Chemo-free regimen appears viable in previously untreated FL

Article Type
Changed
Tue, 01/17/2023 - 11:16

 

– Lenalidomide plus rituximab (R2) had comparable efficacy versus standard chemoimmunotherapy in patients with previously untreated follicular lymphoma, according to results from a phase 3 trial.

RELEVANCE is the first randomized, phase 3 trial to examine a chemotherapy-free regimen in this setting.

Response and progression-free survival (PFS) results were similar for patients who received R2 followed by rituximab maintenance and patients assigned to chemotherapy plus rituximab and rituximab maintenance, in study results presented at the annual meeting of the American Society of Clinical Oncology.

“These results show that lenalidomide plus rituximab, which is a novel immunomodulatory approach, is a potential first-line option for patients with follicular lymphoma that require treatment,” said investigator Nathan H. Fowler, MD, of the University of Texas MD Anderson Cancer Center, Houston.

But since the study was designed as a superiority trial, rather than a noninferiority trial, and it failed to meet its primary endpoint of superior complete remission (CR) or CR unconfirmed (CRu) at 120 weeks, said Bruce D. Cheson, MD, head of hematology at Georgetown University, Washington.

R2 had a similar PFS overall and in all major patient subgroups, similar overall survival, less nonhematologic toxicity aside from rash, less neutropenia, and fewer infections despite increased use of growth factors in the chemoimmunotherapy arm, Dr. Cheson said in a presentation commenting on the results. “Therefore, I agree with Dr. Fowler’s conclusion that R2 can be considered as an option for the front-line therapy of patients with follicular lymphoma,” Dr. Cheson said.

The RELEVANCE study included 1,030 patients (median age, 59 years) with previously untreated, advanced follicular lymphoma requiring treatment. They were randomized 1:1 to either lenalidomide plus rituximab followed by rituximab maintenance, or R-chemotherapy followed by rituximab maintenance.

 

 


For patients randomly assigned to R-chemotherapy, physicians could choose among three standard regimens: rituximab plus bendamustine (R-B), rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP), or rituximab plus cyclophosphamide, vincristine, and prednisone (R-CVP).

There was no statistical difference between treatment approaches in CR/CRu at 120 weeks, which was 48% in the R2 arm and 53% in the R-chemotherapy arm (P = 0.13). Best CR/CRu also was not statistically different between arms (59% and 67%, respectively), as was best overall response rate (84% and 89%). The 3-year duration of response was 77% in the R2 arm and 74% for R-chemotherapy.

With 37.9 months median follow-up, progression-free survival was “nearly identical” between the two groups, Dr. Fowler said, at 77% for R2 and 78% for R-chemotherapy (P = 0.48). The 3-year overall survival was 94% in both the R2 and R-chemotherapy arms, though survival data are still immature, Dr. Fowler noted.

Grade 3/4 neutropenia was more common in the R-chemotherapy arm, resulting in higher rates of febrile neutropenia, according to Dr. Fowler, who also noted that rash and cutaneous reactions were more common with R2. About 70% of patients in each arm were able to tolerate treatment, and reasons for discontinuation were “fairly similar” between arms, Dr. Fowler added.
 

 


Second primary malignancies occurred in 7% of patients in the R2 arm and 10% of the R-chemotherapy arm.

The study was sponsored was Celgene and the Lymphoma Academic Research Organisation. Dr. Fowler reported disclosures related to Abbvie, Celgene, Janssen, Merck, and Roche.

SOURCE: Fowler NH et al. ASCO 2018, Abstract 7500.

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

 

– Lenalidomide plus rituximab (R2) had comparable efficacy versus standard chemoimmunotherapy in patients with previously untreated follicular lymphoma, according to results from a phase 3 trial.

RELEVANCE is the first randomized, phase 3 trial to examine a chemotherapy-free regimen in this setting.

Response and progression-free survival (PFS) results were similar for patients who received R2 followed by rituximab maintenance and patients assigned to chemotherapy plus rituximab and rituximab maintenance, in study results presented at the annual meeting of the American Society of Clinical Oncology.

“These results show that lenalidomide plus rituximab, which is a novel immunomodulatory approach, is a potential first-line option for patients with follicular lymphoma that require treatment,” said investigator Nathan H. Fowler, MD, of the University of Texas MD Anderson Cancer Center, Houston.

But since the study was designed as a superiority trial, rather than a noninferiority trial, and it failed to meet its primary endpoint of superior complete remission (CR) or CR unconfirmed (CRu) at 120 weeks, said Bruce D. Cheson, MD, head of hematology at Georgetown University, Washington.

R2 had a similar PFS overall and in all major patient subgroups, similar overall survival, less nonhematologic toxicity aside from rash, less neutropenia, and fewer infections despite increased use of growth factors in the chemoimmunotherapy arm, Dr. Cheson said in a presentation commenting on the results. “Therefore, I agree with Dr. Fowler’s conclusion that R2 can be considered as an option for the front-line therapy of patients with follicular lymphoma,” Dr. Cheson said.

The RELEVANCE study included 1,030 patients (median age, 59 years) with previously untreated, advanced follicular lymphoma requiring treatment. They were randomized 1:1 to either lenalidomide plus rituximab followed by rituximab maintenance, or R-chemotherapy followed by rituximab maintenance.

 

 


For patients randomly assigned to R-chemotherapy, physicians could choose among three standard regimens: rituximab plus bendamustine (R-B), rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP), or rituximab plus cyclophosphamide, vincristine, and prednisone (R-CVP).

There was no statistical difference between treatment approaches in CR/CRu at 120 weeks, which was 48% in the R2 arm and 53% in the R-chemotherapy arm (P = 0.13). Best CR/CRu also was not statistically different between arms (59% and 67%, respectively), as was best overall response rate (84% and 89%). The 3-year duration of response was 77% in the R2 arm and 74% for R-chemotherapy.

With 37.9 months median follow-up, progression-free survival was “nearly identical” between the two groups, Dr. Fowler said, at 77% for R2 and 78% for R-chemotherapy (P = 0.48). The 3-year overall survival was 94% in both the R2 and R-chemotherapy arms, though survival data are still immature, Dr. Fowler noted.

Grade 3/4 neutropenia was more common in the R-chemotherapy arm, resulting in higher rates of febrile neutropenia, according to Dr. Fowler, who also noted that rash and cutaneous reactions were more common with R2. About 70% of patients in each arm were able to tolerate treatment, and reasons for discontinuation were “fairly similar” between arms, Dr. Fowler added.
 

 


Second primary malignancies occurred in 7% of patients in the R2 arm and 10% of the R-chemotherapy arm.

The study was sponsored was Celgene and the Lymphoma Academic Research Organisation. Dr. Fowler reported disclosures related to Abbvie, Celgene, Janssen, Merck, and Roche.

SOURCE: Fowler NH et al. ASCO 2018, Abstract 7500.

 

– Lenalidomide plus rituximab (R2) had comparable efficacy versus standard chemoimmunotherapy in patients with previously untreated follicular lymphoma, according to results from a phase 3 trial.

RELEVANCE is the first randomized, phase 3 trial to examine a chemotherapy-free regimen in this setting.

Response and progression-free survival (PFS) results were similar for patients who received R2 followed by rituximab maintenance and patients assigned to chemotherapy plus rituximab and rituximab maintenance, in study results presented at the annual meeting of the American Society of Clinical Oncology.

“These results show that lenalidomide plus rituximab, which is a novel immunomodulatory approach, is a potential first-line option for patients with follicular lymphoma that require treatment,” said investigator Nathan H. Fowler, MD, of the University of Texas MD Anderson Cancer Center, Houston.

But since the study was designed as a superiority trial, rather than a noninferiority trial, and it failed to meet its primary endpoint of superior complete remission (CR) or CR unconfirmed (CRu) at 120 weeks, said Bruce D. Cheson, MD, head of hematology at Georgetown University, Washington.

R2 had a similar PFS overall and in all major patient subgroups, similar overall survival, less nonhematologic toxicity aside from rash, less neutropenia, and fewer infections despite increased use of growth factors in the chemoimmunotherapy arm, Dr. Cheson said in a presentation commenting on the results. “Therefore, I agree with Dr. Fowler’s conclusion that R2 can be considered as an option for the front-line therapy of patients with follicular lymphoma,” Dr. Cheson said.

The RELEVANCE study included 1,030 patients (median age, 59 years) with previously untreated, advanced follicular lymphoma requiring treatment. They were randomized 1:1 to either lenalidomide plus rituximab followed by rituximab maintenance, or R-chemotherapy followed by rituximab maintenance.

 

 


For patients randomly assigned to R-chemotherapy, physicians could choose among three standard regimens: rituximab plus bendamustine (R-B), rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP), or rituximab plus cyclophosphamide, vincristine, and prednisone (R-CVP).

There was no statistical difference between treatment approaches in CR/CRu at 120 weeks, which was 48% in the R2 arm and 53% in the R-chemotherapy arm (P = 0.13). Best CR/CRu also was not statistically different between arms (59% and 67%, respectively), as was best overall response rate (84% and 89%). The 3-year duration of response was 77% in the R2 arm and 74% for R-chemotherapy.

With 37.9 months median follow-up, progression-free survival was “nearly identical” between the two groups, Dr. Fowler said, at 77% for R2 and 78% for R-chemotherapy (P = 0.48). The 3-year overall survival was 94% in both the R2 and R-chemotherapy arms, though survival data are still immature, Dr. Fowler noted.

Grade 3/4 neutropenia was more common in the R-chemotherapy arm, resulting in higher rates of febrile neutropenia, according to Dr. Fowler, who also noted that rash and cutaneous reactions were more common with R2. About 70% of patients in each arm were able to tolerate treatment, and reasons for discontinuation were “fairly similar” between arms, Dr. Fowler added.
 

 


Second primary malignancies occurred in 7% of patients in the R2 arm and 10% of the R-chemotherapy arm.

The study was sponsored was Celgene and the Lymphoma Academic Research Organisation. Dr. Fowler reported disclosures related to Abbvie, Celgene, Janssen, Merck, and Roche.

SOURCE: Fowler NH et al. ASCO 2018, Abstract 7500.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASCO 2018

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: Lenalidomide plus rituximab (R2) had comparable efficacy versus standard chemoimmunotherapy in patients with previously untreated follicular lymphoma.

Major finding: With 37.9 months’ median follow-up, progression-free survival was “nearly identical” between the two groups, at 77% for R2 and 78% for rituximab chemotherapy (P = 0.48).

Study details: RELEVANCE, a phase 3, randomized clinical trial including 1,030 patients with previously untreated, advanced follicular lymphoma requiring treatment.

Disclosures: The study was sponsored was Celgene and the Lymphoma Academic Research Organisation. Dr. Fowler reported disclosures related to AbbVie, Celgene, Janssen, Merck, and Roche.

Source: Fowler NH et al. ASCO 2018, Abstract 7500.

Disqus Comments
Default
Use ProPublica

Two agents could take AML therapy in new directions

Article Type
Changed
Thu, 06/07/2018 - 00:10
Display Headline
Two agents could take AML therapy in new directions

©ASCO/Rodney White 2018
Poster session at ASCO 2018

CHICAGO—Two agents targeting novel pathways in myeloid malignancies—mivebresib and bencentinib—are showing promise in early studies, according to a speaker at the 2018 ASCO Annual Meeting.

“Both BET and AXL inhibition appear to be new and exciting targets in myeloid malignancies,” said Alice S. Mims, MD, and both have produced responses as single agents.

Dr Mims, of Ohio State University Wexner Medical Center in Columbus, made these observations in a poster discussion presentation that included commentary on the two agents.

Mivebresib (ABBV-075), an inhibitor of bromodomain and extra terminal (BET) proteins, yielded some responses in relapsed/refractory acute myeloid leukemia (AML) patients in a first-in-human study presented at the meeting (abstract 7019*).

Bemcentinib (BGB324), a first-in class selective inhibitor of the AXL tyrosine kinase, also showed activity in preliminary results of a study including patients with relapsed/refractory disease (abstract 7020*).

“It will be important to know individual patient characteristics to determine the potential response predictors,” Dr Mims said.

Mivebresib (NCT02391480)

Mivebresib is the subject of an ongoing phase 1 dose-escalation study in which 23 patients have been treated. That includes 12 who received the BET inhibitor as monotherapy, and 11 who got it in combination with the BCL-2 inhibitor venetoclax, which is indicated in CLL and has breakthrough therapy designation for AML.

Investigators observed responses in 3 of 17 evaluable patients (17.6%), including 1 complete remission with incomplete blood count recovery in a patient on mivebresib monotherapy, plus 1 partial response and 1 patient achieving a morphologic leukemia-free state with the combination.

The most common grade 3/4 treatment-emergent adverse events included anemia in 52%, thrombocytopenia in 44%, and febrile neutropenia in 26% of patients, with no dose-limiting toxicities noted as of this report.

Bemcentinib (NCT02488408)

Bemcentinib is being evaluated in a phase 1/2 trial including patients with relapsed/refractory AML and myelodysplastic syndromes (MDS).

For 32 patients treated so far, 3 patients achieved a complete remission, including 1 AML and 2 MDS patients.

In addition, 3 patients achieved partial response, including 1 MDS and 2 AML patients.

Treatment with bemcentinib was generally well-tolerated, and most adverse events were mild or moderate, investigators reported in their poster.

Pre-treatment levels of soluble AXL were lower in responders compared with non-responders, investigators also noted.

“Soluble AXL levels may be a predictive biomarker for AXL inhibition, but further assessment is necessary,” Dr Mims said. 

*Data presented at the meeting differ from the abstracts.

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

©ASCO/Rodney White 2018
Poster session at ASCO 2018

CHICAGO—Two agents targeting novel pathways in myeloid malignancies—mivebresib and bencentinib—are showing promise in early studies, according to a speaker at the 2018 ASCO Annual Meeting.

“Both BET and AXL inhibition appear to be new and exciting targets in myeloid malignancies,” said Alice S. Mims, MD, and both have produced responses as single agents.

Dr Mims, of Ohio State University Wexner Medical Center in Columbus, made these observations in a poster discussion presentation that included commentary on the two agents.

Mivebresib (ABBV-075), an inhibitor of bromodomain and extra terminal (BET) proteins, yielded some responses in relapsed/refractory acute myeloid leukemia (AML) patients in a first-in-human study presented at the meeting (abstract 7019*).

Bemcentinib (BGB324), a first-in class selective inhibitor of the AXL tyrosine kinase, also showed activity in preliminary results of a study including patients with relapsed/refractory disease (abstract 7020*).

“It will be important to know individual patient characteristics to determine the potential response predictors,” Dr Mims said.

Mivebresib (NCT02391480)

Mivebresib is the subject of an ongoing phase 1 dose-escalation study in which 23 patients have been treated. That includes 12 who received the BET inhibitor as monotherapy, and 11 who got it in combination with the BCL-2 inhibitor venetoclax, which is indicated in CLL and has breakthrough therapy designation for AML.

Investigators observed responses in 3 of 17 evaluable patients (17.6%), including 1 complete remission with incomplete blood count recovery in a patient on mivebresib monotherapy, plus 1 partial response and 1 patient achieving a morphologic leukemia-free state with the combination.

The most common grade 3/4 treatment-emergent adverse events included anemia in 52%, thrombocytopenia in 44%, and febrile neutropenia in 26% of patients, with no dose-limiting toxicities noted as of this report.

Bemcentinib (NCT02488408)

Bemcentinib is being evaluated in a phase 1/2 trial including patients with relapsed/refractory AML and myelodysplastic syndromes (MDS).

For 32 patients treated so far, 3 patients achieved a complete remission, including 1 AML and 2 MDS patients.

In addition, 3 patients achieved partial response, including 1 MDS and 2 AML patients.

Treatment with bemcentinib was generally well-tolerated, and most adverse events were mild or moderate, investigators reported in their poster.

Pre-treatment levels of soluble AXL were lower in responders compared with non-responders, investigators also noted.

“Soluble AXL levels may be a predictive biomarker for AXL inhibition, but further assessment is necessary,” Dr Mims said. 

*Data presented at the meeting differ from the abstracts.

©ASCO/Rodney White 2018
Poster session at ASCO 2018

CHICAGO—Two agents targeting novel pathways in myeloid malignancies—mivebresib and bencentinib—are showing promise in early studies, according to a speaker at the 2018 ASCO Annual Meeting.

“Both BET and AXL inhibition appear to be new and exciting targets in myeloid malignancies,” said Alice S. Mims, MD, and both have produced responses as single agents.

Dr Mims, of Ohio State University Wexner Medical Center in Columbus, made these observations in a poster discussion presentation that included commentary on the two agents.

Mivebresib (ABBV-075), an inhibitor of bromodomain and extra terminal (BET) proteins, yielded some responses in relapsed/refractory acute myeloid leukemia (AML) patients in a first-in-human study presented at the meeting (abstract 7019*).

Bemcentinib (BGB324), a first-in class selective inhibitor of the AXL tyrosine kinase, also showed activity in preliminary results of a study including patients with relapsed/refractory disease (abstract 7020*).

“It will be important to know individual patient characteristics to determine the potential response predictors,” Dr Mims said.

Mivebresib (NCT02391480)

Mivebresib is the subject of an ongoing phase 1 dose-escalation study in which 23 patients have been treated. That includes 12 who received the BET inhibitor as monotherapy, and 11 who got it in combination with the BCL-2 inhibitor venetoclax, which is indicated in CLL and has breakthrough therapy designation for AML.

Investigators observed responses in 3 of 17 evaluable patients (17.6%), including 1 complete remission with incomplete blood count recovery in a patient on mivebresib monotherapy, plus 1 partial response and 1 patient achieving a morphologic leukemia-free state with the combination.

The most common grade 3/4 treatment-emergent adverse events included anemia in 52%, thrombocytopenia in 44%, and febrile neutropenia in 26% of patients, with no dose-limiting toxicities noted as of this report.

Bemcentinib (NCT02488408)

Bemcentinib is being evaluated in a phase 1/2 trial including patients with relapsed/refractory AML and myelodysplastic syndromes (MDS).

For 32 patients treated so far, 3 patients achieved a complete remission, including 1 AML and 2 MDS patients.

In addition, 3 patients achieved partial response, including 1 MDS and 2 AML patients.

Treatment with bemcentinib was generally well-tolerated, and most adverse events were mild or moderate, investigators reported in their poster.

Pre-treatment levels of soluble AXL were lower in responders compared with non-responders, investigators also noted.

“Soluble AXL levels may be a predictive biomarker for AXL inhibition, but further assessment is necessary,” Dr Mims said. 

*Data presented at the meeting differ from the abstracts.

Publications
Publications
Topics
Article Type
Display Headline
Two agents could take AML therapy in new directions
Display Headline
Two agents could take AML therapy in new directions
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Novel antibody shifts ‘eat me/don’t eat me’ balance in refractory NHL

Article Type
Changed
Tue, 01/17/2023 - 11:16

 

– A first-in-class antibody targeting the macrophage checkpoint CD47 is a promising novel immunotherapy in non-Hodgkin lymphoma, according to Ranjana H. Advani, MD, of Stanford (Calif.) Cancer Institute.

Treatment with Hu5F9-G4 (5F9), an antibody designed to overcome the “don’t eat me” signal associated with CD47, produced “encouraging” antitumor activity in a phase 1b study of 22 patients, Dr. Advani said in an oral abstract presentation at the annual meeting of the American Society of Clinical Oncology.

“5F9 was well tolerated in combination with rituximab, with no maximum tolerated dose achieved,” said Dr. Advani, noting that there were complete remissions in 43% of the refractory follicular lymphoma patients and 33% of refractory diffuse large B-cell lymphoma patients in the phase 1b/2 study.

The antibody has an on-target anemia effect that occurs upon administration, but that was mitigated considerably by a priming and maintenance dosing approach, she added.

The study has demonstrated “excellent” response rates in a highly refractory patient population, said Caron A. Jacobson, MD, of Dana-Farber Cancer Institute and Harvard Medical School in Boston. “Targeting CD47 ... really helps to shift the balance from ‘don’t eat me’ to ‘eat me,’ ” Dr. Jacobson said at the meeting.

“Importantly, we saw very little toxicity in the study, with very few grade 4 adverse events and no immune-related adverse events,” she added.

Most adverse events were grade 1 or 2, with the most common being the expected on-target anemia associated with 5F9. Using an initial priming dose of 5F9 results in a “temporary and mild decline” in hemoglobin due to clearance of aged red blood cells, Dr. Advani said.

 

 


The objective response rate in the study was 50%, with efficacy observed in rituximab-refractory patients, Dr. Advani said. With a median follow-up of greater than 6 months, just 1 of 11 responders had progressed. The median duration of response was not reached, with the longest complete remission lasting more than 14 months.

5F9 is able to selectively eliminate cancer cells through blockade of CD47, while rituximab enhances 5F9’s activity via antibody-dependent cellular phagocytosis, according to Dr. Advani.

“CD47 blockade takes the foot off the brakes, while rituximab puts the foot on the accelerator, leading to maximal tumor phagocytosis,” she said.

The Food and Drug Administration recently granted 5F9 a fast track designation for both diffuse large B-cell lymphoma and follicular lymphoma. Phase 2 investigations of 5F9 in these lymphomas are ongoing, Dr. Advani said.
 

 


The trial is sponsored by Forty Seven. Dr. Advani reported research funding from Forty Seven, which is developing 5F9, as well as disclosures related to AstraZeneca, Bayer, Bristol-Myers Squibb, Cell Medica, Genentech/Roche, Gilead Sciences, Pharmacyclics, and Seattle Genetics, among others.

SOURCE: Advani RH et al. ASCO 2018, abstract 7504.

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

 

– A first-in-class antibody targeting the macrophage checkpoint CD47 is a promising novel immunotherapy in non-Hodgkin lymphoma, according to Ranjana H. Advani, MD, of Stanford (Calif.) Cancer Institute.

Treatment with Hu5F9-G4 (5F9), an antibody designed to overcome the “don’t eat me” signal associated with CD47, produced “encouraging” antitumor activity in a phase 1b study of 22 patients, Dr. Advani said in an oral abstract presentation at the annual meeting of the American Society of Clinical Oncology.

“5F9 was well tolerated in combination with rituximab, with no maximum tolerated dose achieved,” said Dr. Advani, noting that there were complete remissions in 43% of the refractory follicular lymphoma patients and 33% of refractory diffuse large B-cell lymphoma patients in the phase 1b/2 study.

The antibody has an on-target anemia effect that occurs upon administration, but that was mitigated considerably by a priming and maintenance dosing approach, she added.

The study has demonstrated “excellent” response rates in a highly refractory patient population, said Caron A. Jacobson, MD, of Dana-Farber Cancer Institute and Harvard Medical School in Boston. “Targeting CD47 ... really helps to shift the balance from ‘don’t eat me’ to ‘eat me,’ ” Dr. Jacobson said at the meeting.

“Importantly, we saw very little toxicity in the study, with very few grade 4 adverse events and no immune-related adverse events,” she added.

Most adverse events were grade 1 or 2, with the most common being the expected on-target anemia associated with 5F9. Using an initial priming dose of 5F9 results in a “temporary and mild decline” in hemoglobin due to clearance of aged red blood cells, Dr. Advani said.

 

 


The objective response rate in the study was 50%, with efficacy observed in rituximab-refractory patients, Dr. Advani said. With a median follow-up of greater than 6 months, just 1 of 11 responders had progressed. The median duration of response was not reached, with the longest complete remission lasting more than 14 months.

5F9 is able to selectively eliminate cancer cells through blockade of CD47, while rituximab enhances 5F9’s activity via antibody-dependent cellular phagocytosis, according to Dr. Advani.

“CD47 blockade takes the foot off the brakes, while rituximab puts the foot on the accelerator, leading to maximal tumor phagocytosis,” she said.

The Food and Drug Administration recently granted 5F9 a fast track designation for both diffuse large B-cell lymphoma and follicular lymphoma. Phase 2 investigations of 5F9 in these lymphomas are ongoing, Dr. Advani said.
 

 


The trial is sponsored by Forty Seven. Dr. Advani reported research funding from Forty Seven, which is developing 5F9, as well as disclosures related to AstraZeneca, Bayer, Bristol-Myers Squibb, Cell Medica, Genentech/Roche, Gilead Sciences, Pharmacyclics, and Seattle Genetics, among others.

SOURCE: Advani RH et al. ASCO 2018, abstract 7504.

 

– A first-in-class antibody targeting the macrophage checkpoint CD47 is a promising novel immunotherapy in non-Hodgkin lymphoma, according to Ranjana H. Advani, MD, of Stanford (Calif.) Cancer Institute.

Treatment with Hu5F9-G4 (5F9), an antibody designed to overcome the “don’t eat me” signal associated with CD47, produced “encouraging” antitumor activity in a phase 1b study of 22 patients, Dr. Advani said in an oral abstract presentation at the annual meeting of the American Society of Clinical Oncology.

“5F9 was well tolerated in combination with rituximab, with no maximum tolerated dose achieved,” said Dr. Advani, noting that there were complete remissions in 43% of the refractory follicular lymphoma patients and 33% of refractory diffuse large B-cell lymphoma patients in the phase 1b/2 study.

The antibody has an on-target anemia effect that occurs upon administration, but that was mitigated considerably by a priming and maintenance dosing approach, she added.

The study has demonstrated “excellent” response rates in a highly refractory patient population, said Caron A. Jacobson, MD, of Dana-Farber Cancer Institute and Harvard Medical School in Boston. “Targeting CD47 ... really helps to shift the balance from ‘don’t eat me’ to ‘eat me,’ ” Dr. Jacobson said at the meeting.

“Importantly, we saw very little toxicity in the study, with very few grade 4 adverse events and no immune-related adverse events,” she added.

Most adverse events were grade 1 or 2, with the most common being the expected on-target anemia associated with 5F9. Using an initial priming dose of 5F9 results in a “temporary and mild decline” in hemoglobin due to clearance of aged red blood cells, Dr. Advani said.

 

 


The objective response rate in the study was 50%, with efficacy observed in rituximab-refractory patients, Dr. Advani said. With a median follow-up of greater than 6 months, just 1 of 11 responders had progressed. The median duration of response was not reached, with the longest complete remission lasting more than 14 months.

5F9 is able to selectively eliminate cancer cells through blockade of CD47, while rituximab enhances 5F9’s activity via antibody-dependent cellular phagocytosis, according to Dr. Advani.

“CD47 blockade takes the foot off the brakes, while rituximab puts the foot on the accelerator, leading to maximal tumor phagocytosis,” she said.

The Food and Drug Administration recently granted 5F9 a fast track designation for both diffuse large B-cell lymphoma and follicular lymphoma. Phase 2 investigations of 5F9 in these lymphomas are ongoing, Dr. Advani said.
 

 


The trial is sponsored by Forty Seven. Dr. Advani reported research funding from Forty Seven, which is developing 5F9, as well as disclosures related to AstraZeneca, Bayer, Bristol-Myers Squibb, Cell Medica, Genentech/Roche, Gilead Sciences, Pharmacyclics, and Seattle Genetics, among others.

SOURCE: Advani RH et al. ASCO 2018, abstract 7504.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASCO 2018

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: 5F9, a first-in-class macrophage immune checkpoint inhibitor, had promising efficacy in non-Hodgkin lymphomas.

Major finding: Complete responses were seen in 43% of follicular lymphoma (FL) patients and 33% of diffuse large B-cell lymphoma (DLBCL) patients.

Study details: Initial reported results from a phase 1b/2 study of 7 patients with FL and 15 patients with DLBCL.

Disclosures: Forty Seven sponsored the trial. Dr. Advani reported research funding from Forty Seven, which is developing 5F9, as well as disclosures related to AstraZeneca, Bayer, Bristol-Myers Squibb, Cell Medica, Genentech/Roche, Gilead Sciences, Pharmacyclics, and Seattle Genetics, among others.

Source: Advani RH et al. ASCO 2018, abstract 7504.

Disqus Comments
Default
Use ProPublica

Smoking tied to localized prostate cancer recurrence, metastasis, death

Prostate cancer–smoking link more clear
Article Type
Changed
Fri, 01/18/2019 - 17:43

 

Patients with localized prostate cancer who were smokers at the time of local therapy had a higher risk of experiencing adverse outcomes and death related to the disease, a systematic review and meta-analysis has shown.

Current smokers in the study had a higher risk of biochemical recurrence, metastasis, and cancer-specific mortality after undergoing primary radical prostatectomy or radiotherapy.

Brett Mulcahy/ThinkStock
This is the first systematic review and meta-analysis to investigate associations between smoking and outcomes after localized prostate cancer primary therapy, according to investigators led by Beat Foerster, MD, department of urology, Medical University of Vienna.

“Our findings encourage radiation oncologists and urologists to counsel patients to stop smoking, using primary prostate cancer treatment as a teachable moment,” wrote Dr. Foerster and coauthors. The report was published in JAMA Oncology.

The investigators performed a database search of studies published from January 2000 to March 2017 and selected 11 articles for quantitative analysis. Those studies, which were all observational and not randomized, comprised 22,549 patients with prostate cancer undergoing radical prostatectomy or radiotherapy. Of those patients, 4,202 (18.6%) were current smokers.

Current smokers had a higher risk of cancer-specific mortality, the investigators found based on analysis of five studies (hazard ratio, 1.89; 95% confidence interval, 1.37-2.69; P less than .001).

They also had a significantly higher risk of biochemical recurrence, based on 10 studies (HR, 1.40; 95% CI, 1.18-1.66; P less than .001), and high risk of metastasis based on 3 studies (HR, 2.51; 95% CI, 1.80-3.51; P less than .001), the report shows.

 

 


Future studies need to more closely examine the link between smoking cessation and longer-term oncologic outcomes, as well as a more precise assessment of smoking exposure, the researchers concluded.

In the current study, results were inconclusive as to whether former smoking and time to cessation had any relationship to outcomes after radical prostatectomy or radiotherapy.

“Available data were sparse and heterogenous,” they noted.

Dr. Foerster is supported by the Scholarship Foundation of Swiss Urology. One coauthor reported disclosures related to Astellas, Cepheid, Ipsen, Jansen, Lilly, Olympus, Pfizer, Pierre Fabre, Sanochemia, Sanofi, and Wolff.

SOURCE: Foerster B et al. JAMA Oncol. 2018 May 24. doi: 10.1001/jamaoncol.2018.1071.

Body

 

While previous studies have linked smoking to adverse outcomes in prostate cancer, including death, this study argues that the higher rate of prostate cancer–related death among smokers is a real effect with a biological cause, Stephen J. Freedland, MD, said in an editorial.

The current study included only men healthy enough to undergo aggressive treatment, which is an “important and necessary step to level the playing field,” Dr. Freedland wrote.

In that context, current smokers in this study had an 89% increased risk of prostate cancer–related death. “This finding shows that when we treat patients equally and minimize deaths from other causes, there is a stronger link between smoking and death from prostate cancer,” Dr. Freedland noted.

The finding also supports the notion that many smokers won’t live long enough to die from prostate cancer, given its slow-growing nature and the effects of smoking on competing causes of death, he added.

A scenario in which smokers did not live long enough to die of prostate cancer would predict a lower risk of prostate cancer–related death among smokers, he explained.

Because smoking has such clear adverse effects, physicians of all specialties should be hypervigilant about urging patients to quit smoking, Dr. Freedland concluded.

“If all of us remembered we are physicians first and oncologists and/or prostate cancer experts second, we will realize we are uniquely poised to help our patients, as the time of a new cancer diagnosis is a teachable moment when patients are very receptive to overall health advice,” he wrote.

Dr. Freedland is with the Center for Integrated Research on Cancer and Lifestyle, Cedars-Sinai Medical Center, Los Angeles. These comments are derived from his editorial in JAMA Oncology. Dr. Freeland had no reported conflict of interest disclosures.

Publications
Topics
Sections
Body

 

While previous studies have linked smoking to adverse outcomes in prostate cancer, including death, this study argues that the higher rate of prostate cancer–related death among smokers is a real effect with a biological cause, Stephen J. Freedland, MD, said in an editorial.

The current study included only men healthy enough to undergo aggressive treatment, which is an “important and necessary step to level the playing field,” Dr. Freedland wrote.

In that context, current smokers in this study had an 89% increased risk of prostate cancer–related death. “This finding shows that when we treat patients equally and minimize deaths from other causes, there is a stronger link between smoking and death from prostate cancer,” Dr. Freedland noted.

The finding also supports the notion that many smokers won’t live long enough to die from prostate cancer, given its slow-growing nature and the effects of smoking on competing causes of death, he added.

A scenario in which smokers did not live long enough to die of prostate cancer would predict a lower risk of prostate cancer–related death among smokers, he explained.

Because smoking has such clear adverse effects, physicians of all specialties should be hypervigilant about urging patients to quit smoking, Dr. Freedland concluded.

“If all of us remembered we are physicians first and oncologists and/or prostate cancer experts second, we will realize we are uniquely poised to help our patients, as the time of a new cancer diagnosis is a teachable moment when patients are very receptive to overall health advice,” he wrote.

Dr. Freedland is with the Center for Integrated Research on Cancer and Lifestyle, Cedars-Sinai Medical Center, Los Angeles. These comments are derived from his editorial in JAMA Oncology. Dr. Freeland had no reported conflict of interest disclosures.

Body

 

While previous studies have linked smoking to adverse outcomes in prostate cancer, including death, this study argues that the higher rate of prostate cancer–related death among smokers is a real effect with a biological cause, Stephen J. Freedland, MD, said in an editorial.

The current study included only men healthy enough to undergo aggressive treatment, which is an “important and necessary step to level the playing field,” Dr. Freedland wrote.

In that context, current smokers in this study had an 89% increased risk of prostate cancer–related death. “This finding shows that when we treat patients equally and minimize deaths from other causes, there is a stronger link between smoking and death from prostate cancer,” Dr. Freedland noted.

The finding also supports the notion that many smokers won’t live long enough to die from prostate cancer, given its slow-growing nature and the effects of smoking on competing causes of death, he added.

A scenario in which smokers did not live long enough to die of prostate cancer would predict a lower risk of prostate cancer–related death among smokers, he explained.

Because smoking has such clear adverse effects, physicians of all specialties should be hypervigilant about urging patients to quit smoking, Dr. Freedland concluded.

“If all of us remembered we are physicians first and oncologists and/or prostate cancer experts second, we will realize we are uniquely poised to help our patients, as the time of a new cancer diagnosis is a teachable moment when patients are very receptive to overall health advice,” he wrote.

Dr. Freedland is with the Center for Integrated Research on Cancer and Lifestyle, Cedars-Sinai Medical Center, Los Angeles. These comments are derived from his editorial in JAMA Oncology. Dr. Freeland had no reported conflict of interest disclosures.

Title
Prostate cancer–smoking link more clear
Prostate cancer–smoking link more clear

 

Patients with localized prostate cancer who were smokers at the time of local therapy had a higher risk of experiencing adverse outcomes and death related to the disease, a systematic review and meta-analysis has shown.

Current smokers in the study had a higher risk of biochemical recurrence, metastasis, and cancer-specific mortality after undergoing primary radical prostatectomy or radiotherapy.

Brett Mulcahy/ThinkStock
This is the first systematic review and meta-analysis to investigate associations between smoking and outcomes after localized prostate cancer primary therapy, according to investigators led by Beat Foerster, MD, department of urology, Medical University of Vienna.

“Our findings encourage radiation oncologists and urologists to counsel patients to stop smoking, using primary prostate cancer treatment as a teachable moment,” wrote Dr. Foerster and coauthors. The report was published in JAMA Oncology.

The investigators performed a database search of studies published from January 2000 to March 2017 and selected 11 articles for quantitative analysis. Those studies, which were all observational and not randomized, comprised 22,549 patients with prostate cancer undergoing radical prostatectomy or radiotherapy. Of those patients, 4,202 (18.6%) were current smokers.

Current smokers had a higher risk of cancer-specific mortality, the investigators found based on analysis of five studies (hazard ratio, 1.89; 95% confidence interval, 1.37-2.69; P less than .001).

They also had a significantly higher risk of biochemical recurrence, based on 10 studies (HR, 1.40; 95% CI, 1.18-1.66; P less than .001), and high risk of metastasis based on 3 studies (HR, 2.51; 95% CI, 1.80-3.51; P less than .001), the report shows.

 

 


Future studies need to more closely examine the link between smoking cessation and longer-term oncologic outcomes, as well as a more precise assessment of smoking exposure, the researchers concluded.

In the current study, results were inconclusive as to whether former smoking and time to cessation had any relationship to outcomes after radical prostatectomy or radiotherapy.

“Available data were sparse and heterogenous,” they noted.

Dr. Foerster is supported by the Scholarship Foundation of Swiss Urology. One coauthor reported disclosures related to Astellas, Cepheid, Ipsen, Jansen, Lilly, Olympus, Pfizer, Pierre Fabre, Sanochemia, Sanofi, and Wolff.

SOURCE: Foerster B et al. JAMA Oncol. 2018 May 24. doi: 10.1001/jamaoncol.2018.1071.

 

Patients with localized prostate cancer who were smokers at the time of local therapy had a higher risk of experiencing adverse outcomes and death related to the disease, a systematic review and meta-analysis has shown.

Current smokers in the study had a higher risk of biochemical recurrence, metastasis, and cancer-specific mortality after undergoing primary radical prostatectomy or radiotherapy.

Brett Mulcahy/ThinkStock
This is the first systematic review and meta-analysis to investigate associations between smoking and outcomes after localized prostate cancer primary therapy, according to investigators led by Beat Foerster, MD, department of urology, Medical University of Vienna.

“Our findings encourage radiation oncologists and urologists to counsel patients to stop smoking, using primary prostate cancer treatment as a teachable moment,” wrote Dr. Foerster and coauthors. The report was published in JAMA Oncology.

The investigators performed a database search of studies published from January 2000 to March 2017 and selected 11 articles for quantitative analysis. Those studies, which were all observational and not randomized, comprised 22,549 patients with prostate cancer undergoing radical prostatectomy or radiotherapy. Of those patients, 4,202 (18.6%) were current smokers.

Current smokers had a higher risk of cancer-specific mortality, the investigators found based on analysis of five studies (hazard ratio, 1.89; 95% confidence interval, 1.37-2.69; P less than .001).

They also had a significantly higher risk of biochemical recurrence, based on 10 studies (HR, 1.40; 95% CI, 1.18-1.66; P less than .001), and high risk of metastasis based on 3 studies (HR, 2.51; 95% CI, 1.80-3.51; P less than .001), the report shows.

 

 


Future studies need to more closely examine the link between smoking cessation and longer-term oncologic outcomes, as well as a more precise assessment of smoking exposure, the researchers concluded.

In the current study, results were inconclusive as to whether former smoking and time to cessation had any relationship to outcomes after radical prostatectomy or radiotherapy.

“Available data were sparse and heterogenous,” they noted.

Dr. Foerster is supported by the Scholarship Foundation of Swiss Urology. One coauthor reported disclosures related to Astellas, Cepheid, Ipsen, Jansen, Lilly, Olympus, Pfizer, Pierre Fabre, Sanochemia, Sanofi, and Wolff.

SOURCE: Foerster B et al. JAMA Oncol. 2018 May 24. doi: 10.1001/jamaoncol.2018.1071.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM JAMA ONCOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: Patients with localized prostate cancer should be encouraged to quit smoking because of the risk of potentially worse oncologic outcomes.

Major finding: Current smokers had a higher risk of biochemical recurrence, metastasis, and cancer-specific mortality, with hazard ratios of 1.40, 2.51, and 1.89, respectively.

Study details: A systematic review and meta-analysis of 11 studies involving 22,549 patients with prostate cancer undergoing radical prostatectomy or radiotherapy.

Disclosures: The first author is supported by the Scholarship Foundation of Swiss Urology. One coauthor reported disclosures related to Astellas, Cepheid, Ipsen, Jansen, Lilly, Olympus, Pfizer, Pierre Fabre, Sanochemia, Sanofi, and Wolff.

Source: Foerster B et al. JAMA Oncol. 2018 May 24. doi: 10.1001/jamaoncol.2018.1071.

Disqus Comments
Default
Use ProPublica

Single-agent acalabrutinib ‘impressive’ in patients with WM

Article Type
Changed
Wed, 06/06/2018 - 00:10
Display Headline
Single-agent acalabrutinib ‘impressive’ in patients with WM

©ASCO/Rodney White 2018
McCormick Place during ASCO 2018

CHICAGO—Acalabrutinib monotherapy was highly effective in Waldenström’s macroglobulinemia (WM) in a phase 2 study, investigator Roger Owen, MD, reported at the 2018 ASCO Annual Meeting.

The BTK inhibitor was effective in both treatment-naïve and relapsed/refractory patients, with overall response rates for both groups in excess of 90%, and “amazing” major response rates—partial response or better—of approximately 80%, Dr Owen said.

Dr Owen, of the St James's Institute of Oncology, Leeds Teaching Hospitals NHS Trust in Leeds, UK, reported the findings as abstract 7501.*

Durations of response were excellent, according to Dr Owen, who also reported 2-year progression-free survival of 90% in treatment-naïve patients and 82% in relapsed/refractory patients.

The safety profile was favorable, with most adverse events of low grade, and a very limited number of dropouts due to adverse events, according to the investigator.

“I think this study clearly demonstrates the highly effective nature of acalabrutinib in Waldenström’s macroglobulinemia,” Dr Owen stated.

 Acalabrutinib is a selective BTK inhibitor with minimal off-target activity, he said. The BTK inhibitor ibrutinib also has demonstrated activity in Waldenström’s, but has been associated with toxicities such as atrial fibrillation and bleeding, he noted.

In this phase 2 acalabrutinib study (NCT02180724), which included 14 treatment-naïve and 92 relapsed/refractory patients, atrial fibrillation occurred in 5 patients. However, 4 of those cases were grade 1-2, and only one was grade 3, according to Dr Owen.

Investigators observed grade 3 hypertension in 3 relapsed/refractory patients.

Bleeding events occurred in more than half of patients, though only 3 of those events were grade 3, and no patient discontinued treatment due to a bleeding episode.

These efficacy results are “impressive,” and the fact that very few cardiac events were seen is important, said Bruce D. Cheson, MD, of Georgetown University Medical Center in Washington, DC.

Dr Cheson commented on the acalabrutinib results in his presentation during ASCO on non-chemotherapy treatments for lymphoid malignancies.

 One can construct a non-chemo algorithm now for Waldenström’s, for patients who are MYD88 mutated, which is more than 90% of patients,” he said. “Right now ibrutinib, and perhaps in the future acalabrutinib, can be the initial therapy with or without rituximab based on the results of ongoing trials.”

However, single non-chemotherapy agents will not be sufficient to achieve cure of lymphoid malignancies, Dr Cheson added.

“We need to carefully develop rational combinations, identifying biomarkers for response, for resistance, for toxicity,” he said.

The study was sponsored by Acerta Pharma BV. 

*Data presented at the meeting differ from the abstract.

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

©ASCO/Rodney White 2018
McCormick Place during ASCO 2018

CHICAGO—Acalabrutinib monotherapy was highly effective in Waldenström’s macroglobulinemia (WM) in a phase 2 study, investigator Roger Owen, MD, reported at the 2018 ASCO Annual Meeting.

The BTK inhibitor was effective in both treatment-naïve and relapsed/refractory patients, with overall response rates for both groups in excess of 90%, and “amazing” major response rates—partial response or better—of approximately 80%, Dr Owen said.

Dr Owen, of the St James's Institute of Oncology, Leeds Teaching Hospitals NHS Trust in Leeds, UK, reported the findings as abstract 7501.*

Durations of response were excellent, according to Dr Owen, who also reported 2-year progression-free survival of 90% in treatment-naïve patients and 82% in relapsed/refractory patients.

The safety profile was favorable, with most adverse events of low grade, and a very limited number of dropouts due to adverse events, according to the investigator.

“I think this study clearly demonstrates the highly effective nature of acalabrutinib in Waldenström’s macroglobulinemia,” Dr Owen stated.

 Acalabrutinib is a selective BTK inhibitor with minimal off-target activity, he said. The BTK inhibitor ibrutinib also has demonstrated activity in Waldenström’s, but has been associated with toxicities such as atrial fibrillation and bleeding, he noted.

In this phase 2 acalabrutinib study (NCT02180724), which included 14 treatment-naïve and 92 relapsed/refractory patients, atrial fibrillation occurred in 5 patients. However, 4 of those cases were grade 1-2, and only one was grade 3, according to Dr Owen.

Investigators observed grade 3 hypertension in 3 relapsed/refractory patients.

Bleeding events occurred in more than half of patients, though only 3 of those events were grade 3, and no patient discontinued treatment due to a bleeding episode.

These efficacy results are “impressive,” and the fact that very few cardiac events were seen is important, said Bruce D. Cheson, MD, of Georgetown University Medical Center in Washington, DC.

Dr Cheson commented on the acalabrutinib results in his presentation during ASCO on non-chemotherapy treatments for lymphoid malignancies.

 One can construct a non-chemo algorithm now for Waldenström’s, for patients who are MYD88 mutated, which is more than 90% of patients,” he said. “Right now ibrutinib, and perhaps in the future acalabrutinib, can be the initial therapy with or without rituximab based on the results of ongoing trials.”

However, single non-chemotherapy agents will not be sufficient to achieve cure of lymphoid malignancies, Dr Cheson added.

“We need to carefully develop rational combinations, identifying biomarkers for response, for resistance, for toxicity,” he said.

The study was sponsored by Acerta Pharma BV. 

*Data presented at the meeting differ from the abstract.

©ASCO/Rodney White 2018
McCormick Place during ASCO 2018

CHICAGO—Acalabrutinib monotherapy was highly effective in Waldenström’s macroglobulinemia (WM) in a phase 2 study, investigator Roger Owen, MD, reported at the 2018 ASCO Annual Meeting.

The BTK inhibitor was effective in both treatment-naïve and relapsed/refractory patients, with overall response rates for both groups in excess of 90%, and “amazing” major response rates—partial response or better—of approximately 80%, Dr Owen said.

Dr Owen, of the St James's Institute of Oncology, Leeds Teaching Hospitals NHS Trust in Leeds, UK, reported the findings as abstract 7501.*

Durations of response were excellent, according to Dr Owen, who also reported 2-year progression-free survival of 90% in treatment-naïve patients and 82% in relapsed/refractory patients.

The safety profile was favorable, with most adverse events of low grade, and a very limited number of dropouts due to adverse events, according to the investigator.

“I think this study clearly demonstrates the highly effective nature of acalabrutinib in Waldenström’s macroglobulinemia,” Dr Owen stated.

 Acalabrutinib is a selective BTK inhibitor with minimal off-target activity, he said. The BTK inhibitor ibrutinib also has demonstrated activity in Waldenström’s, but has been associated with toxicities such as atrial fibrillation and bleeding, he noted.

In this phase 2 acalabrutinib study (NCT02180724), which included 14 treatment-naïve and 92 relapsed/refractory patients, atrial fibrillation occurred in 5 patients. However, 4 of those cases were grade 1-2, and only one was grade 3, according to Dr Owen.

Investigators observed grade 3 hypertension in 3 relapsed/refractory patients.

Bleeding events occurred in more than half of patients, though only 3 of those events were grade 3, and no patient discontinued treatment due to a bleeding episode.

These efficacy results are “impressive,” and the fact that very few cardiac events were seen is important, said Bruce D. Cheson, MD, of Georgetown University Medical Center in Washington, DC.

Dr Cheson commented on the acalabrutinib results in his presentation during ASCO on non-chemotherapy treatments for lymphoid malignancies.

 One can construct a non-chemo algorithm now for Waldenström’s, for patients who are MYD88 mutated, which is more than 90% of patients,” he said. “Right now ibrutinib, and perhaps in the future acalabrutinib, can be the initial therapy with or without rituximab based on the results of ongoing trials.”

However, single non-chemotherapy agents will not be sufficient to achieve cure of lymphoid malignancies, Dr Cheson added.

“We need to carefully develop rational combinations, identifying biomarkers for response, for resistance, for toxicity,” he said.

The study was sponsored by Acerta Pharma BV. 

*Data presented at the meeting differ from the abstract.

Publications
Publications
Topics
Article Type
Display Headline
Single-agent acalabrutinib ‘impressive’ in patients with WM
Display Headline
Single-agent acalabrutinib ‘impressive’ in patients with WM
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

MRD-negative status signals better outcomes in CAR T–treated ALL

Article Type
Changed
Fri, 01/04/2019 - 10:26

 

– Minimal residual disease (MRD)–negative complete remission was strongly associated with improved survival outcomes in patients with B-cell acute lymphocytic leukemia (ALL) who received CD19 chimeric antigen receptor (CAR) T cells, results of a retrospective study showed.

Allogeneic hematopoietic stem cell transplant (HSCT) appeared to improve both disease-free and overall survival in those patients who had achieved MRD-negative complete remission, according to results of the study, which were presented at the annual meeting of the American Society of Clinical Oncology.

“Based upon our interaction testing, the potential benefit [of transplant] appears to exist in both good-risk and bad-risk patients as identified through multivariate modeling,” said study investigator Kevin Anthony Hay, MD, of Fred Hutchinson Cancer Research Center, Seattle.

In a comment on the results, Sarah Cooley, MD, noted that the benefits of allogeneic transplant were apparent regardless of whether the patients met criteria for the good-risk subgroup, which was defined by levels of lactate dehydrogenase (LDH) and platelets along with exposure to fludarabine as part of the conditioning regimen.

“I think this suggests that the goal at this point is to get patients to an MRD-negative state and to potentially curative transplant,” said Dr. Cooley, director of investigator-initiated research at Masonic Medical Center at the University of Minnesota, Minneapolis.

The retrospective analysis by Dr. Hay and his colleagues included 53 adults with relapsed or refractory ALL who had bone marrow or extramedullary disease at baseline and had received CD19 CAR T cells at or under the maximum tolerated dose at least 1 year prior to this analysis. Of that group, 45 (85%) achieved MRD-negative complete remission.

Those patients who did achieve MRD-negative complete remission had an improved median disease-free survival at 7.6 months versus 0.8 months (P less than .0001) and improved overall survival at 20.0 months versus 5.0 months (P = 0.014).

 

 


Most of the MRD-negative patients who relapsed did so within the first 6 months, an observation that led investigators to consider whether factors exist that could predict better outcomes.

In a multivariate analysis, they found three variables associated with disease free survival: higher LDH prior to lymphodepletion (hazard ratio, 1.39), along with higher platelet count prior to lymphodepletion and incorporation of fludarabine into the regimen, with hazard ratios of 0.65 and 0.34, respectively.

Using those three characteristics, investigators grouped patients as “good risk” if they had normal LDH, platelet count at or above 100 prior to lymphodepletion that included fludarabine. The 24-month disease-free survival for good-risk patients was 78%, and overall survival was 86%.

The role of allogeneic HSCT after ALL patients achieved MRD-negative complete remission with CAR T-cell therapy was one of the “major questions in the field,” Dr. Hay said.
 

 


In this analysis, Dr. Hay and colleagues found that patients who underwent transplant in MRD-negative complete remission had a 24-month disease free survival and overall survival of 61% and 72%, respectively, both of which were significantly higher than in patients with MRD-negative complete remission who had no transplant.

The disease-free survival benefit was not specific to the good-risk group, according to Dr. Hay, who said an interaction test demonstrated no significant interaction between risk group and allogeneic HSCT after CAR T-cell infusion (P = 0.53).

“This is a very important finding that should be further [studied] in an appropriately designed clinical trial,” Dr. Hay said during an oral presentation of the study results.

Dr. Hay and several coauthors reported financial disclosures related to Juno Therapeutics. Other disclosures reported by study coauthors included Cell Medica, Celgene, Eureka Therapeutics, Genentech/Roche, Gilead Sciences, Kite Pharma, Novartis, and others.

SOURCE: Hay KA. ASCO 2018, Abstract 7005.

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

 

– Minimal residual disease (MRD)–negative complete remission was strongly associated with improved survival outcomes in patients with B-cell acute lymphocytic leukemia (ALL) who received CD19 chimeric antigen receptor (CAR) T cells, results of a retrospective study showed.

Allogeneic hematopoietic stem cell transplant (HSCT) appeared to improve both disease-free and overall survival in those patients who had achieved MRD-negative complete remission, according to results of the study, which were presented at the annual meeting of the American Society of Clinical Oncology.

“Based upon our interaction testing, the potential benefit [of transplant] appears to exist in both good-risk and bad-risk patients as identified through multivariate modeling,” said study investigator Kevin Anthony Hay, MD, of Fred Hutchinson Cancer Research Center, Seattle.

In a comment on the results, Sarah Cooley, MD, noted that the benefits of allogeneic transplant were apparent regardless of whether the patients met criteria for the good-risk subgroup, which was defined by levels of lactate dehydrogenase (LDH) and platelets along with exposure to fludarabine as part of the conditioning regimen.

“I think this suggests that the goal at this point is to get patients to an MRD-negative state and to potentially curative transplant,” said Dr. Cooley, director of investigator-initiated research at Masonic Medical Center at the University of Minnesota, Minneapolis.

The retrospective analysis by Dr. Hay and his colleagues included 53 adults with relapsed or refractory ALL who had bone marrow or extramedullary disease at baseline and had received CD19 CAR T cells at or under the maximum tolerated dose at least 1 year prior to this analysis. Of that group, 45 (85%) achieved MRD-negative complete remission.

Those patients who did achieve MRD-negative complete remission had an improved median disease-free survival at 7.6 months versus 0.8 months (P less than .0001) and improved overall survival at 20.0 months versus 5.0 months (P = 0.014).

 

 


Most of the MRD-negative patients who relapsed did so within the first 6 months, an observation that led investigators to consider whether factors exist that could predict better outcomes.

In a multivariate analysis, they found three variables associated with disease free survival: higher LDH prior to lymphodepletion (hazard ratio, 1.39), along with higher platelet count prior to lymphodepletion and incorporation of fludarabine into the regimen, with hazard ratios of 0.65 and 0.34, respectively.

Using those three characteristics, investigators grouped patients as “good risk” if they had normal LDH, platelet count at or above 100 prior to lymphodepletion that included fludarabine. The 24-month disease-free survival for good-risk patients was 78%, and overall survival was 86%.

The role of allogeneic HSCT after ALL patients achieved MRD-negative complete remission with CAR T-cell therapy was one of the “major questions in the field,” Dr. Hay said.
 

 


In this analysis, Dr. Hay and colleagues found that patients who underwent transplant in MRD-negative complete remission had a 24-month disease free survival and overall survival of 61% and 72%, respectively, both of which were significantly higher than in patients with MRD-negative complete remission who had no transplant.

The disease-free survival benefit was not specific to the good-risk group, according to Dr. Hay, who said an interaction test demonstrated no significant interaction between risk group and allogeneic HSCT after CAR T-cell infusion (P = 0.53).

“This is a very important finding that should be further [studied] in an appropriately designed clinical trial,” Dr. Hay said during an oral presentation of the study results.

Dr. Hay and several coauthors reported financial disclosures related to Juno Therapeutics. Other disclosures reported by study coauthors included Cell Medica, Celgene, Eureka Therapeutics, Genentech/Roche, Gilead Sciences, Kite Pharma, Novartis, and others.

SOURCE: Hay KA. ASCO 2018, Abstract 7005.

 

– Minimal residual disease (MRD)–negative complete remission was strongly associated with improved survival outcomes in patients with B-cell acute lymphocytic leukemia (ALL) who received CD19 chimeric antigen receptor (CAR) T cells, results of a retrospective study showed.

Allogeneic hematopoietic stem cell transplant (HSCT) appeared to improve both disease-free and overall survival in those patients who had achieved MRD-negative complete remission, according to results of the study, which were presented at the annual meeting of the American Society of Clinical Oncology.

“Based upon our interaction testing, the potential benefit [of transplant] appears to exist in both good-risk and bad-risk patients as identified through multivariate modeling,” said study investigator Kevin Anthony Hay, MD, of Fred Hutchinson Cancer Research Center, Seattle.

In a comment on the results, Sarah Cooley, MD, noted that the benefits of allogeneic transplant were apparent regardless of whether the patients met criteria for the good-risk subgroup, which was defined by levels of lactate dehydrogenase (LDH) and platelets along with exposure to fludarabine as part of the conditioning regimen.

“I think this suggests that the goal at this point is to get patients to an MRD-negative state and to potentially curative transplant,” said Dr. Cooley, director of investigator-initiated research at Masonic Medical Center at the University of Minnesota, Minneapolis.

The retrospective analysis by Dr. Hay and his colleagues included 53 adults with relapsed or refractory ALL who had bone marrow or extramedullary disease at baseline and had received CD19 CAR T cells at or under the maximum tolerated dose at least 1 year prior to this analysis. Of that group, 45 (85%) achieved MRD-negative complete remission.

Those patients who did achieve MRD-negative complete remission had an improved median disease-free survival at 7.6 months versus 0.8 months (P less than .0001) and improved overall survival at 20.0 months versus 5.0 months (P = 0.014).

 

 


Most of the MRD-negative patients who relapsed did so within the first 6 months, an observation that led investigators to consider whether factors exist that could predict better outcomes.

In a multivariate analysis, they found three variables associated with disease free survival: higher LDH prior to lymphodepletion (hazard ratio, 1.39), along with higher platelet count prior to lymphodepletion and incorporation of fludarabine into the regimen, with hazard ratios of 0.65 and 0.34, respectively.

Using those three characteristics, investigators grouped patients as “good risk” if they had normal LDH, platelet count at or above 100 prior to lymphodepletion that included fludarabine. The 24-month disease-free survival for good-risk patients was 78%, and overall survival was 86%.

The role of allogeneic HSCT after ALL patients achieved MRD-negative complete remission with CAR T-cell therapy was one of the “major questions in the field,” Dr. Hay said.
 

 


In this analysis, Dr. Hay and colleagues found that patients who underwent transplant in MRD-negative complete remission had a 24-month disease free survival and overall survival of 61% and 72%, respectively, both of which were significantly higher than in patients with MRD-negative complete remission who had no transplant.

The disease-free survival benefit was not specific to the good-risk group, according to Dr. Hay, who said an interaction test demonstrated no significant interaction between risk group and allogeneic HSCT after CAR T-cell infusion (P = 0.53).

“This is a very important finding that should be further [studied] in an appropriately designed clinical trial,” Dr. Hay said during an oral presentation of the study results.

Dr. Hay and several coauthors reported financial disclosures related to Juno Therapeutics. Other disclosures reported by study coauthors included Cell Medica, Celgene, Eureka Therapeutics, Genentech/Roche, Gilead Sciences, Kite Pharma, Novartis, and others.

SOURCE: Hay KA. ASCO 2018, Abstract 7005.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASCO 2018

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: Minimal residual disease (MRD)–negative complete remission was strongly associated with improved survival outcomes.

Major finding: Patients who achieved MRD-negative complete remission had an improved median disease-free survival at 7.6 months versus 0.8 months (P less than .0001)

Study details: A retrospective analysis including 53 patients with ALL who had bone marrow or extramedullary disease at baseline and had received CD19 CAR T cells at or under the maximum tolerated dose at least 1 year prior to this analysis.

Disclosures: Researchers reported financial ties to Juno Therapeutics, Cell Medica, Celgene, Eureka Therapeutics, Genentech/Roche, Gilead Sciences, Kite Pharma, Novartis, and others.

Source: Hay KA. ASCO 2018, Abstract 7005.

Disqus Comments
Default
Use ProPublica

Encouraging early results for CB-derived NK cells in MM

Article Type
Changed
Tue, 06/05/2018 - 00:15
Display Headline
Encouraging early results for CB-derived NK cells in MM

©ASCO/Todd Buchanan 2018
Attendees at ASCO 2018

CHICAGO—Cord blood (CB) is a viable source of natural killer (NK) cells for adoptive cellular therapy for multiple myeloma (MM), according to a speaker at the 2018 ASCO Annual Meeting.

Ex-vivo expanded cord blood NK cells were well tolerated without significant graft-versus-host disease (GVHD) or cytokine release syndrome (CRS) in a phase 2 study.

Nina Shah, MD, of the University of California San Francisco, reported these results as abstract 8006.*

The phase 2 study (NCT01729091) included 33 patients with symptomatic MM who were appropriate candidates for autologous stem cell transplant (ASCT).

For each patient, investigators chose cord blood units with at least a 4/6 match at HLA-A, -B and –DR.

Prior to the autologous graft, patients received lenalidomide and melphalan. Lenalidomide was given based on preclinical data suggesting synergy between that immunomodulatory agent and NK cells, Dr Shah said.

Patients were a median age of 59 (range, 25 – 72), 36% had a history of progressive disease or relapse, and 73% had adverse cytogenetics/FISH, were ISS III, or had a history of progressive disease or relapse.

Results

Dr Shah observed that in a generally high-risk population, responses to treatment with cord blood NK cells in the setting of ASCT were “encouraging,” with 79% of patients achieving very good partial response (VGPR) or better.

Twenty-one patients (64%) achieved a complete response (CR) or near CR. And 61% achieved minimal residual disease (MRD) negativity by day 100.

Patients had an estimated 3-year progression-free survival of 52%.

Three patients died, all from disease progression, and 13 patients have progressed.

The investigators observed no infusional toxicities, no GVHD, no CRS, and no neurotoxicity.

One patient experienced graft failure and was rescued with an autologous back-up graft.

"We are able to detect the donor-derived NK cells up to 13 days after infusion,” Dr Shah said, “but I think a more sensitive analysis with flow chimerism will not only allow us to detect more patients, but also better interrogate them to truly understand the in vivo phenotype and activation status of these cells."

Dr Shah indicated she and her colleagues became interested in studying cord blood for NK cell therapy because it is a known source of hematopoietic cells that is immediately available, does not require donor manipulation, and has more flexibility in genetic matching.

Previously, Dr Shah and colleagues conducted a phase 1 study, in which 12 patients received cord blood NK cells up to a dose of 1 x 108 NK cells/kg. “This was determined to be adequate and safe to move on to the phase 2,” she said.

Despite encouraging results, more research needs to be done, according to Dr Shah. “I don't think this is the end-all, be-all for NK cell therapy.”

Some future directions include combination with antibody therapy, improving NK persistence in vivo using cytokine manipulation, and possibly engineering chimeric antigen receptor (CAR)-modified NK cells, Dr Shah observed.

It’s also possible that HLA match may not be needed: “If that is the case, we will truly have an off-the-shelf source of NK cells that we can apply more readily to various patients,” she said.

The study was supported by Celgene Corporation, Stading-Younger Cancer Research Foundation, and the MD Anderson High-Risk Multiple Myeloma Moonshot Project.  

*Data in the presentation differ from the abstract.

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

©ASCO/Todd Buchanan 2018
Attendees at ASCO 2018

CHICAGO—Cord blood (CB) is a viable source of natural killer (NK) cells for adoptive cellular therapy for multiple myeloma (MM), according to a speaker at the 2018 ASCO Annual Meeting.

Ex-vivo expanded cord blood NK cells were well tolerated without significant graft-versus-host disease (GVHD) or cytokine release syndrome (CRS) in a phase 2 study.

Nina Shah, MD, of the University of California San Francisco, reported these results as abstract 8006.*

The phase 2 study (NCT01729091) included 33 patients with symptomatic MM who were appropriate candidates for autologous stem cell transplant (ASCT).

For each patient, investigators chose cord blood units with at least a 4/6 match at HLA-A, -B and –DR.

Prior to the autologous graft, patients received lenalidomide and melphalan. Lenalidomide was given based on preclinical data suggesting synergy between that immunomodulatory agent and NK cells, Dr Shah said.

Patients were a median age of 59 (range, 25 – 72), 36% had a history of progressive disease or relapse, and 73% had adverse cytogenetics/FISH, were ISS III, or had a history of progressive disease or relapse.

Results

Dr Shah observed that in a generally high-risk population, responses to treatment with cord blood NK cells in the setting of ASCT were “encouraging,” with 79% of patients achieving very good partial response (VGPR) or better.

Twenty-one patients (64%) achieved a complete response (CR) or near CR. And 61% achieved minimal residual disease (MRD) negativity by day 100.

Patients had an estimated 3-year progression-free survival of 52%.

Three patients died, all from disease progression, and 13 patients have progressed.

The investigators observed no infusional toxicities, no GVHD, no CRS, and no neurotoxicity.

One patient experienced graft failure and was rescued with an autologous back-up graft.

"We are able to detect the donor-derived NK cells up to 13 days after infusion,” Dr Shah said, “but I think a more sensitive analysis with flow chimerism will not only allow us to detect more patients, but also better interrogate them to truly understand the in vivo phenotype and activation status of these cells."

Dr Shah indicated she and her colleagues became interested in studying cord blood for NK cell therapy because it is a known source of hematopoietic cells that is immediately available, does not require donor manipulation, and has more flexibility in genetic matching.

Previously, Dr Shah and colleagues conducted a phase 1 study, in which 12 patients received cord blood NK cells up to a dose of 1 x 108 NK cells/kg. “This was determined to be adequate and safe to move on to the phase 2,” she said.

Despite encouraging results, more research needs to be done, according to Dr Shah. “I don't think this is the end-all, be-all for NK cell therapy.”

Some future directions include combination with antibody therapy, improving NK persistence in vivo using cytokine manipulation, and possibly engineering chimeric antigen receptor (CAR)-modified NK cells, Dr Shah observed.

It’s also possible that HLA match may not be needed: “If that is the case, we will truly have an off-the-shelf source of NK cells that we can apply more readily to various patients,” she said.

The study was supported by Celgene Corporation, Stading-Younger Cancer Research Foundation, and the MD Anderson High-Risk Multiple Myeloma Moonshot Project.  

*Data in the presentation differ from the abstract.

©ASCO/Todd Buchanan 2018
Attendees at ASCO 2018

CHICAGO—Cord blood (CB) is a viable source of natural killer (NK) cells for adoptive cellular therapy for multiple myeloma (MM), according to a speaker at the 2018 ASCO Annual Meeting.

Ex-vivo expanded cord blood NK cells were well tolerated without significant graft-versus-host disease (GVHD) or cytokine release syndrome (CRS) in a phase 2 study.

Nina Shah, MD, of the University of California San Francisco, reported these results as abstract 8006.*

The phase 2 study (NCT01729091) included 33 patients with symptomatic MM who were appropriate candidates for autologous stem cell transplant (ASCT).

For each patient, investigators chose cord blood units with at least a 4/6 match at HLA-A, -B and –DR.

Prior to the autologous graft, patients received lenalidomide and melphalan. Lenalidomide was given based on preclinical data suggesting synergy between that immunomodulatory agent and NK cells, Dr Shah said.

Patients were a median age of 59 (range, 25 – 72), 36% had a history of progressive disease or relapse, and 73% had adverse cytogenetics/FISH, were ISS III, or had a history of progressive disease or relapse.

Results

Dr Shah observed that in a generally high-risk population, responses to treatment with cord blood NK cells in the setting of ASCT were “encouraging,” with 79% of patients achieving very good partial response (VGPR) or better.

Twenty-one patients (64%) achieved a complete response (CR) or near CR. And 61% achieved minimal residual disease (MRD) negativity by day 100.

Patients had an estimated 3-year progression-free survival of 52%.

Three patients died, all from disease progression, and 13 patients have progressed.

The investigators observed no infusional toxicities, no GVHD, no CRS, and no neurotoxicity.

One patient experienced graft failure and was rescued with an autologous back-up graft.

"We are able to detect the donor-derived NK cells up to 13 days after infusion,” Dr Shah said, “but I think a more sensitive analysis with flow chimerism will not only allow us to detect more patients, but also better interrogate them to truly understand the in vivo phenotype and activation status of these cells."

Dr Shah indicated she and her colleagues became interested in studying cord blood for NK cell therapy because it is a known source of hematopoietic cells that is immediately available, does not require donor manipulation, and has more flexibility in genetic matching.

Previously, Dr Shah and colleagues conducted a phase 1 study, in which 12 patients received cord blood NK cells up to a dose of 1 x 108 NK cells/kg. “This was determined to be adequate and safe to move on to the phase 2,” she said.

Despite encouraging results, more research needs to be done, according to Dr Shah. “I don't think this is the end-all, be-all for NK cell therapy.”

Some future directions include combination with antibody therapy, improving NK persistence in vivo using cytokine manipulation, and possibly engineering chimeric antigen receptor (CAR)-modified NK cells, Dr Shah observed.

It’s also possible that HLA match may not be needed: “If that is the case, we will truly have an off-the-shelf source of NK cells that we can apply more readily to various patients,” she said.

The study was supported by Celgene Corporation, Stading-Younger Cancer Research Foundation, and the MD Anderson High-Risk Multiple Myeloma Moonshot Project.  

*Data in the presentation differ from the abstract.

Publications
Publications
Topics
Article Type
Display Headline
Encouraging early results for CB-derived NK cells in MM
Display Headline
Encouraging early results for CB-derived NK cells in MM
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

ACS: Start colorectal cancer screening at age 45

Dr. John M. Inadomi, AGAF, of the University of Washington provides a perspective on the recommendation to start screening younger
Article Type
Changed
Wed, 05/26/2021 - 13:49

The American Cancer Society recommends all U.S. adults at average risk of colorectal cancer (CRC) undergo screening starting at age 45 years.

That update to ACS recommendations is based on an increasing burden of CRC in younger individuals, microsimulation modeling results, and a “reasonable expectation” that screening tests will perform as well in adults aged 45-49 years as they do in older adults, members of the ACS Guideline Development Group said in the guideline, which was published in CA: A Cancer Journal for Clinicians.

Starting screening at age 45 contrasts with recommendations from the U.S. Preventive Services Task Force (USPSTF), which in 2016 gave an “A” recommendation for CRC screening from 50 to 75 years of age. At the time, the USPSTF noted a modest increase in life-years gained by starting earlier, based on microsimulation modeling. But it concluded that available evidence best supported starting at age 50.

The updated ACS guidelines are based in part on a modeling study that the authors say extends the previous analysis conducted for the USPSTF.

“The recommendation places a high value on the potential years of life saved, addresses anticipated rising incidence going forward, and is expected to contribute to the reduction in disparities in incidence before age 50 years in some racial groups,” the ACS guideline authors added.

The recommendation to start screening at age 45 is a “qualified” recommendation, the authors said, given the limitations of the current evidence base. Most studies to date have been focused on older individuals, in keeping with long-standing recommendations to start screening at age 50.

The move downward in screening age recommendation acknowledges one of the most “significant and disturbing” developments in CRC, the guideline’s authors said: the marked increase in incidence among younger individuals.

While CRC incidence and mortality have been declining in adults aged 55 years and older, recent studies cited in the ACS guideline document show a 51% increase in incidence from 1994 to 2014 – and an 11% increase in mortality from 2005 to 2015 – for adults younger than 55 years.

The current age-specific incidence rate for adults 45-49 years is 31.4 per 100,000, compared with 58.4 per 100,000 in adults 50-54 years. However, the ACS guideline authors said the higher rate in the older cohort is partly influenced by more frequent screening. “The true underlying risk in adults aged 45-49 years is likely closer to the risk in adults aged 50-54 years than the most recent age-specific rates would suggest,” they wrote.

Since patients in this age range have not been routinely screened before, the ACS recommendation is based on modeling. Now we need to analyze the outcomes of early screening to identify which patients will benefit most. Choices for screening include either a structural examination or a high-sensitivity stool-based test, according to the guideline, which doesn’t state a preference for any particular test.

The AGA, in their statement in response, noted that with CRC rates rising in people younger than age 50, it is appropriate to consider beginning routine screening at age 45. The statement continues “Since patients in this age range have not been previously routinely screened, the ACS recommendation is based on modeling. Now we need to analyze the outcomes of early screening to identify which patients will benefit most.”

 

 

In addition to Dr. Wolf, members of the ACS Guideline Development Group received no compensation.

SOURCE: Wolf AMD et al. CA Cancer J Clin. 2018 May 30. doi: 10.3322/caac.21457.

Body

The latest recommendations from the American Cancer Society added individuals 45 years and older to the population for whom CRC screeningshould be performed. The change from a start age of 50 was prompted by the increase in CRC reported in younger adults and was based on a computer simulation that predicted a greater number of life-years saved using an earlier age for initiation of screening among adults at average risk for development of colorectal cancer. It is likely that screening will reduce cancer mortality even in this younger age group; however, several issues should be considered when implementing this policy. 
 

Differences in screening tests: The reason for the increase in CRC in younger adults is not known. Nor is it understood why this increase is far greater for rectal cancer than cancers more proximal in the colon. Based on this observation, however, it is possible that flexible sigmoidoscopy may be a more appropriate test than colonoscopy for younger adults. Conversely, we do not know if the precursor of early-age CRC is more likely to be a flat lesion that is more difficult to detect using endoscopy, or less likely to bleed that may make FIT less able to detect, or have a genetic mechanism different from proximal CRC that is not part of the current DNA stool testing. 

Robert Lodge/MDedge News
Dr. John M. Inadomi
The evidence supporting screening tests are not equal. No randomized trial confirming the effectiveness of screening colonoscopy to reduce CRC mortality has been completed, although at least four studies are ongoing. More importantly, a recent study of one-time screening flexible sigmoidoscopy published in JAMA reported a significant reduction in CRC incidence and mortality among men that was not seen among women. A variety of factors may have caused this observation, one of which is that the age-related incidence of CRC among women is lower compared with men. One-time screening will prevent fewer cancers in women since the majority of cancers precursors have not developed at a younger age. Starting screening at age 45 years may miss even more cancers among women. 

Value is the benefit gained with screening compared with the resources required to implement screening. The value of screening is greater in older individuals than in younger individuals because the risk of CRC is increased and for this reason, population-based screening should focus on screening older adults who have not undergone screening. Unfortunately, U.S. population adherence to CRC screening remains below 70% with little improvement since 2010. Only after the older population is fully screened should our attention shift to younger populations.  
 

Disparities: The individuals most likely to undergo screening are unlikely to be the individuals most likely to benefit. African Americans have a higher age-related incidence of CRC but have the lowest screening rates in the U.S. compared with other racial and ethnic groups. This relates to not only reduced access but also reduced utilization. It is a concern that, by increasing the pool of individuals recommended for screening, we may also reduce access to those who may benefit most.  

The ACS recommendations to go low may reduce colorectal cancer mortality in younger adults; however, our lack of understanding about the biology of the cancer hampers our ability to recommend the optimal screening strategy, sacrifices value, and may increase disparities in cancer outcomes.  
 
John M. Inadomi, MD, AGAF, is a Cyrus E. Rubin Professor of Medicine and head of the division of gastroenterology at the University of Washington School of Medicine, Seattle. He has no conflicts. 

Publications
Topics
Sections
Body

The latest recommendations from the American Cancer Society added individuals 45 years and older to the population for whom CRC screeningshould be performed. The change from a start age of 50 was prompted by the increase in CRC reported in younger adults and was based on a computer simulation that predicted a greater number of life-years saved using an earlier age for initiation of screening among adults at average risk for development of colorectal cancer. It is likely that screening will reduce cancer mortality even in this younger age group; however, several issues should be considered when implementing this policy. 
 

Differences in screening tests: The reason for the increase in CRC in younger adults is not known. Nor is it understood why this increase is far greater for rectal cancer than cancers more proximal in the colon. Based on this observation, however, it is possible that flexible sigmoidoscopy may be a more appropriate test than colonoscopy for younger adults. Conversely, we do not know if the precursor of early-age CRC is more likely to be a flat lesion that is more difficult to detect using endoscopy, or less likely to bleed that may make FIT less able to detect, or have a genetic mechanism different from proximal CRC that is not part of the current DNA stool testing. 

Robert Lodge/MDedge News
Dr. John M. Inadomi
The evidence supporting screening tests are not equal. No randomized trial confirming the effectiveness of screening colonoscopy to reduce CRC mortality has been completed, although at least four studies are ongoing. More importantly, a recent study of one-time screening flexible sigmoidoscopy published in JAMA reported a significant reduction in CRC incidence and mortality among men that was not seen among women. A variety of factors may have caused this observation, one of which is that the age-related incidence of CRC among women is lower compared with men. One-time screening will prevent fewer cancers in women since the majority of cancers precursors have not developed at a younger age. Starting screening at age 45 years may miss even more cancers among women. 

Value is the benefit gained with screening compared with the resources required to implement screening. The value of screening is greater in older individuals than in younger individuals because the risk of CRC is increased and for this reason, population-based screening should focus on screening older adults who have not undergone screening. Unfortunately, U.S. population adherence to CRC screening remains below 70% with little improvement since 2010. Only after the older population is fully screened should our attention shift to younger populations.  
 

Disparities: The individuals most likely to undergo screening are unlikely to be the individuals most likely to benefit. African Americans have a higher age-related incidence of CRC but have the lowest screening rates in the U.S. compared with other racial and ethnic groups. This relates to not only reduced access but also reduced utilization. It is a concern that, by increasing the pool of individuals recommended for screening, we may also reduce access to those who may benefit most.  

The ACS recommendations to go low may reduce colorectal cancer mortality in younger adults; however, our lack of understanding about the biology of the cancer hampers our ability to recommend the optimal screening strategy, sacrifices value, and may increase disparities in cancer outcomes.  
 
John M. Inadomi, MD, AGAF, is a Cyrus E. Rubin Professor of Medicine and head of the division of gastroenterology at the University of Washington School of Medicine, Seattle. He has no conflicts. 

Body

The latest recommendations from the American Cancer Society added individuals 45 years and older to the population for whom CRC screeningshould be performed. The change from a start age of 50 was prompted by the increase in CRC reported in younger adults and was based on a computer simulation that predicted a greater number of life-years saved using an earlier age for initiation of screening among adults at average risk for development of colorectal cancer. It is likely that screening will reduce cancer mortality even in this younger age group; however, several issues should be considered when implementing this policy. 
 

Differences in screening tests: The reason for the increase in CRC in younger adults is not known. Nor is it understood why this increase is far greater for rectal cancer than cancers more proximal in the colon. Based on this observation, however, it is possible that flexible sigmoidoscopy may be a more appropriate test than colonoscopy for younger adults. Conversely, we do not know if the precursor of early-age CRC is more likely to be a flat lesion that is more difficult to detect using endoscopy, or less likely to bleed that may make FIT less able to detect, or have a genetic mechanism different from proximal CRC that is not part of the current DNA stool testing. 

Robert Lodge/MDedge News
Dr. John M. Inadomi
The evidence supporting screening tests are not equal. No randomized trial confirming the effectiveness of screening colonoscopy to reduce CRC mortality has been completed, although at least four studies are ongoing. More importantly, a recent study of one-time screening flexible sigmoidoscopy published in JAMA reported a significant reduction in CRC incidence and mortality among men that was not seen among women. A variety of factors may have caused this observation, one of which is that the age-related incidence of CRC among women is lower compared with men. One-time screening will prevent fewer cancers in women since the majority of cancers precursors have not developed at a younger age. Starting screening at age 45 years may miss even more cancers among women. 

Value is the benefit gained with screening compared with the resources required to implement screening. The value of screening is greater in older individuals than in younger individuals because the risk of CRC is increased and for this reason, population-based screening should focus on screening older adults who have not undergone screening. Unfortunately, U.S. population adherence to CRC screening remains below 70% with little improvement since 2010. Only after the older population is fully screened should our attention shift to younger populations.  
 

Disparities: The individuals most likely to undergo screening are unlikely to be the individuals most likely to benefit. African Americans have a higher age-related incidence of CRC but have the lowest screening rates in the U.S. compared with other racial and ethnic groups. This relates to not only reduced access but also reduced utilization. It is a concern that, by increasing the pool of individuals recommended for screening, we may also reduce access to those who may benefit most.  

The ACS recommendations to go low may reduce colorectal cancer mortality in younger adults; however, our lack of understanding about the biology of the cancer hampers our ability to recommend the optimal screening strategy, sacrifices value, and may increase disparities in cancer outcomes.  
 
John M. Inadomi, MD, AGAF, is a Cyrus E. Rubin Professor of Medicine and head of the division of gastroenterology at the University of Washington School of Medicine, Seattle. He has no conflicts. 

Title
Dr. John M. Inadomi, AGAF, of the University of Washington provides a perspective on the recommendation to start screening younger
Dr. John M. Inadomi, AGAF, of the University of Washington provides a perspective on the recommendation to start screening younger

The American Cancer Society recommends all U.S. adults at average risk of colorectal cancer (CRC) undergo screening starting at age 45 years.

That update to ACS recommendations is based on an increasing burden of CRC in younger individuals, microsimulation modeling results, and a “reasonable expectation” that screening tests will perform as well in adults aged 45-49 years as they do in older adults, members of the ACS Guideline Development Group said in the guideline, which was published in CA: A Cancer Journal for Clinicians.

Starting screening at age 45 contrasts with recommendations from the U.S. Preventive Services Task Force (USPSTF), which in 2016 gave an “A” recommendation for CRC screening from 50 to 75 years of age. At the time, the USPSTF noted a modest increase in life-years gained by starting earlier, based on microsimulation modeling. But it concluded that available evidence best supported starting at age 50.

The updated ACS guidelines are based in part on a modeling study that the authors say extends the previous analysis conducted for the USPSTF.

“The recommendation places a high value on the potential years of life saved, addresses anticipated rising incidence going forward, and is expected to contribute to the reduction in disparities in incidence before age 50 years in some racial groups,” the ACS guideline authors added.

The recommendation to start screening at age 45 is a “qualified” recommendation, the authors said, given the limitations of the current evidence base. Most studies to date have been focused on older individuals, in keeping with long-standing recommendations to start screening at age 50.

The move downward in screening age recommendation acknowledges one of the most “significant and disturbing” developments in CRC, the guideline’s authors said: the marked increase in incidence among younger individuals.

While CRC incidence and mortality have been declining in adults aged 55 years and older, recent studies cited in the ACS guideline document show a 51% increase in incidence from 1994 to 2014 – and an 11% increase in mortality from 2005 to 2015 – for adults younger than 55 years.

The current age-specific incidence rate for adults 45-49 years is 31.4 per 100,000, compared with 58.4 per 100,000 in adults 50-54 years. However, the ACS guideline authors said the higher rate in the older cohort is partly influenced by more frequent screening. “The true underlying risk in adults aged 45-49 years is likely closer to the risk in adults aged 50-54 years than the most recent age-specific rates would suggest,” they wrote.

Since patients in this age range have not been routinely screened before, the ACS recommendation is based on modeling. Now we need to analyze the outcomes of early screening to identify which patients will benefit most. Choices for screening include either a structural examination or a high-sensitivity stool-based test, according to the guideline, which doesn’t state a preference for any particular test.

The AGA, in their statement in response, noted that with CRC rates rising in people younger than age 50, it is appropriate to consider beginning routine screening at age 45. The statement continues “Since patients in this age range have not been previously routinely screened, the ACS recommendation is based on modeling. Now we need to analyze the outcomes of early screening to identify which patients will benefit most.”

 

 

In addition to Dr. Wolf, members of the ACS Guideline Development Group received no compensation.

SOURCE: Wolf AMD et al. CA Cancer J Clin. 2018 May 30. doi: 10.3322/caac.21457.

The American Cancer Society recommends all U.S. adults at average risk of colorectal cancer (CRC) undergo screening starting at age 45 years.

That update to ACS recommendations is based on an increasing burden of CRC in younger individuals, microsimulation modeling results, and a “reasonable expectation” that screening tests will perform as well in adults aged 45-49 years as they do in older adults, members of the ACS Guideline Development Group said in the guideline, which was published in CA: A Cancer Journal for Clinicians.

Starting screening at age 45 contrasts with recommendations from the U.S. Preventive Services Task Force (USPSTF), which in 2016 gave an “A” recommendation for CRC screening from 50 to 75 years of age. At the time, the USPSTF noted a modest increase in life-years gained by starting earlier, based on microsimulation modeling. But it concluded that available evidence best supported starting at age 50.

The updated ACS guidelines are based in part on a modeling study that the authors say extends the previous analysis conducted for the USPSTF.

“The recommendation places a high value on the potential years of life saved, addresses anticipated rising incidence going forward, and is expected to contribute to the reduction in disparities in incidence before age 50 years in some racial groups,” the ACS guideline authors added.

The recommendation to start screening at age 45 is a “qualified” recommendation, the authors said, given the limitations of the current evidence base. Most studies to date have been focused on older individuals, in keeping with long-standing recommendations to start screening at age 50.

The move downward in screening age recommendation acknowledges one of the most “significant and disturbing” developments in CRC, the guideline’s authors said: the marked increase in incidence among younger individuals.

While CRC incidence and mortality have been declining in adults aged 55 years and older, recent studies cited in the ACS guideline document show a 51% increase in incidence from 1994 to 2014 – and an 11% increase in mortality from 2005 to 2015 – for adults younger than 55 years.

The current age-specific incidence rate for adults 45-49 years is 31.4 per 100,000, compared with 58.4 per 100,000 in adults 50-54 years. However, the ACS guideline authors said the higher rate in the older cohort is partly influenced by more frequent screening. “The true underlying risk in adults aged 45-49 years is likely closer to the risk in adults aged 50-54 years than the most recent age-specific rates would suggest,” they wrote.

Since patients in this age range have not been routinely screened before, the ACS recommendation is based on modeling. Now we need to analyze the outcomes of early screening to identify which patients will benefit most. Choices for screening include either a structural examination or a high-sensitivity stool-based test, according to the guideline, which doesn’t state a preference for any particular test.

The AGA, in their statement in response, noted that with CRC rates rising in people younger than age 50, it is appropriate to consider beginning routine screening at age 45. The statement continues “Since patients in this age range have not been previously routinely screened, the ACS recommendation is based on modeling. Now we need to analyze the outcomes of early screening to identify which patients will benefit most.”

 

 

In addition to Dr. Wolf, members of the ACS Guideline Development Group received no compensation.

SOURCE: Wolf AMD et al. CA Cancer J Clin. 2018 May 30. doi: 10.3322/caac.21457.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CA: A CANCER JOURNAL FOR CLINICIANS

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

When is denosumab an option in myeloma?

Article Type
Changed
Fri, 01/04/2019 - 10:26

 

Denosumab may be preferable to bisphosphonates in myeloma patients in specific scenarios, G. David Roodman, MD, PhD, reported at the annual meeting of the American Society of Clinical Oncology.

“We use denosumab in patients with compromised renal function,” said Dr. Roodman, director of the Division of Hematology-Oncology at Indiana University, Indianapolis, noting one such scenario. That use of denosumab echoes recently published ASCO guidelines on bone-modifying therapy.

Andrew D. Bowser/MDedge News
Dr. G. David Roodman
Those guidelines recommend pamidronate or zoledronic acid for patients with active symptomatic myeloma who need systemic therapy. They describe denosumab as an “alternative” based on recent noninferiority data; however, they add that denosumab is associated with less renal toxicity compared with zoledronic acid or pamidronate, and thus, “may be preferred” in that setting.

The second scenario for denosumab use is in patients who aren’t tolerating bisphosphonates: “We switch them from zoledronic acid to pamidronate, and they still have terrible acute phase reactions,” Dr. Roodman said.

Dr. Roodman’s comments on use of denosumab were in response to an audience question about when he would use denosumab, given the considerable cost difference between the RANK ligand inhibitor and bisphosphonates.

The recent ASCO guidelines, of which Dr. Roodman is a coauthor, state that denosumab “is more expensive than zoledronic acid or pamidronate and must be considered in treatment decisions.”

Previously, ASCO guidelines recommended use of intravenous bisphosphonates for patients with myeloma and evidence of bone disease. Based on consideration of new evidence, the guideline authors eliminated the requirement for evidence of bone disease and added denosumab as an alternative treatment choice.

 

 


The addition of denosumab was based in part on results of a recent randomized phase 3 trial that comprised 1,718 myeloma patients who were treated with either denosumab or zoledronic acid.

The primary endpoint, time to first on-study skeletal-related event, was evaluated after 676 skeletal-related events had accrued on study. The investigators found no difference in time to first event (hazard ratio [HR], 0.98; 95% confidence interval, 0.85-1.14; P = 0.82).



Likewise, the secondary endpoint of overall survival showed no difference between arms (HR, 0.90; 95% CI, 0.70-1.16), though an exploratory analysis did suggest denosumab was superior on the endpoint of progression-free survival (HR, 0.82; 95% CI, 0.68-0.99).

The ASCO guidelines also recommend that clinicians consider less-frequent dosing in patients with responsive or stable disease. That recommendation is based on results of two studies of less-frequent dosing prompted by concerns over the risk of osteonecrosis of the jaw, an uncommon but potentially serious complication associated with bone-modifying agents.

 

 


Both studies suggested every-3-months dosing of zoledronic acid could be effective. However, Dr. Roodman noted that both studies had limitations that need to be considered, including small numbers of myeloma patients, limited duration of therapy studied, and a high dropout rate in the case of one study. Due to those limitations, “it’s very difficult to draw conclusions about this today,” Dr. Roodman said.

Dr. Roodman reported that he had a consulting or advisory role with Amgen.

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

 

Denosumab may be preferable to bisphosphonates in myeloma patients in specific scenarios, G. David Roodman, MD, PhD, reported at the annual meeting of the American Society of Clinical Oncology.

“We use denosumab in patients with compromised renal function,” said Dr. Roodman, director of the Division of Hematology-Oncology at Indiana University, Indianapolis, noting one such scenario. That use of denosumab echoes recently published ASCO guidelines on bone-modifying therapy.

Andrew D. Bowser/MDedge News
Dr. G. David Roodman
Those guidelines recommend pamidronate or zoledronic acid for patients with active symptomatic myeloma who need systemic therapy. They describe denosumab as an “alternative” based on recent noninferiority data; however, they add that denosumab is associated with less renal toxicity compared with zoledronic acid or pamidronate, and thus, “may be preferred” in that setting.

The second scenario for denosumab use is in patients who aren’t tolerating bisphosphonates: “We switch them from zoledronic acid to pamidronate, and they still have terrible acute phase reactions,” Dr. Roodman said.

Dr. Roodman’s comments on use of denosumab were in response to an audience question about when he would use denosumab, given the considerable cost difference between the RANK ligand inhibitor and bisphosphonates.

The recent ASCO guidelines, of which Dr. Roodman is a coauthor, state that denosumab “is more expensive than zoledronic acid or pamidronate and must be considered in treatment decisions.”

Previously, ASCO guidelines recommended use of intravenous bisphosphonates for patients with myeloma and evidence of bone disease. Based on consideration of new evidence, the guideline authors eliminated the requirement for evidence of bone disease and added denosumab as an alternative treatment choice.

 

 


The addition of denosumab was based in part on results of a recent randomized phase 3 trial that comprised 1,718 myeloma patients who were treated with either denosumab or zoledronic acid.

The primary endpoint, time to first on-study skeletal-related event, was evaluated after 676 skeletal-related events had accrued on study. The investigators found no difference in time to first event (hazard ratio [HR], 0.98; 95% confidence interval, 0.85-1.14; P = 0.82).



Likewise, the secondary endpoint of overall survival showed no difference between arms (HR, 0.90; 95% CI, 0.70-1.16), though an exploratory analysis did suggest denosumab was superior on the endpoint of progression-free survival (HR, 0.82; 95% CI, 0.68-0.99).

The ASCO guidelines also recommend that clinicians consider less-frequent dosing in patients with responsive or stable disease. That recommendation is based on results of two studies of less-frequent dosing prompted by concerns over the risk of osteonecrosis of the jaw, an uncommon but potentially serious complication associated with bone-modifying agents.

 

 


Both studies suggested every-3-months dosing of zoledronic acid could be effective. However, Dr. Roodman noted that both studies had limitations that need to be considered, including small numbers of myeloma patients, limited duration of therapy studied, and a high dropout rate in the case of one study. Due to those limitations, “it’s very difficult to draw conclusions about this today,” Dr. Roodman said.

Dr. Roodman reported that he had a consulting or advisory role with Amgen.

 

Denosumab may be preferable to bisphosphonates in myeloma patients in specific scenarios, G. David Roodman, MD, PhD, reported at the annual meeting of the American Society of Clinical Oncology.

“We use denosumab in patients with compromised renal function,” said Dr. Roodman, director of the Division of Hematology-Oncology at Indiana University, Indianapolis, noting one such scenario. That use of denosumab echoes recently published ASCO guidelines on bone-modifying therapy.

Andrew D. Bowser/MDedge News
Dr. G. David Roodman
Those guidelines recommend pamidronate or zoledronic acid for patients with active symptomatic myeloma who need systemic therapy. They describe denosumab as an “alternative” based on recent noninferiority data; however, they add that denosumab is associated with less renal toxicity compared with zoledronic acid or pamidronate, and thus, “may be preferred” in that setting.

The second scenario for denosumab use is in patients who aren’t tolerating bisphosphonates: “We switch them from zoledronic acid to pamidronate, and they still have terrible acute phase reactions,” Dr. Roodman said.

Dr. Roodman’s comments on use of denosumab were in response to an audience question about when he would use denosumab, given the considerable cost difference between the RANK ligand inhibitor and bisphosphonates.

The recent ASCO guidelines, of which Dr. Roodman is a coauthor, state that denosumab “is more expensive than zoledronic acid or pamidronate and must be considered in treatment decisions.”

Previously, ASCO guidelines recommended use of intravenous bisphosphonates for patients with myeloma and evidence of bone disease. Based on consideration of new evidence, the guideline authors eliminated the requirement for evidence of bone disease and added denosumab as an alternative treatment choice.

 

 


The addition of denosumab was based in part on results of a recent randomized phase 3 trial that comprised 1,718 myeloma patients who were treated with either denosumab or zoledronic acid.

The primary endpoint, time to first on-study skeletal-related event, was evaluated after 676 skeletal-related events had accrued on study. The investigators found no difference in time to first event (hazard ratio [HR], 0.98; 95% confidence interval, 0.85-1.14; P = 0.82).



Likewise, the secondary endpoint of overall survival showed no difference between arms (HR, 0.90; 95% CI, 0.70-1.16), though an exploratory analysis did suggest denosumab was superior on the endpoint of progression-free survival (HR, 0.82; 95% CI, 0.68-0.99).

The ASCO guidelines also recommend that clinicians consider less-frequent dosing in patients with responsive or stable disease. That recommendation is based on results of two studies of less-frequent dosing prompted by concerns over the risk of osteonecrosis of the jaw, an uncommon but potentially serious complication associated with bone-modifying agents.

 

 


Both studies suggested every-3-months dosing of zoledronic acid could be effective. However, Dr. Roodman noted that both studies had limitations that need to be considered, including small numbers of myeloma patients, limited duration of therapy studied, and a high dropout rate in the case of one study. Due to those limitations, “it’s very difficult to draw conclusions about this today,” Dr. Roodman said.

Dr. Roodman reported that he had a consulting or advisory role with Amgen.

Publications
Publications
Topics
Article Type
Sections
Article Source

EXPERT ANALYSIS FROM ASCO 2018

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Ruxolitinib overcame lenalidomide resistance in myeloma

Article Type
Changed
Fri, 01/04/2019 - 10:26

 

– The JAK 1/2 inhibitor ruxolitinib (Jakafi), in combination with lenalidomide and methylprednisolone, overcame resistance to lenalidomide in about half of 28 heavily pre-treated patients with relapsed/refractory multiple myeloma, based on phase I trial results presented at the annual meeting of the American Society of Clinical Oncology (ASCO).

The clinical trial is the first to demonstrate the activity of a JAK inhibitor in the treatment of myeloma patients, according to investigator James R. Berenson, MD, medical and scientific director for the Institute for Myeloma & Bone Cancer Research (IMBCR), West Hollywood, Calif.

Andrew Bowser/MDedge News
Dr. James R. Berenson
The all-oral combination was “very well tolerated” with few grade 3 or greater adverse events, including cytopenias, Dr. Berenson reported. All patients who had a response to the ruxolitinib, lenalidomide, and methylprednisolone combination were lenalidomide refractory.

“These promising results have led to the expansion of the current clinical trial and provide the basis for exploration of this and other JAK inhibitor-containing combinations for treating patients with myeloma and other malignant diseases,” he added.

The dose-escalation study enrolled 28 patients with relapsed/refractory multiple myeloma who had previous treatment with lenalidomide/steroids and a proteasome inhibitor. Subjects received ruxolitinib twice daily continuously, lenalidomide daily on days 1-21 of a 28-day cycle, and methylprednisolone orally every other day. A traditional 3+3 dose escalation design was used to enroll subjects in four cohorts. In DL0, patients received ruxolitinib 5 mg, lenalidomide 5 mg, and methylprednisolone 40 mg. In DL+1 and +2, both doses of lenalidomide and methylprednisolone remained unchanged, and ruxolitinib was escalated to 10 and 15 mg, respectively. In DL+3, lenalidomide was escalated to 10 mg with methylprednisolone unchanged and ruxolitinib at 15 mg. A total of 19 patients were treated at the highest dose level, which was ruxolitinib 15 mg twice daily on days 1-28, lenalidomide 10 mg daily on days 1-21, and methylprednisolone 40 mg every other day.

The overall response rate was 39% (10 of 26 evaluable patients), Dr. Berenson reported. The clinical benefit rate was 50% (13 of 26 patients), with a median duration of response of 5.6 months in that group.

There were no dose-limiting toxicities. Grade 3 toxicities reported included thrombocytopenia in 11% (3 patients, gastrointestinal bleeding in 11% (3 patients), and anemia in 7% (2 patients).

 

 


These results are encouraging, though challenges remain, said Craig Hofmeister, MD, MPH, of Winship Cancer Institute, Emory University, Atlanta.

“Clearly in these patients who are 100% lenalidomide refractory, the overall response rate of anything greater than or close to 20, 30, 40% is very appealing,” Dr. Hofmeister said in an ASCO presentation discussing the results of this trial.

The usual rationale for JAK inhibition is targeting of the bone microenvironment, but the microenvironment is a formidable opponent, Dr. Hofmeister said in his presentation.

“That’s an uphill battle,” he said. “There is an upcoming carfilzomib and ruxolitinib trial in multiple myeloma moving forward, and I’d be excited to see” the results.

The study (NCT03110822) was sponsored by Oncotherapeutics in collaboration with Incyte, the maker of ruxolitinib (Jakafi). Dr. Berenson, the presenting author, had disclosures related to Incyte, as well as Amgen, Bristol-Myers Squibb, Celgene, Janssen, Takeda, and OncoTracker.

SOURCE: Berenson JR, et al. J Clin Oncol 36, 2018 (suppl; abstr 8005).

Publications
Topics
Sections

 

– The JAK 1/2 inhibitor ruxolitinib (Jakafi), in combination with lenalidomide and methylprednisolone, overcame resistance to lenalidomide in about half of 28 heavily pre-treated patients with relapsed/refractory multiple myeloma, based on phase I trial results presented at the annual meeting of the American Society of Clinical Oncology (ASCO).

The clinical trial is the first to demonstrate the activity of a JAK inhibitor in the treatment of myeloma patients, according to investigator James R. Berenson, MD, medical and scientific director for the Institute for Myeloma & Bone Cancer Research (IMBCR), West Hollywood, Calif.

Andrew Bowser/MDedge News
Dr. James R. Berenson
The all-oral combination was “very well tolerated” with few grade 3 or greater adverse events, including cytopenias, Dr. Berenson reported. All patients who had a response to the ruxolitinib, lenalidomide, and methylprednisolone combination were lenalidomide refractory.

“These promising results have led to the expansion of the current clinical trial and provide the basis for exploration of this and other JAK inhibitor-containing combinations for treating patients with myeloma and other malignant diseases,” he added.

The dose-escalation study enrolled 28 patients with relapsed/refractory multiple myeloma who had previous treatment with lenalidomide/steroids and a proteasome inhibitor. Subjects received ruxolitinib twice daily continuously, lenalidomide daily on days 1-21 of a 28-day cycle, and methylprednisolone orally every other day. A traditional 3+3 dose escalation design was used to enroll subjects in four cohorts. In DL0, patients received ruxolitinib 5 mg, lenalidomide 5 mg, and methylprednisolone 40 mg. In DL+1 and +2, both doses of lenalidomide and methylprednisolone remained unchanged, and ruxolitinib was escalated to 10 and 15 mg, respectively. In DL+3, lenalidomide was escalated to 10 mg with methylprednisolone unchanged and ruxolitinib at 15 mg. A total of 19 patients were treated at the highest dose level, which was ruxolitinib 15 mg twice daily on days 1-28, lenalidomide 10 mg daily on days 1-21, and methylprednisolone 40 mg every other day.

The overall response rate was 39% (10 of 26 evaluable patients), Dr. Berenson reported. The clinical benefit rate was 50% (13 of 26 patients), with a median duration of response of 5.6 months in that group.

There were no dose-limiting toxicities. Grade 3 toxicities reported included thrombocytopenia in 11% (3 patients, gastrointestinal bleeding in 11% (3 patients), and anemia in 7% (2 patients).

 

 


These results are encouraging, though challenges remain, said Craig Hofmeister, MD, MPH, of Winship Cancer Institute, Emory University, Atlanta.

“Clearly in these patients who are 100% lenalidomide refractory, the overall response rate of anything greater than or close to 20, 30, 40% is very appealing,” Dr. Hofmeister said in an ASCO presentation discussing the results of this trial.

The usual rationale for JAK inhibition is targeting of the bone microenvironment, but the microenvironment is a formidable opponent, Dr. Hofmeister said in his presentation.

“That’s an uphill battle,” he said. “There is an upcoming carfilzomib and ruxolitinib trial in multiple myeloma moving forward, and I’d be excited to see” the results.

The study (NCT03110822) was sponsored by Oncotherapeutics in collaboration with Incyte, the maker of ruxolitinib (Jakafi). Dr. Berenson, the presenting author, had disclosures related to Incyte, as well as Amgen, Bristol-Myers Squibb, Celgene, Janssen, Takeda, and OncoTracker.

SOURCE: Berenson JR, et al. J Clin Oncol 36, 2018 (suppl; abstr 8005).

 

– The JAK 1/2 inhibitor ruxolitinib (Jakafi), in combination with lenalidomide and methylprednisolone, overcame resistance to lenalidomide in about half of 28 heavily pre-treated patients with relapsed/refractory multiple myeloma, based on phase I trial results presented at the annual meeting of the American Society of Clinical Oncology (ASCO).

The clinical trial is the first to demonstrate the activity of a JAK inhibitor in the treatment of myeloma patients, according to investigator James R. Berenson, MD, medical and scientific director for the Institute for Myeloma & Bone Cancer Research (IMBCR), West Hollywood, Calif.

Andrew Bowser/MDedge News
Dr. James R. Berenson
The all-oral combination was “very well tolerated” with few grade 3 or greater adverse events, including cytopenias, Dr. Berenson reported. All patients who had a response to the ruxolitinib, lenalidomide, and methylprednisolone combination were lenalidomide refractory.

“These promising results have led to the expansion of the current clinical trial and provide the basis for exploration of this and other JAK inhibitor-containing combinations for treating patients with myeloma and other malignant diseases,” he added.

The dose-escalation study enrolled 28 patients with relapsed/refractory multiple myeloma who had previous treatment with lenalidomide/steroids and a proteasome inhibitor. Subjects received ruxolitinib twice daily continuously, lenalidomide daily on days 1-21 of a 28-day cycle, and methylprednisolone orally every other day. A traditional 3+3 dose escalation design was used to enroll subjects in four cohorts. In DL0, patients received ruxolitinib 5 mg, lenalidomide 5 mg, and methylprednisolone 40 mg. In DL+1 and +2, both doses of lenalidomide and methylprednisolone remained unchanged, and ruxolitinib was escalated to 10 and 15 mg, respectively. In DL+3, lenalidomide was escalated to 10 mg with methylprednisolone unchanged and ruxolitinib at 15 mg. A total of 19 patients were treated at the highest dose level, which was ruxolitinib 15 mg twice daily on days 1-28, lenalidomide 10 mg daily on days 1-21, and methylprednisolone 40 mg every other day.

The overall response rate was 39% (10 of 26 evaluable patients), Dr. Berenson reported. The clinical benefit rate was 50% (13 of 26 patients), with a median duration of response of 5.6 months in that group.

There were no dose-limiting toxicities. Grade 3 toxicities reported included thrombocytopenia in 11% (3 patients, gastrointestinal bleeding in 11% (3 patients), and anemia in 7% (2 patients).

 

 


These results are encouraging, though challenges remain, said Craig Hofmeister, MD, MPH, of Winship Cancer Institute, Emory University, Atlanta.

“Clearly in these patients who are 100% lenalidomide refractory, the overall response rate of anything greater than or close to 20, 30, 40% is very appealing,” Dr. Hofmeister said in an ASCO presentation discussing the results of this trial.

The usual rationale for JAK inhibition is targeting of the bone microenvironment, but the microenvironment is a formidable opponent, Dr. Hofmeister said in his presentation.

“That’s an uphill battle,” he said. “There is an upcoming carfilzomib and ruxolitinib trial in multiple myeloma moving forward, and I’d be excited to see” the results.

The study (NCT03110822) was sponsored by Oncotherapeutics in collaboration with Incyte, the maker of ruxolitinib (Jakafi). Dr. Berenson, the presenting author, had disclosures related to Incyte, as well as Amgen, Bristol-Myers Squibb, Celgene, Janssen, Takeda, and OncoTracker.

SOURCE: Berenson JR, et al. J Clin Oncol 36, 2018 (suppl; abstr 8005).

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASCO 2018

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: The JAK 1/2 inhibitor ruxolitinib, in combination with lenalidomide and methylprednisolone, overcame resistance to lenalidomide in about half of heavily pre-treated patients with relapsed/refractory multiple myeloma in a phase I trial.

Major finding: The overall response rate was 39%, and the clinical benefit rate was 50% (13 of 26 patients).

Study details: A phase 1 study including 28 patients with relapsed/refractory multiple myeloma who had previous treatment with lenalidomide/steroids and a proteasome inhibitor.

Disclosures: Dr. Berenson, the presenting author, had disclosures related to Amgen, Bristol-Myers Squibb, Celgene, Incyte, Janssen, Takeda, and OncoTracker.

Source: Berenson JR, et al. J Clin Oncol 36, 2018 (suppl; abstr 8005).

Disqus Comments
Default
Use ProPublica