Targeting inflammation improved survival for some patients with advanced pancreatic cancer

Article Type
Changed
Wed, 05/26/2021 - 13:59
Display Headline
Targeting inflammation improved survival for some patients with advanced pancreatic cancer

CHICAGO – Adding the kinase inhibitor ruxolitinib to capecitabine improved overall survival of a subset of patients with metastatic pancreatic cancer, compared with capecitabine and placebo.

In a randomized phase II trial in 127 patients with metastatic pancreatic ductal adenocarcinoma, the second-line therapy combination of ruxolitinib (Jakafi) and capecitabine (Xeloda) was not associated with better outcomes than capecitabine and placebo in the overall population.

But among 60 patients with high levels of C-reactive protein indicative of systemic inflammation, adding ruxolitinib improved median overall survival to 83 days, compared with 55 days for controls, reported Dr. Herbert I. Hurwitz, professor of medicine in the division of oncology at Duke University, Durham, N.C.

Dr. Herbert Hurwitz

"These data support the role of inflammation and the JAK-STAT pathway in particular for patients with pancreatic cancer," Dr. Hurwitz said at the annual meeting of the American Society of Clinical Oncology.

Ruxolitinib is an inhibitor of the Janus kinases (JAK) 1 and 2, which have been shown to mediate cytokine signaling through activation of STAT transcription factors. In preclinical models, pro-inflammatory cytokine signaling has been shown to contribute to pancreatic cancer initiation and progression.

Systemic inflammation is also associated with weight loss, decreased muscle mass, and poor performance status, all of which can shorten survival in patients with advanced pancreatic cancer.

The investigators hypothesized that ruxolitinib could improve survival of advanced pancreatic cancer patients when added to standard chemotherapy by dampening inflammation and thereby reducing cachexia and its related effects on patients’ overall health.

In tumor xenograft models of pancreatic cancer, the combination of ruxolitinib and capecitabine showed antitumor activity, prompting investigators to evaluate the drugs in human clinical trials.

Recap of RECAP

The phase II RECAP study (A Randomized Phase II Study of Ruxolitinib Efficacy and Safety in Combination With Capecitabine for Subjects With Recurrent or Treatment Refractory Metastatic Pancreatic Cancer) enrolled 127 patients with histologically confirmed metastatic pancreatic ductal adenocarcinoma and a Karnofsky performance score of 60 or greater who had disease progression on gemcitabine (Gemzar).

The patients were randomized to capecitabine 1,000 mg/m2 twice daily for 14 days, plus either ruxolitinib 15 mg twice daily for 21 days, or placebo.

The median overall survival was 136.5 days for patients treated with the combination, and 129.5 days for those treated with capecitabine/placebo. The hazard ratio (HR) was 0.79, but the difference was not statistically significant (P = .25, above the prespecified 2-sided P value of .20).

However, when the authors performed the prespecified analysis of patients with C-reactive protein (CRP) levels above 13 mg/L, they saw a significant difference: 83.0 days for patients on the combination, vs. 55.0 days for capecitabine/placebo (HR, 0.47; 2-sided P = .01).

The 6-month overall survival rate for patients treated with the combination was 42%, compared with 11% for patients treated with capecitabine/placebo.

In a multivariate analysis of patients with high CRP levels – controlling for age, serum lactate dehydrogenase and albumin levels, liver and lung metastases, performance score, prior erlotinib (Tarceva), prior radiation or surgery, and sex – they found that the association of the combination therapy with better overall survival remained (adjusted HR, 0.50; 2-sided P = .037).

In addition, an analysis of survival according to modified Glasgow Prognostic Score (mGPS), a combination of CRP and albumin measures, showed that patients with a higher score indicative of higher degrees of systemic inflammation had better survival when treated with ruxolitinib than with placebo.

The median progression-free survival (PFS), a secondary endpoint, was in the overall population (intention to treat) 51.0 days for combination-treated patients, compared with 46.9 days for those treated with capecitabine/placebo (HR, 0.75; 2-sided P = .14). In the prespecified population with a high CRP level, the respective PFS was 48.0, vs. 41.5 days (HR, 0.62; 2-sided P = .10).

Grade 3 or 4 adverse events occurred in 74.6% of patients on the combination and 81.7% of those on capecitabine/placebo. More patients discontinued the study drug because of adverse events in the placebo group (12 patients, vs. 7 in the ruxolitinib group).

Adverse events were generally lower in frequency among patients treated with the combination, except for pulmonary embolism (a common side effect of JAK/STAT inhibitors), which occurred in seven patients on the combination, compared with three on placebo, and anemia (9 vs. 1 patient, respectively).

Invited discussant Dr. Andrew Ko of the Helen Diller Family Comprehensive Cancer Center at the University of California, San Francisco, applauded the investigators for their "smart trial design and drug development strategy."

He said that the preplanned analysis of a subgroup of interest allowed investigators to better plan for patient selection in a phase III trial, called JANUS.

 

 

He noted, however, that the cutoff point for CRP is lower for the phase III study, which could allow for greater enrollment but with the potential trade-off in lower overall efficacy.

In addition, he questioned the wisdom of a capecitabine-only reference arm, "which may be a deterrent to study enrollment and may even be obsolete at some point."

Results of the RECAP trials also suggest that the "survival benefit of ruxolitinib may relate to alleviating cachexia and inanition, as much as reducing tumor burden," he added.

The study was sponsored by Incyte. Dr Hurwitz disclosed serving as a consultant/adviser to, and receiving research funding from, Genentech, maker of capecitabine, and Novartis, which markets ruxolitinib in Europe. Dr. Ko reported no disclosures relevant to the study.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
kinase inhibitor, ruxolitinib, capecitabine, metastatic pancreatic cancer, pancreatic ductal adenocarcinoma, Jakafi, Xeloda, Dr. Herbert I. Hurwitz,
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

CHICAGO – Adding the kinase inhibitor ruxolitinib to capecitabine improved overall survival of a subset of patients with metastatic pancreatic cancer, compared with capecitabine and placebo.

In a randomized phase II trial in 127 patients with metastatic pancreatic ductal adenocarcinoma, the second-line therapy combination of ruxolitinib (Jakafi) and capecitabine (Xeloda) was not associated with better outcomes than capecitabine and placebo in the overall population.

But among 60 patients with high levels of C-reactive protein indicative of systemic inflammation, adding ruxolitinib improved median overall survival to 83 days, compared with 55 days for controls, reported Dr. Herbert I. Hurwitz, professor of medicine in the division of oncology at Duke University, Durham, N.C.

Dr. Herbert Hurwitz

"These data support the role of inflammation and the JAK-STAT pathway in particular for patients with pancreatic cancer," Dr. Hurwitz said at the annual meeting of the American Society of Clinical Oncology.

Ruxolitinib is an inhibitor of the Janus kinases (JAK) 1 and 2, which have been shown to mediate cytokine signaling through activation of STAT transcription factors. In preclinical models, pro-inflammatory cytokine signaling has been shown to contribute to pancreatic cancer initiation and progression.

Systemic inflammation is also associated with weight loss, decreased muscle mass, and poor performance status, all of which can shorten survival in patients with advanced pancreatic cancer.

The investigators hypothesized that ruxolitinib could improve survival of advanced pancreatic cancer patients when added to standard chemotherapy by dampening inflammation and thereby reducing cachexia and its related effects on patients’ overall health.

In tumor xenograft models of pancreatic cancer, the combination of ruxolitinib and capecitabine showed antitumor activity, prompting investigators to evaluate the drugs in human clinical trials.

Recap of RECAP

The phase II RECAP study (A Randomized Phase II Study of Ruxolitinib Efficacy and Safety in Combination With Capecitabine for Subjects With Recurrent or Treatment Refractory Metastatic Pancreatic Cancer) enrolled 127 patients with histologically confirmed metastatic pancreatic ductal adenocarcinoma and a Karnofsky performance score of 60 or greater who had disease progression on gemcitabine (Gemzar).

The patients were randomized to capecitabine 1,000 mg/m2 twice daily for 14 days, plus either ruxolitinib 15 mg twice daily for 21 days, or placebo.

The median overall survival was 136.5 days for patients treated with the combination, and 129.5 days for those treated with capecitabine/placebo. The hazard ratio (HR) was 0.79, but the difference was not statistically significant (P = .25, above the prespecified 2-sided P value of .20).

However, when the authors performed the prespecified analysis of patients with C-reactive protein (CRP) levels above 13 mg/L, they saw a significant difference: 83.0 days for patients on the combination, vs. 55.0 days for capecitabine/placebo (HR, 0.47; 2-sided P = .01).

The 6-month overall survival rate for patients treated with the combination was 42%, compared with 11% for patients treated with capecitabine/placebo.

In a multivariate analysis of patients with high CRP levels – controlling for age, serum lactate dehydrogenase and albumin levels, liver and lung metastases, performance score, prior erlotinib (Tarceva), prior radiation or surgery, and sex – they found that the association of the combination therapy with better overall survival remained (adjusted HR, 0.50; 2-sided P = .037).

In addition, an analysis of survival according to modified Glasgow Prognostic Score (mGPS), a combination of CRP and albumin measures, showed that patients with a higher score indicative of higher degrees of systemic inflammation had better survival when treated with ruxolitinib than with placebo.

The median progression-free survival (PFS), a secondary endpoint, was in the overall population (intention to treat) 51.0 days for combination-treated patients, compared with 46.9 days for those treated with capecitabine/placebo (HR, 0.75; 2-sided P = .14). In the prespecified population with a high CRP level, the respective PFS was 48.0, vs. 41.5 days (HR, 0.62; 2-sided P = .10).

Grade 3 or 4 adverse events occurred in 74.6% of patients on the combination and 81.7% of those on capecitabine/placebo. More patients discontinued the study drug because of adverse events in the placebo group (12 patients, vs. 7 in the ruxolitinib group).

Adverse events were generally lower in frequency among patients treated with the combination, except for pulmonary embolism (a common side effect of JAK/STAT inhibitors), which occurred in seven patients on the combination, compared with three on placebo, and anemia (9 vs. 1 patient, respectively).

Invited discussant Dr. Andrew Ko of the Helen Diller Family Comprehensive Cancer Center at the University of California, San Francisco, applauded the investigators for their "smart trial design and drug development strategy."

He said that the preplanned analysis of a subgroup of interest allowed investigators to better plan for patient selection in a phase III trial, called JANUS.

 

 

He noted, however, that the cutoff point for CRP is lower for the phase III study, which could allow for greater enrollment but with the potential trade-off in lower overall efficacy.

In addition, he questioned the wisdom of a capecitabine-only reference arm, "which may be a deterrent to study enrollment and may even be obsolete at some point."

Results of the RECAP trials also suggest that the "survival benefit of ruxolitinib may relate to alleviating cachexia and inanition, as much as reducing tumor burden," he added.

The study was sponsored by Incyte. Dr Hurwitz disclosed serving as a consultant/adviser to, and receiving research funding from, Genentech, maker of capecitabine, and Novartis, which markets ruxolitinib in Europe. Dr. Ko reported no disclosures relevant to the study.

CHICAGO – Adding the kinase inhibitor ruxolitinib to capecitabine improved overall survival of a subset of patients with metastatic pancreatic cancer, compared with capecitabine and placebo.

In a randomized phase II trial in 127 patients with metastatic pancreatic ductal adenocarcinoma, the second-line therapy combination of ruxolitinib (Jakafi) and capecitabine (Xeloda) was not associated with better outcomes than capecitabine and placebo in the overall population.

But among 60 patients with high levels of C-reactive protein indicative of systemic inflammation, adding ruxolitinib improved median overall survival to 83 days, compared with 55 days for controls, reported Dr. Herbert I. Hurwitz, professor of medicine in the division of oncology at Duke University, Durham, N.C.

Dr. Herbert Hurwitz

"These data support the role of inflammation and the JAK-STAT pathway in particular for patients with pancreatic cancer," Dr. Hurwitz said at the annual meeting of the American Society of Clinical Oncology.

Ruxolitinib is an inhibitor of the Janus kinases (JAK) 1 and 2, which have been shown to mediate cytokine signaling through activation of STAT transcription factors. In preclinical models, pro-inflammatory cytokine signaling has been shown to contribute to pancreatic cancer initiation and progression.

Systemic inflammation is also associated with weight loss, decreased muscle mass, and poor performance status, all of which can shorten survival in patients with advanced pancreatic cancer.

The investigators hypothesized that ruxolitinib could improve survival of advanced pancreatic cancer patients when added to standard chemotherapy by dampening inflammation and thereby reducing cachexia and its related effects on patients’ overall health.

In tumor xenograft models of pancreatic cancer, the combination of ruxolitinib and capecitabine showed antitumor activity, prompting investigators to evaluate the drugs in human clinical trials.

Recap of RECAP

The phase II RECAP study (A Randomized Phase II Study of Ruxolitinib Efficacy and Safety in Combination With Capecitabine for Subjects With Recurrent or Treatment Refractory Metastatic Pancreatic Cancer) enrolled 127 patients with histologically confirmed metastatic pancreatic ductal adenocarcinoma and a Karnofsky performance score of 60 or greater who had disease progression on gemcitabine (Gemzar).

The patients were randomized to capecitabine 1,000 mg/m2 twice daily for 14 days, plus either ruxolitinib 15 mg twice daily for 21 days, or placebo.

The median overall survival was 136.5 days for patients treated with the combination, and 129.5 days for those treated with capecitabine/placebo. The hazard ratio (HR) was 0.79, but the difference was not statistically significant (P = .25, above the prespecified 2-sided P value of .20).

However, when the authors performed the prespecified analysis of patients with C-reactive protein (CRP) levels above 13 mg/L, they saw a significant difference: 83.0 days for patients on the combination, vs. 55.0 days for capecitabine/placebo (HR, 0.47; 2-sided P = .01).

The 6-month overall survival rate for patients treated with the combination was 42%, compared with 11% for patients treated with capecitabine/placebo.

In a multivariate analysis of patients with high CRP levels – controlling for age, serum lactate dehydrogenase and albumin levels, liver and lung metastases, performance score, prior erlotinib (Tarceva), prior radiation or surgery, and sex – they found that the association of the combination therapy with better overall survival remained (adjusted HR, 0.50; 2-sided P = .037).

In addition, an analysis of survival according to modified Glasgow Prognostic Score (mGPS), a combination of CRP and albumin measures, showed that patients with a higher score indicative of higher degrees of systemic inflammation had better survival when treated with ruxolitinib than with placebo.

The median progression-free survival (PFS), a secondary endpoint, was in the overall population (intention to treat) 51.0 days for combination-treated patients, compared with 46.9 days for those treated with capecitabine/placebo (HR, 0.75; 2-sided P = .14). In the prespecified population with a high CRP level, the respective PFS was 48.0, vs. 41.5 days (HR, 0.62; 2-sided P = .10).

Grade 3 or 4 adverse events occurred in 74.6% of patients on the combination and 81.7% of those on capecitabine/placebo. More patients discontinued the study drug because of adverse events in the placebo group (12 patients, vs. 7 in the ruxolitinib group).

Adverse events were generally lower in frequency among patients treated with the combination, except for pulmonary embolism (a common side effect of JAK/STAT inhibitors), which occurred in seven patients on the combination, compared with three on placebo, and anemia (9 vs. 1 patient, respectively).

Invited discussant Dr. Andrew Ko of the Helen Diller Family Comprehensive Cancer Center at the University of California, San Francisco, applauded the investigators for their "smart trial design and drug development strategy."

He said that the preplanned analysis of a subgroup of interest allowed investigators to better plan for patient selection in a phase III trial, called JANUS.

 

 

He noted, however, that the cutoff point for CRP is lower for the phase III study, which could allow for greater enrollment but with the potential trade-off in lower overall efficacy.

In addition, he questioned the wisdom of a capecitabine-only reference arm, "which may be a deterrent to study enrollment and may even be obsolete at some point."

Results of the RECAP trials also suggest that the "survival benefit of ruxolitinib may relate to alleviating cachexia and inanition, as much as reducing tumor burden," he added.

The study was sponsored by Incyte. Dr Hurwitz disclosed serving as a consultant/adviser to, and receiving research funding from, Genentech, maker of capecitabine, and Novartis, which markets ruxolitinib in Europe. Dr. Ko reported no disclosures relevant to the study.

Publications
Publications
Topics
Article Type
Display Headline
Targeting inflammation improved survival for some patients with advanced pancreatic cancer
Display Headline
Targeting inflammation improved survival for some patients with advanced pancreatic cancer
Legacy Keywords
kinase inhibitor, ruxolitinib, capecitabine, metastatic pancreatic cancer, pancreatic ductal adenocarcinoma, Jakafi, Xeloda, Dr. Herbert I. Hurwitz,
Legacy Keywords
kinase inhibitor, ruxolitinib, capecitabine, metastatic pancreatic cancer, pancreatic ductal adenocarcinoma, Jakafi, Xeloda, Dr. Herbert I. Hurwitz,
Article Source

AT THE ASCO ANNUAL MEETING 2014

PURLs Copyright

Inside the Article

Vitals

Major finding: Median overall survival of patients with metastatic pancreatic cancer with CRP levels above 13 mg/L treated with ruxolitinib and capecitabine was 83 days, vs. 55 days for capecitabine/placebo-treated patients.

Data source: A randomized phase II trial with 127 patients, 60 of whom had serum C-reactive protein levels above 13 mg/L.

Disclosures: The study was sponsored by Incyte. Dr Hurwitz disclosed serving as a consultant/adviser to, and receiving research funding from, Genentech, maker of capecitabine, and Novartis, which markets ruxolitinib in Europe. Dr. Ko reported no disclosures relevant to the study.

Time to look beyond wild-type KRAS in metastatic CRC?

Article Type
Changed
Wed, 05/26/2021 - 13:59
Display Headline
Time to look beyond wild-type KRAS in metastatic CRC?

CHICAGO – Molecular testing of patients with metastatic colorectal cancer for activating mutations in KRAS and NRAS oncogenes can help clinicians choose the most appropriate first-line therapy, findings of a mutational analysis study suggest.

In the CRYSTAL study, the addition of cetuximab (Erbitux) to the FOLFIRI regimen (irinotecan, 5-fluorouracil, and leucovorin) improved survival in patients with wild-type KRAS (codons 12 and 13 in exon 2) compared with FOLFIRI alone, but not those with mutations in KRAS exon 2.

The current study, a retrospective analysis of a subgroup of patients with wild-type KRAS who had mutations in other KRAS or NRAS exons, showed that any RAS mutation neutralized the benefit of cetuximab, reported Dr. Eric Van Cutsem of University Hospitals Gasthuisberg Leuven and KU Leuven, Belgium.

"I think it’s fair to say that exclusion of patients with other RAS mutations from the KRAS codon 12/13 wild-type exon 2 treatment population improves the benefit-risk ratio associated with the addition of cetuximab to FOLFIRI. We believe therefore that all patients with colorectal cancer should be treated for a full, extensive RAS analysis from today on, and that this marker testing of tumors for all activating mutations of KRAS and NRAS is therefore essential in selecting the most appropriate first-line treatment in patients with metastatic colorectal cancer," he said at the annual meeting of the American Society of Clinical Oncology.

Dr. Van Cutsem and his colleagues conducted an exploratory analysis looking at the treatment effect of adding cetuximab to FOLFIRI in patients with KRAS codon 12/13 wild-type metastatic colorectal cancer.

BEAMing up RAS

They identified 430 patients with tumor samples evaluable for other RAS mutations using BEAMing (Beads, Emulsions, Amplification, Magnetics) technology. The technique, which some investigators have dubbed "liquid biopsy," involves treating plasma with beads coated with DNA sequences that are complementary to target mutational sequences to create an emulsion polymerase chain reaction (PCR). The PCR amplifies the circulating DNA, which can then be identified with flow cytometry. The test can detect one circulating DNA molecule per 10,000 molecules in plasma, according to the American Association for Cancer Research.

Among all 430 RAS evaluable patients, FOLFIRI plus cetuximab was associated with better progression-free survival (PFS; 11.3 vs. 7.7 months for FOLFIRI alone, hazard ratio [HR] 0.58, P = .0001), overall survival (OS; 26.1 vs. 20.2 months, HR 0.75, P = .0080), and objective response rate (ORR; 61.4% vs. 38.2%, HR 2.64, P less than .0001).

But in a subanalysis of the 63 patients (14.7%) with wild-type KRAS exon 2 but new RAS mutations in KRAS exons 3 and 4, or NRAS exons, 2, 3 and 4, they found that any RAS mutation appeared to nullify the benefit of adding cetuximab, with no significant differences between FOLFIRI with or without cetuximab in either PFS, OS, or ORR.

For example, for patients with RAS wild type, median PFS for those treated with FOLFIRI plus cetuximab was 11.4 months vs. 8.4 months for those treated with FOLFIRI only (HR 0.50, 95% confidence interval [CI] 0.41-0.76). But for patients with any RAS mutation, the PFS was 7.4 and 7.5 months respectively (HR 1.10, CI, 0.85-1.4).

Similarly, median overall survival among all RAS wild-type patients in the subgroup was 28.4 months vs. 20.2 months (HR 0.69, CI 0.54-0.88), but among those with any RAS mutation was 16.4 vs. 17.7 months (HR 1.05, CI, 0.86-1.28)

Although cetuximab did not benefit patients with RAS mutations, neither did it cause harm, as evidenced by a similar safety profile among RAS wild-type and mutant subgroups, Dr Van Cutsem said.

Evidence from this and five other clinical trials suggests that clinicians should not use epidermal growth factor receptor (EGFR) inhibitors in patients with tumors that harbor any RAS mutations, said Dr. Neal J. Meropol, chief of hematology and oncology at the University Hospitals Seidman Cancer Center at Case Western Reserve University, Cleveland.

"I view this as a four-star [out of five] recommendation. As each of the new mutations are rare, and real differences in biologic behavior and response to treatment are still possible within these rare sub [mutations]," he said.

Although the differences in outcomes between various studies may be attributable to the use of different testing platforms, "it seems very unlikely to me that the benefit of treatment in one of these rare [mutations] will be great enough to conclude that this is a high-value treatment for these patients. Further pooled analyses across studies are needed to provide additional insight into this question," he said.

The study was supported by Merck. Dr. Van Cutsem reported receiving research funding from Merck Serono. Dr. Meropol disclosed serving as consultant/adviser to Precision Therapeutics.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

CHICAGO – Molecular testing of patients with metastatic colorectal cancer for activating mutations in KRAS and NRAS oncogenes can help clinicians choose the most appropriate first-line therapy, findings of a mutational analysis study suggest.

In the CRYSTAL study, the addition of cetuximab (Erbitux) to the FOLFIRI regimen (irinotecan, 5-fluorouracil, and leucovorin) improved survival in patients with wild-type KRAS (codons 12 and 13 in exon 2) compared with FOLFIRI alone, but not those with mutations in KRAS exon 2.

The current study, a retrospective analysis of a subgroup of patients with wild-type KRAS who had mutations in other KRAS or NRAS exons, showed that any RAS mutation neutralized the benefit of cetuximab, reported Dr. Eric Van Cutsem of University Hospitals Gasthuisberg Leuven and KU Leuven, Belgium.

"I think it’s fair to say that exclusion of patients with other RAS mutations from the KRAS codon 12/13 wild-type exon 2 treatment population improves the benefit-risk ratio associated with the addition of cetuximab to FOLFIRI. We believe therefore that all patients with colorectal cancer should be treated for a full, extensive RAS analysis from today on, and that this marker testing of tumors for all activating mutations of KRAS and NRAS is therefore essential in selecting the most appropriate first-line treatment in patients with metastatic colorectal cancer," he said at the annual meeting of the American Society of Clinical Oncology.

Dr. Van Cutsem and his colleagues conducted an exploratory analysis looking at the treatment effect of adding cetuximab to FOLFIRI in patients with KRAS codon 12/13 wild-type metastatic colorectal cancer.

BEAMing up RAS

They identified 430 patients with tumor samples evaluable for other RAS mutations using BEAMing (Beads, Emulsions, Amplification, Magnetics) technology. The technique, which some investigators have dubbed "liquid biopsy," involves treating plasma with beads coated with DNA sequences that are complementary to target mutational sequences to create an emulsion polymerase chain reaction (PCR). The PCR amplifies the circulating DNA, which can then be identified with flow cytometry. The test can detect one circulating DNA molecule per 10,000 molecules in plasma, according to the American Association for Cancer Research.

Among all 430 RAS evaluable patients, FOLFIRI plus cetuximab was associated with better progression-free survival (PFS; 11.3 vs. 7.7 months for FOLFIRI alone, hazard ratio [HR] 0.58, P = .0001), overall survival (OS; 26.1 vs. 20.2 months, HR 0.75, P = .0080), and objective response rate (ORR; 61.4% vs. 38.2%, HR 2.64, P less than .0001).

But in a subanalysis of the 63 patients (14.7%) with wild-type KRAS exon 2 but new RAS mutations in KRAS exons 3 and 4, or NRAS exons, 2, 3 and 4, they found that any RAS mutation appeared to nullify the benefit of adding cetuximab, with no significant differences between FOLFIRI with or without cetuximab in either PFS, OS, or ORR.

For example, for patients with RAS wild type, median PFS for those treated with FOLFIRI plus cetuximab was 11.4 months vs. 8.4 months for those treated with FOLFIRI only (HR 0.50, 95% confidence interval [CI] 0.41-0.76). But for patients with any RAS mutation, the PFS was 7.4 and 7.5 months respectively (HR 1.10, CI, 0.85-1.4).

Similarly, median overall survival among all RAS wild-type patients in the subgroup was 28.4 months vs. 20.2 months (HR 0.69, CI 0.54-0.88), but among those with any RAS mutation was 16.4 vs. 17.7 months (HR 1.05, CI, 0.86-1.28)

Although cetuximab did not benefit patients with RAS mutations, neither did it cause harm, as evidenced by a similar safety profile among RAS wild-type and mutant subgroups, Dr Van Cutsem said.

Evidence from this and five other clinical trials suggests that clinicians should not use epidermal growth factor receptor (EGFR) inhibitors in patients with tumors that harbor any RAS mutations, said Dr. Neal J. Meropol, chief of hematology and oncology at the University Hospitals Seidman Cancer Center at Case Western Reserve University, Cleveland.

"I view this as a four-star [out of five] recommendation. As each of the new mutations are rare, and real differences in biologic behavior and response to treatment are still possible within these rare sub [mutations]," he said.

Although the differences in outcomes between various studies may be attributable to the use of different testing platforms, "it seems very unlikely to me that the benefit of treatment in one of these rare [mutations] will be great enough to conclude that this is a high-value treatment for these patients. Further pooled analyses across studies are needed to provide additional insight into this question," he said.

The study was supported by Merck. Dr. Van Cutsem reported receiving research funding from Merck Serono. Dr. Meropol disclosed serving as consultant/adviser to Precision Therapeutics.

CHICAGO – Molecular testing of patients with metastatic colorectal cancer for activating mutations in KRAS and NRAS oncogenes can help clinicians choose the most appropriate first-line therapy, findings of a mutational analysis study suggest.

In the CRYSTAL study, the addition of cetuximab (Erbitux) to the FOLFIRI regimen (irinotecan, 5-fluorouracil, and leucovorin) improved survival in patients with wild-type KRAS (codons 12 and 13 in exon 2) compared with FOLFIRI alone, but not those with mutations in KRAS exon 2.

The current study, a retrospective analysis of a subgroup of patients with wild-type KRAS who had mutations in other KRAS or NRAS exons, showed that any RAS mutation neutralized the benefit of cetuximab, reported Dr. Eric Van Cutsem of University Hospitals Gasthuisberg Leuven and KU Leuven, Belgium.

"I think it’s fair to say that exclusion of patients with other RAS mutations from the KRAS codon 12/13 wild-type exon 2 treatment population improves the benefit-risk ratio associated with the addition of cetuximab to FOLFIRI. We believe therefore that all patients with colorectal cancer should be treated for a full, extensive RAS analysis from today on, and that this marker testing of tumors for all activating mutations of KRAS and NRAS is therefore essential in selecting the most appropriate first-line treatment in patients with metastatic colorectal cancer," he said at the annual meeting of the American Society of Clinical Oncology.

Dr. Van Cutsem and his colleagues conducted an exploratory analysis looking at the treatment effect of adding cetuximab to FOLFIRI in patients with KRAS codon 12/13 wild-type metastatic colorectal cancer.

BEAMing up RAS

They identified 430 patients with tumor samples evaluable for other RAS mutations using BEAMing (Beads, Emulsions, Amplification, Magnetics) technology. The technique, which some investigators have dubbed "liquid biopsy," involves treating plasma with beads coated with DNA sequences that are complementary to target mutational sequences to create an emulsion polymerase chain reaction (PCR). The PCR amplifies the circulating DNA, which can then be identified with flow cytometry. The test can detect one circulating DNA molecule per 10,000 molecules in plasma, according to the American Association for Cancer Research.

Among all 430 RAS evaluable patients, FOLFIRI plus cetuximab was associated with better progression-free survival (PFS; 11.3 vs. 7.7 months for FOLFIRI alone, hazard ratio [HR] 0.58, P = .0001), overall survival (OS; 26.1 vs. 20.2 months, HR 0.75, P = .0080), and objective response rate (ORR; 61.4% vs. 38.2%, HR 2.64, P less than .0001).

But in a subanalysis of the 63 patients (14.7%) with wild-type KRAS exon 2 but new RAS mutations in KRAS exons 3 and 4, or NRAS exons, 2, 3 and 4, they found that any RAS mutation appeared to nullify the benefit of adding cetuximab, with no significant differences between FOLFIRI with or without cetuximab in either PFS, OS, or ORR.

For example, for patients with RAS wild type, median PFS for those treated with FOLFIRI plus cetuximab was 11.4 months vs. 8.4 months for those treated with FOLFIRI only (HR 0.50, 95% confidence interval [CI] 0.41-0.76). But for patients with any RAS mutation, the PFS was 7.4 and 7.5 months respectively (HR 1.10, CI, 0.85-1.4).

Similarly, median overall survival among all RAS wild-type patients in the subgroup was 28.4 months vs. 20.2 months (HR 0.69, CI 0.54-0.88), but among those with any RAS mutation was 16.4 vs. 17.7 months (HR 1.05, CI, 0.86-1.28)

Although cetuximab did not benefit patients with RAS mutations, neither did it cause harm, as evidenced by a similar safety profile among RAS wild-type and mutant subgroups, Dr Van Cutsem said.

Evidence from this and five other clinical trials suggests that clinicians should not use epidermal growth factor receptor (EGFR) inhibitors in patients with tumors that harbor any RAS mutations, said Dr. Neal J. Meropol, chief of hematology and oncology at the University Hospitals Seidman Cancer Center at Case Western Reserve University, Cleveland.

"I view this as a four-star [out of five] recommendation. As each of the new mutations are rare, and real differences in biologic behavior and response to treatment are still possible within these rare sub [mutations]," he said.

Although the differences in outcomes between various studies may be attributable to the use of different testing platforms, "it seems very unlikely to me that the benefit of treatment in one of these rare [mutations] will be great enough to conclude that this is a high-value treatment for these patients. Further pooled analyses across studies are needed to provide additional insight into this question," he said.

The study was supported by Merck. Dr. Van Cutsem reported receiving research funding from Merck Serono. Dr. Meropol disclosed serving as consultant/adviser to Precision Therapeutics.

Publications
Publications
Topics
Article Type
Display Headline
Time to look beyond wild-type KRAS in metastatic CRC?
Display Headline
Time to look beyond wild-type KRAS in metastatic CRC?
Article Source

AT THE ASCO ANNUAL MEETING 2014

PURLs Copyright

Inside the Article

Vitals

Major finding: Among 63 patients with metastatic colorectal cancer with wild-type KRAS exon 2 but new RAS mutations in other exons, there was no benefit in either progression-free or overall survival from the addition of cetuximab to FOLFIRI.

Data source: Subgroup analysis from the CRYSTAL trial of 430 patients with tumor samples evaluable for RAS mutational status.

Disclosures: The study was supported by Merck. Dr. Van Cutsem reported receiving research funding from Merck Serono. Dr. Meropol disclosed serving as consultant/adviser to Precision Therapeutics.

Maintenance improves PFS in patients with metastatic colorectal cancer

Article Type
Changed
Wed, 05/26/2021 - 13:59
Display Headline
Maintenance improves PFS in patients with metastatic colorectal cancer

CHICAGO – Patients with metastatic colorectal cancer who had at least stable disease after induction chemotherapy fared better on maintenance therapy with capecitabine and bevacizumab than on observation alone.

That’s the conclusion reached by investigators in a phase III trial conducted in the Netherlands. They found that patients who were randomly assigned to maintenance therapy after six cycles of induction therapy with capecitabine (Xeloda), oxaliplatin (Eloxatin), and bevacizumab (Avastin) – the CAPOX-B regimen – had better progression-free survival (PFS) and time to progression (TTP) than did patients assigned to observation alone.

With maintenance therapy, "the quality of life is maintained and clinically not inferior to observation," said lead author Dr. Miriam Koopman of the University Medical Center Utrecht Cancer Center, the Netherlands. She presented the final results of the CAIRO3 (Maintenance Treatment With Capecitabine and Bevacizumab Versus Observation After Induction Treatment With Chemotherapy and Bevacizumab as First-line Treatment) trial at the annual meeting of the American Society of Clinical Oncology.

The investigators enrolled 588 patients with metastatic colorectal cancer who had a response of stable-disease or better following six cycles of CAPOX-B to either observation or maintenance with oral capecitabine 625 mg/m2 twice daily and intravenous bevacizumab 7.5 mg/kg on day 1 every 3 weeks. The patients were stratified by prior adjuvant therapy, serum lactate dehydrogenase, response to induction therapy, World Health Organization performance status, and institution.

In both arms, CAPOX-B reintroduction was planned at the time of first progression. The primary endpoint was progression-free survival following reintroduction of CAPOX-B (dubbed PFS2). For those patients who for any reason did not receive CAPOX-B after the first PFS, PFS2 was considered to be equal to PFS1, Dr. Koopman explained.

Of the 279 patients assigned to observation, 168 (60%) had CAPOX-B reintroduced. Of the remaining 111 (40%) of patients in this arm, seven had ongoing observation, 31 received no treatment, and 73 received treatment other than CAPOX-B.

Of the 279 assigned to maintenance, 132 (47%) had CAPOX-B reintroduced. Of the remaining 147 patients (53%) in this arm, 13 continued on maintenance, 45 had no further treatment, 88 had other treatments, and 1 withdrew.

The median PFS from the time of randomization (not including induction) to first progression was 4.1 months in the observation arm and 8.5 months in the maintenance arm (stratified hazard ratio 0.43, P less than .0001).

The benefits of maintenance were also seen with the primary endpoint of PFS2, with a median of 8.5 months for observation, compared with 11.7 months for maintenance (stratified HR 0.67, P less than .0001)

The median time to second progression was 11.1 months among patients randomized to observation, vs. 13. 9 months for those randomized to maintenance (stratified HR 0.68, P less than .0001).

There were no significant differences in median overall survival (OS), however, at 18.1 months for the observation arm and 21.6 months for the maintenance arm.

There was a small but significant difference in quality of life scores between the groups (3.9 point difference on a 100-point scale, P = .004), but this difference was too small to be considered clinically meaningful, Dr. Koopman said.

In preplanned subgroup analyses, the authors found evidence to suggest that patients with synchronous metastases who had resection of the primary tumor had a greater OS benefit from maintenance therapy than did patients with metachronous disease, and that those who had complete or partial response as best response to induction therapy seemed to do better than did patients with stable disease after induction.

"We do not have a clear-cut explanation for this subgroup analysis," Dr. Koopman said.

The investigators hypothesize that the differences between patients with synchronous and metachronous disease could be due to differences in sensitivity to systemic treatment, the prognostic role of resection of the primary tumor, dependence of the angiogenic environment in metastases on the resection status of the primary tumor, or co-option of the local vasculature by the tumor, leading to decreased sensitivity of metachronous metastases to bevacizumab.

The CAIRO3 results suggest that "treatment breaks for all patients at 4 months may be too many breaks, and too early," said Dr. Leonard Saltz, chief of the gastrointestinal oncology service at Memorial Sloan-Kettering Cancer Center, New York.

"Responding patients likely benefit from treatment at least until maximal response, and that’s an aspect to consider in terms of figuring out how to utilize a treatment strategy. And treatment-break strategies will need to be individualized, but should not be abandoned," said Dr. Saltz, who was the invited discussant.

He also cautioned that the quality of life results reported by Dr. Koopman and colleagues may not accurately reflect how patients feel.

 

 

"In terms of the quality of life, what we have to conclude is that as our instruments can measure it we do not see a detriment in quality of life. But intuitively, we can conjecture that being on chemotherapy has some negative aspects – just ask any of our patients. So the idea that there is no detriment in quality of life when we’re comparing chemotherapy to nonchemotherapy suggests that we need more sensitive and specific tools for the question," he said.

The CAIRO3 study was supported by the Dutch Cancer Foundation and by unrestricted scientific grants from Roche and Sanofi-Aventis. Dr. Koopman disclosed ties with Roche/Genentech and Sanofi. Dr. Saltz disclosed ties with Roche, Genentech, and Sanofi.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
metastatic colorectal cancer, capecitabine, bevacizumab, Xeloda, oxaliplatin, Eloxatin, Avastin, CAPOX-B regimen,
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

CHICAGO – Patients with metastatic colorectal cancer who had at least stable disease after induction chemotherapy fared better on maintenance therapy with capecitabine and bevacizumab than on observation alone.

That’s the conclusion reached by investigators in a phase III trial conducted in the Netherlands. They found that patients who were randomly assigned to maintenance therapy after six cycles of induction therapy with capecitabine (Xeloda), oxaliplatin (Eloxatin), and bevacizumab (Avastin) – the CAPOX-B regimen – had better progression-free survival (PFS) and time to progression (TTP) than did patients assigned to observation alone.

With maintenance therapy, "the quality of life is maintained and clinically not inferior to observation," said lead author Dr. Miriam Koopman of the University Medical Center Utrecht Cancer Center, the Netherlands. She presented the final results of the CAIRO3 (Maintenance Treatment With Capecitabine and Bevacizumab Versus Observation After Induction Treatment With Chemotherapy and Bevacizumab as First-line Treatment) trial at the annual meeting of the American Society of Clinical Oncology.

The investigators enrolled 588 patients with metastatic colorectal cancer who had a response of stable-disease or better following six cycles of CAPOX-B to either observation or maintenance with oral capecitabine 625 mg/m2 twice daily and intravenous bevacizumab 7.5 mg/kg on day 1 every 3 weeks. The patients were stratified by prior adjuvant therapy, serum lactate dehydrogenase, response to induction therapy, World Health Organization performance status, and institution.

In both arms, CAPOX-B reintroduction was planned at the time of first progression. The primary endpoint was progression-free survival following reintroduction of CAPOX-B (dubbed PFS2). For those patients who for any reason did not receive CAPOX-B after the first PFS, PFS2 was considered to be equal to PFS1, Dr. Koopman explained.

Of the 279 patients assigned to observation, 168 (60%) had CAPOX-B reintroduced. Of the remaining 111 (40%) of patients in this arm, seven had ongoing observation, 31 received no treatment, and 73 received treatment other than CAPOX-B.

Of the 279 assigned to maintenance, 132 (47%) had CAPOX-B reintroduced. Of the remaining 147 patients (53%) in this arm, 13 continued on maintenance, 45 had no further treatment, 88 had other treatments, and 1 withdrew.

The median PFS from the time of randomization (not including induction) to first progression was 4.1 months in the observation arm and 8.5 months in the maintenance arm (stratified hazard ratio 0.43, P less than .0001).

The benefits of maintenance were also seen with the primary endpoint of PFS2, with a median of 8.5 months for observation, compared with 11.7 months for maintenance (stratified HR 0.67, P less than .0001)

The median time to second progression was 11.1 months among patients randomized to observation, vs. 13. 9 months for those randomized to maintenance (stratified HR 0.68, P less than .0001).

There were no significant differences in median overall survival (OS), however, at 18.1 months for the observation arm and 21.6 months for the maintenance arm.

There was a small but significant difference in quality of life scores between the groups (3.9 point difference on a 100-point scale, P = .004), but this difference was too small to be considered clinically meaningful, Dr. Koopman said.

In preplanned subgroup analyses, the authors found evidence to suggest that patients with synchronous metastases who had resection of the primary tumor had a greater OS benefit from maintenance therapy than did patients with metachronous disease, and that those who had complete or partial response as best response to induction therapy seemed to do better than did patients with stable disease after induction.

"We do not have a clear-cut explanation for this subgroup analysis," Dr. Koopman said.

The investigators hypothesize that the differences between patients with synchronous and metachronous disease could be due to differences in sensitivity to systemic treatment, the prognostic role of resection of the primary tumor, dependence of the angiogenic environment in metastases on the resection status of the primary tumor, or co-option of the local vasculature by the tumor, leading to decreased sensitivity of metachronous metastases to bevacizumab.

The CAIRO3 results suggest that "treatment breaks for all patients at 4 months may be too many breaks, and too early," said Dr. Leonard Saltz, chief of the gastrointestinal oncology service at Memorial Sloan-Kettering Cancer Center, New York.

"Responding patients likely benefit from treatment at least until maximal response, and that’s an aspect to consider in terms of figuring out how to utilize a treatment strategy. And treatment-break strategies will need to be individualized, but should not be abandoned," said Dr. Saltz, who was the invited discussant.

He also cautioned that the quality of life results reported by Dr. Koopman and colleagues may not accurately reflect how patients feel.

 

 

"In terms of the quality of life, what we have to conclude is that as our instruments can measure it we do not see a detriment in quality of life. But intuitively, we can conjecture that being on chemotherapy has some negative aspects – just ask any of our patients. So the idea that there is no detriment in quality of life when we’re comparing chemotherapy to nonchemotherapy suggests that we need more sensitive and specific tools for the question," he said.

The CAIRO3 study was supported by the Dutch Cancer Foundation and by unrestricted scientific grants from Roche and Sanofi-Aventis. Dr. Koopman disclosed ties with Roche/Genentech and Sanofi. Dr. Saltz disclosed ties with Roche, Genentech, and Sanofi.

CHICAGO – Patients with metastatic colorectal cancer who had at least stable disease after induction chemotherapy fared better on maintenance therapy with capecitabine and bevacizumab than on observation alone.

That’s the conclusion reached by investigators in a phase III trial conducted in the Netherlands. They found that patients who were randomly assigned to maintenance therapy after six cycles of induction therapy with capecitabine (Xeloda), oxaliplatin (Eloxatin), and bevacizumab (Avastin) – the CAPOX-B regimen – had better progression-free survival (PFS) and time to progression (TTP) than did patients assigned to observation alone.

With maintenance therapy, "the quality of life is maintained and clinically not inferior to observation," said lead author Dr. Miriam Koopman of the University Medical Center Utrecht Cancer Center, the Netherlands. She presented the final results of the CAIRO3 (Maintenance Treatment With Capecitabine and Bevacizumab Versus Observation After Induction Treatment With Chemotherapy and Bevacizumab as First-line Treatment) trial at the annual meeting of the American Society of Clinical Oncology.

The investigators enrolled 588 patients with metastatic colorectal cancer who had a response of stable-disease or better following six cycles of CAPOX-B to either observation or maintenance with oral capecitabine 625 mg/m2 twice daily and intravenous bevacizumab 7.5 mg/kg on day 1 every 3 weeks. The patients were stratified by prior adjuvant therapy, serum lactate dehydrogenase, response to induction therapy, World Health Organization performance status, and institution.

In both arms, CAPOX-B reintroduction was planned at the time of first progression. The primary endpoint was progression-free survival following reintroduction of CAPOX-B (dubbed PFS2). For those patients who for any reason did not receive CAPOX-B after the first PFS, PFS2 was considered to be equal to PFS1, Dr. Koopman explained.

Of the 279 patients assigned to observation, 168 (60%) had CAPOX-B reintroduced. Of the remaining 111 (40%) of patients in this arm, seven had ongoing observation, 31 received no treatment, and 73 received treatment other than CAPOX-B.

Of the 279 assigned to maintenance, 132 (47%) had CAPOX-B reintroduced. Of the remaining 147 patients (53%) in this arm, 13 continued on maintenance, 45 had no further treatment, 88 had other treatments, and 1 withdrew.

The median PFS from the time of randomization (not including induction) to first progression was 4.1 months in the observation arm and 8.5 months in the maintenance arm (stratified hazard ratio 0.43, P less than .0001).

The benefits of maintenance were also seen with the primary endpoint of PFS2, with a median of 8.5 months for observation, compared with 11.7 months for maintenance (stratified HR 0.67, P less than .0001)

The median time to second progression was 11.1 months among patients randomized to observation, vs. 13. 9 months for those randomized to maintenance (stratified HR 0.68, P less than .0001).

There were no significant differences in median overall survival (OS), however, at 18.1 months for the observation arm and 21.6 months for the maintenance arm.

There was a small but significant difference in quality of life scores between the groups (3.9 point difference on a 100-point scale, P = .004), but this difference was too small to be considered clinically meaningful, Dr. Koopman said.

In preplanned subgroup analyses, the authors found evidence to suggest that patients with synchronous metastases who had resection of the primary tumor had a greater OS benefit from maintenance therapy than did patients with metachronous disease, and that those who had complete or partial response as best response to induction therapy seemed to do better than did patients with stable disease after induction.

"We do not have a clear-cut explanation for this subgroup analysis," Dr. Koopman said.

The investigators hypothesize that the differences between patients with synchronous and metachronous disease could be due to differences in sensitivity to systemic treatment, the prognostic role of resection of the primary tumor, dependence of the angiogenic environment in metastases on the resection status of the primary tumor, or co-option of the local vasculature by the tumor, leading to decreased sensitivity of metachronous metastases to bevacizumab.

The CAIRO3 results suggest that "treatment breaks for all patients at 4 months may be too many breaks, and too early," said Dr. Leonard Saltz, chief of the gastrointestinal oncology service at Memorial Sloan-Kettering Cancer Center, New York.

"Responding patients likely benefit from treatment at least until maximal response, and that’s an aspect to consider in terms of figuring out how to utilize a treatment strategy. And treatment-break strategies will need to be individualized, but should not be abandoned," said Dr. Saltz, who was the invited discussant.

He also cautioned that the quality of life results reported by Dr. Koopman and colleagues may not accurately reflect how patients feel.

 

 

"In terms of the quality of life, what we have to conclude is that as our instruments can measure it we do not see a detriment in quality of life. But intuitively, we can conjecture that being on chemotherapy has some negative aspects – just ask any of our patients. So the idea that there is no detriment in quality of life when we’re comparing chemotherapy to nonchemotherapy suggests that we need more sensitive and specific tools for the question," he said.

The CAIRO3 study was supported by the Dutch Cancer Foundation and by unrestricted scientific grants from Roche and Sanofi-Aventis. Dr. Koopman disclosed ties with Roche/Genentech and Sanofi. Dr. Saltz disclosed ties with Roche, Genentech, and Sanofi.

Publications
Publications
Topics
Article Type
Display Headline
Maintenance improves PFS in patients with metastatic colorectal cancer
Display Headline
Maintenance improves PFS in patients with metastatic colorectal cancer
Legacy Keywords
metastatic colorectal cancer, capecitabine, bevacizumab, Xeloda, oxaliplatin, Eloxatin, Avastin, CAPOX-B regimen,
Legacy Keywords
metastatic colorectal cancer, capecitabine, bevacizumab, Xeloda, oxaliplatin, Eloxatin, Avastin, CAPOX-B regimen,
Article Source

AT THE ASCO ANNUAL MEETING 2014

PURLs Copyright

Inside the Article

Vitals

Key clinical point: Patients with metastatic colorectal cancer who have at least stable disease after induction chemotherapy may benefit from maintenance therapy with capecitabine and bevacizumab, though further studies on quality of life are needed.

Major finding: The median time to second progression (PFS2) for patients with metastatic colorectal cancer following induction and retreatment was a median of 8.5 months for observation, compared with 11.7 months for maintenance with capecitabine and bevacizumab.

Data source: Randomized controlled trial of 588 patients from 64 hospitals in the Netherlands.

Disclosures: The CAIRO3 study was supported by the Dutch Cancer Foundation and by unrestricted scientific grants from Roche and Sanofi-Aventis. Dr. Koopman disclosed ties with Roche/Genentech and Sanofi. Dr. Saltz disclosed ties with Roche, Genentech, and Sanofi.

CAR T-cell therapy successfully used frontline as consolidation in CLL

Article Type
Changed
Fri, 06/13/2014 - 05:00
Display Headline
CAR T-cell therapy successfully used frontline as consolidation in CLL

Poster hall at ASCO 2014

©ASCO/Todd Buchanan

CHICAGO—Infusion of autologous CD19-targeted chimeric antigen receptor (CAR)-modified T cells appears to have promising anti-tumor activity and be well-tolerated as a consolidation to frontline chemotherapy in patients with high-risk chronic lymphocytic leukemia (CLL), researchers report.

In a phase 1 trial, the modified T cells benefitted 43% of patients, including 1 who achieved a complete response and 2 who achieved marrow responses only.

Jae Park, MD, of Memorial Sloan-Kettering Cancer Center in New York, reported the findings at the 2014 ASCO Annual Meeting as abstract 7020.

Dr Park and colleagues enrolled 7 CLL patients who had detectable minimal residual disease after achieving either a partial or complete response following 6 cycles of pentostatin, cyclophosphamide, and rituximab.

Patients then received cyclophosphamide conditioning therapy, followed by 3 escalating doses of CAR T cells in 3 dose cohorts.

Six patients had unmutated IgHV, and 2 patients had del 11q. Four patients had palpable lymphadenopathy, including 1 patient with bulky lymph nodes, prior to T-cell infusion.

After a median follow-up of 11 months, 1 patient achieved a complete response, and 2 patients achieved complete responses in the bone marrow but had progressive disease in the lymph nodes.

Three patients achieved a partial response, and 1 patient had progressive disease, but this patient had rapidly rising absolute lymphocyte count at the time of T-cell infusion, Dr Park noted.

The investigators observed no dose-limiting toxicity. Mild and self-limiting cytokine release syndrome occurred in 3 patients.

“There was a positive correlation between the development of cytokine release syndrome and the CAR T-cell persistence,” Dr Park said.

“Our findings suggest that the CD19-targeted CAR T cells are more effective in eradicating disease in the marrow versus lymph nodes,” he added. “And further studies are being conducted to better understand the mechanism of resistance.”

ASCO discussant Veronika Bachanova, MD, of the University of Minnesota, noted that all previous studies of CAR T-cell therapy in CLL were conducted with relapsed/refractory patients.

“The novelty of this study,” she commented, “is the use of CAR T cells upfront.”

She noted that it can take as long as 8 months to develop the CAR T cells for therapy, adding that “the vector design is of critical importance, since inhibitory signals influence therapy.”

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

Poster hall at ASCO 2014

©ASCO/Todd Buchanan

CHICAGO—Infusion of autologous CD19-targeted chimeric antigen receptor (CAR)-modified T cells appears to have promising anti-tumor activity and be well-tolerated as a consolidation to frontline chemotherapy in patients with high-risk chronic lymphocytic leukemia (CLL), researchers report.

In a phase 1 trial, the modified T cells benefitted 43% of patients, including 1 who achieved a complete response and 2 who achieved marrow responses only.

Jae Park, MD, of Memorial Sloan-Kettering Cancer Center in New York, reported the findings at the 2014 ASCO Annual Meeting as abstract 7020.

Dr Park and colleagues enrolled 7 CLL patients who had detectable minimal residual disease after achieving either a partial or complete response following 6 cycles of pentostatin, cyclophosphamide, and rituximab.

Patients then received cyclophosphamide conditioning therapy, followed by 3 escalating doses of CAR T cells in 3 dose cohorts.

Six patients had unmutated IgHV, and 2 patients had del 11q. Four patients had palpable lymphadenopathy, including 1 patient with bulky lymph nodes, prior to T-cell infusion.

After a median follow-up of 11 months, 1 patient achieved a complete response, and 2 patients achieved complete responses in the bone marrow but had progressive disease in the lymph nodes.

Three patients achieved a partial response, and 1 patient had progressive disease, but this patient had rapidly rising absolute lymphocyte count at the time of T-cell infusion, Dr Park noted.

The investigators observed no dose-limiting toxicity. Mild and self-limiting cytokine release syndrome occurred in 3 patients.

“There was a positive correlation between the development of cytokine release syndrome and the CAR T-cell persistence,” Dr Park said.

“Our findings suggest that the CD19-targeted CAR T cells are more effective in eradicating disease in the marrow versus lymph nodes,” he added. “And further studies are being conducted to better understand the mechanism of resistance.”

ASCO discussant Veronika Bachanova, MD, of the University of Minnesota, noted that all previous studies of CAR T-cell therapy in CLL were conducted with relapsed/refractory patients.

“The novelty of this study,” she commented, “is the use of CAR T cells upfront.”

She noted that it can take as long as 8 months to develop the CAR T cells for therapy, adding that “the vector design is of critical importance, since inhibitory signals influence therapy.”

Poster hall at ASCO 2014

©ASCO/Todd Buchanan

CHICAGO—Infusion of autologous CD19-targeted chimeric antigen receptor (CAR)-modified T cells appears to have promising anti-tumor activity and be well-tolerated as a consolidation to frontline chemotherapy in patients with high-risk chronic lymphocytic leukemia (CLL), researchers report.

In a phase 1 trial, the modified T cells benefitted 43% of patients, including 1 who achieved a complete response and 2 who achieved marrow responses only.

Jae Park, MD, of Memorial Sloan-Kettering Cancer Center in New York, reported the findings at the 2014 ASCO Annual Meeting as abstract 7020.

Dr Park and colleagues enrolled 7 CLL patients who had detectable minimal residual disease after achieving either a partial or complete response following 6 cycles of pentostatin, cyclophosphamide, and rituximab.

Patients then received cyclophosphamide conditioning therapy, followed by 3 escalating doses of CAR T cells in 3 dose cohorts.

Six patients had unmutated IgHV, and 2 patients had del 11q. Four patients had palpable lymphadenopathy, including 1 patient with bulky lymph nodes, prior to T-cell infusion.

After a median follow-up of 11 months, 1 patient achieved a complete response, and 2 patients achieved complete responses in the bone marrow but had progressive disease in the lymph nodes.

Three patients achieved a partial response, and 1 patient had progressive disease, but this patient had rapidly rising absolute lymphocyte count at the time of T-cell infusion, Dr Park noted.

The investigators observed no dose-limiting toxicity. Mild and self-limiting cytokine release syndrome occurred in 3 patients.

“There was a positive correlation between the development of cytokine release syndrome and the CAR T-cell persistence,” Dr Park said.

“Our findings suggest that the CD19-targeted CAR T cells are more effective in eradicating disease in the marrow versus lymph nodes,” he added. “And further studies are being conducted to better understand the mechanism of resistance.”

ASCO discussant Veronika Bachanova, MD, of the University of Minnesota, noted that all previous studies of CAR T-cell therapy in CLL were conducted with relapsed/refractory patients.

“The novelty of this study,” she commented, “is the use of CAR T cells upfront.”

She noted that it can take as long as 8 months to develop the CAR T cells for therapy, adding that “the vector design is of critical importance, since inhibitory signals influence therapy.”

Publications
Publications
Topics
Article Type
Display Headline
CAR T-cell therapy successfully used frontline as consolidation in CLL
Display Headline
CAR T-cell therapy successfully used frontline as consolidation in CLL
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Checkpoint inhibitors produce durable responses in metastatic RCC

Article Type
Changed
Fri, 01/04/2019 - 12:33
Display Headline
Checkpoint inhibitors produce durable responses in metastatic RCC

CHICAGO – A combination of two immune checkpoint inhibitors – ipilimumab and nivolumab – produced durable responses in patients with metastatic renal cell carcinoma in a phase I trial.

At 40.1 weeks of follow-up, the median duration of response for patients treated with nivolumab 3 mg/kg and ipilimumab (Yervoy) 1 mg/kg for four cycles followed by nivolumab maintenance was 31weeks, and for patients treated with nivolumab 1 mg/kg/ and ipilimumab 3 mg/kg the median duration of response had not been reached, reported Dr. Hans J. Hammers of the Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore.

"The objective response rate suggests greater activity than reported previously with nivolumab or ipilimumab monotherapy," Dr. Hammers said at the annual meeting of the American Society of Clinical Oncology.

"Responses appear durable even after discontinuation of study drug," he added.

Nonredundant checkpoints

Both drugs are monoclonal antibodies directed against receptors in immune system checkpoints. Ipilimumab is a cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitor that acts as an early brake point in the immune response; nivolumab is a programmed cell death-1 (PD-1) inhibitor that serves as a late brake. By releasing the brakes, the drugs allow the immune system to operate full throttle against cancer.

Ipilimumab is approved by the Food and Drug Administration for the treatment of metastatic melanoma. Nivolumab is an investigational agent that has been shown to have good antitumor activity in monotherapy against melanoma and other malignancies.

At ASCO 2014, investigators reported that the combinations resulted in "unprecedented" 2-year overall survival rates for patients with metastatic melanoma.

Dr. Hammers and his colleagues investigated the combination in a phase I trial comparing two different combinations of the checkpoint inhibitors.

Patients with untreated or previously treated metastatic renal cell carcinoma (mRCC) with clear-cell histology were randomly assigned to receive intravenous induction therapy every 3 weeks for four cycles with either nivolumab 3 mg/kg and ipilimumab 1 mg/kg (N3/I1) or nivolumab 1 mg/kg and ipilimumab 3 mg/kg (N1/I3). Patients received two infusions during each induction, with nivolumab first, followed by ipilimumab started at least 30 minutes after completion of the nivolumab infusion.

Following induction, all patients went on to continuous nivolumab 3 mg/kg every 2 weeks until disease progression.

Adverse events

Treatment-related adverse events, the primary endpoint, occurred in 16 of the 21 patients (76.2%) assigned to N3/I1) and in all 23 patients assigned to N1/I3. Grade 3 or 4 adverse events occurred in 5 of 21 (23.8%) and 14 of 23 (60.9%), respectively. There were no treatment-related deaths.

The grade 3 or 4 adverse events included diarrhea in one patient in N3/I1 and eight patients in N1/I3, increased lipase in three and six patients, respectively, and increased amylase in one and three patients. There were no other grade 3 or 4 adverse events in either study arm. In addition, there were no high-grade pulmonary adverse events or cases of pneumonitis, which are often seen with immunotherapy, Dr. Hammers noted.

The confirmed objective response rate (ORR) was 43% (9 of 21) in N3/I1 and 48% (11 of 23) in N1/I3. As noted before, the median duration of response was 31.1 weeks in N3/I1 and not reached in N1/I3.

Of the patients who had objective response, the responses were ongoing at last follow-up in 7 of 9 on N3/I1 and 9 of 11 on N1/I3.

There was only one complete response, however, occurring in a patient who received N1/I3. Partial responses occurred in nine patients on N3/I1 and 10 in N1/I3.

The respective progression-free survival rates at 24 weeks were 65% and 64%, which "compares favorably with the nivolumab monotherapy experience," Dr. Hammers said.

The majority of patients in each study arm had significant reductions in tumor burden of the target lesions, he added.

Of the patients with ongoing responses, 3 of 9 in the N3/I1 arm and 5 of 11 in the N1/I3 arm continued to have responses after discontinuing therapy for reasons other than disease progression.

"This encouraging antitumor activity reported with this combination is the basis for a planned phase III combination trial in the first-line setting for the treatment of metastatic renal cell carcinoma patients," Dr. Hammers said.

‘Gutsy move’

Dr. Primo Lara, professor of medicine at the University of California, Davis, who was the invited discussant, commented that, "at least for now, I think we could say that combination checkpoint blockade in RCC is at least additive, recalling that the single-agent response rate in this disease for nivo[lumab] is about 20%-29%, and for ipi[limumab] is about 13%, and some of these responses are encouragingly durable."

 

 

He cautioned, however, that the toxicities with the combined drugs, primarily driven by ipilimumab, "are not inconsequential."

"We heard today that a phase III trial has been initiated, presumably with a lower-dose ipi[limumab] arm, but I think that’s really a gutsy move, considering that there were only 21 patients in that subset of patients that led to this phase III decision. Just a fair warning to everyone that toxicities observed in a phase I trial tend to magnify in a larger phase III when you have more centers, different eligibility criteria, and less experienced folks administering a pretty toxic combination," he said.

The study was funded by Bristol-Myers Squibb and Ono Pharmaceutical. Dr. Hammers has received research funding from BMS. Dr. Lara disclosed serving as a consultant/advisor, and receiving honoraria and research funding from many companies, but not BMS.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
immune checkpoint inhibitor, ipilimumab, nivolumab, metastatic renal cell carcinoma, Yervoy, Dr. Hans J. Hammers, ipilimumab monotherapy,
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

CHICAGO – A combination of two immune checkpoint inhibitors – ipilimumab and nivolumab – produced durable responses in patients with metastatic renal cell carcinoma in a phase I trial.

At 40.1 weeks of follow-up, the median duration of response for patients treated with nivolumab 3 mg/kg and ipilimumab (Yervoy) 1 mg/kg for four cycles followed by nivolumab maintenance was 31weeks, and for patients treated with nivolumab 1 mg/kg/ and ipilimumab 3 mg/kg the median duration of response had not been reached, reported Dr. Hans J. Hammers of the Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore.

"The objective response rate suggests greater activity than reported previously with nivolumab or ipilimumab monotherapy," Dr. Hammers said at the annual meeting of the American Society of Clinical Oncology.

"Responses appear durable even after discontinuation of study drug," he added.

Nonredundant checkpoints

Both drugs are monoclonal antibodies directed against receptors in immune system checkpoints. Ipilimumab is a cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitor that acts as an early brake point in the immune response; nivolumab is a programmed cell death-1 (PD-1) inhibitor that serves as a late brake. By releasing the brakes, the drugs allow the immune system to operate full throttle against cancer.

Ipilimumab is approved by the Food and Drug Administration for the treatment of metastatic melanoma. Nivolumab is an investigational agent that has been shown to have good antitumor activity in monotherapy against melanoma and other malignancies.

At ASCO 2014, investigators reported that the combinations resulted in "unprecedented" 2-year overall survival rates for patients with metastatic melanoma.

Dr. Hammers and his colleagues investigated the combination in a phase I trial comparing two different combinations of the checkpoint inhibitors.

Patients with untreated or previously treated metastatic renal cell carcinoma (mRCC) with clear-cell histology were randomly assigned to receive intravenous induction therapy every 3 weeks for four cycles with either nivolumab 3 mg/kg and ipilimumab 1 mg/kg (N3/I1) or nivolumab 1 mg/kg and ipilimumab 3 mg/kg (N1/I3). Patients received two infusions during each induction, with nivolumab first, followed by ipilimumab started at least 30 minutes after completion of the nivolumab infusion.

Following induction, all patients went on to continuous nivolumab 3 mg/kg every 2 weeks until disease progression.

Adverse events

Treatment-related adverse events, the primary endpoint, occurred in 16 of the 21 patients (76.2%) assigned to N3/I1) and in all 23 patients assigned to N1/I3. Grade 3 or 4 adverse events occurred in 5 of 21 (23.8%) and 14 of 23 (60.9%), respectively. There were no treatment-related deaths.

The grade 3 or 4 adverse events included diarrhea in one patient in N3/I1 and eight patients in N1/I3, increased lipase in three and six patients, respectively, and increased amylase in one and three patients. There were no other grade 3 or 4 adverse events in either study arm. In addition, there were no high-grade pulmonary adverse events or cases of pneumonitis, which are often seen with immunotherapy, Dr. Hammers noted.

The confirmed objective response rate (ORR) was 43% (9 of 21) in N3/I1 and 48% (11 of 23) in N1/I3. As noted before, the median duration of response was 31.1 weeks in N3/I1 and not reached in N1/I3.

Of the patients who had objective response, the responses were ongoing at last follow-up in 7 of 9 on N3/I1 and 9 of 11 on N1/I3.

There was only one complete response, however, occurring in a patient who received N1/I3. Partial responses occurred in nine patients on N3/I1 and 10 in N1/I3.

The respective progression-free survival rates at 24 weeks were 65% and 64%, which "compares favorably with the nivolumab monotherapy experience," Dr. Hammers said.

The majority of patients in each study arm had significant reductions in tumor burden of the target lesions, he added.

Of the patients with ongoing responses, 3 of 9 in the N3/I1 arm and 5 of 11 in the N1/I3 arm continued to have responses after discontinuing therapy for reasons other than disease progression.

"This encouraging antitumor activity reported with this combination is the basis for a planned phase III combination trial in the first-line setting for the treatment of metastatic renal cell carcinoma patients," Dr. Hammers said.

‘Gutsy move’

Dr. Primo Lara, professor of medicine at the University of California, Davis, who was the invited discussant, commented that, "at least for now, I think we could say that combination checkpoint blockade in RCC is at least additive, recalling that the single-agent response rate in this disease for nivo[lumab] is about 20%-29%, and for ipi[limumab] is about 13%, and some of these responses are encouragingly durable."

 

 

He cautioned, however, that the toxicities with the combined drugs, primarily driven by ipilimumab, "are not inconsequential."

"We heard today that a phase III trial has been initiated, presumably with a lower-dose ipi[limumab] arm, but I think that’s really a gutsy move, considering that there were only 21 patients in that subset of patients that led to this phase III decision. Just a fair warning to everyone that toxicities observed in a phase I trial tend to magnify in a larger phase III when you have more centers, different eligibility criteria, and less experienced folks administering a pretty toxic combination," he said.

The study was funded by Bristol-Myers Squibb and Ono Pharmaceutical. Dr. Hammers has received research funding from BMS. Dr. Lara disclosed serving as a consultant/advisor, and receiving honoraria and research funding from many companies, but not BMS.

CHICAGO – A combination of two immune checkpoint inhibitors – ipilimumab and nivolumab – produced durable responses in patients with metastatic renal cell carcinoma in a phase I trial.

At 40.1 weeks of follow-up, the median duration of response for patients treated with nivolumab 3 mg/kg and ipilimumab (Yervoy) 1 mg/kg for four cycles followed by nivolumab maintenance was 31weeks, and for patients treated with nivolumab 1 mg/kg/ and ipilimumab 3 mg/kg the median duration of response had not been reached, reported Dr. Hans J. Hammers of the Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore.

"The objective response rate suggests greater activity than reported previously with nivolumab or ipilimumab monotherapy," Dr. Hammers said at the annual meeting of the American Society of Clinical Oncology.

"Responses appear durable even after discontinuation of study drug," he added.

Nonredundant checkpoints

Both drugs are monoclonal antibodies directed against receptors in immune system checkpoints. Ipilimumab is a cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitor that acts as an early brake point in the immune response; nivolumab is a programmed cell death-1 (PD-1) inhibitor that serves as a late brake. By releasing the brakes, the drugs allow the immune system to operate full throttle against cancer.

Ipilimumab is approved by the Food and Drug Administration for the treatment of metastatic melanoma. Nivolumab is an investigational agent that has been shown to have good antitumor activity in monotherapy against melanoma and other malignancies.

At ASCO 2014, investigators reported that the combinations resulted in "unprecedented" 2-year overall survival rates for patients with metastatic melanoma.

Dr. Hammers and his colleagues investigated the combination in a phase I trial comparing two different combinations of the checkpoint inhibitors.

Patients with untreated or previously treated metastatic renal cell carcinoma (mRCC) with clear-cell histology were randomly assigned to receive intravenous induction therapy every 3 weeks for four cycles with either nivolumab 3 mg/kg and ipilimumab 1 mg/kg (N3/I1) or nivolumab 1 mg/kg and ipilimumab 3 mg/kg (N1/I3). Patients received two infusions during each induction, with nivolumab first, followed by ipilimumab started at least 30 minutes after completion of the nivolumab infusion.

Following induction, all patients went on to continuous nivolumab 3 mg/kg every 2 weeks until disease progression.

Adverse events

Treatment-related adverse events, the primary endpoint, occurred in 16 of the 21 patients (76.2%) assigned to N3/I1) and in all 23 patients assigned to N1/I3. Grade 3 or 4 adverse events occurred in 5 of 21 (23.8%) and 14 of 23 (60.9%), respectively. There were no treatment-related deaths.

The grade 3 or 4 adverse events included diarrhea in one patient in N3/I1 and eight patients in N1/I3, increased lipase in three and six patients, respectively, and increased amylase in one and three patients. There were no other grade 3 or 4 adverse events in either study arm. In addition, there were no high-grade pulmonary adverse events or cases of pneumonitis, which are often seen with immunotherapy, Dr. Hammers noted.

The confirmed objective response rate (ORR) was 43% (9 of 21) in N3/I1 and 48% (11 of 23) in N1/I3. As noted before, the median duration of response was 31.1 weeks in N3/I1 and not reached in N1/I3.

Of the patients who had objective response, the responses were ongoing at last follow-up in 7 of 9 on N3/I1 and 9 of 11 on N1/I3.

There was only one complete response, however, occurring in a patient who received N1/I3. Partial responses occurred in nine patients on N3/I1 and 10 in N1/I3.

The respective progression-free survival rates at 24 weeks were 65% and 64%, which "compares favorably with the nivolumab monotherapy experience," Dr. Hammers said.

The majority of patients in each study arm had significant reductions in tumor burden of the target lesions, he added.

Of the patients with ongoing responses, 3 of 9 in the N3/I1 arm and 5 of 11 in the N1/I3 arm continued to have responses after discontinuing therapy for reasons other than disease progression.

"This encouraging antitumor activity reported with this combination is the basis for a planned phase III combination trial in the first-line setting for the treatment of metastatic renal cell carcinoma patients," Dr. Hammers said.

‘Gutsy move’

Dr. Primo Lara, professor of medicine at the University of California, Davis, who was the invited discussant, commented that, "at least for now, I think we could say that combination checkpoint blockade in RCC is at least additive, recalling that the single-agent response rate in this disease for nivo[lumab] is about 20%-29%, and for ipi[limumab] is about 13%, and some of these responses are encouragingly durable."

 

 

He cautioned, however, that the toxicities with the combined drugs, primarily driven by ipilimumab, "are not inconsequential."

"We heard today that a phase III trial has been initiated, presumably with a lower-dose ipi[limumab] arm, but I think that’s really a gutsy move, considering that there were only 21 patients in that subset of patients that led to this phase III decision. Just a fair warning to everyone that toxicities observed in a phase I trial tend to magnify in a larger phase III when you have more centers, different eligibility criteria, and less experienced folks administering a pretty toxic combination," he said.

The study was funded by Bristol-Myers Squibb and Ono Pharmaceutical. Dr. Hammers has received research funding from BMS. Dr. Lara disclosed serving as a consultant/advisor, and receiving honoraria and research funding from many companies, but not BMS.

Publications
Publications
Topics
Article Type
Display Headline
Checkpoint inhibitors produce durable responses in metastatic RCC
Display Headline
Checkpoint inhibitors produce durable responses in metastatic RCC
Legacy Keywords
immune checkpoint inhibitor, ipilimumab, nivolumab, metastatic renal cell carcinoma, Yervoy, Dr. Hans J. Hammers, ipilimumab monotherapy,
Legacy Keywords
immune checkpoint inhibitor, ipilimumab, nivolumab, metastatic renal cell carcinoma, Yervoy, Dr. Hans J. Hammers, ipilimumab monotherapy,
Article Source

AT THE ASCO ANNUAL MEETING 2014

PURLs Copyright

Inside the Article

Vitals

Major finding: At 40.1 weeks of follow-up, the median duration of response for patients with metastatic renal cell carcinoma treated with nivolumab 3 mg/kg and ipilimumab 1 mg/kg for four cycles followed by nivolumab maintenance was 31 weeks.

Data source: Randomized phase I study comparing two different combinations of nivolumab and ipilimumab in 44 patients with metastatic renal cell carcinoma with a clear cell component.

Disclosures: The study was funded by Bristol-Myers Squibb and Ono Pharmaceutical. Dr. Hammers has received research funding from BMS. Dr. Lara disclosed serving as a consultant/advisor, and receiving honoraria and research funding from many companies, but not BMS.

Ramucirumab briefly prolonged advanced NSCLC survival

Article Type
Changed
Tue, 12/13/2016 - 12:08
Display Headline
Ramucirumab briefly prolonged advanced NSCLC survival

CHICAGO – Adding the recently approved stomach cancer drug, ramucirumab (Cyramza) to docetaxel reduced the risk of death in second-line, non–small cell lung cancer by 14% in the phase III REVEL study.

This is the first therapy in roughly a decade to improve the outcome of lung cancer patients in the second-line setting, according to study author Dr. Maurice Perol, head of thoracic oncology at Cancer Research Center of Lyon, France.

In addition, the recombinant human monoclonal antibody showed benefits in nonsquamous carcinoma as well as the squamous subtype, where treatments are limited.

Still, the median survival gain of just 1.4 months did not impress all at the annual meeting of the American Society of Clinical Oncology, where the study was highlighted in a press briefing before its formal presentation.

To hear Dr. Perol wade through the data and controversy, listen to our interview online.

The study was supported by ImClone, a subsidiary of Eli Lilly, which markets ramucirumab. Dr. Perol reported serving as a consultant or adviser with Lilly, Pfizer, Roche, Boehringer Ingelheim, and Genentech.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
Meeting/Event
Author and Disclosure Information

Publications
Topics
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

CHICAGO – Adding the recently approved stomach cancer drug, ramucirumab (Cyramza) to docetaxel reduced the risk of death in second-line, non–small cell lung cancer by 14% in the phase III REVEL study.

This is the first therapy in roughly a decade to improve the outcome of lung cancer patients in the second-line setting, according to study author Dr. Maurice Perol, head of thoracic oncology at Cancer Research Center of Lyon, France.

In addition, the recombinant human monoclonal antibody showed benefits in nonsquamous carcinoma as well as the squamous subtype, where treatments are limited.

Still, the median survival gain of just 1.4 months did not impress all at the annual meeting of the American Society of Clinical Oncology, where the study was highlighted in a press briefing before its formal presentation.

To hear Dr. Perol wade through the data and controversy, listen to our interview online.

The study was supported by ImClone, a subsidiary of Eli Lilly, which markets ramucirumab. Dr. Perol reported serving as a consultant or adviser with Lilly, Pfizer, Roche, Boehringer Ingelheim, and Genentech.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel

CHICAGO – Adding the recently approved stomach cancer drug, ramucirumab (Cyramza) to docetaxel reduced the risk of death in second-line, non–small cell lung cancer by 14% in the phase III REVEL study.

This is the first therapy in roughly a decade to improve the outcome of lung cancer patients in the second-line setting, according to study author Dr. Maurice Perol, head of thoracic oncology at Cancer Research Center of Lyon, France.

In addition, the recombinant human monoclonal antibody showed benefits in nonsquamous carcinoma as well as the squamous subtype, where treatments are limited.

Still, the median survival gain of just 1.4 months did not impress all at the annual meeting of the American Society of Clinical Oncology, where the study was highlighted in a press briefing before its formal presentation.

To hear Dr. Perol wade through the data and controversy, listen to our interview online.

The study was supported by ImClone, a subsidiary of Eli Lilly, which markets ramucirumab. Dr. Perol reported serving as a consultant or adviser with Lilly, Pfizer, Roche, Boehringer Ingelheim, and Genentech.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
Publications
Publications
Topics
Article Type
Display Headline
Ramucirumab briefly prolonged advanced NSCLC survival
Display Headline
Ramucirumab briefly prolonged advanced NSCLC survival
Article Source

PURLs Copyright

Inside the Article

Lenalidomide combination improves QOL in newly diagnosed MM

Article Type
Changed
Thu, 06/12/2014 - 05:00
Display Headline
Lenalidomide combination improves QOL in newly diagnosed MM

Info booth at ASCO 2014

©ASCO/Scott Morgan

CHICAGO—Substituting lenalidomide for thalidomide in the standard treatment of newly diagnosed multiple myeloma (MM) improves quality of life and

lowers toxicity without significant loss of response, results of a phase 3 study suggest.

Combination melphalan, prednisone, and thalidomide (MPT) is considered a standard treatment option for transplant ineligible, newly diagnosed MM.

Early phase 1/2 studies suggested substituting lenalidomide for thalidomide might result in similar efficacy and less toxicity.

“However, neither feature can be confidently predicted, since long-term follow-up of the melphalan, prednisone, and lenalidomide (MPR) regimen is lacking, and myelosuppression may prove limiting, compromising drug dose and efficacy,” said A. Keith Stewart, MD, from the Mayo Clinic in Scottsdale, Arizona.

So Dr Stewart and his colleagues conducted a randomized, multicenter, phase 3 trial comparing MPT to MPR in untreated, symptomatic, transplant-ineligible MM patients. Dr Stewart presented the results at the 2014 ASCO Annual Meeting as abstract 8511.

The study included 306 patients with a median age of 76 years. The primary objective was to evaluate the  difference in progression-free survival (PFS) between patients receiving MPT and those treated with MPR.

Patients received melphalan at 9 mg/m2 and prednisone at 100 mg orally on days 1-4, with thalidomide at 100 mg daily. Or they received melphalan at 5 mg/m2 and prednisone at 100 mg orally on days 1-4, with lenalidomide at 10 mg orally on days 1-21.

MPT or MPR therapy was continued for twelve 28-day cycles, followed by thalidomide at 100 mg or lenalidomide at 10 mg daily until relapse. Patients were required to have aspirin prophylaxis.

Secondary study objectives included overall survival, toxicities, response rates, depth of response, and quality of life change. Treatment arms were balanced for age, ISS stage, and other major prognostic factors.

The median follow-up was 40.7 months. The median time on therapy was 12 months overall and 23 months for the 46% of patients on maintenance therapy, with no differences by arm. Some 7% of patients remained on treatment through cycle 60.

The results showed similar response rates between the 2 arms. The partial response rate was 64% for the MPT group, compared with 60% for the MPR group, with no difference in very good partial response/complete response rates (18.8% vs 23%).

The median PFS was 21 months for patients receiving MPT and 18.7 months for those receiving MPR. The median overall survival was not significantly different between the arms—52.6% for the MPT arm and 47.7% for the MPR arm.

Toxicities of grade 3 or higher were significantly more likely in the MPT arm (73%) than in the MPR arm (58%), as was non-hematologic toxicity (59% and 40%, respectively).

The incidence of second primary malignancies was higher with MPT (3.47/100 person years) than with MPR (2.01/100 person years). And deep vein thrombosis or pulmonary embolism occurred in 8.8% of MPT patients and 6.7% MPR patients.

“The quality of life analysis favored MPR by induction end,” Dr Stewart said. “Response rates, PFS, and overall survival were similar between the 2 arms. However, there was significantly better quality of life at 12 months and lower toxicity with MPR.”

Dr Stewart noted, however, that in the US, “melphalan-based regimens are now seldom utilized due to availability of lenalidomide and bortezomib in newly diagnosed patients.”

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

Info booth at ASCO 2014

©ASCO/Scott Morgan

CHICAGO—Substituting lenalidomide for thalidomide in the standard treatment of newly diagnosed multiple myeloma (MM) improves quality of life and

lowers toxicity without significant loss of response, results of a phase 3 study suggest.

Combination melphalan, prednisone, and thalidomide (MPT) is considered a standard treatment option for transplant ineligible, newly diagnosed MM.

Early phase 1/2 studies suggested substituting lenalidomide for thalidomide might result in similar efficacy and less toxicity.

“However, neither feature can be confidently predicted, since long-term follow-up of the melphalan, prednisone, and lenalidomide (MPR) regimen is lacking, and myelosuppression may prove limiting, compromising drug dose and efficacy,” said A. Keith Stewart, MD, from the Mayo Clinic in Scottsdale, Arizona.

So Dr Stewart and his colleagues conducted a randomized, multicenter, phase 3 trial comparing MPT to MPR in untreated, symptomatic, transplant-ineligible MM patients. Dr Stewart presented the results at the 2014 ASCO Annual Meeting as abstract 8511.

The study included 306 patients with a median age of 76 years. The primary objective was to evaluate the  difference in progression-free survival (PFS) between patients receiving MPT and those treated with MPR.

Patients received melphalan at 9 mg/m2 and prednisone at 100 mg orally on days 1-4, with thalidomide at 100 mg daily. Or they received melphalan at 5 mg/m2 and prednisone at 100 mg orally on days 1-4, with lenalidomide at 10 mg orally on days 1-21.

MPT or MPR therapy was continued for twelve 28-day cycles, followed by thalidomide at 100 mg or lenalidomide at 10 mg daily until relapse. Patients were required to have aspirin prophylaxis.

Secondary study objectives included overall survival, toxicities, response rates, depth of response, and quality of life change. Treatment arms were balanced for age, ISS stage, and other major prognostic factors.

The median follow-up was 40.7 months. The median time on therapy was 12 months overall and 23 months for the 46% of patients on maintenance therapy, with no differences by arm. Some 7% of patients remained on treatment through cycle 60.

The results showed similar response rates between the 2 arms. The partial response rate was 64% for the MPT group, compared with 60% for the MPR group, with no difference in very good partial response/complete response rates (18.8% vs 23%).

The median PFS was 21 months for patients receiving MPT and 18.7 months for those receiving MPR. The median overall survival was not significantly different between the arms—52.6% for the MPT arm and 47.7% for the MPR arm.

Toxicities of grade 3 or higher were significantly more likely in the MPT arm (73%) than in the MPR arm (58%), as was non-hematologic toxicity (59% and 40%, respectively).

The incidence of second primary malignancies was higher with MPT (3.47/100 person years) than with MPR (2.01/100 person years). And deep vein thrombosis or pulmonary embolism occurred in 8.8% of MPT patients and 6.7% MPR patients.

“The quality of life analysis favored MPR by induction end,” Dr Stewart said. “Response rates, PFS, and overall survival were similar between the 2 arms. However, there was significantly better quality of life at 12 months and lower toxicity with MPR.”

Dr Stewart noted, however, that in the US, “melphalan-based regimens are now seldom utilized due to availability of lenalidomide and bortezomib in newly diagnosed patients.”

Info booth at ASCO 2014

©ASCO/Scott Morgan

CHICAGO—Substituting lenalidomide for thalidomide in the standard treatment of newly diagnosed multiple myeloma (MM) improves quality of life and

lowers toxicity without significant loss of response, results of a phase 3 study suggest.

Combination melphalan, prednisone, and thalidomide (MPT) is considered a standard treatment option for transplant ineligible, newly diagnosed MM.

Early phase 1/2 studies suggested substituting lenalidomide for thalidomide might result in similar efficacy and less toxicity.

“However, neither feature can be confidently predicted, since long-term follow-up of the melphalan, prednisone, and lenalidomide (MPR) regimen is lacking, and myelosuppression may prove limiting, compromising drug dose and efficacy,” said A. Keith Stewart, MD, from the Mayo Clinic in Scottsdale, Arizona.

So Dr Stewart and his colleagues conducted a randomized, multicenter, phase 3 trial comparing MPT to MPR in untreated, symptomatic, transplant-ineligible MM patients. Dr Stewart presented the results at the 2014 ASCO Annual Meeting as abstract 8511.

The study included 306 patients with a median age of 76 years. The primary objective was to evaluate the  difference in progression-free survival (PFS) between patients receiving MPT and those treated with MPR.

Patients received melphalan at 9 mg/m2 and prednisone at 100 mg orally on days 1-4, with thalidomide at 100 mg daily. Or they received melphalan at 5 mg/m2 and prednisone at 100 mg orally on days 1-4, with lenalidomide at 10 mg orally on days 1-21.

MPT or MPR therapy was continued for twelve 28-day cycles, followed by thalidomide at 100 mg or lenalidomide at 10 mg daily until relapse. Patients were required to have aspirin prophylaxis.

Secondary study objectives included overall survival, toxicities, response rates, depth of response, and quality of life change. Treatment arms were balanced for age, ISS stage, and other major prognostic factors.

The median follow-up was 40.7 months. The median time on therapy was 12 months overall and 23 months for the 46% of patients on maintenance therapy, with no differences by arm. Some 7% of patients remained on treatment through cycle 60.

The results showed similar response rates between the 2 arms. The partial response rate was 64% for the MPT group, compared with 60% for the MPR group, with no difference in very good partial response/complete response rates (18.8% vs 23%).

The median PFS was 21 months for patients receiving MPT and 18.7 months for those receiving MPR. The median overall survival was not significantly different between the arms—52.6% for the MPT arm and 47.7% for the MPR arm.

Toxicities of grade 3 or higher were significantly more likely in the MPT arm (73%) than in the MPR arm (58%), as was non-hematologic toxicity (59% and 40%, respectively).

The incidence of second primary malignancies was higher with MPT (3.47/100 person years) than with MPR (2.01/100 person years). And deep vein thrombosis or pulmonary embolism occurred in 8.8% of MPT patients and 6.7% MPR patients.

“The quality of life analysis favored MPR by induction end,” Dr Stewart said. “Response rates, PFS, and overall survival were similar between the 2 arms. However, there was significantly better quality of life at 12 months and lower toxicity with MPR.”

Dr Stewart noted, however, that in the US, “melphalan-based regimens are now seldom utilized due to availability of lenalidomide and bortezomib in newly diagnosed patients.”

Publications
Publications
Topics
Article Type
Display Headline
Lenalidomide combination improves QOL in newly diagnosed MM
Display Headline
Lenalidomide combination improves QOL in newly diagnosed MM
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

VIDEO: Metastatic cervical cancer yields to immunotherapy

Article Type
Changed
Fri, 01/04/2019 - 12:33
Display Headline
VIDEO: Metastatic cervical cancer yields to immunotherapy

CHICAGO – For the first time, researchers have shown that immunotherapy using adoptive T-cell therapy can induce complete remission in metastatic cervical cancer.

The study involved just nine patients given a single infusion of human papillomavirus (HPV)-targeted T cell therapy, but is sparking enormous excitement.

To learn more, watch our interview at the annual meeting of the American Society of Clinical Oncology with lead study author Dr. Christian Hinrichs of the National Cancer Institute.

The study was supported by the National Cancer Institute, National Institutes of Health. The authors reported no financial disclosures.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel

pwendling@frontlinemedcom.com

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
adoptive T-cell therapy, metastatic cervical cancer, Dr. Christian Hinrichs,
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

CHICAGO – For the first time, researchers have shown that immunotherapy using adoptive T-cell therapy can induce complete remission in metastatic cervical cancer.

The study involved just nine patients given a single infusion of human papillomavirus (HPV)-targeted T cell therapy, but is sparking enormous excitement.

To learn more, watch our interview at the annual meeting of the American Society of Clinical Oncology with lead study author Dr. Christian Hinrichs of the National Cancer Institute.

The study was supported by the National Cancer Institute, National Institutes of Health. The authors reported no financial disclosures.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel

pwendling@frontlinemedcom.com

CHICAGO – For the first time, researchers have shown that immunotherapy using adoptive T-cell therapy can induce complete remission in metastatic cervical cancer.

The study involved just nine patients given a single infusion of human papillomavirus (HPV)-targeted T cell therapy, but is sparking enormous excitement.

To learn more, watch our interview at the annual meeting of the American Society of Clinical Oncology with lead study author Dr. Christian Hinrichs of the National Cancer Institute.

The study was supported by the National Cancer Institute, National Institutes of Health. The authors reported no financial disclosures.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel

pwendling@frontlinemedcom.com

Publications
Publications
Topics
Article Type
Display Headline
VIDEO: Metastatic cervical cancer yields to immunotherapy
Display Headline
VIDEO: Metastatic cervical cancer yields to immunotherapy
Legacy Keywords
adoptive T-cell therapy, metastatic cervical cancer, Dr. Christian Hinrichs,
Legacy Keywords
adoptive T-cell therapy, metastatic cervical cancer, Dr. Christian Hinrichs,
Article Source

AT THE ASCO ANNUAL MEETING 2014

PURLs Copyright

Inside the Article

VIDEO: Immunotherapy as treatment for cervical cancer

Article Type
Changed
Fri, 01/04/2019 - 12:33
Display Headline
VIDEO: Immunotherapy as treatment for cervical cancer

CHICAGO – Research demonstrating that immunotherapy can be harnessed to treat metastatic cervical cancer was one of the highlights of the annual meeting of the American Society of Clinical Oncology.

We spoke with gynecologic cancer expert Dr. Don Dizon to learn more about why human papillomavirus-targeted T-cell therapy could work in cervical cancer and if this same cellular-based approach could be used in other virally mediated tumors.

Dr. Dizon is director of the Oncology Sexual Health Clinic at Massachusetts General Hospital Cancer Center in Boston.

He reported having no financial disclosures.

pwendling@frontlinemedcom.com

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
immunotherapy, metastatic cervical cancer, American Society of Clinical Oncology, gynecologic cancer, Dr. Don Dizon, human papillomavirus, T-cell therapy, cervical cancer,
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event
Related Articles

CHICAGO – Research demonstrating that immunotherapy can be harnessed to treat metastatic cervical cancer was one of the highlights of the annual meeting of the American Society of Clinical Oncology.

We spoke with gynecologic cancer expert Dr. Don Dizon to learn more about why human papillomavirus-targeted T-cell therapy could work in cervical cancer and if this same cellular-based approach could be used in other virally mediated tumors.

Dr. Dizon is director of the Oncology Sexual Health Clinic at Massachusetts General Hospital Cancer Center in Boston.

He reported having no financial disclosures.

pwendling@frontlinemedcom.com

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel

CHICAGO – Research demonstrating that immunotherapy can be harnessed to treat metastatic cervical cancer was one of the highlights of the annual meeting of the American Society of Clinical Oncology.

We spoke with gynecologic cancer expert Dr. Don Dizon to learn more about why human papillomavirus-targeted T-cell therapy could work in cervical cancer and if this same cellular-based approach could be used in other virally mediated tumors.

Dr. Dizon is director of the Oncology Sexual Health Clinic at Massachusetts General Hospital Cancer Center in Boston.

He reported having no financial disclosures.

pwendling@frontlinemedcom.com

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
Publications
Publications
Topics
Article Type
Display Headline
VIDEO: Immunotherapy as treatment for cervical cancer
Display Headline
VIDEO: Immunotherapy as treatment for cervical cancer
Legacy Keywords
immunotherapy, metastatic cervical cancer, American Society of Clinical Oncology, gynecologic cancer, Dr. Don Dizon, human papillomavirus, T-cell therapy, cervical cancer,
Legacy Keywords
immunotherapy, metastatic cervical cancer, American Society of Clinical Oncology, gynecologic cancer, Dr. Don Dizon, human papillomavirus, T-cell therapy, cervical cancer,
Article Source

AT ASCO ANNUAL MEETING 2014

PURLs Copyright

Inside the Article

Novel agent shows promising activity in heavily pretreated NHL

Article Type
Changed
Tue, 01/17/2023 - 11:18
Display Headline
Novel agent shows promising activity in heavily pretreated NHL

 

 

 

Lobby view at ASCO 2014

©ASCO/Rodney White

 

CHICAGO—The novel, oral selective inhibitor of nuclear transport known as selinexor (KPT-330) can safely be given as monotherapy to patients with heavily pretreated non-Hodgkin lymphoma (NHL), according to a presentation at the 2014 ASCO Annual Meeting.

 

“Selinexor has favorable pharmacokinetic and pharmacodynamic characteristics,” said presenter Martin Gutierrez, MD, of the John Theurer Cancer Center at Hackensack University Medical Center in New Jersey.

 

Selinexor is a slowly reversible, selective inhibitor of nuclear transport that inhibits XPO1, which is elevated in NHL, chronic lymphocytic leukemia (CLL), and other malignances.

 

“It shows single-agent anti-tumor activity across all NHL types, with durable cancer control of more than 9 months [and] marked activity across germ cell B (GCB), non-germ cell B, and double-hit diffuse large B-cell lymphoma (DLBCL),” Dr Gutierrez said.

 

He provided an update of the ongoing phase 1 dose-escalation study at the meeting as abstract 8518.

 

The study now includes 51 patients, half of whom have NHL. Their median age is 60 years.

 

The patients received selinexor across 8 dose levels, ranging from 3 mg/m2 to 60 mg/m2. Dosing at 60 mg/m2 twice weekly is ongoing, and the maximum-tolerated dose has not been reached.

 

Among the 43 evaluable patients, the disease control rate was 74%, the overall response rate was 28%, and the complete response rate was 5%.

 

“All patients who had their disease controlled had a reduction in lymph nodes and some degree of activity across all dose levels,” Dr Gutierrez said. “GCB and non-GCB patients responded similarly.”

 

The length of response was up to 632 days in the DLBCL group.

 

Most adverse events were gastrointestinal in nature, and most of them were grade 1 or 2. The most common adverse events were nausea, anorexia, and fatigue.

 

The investigators found a higher incidence of side effects in the first treatment cycle. The dosing schedule was interrupted and reduced to maintain steady state levels.

 

“The results suggest that an intermittent dosing schedule optimally induces a steady state with maximal induction of XPO1 mRNA,” Dr Gutierrez said.

 

There were 3 dose-limiting toxicities, including 1 multiple myeloma patient with grade 4 thrombocytopenia, 1 follicular lymphoma patient with grade 4 thrombocytopenia, and 1 CLL patient with grade 2 fatigue.

 

ASCO discussant Owen O’Connor, MD, from Columbia University Medical Center in New York, commented, “These clinical data are interesting with a provocative target. I applaud the investigators for doing a trial across a diversity of B- and T-cell lymphomas . . . . The results suggest a potential effect in a rare subset of lymphoma patients that have little treatment options.”

 

Frontline trials of selinexor are planned, including patients with Richter transformation and follicular lymphoma.

 

Selinexor recently received orphan drug status from the US Food and Drug Administration for the treatment of DLBCL.

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

 

 

 

Lobby view at ASCO 2014

©ASCO/Rodney White

 

CHICAGO—The novel, oral selective inhibitor of nuclear transport known as selinexor (KPT-330) can safely be given as monotherapy to patients with heavily pretreated non-Hodgkin lymphoma (NHL), according to a presentation at the 2014 ASCO Annual Meeting.

 

“Selinexor has favorable pharmacokinetic and pharmacodynamic characteristics,” said presenter Martin Gutierrez, MD, of the John Theurer Cancer Center at Hackensack University Medical Center in New Jersey.

 

Selinexor is a slowly reversible, selective inhibitor of nuclear transport that inhibits XPO1, which is elevated in NHL, chronic lymphocytic leukemia (CLL), and other malignances.

 

“It shows single-agent anti-tumor activity across all NHL types, with durable cancer control of more than 9 months [and] marked activity across germ cell B (GCB), non-germ cell B, and double-hit diffuse large B-cell lymphoma (DLBCL),” Dr Gutierrez said.

 

He provided an update of the ongoing phase 1 dose-escalation study at the meeting as abstract 8518.

 

The study now includes 51 patients, half of whom have NHL. Their median age is 60 years.

 

The patients received selinexor across 8 dose levels, ranging from 3 mg/m2 to 60 mg/m2. Dosing at 60 mg/m2 twice weekly is ongoing, and the maximum-tolerated dose has not been reached.

 

Among the 43 evaluable patients, the disease control rate was 74%, the overall response rate was 28%, and the complete response rate was 5%.

 

“All patients who had their disease controlled had a reduction in lymph nodes and some degree of activity across all dose levels,” Dr Gutierrez said. “GCB and non-GCB patients responded similarly.”

 

The length of response was up to 632 days in the DLBCL group.

 

Most adverse events were gastrointestinal in nature, and most of them were grade 1 or 2. The most common adverse events were nausea, anorexia, and fatigue.

 

The investigators found a higher incidence of side effects in the first treatment cycle. The dosing schedule was interrupted and reduced to maintain steady state levels.

 

“The results suggest that an intermittent dosing schedule optimally induces a steady state with maximal induction of XPO1 mRNA,” Dr Gutierrez said.

 

There were 3 dose-limiting toxicities, including 1 multiple myeloma patient with grade 4 thrombocytopenia, 1 follicular lymphoma patient with grade 4 thrombocytopenia, and 1 CLL patient with grade 2 fatigue.

 

ASCO discussant Owen O’Connor, MD, from Columbia University Medical Center in New York, commented, “These clinical data are interesting with a provocative target. I applaud the investigators for doing a trial across a diversity of B- and T-cell lymphomas . . . . The results suggest a potential effect in a rare subset of lymphoma patients that have little treatment options.”

 

Frontline trials of selinexor are planned, including patients with Richter transformation and follicular lymphoma.

 

Selinexor recently received orphan drug status from the US Food and Drug Administration for the treatment of DLBCL.

 

 

 

Lobby view at ASCO 2014

©ASCO/Rodney White

 

CHICAGO—The novel, oral selective inhibitor of nuclear transport known as selinexor (KPT-330) can safely be given as monotherapy to patients with heavily pretreated non-Hodgkin lymphoma (NHL), according to a presentation at the 2014 ASCO Annual Meeting.

 

“Selinexor has favorable pharmacokinetic and pharmacodynamic characteristics,” said presenter Martin Gutierrez, MD, of the John Theurer Cancer Center at Hackensack University Medical Center in New Jersey.

 

Selinexor is a slowly reversible, selective inhibitor of nuclear transport that inhibits XPO1, which is elevated in NHL, chronic lymphocytic leukemia (CLL), and other malignances.

 

“It shows single-agent anti-tumor activity across all NHL types, with durable cancer control of more than 9 months [and] marked activity across germ cell B (GCB), non-germ cell B, and double-hit diffuse large B-cell lymphoma (DLBCL),” Dr Gutierrez said.

 

He provided an update of the ongoing phase 1 dose-escalation study at the meeting as abstract 8518.

 

The study now includes 51 patients, half of whom have NHL. Their median age is 60 years.

 

The patients received selinexor across 8 dose levels, ranging from 3 mg/m2 to 60 mg/m2. Dosing at 60 mg/m2 twice weekly is ongoing, and the maximum-tolerated dose has not been reached.

 

Among the 43 evaluable patients, the disease control rate was 74%, the overall response rate was 28%, and the complete response rate was 5%.

 

“All patients who had their disease controlled had a reduction in lymph nodes and some degree of activity across all dose levels,” Dr Gutierrez said. “GCB and non-GCB patients responded similarly.”

 

The length of response was up to 632 days in the DLBCL group.

 

Most adverse events were gastrointestinal in nature, and most of them were grade 1 or 2. The most common adverse events were nausea, anorexia, and fatigue.

 

The investigators found a higher incidence of side effects in the first treatment cycle. The dosing schedule was interrupted and reduced to maintain steady state levels.

 

“The results suggest that an intermittent dosing schedule optimally induces a steady state with maximal induction of XPO1 mRNA,” Dr Gutierrez said.

 

There were 3 dose-limiting toxicities, including 1 multiple myeloma patient with grade 4 thrombocytopenia, 1 follicular lymphoma patient with grade 4 thrombocytopenia, and 1 CLL patient with grade 2 fatigue.

 

ASCO discussant Owen O’Connor, MD, from Columbia University Medical Center in New York, commented, “These clinical data are interesting with a provocative target. I applaud the investigators for doing a trial across a diversity of B- and T-cell lymphomas . . . . The results suggest a potential effect in a rare subset of lymphoma patients that have little treatment options.”

 

Frontline trials of selinexor are planned, including patients with Richter transformation and follicular lymphoma.

 

Selinexor recently received orphan drug status from the US Food and Drug Administration for the treatment of DLBCL.

Publications
Publications
Topics
Article Type
Display Headline
Novel agent shows promising activity in heavily pretreated NHL
Display Headline
Novel agent shows promising activity in heavily pretreated NHL
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica