User login
Baseline steroids may reduce efficacy of checkpoint inhibitors
Immune checkpoint inhibitor therapy may be less effective if the patient is receiving corticosteroids at the time that treatment is initiated, results of a retrospective, two-center analysis suggest.
Corticosteroid use at the time of starting a PD-1 or PD-L1 inhibitor was associated with significantly reduced overall survival and progression-free survival in patients with non–small-cell lung cancer, the authors of the analysis reported.
“Prudent use of corticosteroids at the time of initiating PD-1/PD-L1 blockade is warranted,” said Matthew D. Hellmann, MD, of Memorial Sloan Kettering Cancer Center, New York, and his coauthors. The report is in the Journal of Clinical Oncology.
The retrospective analysis from Dr. Hellmann and his colleagues comprised 640 patients treated with single-agent atezolizumab, durvalumab, nivolumab, or pembrolizumab at Memorial Sloan Kettering Cancer Center or Gustave Roussy Cancer Center, Villejuif, France, between April 2011 and September 2017.
Ninety of those patients (14%) were receiving at least 10 mg of prednisone equivalent at the time the immune checkpoint inhibitor was started, a review of patient records revealed. About one-third were receiving corticosteroids for dyspnea or other respiratory symptoms. Another 21% had fatigue and 19% had brain metastases prompting corticosteroid treatment.
Corticosteroid use at the time of PD-1/PD-L1 blockade was associated with decreased progression-free survival (hazard ratio, 1.31; P = .03) and overall survival (HR, 1.66; P less than .001), as well as decreased overall response rate in a multivariable analysis adjusted for performance status, history of smoking, and history of brain metastases, the investigators reported.
Reduced efficacy also was seen in analyses that looked at each center separately. For the Memorial Sloan Kettering cohort (455 patients), baseline corticosteroid use was associated with significantly decreased overall response rate, shorter progression-free survival, and shorter overall survival.
For the smaller French cohort, (185 patients), there was a statistically nonsignificant decrease in overall response rate, along with significant reductions in progression-free survival and overall survival.
Based on these data, it may be prudent to try other pharmacologic or nonpharmacologic strategies to manage cancer symptoms if treatment with a PD-1/PD-L1 blocker is planned, Dr. Hellmann and his coauthors said.
“These strategies could enable patients to be tapered off corticosteroids before the start of PD-1/PD-L1 blockade to potentially achieve maximum benefit from these agents,” they wrote.
However, medically necessary corticosteroids should not be avoided, such as those given for management of brain metastases, they added.
Interestingly, other recent studies have suggested that patients already on PD-1/PD-L1 inhibitors do not seem to have decreased efficacy when corticosteroids are prescribed to manage emergent immune-related adverse events.
That’s “fortunate,” given that corticosteroids are a mainstay for the management of these characteristic adverse events in patients receiving immune checkpoint inhibitors, they said.
Dr. Hellmann reported research funding from Bristol-Myers Squibb, and a consulting or advisory role with AstraZeneca, Bristol-Myers Squibb, Genentech, Janssen Pharmaceuticals, MedImmune, Merck, Mirati Therapeutics, Shattuck Labs, and Syndax. Coauthors reported disclosures related to Amgen, Boehringer Ingelheim, Pfizer, and Roche, among others.
SOURCE: Arbour KC et al. J Clin Oncol. 2018 Aug 20. doi: 10.1200/JCO.2018.79.0006.
Immune checkpoint inhibitor therapy may be less effective if the patient is receiving corticosteroids at the time that treatment is initiated, results of a retrospective, two-center analysis suggest.
Corticosteroid use at the time of starting a PD-1 or PD-L1 inhibitor was associated with significantly reduced overall survival and progression-free survival in patients with non–small-cell lung cancer, the authors of the analysis reported.
“Prudent use of corticosteroids at the time of initiating PD-1/PD-L1 blockade is warranted,” said Matthew D. Hellmann, MD, of Memorial Sloan Kettering Cancer Center, New York, and his coauthors. The report is in the Journal of Clinical Oncology.
The retrospective analysis from Dr. Hellmann and his colleagues comprised 640 patients treated with single-agent atezolizumab, durvalumab, nivolumab, or pembrolizumab at Memorial Sloan Kettering Cancer Center or Gustave Roussy Cancer Center, Villejuif, France, between April 2011 and September 2017.
Ninety of those patients (14%) were receiving at least 10 mg of prednisone equivalent at the time the immune checkpoint inhibitor was started, a review of patient records revealed. About one-third were receiving corticosteroids for dyspnea or other respiratory symptoms. Another 21% had fatigue and 19% had brain metastases prompting corticosteroid treatment.
Corticosteroid use at the time of PD-1/PD-L1 blockade was associated with decreased progression-free survival (hazard ratio, 1.31; P = .03) and overall survival (HR, 1.66; P less than .001), as well as decreased overall response rate in a multivariable analysis adjusted for performance status, history of smoking, and history of brain metastases, the investigators reported.
Reduced efficacy also was seen in analyses that looked at each center separately. For the Memorial Sloan Kettering cohort (455 patients), baseline corticosteroid use was associated with significantly decreased overall response rate, shorter progression-free survival, and shorter overall survival.
For the smaller French cohort, (185 patients), there was a statistically nonsignificant decrease in overall response rate, along with significant reductions in progression-free survival and overall survival.
Based on these data, it may be prudent to try other pharmacologic or nonpharmacologic strategies to manage cancer symptoms if treatment with a PD-1/PD-L1 blocker is planned, Dr. Hellmann and his coauthors said.
“These strategies could enable patients to be tapered off corticosteroids before the start of PD-1/PD-L1 blockade to potentially achieve maximum benefit from these agents,” they wrote.
However, medically necessary corticosteroids should not be avoided, such as those given for management of brain metastases, they added.
Interestingly, other recent studies have suggested that patients already on PD-1/PD-L1 inhibitors do not seem to have decreased efficacy when corticosteroids are prescribed to manage emergent immune-related adverse events.
That’s “fortunate,” given that corticosteroids are a mainstay for the management of these characteristic adverse events in patients receiving immune checkpoint inhibitors, they said.
Dr. Hellmann reported research funding from Bristol-Myers Squibb, and a consulting or advisory role with AstraZeneca, Bristol-Myers Squibb, Genentech, Janssen Pharmaceuticals, MedImmune, Merck, Mirati Therapeutics, Shattuck Labs, and Syndax. Coauthors reported disclosures related to Amgen, Boehringer Ingelheim, Pfizer, and Roche, among others.
SOURCE: Arbour KC et al. J Clin Oncol. 2018 Aug 20. doi: 10.1200/JCO.2018.79.0006.
Immune checkpoint inhibitor therapy may be less effective if the patient is receiving corticosteroids at the time that treatment is initiated, results of a retrospective, two-center analysis suggest.
Corticosteroid use at the time of starting a PD-1 or PD-L1 inhibitor was associated with significantly reduced overall survival and progression-free survival in patients with non–small-cell lung cancer, the authors of the analysis reported.
“Prudent use of corticosteroids at the time of initiating PD-1/PD-L1 blockade is warranted,” said Matthew D. Hellmann, MD, of Memorial Sloan Kettering Cancer Center, New York, and his coauthors. The report is in the Journal of Clinical Oncology.
The retrospective analysis from Dr. Hellmann and his colleagues comprised 640 patients treated with single-agent atezolizumab, durvalumab, nivolumab, or pembrolizumab at Memorial Sloan Kettering Cancer Center or Gustave Roussy Cancer Center, Villejuif, France, between April 2011 and September 2017.
Ninety of those patients (14%) were receiving at least 10 mg of prednisone equivalent at the time the immune checkpoint inhibitor was started, a review of patient records revealed. About one-third were receiving corticosteroids for dyspnea or other respiratory symptoms. Another 21% had fatigue and 19% had brain metastases prompting corticosteroid treatment.
Corticosteroid use at the time of PD-1/PD-L1 blockade was associated with decreased progression-free survival (hazard ratio, 1.31; P = .03) and overall survival (HR, 1.66; P less than .001), as well as decreased overall response rate in a multivariable analysis adjusted for performance status, history of smoking, and history of brain metastases, the investigators reported.
Reduced efficacy also was seen in analyses that looked at each center separately. For the Memorial Sloan Kettering cohort (455 patients), baseline corticosteroid use was associated with significantly decreased overall response rate, shorter progression-free survival, and shorter overall survival.
For the smaller French cohort, (185 patients), there was a statistically nonsignificant decrease in overall response rate, along with significant reductions in progression-free survival and overall survival.
Based on these data, it may be prudent to try other pharmacologic or nonpharmacologic strategies to manage cancer symptoms if treatment with a PD-1/PD-L1 blocker is planned, Dr. Hellmann and his coauthors said.
“These strategies could enable patients to be tapered off corticosteroids before the start of PD-1/PD-L1 blockade to potentially achieve maximum benefit from these agents,” they wrote.
However, medically necessary corticosteroids should not be avoided, such as those given for management of brain metastases, they added.
Interestingly, other recent studies have suggested that patients already on PD-1/PD-L1 inhibitors do not seem to have decreased efficacy when corticosteroids are prescribed to manage emergent immune-related adverse events.
That’s “fortunate,” given that corticosteroids are a mainstay for the management of these characteristic adverse events in patients receiving immune checkpoint inhibitors, they said.
Dr. Hellmann reported research funding from Bristol-Myers Squibb, and a consulting or advisory role with AstraZeneca, Bristol-Myers Squibb, Genentech, Janssen Pharmaceuticals, MedImmune, Merck, Mirati Therapeutics, Shattuck Labs, and Syndax. Coauthors reported disclosures related to Amgen, Boehringer Ingelheim, Pfizer, and Roche, among others.
SOURCE: Arbour KC et al. J Clin Oncol. 2018 Aug 20. doi: 10.1200/JCO.2018.79.0006.
FROM THE JOURNAL OF CLINICAL ONCOLOGY
Key clinical point: Immune checkpoint inhibitors may be less effective if the patient is receiving corticosteroids at the time treatment is started.
Major finding: Corticosteroid use at the time of PD-1/PD-L1 blockade was associated with decreased progression-free survival (hazard ratio, 1.31; P = .03) and overall survival (HR, 1.66; P less than .001) in multivariate analysis.
Study details: Retrospective analysis of 640 patients treated with atezolizumab, durvalumab, nivolumab, or pembrolizumab at Memorial Sloan Kettering Cancer Center or Gustave Roussy Cancer Center.
Disclosures: The authors reported disclosures related to Amgen, AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Genentech, Janssen Pharmaceuticals, MedImmune, Merck, Mirati Therapeutics, Pfizer, Roche, Shattuck Labs, and Syndax, among others.
Source: Arbour KC et al. J Clin Oncol. 2018 Aug 20. doi: 10.1200/JCO.2018.79.0006.
Nivolumab plus ipilimumab boosts response rate in refractory esophagogastric cancer
Nivolumab alone or in combination with ipilimumab met multiple endpoints against metastatic or locally advanced chemotherapy-refractory esophagogastric cancer in the recent phase 1/2 CheckMate-032 trial, thereby opening doors to a future phase 3 trial.
The agents demonstrated “clinically meaningful antitumor activity,” reported Yelena Y. Janjigian, MD, of Memorial Sloan Kettering Cancer Center, New York, and her coauthors.
After the 2017 ATTRACTION-2 trial demonstrated improved survival rates, “nivolumab was approved in Japan for the treatment of patients with chemotherapy-refractory gastric and gastroesophageal junction [GEJ] cancers regardless of programmed death-ligand 1 [PD-L1] status,” the authors wrote in the Journal of Clinical Oncology.
Nivolumab is a checkpoint inhibitor, like pembrolizumab, which “was approved for the treatment of patients with chemotherapy-refractory PD-L1–positive gastric/GEJ cancer on the basis of the promising clinical activity observed in the KEYNOTE-059 trial,” the authors noted. Testing nivolumab in a Western population would therefore build on these previous trials. Combining nivolumab, a PD-l inhibitor, with ipilimumab, a monoclonal antibody targeting cytotoxic T-lymphocyte antigen 4, was based on “synergistic activity” reported in preclinical models, the authors wrote.
Results from the ongoing CheckMate-032 trial included 160 patients with metastatic or locally advanced chemotherapy-refractory esophageal, gastric, or gastroesophageal junction cancer treated at centers in Europe and the United States. Just under 80% of patients had received two or more prior therapies.
In the present trial, patients were given one of three treatment regimens: nivolumab 3 mg/kg every 2 weeks, nivolumab 1 mg/kg plus ipilimumab 3 mg/kg every 3 weeks for four cycles (NIVO1 + IPI3), or nivolumab 3 mg/kg plus ipilimumab 1 mg/kg every 3 weeks for four cycles (NIVO3 + IPI1). The primary endpoint was objective response rate (ORR). Secondary endpoints included 12-month progression-free survival and 12-month overall survival (OS).
Patients in the NIVO1 + IPI3 group achieved the best ORR (24%) and 12-month progression-free survival (17%) and also showed a promising 12-month OS (35%), second only to nivolumab monotherapy (39%). PD-L1 status was not predictive of treatment response.
Although NIVO1 + IPI3 was the most clinically effective, almost half (47%) of these patients also had grade 3 or higher adverse events, compared with more favorable rates of 17% and 27% for nivolumab monotherapy and NIVO3 + IPI1, respectively.
Still, the authors concluded, “on the basis of the numerically higher overall response and landmark OS rates in the NIVO1 + IPI3 arm, this combination was considered more likely to offer clinical benefit relative to currently available treatment regimens for first-line metastatic esophagogastric cancer and was selected for further evaluation in the phase 3 CheckMate-649 study (NCT02872116).” This trial, along with another to investigate nivolumab in the adjuvant setting (NCT02743494), are ongoing.
CheckMate-032 was supported by Bristol-Myers Squibb. The authors also reported funding from Merck, Incyte, Gilead Sciences, and others.
SOURCE: Janjigian YY et al. J Clin Oncol. 2018 Aug 15. doi: 10.1200/JCO.2017.76.6212.
Nivolumab alone or in combination with ipilimumab met multiple endpoints against metastatic or locally advanced chemotherapy-refractory esophagogastric cancer in the recent phase 1/2 CheckMate-032 trial, thereby opening doors to a future phase 3 trial.
The agents demonstrated “clinically meaningful antitumor activity,” reported Yelena Y. Janjigian, MD, of Memorial Sloan Kettering Cancer Center, New York, and her coauthors.
After the 2017 ATTRACTION-2 trial demonstrated improved survival rates, “nivolumab was approved in Japan for the treatment of patients with chemotherapy-refractory gastric and gastroesophageal junction [GEJ] cancers regardless of programmed death-ligand 1 [PD-L1] status,” the authors wrote in the Journal of Clinical Oncology.
Nivolumab is a checkpoint inhibitor, like pembrolizumab, which “was approved for the treatment of patients with chemotherapy-refractory PD-L1–positive gastric/GEJ cancer on the basis of the promising clinical activity observed in the KEYNOTE-059 trial,” the authors noted. Testing nivolumab in a Western population would therefore build on these previous trials. Combining nivolumab, a PD-l inhibitor, with ipilimumab, a monoclonal antibody targeting cytotoxic T-lymphocyte antigen 4, was based on “synergistic activity” reported in preclinical models, the authors wrote.
Results from the ongoing CheckMate-032 trial included 160 patients with metastatic or locally advanced chemotherapy-refractory esophageal, gastric, or gastroesophageal junction cancer treated at centers in Europe and the United States. Just under 80% of patients had received two or more prior therapies.
In the present trial, patients were given one of three treatment regimens: nivolumab 3 mg/kg every 2 weeks, nivolumab 1 mg/kg plus ipilimumab 3 mg/kg every 3 weeks for four cycles (NIVO1 + IPI3), or nivolumab 3 mg/kg plus ipilimumab 1 mg/kg every 3 weeks for four cycles (NIVO3 + IPI1). The primary endpoint was objective response rate (ORR). Secondary endpoints included 12-month progression-free survival and 12-month overall survival (OS).
Patients in the NIVO1 + IPI3 group achieved the best ORR (24%) and 12-month progression-free survival (17%) and also showed a promising 12-month OS (35%), second only to nivolumab monotherapy (39%). PD-L1 status was not predictive of treatment response.
Although NIVO1 + IPI3 was the most clinically effective, almost half (47%) of these patients also had grade 3 or higher adverse events, compared with more favorable rates of 17% and 27% for nivolumab monotherapy and NIVO3 + IPI1, respectively.
Still, the authors concluded, “on the basis of the numerically higher overall response and landmark OS rates in the NIVO1 + IPI3 arm, this combination was considered more likely to offer clinical benefit relative to currently available treatment regimens for first-line metastatic esophagogastric cancer and was selected for further evaluation in the phase 3 CheckMate-649 study (NCT02872116).” This trial, along with another to investigate nivolumab in the adjuvant setting (NCT02743494), are ongoing.
CheckMate-032 was supported by Bristol-Myers Squibb. The authors also reported funding from Merck, Incyte, Gilead Sciences, and others.
SOURCE: Janjigian YY et al. J Clin Oncol. 2018 Aug 15. doi: 10.1200/JCO.2017.76.6212.
Nivolumab alone or in combination with ipilimumab met multiple endpoints against metastatic or locally advanced chemotherapy-refractory esophagogastric cancer in the recent phase 1/2 CheckMate-032 trial, thereby opening doors to a future phase 3 trial.
The agents demonstrated “clinically meaningful antitumor activity,” reported Yelena Y. Janjigian, MD, of Memorial Sloan Kettering Cancer Center, New York, and her coauthors.
After the 2017 ATTRACTION-2 trial demonstrated improved survival rates, “nivolumab was approved in Japan for the treatment of patients with chemotherapy-refractory gastric and gastroesophageal junction [GEJ] cancers regardless of programmed death-ligand 1 [PD-L1] status,” the authors wrote in the Journal of Clinical Oncology.
Nivolumab is a checkpoint inhibitor, like pembrolizumab, which “was approved for the treatment of patients with chemotherapy-refractory PD-L1–positive gastric/GEJ cancer on the basis of the promising clinical activity observed in the KEYNOTE-059 trial,” the authors noted. Testing nivolumab in a Western population would therefore build on these previous trials. Combining nivolumab, a PD-l inhibitor, with ipilimumab, a monoclonal antibody targeting cytotoxic T-lymphocyte antigen 4, was based on “synergistic activity” reported in preclinical models, the authors wrote.
Results from the ongoing CheckMate-032 trial included 160 patients with metastatic or locally advanced chemotherapy-refractory esophageal, gastric, or gastroesophageal junction cancer treated at centers in Europe and the United States. Just under 80% of patients had received two or more prior therapies.
In the present trial, patients were given one of three treatment regimens: nivolumab 3 mg/kg every 2 weeks, nivolumab 1 mg/kg plus ipilimumab 3 mg/kg every 3 weeks for four cycles (NIVO1 + IPI3), or nivolumab 3 mg/kg plus ipilimumab 1 mg/kg every 3 weeks for four cycles (NIVO3 + IPI1). The primary endpoint was objective response rate (ORR). Secondary endpoints included 12-month progression-free survival and 12-month overall survival (OS).
Patients in the NIVO1 + IPI3 group achieved the best ORR (24%) and 12-month progression-free survival (17%) and also showed a promising 12-month OS (35%), second only to nivolumab monotherapy (39%). PD-L1 status was not predictive of treatment response.
Although NIVO1 + IPI3 was the most clinically effective, almost half (47%) of these patients also had grade 3 or higher adverse events, compared with more favorable rates of 17% and 27% for nivolumab monotherapy and NIVO3 + IPI1, respectively.
Still, the authors concluded, “on the basis of the numerically higher overall response and landmark OS rates in the NIVO1 + IPI3 arm, this combination was considered more likely to offer clinical benefit relative to currently available treatment regimens for first-line metastatic esophagogastric cancer and was selected for further evaluation in the phase 3 CheckMate-649 study (NCT02872116).” This trial, along with another to investigate nivolumab in the adjuvant setting (NCT02743494), are ongoing.
CheckMate-032 was supported by Bristol-Myers Squibb. The authors also reported funding from Merck, Incyte, Gilead Sciences, and others.
SOURCE: Janjigian YY et al. J Clin Oncol. 2018 Aug 15. doi: 10.1200/JCO.2017.76.6212.
FROM THE JOURNAL OF CLINICAL ONCOLOGY
Key clinical point: Both nivolumab and nivolumab plus ipilimumab were effective in patients with chemotherapy-refractory esophagogastric cancer.
Major finding: Treatment with nivolumab plus ipilimumab was associated with an objective response rate of 24%.
Study details: CheckMate-032 is an ongoing phase 1/2 trial involving 160 patients with metastatic or locally advanced chemotherapy-refractory esophageal, gastric, or gastroesophageal junction cancer from centers in Europe and the United States.
Disclosures: The study was supported by Bristol-Myers Squibb. The authors also reported funding from Merck, Incyte, Gilead Sciences, and others.
Source: Janjigian YY et al. J Clin Oncol. 2018 Aug 15. doi: 10.1200/JCO.2017.76.6212.
Signal strength may limit potency of CAR T-cell therapy
Contrary to what might be expected, chimeric antigen receptor (CAR) T cells with stronger signaling capabilities were less effective against lymphoma cells in a mouse model, investigators reported.
Intracellular signaling strength was a key determinant of T cell fate in the study, which was published in the journal Science Signaling.
By contrast, CAR signaling pathways could not be predicted solely by the costimulatory domains used to construct the receptor, investigators said.
Based on those findings, tailoring CAR design based on signal strength might improve the efficacy and reduce the toxicity of CAR T-cell therapy, according to Alexander Salter, an MD/PhD student at Fred Hutchinson Cancer Research Center, Seattle, Wash.
In a press conference, Mr. Salter described results of the study, which used mass spectrometry to evaluate CARs encoding CD28 or 4-1BB costimulatory domains in primary human T cells.
While CARs with CD28 domains elicited more robust intracellular signaling than those with 4-1BB domains, there was considerable overlap in activation of T cell signaling pathways, Mr. Salter said.
That overlap was somewhat surprising, according to Mr. Salter, since researchers have generally assumed that CARs with CD28 and 4-1BB costimulatory domains will primarily signal through those respective pathways.
“No matter what costimulatory domain was encoded by the receptor, both CARs… activated both CD28 and 41BB signaling pathways,” Mr. Salter said.
The major determinant of efficacy in the study turned out to be not the domain used to construct the receptor, but the speed and strength of signaling, he added. In particular, the CARs that evoked stronger signals also had increased T cell dysfunction, decreasing their potency in the mouse lymphoma model.
The T cells with a CD28 CAR had very strong initial antitumor function that quickly waned in the mouse model of lymphoma; by contrast, the “slower burning” 4-1BB CAR signal led to T cells that better retained their function in vivo and were associated with longer median survival in the model, he said.
Those findings suggest tailoring CAR design based on signal strength may improve clinical efficacy and reduce toxicity.
As part of the study, Mr. Salter and his co-investigators were able to modify the CAR CD28 domain to make the signaling of the CD28 CARs less intense. “This is a modification that we think should be considered in future CAR design,” Mr. Salter said.
While the alterations in the CD28 signaling domain were able to reduce levels of cytokines produced by T cells, the study was primarily designed to look at the efficacy, noted Stanley Riddell, MD, scientific director of the Immunotherapy Integrated Research Center at Fred Hutchinson Cancer Research Center.
“Our models were not set up to address the question of toxicity, so we can’t directly say this would translate to what we would see in patients,” Dr. Riddell said during the press conference. “But I think we gleaned a lot of insights as to why cytokines are produced at greater or lesser levels with various CAR designs, and insights as to how to redesign these receptors to lower the levels of cytokines they make without compromising their ability to kill.”
Dr. Riddell is a founder, shareholder, and scientific advisor of Juno Therapeutics, and together with Mr. Salter, he has filed a patent application on the use of mutant CD28 CARs for cellular therapy. Co-author Raphael Gottardo, PhD, also with Fred Hutchinson Cancer Research Center, is a consultant for Juno Therapeutics. No other competing interests were reported.
SOURCE: Salter AI et al., Sci Signal. 2018 Aug 21;11. pii:eaat6753.
Contrary to what might be expected, chimeric antigen receptor (CAR) T cells with stronger signaling capabilities were less effective against lymphoma cells in a mouse model, investigators reported.
Intracellular signaling strength was a key determinant of T cell fate in the study, which was published in the journal Science Signaling.
By contrast, CAR signaling pathways could not be predicted solely by the costimulatory domains used to construct the receptor, investigators said.
Based on those findings, tailoring CAR design based on signal strength might improve the efficacy and reduce the toxicity of CAR T-cell therapy, according to Alexander Salter, an MD/PhD student at Fred Hutchinson Cancer Research Center, Seattle, Wash.
In a press conference, Mr. Salter described results of the study, which used mass spectrometry to evaluate CARs encoding CD28 or 4-1BB costimulatory domains in primary human T cells.
While CARs with CD28 domains elicited more robust intracellular signaling than those with 4-1BB domains, there was considerable overlap in activation of T cell signaling pathways, Mr. Salter said.
That overlap was somewhat surprising, according to Mr. Salter, since researchers have generally assumed that CARs with CD28 and 4-1BB costimulatory domains will primarily signal through those respective pathways.
“No matter what costimulatory domain was encoded by the receptor, both CARs… activated both CD28 and 41BB signaling pathways,” Mr. Salter said.
The major determinant of efficacy in the study turned out to be not the domain used to construct the receptor, but the speed and strength of signaling, he added. In particular, the CARs that evoked stronger signals also had increased T cell dysfunction, decreasing their potency in the mouse lymphoma model.
The T cells with a CD28 CAR had very strong initial antitumor function that quickly waned in the mouse model of lymphoma; by contrast, the “slower burning” 4-1BB CAR signal led to T cells that better retained their function in vivo and were associated with longer median survival in the model, he said.
Those findings suggest tailoring CAR design based on signal strength may improve clinical efficacy and reduce toxicity.
As part of the study, Mr. Salter and his co-investigators were able to modify the CAR CD28 domain to make the signaling of the CD28 CARs less intense. “This is a modification that we think should be considered in future CAR design,” Mr. Salter said.
While the alterations in the CD28 signaling domain were able to reduce levels of cytokines produced by T cells, the study was primarily designed to look at the efficacy, noted Stanley Riddell, MD, scientific director of the Immunotherapy Integrated Research Center at Fred Hutchinson Cancer Research Center.
“Our models were not set up to address the question of toxicity, so we can’t directly say this would translate to what we would see in patients,” Dr. Riddell said during the press conference. “But I think we gleaned a lot of insights as to why cytokines are produced at greater or lesser levels with various CAR designs, and insights as to how to redesign these receptors to lower the levels of cytokines they make without compromising their ability to kill.”
Dr. Riddell is a founder, shareholder, and scientific advisor of Juno Therapeutics, and together with Mr. Salter, he has filed a patent application on the use of mutant CD28 CARs for cellular therapy. Co-author Raphael Gottardo, PhD, also with Fred Hutchinson Cancer Research Center, is a consultant for Juno Therapeutics. No other competing interests were reported.
SOURCE: Salter AI et al., Sci Signal. 2018 Aug 21;11. pii:eaat6753.
Contrary to what might be expected, chimeric antigen receptor (CAR) T cells with stronger signaling capabilities were less effective against lymphoma cells in a mouse model, investigators reported.
Intracellular signaling strength was a key determinant of T cell fate in the study, which was published in the journal Science Signaling.
By contrast, CAR signaling pathways could not be predicted solely by the costimulatory domains used to construct the receptor, investigators said.
Based on those findings, tailoring CAR design based on signal strength might improve the efficacy and reduce the toxicity of CAR T-cell therapy, according to Alexander Salter, an MD/PhD student at Fred Hutchinson Cancer Research Center, Seattle, Wash.
In a press conference, Mr. Salter described results of the study, which used mass spectrometry to evaluate CARs encoding CD28 or 4-1BB costimulatory domains in primary human T cells.
While CARs with CD28 domains elicited more robust intracellular signaling than those with 4-1BB domains, there was considerable overlap in activation of T cell signaling pathways, Mr. Salter said.
That overlap was somewhat surprising, according to Mr. Salter, since researchers have generally assumed that CARs with CD28 and 4-1BB costimulatory domains will primarily signal through those respective pathways.
“No matter what costimulatory domain was encoded by the receptor, both CARs… activated both CD28 and 41BB signaling pathways,” Mr. Salter said.
The major determinant of efficacy in the study turned out to be not the domain used to construct the receptor, but the speed and strength of signaling, he added. In particular, the CARs that evoked stronger signals also had increased T cell dysfunction, decreasing their potency in the mouse lymphoma model.
The T cells with a CD28 CAR had very strong initial antitumor function that quickly waned in the mouse model of lymphoma; by contrast, the “slower burning” 4-1BB CAR signal led to T cells that better retained their function in vivo and were associated with longer median survival in the model, he said.
Those findings suggest tailoring CAR design based on signal strength may improve clinical efficacy and reduce toxicity.
As part of the study, Mr. Salter and his co-investigators were able to modify the CAR CD28 domain to make the signaling of the CD28 CARs less intense. “This is a modification that we think should be considered in future CAR design,” Mr. Salter said.
While the alterations in the CD28 signaling domain were able to reduce levels of cytokines produced by T cells, the study was primarily designed to look at the efficacy, noted Stanley Riddell, MD, scientific director of the Immunotherapy Integrated Research Center at Fred Hutchinson Cancer Research Center.
“Our models were not set up to address the question of toxicity, so we can’t directly say this would translate to what we would see in patients,” Dr. Riddell said during the press conference. “But I think we gleaned a lot of insights as to why cytokines are produced at greater or lesser levels with various CAR designs, and insights as to how to redesign these receptors to lower the levels of cytokines they make without compromising their ability to kill.”
Dr. Riddell is a founder, shareholder, and scientific advisor of Juno Therapeutics, and together with Mr. Salter, he has filed a patent application on the use of mutant CD28 CARs for cellular therapy. Co-author Raphael Gottardo, PhD, also with Fred Hutchinson Cancer Research Center, is a consultant for Juno Therapeutics. No other competing interests were reported.
SOURCE: Salter AI et al., Sci Signal. 2018 Aug 21;11. pii:eaat6753.
FROM SCIENCE SIGNALING
Key clinical point:
Major finding: T cells with a CD28 CAR had very strong initial antitumor function that quickly waned in a mouse model of lymphoma, while the 4-1BB CAR signal led to T cells that better retained their function in vivo and had a longer median survival in the model.
Study details: Analysis of CARs encoding CD28 or 4-1BB costimulatory domains in primary human T cells using mass spectrometry, plus analysis of efficacy in a mouse model of lymphoma.
Disclosures: Study authors reported disclosures related to Juno therapeutics and a patent application related to use of mutant CD28 CARs for cellular therapy.
Source: Salter AI et al., Sci Signal. 2018 Aug 21;11. pii:eaat6753.
CMS finalizes CAR T-cell therapy inpatient payments
Medical associations are expressing disappointment at the new payment scheme put forward by the Centers for Medicare & Medicaid Services for inpatient administration of two chimeric antigen receptor (CAR) T-cell therapies, calling the reimbursement insufficient for use of the expensive medications.
Under its Aug. 17 final rule, CMS will now categorize CAR T-cell therapies under the umbrella of the renamed Medicare Severity–Diagnosis Related Groups (MS-DRG) 016 – Autologous Bone Marrow Transplant with CC/MCC or T-cell Immunotherapy – and assign ICD-10-PCS procedure codes XW033C3 and XW043C3 to the use of axicabtagene ciloleucel (Yescarta) and tisagenlecleucel (Kymriah) in the inpatient setting for fiscal year 2019, which begins in October 2018.
CMS also approved a temporary New Technology Add-On Payment (NTAP) for use of the therapies with a maximum threshold of $186,500, according to the rule.
According to the American Society of Hematology (ASH), this payment structure is an improvement, but it hardly covers the cost of the products, nor does it account for full hospitalization costs. ASH noted that the revised MS-DRG 016 has a base payment rate of $36,000 and that the maximum NTAP payment ($186,500) is only about half of the cost for a CAR T-cell product.
“ASH is concerned that this final policy may impede access to care to this cutting-edge therapy because hospitals and academic medical centers that provide this personalized treatment will simply not be able to withstand the negative financial impact,” the society said in a statement. “While this final policy represents an improvement over current CAR T therapy reimbursement rates, ASH believes patient access to care will be jeopardized as providers and hospitals will not be able to afford to deliver the therapy at this reimbursement rate, particularly as other CAR T products receive FDA [Food and Drug Administration] approval.”
ASH and the American Society for Blood and Marrow Transplantation (ASBMT) had strongly urged CMS to develop a site-neutral, equitable payment structure that would have allowed providers to recover more product acquisition costs from CAR T-cell therapies. In its final rule, CMS stated that it was too early to develop a novel payment structure for CAR T-cell treatments and that more research is needed before such changes are made. The agency noted that in May CMS opened a national coverage determination analysis on CAR T-cell therapy for Medicare patients with advanced cancer, which is expected to be completed by May 2019.
“[CMS] is soliciting public comment … on key design considerations for developing a potential model that would test private market strategies and introduce competition to improve quality of care for beneficiaries,” the agency said in the rule. “Given the relative newness of CAR T-cell therapy, the potential model, and our request for feedback on this model approach, we believe it would be premature to adopt changes to our existing payment mechanisms.”
The payment outline by CMS is essentially the bare minimum it could have extended to CAR T-cell therapies for 2019, said Stephanie Farnia, director of health policy and strategic relations for the ASBMT.
“[ASBMT] and a number of stakeholders have been very clear in our comment letters that that would not be enough and the reasons why,” Ms. Farnia said in an interview. “It’s not going to be sufficient to cover the cost of care or the product.”
The rule also fails to address the cancer centers that are exempt from the DRG payment system, Ms. Farnia said. Eleven centers are excluded from the payment system because of past legislation that excludes exclusive cancer hospitals that do not provide noncancer services. The exempt cancer centers cannot receive additional money for new or expensive drugs and therefore will not gain any financial relief from the CAR T-cell therapy payment changes in the CMS final rule.
ASH officials plan to follow up with congressional leaders to identify ways to improve future CAR T-cell therapy payments, including a potential legislative solution. An ASH spokesperson declined to elaborate on its ideal legislative remedy.
Hospital administrators and physicians will need to have difficult conversations in the upcoming year about whether treating patients with CAR T-cell therapies is worth the cost deficits, Ms. Farnia said.
“Everyone was really counting on it being a different reimbursement scenario for the upcoming fiscal year, and it is, but again, it’s that bare minimum difference,” Ms. Farnia said. “I think a number of programs are going to be taking a look at their financial experience thus far and comparing that to the reimbursement and deciding on if they [should] continue to offer it and how to do that.”
In April 2018, CMS announced payment rates for outpatient administration of the two drugs, settling on $395,380 for axicabtagene ciloleucel and $500,839 for tisagenlecleucel. The two medications have list prices of $373,000 and $475,000, respectively.
However, physicians have raised concerns that even if the drugs are first administered in the outpatient setting, inpatient care is likely to occur with CAR T-cell therapies because some patients will need to be admitted in order to be monitored for serious side effects. In such cases, all payments will become part of the inpatient stay under CMS’s 3-day payment window rule.
Medical associations are expressing disappointment at the new payment scheme put forward by the Centers for Medicare & Medicaid Services for inpatient administration of two chimeric antigen receptor (CAR) T-cell therapies, calling the reimbursement insufficient for use of the expensive medications.
Under its Aug. 17 final rule, CMS will now categorize CAR T-cell therapies under the umbrella of the renamed Medicare Severity–Diagnosis Related Groups (MS-DRG) 016 – Autologous Bone Marrow Transplant with CC/MCC or T-cell Immunotherapy – and assign ICD-10-PCS procedure codes XW033C3 and XW043C3 to the use of axicabtagene ciloleucel (Yescarta) and tisagenlecleucel (Kymriah) in the inpatient setting for fiscal year 2019, which begins in October 2018.
CMS also approved a temporary New Technology Add-On Payment (NTAP) for use of the therapies with a maximum threshold of $186,500, according to the rule.
According to the American Society of Hematology (ASH), this payment structure is an improvement, but it hardly covers the cost of the products, nor does it account for full hospitalization costs. ASH noted that the revised MS-DRG 016 has a base payment rate of $36,000 and that the maximum NTAP payment ($186,500) is only about half of the cost for a CAR T-cell product.
“ASH is concerned that this final policy may impede access to care to this cutting-edge therapy because hospitals and academic medical centers that provide this personalized treatment will simply not be able to withstand the negative financial impact,” the society said in a statement. “While this final policy represents an improvement over current CAR T therapy reimbursement rates, ASH believes patient access to care will be jeopardized as providers and hospitals will not be able to afford to deliver the therapy at this reimbursement rate, particularly as other CAR T products receive FDA [Food and Drug Administration] approval.”
ASH and the American Society for Blood and Marrow Transplantation (ASBMT) had strongly urged CMS to develop a site-neutral, equitable payment structure that would have allowed providers to recover more product acquisition costs from CAR T-cell therapies. In its final rule, CMS stated that it was too early to develop a novel payment structure for CAR T-cell treatments and that more research is needed before such changes are made. The agency noted that in May CMS opened a national coverage determination analysis on CAR T-cell therapy for Medicare patients with advanced cancer, which is expected to be completed by May 2019.
“[CMS] is soliciting public comment … on key design considerations for developing a potential model that would test private market strategies and introduce competition to improve quality of care for beneficiaries,” the agency said in the rule. “Given the relative newness of CAR T-cell therapy, the potential model, and our request for feedback on this model approach, we believe it would be premature to adopt changes to our existing payment mechanisms.”
The payment outline by CMS is essentially the bare minimum it could have extended to CAR T-cell therapies for 2019, said Stephanie Farnia, director of health policy and strategic relations for the ASBMT.
“[ASBMT] and a number of stakeholders have been very clear in our comment letters that that would not be enough and the reasons why,” Ms. Farnia said in an interview. “It’s not going to be sufficient to cover the cost of care or the product.”
The rule also fails to address the cancer centers that are exempt from the DRG payment system, Ms. Farnia said. Eleven centers are excluded from the payment system because of past legislation that excludes exclusive cancer hospitals that do not provide noncancer services. The exempt cancer centers cannot receive additional money for new or expensive drugs and therefore will not gain any financial relief from the CAR T-cell therapy payment changes in the CMS final rule.
ASH officials plan to follow up with congressional leaders to identify ways to improve future CAR T-cell therapy payments, including a potential legislative solution. An ASH spokesperson declined to elaborate on its ideal legislative remedy.
Hospital administrators and physicians will need to have difficult conversations in the upcoming year about whether treating patients with CAR T-cell therapies is worth the cost deficits, Ms. Farnia said.
“Everyone was really counting on it being a different reimbursement scenario for the upcoming fiscal year, and it is, but again, it’s that bare minimum difference,” Ms. Farnia said. “I think a number of programs are going to be taking a look at their financial experience thus far and comparing that to the reimbursement and deciding on if they [should] continue to offer it and how to do that.”
In April 2018, CMS announced payment rates for outpatient administration of the two drugs, settling on $395,380 for axicabtagene ciloleucel and $500,839 for tisagenlecleucel. The two medications have list prices of $373,000 and $475,000, respectively.
However, physicians have raised concerns that even if the drugs are first administered in the outpatient setting, inpatient care is likely to occur with CAR T-cell therapies because some patients will need to be admitted in order to be monitored for serious side effects. In such cases, all payments will become part of the inpatient stay under CMS’s 3-day payment window rule.
Medical associations are expressing disappointment at the new payment scheme put forward by the Centers for Medicare & Medicaid Services for inpatient administration of two chimeric antigen receptor (CAR) T-cell therapies, calling the reimbursement insufficient for use of the expensive medications.
Under its Aug. 17 final rule, CMS will now categorize CAR T-cell therapies under the umbrella of the renamed Medicare Severity–Diagnosis Related Groups (MS-DRG) 016 – Autologous Bone Marrow Transplant with CC/MCC or T-cell Immunotherapy – and assign ICD-10-PCS procedure codes XW033C3 and XW043C3 to the use of axicabtagene ciloleucel (Yescarta) and tisagenlecleucel (Kymriah) in the inpatient setting for fiscal year 2019, which begins in October 2018.
CMS also approved a temporary New Technology Add-On Payment (NTAP) for use of the therapies with a maximum threshold of $186,500, according to the rule.
According to the American Society of Hematology (ASH), this payment structure is an improvement, but it hardly covers the cost of the products, nor does it account for full hospitalization costs. ASH noted that the revised MS-DRG 016 has a base payment rate of $36,000 and that the maximum NTAP payment ($186,500) is only about half of the cost for a CAR T-cell product.
“ASH is concerned that this final policy may impede access to care to this cutting-edge therapy because hospitals and academic medical centers that provide this personalized treatment will simply not be able to withstand the negative financial impact,” the society said in a statement. “While this final policy represents an improvement over current CAR T therapy reimbursement rates, ASH believes patient access to care will be jeopardized as providers and hospitals will not be able to afford to deliver the therapy at this reimbursement rate, particularly as other CAR T products receive FDA [Food and Drug Administration] approval.”
ASH and the American Society for Blood and Marrow Transplantation (ASBMT) had strongly urged CMS to develop a site-neutral, equitable payment structure that would have allowed providers to recover more product acquisition costs from CAR T-cell therapies. In its final rule, CMS stated that it was too early to develop a novel payment structure for CAR T-cell treatments and that more research is needed before such changes are made. The agency noted that in May CMS opened a national coverage determination analysis on CAR T-cell therapy for Medicare patients with advanced cancer, which is expected to be completed by May 2019.
“[CMS] is soliciting public comment … on key design considerations for developing a potential model that would test private market strategies and introduce competition to improve quality of care for beneficiaries,” the agency said in the rule. “Given the relative newness of CAR T-cell therapy, the potential model, and our request for feedback on this model approach, we believe it would be premature to adopt changes to our existing payment mechanisms.”
The payment outline by CMS is essentially the bare minimum it could have extended to CAR T-cell therapies for 2019, said Stephanie Farnia, director of health policy and strategic relations for the ASBMT.
“[ASBMT] and a number of stakeholders have been very clear in our comment letters that that would not be enough and the reasons why,” Ms. Farnia said in an interview. “It’s not going to be sufficient to cover the cost of care or the product.”
The rule also fails to address the cancer centers that are exempt from the DRG payment system, Ms. Farnia said. Eleven centers are excluded from the payment system because of past legislation that excludes exclusive cancer hospitals that do not provide noncancer services. The exempt cancer centers cannot receive additional money for new or expensive drugs and therefore will not gain any financial relief from the CAR T-cell therapy payment changes in the CMS final rule.
ASH officials plan to follow up with congressional leaders to identify ways to improve future CAR T-cell therapy payments, including a potential legislative solution. An ASH spokesperson declined to elaborate on its ideal legislative remedy.
Hospital administrators and physicians will need to have difficult conversations in the upcoming year about whether treating patients with CAR T-cell therapies is worth the cost deficits, Ms. Farnia said.
“Everyone was really counting on it being a different reimbursement scenario for the upcoming fiscal year, and it is, but again, it’s that bare minimum difference,” Ms. Farnia said. “I think a number of programs are going to be taking a look at their financial experience thus far and comparing that to the reimbursement and deciding on if they [should] continue to offer it and how to do that.”
In April 2018, CMS announced payment rates for outpatient administration of the two drugs, settling on $395,380 for axicabtagene ciloleucel and $500,839 for tisagenlecleucel. The two medications have list prices of $373,000 and $475,000, respectively.
However, physicians have raised concerns that even if the drugs are first administered in the outpatient setting, inpatient care is likely to occur with CAR T-cell therapies because some patients will need to be admitted in order to be monitored for serious side effects. In such cases, all payments will become part of the inpatient stay under CMS’s 3-day payment window rule.
Feds aim to streamline gene therapy oversight
“As the NIH, the FDA, and research entities have moved to strengthen their individual oversight efforts, some overlaps have occurred,” Dr. Collins and Dr. Gottlieb wrote Aug. 15 in the New England Journal of Medicine. “Substantial duplication has arisen in the submission of initial protocols, annual reports, amendments, and reports of serious adverse events. Originally, these overlaps – which affect no other field in biomedical research – were viewed as harmonized reporting that enabled FDA to conduct regulatory oversight while maintaining confidentiality with sponsors and allowed NIH to provide transparency with regard to the research.”
With three approved treatments – two CAR T therapies and a treatment for a genetic retinal dystrophy – and more than 700 active investigational new drug applications in front of the FDA, “it seems reasonable to envision a day when gene therapy will be a mainstay of treatment for many diseases,” according to the editorial.
To remedy the situation, NIH officials Aug. 16 posted a proposed update to NIH guidance that seeks “to reduce the duplicative oversight burden by further limiting the role of NIH and RAC [Recombinant DNA Advisory Committee] in assessing gene therapy protocols and reviewing their safety information,” Dr. Collins and Dr. Gottlieb wrote. “Specifically, these proposals will eliminate RAC review and reporting requirements to the NIH for human gene therapy protocols. They will also revise the responsibility of institutional Biosafety Committees, which have local oversight for this research, making their review of human gene therapy protocols consistent with review of other research subject to the NIH Guidelines. Such streamlining will also appropriately place the focus of the NIH Guidelines squarely back on laboratory biosafety.”
RAC was originally established in 1974 to advise the NIH director on research involving manipulation of nucleic acids, but was later expanded to review and discuss protocols for gene therapy in humans.
The proposed structure provides an opportunity “to return the RAC to the spirit in which it was founded. ... The NIH envisions using the RAC as an advisory board on today’s emerging technologies, such as gene editing, synthetic biology, and neurotechnology, while harnessing the attributes that have long ensured its transparency,” they wrote.
This latest proposal follows a suite of FDA draft guidances on gene therapies issued in July that proposed new guidance on manufacturing issues, long-term follow-up, and pathways for clinical development in certain areas.
The guidance is scheduled for publication in the Federal Register on Aug. 17.
SOURCE: Collins FS and Gottlieb S. N Engl J Med. doi: 10.1056/NEJMp1810628.
“As the NIH, the FDA, and research entities have moved to strengthen their individual oversight efforts, some overlaps have occurred,” Dr. Collins and Dr. Gottlieb wrote Aug. 15 in the New England Journal of Medicine. “Substantial duplication has arisen in the submission of initial protocols, annual reports, amendments, and reports of serious adverse events. Originally, these overlaps – which affect no other field in biomedical research – were viewed as harmonized reporting that enabled FDA to conduct regulatory oversight while maintaining confidentiality with sponsors and allowed NIH to provide transparency with regard to the research.”
With three approved treatments – two CAR T therapies and a treatment for a genetic retinal dystrophy – and more than 700 active investigational new drug applications in front of the FDA, “it seems reasonable to envision a day when gene therapy will be a mainstay of treatment for many diseases,” according to the editorial.
To remedy the situation, NIH officials Aug. 16 posted a proposed update to NIH guidance that seeks “to reduce the duplicative oversight burden by further limiting the role of NIH and RAC [Recombinant DNA Advisory Committee] in assessing gene therapy protocols and reviewing their safety information,” Dr. Collins and Dr. Gottlieb wrote. “Specifically, these proposals will eliminate RAC review and reporting requirements to the NIH for human gene therapy protocols. They will also revise the responsibility of institutional Biosafety Committees, which have local oversight for this research, making their review of human gene therapy protocols consistent with review of other research subject to the NIH Guidelines. Such streamlining will also appropriately place the focus of the NIH Guidelines squarely back on laboratory biosafety.”
RAC was originally established in 1974 to advise the NIH director on research involving manipulation of nucleic acids, but was later expanded to review and discuss protocols for gene therapy in humans.
The proposed structure provides an opportunity “to return the RAC to the spirit in which it was founded. ... The NIH envisions using the RAC as an advisory board on today’s emerging technologies, such as gene editing, synthetic biology, and neurotechnology, while harnessing the attributes that have long ensured its transparency,” they wrote.
This latest proposal follows a suite of FDA draft guidances on gene therapies issued in July that proposed new guidance on manufacturing issues, long-term follow-up, and pathways for clinical development in certain areas.
The guidance is scheduled for publication in the Federal Register on Aug. 17.
SOURCE: Collins FS and Gottlieb S. N Engl J Med. doi: 10.1056/NEJMp1810628.
“As the NIH, the FDA, and research entities have moved to strengthen their individual oversight efforts, some overlaps have occurred,” Dr. Collins and Dr. Gottlieb wrote Aug. 15 in the New England Journal of Medicine. “Substantial duplication has arisen in the submission of initial protocols, annual reports, amendments, and reports of serious adverse events. Originally, these overlaps – which affect no other field in biomedical research – were viewed as harmonized reporting that enabled FDA to conduct regulatory oversight while maintaining confidentiality with sponsors and allowed NIH to provide transparency with regard to the research.”
With three approved treatments – two CAR T therapies and a treatment for a genetic retinal dystrophy – and more than 700 active investigational new drug applications in front of the FDA, “it seems reasonable to envision a day when gene therapy will be a mainstay of treatment for many diseases,” according to the editorial.
To remedy the situation, NIH officials Aug. 16 posted a proposed update to NIH guidance that seeks “to reduce the duplicative oversight burden by further limiting the role of NIH and RAC [Recombinant DNA Advisory Committee] in assessing gene therapy protocols and reviewing their safety information,” Dr. Collins and Dr. Gottlieb wrote. “Specifically, these proposals will eliminate RAC review and reporting requirements to the NIH for human gene therapy protocols. They will also revise the responsibility of institutional Biosafety Committees, which have local oversight for this research, making their review of human gene therapy protocols consistent with review of other research subject to the NIH Guidelines. Such streamlining will also appropriately place the focus of the NIH Guidelines squarely back on laboratory biosafety.”
RAC was originally established in 1974 to advise the NIH director on research involving manipulation of nucleic acids, but was later expanded to review and discuss protocols for gene therapy in humans.
The proposed structure provides an opportunity “to return the RAC to the spirit in which it was founded. ... The NIH envisions using the RAC as an advisory board on today’s emerging technologies, such as gene editing, synthetic biology, and neurotechnology, while harnessing the attributes that have long ensured its transparency,” they wrote.
This latest proposal follows a suite of FDA draft guidances on gene therapies issued in July that proposed new guidance on manufacturing issues, long-term follow-up, and pathways for clinical development in certain areas.
The guidance is scheduled for publication in the Federal Register on Aug. 17.
SOURCE: Collins FS and Gottlieb S. N Engl J Med. doi: 10.1056/NEJMp1810628.
Key clinical point: NIH and FDA are looking to streamline gene therapy development oversight.
Major finding: The proposal would return the function of the Recombinant DNA Advisory Committee (RAC) to a function more in line with its original mission.
Study details: Full proposal details are to be published in the Federal Register on Aug. 17.
Disclosures: The agency leaders reported no relevant disclosures in the production of the proposal.
Source: Collins FS and Gottlieb S. N Engl J Med. doi: 10.1056/NEJMp1810628.
EGFR-mutant NSCLC may still respond to PD-1 blockade
PD-1 blockade should still be considered as a second-line approach in patients with EGFR-mutant non–small-cell lung cancer (NSCLC), as some may still respond to this therapy, investigators reported.
The researchers described a patient who was diagnosed with epidermal growth factor receptor (EGFR)-mutant NSCLC but who also showed high expression of PD-L1.
The 62-year-old woman first presented with a lung nodule and metastasis in the right hip, which were EGFR-mutant and had a PD-L1 tumor proportion score of 90%. She was treated with erlotinib – an EGFR tyrosine kinase inhibitor – and radiotherapy, according to the report, published in Annals of Oncology.
Two months later, she developed multiple new metastases in the chest wall. These also showed high PD-L1 expression, but no EGFR mutations, so she was treated with pembrolizumab as second-line therapy.
After the first course of treatment with pembrolizumab, the hip pain improved dramatically, and after three cycles the primary and metastatic lesions disappeared completely.
“Immunofluorescent analysis of both right ilium and chest wall lesions with an anti-EGFR antibody specific to Ex.19 del and an anti-PD-L1 antibody revealed the presence of distinct heterogeneity of EGFR-mutant clones and PD-L1 highly-expressing clones,” wrote Kei Kunimasa, MD, of Osaka International Cancer Institute, and coauthors.
Current clinical trials suggested that patients with EGFR mutations are likely to have a poor response to PD-1 blockade therapies for NSCLC, and the authors agreed that patients with EGFR mutations should be treated with EGFR tyrosine kinase inhibitors as first-line therapy.
“However, the present case suggested that shorter PFS in EGFR-TKI treatment for EGFR-mutant NSCLC patients with high PD-L1 TPS and without acquired T790M mutation could encourage us to try PD-1 blockade therapy as second line therapy,” they wrote.
The investigators suggested that certain clinical factors could identify patients with EGFR-mutant NSCLC who might respond well to PD-1 blockade, such as progression-free survival on EGFR tyrosine kinase inhibitor treatment, other targetable resistant mutations, tumor mutation burden, and PD-L1 expression.
Two authors reported grants and personal fees from pharmaceutical companies outside the submitted work. No other conflicts of interest were declared.
SOURCE: Kunimasa K et al. Ann Oncol, 2018 Aug 7. doi: 10.1093/annonc/mdy312.
PD-1 blockade should still be considered as a second-line approach in patients with EGFR-mutant non–small-cell lung cancer (NSCLC), as some may still respond to this therapy, investigators reported.
The researchers described a patient who was diagnosed with epidermal growth factor receptor (EGFR)-mutant NSCLC but who also showed high expression of PD-L1.
The 62-year-old woman first presented with a lung nodule and metastasis in the right hip, which were EGFR-mutant and had a PD-L1 tumor proportion score of 90%. She was treated with erlotinib – an EGFR tyrosine kinase inhibitor – and radiotherapy, according to the report, published in Annals of Oncology.
Two months later, she developed multiple new metastases in the chest wall. These also showed high PD-L1 expression, but no EGFR mutations, so she was treated with pembrolizumab as second-line therapy.
After the first course of treatment with pembrolizumab, the hip pain improved dramatically, and after three cycles the primary and metastatic lesions disappeared completely.
“Immunofluorescent analysis of both right ilium and chest wall lesions with an anti-EGFR antibody specific to Ex.19 del and an anti-PD-L1 antibody revealed the presence of distinct heterogeneity of EGFR-mutant clones and PD-L1 highly-expressing clones,” wrote Kei Kunimasa, MD, of Osaka International Cancer Institute, and coauthors.
Current clinical trials suggested that patients with EGFR mutations are likely to have a poor response to PD-1 blockade therapies for NSCLC, and the authors agreed that patients with EGFR mutations should be treated with EGFR tyrosine kinase inhibitors as first-line therapy.
“However, the present case suggested that shorter PFS in EGFR-TKI treatment for EGFR-mutant NSCLC patients with high PD-L1 TPS and without acquired T790M mutation could encourage us to try PD-1 blockade therapy as second line therapy,” they wrote.
The investigators suggested that certain clinical factors could identify patients with EGFR-mutant NSCLC who might respond well to PD-1 blockade, such as progression-free survival on EGFR tyrosine kinase inhibitor treatment, other targetable resistant mutations, tumor mutation burden, and PD-L1 expression.
Two authors reported grants and personal fees from pharmaceutical companies outside the submitted work. No other conflicts of interest were declared.
SOURCE: Kunimasa K et al. Ann Oncol, 2018 Aug 7. doi: 10.1093/annonc/mdy312.
PD-1 blockade should still be considered as a second-line approach in patients with EGFR-mutant non–small-cell lung cancer (NSCLC), as some may still respond to this therapy, investigators reported.
The researchers described a patient who was diagnosed with epidermal growth factor receptor (EGFR)-mutant NSCLC but who also showed high expression of PD-L1.
The 62-year-old woman first presented with a lung nodule and metastasis in the right hip, which were EGFR-mutant and had a PD-L1 tumor proportion score of 90%. She was treated with erlotinib – an EGFR tyrosine kinase inhibitor – and radiotherapy, according to the report, published in Annals of Oncology.
Two months later, she developed multiple new metastases in the chest wall. These also showed high PD-L1 expression, but no EGFR mutations, so she was treated with pembrolizumab as second-line therapy.
After the first course of treatment with pembrolizumab, the hip pain improved dramatically, and after three cycles the primary and metastatic lesions disappeared completely.
“Immunofluorescent analysis of both right ilium and chest wall lesions with an anti-EGFR antibody specific to Ex.19 del and an anti-PD-L1 antibody revealed the presence of distinct heterogeneity of EGFR-mutant clones and PD-L1 highly-expressing clones,” wrote Kei Kunimasa, MD, of Osaka International Cancer Institute, and coauthors.
Current clinical trials suggested that patients with EGFR mutations are likely to have a poor response to PD-1 blockade therapies for NSCLC, and the authors agreed that patients with EGFR mutations should be treated with EGFR tyrosine kinase inhibitors as first-line therapy.
“However, the present case suggested that shorter PFS in EGFR-TKI treatment for EGFR-mutant NSCLC patients with high PD-L1 TPS and without acquired T790M mutation could encourage us to try PD-1 blockade therapy as second line therapy,” they wrote.
The investigators suggested that certain clinical factors could identify patients with EGFR-mutant NSCLC who might respond well to PD-1 blockade, such as progression-free survival on EGFR tyrosine kinase inhibitor treatment, other targetable resistant mutations, tumor mutation burden, and PD-L1 expression.
Two authors reported grants and personal fees from pharmaceutical companies outside the submitted work. No other conflicts of interest were declared.
SOURCE: Kunimasa K et al. Ann Oncol, 2018 Aug 7. doi: 10.1093/annonc/mdy312.
FROM ANNALS OF ONCOLOGY
Key clinical point: Consider PD-1 blockade in EGFR-mutant non–small-cell lung cancer with high PD-L1 expression.Major finding: EGFR-mutant non–small-cell lung cancer may still respond to PD-1 blockade.
Study details: Case study.
Disclosures: Two authors reported grants and personal fees from pharmaceutical companies outside the submitted work. No other conflicts of interest were declared.
Source: Kunimasa K et al. Ann Oncol. 2018 Aug 7. doi: 10.1093/annonc/mdy312.
Phase 2 trial: Dendritic cell vaccine maintenance prolongs PFS in EOC
CHICAGO – who have undergone primary debulking surgery, according to interim findings from a randomized, phase 2, open-label trial.
Of 99 patients enrolled in the international, multicenter trial between November 2013 and March 2016, 92 received at least one dose of therapy and had a postbaseline endpoint assessment (the modified intention-to-treat population). Median progression-free survival was 18.3 months in the 31 patients who received autologous dendritic cell vaccine (DCVAC) given concomitantly with chemotherapy (group A), 24.3 months in the 30 patients who received DCVAC given sequentially as maintenance therapy after chemotherapy (group B), and 18.6 months in the 31 controls who received chemotherapy alone (group C), Lukas Rob, MD, reported at the annual meeting of the American Society of Clinical Oncology.
“When DCVAC was administered as maintenance [therapy] after the chemotherapy, the time to disease progression was prolonged by almost 6 months. Based on the hazard ratio, there was a 57% decrease in the hazard of progression in favor of arm B,” said Dr. Rob, of University Hospital Kralovske Vinohrady, Prague.
Compared with group C, progression-free survival hazard ratios were 0.64 and 0.43 in the modified intention-to-treat population in groups A and B, respectively, he said.
“These results are statistically significant and the clinical benefit is even more significant in patients who received more doses of DCVAC,” he added, explaining that the hazard ratios for patients in groups A and B who received at least eight doses of DCVAC and/or three cycles of chemotherapy (the per protocol population, including 87 patients) were 1.01 and 0.32, respectively.
Although the overall survival data in this study is immature, there is “a strong trend in favor of DCVAC in the maintenance treatment arm,” he said.
Study participants had FIGO stage III epithelial ovarian cancer (EOC) and a performance status score of 0-2. All had undergone primary debulking surgery and had less than 1 cm maximal residuum and no prior systemic therapy. Patients were randomized up to 6 weeks after surgery. Chemotherapy in all three groups included six cycles of carboplatin and paclitaxel, and DCVAC was given at doses of 1 x 107 dendritic cells per dose for a planned 10 doses.
Most patients with EOC – about 70% of those with stage III/IV disease – relapse after optimal debulking surgery and chemotherapy.
“There is no doubt that we need a new treatment modality,” he said. Because autologous DCVAC can present tumor antigens to elicit a durable immune response, he and his colleagues hypothesized that adding DCVAC to chemotherapy could improve outcomes – either when used concomitantly to target tumor-induced immune suppression and allowing for partial recovery of the immune system after each cycle, or when used sequentially as maintenance therapy, as minimal tumor burden after chemotherapy would provide optimal conditions for immune stimulation and the immune system would be fully recovered after completing cytotoxic therapy.
The treatments in this study were well tolerated; no grade 3 or greater adverse events were related solely to DCVAC, and the vaccine did not worsen the side effects of chemotherapy, Dr. Rob said.
The findings suggested that sequential DCVAC after chemotherapy in patients with EOC is a promising maintenance treatment option that can delay disease progression, he concluded, noting that “due to the excellent result in the maintenance arm we decided ... to enroll more patients in arms B and C to increase the power of this study.”
Additionally, a phase 3 trial is planned and enrollment should begin in early 2019, he said.
Dr. Rob reported having no disclosures.
SOURCE: Rob L et al. ASCO 2018, Abstract 5509.
CHICAGO – who have undergone primary debulking surgery, according to interim findings from a randomized, phase 2, open-label trial.
Of 99 patients enrolled in the international, multicenter trial between November 2013 and March 2016, 92 received at least one dose of therapy and had a postbaseline endpoint assessment (the modified intention-to-treat population). Median progression-free survival was 18.3 months in the 31 patients who received autologous dendritic cell vaccine (DCVAC) given concomitantly with chemotherapy (group A), 24.3 months in the 30 patients who received DCVAC given sequentially as maintenance therapy after chemotherapy (group B), and 18.6 months in the 31 controls who received chemotherapy alone (group C), Lukas Rob, MD, reported at the annual meeting of the American Society of Clinical Oncology.
“When DCVAC was administered as maintenance [therapy] after the chemotherapy, the time to disease progression was prolonged by almost 6 months. Based on the hazard ratio, there was a 57% decrease in the hazard of progression in favor of arm B,” said Dr. Rob, of University Hospital Kralovske Vinohrady, Prague.
Compared with group C, progression-free survival hazard ratios were 0.64 and 0.43 in the modified intention-to-treat population in groups A and B, respectively, he said.
“These results are statistically significant and the clinical benefit is even more significant in patients who received more doses of DCVAC,” he added, explaining that the hazard ratios for patients in groups A and B who received at least eight doses of DCVAC and/or three cycles of chemotherapy (the per protocol population, including 87 patients) were 1.01 and 0.32, respectively.
Although the overall survival data in this study is immature, there is “a strong trend in favor of DCVAC in the maintenance treatment arm,” he said.
Study participants had FIGO stage III epithelial ovarian cancer (EOC) and a performance status score of 0-2. All had undergone primary debulking surgery and had less than 1 cm maximal residuum and no prior systemic therapy. Patients were randomized up to 6 weeks after surgery. Chemotherapy in all three groups included six cycles of carboplatin and paclitaxel, and DCVAC was given at doses of 1 x 107 dendritic cells per dose for a planned 10 doses.
Most patients with EOC – about 70% of those with stage III/IV disease – relapse after optimal debulking surgery and chemotherapy.
“There is no doubt that we need a new treatment modality,” he said. Because autologous DCVAC can present tumor antigens to elicit a durable immune response, he and his colleagues hypothesized that adding DCVAC to chemotherapy could improve outcomes – either when used concomitantly to target tumor-induced immune suppression and allowing for partial recovery of the immune system after each cycle, or when used sequentially as maintenance therapy, as minimal tumor burden after chemotherapy would provide optimal conditions for immune stimulation and the immune system would be fully recovered after completing cytotoxic therapy.
The treatments in this study were well tolerated; no grade 3 or greater adverse events were related solely to DCVAC, and the vaccine did not worsen the side effects of chemotherapy, Dr. Rob said.
The findings suggested that sequential DCVAC after chemotherapy in patients with EOC is a promising maintenance treatment option that can delay disease progression, he concluded, noting that “due to the excellent result in the maintenance arm we decided ... to enroll more patients in arms B and C to increase the power of this study.”
Additionally, a phase 3 trial is planned and enrollment should begin in early 2019, he said.
Dr. Rob reported having no disclosures.
SOURCE: Rob L et al. ASCO 2018, Abstract 5509.
CHICAGO – who have undergone primary debulking surgery, according to interim findings from a randomized, phase 2, open-label trial.
Of 99 patients enrolled in the international, multicenter trial between November 2013 and March 2016, 92 received at least one dose of therapy and had a postbaseline endpoint assessment (the modified intention-to-treat population). Median progression-free survival was 18.3 months in the 31 patients who received autologous dendritic cell vaccine (DCVAC) given concomitantly with chemotherapy (group A), 24.3 months in the 30 patients who received DCVAC given sequentially as maintenance therapy after chemotherapy (group B), and 18.6 months in the 31 controls who received chemotherapy alone (group C), Lukas Rob, MD, reported at the annual meeting of the American Society of Clinical Oncology.
“When DCVAC was administered as maintenance [therapy] after the chemotherapy, the time to disease progression was prolonged by almost 6 months. Based on the hazard ratio, there was a 57% decrease in the hazard of progression in favor of arm B,” said Dr. Rob, of University Hospital Kralovske Vinohrady, Prague.
Compared with group C, progression-free survival hazard ratios were 0.64 and 0.43 in the modified intention-to-treat population in groups A and B, respectively, he said.
“These results are statistically significant and the clinical benefit is even more significant in patients who received more doses of DCVAC,” he added, explaining that the hazard ratios for patients in groups A and B who received at least eight doses of DCVAC and/or three cycles of chemotherapy (the per protocol population, including 87 patients) were 1.01 and 0.32, respectively.
Although the overall survival data in this study is immature, there is “a strong trend in favor of DCVAC in the maintenance treatment arm,” he said.
Study participants had FIGO stage III epithelial ovarian cancer (EOC) and a performance status score of 0-2. All had undergone primary debulking surgery and had less than 1 cm maximal residuum and no prior systemic therapy. Patients were randomized up to 6 weeks after surgery. Chemotherapy in all three groups included six cycles of carboplatin and paclitaxel, and DCVAC was given at doses of 1 x 107 dendritic cells per dose for a planned 10 doses.
Most patients with EOC – about 70% of those with stage III/IV disease – relapse after optimal debulking surgery and chemotherapy.
“There is no doubt that we need a new treatment modality,” he said. Because autologous DCVAC can present tumor antigens to elicit a durable immune response, he and his colleagues hypothesized that adding DCVAC to chemotherapy could improve outcomes – either when used concomitantly to target tumor-induced immune suppression and allowing for partial recovery of the immune system after each cycle, or when used sequentially as maintenance therapy, as minimal tumor burden after chemotherapy would provide optimal conditions for immune stimulation and the immune system would be fully recovered after completing cytotoxic therapy.
The treatments in this study were well tolerated; no grade 3 or greater adverse events were related solely to DCVAC, and the vaccine did not worsen the side effects of chemotherapy, Dr. Rob said.
The findings suggested that sequential DCVAC after chemotherapy in patients with EOC is a promising maintenance treatment option that can delay disease progression, he concluded, noting that “due to the excellent result in the maintenance arm we decided ... to enroll more patients in arms B and C to increase the power of this study.”
Additionally, a phase 3 trial is planned and enrollment should begin in early 2019, he said.
Dr. Rob reported having no disclosures.
SOURCE: Rob L et al. ASCO 2018, Abstract 5509.
REPORTING FROM ASCO 2018
Key clinical point: Dendritic cell vaccine maintenance improves progression-free survival in patients with epithelial ovarian carcinoma who have undergone primary debulking surgery.
Major finding: DCVAC maintenance after chemotherapy prolonged progression-free survival by almost 6 months (hazard ratio, 0.43 vs. chemotherapy alone).
Study details: A randomized, phase 2, open-label trial including 99 patients.
Disclosures: Dr. Rob reported having no disclosures.
Source: Rob L et al. ASCO 2018, Abstract 5509.
Third-line avelumab for gastric cancer safer than chemotherapy
according to a recent study.
Although no primary or secondary endpoints were met, avelumab demonstrated similar antitumor activity compared with chemotherapy, reported Yung-Jue Bang, MD, PhD, of Seoul National University College of Medicine, South Korea, and his coauthors.
“There currently are no internationally recognized treatment guidelines for patients with advanced gastric cancer/gastroesophageal junction cancer [GC/GEJC] in whom two prior lines of therapy have failed,” the investigators wrote in Annals of Oncology.
The phase 3, randomized JAVELIN Gastric 300 trial compared avelumab, an anti–programmed death ligand 1 (PD-L1) monoclonal antibody, with physician’s choice of chemotherapy in 371 patients with metastatic GC/GEJC who had received two previous lines of systemic therapy. A total of 185 patients received avelumab and 186 patients received chemotherapy. Of those in the chemotherapy group, 120 (64.5%) were given irinotecan, 50 (29.0%) were given paclitaxel, and 3 (1.6%) received best supportive care only. The primary endpoint was overall survival, and secondary endpoints were objective response rate and progression-free survival. Safety and tolerability were also evaluated.
Neither primary nor secondary endpoints were met, and no significant difference in efficacy was found between treatment arms. However, tolerability was notably better in the avelumab treatment group. Treatment-related adverse events occurred in 90 patients (48.9%) in the avelumab group, compared with 131 patients (74.0%) in the chemotherapy group. Grade 3 or higher treatment-related adverse events were also less common in the avelumab group (9.2% vs. 31.6%). In the avelumab treatment arm, 4 patients (2.2%) had grade 3 or higher immune-related adverse events (autoimmune hypothyroidism, autoimmune hepatitis, elevated AST, and colitis).
“These results demonstrate that avelumab is better tolerated than chemotherapy in patients with heavily pretreated GC/GEJC, supporting the potential of avelumab for combination or maintenance therapy, even in later stages of disease,” the investigators wrote. “Nevertheless, the optimal strategy for incorporating checkpoint inhibitors into the continuum of care for patients with advanced GC/GEJC is still unknown, and studies of alternative anti–PD-1/PD-L1 treatment strategies in earlier lines of therapy are warranted.”
The study was funded by Merck and Pfizer. Authors reported compensation from AstraZeneca, Eli Lilly, Novartis, and others.
SOURCE: Bang YJ et al. Ann Oncol. 2018 Jul 24. doi:10.1093/annonc/mdy264.
according to a recent study.
Although no primary or secondary endpoints were met, avelumab demonstrated similar antitumor activity compared with chemotherapy, reported Yung-Jue Bang, MD, PhD, of Seoul National University College of Medicine, South Korea, and his coauthors.
“There currently are no internationally recognized treatment guidelines for patients with advanced gastric cancer/gastroesophageal junction cancer [GC/GEJC] in whom two prior lines of therapy have failed,” the investigators wrote in Annals of Oncology.
The phase 3, randomized JAVELIN Gastric 300 trial compared avelumab, an anti–programmed death ligand 1 (PD-L1) monoclonal antibody, with physician’s choice of chemotherapy in 371 patients with metastatic GC/GEJC who had received two previous lines of systemic therapy. A total of 185 patients received avelumab and 186 patients received chemotherapy. Of those in the chemotherapy group, 120 (64.5%) were given irinotecan, 50 (29.0%) were given paclitaxel, and 3 (1.6%) received best supportive care only. The primary endpoint was overall survival, and secondary endpoints were objective response rate and progression-free survival. Safety and tolerability were also evaluated.
Neither primary nor secondary endpoints were met, and no significant difference in efficacy was found between treatment arms. However, tolerability was notably better in the avelumab treatment group. Treatment-related adverse events occurred in 90 patients (48.9%) in the avelumab group, compared with 131 patients (74.0%) in the chemotherapy group. Grade 3 or higher treatment-related adverse events were also less common in the avelumab group (9.2% vs. 31.6%). In the avelumab treatment arm, 4 patients (2.2%) had grade 3 or higher immune-related adverse events (autoimmune hypothyroidism, autoimmune hepatitis, elevated AST, and colitis).
“These results demonstrate that avelumab is better tolerated than chemotherapy in patients with heavily pretreated GC/GEJC, supporting the potential of avelumab for combination or maintenance therapy, even in later stages of disease,” the investigators wrote. “Nevertheless, the optimal strategy for incorporating checkpoint inhibitors into the continuum of care for patients with advanced GC/GEJC is still unknown, and studies of alternative anti–PD-1/PD-L1 treatment strategies in earlier lines of therapy are warranted.”
The study was funded by Merck and Pfizer. Authors reported compensation from AstraZeneca, Eli Lilly, Novartis, and others.
SOURCE: Bang YJ et al. Ann Oncol. 2018 Jul 24. doi:10.1093/annonc/mdy264.
according to a recent study.
Although no primary or secondary endpoints were met, avelumab demonstrated similar antitumor activity compared with chemotherapy, reported Yung-Jue Bang, MD, PhD, of Seoul National University College of Medicine, South Korea, and his coauthors.
“There currently are no internationally recognized treatment guidelines for patients with advanced gastric cancer/gastroesophageal junction cancer [GC/GEJC] in whom two prior lines of therapy have failed,” the investigators wrote in Annals of Oncology.
The phase 3, randomized JAVELIN Gastric 300 trial compared avelumab, an anti–programmed death ligand 1 (PD-L1) monoclonal antibody, with physician’s choice of chemotherapy in 371 patients with metastatic GC/GEJC who had received two previous lines of systemic therapy. A total of 185 patients received avelumab and 186 patients received chemotherapy. Of those in the chemotherapy group, 120 (64.5%) were given irinotecan, 50 (29.0%) were given paclitaxel, and 3 (1.6%) received best supportive care only. The primary endpoint was overall survival, and secondary endpoints were objective response rate and progression-free survival. Safety and tolerability were also evaluated.
Neither primary nor secondary endpoints were met, and no significant difference in efficacy was found between treatment arms. However, tolerability was notably better in the avelumab treatment group. Treatment-related adverse events occurred in 90 patients (48.9%) in the avelumab group, compared with 131 patients (74.0%) in the chemotherapy group. Grade 3 or higher treatment-related adverse events were also less common in the avelumab group (9.2% vs. 31.6%). In the avelumab treatment arm, 4 patients (2.2%) had grade 3 or higher immune-related adverse events (autoimmune hypothyroidism, autoimmune hepatitis, elevated AST, and colitis).
“These results demonstrate that avelumab is better tolerated than chemotherapy in patients with heavily pretreated GC/GEJC, supporting the potential of avelumab for combination or maintenance therapy, even in later stages of disease,” the investigators wrote. “Nevertheless, the optimal strategy for incorporating checkpoint inhibitors into the continuum of care for patients with advanced GC/GEJC is still unknown, and studies of alternative anti–PD-1/PD-L1 treatment strategies in earlier lines of therapy are warranted.”
The study was funded by Merck and Pfizer. Authors reported compensation from AstraZeneca, Eli Lilly, Novartis, and others.
SOURCE: Bang YJ et al. Ann Oncol. 2018 Jul 24. doi:10.1093/annonc/mdy264.
FROM ANNALS OF ONCOLOGY
Key clinical point: Third-line avelumab for patients with gastric cancer is safer, but no more effective, than chemotherapy.
Major finding: Grade 3 or higher treatment-related adverse events occurred in 9.2% of patients treated with avelumab, compared with 31.6% of patients treated with chemotherapy.
Study details: The phase 3 JAVELIN Gastric 300 trial involved 371 patients with metastatic, nonresectable gastric cancer who had received two previous lines of systemic therapy.
Disclosures: The study was funded by Merck and Pfizer. Authors reported compensation from AstraZeneca, Eli Lilly, Novartis, and others.
Source: Bang YJ et al. Ann Oncol. 2018 Jul 24. doi:10.1093/annonc/mdy264.
No survival boost with atezolizumab vs. regorafenib in mCRC
BARCELONA – There was no overall survival advantage for patients with metastatic colorectal cancer treated with the immune checkpoint inhibitor atezolizumab (Tencentriq) either in combination with cobimetinib (Cotellic) or as monotherapy compared with regorafenib (Stivarga), results of the randomized phase 3 IMblaze370 trial showed.
Median overall survival (OS, the primary endpoint) for 183 patients treated with atezolizumab and cobimetinib was 8.9 months, compared with 7.1 months for 90 patients treated with atezolizumab monotherapy, and 8.5 months for 90 patients treated with regorafenib. None of the differences were statistically significant, reported Johanna C. Bendell, MD, of the Sarah Cannon Research Institute in Nashville, Tenn.
“The lack of clinical activity may be due to the immune-excluded phenotype of metastatic colorectal cancer. The dual inhibition of the PD-L1 immune checkpoint and MAP kinase–mediated immune suppression may not be sufficient to generate that immune response that we need for antitumor activity,” she said at the European Society for Medical Oncology World Congress on Gastrointestinal Cancer.
In an interview, Dr. Bendell said that “we were all hoping that we were going to see some benefit, and we had some preliminary data suggesting that we would. What we still need to see is whether there are potentially populations of patients who might have benefited more, which hopefully will come out of the biomarker analysis.”
The results demonstrate the need for controlled comparison trials to detect true clinical benefits from a specific agent or combination, she added.
Microsatellite-stable disease, which accounts for approximately 95% of cases of metastatic colorectal cancer (MSS mCRC) is considered to be an immune-excluded or “cold” tumor type due to a lack of tumor-infiltrating lymphocytes. Single agent inhibitors of the programmed death 1 protein and its ligand (PD1/PD-L1) have only minimal activity in MSS mCRC, she noted.
In preclinical studies, there was evidence to suggest that the combination of atezolizumab and cobimetinib could augment antitumor T-cell responses. The combination also was shown to be safe and to demonstrate “promising” clinical activity in a phase 1b trial conducted by Dr. Bendell and her colleagues.
To see whether the initial promise of atezolizumab in this population held up under closer scrutiny, the investigators enrolled patients with MSS mCRC refractory to chemotherapy and randomly assigned them on a 2:1:1 basis to receive either atezolizumab plus cobimetinib, atezolizumab monotherapy, or regorafenib.
Atezolizumab was administered intravenously at 840 mg every 2 weeks in the combination arm, and at 1,200 mg every 3 weeks in the monotherapy arm. Cobimetinib was administered orally at 60 mg on a 21-days-on/7-days-off schedule. Regorafenib was administered orally at 160 mg on a 21-days-on/7-days-off schedule.
As noted, the trial did not meet its primary endpoint of an OS benefit in the intention-to-treat population. In addition, there were no significant differences in either median progression-free survival, overall response rates, or duration of response.
Response rates for both the atezolizumab/cobimetinib combination and atezolizumab monotherapy arms were consistent with published data for other PD-1/PD-L1 inhibitors, and the safety of the combination was consistent with the known safety profiles of the individual agents, Dr. Bendell said.
The investigators are conducting extensive biomarker evaluations and gene expression profiles, and hope to present the results of these studies at a future conference, she added.
SOURCE: Bendell J et al. ESMO GI 2018. Abstract LBA-004.
BARCELONA – There was no overall survival advantage for patients with metastatic colorectal cancer treated with the immune checkpoint inhibitor atezolizumab (Tencentriq) either in combination with cobimetinib (Cotellic) or as monotherapy compared with regorafenib (Stivarga), results of the randomized phase 3 IMblaze370 trial showed.
Median overall survival (OS, the primary endpoint) for 183 patients treated with atezolizumab and cobimetinib was 8.9 months, compared with 7.1 months for 90 patients treated with atezolizumab monotherapy, and 8.5 months for 90 patients treated with regorafenib. None of the differences were statistically significant, reported Johanna C. Bendell, MD, of the Sarah Cannon Research Institute in Nashville, Tenn.
“The lack of clinical activity may be due to the immune-excluded phenotype of metastatic colorectal cancer. The dual inhibition of the PD-L1 immune checkpoint and MAP kinase–mediated immune suppression may not be sufficient to generate that immune response that we need for antitumor activity,” she said at the European Society for Medical Oncology World Congress on Gastrointestinal Cancer.
In an interview, Dr. Bendell said that “we were all hoping that we were going to see some benefit, and we had some preliminary data suggesting that we would. What we still need to see is whether there are potentially populations of patients who might have benefited more, which hopefully will come out of the biomarker analysis.”
The results demonstrate the need for controlled comparison trials to detect true clinical benefits from a specific agent or combination, she added.
Microsatellite-stable disease, which accounts for approximately 95% of cases of metastatic colorectal cancer (MSS mCRC) is considered to be an immune-excluded or “cold” tumor type due to a lack of tumor-infiltrating lymphocytes. Single agent inhibitors of the programmed death 1 protein and its ligand (PD1/PD-L1) have only minimal activity in MSS mCRC, she noted.
In preclinical studies, there was evidence to suggest that the combination of atezolizumab and cobimetinib could augment antitumor T-cell responses. The combination also was shown to be safe and to demonstrate “promising” clinical activity in a phase 1b trial conducted by Dr. Bendell and her colleagues.
To see whether the initial promise of atezolizumab in this population held up under closer scrutiny, the investigators enrolled patients with MSS mCRC refractory to chemotherapy and randomly assigned them on a 2:1:1 basis to receive either atezolizumab plus cobimetinib, atezolizumab monotherapy, or regorafenib.
Atezolizumab was administered intravenously at 840 mg every 2 weeks in the combination arm, and at 1,200 mg every 3 weeks in the monotherapy arm. Cobimetinib was administered orally at 60 mg on a 21-days-on/7-days-off schedule. Regorafenib was administered orally at 160 mg on a 21-days-on/7-days-off schedule.
As noted, the trial did not meet its primary endpoint of an OS benefit in the intention-to-treat population. In addition, there were no significant differences in either median progression-free survival, overall response rates, or duration of response.
Response rates for both the atezolizumab/cobimetinib combination and atezolizumab monotherapy arms were consistent with published data for other PD-1/PD-L1 inhibitors, and the safety of the combination was consistent with the known safety profiles of the individual agents, Dr. Bendell said.
The investigators are conducting extensive biomarker evaluations and gene expression profiles, and hope to present the results of these studies at a future conference, she added.
SOURCE: Bendell J et al. ESMO GI 2018. Abstract LBA-004.
BARCELONA – There was no overall survival advantage for patients with metastatic colorectal cancer treated with the immune checkpoint inhibitor atezolizumab (Tencentriq) either in combination with cobimetinib (Cotellic) or as monotherapy compared with regorafenib (Stivarga), results of the randomized phase 3 IMblaze370 trial showed.
Median overall survival (OS, the primary endpoint) for 183 patients treated with atezolizumab and cobimetinib was 8.9 months, compared with 7.1 months for 90 patients treated with atezolizumab monotherapy, and 8.5 months for 90 patients treated with regorafenib. None of the differences were statistically significant, reported Johanna C. Bendell, MD, of the Sarah Cannon Research Institute in Nashville, Tenn.
“The lack of clinical activity may be due to the immune-excluded phenotype of metastatic colorectal cancer. The dual inhibition of the PD-L1 immune checkpoint and MAP kinase–mediated immune suppression may not be sufficient to generate that immune response that we need for antitumor activity,” she said at the European Society for Medical Oncology World Congress on Gastrointestinal Cancer.
In an interview, Dr. Bendell said that “we were all hoping that we were going to see some benefit, and we had some preliminary data suggesting that we would. What we still need to see is whether there are potentially populations of patients who might have benefited more, which hopefully will come out of the biomarker analysis.”
The results demonstrate the need for controlled comparison trials to detect true clinical benefits from a specific agent or combination, she added.
Microsatellite-stable disease, which accounts for approximately 95% of cases of metastatic colorectal cancer (MSS mCRC) is considered to be an immune-excluded or “cold” tumor type due to a lack of tumor-infiltrating lymphocytes. Single agent inhibitors of the programmed death 1 protein and its ligand (PD1/PD-L1) have only minimal activity in MSS mCRC, she noted.
In preclinical studies, there was evidence to suggest that the combination of atezolizumab and cobimetinib could augment antitumor T-cell responses. The combination also was shown to be safe and to demonstrate “promising” clinical activity in a phase 1b trial conducted by Dr. Bendell and her colleagues.
To see whether the initial promise of atezolizumab in this population held up under closer scrutiny, the investigators enrolled patients with MSS mCRC refractory to chemotherapy and randomly assigned them on a 2:1:1 basis to receive either atezolizumab plus cobimetinib, atezolizumab monotherapy, or regorafenib.
Atezolizumab was administered intravenously at 840 mg every 2 weeks in the combination arm, and at 1,200 mg every 3 weeks in the monotherapy arm. Cobimetinib was administered orally at 60 mg on a 21-days-on/7-days-off schedule. Regorafenib was administered orally at 160 mg on a 21-days-on/7-days-off schedule.
As noted, the trial did not meet its primary endpoint of an OS benefit in the intention-to-treat population. In addition, there were no significant differences in either median progression-free survival, overall response rates, or duration of response.
Response rates for both the atezolizumab/cobimetinib combination and atezolizumab monotherapy arms were consistent with published data for other PD-1/PD-L1 inhibitors, and the safety of the combination was consistent with the known safety profiles of the individual agents, Dr. Bendell said.
The investigators are conducting extensive biomarker evaluations and gene expression profiles, and hope to present the results of these studies at a future conference, she added.
SOURCE: Bendell J et al. ESMO GI 2018. Abstract LBA-004.
REPORTING FROM ESMO GI 2018
Key clinical point: Microsatellite stable metastatic colorectal cancer (mCRC) has a poor survival prognosis; better agents or combinations are needed.
Major finding: There were no significant differences in overall or progression-free survival, overall response rates, or duration of responses between atezolizumab with or without cobimetinib vs. regorafenib.
Study details: Randomized phase 3 trial of 363 patients with mCRC.
Disclosures: Hoffman-La Roche sponsored the study. Dr. Bendell reported no relevant disclosures. She is a member of the Oncology Practice editorial advisory board.
Source: Bendell J et al. ESMO GI 2018, Abstract LBA-004.
FDA grants accelerated approval to ipilimumab/nivolumab combo for CRC
The Food and Drug Administration has granted accelerated approval to ipilimumab (Yervoy) for the treatment of colorectal cancer (CRC) in patients aged 12 years and older, in combination with nivolumab (Opdivo). Specifically, this indication is for microsatellite instability–high or mismatch repair deficient metastatic CRC that has progressed after treatment with a fluoropyrimidine, oxaliplatin, or irinotecan, the FDA said in a press announcement.
This new use has been added to the nivolumab labeling; nivolumab received an accelerated approval of its own on July 31, 2017, as a single-agent treatment for these kinds of CRCs.
These approvals were based on the overall response rate in the nonrandomized CheckMate 142 trial. In the multiple parallel-cohort, open-label study, 82 patients received 1 mg/kg of IV ipilimumab and 3 mg/kg of IV nivolumab every 3 weeks for four doses, followed by 3 mg/kg of IV nivolumab alone every 2 weeks until either radiographic progression or unacceptable toxicity.
Their overall response rate was 46%, and 89% of those responding had response durations greater than 6 months. These rates were higher than those observed in a separate cohort of 58 patients who received only nivolumab: 28% and 67%, respectively.
The most common adverse events were fatigue, diarrhea, pyrexia, musculoskeletal pain, abdominal pain, pruritus, nausea, rash, dyspnea, decreased appetite, and vomiting.
Full prescribing information for ipilimumab and nivolumab can be found on the FDA website.
The Food and Drug Administration has granted accelerated approval to ipilimumab (Yervoy) for the treatment of colorectal cancer (CRC) in patients aged 12 years and older, in combination with nivolumab (Opdivo). Specifically, this indication is for microsatellite instability–high or mismatch repair deficient metastatic CRC that has progressed after treatment with a fluoropyrimidine, oxaliplatin, or irinotecan, the FDA said in a press announcement.
This new use has been added to the nivolumab labeling; nivolumab received an accelerated approval of its own on July 31, 2017, as a single-agent treatment for these kinds of CRCs.
These approvals were based on the overall response rate in the nonrandomized CheckMate 142 trial. In the multiple parallel-cohort, open-label study, 82 patients received 1 mg/kg of IV ipilimumab and 3 mg/kg of IV nivolumab every 3 weeks for four doses, followed by 3 mg/kg of IV nivolumab alone every 2 weeks until either radiographic progression or unacceptable toxicity.
Their overall response rate was 46%, and 89% of those responding had response durations greater than 6 months. These rates were higher than those observed in a separate cohort of 58 patients who received only nivolumab: 28% and 67%, respectively.
The most common adverse events were fatigue, diarrhea, pyrexia, musculoskeletal pain, abdominal pain, pruritus, nausea, rash, dyspnea, decreased appetite, and vomiting.
Full prescribing information for ipilimumab and nivolumab can be found on the FDA website.
The Food and Drug Administration has granted accelerated approval to ipilimumab (Yervoy) for the treatment of colorectal cancer (CRC) in patients aged 12 years and older, in combination with nivolumab (Opdivo). Specifically, this indication is for microsatellite instability–high or mismatch repair deficient metastatic CRC that has progressed after treatment with a fluoropyrimidine, oxaliplatin, or irinotecan, the FDA said in a press announcement.
This new use has been added to the nivolumab labeling; nivolumab received an accelerated approval of its own on July 31, 2017, as a single-agent treatment for these kinds of CRCs.
These approvals were based on the overall response rate in the nonrandomized CheckMate 142 trial. In the multiple parallel-cohort, open-label study, 82 patients received 1 mg/kg of IV ipilimumab and 3 mg/kg of IV nivolumab every 3 weeks for four doses, followed by 3 mg/kg of IV nivolumab alone every 2 weeks until either radiographic progression or unacceptable toxicity.
Their overall response rate was 46%, and 89% of those responding had response durations greater than 6 months. These rates were higher than those observed in a separate cohort of 58 patients who received only nivolumab: 28% and 67%, respectively.
The most common adverse events were fatigue, diarrhea, pyrexia, musculoskeletal pain, abdominal pain, pruritus, nausea, rash, dyspnea, decreased appetite, and vomiting.
Full prescribing information for ipilimumab and nivolumab can be found on the FDA website.