Hodgkin lymphomas, and two were T-cell large granular lymphocytic leukemia.
  • Among the reported 536 second primary malignancies, the most frequent cancers were leukemias (333 reports, or 62%), followed by skin neoplasms (54 reports, or 10.1%), hematopoietic neoplasms excluding leukemias and lymphomas (26 reports, 4.85%), nervous system tumors (21 reports, 3.92%), and respiratory neoplasms (20 reports, 3.73%).

IN PRACTICE:

“We will continue to monitor the data released by the FDA to learn more about CAR T-associated risks. However, it’s crucial to stress that the benefits of CAR T-cell therapies still outweigh the risks for the approved indications,” Magdi Elsallab, MD, the study’s co-lead author, said in a news release.

SOURCE:

This work, led by Dr. Elsallab from Harvard Medical School in Boston, was published online on March 14 in Blood.

LIMITATIONS:

The limitations of the analysis include the presence of duplicate report submissions, incomplete data, difficulty establishing causal relationships, and the potential for both underreporting and overreporting based on the severity of adverse events. Furthermore, without the total number of prescribed products, it was difficult to determine the adverse event frequency.

DISCLOSURES:

The study funding source was not disclosed. Some of the authors reported financial ties with various organizations outside this work, including Bristol Myers Squibb, Janssen Biotech, Johnson & Johnson, Kite Pharma, and Novartis.
 

A version of this article appeared on Medscape.com.

 

TOPLINE:

Secondary cancers were flagged in 4.3% of all adverse event reports among patients who received CAR T therapy, with T-cell malignancies comprising only 0.15% of the total reports and 3.54% of all second primary malignancies, according to an analysis of adverse event reports submitted to the US Food and Drug Administration (FDA).

METHODOLOGY:

  • In November 2023, the FDA announced its investigation into whether chimeric antigen receptor (CAR) T-cell immunotherapies can cause secondary blood cancers, specifically T-cell malignancies. At the time, the agency said: “Although the overall benefits of these products continue to outweigh their potential risks for their approved uses, FDA is investigating the identified risk of T-cell malignancy with serious outcomes.”
  • In January 2024, the FDA issued boxed warnings on the six approved CART cell therapies, citing the possibility of second primary malignancies, including CAR-positive lymphomas, in patients who had received a CAR T agent.
  • To evaluate the extent of these secondary cancers, researchers analyzed the FDA Adverse Event Reporting System database for CAR T-cell reports citing second primary malignancies.

TAKEAWAY:

  • Overall, the authors identified 12,394 unique adverse events associated with CAR T therapy; of these, 536 adverse events (4.3%) were second primary malignancies.
  • Axicabtagene ciloleucel (axi-cel) and tisagenlecleucel (tis-cel) accounted for most of the second primary malignancies reports — 51.7% (277 of 536 patients) for axi-cel and 33% (177 of 536 patients) for tis-cel.
  • The researchers identified 19 cases of T-cell malignancies, representing only 0.15% of all unique adverse events and 3.54% of all second primary malignancies (19 of 536 patients); 17 of these cases were T-cell non-Hodgkin lymphomas, and two were T-cell large granular lymphocytic leukemia.
  • Among the reported 536 second primary malignancies, the most frequent cancers were leukemias (333 reports, or 62%), followed by skin neoplasms (54 reports, or 10.1%), hematopoietic neoplasms excluding leukemias and lymphomas (26 reports, 4.85%), nervous system tumors (21 reports, 3.92%), and respiratory neoplasms (20 reports, 3.73%).

IN PRACTICE:

“We will continue to monitor the data released by the FDA to learn more about CAR T-associated risks. However, it’s crucial to stress that the benefits of CAR T-cell therapies still outweigh the risks for the approved indications,” Magdi Elsallab, MD, the study’s co-lead author, said in a news release.

SOURCE:

This work, led by Dr. Elsallab from Harvard Medical School in Boston, was published online on March 14 in Blood.

LIMITATIONS:

The limitations of the analysis include the presence of duplicate report submissions, incomplete data, difficulty establishing causal relationships, and the potential for both underreporting and overreporting based on the severity of adverse events. Furthermore, without the total number of prescribed products, it was difficult to determine the adverse event frequency.

DISCLOSURES:

The study funding source was not disclosed. Some of the authors reported financial ties with various organizations outside this work, including Bristol Myers Squibb, Janssen Biotech, Johnson & Johnson, Kite Pharma, and Novartis.
 

A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167395</fileName> <TBEID>0C04F27A.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F27A</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240320T151912</QCDate> <firstPublished>20240320T152011</firstPublished> <LastPublished>20240320T152011</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240320T152010</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Deepa Varma</byline> <bylineText>DEEPA VARMA</bylineText> <bylineFull>DEEPA VARMA</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>Secondary cancers were flagged in 4.3% of all adverse event reports among patients who received CAR T therapy, with T-cell malignancies comprising only 0.15% of</metaDescription> <articlePDF/> <teaserImage/> <teaser>FDA analysis of adverse events following CAR T-cell therapy reports on the incidence of secondary blood cancers.</teaser> <title>Secondary Cancers Post CAR T Therapy: A Concern?</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>hemn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">18</term> </publications> <sections> <term canonical="true">37225</term> <term>39313</term> </sections> <topics> <term>178</term> <term>179</term> <term>181</term> <term>182</term> <term canonical="true">195</term> <term>196</term> <term>197</term> <term>243</term> <term>27442</term> <term>303</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Secondary Cancers Post CAR T Therapy: A Concern?</title> <deck/> </itemMeta> <itemContent> <h2>TOPLINE:</h2> <p> <span class="tag metaDescription">Secondary cancers were flagged in 4.3% of all adverse event reports among patients who received CAR T therapy, with T-cell malignancies comprising only 0.15% of the total reports and 3.54% of all second primary malignancies, according to an analysis of adverse event reports submitted to the US Food and Drug Administration (FDA).</span> </p> <h2>METHODOLOGY:</h2> <ul class="body"> <li>In November 2023, the FDA announced its investigation into whether chimeric antigen receptor (CAR) T-cell immunotherapies can cause secondary blood cancers, specifically T-cell malignancies. At the time, the agency said: “Although the overall benefits of these products continue to outweigh their potential risks for their approved uses, FDA is investigating the identified risk of T-cell malignancy with serious outcomes.”</li> <li>In January 2024, the FDA issued boxed warnings on the six approved CART cell therapies, citing the possibility of second primary malignancies, including CAR-positive lymphomas, in patients who had received a CAR T agent.</li> <li>To evaluate the extent of these secondary cancers, researchers analyzed the FDA Adverse Event Reporting System database for CAR T-cell reports citing second primary malignancies.</li> </ul> <h2>TAKEAWAY:</h2> <ul class="body"> <li>Overall, the authors identified 12,394 unique adverse events associated with CAR T therapy; of these, 536 adverse events (4.3%) were second primary malignancies.</li> <li><a href="https://reference.medscape.com/drug/yescarta-axicabtagene-ciloleucel-1000187">Axicabtagene ciloleucel</a> (axi-cel) and <a href="https://reference.medscape.com/drug/kymriah-tisagenlecleucel-1000169">tisagenlecleucel</a> (tis-cel) accounted for most of the second primary malignancies reports — 51.7% (277 of 536 patients) for axi-cel and 33% (177 of 536 patients) for tis-cel.</li> <li>The researchers identified 19 cases of T-cell malignancies, representing only 0.15% of all unique adverse events and 3.54% of all second primary malignancies (19 of 536 patients); 17 of these cases were T-cell non-Hodgkin lymphomas, and two were T-cell large granular lymphocytic leukemia.</li> <li>Among the reported 536 second primary malignancies, the most frequent cancers were leukemias (333 reports, or 62%), followed by skin neoplasms (54 reports, or 10.1%), hematopoietic neoplasms excluding leukemias and lymphomas (26 reports, 4.85%), nervous system tumors (21 reports, 3.92%), and respiratory neoplasms (20 reports, 3.73%).</li> </ul> <h2>IN PRACTICE:</h2> <p>“We will continue to monitor the data released by the FDA to learn more about CAR T-associated risks. However, it’s crucial to stress that the benefits of CAR T-cell therapies still outweigh the risks for the approved indications,” Magdi Elsallab, MD, the study’s co-lead author, said in a news release.</p> <h2>SOURCE:</h2> <p>This work, led by Dr. Elsallab from Harvard Medical School in Boston, was published <span class="Hyperlink"><a href="https://ashpublications.org/blood/article/doi/10.1182/blood.2024024166/515310/Second-Primary-Malignancies-After-Commercial-CAR-T">online</a></span> on March 14 in <em>Blood</em>.</p> <h2>LIMITATIONS:</h2> <p>The limitations of the analysis include the presence of duplicate report submissions, incomplete data, difficulty establishing causal relationships, and the potential for both underreporting and overreporting based on the severity of adverse events. Furthermore, without the total number of prescribed products, it was difficult to determine the adverse event frequency.</p> <h2>DISCLOSURES:</h2> <p>The study funding source was not disclosed. Some of the authors reported financial ties with various organizations outside this work, including Bristol Myers Squibb, Janssen Biotech, Johnson &amp; Johnson, Kite Pharma, and Novartis.<br/><br/></p> <p> <em>A version of this article appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/secondary-cancers-post-car-t-therapy-concern-2024a100056a">Medscape.com</a></span>.</em> </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Myeloma: FDA Advisers Greenlight Early CAR-T

Article Type
Changed
Mon, 03/18/2024 - 15:34

An advisory panel at the US Food and Drug Administration (FDA) lent support to bids that allow for earlier use of chimeric antigen receptor (CAR-T) therapies in treating multiple myeloma, while also calling for clear disclosure to patients of potential risks of these treatments.

The FDA asked its Oncologic Drugs Advisory Committee (ODAC) to vote on two separate but similar questions at the March 15 meeting. Much of their discussion centered on higher rates of deaths for patients on the CAR-T therapies during early stages of key studies.

ODAC voted 11-0 to say the risk-benefit assessment appeared favorable for a requested broadening of the patient pool for ciltacabtagene autoleucel (cilta-cel, Carvykti, Johnson & Johnson’s Janssen). J&J is seeking approval for use of the drug for adults with relapsed or refractory multiple myeloma (RRMM) who have received at least one prior line of therapy, including a proteasome inhibitor (PI) and an immunomodulatory agent (IMiD), and are refractory to lenalidomide.

ODAC voted 8-3 to say the risk-benefit assessment appeared favorable for a requested broadening of the patient pool for idecabtagene vicleucel (ide-cel, Abecma, Bristol Myers Squibb). The company is seeking approval of the drug for people with relapsed or refractory multiple myeloma (RRMM) who have received an IMiD, a PI, and an anti-CD38 antibody.

The FDA staff will consider ODAC’s votes and recommendations, but is not bound by them. Janssen’s parent company, J&J, said the FDA’s deadline for deciding on the request to change the cilta-cel label is April 5. Bristol Myers Squibb (BMS) said there is not a PDUFA deadline at this time for its application.

Both CAR-T treatments currently are approved for RRMM after 4 or more prior lines of therapy, including an IMiD, PI and an anti-CD38 monoclonal antibody. Last year BMS and Janssen filed their separate applications, both seeking to have their drugs used earlier in the course of RRMM.

Data provided in support of both requests for expanded use raised alarms at the FDA, with more deaths seen in the early stage of testing among patients given the CAR-T drugs compared to those given standard-of-care regimens, the agency staff said.

The application for cilta-cel rests heavily on the data from the CARTITUDE-4 trial. As reported in The New England Journal of Medicine last year, progression-free survival (PFS) at 12 months was 75.9% (95% CI, 69.4 to 81.1) in the cilta-cel group and 48.6% (95% CI, 41.5 to 55.3) in the standard-care group.

But the FDA staff review focused on worrying signs in the early months of this study. For example, the rate of death in the first 10 months post randomization was higher in the cilta-cel arm (29 of 208; 14%) than in the standard therapy arm (25 of 211; 12%) based on an analysis of the intent-to-treat (ITT) population, the FDA said.

In its review of the ide-cel application, the FDA staff said the median PFS was 13.3 months in the ide-cel arm (95% CI: 11.8, 16.1), and 4.4 months (95% CI: 3.4, 5.9) in the standard of care (SOC) arm.

However, the rate of deaths in the first 9 months post randomization was higher in the ide-cel arm (45/254; 18%) than in the comparator standard-of-care group (15/132; 11%) in the ITT population, the FDA staff said. In the safety analysis population, the rate of deaths from adverse events that occurred within 90 days from starting treatment was 2.7% in the ide-cel arm and 1.6 % in the standard-regimen group.

ODAC ultimately appeared more impressed by data indicating the potential benefit, measured as progression-free survival (PFS), of the two drugs under review, than they were concerned about the issues about early deaths raised by FDA staff.

Panelist Jorge J. Nieva, MD, of the University of Southern California said the CAR-T drugs may present another case of “front-loaded risk” as has been noted for other treatments for serious medical procedures, such as allogeneic transplantations and thoracic surgeries.

In response, Robert Sokolic, MD, the branch chief for malignant hematology at FDA, replied that the data raised concerns that did in fact remind him of these procedures.

“I’m a bone marrow transplant physician. And that’s exactly what I said when I saw these curves. This looks like an allogeneic transplant curve,” Dr. Sokolic said.

But there’s a major difference between that procedure and CAR-T in the context being considered at the ODAC meeting, he said.

With allogeneic transplant, physicians “counsel patients. We ask them to accept an upfront burden of increased mortality, because we know that down the line, overall, there’s a benefit in survival,” Dr. Sokolic said.

In contrast, the primary endpoint in the key studies for expansion of CAR-T drugs was progression-free survival (PFS), with overall survival as a second endpoint. The FDA staff in briefing documents noted how overall survival, the gold standard in research, delivers far more reliable answers for patients and doctors in assessing treatments.

In the exchange with Dr. Nieva, Dr. Sokolic noted that there’s far less certainty of benefit at this time when asking patients to consider CAR-T earlier in the progression of MM, especially given the safety concerns.

“We know there’s benefit in PFS. We know there’s a safety concern,” Dr. Sokolic said.“That’s not balanced by an overall survival balance on the tail end. It may be when the data are more mature, but it’s not there yet.”
 

 

 

Describing Risks to Patients

ODAC panelists also stressed a need to help patients understand what’s known — and not yet known — about these CAR-T therapies. It will be very challenging for patients to understand and interpret the data from key studies on these medicines, said ODAC panelist Susan Lattimore, RN, of Oregon Health & Science University. She suggested the FDA seek labeling that would be “overtly transparent” and use lay terms to describe the potential risks and benefits.

In its presentations to the FDA and ODAC, J&J noted that the COVID pandemic has affected testing and that the rate of deaths flips in time to be higher in the comparator group.

In its briefing document for the meeting, BMS emphasized that most of the patients in the ide-cel arm who died in the first 6 months of its trial did not get the study drug. There were 9 deaths in the standard-regimen arm, or 6.8% of the group, compared with 30, or 11.8% in the ide-cel group.

In the ide-cel arm, the majority of early deaths (17/30; 56.7%) occurred in patients who never received ide-cel treatment, with 13 of those 17 dying from disease progression, the company said in its briefing document. The early death rate among patients who received the allocated study treatment was similar between arms (5.1% in the ide-cel arm vs 6.8% in the standard regimen arm),the company said.

In the staff briefing, the FDA said the median PFS was 13.3 months in the ide-cel arm, compared with 4.4 months in the standard of care (SOC) arm. But there was a “clear and persistent increased mortality” for the ide-cel group, compared with the standard regimen arm, with increased rates of death up to 9 months. In addition, the overall survival disadvantage persisted to 15 months after randomization, when the survival curves finally crossed, the FDA staff said in its March 15 presentation.

ODAC Chairman Ravi A. Madan, MD, of the National Cancer Institute, was among the panelists who voted “no” in the ide-cel question. He said the risk-benefit profile of the drug does not appear favorable at this time for expanded use.

“There’s a lot of optimism about moving these therapies earlier in the disease states of multiple myeloma,” Dr. Madan said, calling the PFS data “quite remarkable.

“But for me this data at this level of maturity really didn’t provide convincing evidence that ide-cel earlier had a favorable risk benefit assessment in a proposed indication.”

ODAC panelist Christopher H. Lieu, MD, of the University of Colorado, said he struggled to decide how to vote on the ide-cel question and in the end voted yes.

He said the response to the treatment doesn’t appear to be as durable as hoped, considering the significant burden that CAR-T therapy imposes on patients. However, the PFS data suggest that ide-cel could offer patients with RRMM a chance for significant times off therapy with associated quality of life improvement.

“I do believe that the risk-benefit profile is favorable for this population as a whole,” he said. “But it’s a closer margin than I think we would like and patients will need to have in-depth discussions about the risks and benefits and balance that with the possible benefits with their provider.”

Publications
Topics
Sections

An advisory panel at the US Food and Drug Administration (FDA) lent support to bids that allow for earlier use of chimeric antigen receptor (CAR-T) therapies in treating multiple myeloma, while also calling for clear disclosure to patients of potential risks of these treatments.

The FDA asked its Oncologic Drugs Advisory Committee (ODAC) to vote on two separate but similar questions at the March 15 meeting. Much of their discussion centered on higher rates of deaths for patients on the CAR-T therapies during early stages of key studies.

ODAC voted 11-0 to say the risk-benefit assessment appeared favorable for a requested broadening of the patient pool for ciltacabtagene autoleucel (cilta-cel, Carvykti, Johnson & Johnson’s Janssen). J&J is seeking approval for use of the drug for adults with relapsed or refractory multiple myeloma (RRMM) who have received at least one prior line of therapy, including a proteasome inhibitor (PI) and an immunomodulatory agent (IMiD), and are refractory to lenalidomide.

ODAC voted 8-3 to say the risk-benefit assessment appeared favorable for a requested broadening of the patient pool for idecabtagene vicleucel (ide-cel, Abecma, Bristol Myers Squibb). The company is seeking approval of the drug for people with relapsed or refractory multiple myeloma (RRMM) who have received an IMiD, a PI, and an anti-CD38 antibody.

The FDA staff will consider ODAC’s votes and recommendations, but is not bound by them. Janssen’s parent company, J&J, said the FDA’s deadline for deciding on the request to change the cilta-cel label is April 5. Bristol Myers Squibb (BMS) said there is not a PDUFA deadline at this time for its application.

Both CAR-T treatments currently are approved for RRMM after 4 or more prior lines of therapy, including an IMiD, PI and an anti-CD38 monoclonal antibody. Last year BMS and Janssen filed their separate applications, both seeking to have their drugs used earlier in the course of RRMM.

Data provided in support of both requests for expanded use raised alarms at the FDA, with more deaths seen in the early stage of testing among patients given the CAR-T drugs compared to those given standard-of-care regimens, the agency staff said.

The application for cilta-cel rests heavily on the data from the CARTITUDE-4 trial. As reported in The New England Journal of Medicine last year, progression-free survival (PFS) at 12 months was 75.9% (95% CI, 69.4 to 81.1) in the cilta-cel group and 48.6% (95% CI, 41.5 to 55.3) in the standard-care group.

But the FDA staff review focused on worrying signs in the early months of this study. For example, the rate of death in the first 10 months post randomization was higher in the cilta-cel arm (29 of 208; 14%) than in the standard therapy arm (25 of 211; 12%) based on an analysis of the intent-to-treat (ITT) population, the FDA said.

In its review of the ide-cel application, the FDA staff said the median PFS was 13.3 months in the ide-cel arm (95% CI: 11.8, 16.1), and 4.4 months (95% CI: 3.4, 5.9) in the standard of care (SOC) arm.

However, the rate of deaths in the first 9 months post randomization was higher in the ide-cel arm (45/254; 18%) than in the comparator standard-of-care group (15/132; 11%) in the ITT population, the FDA staff said. In the safety analysis population, the rate of deaths from adverse events that occurred within 90 days from starting treatment was 2.7% in the ide-cel arm and 1.6 % in the standard-regimen group.

ODAC ultimately appeared more impressed by data indicating the potential benefit, measured as progression-free survival (PFS), of the two drugs under review, than they were concerned about the issues about early deaths raised by FDA staff.

Panelist Jorge J. Nieva, MD, of the University of Southern California said the CAR-T drugs may present another case of “front-loaded risk” as has been noted for other treatments for serious medical procedures, such as allogeneic transplantations and thoracic surgeries.

In response, Robert Sokolic, MD, the branch chief for malignant hematology at FDA, replied that the data raised concerns that did in fact remind him of these procedures.

“I’m a bone marrow transplant physician. And that’s exactly what I said when I saw these curves. This looks like an allogeneic transplant curve,” Dr. Sokolic said.

But there’s a major difference between that procedure and CAR-T in the context being considered at the ODAC meeting, he said.

With allogeneic transplant, physicians “counsel patients. We ask them to accept an upfront burden of increased mortality, because we know that down the line, overall, there’s a benefit in survival,” Dr. Sokolic said.

In contrast, the primary endpoint in the key studies for expansion of CAR-T drugs was progression-free survival (PFS), with overall survival as a second endpoint. The FDA staff in briefing documents noted how overall survival, the gold standard in research, delivers far more reliable answers for patients and doctors in assessing treatments.

In the exchange with Dr. Nieva, Dr. Sokolic noted that there’s far less certainty of benefit at this time when asking patients to consider CAR-T earlier in the progression of MM, especially given the safety concerns.

“We know there’s benefit in PFS. We know there’s a safety concern,” Dr. Sokolic said.“That’s not balanced by an overall survival balance on the tail end. It may be when the data are more mature, but it’s not there yet.”
 

 

 

Describing Risks to Patients

ODAC panelists also stressed a need to help patients understand what’s known — and not yet known — about these CAR-T therapies. It will be very challenging for patients to understand and interpret the data from key studies on these medicines, said ODAC panelist Susan Lattimore, RN, of Oregon Health & Science University. She suggested the FDA seek labeling that would be “overtly transparent” and use lay terms to describe the potential risks and benefits.

In its presentations to the FDA and ODAC, J&J noted that the COVID pandemic has affected testing and that the rate of deaths flips in time to be higher in the comparator group.

In its briefing document for the meeting, BMS emphasized that most of the patients in the ide-cel arm who died in the first 6 months of its trial did not get the study drug. There were 9 deaths in the standard-regimen arm, or 6.8% of the group, compared with 30, or 11.8% in the ide-cel group.

In the ide-cel arm, the majority of early deaths (17/30; 56.7%) occurred in patients who never received ide-cel treatment, with 13 of those 17 dying from disease progression, the company said in its briefing document. The early death rate among patients who received the allocated study treatment was similar between arms (5.1% in the ide-cel arm vs 6.8% in the standard regimen arm),the company said.

In the staff briefing, the FDA said the median PFS was 13.3 months in the ide-cel arm, compared with 4.4 months in the standard of care (SOC) arm. But there was a “clear and persistent increased mortality” for the ide-cel group, compared with the standard regimen arm, with increased rates of death up to 9 months. In addition, the overall survival disadvantage persisted to 15 months after randomization, when the survival curves finally crossed, the FDA staff said in its March 15 presentation.

ODAC Chairman Ravi A. Madan, MD, of the National Cancer Institute, was among the panelists who voted “no” in the ide-cel question. He said the risk-benefit profile of the drug does not appear favorable at this time for expanded use.

“There’s a lot of optimism about moving these therapies earlier in the disease states of multiple myeloma,” Dr. Madan said, calling the PFS data “quite remarkable.

“But for me this data at this level of maturity really didn’t provide convincing evidence that ide-cel earlier had a favorable risk benefit assessment in a proposed indication.”

ODAC panelist Christopher H. Lieu, MD, of the University of Colorado, said he struggled to decide how to vote on the ide-cel question and in the end voted yes.

He said the response to the treatment doesn’t appear to be as durable as hoped, considering the significant burden that CAR-T therapy imposes on patients. However, the PFS data suggest that ide-cel could offer patients with RRMM a chance for significant times off therapy with associated quality of life improvement.

“I do believe that the risk-benefit profile is favorable for this population as a whole,” he said. “But it’s a closer margin than I think we would like and patients will need to have in-depth discussions about the risks and benefits and balance that with the possible benefits with their provider.”

An advisory panel at the US Food and Drug Administration (FDA) lent support to bids that allow for earlier use of chimeric antigen receptor (CAR-T) therapies in treating multiple myeloma, while also calling for clear disclosure to patients of potential risks of these treatments.

The FDA asked its Oncologic Drugs Advisory Committee (ODAC) to vote on two separate but similar questions at the March 15 meeting. Much of their discussion centered on higher rates of deaths for patients on the CAR-T therapies during early stages of key studies.

ODAC voted 11-0 to say the risk-benefit assessment appeared favorable for a requested broadening of the patient pool for ciltacabtagene autoleucel (cilta-cel, Carvykti, Johnson & Johnson’s Janssen). J&J is seeking approval for use of the drug for adults with relapsed or refractory multiple myeloma (RRMM) who have received at least one prior line of therapy, including a proteasome inhibitor (PI) and an immunomodulatory agent (IMiD), and are refractory to lenalidomide.

ODAC voted 8-3 to say the risk-benefit assessment appeared favorable for a requested broadening of the patient pool for idecabtagene vicleucel (ide-cel, Abecma, Bristol Myers Squibb). The company is seeking approval of the drug for people with relapsed or refractory multiple myeloma (RRMM) who have received an IMiD, a PI, and an anti-CD38 antibody.

The FDA staff will consider ODAC’s votes and recommendations, but is not bound by them. Janssen’s parent company, J&J, said the FDA’s deadline for deciding on the request to change the cilta-cel label is April 5. Bristol Myers Squibb (BMS) said there is not a PDUFA deadline at this time for its application.

Both CAR-T treatments currently are approved for RRMM after 4 or more prior lines of therapy, including an IMiD, PI and an anti-CD38 monoclonal antibody. Last year BMS and Janssen filed their separate applications, both seeking to have their drugs used earlier in the course of RRMM.

Data provided in support of both requests for expanded use raised alarms at the FDA, with more deaths seen in the early stage of testing among patients given the CAR-T drugs compared to those given standard-of-care regimens, the agency staff said.

The application for cilta-cel rests heavily on the data from the CARTITUDE-4 trial. As reported in The New England Journal of Medicine last year, progression-free survival (PFS) at 12 months was 75.9% (95% CI, 69.4 to 81.1) in the cilta-cel group and 48.6% (95% CI, 41.5 to 55.3) in the standard-care group.

But the FDA staff review focused on worrying signs in the early months of this study. For example, the rate of death in the first 10 months post randomization was higher in the cilta-cel arm (29 of 208; 14%) than in the standard therapy arm (25 of 211; 12%) based on an analysis of the intent-to-treat (ITT) population, the FDA said.

In its review of the ide-cel application, the FDA staff said the median PFS was 13.3 months in the ide-cel arm (95% CI: 11.8, 16.1), and 4.4 months (95% CI: 3.4, 5.9) in the standard of care (SOC) arm.

However, the rate of deaths in the first 9 months post randomization was higher in the ide-cel arm (45/254; 18%) than in the comparator standard-of-care group (15/132; 11%) in the ITT population, the FDA staff said. In the safety analysis population, the rate of deaths from adverse events that occurred within 90 days from starting treatment was 2.7% in the ide-cel arm and 1.6 % in the standard-regimen group.

ODAC ultimately appeared more impressed by data indicating the potential benefit, measured as progression-free survival (PFS), of the two drugs under review, than they were concerned about the issues about early deaths raised by FDA staff.

Panelist Jorge J. Nieva, MD, of the University of Southern California said the CAR-T drugs may present another case of “front-loaded risk” as has been noted for other treatments for serious medical procedures, such as allogeneic transplantations and thoracic surgeries.

In response, Robert Sokolic, MD, the branch chief for malignant hematology at FDA, replied that the data raised concerns that did in fact remind him of these procedures.

“I’m a bone marrow transplant physician. And that’s exactly what I said when I saw these curves. This looks like an allogeneic transplant curve,” Dr. Sokolic said.

But there’s a major difference between that procedure and CAR-T in the context being considered at the ODAC meeting, he said.

With allogeneic transplant, physicians “counsel patients. We ask them to accept an upfront burden of increased mortality, because we know that down the line, overall, there’s a benefit in survival,” Dr. Sokolic said.

In contrast, the primary endpoint in the key studies for expansion of CAR-T drugs was progression-free survival (PFS), with overall survival as a second endpoint. The FDA staff in briefing documents noted how overall survival, the gold standard in research, delivers far more reliable answers for patients and doctors in assessing treatments.

In the exchange with Dr. Nieva, Dr. Sokolic noted that there’s far less certainty of benefit at this time when asking patients to consider CAR-T earlier in the progression of MM, especially given the safety concerns.

“We know there’s benefit in PFS. We know there’s a safety concern,” Dr. Sokolic said.“That’s not balanced by an overall survival balance on the tail end. It may be when the data are more mature, but it’s not there yet.”
 

 

 

Describing Risks to Patients

ODAC panelists also stressed a need to help patients understand what’s known — and not yet known — about these CAR-T therapies. It will be very challenging for patients to understand and interpret the data from key studies on these medicines, said ODAC panelist Susan Lattimore, RN, of Oregon Health & Science University. She suggested the FDA seek labeling that would be “overtly transparent” and use lay terms to describe the potential risks and benefits.

In its presentations to the FDA and ODAC, J&J noted that the COVID pandemic has affected testing and that the rate of deaths flips in time to be higher in the comparator group.

In its briefing document for the meeting, BMS emphasized that most of the patients in the ide-cel arm who died in the first 6 months of its trial did not get the study drug. There were 9 deaths in the standard-regimen arm, or 6.8% of the group, compared with 30, or 11.8% in the ide-cel group.

In the ide-cel arm, the majority of early deaths (17/30; 56.7%) occurred in patients who never received ide-cel treatment, with 13 of those 17 dying from disease progression, the company said in its briefing document. The early death rate among patients who received the allocated study treatment was similar between arms (5.1% in the ide-cel arm vs 6.8% in the standard regimen arm),the company said.

In the staff briefing, the FDA said the median PFS was 13.3 months in the ide-cel arm, compared with 4.4 months in the standard of care (SOC) arm. But there was a “clear and persistent increased mortality” for the ide-cel group, compared with the standard regimen arm, with increased rates of death up to 9 months. In addition, the overall survival disadvantage persisted to 15 months after randomization, when the survival curves finally crossed, the FDA staff said in its March 15 presentation.

ODAC Chairman Ravi A. Madan, MD, of the National Cancer Institute, was among the panelists who voted “no” in the ide-cel question. He said the risk-benefit profile of the drug does not appear favorable at this time for expanded use.

“There’s a lot of optimism about moving these therapies earlier in the disease states of multiple myeloma,” Dr. Madan said, calling the PFS data “quite remarkable.

“But for me this data at this level of maturity really didn’t provide convincing evidence that ide-cel earlier had a favorable risk benefit assessment in a proposed indication.”

ODAC panelist Christopher H. Lieu, MD, of the University of Colorado, said he struggled to decide how to vote on the ide-cel question and in the end voted yes.

He said the response to the treatment doesn’t appear to be as durable as hoped, considering the significant burden that CAR-T therapy imposes on patients. However, the PFS data suggest that ide-cel could offer patients with RRMM a chance for significant times off therapy with associated quality of life improvement.

“I do believe that the risk-benefit profile is favorable for this population as a whole,” he said. “But it’s a closer margin than I think we would like and patients will need to have in-depth discussions about the risks and benefits and balance that with the possible benefits with their provider.”

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167343</fileName> <TBEID>0C04F16E.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F16E</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240318T152441</QCDate> <firstPublished>20240318T153203</firstPublished> <LastPublished>20240318T153203</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240318T153203</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>K.D. Young</byline> <bylineText>KERRY DOOLEY YOUNG</bylineText> <bylineFull>KERRY DOOLEY YOUNG</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>An advisory panel at the US Food and Drug Administration (FDA) lent support to bids that allow for earlier use of chimeric antigen receptor (CAR-T) therapies in</metaDescription> <articlePDF/> <teaserImage/> <teaser>ODAC voted 11-0 in support of expanded cilta-cel use, 8-3 for expanded ide-cel use, while urging clearer warning about the risks of these drugs.</teaser> <title>Myeloma: FDA Advisers Greenlight Early CAR-T</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>hemn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">18</term> </publications> <sections> <term canonical="true">37225</term> <term>39313</term> </sections> <topics> <term canonical="true">250</term> <term>195</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Myeloma: FDA Advisers Greenlight Early CAR-T</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">An advisory panel at the US Food and Drug Administration (FDA) lent support to bids that allow for earlier use of chimeric antigen receptor (CAR-T) therapies in treating multiple myeloma, while also calling for clear disclosure to patients of potential risks of these treatments.</span><br/><br/>The FDA asked its Oncologic Drugs Advisory Committee (ODAC) to vote on two separate but similar questions at the March 15 meeting. Much of their discussion centered on higher rates of deaths for patients on the CAR-T therapies during early stages of key studies.<br/><br/>ODAC voted 11-0 to say the risk-benefit assessment appeared favorable for a requested broadening of the patient pool for ciltacabtagene autoleucel (cilta-cel, Carvykti, Johnson &amp; Johnson’s Janssen). J&amp;J is seeking approval for use of the drug for adults with relapsed or refractory multiple myeloma (RRMM) who have received at least one prior line of therapy, including a proteasome inhibitor (PI) and an immunomodulatory agent (IMiD), and are refractory to lenalidomide. <br/><br/>ODAC voted 8-3 to say the risk-benefit assessment appeared favorable for a requested broadening of the patient pool for idecabtagene vicleucel (ide-cel, Abecma, Bristol Myers Squibb). The company is seeking approval of the drug for people with relapsed or refractory multiple myeloma (RRMM) who have received an IMiD, a PI, and an anti-CD38 antibody.<br/><br/>The FDA staff will consider ODAC’s votes and recommendations, but is not bound by them. Janssen’s parent company, J&amp;J, said the FDA’s deadline for deciding on the request to change the cilta-cel label is April 5. Bristol Myers Squibb (BMS) said there is not a PDUFA deadline at this time for its application.<br/><br/>Both CAR-T treatments currently are approved for RRMM after 4 or more prior lines of therapy, including an IMiD, PI and an anti-CD38 monoclonal antibody. Last year BMS and Janssen filed their separate applications, both seeking to have their drugs used earlier in the course of RRMM.<br/><br/>Data provided in support of both requests for expanded use raised alarms at the FDA, with more deaths seen in the early stage of testing among patients given the CAR-T drugs compared to those given standard-of-care regimens, the agency staff said. <br/><br/>The application for cilta-cel rests heavily on the data from the CARTITUDE-4 trial. <span class="Hyperlink"><a href="https://www.nejm.org/doi/10.1056/NEJMoa2303379?url_ver=Z39.88-2003&amp;amp;rfr_id=ori:rid:crossref.org&amp;amp;rfr_dat=cr_pub%20%200pubmed">As reported</a></span> in <em>The New England Journal of Medicine</em> last year, progression-free survival (PFS) at 12 months was 75.9% (95% CI, 69.4 to 81.1) in the cilta-cel group and 48.6% (95% CI, 41.5 to 55.3) in the standard-care group. <br/><br/>But the FDA staff review focused on worrying signs in the early months of this study. For example, the rate of death in the first 10 months post randomization was higher in the cilta-cel arm (29 of 208; 14%) than in the standard therapy arm (25 of 211; 12%) based on an analysis of the intent-to-treat (ITT) population, the FDA said.<br/><br/>In its review of the ide-cel application, the FDA staff said the median PFS was 13.3 months in the ide-cel arm (95% CI: 11.8, 16.1), and 4.4 months (95% CI: 3.4, 5.9) in the standard of care (SOC) arm. <br/><br/>However, the rate of deaths in the first 9 months post randomization was higher in the ide-cel arm (45/254; 18%) than in the comparator standard-of-care group (15/132; 11%) in the ITT population, the FDA staff said. In the safety analysis population, the rate of deaths from adverse events that occurred within 90 days from starting treatment was 2.7% in the ide-cel arm and 1.6 % in the standard-regimen group. <br/><br/>ODAC ultimately appeared more impressed by data indicating the potential benefit, measured as progression-free survival (PFS), of the two drugs under review, than they were concerned about the issues about early deaths raised by FDA staff. <br/><br/>Panelist Jorge J. Nieva, MD, of the University of Southern California said the CAR-T drugs may present another case of “front-loaded risk” as has been noted for other treatments for serious medical procedures, such as allogeneic transplantations and thoracic surgeries.<br/><br/>In response, Robert Sokolic, MD, the branch chief for malignant hematology at FDA, replied that the data raised concerns that did in fact remind him of these procedures.<br/><br/>“I’m a bone marrow transplant physician. And that’s exactly what I said when I saw these curves. This looks like an allogeneic transplant curve,” Dr. Sokolic said.<br/><br/>But there’s a major difference between that procedure and CAR-T in the context being considered at the ODAC meeting, he said.<br/><br/>With allogeneic transplant, physicians “counsel patients. We ask them to accept an upfront burden of increased mortality, because we know that down the line, overall, there’s a benefit in survival,” Dr. Sokolic said.<br/><br/>In contrast, the primary endpoint in the key studies for expansion of CAR-T drugs was progression-free survival (PFS), with overall survival as a second endpoint. The FDA staff in briefing documents noted how overall survival, the gold standard in research, delivers far more reliable answers for patients and doctors in assessing treatments.<br/><br/>In the exchange with Dr. Nieva, Dr. Sokolic noted that there’s far less certainty of benefit at this time when asking patients to consider CAR-T earlier in the progression of MM, especially given the safety concerns.<br/><br/>“We know there’s benefit in PFS. We know there’s a safety concern,” Dr. Sokolic said.“That’s not balanced by an overall survival balance on the tail end. It may be when the data are more mature, but it’s not there yet.”<br/><br/></p> <h2>Describing Risks to Patients</h2> <p>ODAC panelists also stressed a need to help patients understand what’s known — and not yet known — about these CAR-T therapies. It will be very challenging for patients to understand and interpret the data from key studies on these medicines, said ODAC panelist Susan Lattimore, RN, of Oregon Health &amp; Science University. She suggested the FDA seek labeling that would be “overtly transparent” and use lay terms to describe the potential risks and benefits.<br/><br/>In its presentations to the FDA and ODAC, J&amp;J noted that the COVID pandemic has affected testing and that the rate of deaths flips in time to be higher in the comparator group. <br/><br/>In its briefing document for the meeting, BMS emphasized that most of the patients in the ide-cel arm who died in the first 6 months of its trial did not get the study drug. There were 9 deaths in the standard-regimen arm, or 6.8% of the group, compared with 30, or 11.8% in the ide-cel group. <br/><br/>In the ide-cel arm, the majority of early deaths (17/30; 56.7%) occurred in patients who never received ide-cel treatment, with 13 of those 17 dying from disease progression, the company said in its briefing document. The early death rate among patients who received the allocated study treatment was similar between arms (5.1% in the ide-cel arm vs 6.8% in the standard regimen arm),the company said.<br/><br/>In the staff briefing, the FDA said the median PFS was 13.3 months in the ide-cel arm, compared with 4.4 months in the standard of care (SOC) arm. But there was a “clear and persistent increased mortality” for the ide-cel group, compared with the standard regimen arm, with increased rates of death up to 9 months. In addition, the overall survival disadvantage persisted to 15 months after randomization, when the survival curves finally crossed, the FDA staff said in its March 15 presentation.<br/><br/>ODAC Chairman Ravi A. Madan, MD, of the National Cancer Institute, was among the panelists who voted “no” in the ide-cel question. He said the risk-benefit profile of the drug does not appear favorable at this time for expanded use.<br/><br/>“There’s a lot of optimism about moving these therapies earlier in the disease states of multiple myeloma,” Dr. Madan said, calling the PFS data “quite remarkable.<br/><br/>“But for me this data at this level of maturity really didn’t provide convincing evidence that ide-cel earlier had a favorable risk benefit assessment in a proposed indication.”<br/><br/>ODAC panelist Christopher H. Lieu, MD, of the University of Colorado, said he struggled to decide how to vote on the ide-cel question and in the end voted yes. <br/><br/>He said the response to the treatment doesn’t appear to be as durable as hoped, considering the significant burden that CAR-T therapy imposes on patients. However, the PFS data suggest that ide-cel could offer patients with RRMM a chance for significant times off therapy with associated quality of life improvement.<br/><br/>“I do believe that the risk-benefit profile is favorable for this population as a whole,” he said. “But it’s a closer margin than I think we would like and patients will need to have in-depth discussions about the risks and benefits and balance that with the possible benefits with their provider.”</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article