BET inhibitor exhibits activity in myelofibrosis

Article Type
Changed
Tue, 01/14/2020 - 10:44

– The BET inhibitor CPI-0610, given alone or in combination with ruxolitinib, has demonstrated activity in a phase 2 trial of patients with relapsed/refractory myelofibrosis (MF).

Dr. John Mascarenhas of  Icahn School of Medicine at Mount Sinai, New York
Jennifer Smith/MDedge News
Dr. John Mascarenhas

Responses were best among transfusion-dependent patients who received CPI-0610 and ruxolitinib. All but one of these patients experienced symptom improvement.

In the monotherapy group, results were best among transfusion-independent patients. All of these patients had an improvement in Patient Global Impression of Change (PGIC) score, and most had a 50% or greater improvement in total symptom score (TSS).

John Mascarenhas, MD, of the Icahn School of Medicine at Mount Sinai, New York, presented these results from the phase 2 MANIFEST trial (NCT02158858) at the annual meeting of the American Society of Hematology.

Dr. Mascarenhas presented data on 90 patients – 59 with primary MF, 16 with post–polycythemia vera MF, 13 with post–essential thrombocythemia MF, and 2 whose type of MF was unknown. At baseline, the patients’ median age was 69 years and 76.7% of patients had received at least 6 months of ruxolitinib treatment.

Of the 36 patients who received CPI-0610 monotherapy, 34 were still receiving the treatment as of Oct. 17, 2019. Of the 54 patients treated with CPI-0610 and ruxolitinib, 41 were still receiving the combination at that time. The median duration of treatment was 11.3 weeks in the monotherapy arm and 25.9 weeks in the combination arm. Responses were assessed at 24 weeks.

“CPI-0610 monotherapy or added on to ruxolitinib in a relapsed/refractory MF population demonstrated antitumor activity, as evidenced by spleen and symptom improvement,” Dr. Mascarenhas said. “Symptom responses were observed in a majority of patients. We treated these patients, and they felt much better. It was impressive.”

Efficacy of monotherapy

There were two evaluable patients who had been transfusion dependent at baseline and received CPI-0610 monotherapy for at least 24 weeks. Neither patient achieved transfusion independence, and neither had a spleen volume reduction of at least 35% (SVR35). One patient had a 50% or greater improvement in TSS, and one had an improvement in PGIC score.

There were seven evaluable patients who were transfusion independent at baseline and received CPI-0610 for at least 24 weeks. None of these patients had an SVR35 response, three of five evaluable patients had a 50% or greater improvement in TSS, and all seven had an improvement in PGIC score.
 

Efficacy of the combination

There were 14 patients who had been transfusion dependent at baseline and received CPI-0610 and ruxolitinib for at least 24 weeks. Six of these patients had become transfusion independent at week 24.

Among the patients who were transfusion dependent at baseline, 25% (3/12) had an SVR35 response at week 24, 54% (7/13) had a 50% or greater improvement in TSS, and 75% (9/12) had an improvement in PGIC score.

There were 13 evaluable patients who were transfusion independent at baseline and received CPI-0610 and ruxolitinib for at least 24 weeks. None of these patients had an SVR35 response, 38% had a 50% or greater improvement in TSS, and 69% had an improvement in PGIC score.
 

 

 

Safety

“CPI-0610 monotherapy or as an add-on to [ruxolitinib] was generally well tolerated,” Dr. Mascarenhas said. “Thrombocytopenia was asymptomatic, generally reversible, and manageable. There were no other unanticipated safety concerns.”

All 90 patients were evaluable for safety. Hematologic adverse events included thrombocytopenia (23.3%) and anemia (8.9%).

The most common nonhematologic adverse events were diarrhea (32.2%), nausea (22.2%), cough (16.7%), fatigue (14.4%), vomiting (14.4%), and upper respiratory tract infection (14.4%).

Eight patients (8.9%) experienced grade 4 adverse events, but all events resolved. Four events occurred in the monotherapy arm, and one (rash) required dose interruption. Of the four events in the combination arm, one (anemia) was considered treatment related.

There were three fatal adverse events – acute kidney injury, traumatic subdural hematoma, and brain stem hemorrhage. None of these events were considered related to CPI-0610.

Based on these preliminary results, the cohort of transfusion-dependent patients receiving CPI-0610 and ruxolitinib has been expanded. A cohort of ruxolitinib-naive patients receiving CPI-0610 and ruxolitinib has been expanded as well.

The MANIFEST trial is funded by Constellation Pharmaceuticals in collaboration with the Leukemia & Lymphoma Society. Dr. Mascarenhas reported relationships with Incyte, Janssen, CTI Biopharma, Novartis, Roche, Merck, Celgene, Promedior, Merus, and PharmaEssentia.

SOURCE: Mascarenhas J et al. ASH 2019, Abstract 670.
 

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

– The BET inhibitor CPI-0610, given alone or in combination with ruxolitinib, has demonstrated activity in a phase 2 trial of patients with relapsed/refractory myelofibrosis (MF).

Dr. John Mascarenhas of  Icahn School of Medicine at Mount Sinai, New York
Jennifer Smith/MDedge News
Dr. John Mascarenhas

Responses were best among transfusion-dependent patients who received CPI-0610 and ruxolitinib. All but one of these patients experienced symptom improvement.

In the monotherapy group, results were best among transfusion-independent patients. All of these patients had an improvement in Patient Global Impression of Change (PGIC) score, and most had a 50% or greater improvement in total symptom score (TSS).

John Mascarenhas, MD, of the Icahn School of Medicine at Mount Sinai, New York, presented these results from the phase 2 MANIFEST trial (NCT02158858) at the annual meeting of the American Society of Hematology.

Dr. Mascarenhas presented data on 90 patients – 59 with primary MF, 16 with post–polycythemia vera MF, 13 with post–essential thrombocythemia MF, and 2 whose type of MF was unknown. At baseline, the patients’ median age was 69 years and 76.7% of patients had received at least 6 months of ruxolitinib treatment.

Of the 36 patients who received CPI-0610 monotherapy, 34 were still receiving the treatment as of Oct. 17, 2019. Of the 54 patients treated with CPI-0610 and ruxolitinib, 41 were still receiving the combination at that time. The median duration of treatment was 11.3 weeks in the monotherapy arm and 25.9 weeks in the combination arm. Responses were assessed at 24 weeks.

“CPI-0610 monotherapy or added on to ruxolitinib in a relapsed/refractory MF population demonstrated antitumor activity, as evidenced by spleen and symptom improvement,” Dr. Mascarenhas said. “Symptom responses were observed in a majority of patients. We treated these patients, and they felt much better. It was impressive.”

Efficacy of monotherapy

There were two evaluable patients who had been transfusion dependent at baseline and received CPI-0610 monotherapy for at least 24 weeks. Neither patient achieved transfusion independence, and neither had a spleen volume reduction of at least 35% (SVR35). One patient had a 50% or greater improvement in TSS, and one had an improvement in PGIC score.

There were seven evaluable patients who were transfusion independent at baseline and received CPI-0610 for at least 24 weeks. None of these patients had an SVR35 response, three of five evaluable patients had a 50% or greater improvement in TSS, and all seven had an improvement in PGIC score.
 

Efficacy of the combination

There were 14 patients who had been transfusion dependent at baseline and received CPI-0610 and ruxolitinib for at least 24 weeks. Six of these patients had become transfusion independent at week 24.

Among the patients who were transfusion dependent at baseline, 25% (3/12) had an SVR35 response at week 24, 54% (7/13) had a 50% or greater improvement in TSS, and 75% (9/12) had an improvement in PGIC score.

There were 13 evaluable patients who were transfusion independent at baseline and received CPI-0610 and ruxolitinib for at least 24 weeks. None of these patients had an SVR35 response, 38% had a 50% or greater improvement in TSS, and 69% had an improvement in PGIC score.
 

 

 

Safety

“CPI-0610 monotherapy or as an add-on to [ruxolitinib] was generally well tolerated,” Dr. Mascarenhas said. “Thrombocytopenia was asymptomatic, generally reversible, and manageable. There were no other unanticipated safety concerns.”

All 90 patients were evaluable for safety. Hematologic adverse events included thrombocytopenia (23.3%) and anemia (8.9%).

The most common nonhematologic adverse events were diarrhea (32.2%), nausea (22.2%), cough (16.7%), fatigue (14.4%), vomiting (14.4%), and upper respiratory tract infection (14.4%).

Eight patients (8.9%) experienced grade 4 adverse events, but all events resolved. Four events occurred in the monotherapy arm, and one (rash) required dose interruption. Of the four events in the combination arm, one (anemia) was considered treatment related.

There were three fatal adverse events – acute kidney injury, traumatic subdural hematoma, and brain stem hemorrhage. None of these events were considered related to CPI-0610.

Based on these preliminary results, the cohort of transfusion-dependent patients receiving CPI-0610 and ruxolitinib has been expanded. A cohort of ruxolitinib-naive patients receiving CPI-0610 and ruxolitinib has been expanded as well.

The MANIFEST trial is funded by Constellation Pharmaceuticals in collaboration with the Leukemia & Lymphoma Society. Dr. Mascarenhas reported relationships with Incyte, Janssen, CTI Biopharma, Novartis, Roche, Merck, Celgene, Promedior, Merus, and PharmaEssentia.

SOURCE: Mascarenhas J et al. ASH 2019, Abstract 670.
 

– The BET inhibitor CPI-0610, given alone or in combination with ruxolitinib, has demonstrated activity in a phase 2 trial of patients with relapsed/refractory myelofibrosis (MF).

Dr. John Mascarenhas of  Icahn School of Medicine at Mount Sinai, New York
Jennifer Smith/MDedge News
Dr. John Mascarenhas

Responses were best among transfusion-dependent patients who received CPI-0610 and ruxolitinib. All but one of these patients experienced symptom improvement.

In the monotherapy group, results were best among transfusion-independent patients. All of these patients had an improvement in Patient Global Impression of Change (PGIC) score, and most had a 50% or greater improvement in total symptom score (TSS).

John Mascarenhas, MD, of the Icahn School of Medicine at Mount Sinai, New York, presented these results from the phase 2 MANIFEST trial (NCT02158858) at the annual meeting of the American Society of Hematology.

Dr. Mascarenhas presented data on 90 patients – 59 with primary MF, 16 with post–polycythemia vera MF, 13 with post–essential thrombocythemia MF, and 2 whose type of MF was unknown. At baseline, the patients’ median age was 69 years and 76.7% of patients had received at least 6 months of ruxolitinib treatment.

Of the 36 patients who received CPI-0610 monotherapy, 34 were still receiving the treatment as of Oct. 17, 2019. Of the 54 patients treated with CPI-0610 and ruxolitinib, 41 were still receiving the combination at that time. The median duration of treatment was 11.3 weeks in the monotherapy arm and 25.9 weeks in the combination arm. Responses were assessed at 24 weeks.

“CPI-0610 monotherapy or added on to ruxolitinib in a relapsed/refractory MF population demonstrated antitumor activity, as evidenced by spleen and symptom improvement,” Dr. Mascarenhas said. “Symptom responses were observed in a majority of patients. We treated these patients, and they felt much better. It was impressive.”

Efficacy of monotherapy

There were two evaluable patients who had been transfusion dependent at baseline and received CPI-0610 monotherapy for at least 24 weeks. Neither patient achieved transfusion independence, and neither had a spleen volume reduction of at least 35% (SVR35). One patient had a 50% or greater improvement in TSS, and one had an improvement in PGIC score.

There were seven evaluable patients who were transfusion independent at baseline and received CPI-0610 for at least 24 weeks. None of these patients had an SVR35 response, three of five evaluable patients had a 50% or greater improvement in TSS, and all seven had an improvement in PGIC score.
 

Efficacy of the combination

There were 14 patients who had been transfusion dependent at baseline and received CPI-0610 and ruxolitinib for at least 24 weeks. Six of these patients had become transfusion independent at week 24.

Among the patients who were transfusion dependent at baseline, 25% (3/12) had an SVR35 response at week 24, 54% (7/13) had a 50% or greater improvement in TSS, and 75% (9/12) had an improvement in PGIC score.

There were 13 evaluable patients who were transfusion independent at baseline and received CPI-0610 and ruxolitinib for at least 24 weeks. None of these patients had an SVR35 response, 38% had a 50% or greater improvement in TSS, and 69% had an improvement in PGIC score.
 

 

 

Safety

“CPI-0610 monotherapy or as an add-on to [ruxolitinib] was generally well tolerated,” Dr. Mascarenhas said. “Thrombocytopenia was asymptomatic, generally reversible, and manageable. There were no other unanticipated safety concerns.”

All 90 patients were evaluable for safety. Hematologic adverse events included thrombocytopenia (23.3%) and anemia (8.9%).

The most common nonhematologic adverse events were diarrhea (32.2%), nausea (22.2%), cough (16.7%), fatigue (14.4%), vomiting (14.4%), and upper respiratory tract infection (14.4%).

Eight patients (8.9%) experienced grade 4 adverse events, but all events resolved. Four events occurred in the monotherapy arm, and one (rash) required dose interruption. Of the four events in the combination arm, one (anemia) was considered treatment related.

There were three fatal adverse events – acute kidney injury, traumatic subdural hematoma, and brain stem hemorrhage. None of these events were considered related to CPI-0610.

Based on these preliminary results, the cohort of transfusion-dependent patients receiving CPI-0610 and ruxolitinib has been expanded. A cohort of ruxolitinib-naive patients receiving CPI-0610 and ruxolitinib has been expanded as well.

The MANIFEST trial is funded by Constellation Pharmaceuticals in collaboration with the Leukemia & Lymphoma Society. Dr. Mascarenhas reported relationships with Incyte, Janssen, CTI Biopharma, Novartis, Roche, Merck, Celgene, Promedior, Merus, and PharmaEssentia.

SOURCE: Mascarenhas J et al. ASH 2019, Abstract 670.
 

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASH 2019

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Navitoclax may overcome ruxolitinib resistance in MF

Article Type
Changed
Thu, 01/09/2020 - 15:30

– Adding navitoclax to ruxolitinib improved responses in a phase 2 trial of patients with uncontrolled myelofibrosis (MF) and prolonged exposure to ruxolitinib.

Dr. Jacqueline S. Garcia, Dana-Farber Cancer Institute, Boston
Dr. Jacqueline S. Garcia

Navitoclax and ruxolitinib yielded “clinically meaningful” spleen responses, improved total symptom scores, and produced “encouraging” reductions in bone marrow fibrosis among patients with primary or secondary MF, according to Jacqueline S. Garcia, MD, of Dana-Farber Cancer Institute in Boston.

Dr. Garcia presented these results at the annual meeting of the American Society of Hematology.

Navitoclax binds with high affinity to BCL-XL, BCL-2, and BCL-W, Dr. Garcia noted. Preclinical research has shown that combining Janus kinase 2 (JAK2) inhibition with BCL-XL/BCL-2 inhibition has a synergistic cytotoxic effect on JAK2-mutated cells (Blood. 2009 Feb 12;113[7]:1522-5), and BCL-XL inhibition can overcome resistance to JAK2 inhibition (Cell Rep. 2013 Nov 27;5[4]:1047-59).

These findings led to the theory that combining navitoclax and ruxolitinib could overcome resistance to JAK2 inhibition in MF. The researchers tested this theory in a phase 2 trial (NCT03222609) of 34 MF patients.

The patients had primary MF (n = 16), post–polycythemia vera MF (n = 13), and post–essential thrombocythemia MF (n = 5). At baseline, their median age was 68 years (range 42-86 years), and 68% of them were men.

There were 33 patients with genetic testing results available. None of them were triple negative, 27 had JAK2 mutations, and 7 had CALR mutations. Roughly half of patients (n = 17) were classified as high molecular risk, with mutations in ASXL1, EZH2, IDH1/2, SRSF2, or U2AF1.
 

Treatment

All patients had received ruxolitinib for at least 12 weeks prior to their first dose of navitoclax. They had been receiving a stable dose of 10 mg or greater, twice daily, for at least 8 weeks. The median duration of prior ruxolitinib exposure was 21 months (range, 4-71 months).

On study, patients received navitoclax once daily plus the current stable dose of ruxolitinib (10 mg or greater twice daily). Navitoclax dosing started at 50 mg, but weekly dose escalation was allowed to a maximum daily dose of 300 mg. Treatment could continue until the loss of clinical benefit, unacceptable toxicity, or discontinuation.

There were 23 patients who received the maximum dose of navitoclax, and the median duration of navitoclax treatment was 330 days (range, 29-588 days). Of the 25 patients who started the study on a ruxolitinib dose higher than 10 mg twice daily, 22 had their dose reduced to 10 mg twice daily.

Nine patients discontinued study treatment – three due to adverse events, two due to progressive disease, and four for other reasons.
 

Efficacy

Navitoclax appears to overcome ruxolitinib resistance, Dr. Garcia said, citing improvements in spleen size, symptom scores, bone marrow fibrosis, white blood cell counts, and transfusion needs.

Thirty patients were evaluable for spleen response. At week 24, 30% had a spleen volume reduction of 35% or greater from baseline. At any time on study, 43% of patients had a spleen volume reduction of 35% or greater from baseline. More than half of patients (53%) had a resolution of palpable splenomegaly.

Eight of 32 patients (25%) had a reduction in bone marrow fibrosis, four with a one-grade reduction and four with a two-grade reduction.

Seventeen patients were evaluable for change in total symptom score. Eleven patients (65%) experienced a reduction in symptoms, and six (35%) had a 50% or greater reduction in symptoms. The median total symptom score was 12 (range, 0-30) at baseline and 7 (range, 0-23) at 24 weeks.

Patients had a significant reduction in white blood cells on study. The mean white blood cell reduction at week 24 was 25.8 x 109/L.

Patients’ hemoglobin levels remained stable over time, but a few patients had a decreased need for transfusions on study. Seven patients entered the study having received at least one unit of packed red blood cells in the prior 12 weeks. Four of them (57%) have had a transfusion-free period of at least 12 weeks on study.
 

 

 

Safety

“Navitoclax in combination with ruxolitinib appears to be well tolerated,” Dr. Garcia said.

She noted that treatment resulted in reduced platelet counts, but counts stabilized after 6-8 weeks. The mean platelet count was 232 x 109/L at baseline and 95 x 109/L at week 8.

In fact, the most common adverse event was thrombocytopenia, with any-grade thrombocytopenia occurring in 85% of patients and grade 3/4 occurring in 44%. One patient had grade 4 thrombocytopenia, but it was reversed by withholding treatment and subsequent dose modification.

Other common treatment-emergent adverse events were diarrhea (68%), fatigue (53%), nausea (35%), anemia (29%), dizziness (27%), confusion (27%), and vomiting (24%).

All 34 patients experienced at least one adverse event. Eight patients (24%) had serious adverse events, including anemia, pancytopenia, splenic infarction, upper abdominal pain, vomiting, chest pain, pneumonia, and abnormal liver function test.

One patient had a grade 5 adverse event – pneumonia – that was deemed unrelated to navitoclax.

This trial is sponsored by AbbVie. Dr. Garcia reported relationships with AbbVie, Genentech, and Pfizer.

SOURCE: Garcia JS et al. ASH 2019, Abstract 671.

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

– Adding navitoclax to ruxolitinib improved responses in a phase 2 trial of patients with uncontrolled myelofibrosis (MF) and prolonged exposure to ruxolitinib.

Dr. Jacqueline S. Garcia, Dana-Farber Cancer Institute, Boston
Dr. Jacqueline S. Garcia

Navitoclax and ruxolitinib yielded “clinically meaningful” spleen responses, improved total symptom scores, and produced “encouraging” reductions in bone marrow fibrosis among patients with primary or secondary MF, according to Jacqueline S. Garcia, MD, of Dana-Farber Cancer Institute in Boston.

Dr. Garcia presented these results at the annual meeting of the American Society of Hematology.

Navitoclax binds with high affinity to BCL-XL, BCL-2, and BCL-W, Dr. Garcia noted. Preclinical research has shown that combining Janus kinase 2 (JAK2) inhibition with BCL-XL/BCL-2 inhibition has a synergistic cytotoxic effect on JAK2-mutated cells (Blood. 2009 Feb 12;113[7]:1522-5), and BCL-XL inhibition can overcome resistance to JAK2 inhibition (Cell Rep. 2013 Nov 27;5[4]:1047-59).

These findings led to the theory that combining navitoclax and ruxolitinib could overcome resistance to JAK2 inhibition in MF. The researchers tested this theory in a phase 2 trial (NCT03222609) of 34 MF patients.

The patients had primary MF (n = 16), post–polycythemia vera MF (n = 13), and post–essential thrombocythemia MF (n = 5). At baseline, their median age was 68 years (range 42-86 years), and 68% of them were men.

There were 33 patients with genetic testing results available. None of them were triple negative, 27 had JAK2 mutations, and 7 had CALR mutations. Roughly half of patients (n = 17) were classified as high molecular risk, with mutations in ASXL1, EZH2, IDH1/2, SRSF2, or U2AF1.
 

Treatment

All patients had received ruxolitinib for at least 12 weeks prior to their first dose of navitoclax. They had been receiving a stable dose of 10 mg or greater, twice daily, for at least 8 weeks. The median duration of prior ruxolitinib exposure was 21 months (range, 4-71 months).

On study, patients received navitoclax once daily plus the current stable dose of ruxolitinib (10 mg or greater twice daily). Navitoclax dosing started at 50 mg, but weekly dose escalation was allowed to a maximum daily dose of 300 mg. Treatment could continue until the loss of clinical benefit, unacceptable toxicity, or discontinuation.

There were 23 patients who received the maximum dose of navitoclax, and the median duration of navitoclax treatment was 330 days (range, 29-588 days). Of the 25 patients who started the study on a ruxolitinib dose higher than 10 mg twice daily, 22 had their dose reduced to 10 mg twice daily.

Nine patients discontinued study treatment – three due to adverse events, two due to progressive disease, and four for other reasons.
 

Efficacy

Navitoclax appears to overcome ruxolitinib resistance, Dr. Garcia said, citing improvements in spleen size, symptom scores, bone marrow fibrosis, white blood cell counts, and transfusion needs.

Thirty patients were evaluable for spleen response. At week 24, 30% had a spleen volume reduction of 35% or greater from baseline. At any time on study, 43% of patients had a spleen volume reduction of 35% or greater from baseline. More than half of patients (53%) had a resolution of palpable splenomegaly.

Eight of 32 patients (25%) had a reduction in bone marrow fibrosis, four with a one-grade reduction and four with a two-grade reduction.

Seventeen patients were evaluable for change in total symptom score. Eleven patients (65%) experienced a reduction in symptoms, and six (35%) had a 50% or greater reduction in symptoms. The median total symptom score was 12 (range, 0-30) at baseline and 7 (range, 0-23) at 24 weeks.

Patients had a significant reduction in white blood cells on study. The mean white blood cell reduction at week 24 was 25.8 x 109/L.

Patients’ hemoglobin levels remained stable over time, but a few patients had a decreased need for transfusions on study. Seven patients entered the study having received at least one unit of packed red blood cells in the prior 12 weeks. Four of them (57%) have had a transfusion-free period of at least 12 weeks on study.
 

 

 

Safety

“Navitoclax in combination with ruxolitinib appears to be well tolerated,” Dr. Garcia said.

She noted that treatment resulted in reduced platelet counts, but counts stabilized after 6-8 weeks. The mean platelet count was 232 x 109/L at baseline and 95 x 109/L at week 8.

In fact, the most common adverse event was thrombocytopenia, with any-grade thrombocytopenia occurring in 85% of patients and grade 3/4 occurring in 44%. One patient had grade 4 thrombocytopenia, but it was reversed by withholding treatment and subsequent dose modification.

Other common treatment-emergent adverse events were diarrhea (68%), fatigue (53%), nausea (35%), anemia (29%), dizziness (27%), confusion (27%), and vomiting (24%).

All 34 patients experienced at least one adverse event. Eight patients (24%) had serious adverse events, including anemia, pancytopenia, splenic infarction, upper abdominal pain, vomiting, chest pain, pneumonia, and abnormal liver function test.

One patient had a grade 5 adverse event – pneumonia – that was deemed unrelated to navitoclax.

This trial is sponsored by AbbVie. Dr. Garcia reported relationships with AbbVie, Genentech, and Pfizer.

SOURCE: Garcia JS et al. ASH 2019, Abstract 671.

– Adding navitoclax to ruxolitinib improved responses in a phase 2 trial of patients with uncontrolled myelofibrosis (MF) and prolonged exposure to ruxolitinib.

Dr. Jacqueline S. Garcia, Dana-Farber Cancer Institute, Boston
Dr. Jacqueline S. Garcia

Navitoclax and ruxolitinib yielded “clinically meaningful” spleen responses, improved total symptom scores, and produced “encouraging” reductions in bone marrow fibrosis among patients with primary or secondary MF, according to Jacqueline S. Garcia, MD, of Dana-Farber Cancer Institute in Boston.

Dr. Garcia presented these results at the annual meeting of the American Society of Hematology.

Navitoclax binds with high affinity to BCL-XL, BCL-2, and BCL-W, Dr. Garcia noted. Preclinical research has shown that combining Janus kinase 2 (JAK2) inhibition with BCL-XL/BCL-2 inhibition has a synergistic cytotoxic effect on JAK2-mutated cells (Blood. 2009 Feb 12;113[7]:1522-5), and BCL-XL inhibition can overcome resistance to JAK2 inhibition (Cell Rep. 2013 Nov 27;5[4]:1047-59).

These findings led to the theory that combining navitoclax and ruxolitinib could overcome resistance to JAK2 inhibition in MF. The researchers tested this theory in a phase 2 trial (NCT03222609) of 34 MF patients.

The patients had primary MF (n = 16), post–polycythemia vera MF (n = 13), and post–essential thrombocythemia MF (n = 5). At baseline, their median age was 68 years (range 42-86 years), and 68% of them were men.

There were 33 patients with genetic testing results available. None of them were triple negative, 27 had JAK2 mutations, and 7 had CALR mutations. Roughly half of patients (n = 17) were classified as high molecular risk, with mutations in ASXL1, EZH2, IDH1/2, SRSF2, or U2AF1.
 

Treatment

All patients had received ruxolitinib for at least 12 weeks prior to their first dose of navitoclax. They had been receiving a stable dose of 10 mg or greater, twice daily, for at least 8 weeks. The median duration of prior ruxolitinib exposure was 21 months (range, 4-71 months).

On study, patients received navitoclax once daily plus the current stable dose of ruxolitinib (10 mg or greater twice daily). Navitoclax dosing started at 50 mg, but weekly dose escalation was allowed to a maximum daily dose of 300 mg. Treatment could continue until the loss of clinical benefit, unacceptable toxicity, or discontinuation.

There were 23 patients who received the maximum dose of navitoclax, and the median duration of navitoclax treatment was 330 days (range, 29-588 days). Of the 25 patients who started the study on a ruxolitinib dose higher than 10 mg twice daily, 22 had their dose reduced to 10 mg twice daily.

Nine patients discontinued study treatment – three due to adverse events, two due to progressive disease, and four for other reasons.
 

Efficacy

Navitoclax appears to overcome ruxolitinib resistance, Dr. Garcia said, citing improvements in spleen size, symptom scores, bone marrow fibrosis, white blood cell counts, and transfusion needs.

Thirty patients were evaluable for spleen response. At week 24, 30% had a spleen volume reduction of 35% or greater from baseline. At any time on study, 43% of patients had a spleen volume reduction of 35% or greater from baseline. More than half of patients (53%) had a resolution of palpable splenomegaly.

Eight of 32 patients (25%) had a reduction in bone marrow fibrosis, four with a one-grade reduction and four with a two-grade reduction.

Seventeen patients were evaluable for change in total symptom score. Eleven patients (65%) experienced a reduction in symptoms, and six (35%) had a 50% or greater reduction in symptoms. The median total symptom score was 12 (range, 0-30) at baseline and 7 (range, 0-23) at 24 weeks.

Patients had a significant reduction in white blood cells on study. The mean white blood cell reduction at week 24 was 25.8 x 109/L.

Patients’ hemoglobin levels remained stable over time, but a few patients had a decreased need for transfusions on study. Seven patients entered the study having received at least one unit of packed red blood cells in the prior 12 weeks. Four of them (57%) have had a transfusion-free period of at least 12 weeks on study.
 

 

 

Safety

“Navitoclax in combination with ruxolitinib appears to be well tolerated,” Dr. Garcia said.

She noted that treatment resulted in reduced platelet counts, but counts stabilized after 6-8 weeks. The mean platelet count was 232 x 109/L at baseline and 95 x 109/L at week 8.

In fact, the most common adverse event was thrombocytopenia, with any-grade thrombocytopenia occurring in 85% of patients and grade 3/4 occurring in 44%. One patient had grade 4 thrombocytopenia, but it was reversed by withholding treatment and subsequent dose modification.

Other common treatment-emergent adverse events were diarrhea (68%), fatigue (53%), nausea (35%), anemia (29%), dizziness (27%), confusion (27%), and vomiting (24%).

All 34 patients experienced at least one adverse event. Eight patients (24%) had serious adverse events, including anemia, pancytopenia, splenic infarction, upper abdominal pain, vomiting, chest pain, pneumonia, and abnormal liver function test.

One patient had a grade 5 adverse event – pneumonia – that was deemed unrelated to navitoclax.

This trial is sponsored by AbbVie. Dr. Garcia reported relationships with AbbVie, Genentech, and Pfizer.

SOURCE: Garcia JS et al. ASH 2019, Abstract 671.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASH 2019

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Genomic profiling of AML and MDS yields prognostic clues

Article Type
Changed
Tue, 01/14/2020 - 14:43

– A genome-wide study of blood and bone marrow samples from more than 1,300 adults with myeloid disorders has both confirmed the role of known or suspected driver mutations and uncovered new associations that could inform clinical care for patients with acute myeloid leukemia and myelodysplastic syndrome.

Dr. Ilaria Iacobucci, St. Jude Children’s Research Hospital, Memphis, Tenn.
Neil Osterweil/MDedge News
Dr. Ilaria Iacobucci

“Integration of mutational and expression data is important to refine subytpes and constellations of mutations with prognostic significance,” Ilaria Iacobucci, PhD, of St. Jude Children’s Research Hospital in Memphis said during a late-breaking abstract session at the annual meeting of the American Society of Hematology.

Her team conducted an analysis combining full genomic sequencing and gene-expression profiles in blood and bone marrow samples from 598 adults with acute myeloid leukemia (AML) and 706 with myelodysplastic syndrome (MDS).

The goals of the study were to provide “unbiased analysis of AML and MDS by integrated genomic and transcriptome data and clinico-pathologic features and clinical outcome” and to identify and define myeloid leukemia subtypes with diagnostic, prognostic, and therapeutic significance, she said.

The median age of the MDS cohort was 73.2 years (range 23.3-93.1). According to 2016 World Health Organization criteria, 37% had a diagnosis of MDS with excess blasts, 26.3% had MDS with ring sideroblasts, 20.9% had MDS with multilineage dysplasia, 14.6% had MDS with deletion 5q, and 1.1% had unclassifiable MDS.

The median age of the AML cohort was 68 years. Of this group, 31.7% had a diagnosis of AML not otherwise specified, 29.9% had known cytogenetic alterations, 27.3% had NPM1-mutated AML, and 9.7% had RUNX1-mutated disease.

Samples from all patients underwent tumor whole-genome sequencing and whole-transcriptome sequencing.

The combined sequencing confirmed a diagnosis of AML with recurrent genetic abnormalities in 11% of cases. These patients had disease with distinct gene-expression profiles and favorable prognosis. The sequencing identified combinations of mutations in genes linked with specific AML subtypes.

For example, combinations of mutations in KIT, ZBTB7A, ASXL2, RAD21, CSF3R, and DNM2 were associated with RUNX1-RUNXT1 leukemia, whereas mutations in FLT3, DDX54, WT1, and CALR in promyelocytic leukemia/retinoic acid receptor alpha were associated with promyelocytic leukemia, and KIT and BCORL1 mutations were associated with CBFB-rearranged leukemia.

In addition to rounding up the usual genomic suspects, the investigators also identified combinations that are associated with prognosis. Notably, NPM1 mutations were found in 27.4% of AML and 1% of MDS cases, and these mutations were characterized by four gene-expression signatures that were associated with different combinations of cooperating mutations in cohesin and signaling genes, and with outcome.

They found that patients with co-occurring NPM1 and FLT3 mutations had worse prognosis than those with mutations only in NPM1, whereas patients with NPM1 mutations co-occurring with cohesin gene mutations had better outcomes.

At a briefing prior to her presentation of the data, Dr. Iacobucci explained how her group’s findings might inform treatment, including the possibility of preventing development of AML in patients with MDS.

“What we are doing, in addition to the genomic part, is also establishing a repository of patient-derived xenografts, so in this way we can have the genome information, and we can have the biological material in vivo to test different therapies,” she said.

Dr. Andrew Wei of Alfred Hospital in Melbourne
Benjamin Pena/Medscape
Dr. Andrew H. Wei

In an interview, Andrew H. Wei, MBBS, PhD, from the Alfred Hospital in Melbourne, who was not involved in the genomic study, commented on the role of sequencing in treatment of patients with myeloid malignancies.

“I think the future is that as the leukemia evolves, our therapy will evolve along with it. Furthermore, we now have the potential to measure many of these mutations with much higher sensitivity than just whole-genome sequencing, so we can imagine a future whereby we can track and measure these mutations as they rise in the patient’s bone marrow or blood before the patients becomes sick with florid leukemia, and it gives us the potential to predictably alter our management before they become sick,” he said.

The study was supported by St. Jude Children’s Research Hospital and the Leukemia and Lymphoma Society. Dr. Iacobucci and Dr. Wei reported having no relevant disclosures.

SOURCE: Iacobucci I et al. ASH 2019, Abstract LBA-4.

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

– A genome-wide study of blood and bone marrow samples from more than 1,300 adults with myeloid disorders has both confirmed the role of known or suspected driver mutations and uncovered new associations that could inform clinical care for patients with acute myeloid leukemia and myelodysplastic syndrome.

Dr. Ilaria Iacobucci, St. Jude Children’s Research Hospital, Memphis, Tenn.
Neil Osterweil/MDedge News
Dr. Ilaria Iacobucci

“Integration of mutational and expression data is important to refine subytpes and constellations of mutations with prognostic significance,” Ilaria Iacobucci, PhD, of St. Jude Children’s Research Hospital in Memphis said during a late-breaking abstract session at the annual meeting of the American Society of Hematology.

Her team conducted an analysis combining full genomic sequencing and gene-expression profiles in blood and bone marrow samples from 598 adults with acute myeloid leukemia (AML) and 706 with myelodysplastic syndrome (MDS).

The goals of the study were to provide “unbiased analysis of AML and MDS by integrated genomic and transcriptome data and clinico-pathologic features and clinical outcome” and to identify and define myeloid leukemia subtypes with diagnostic, prognostic, and therapeutic significance, she said.

The median age of the MDS cohort was 73.2 years (range 23.3-93.1). According to 2016 World Health Organization criteria, 37% had a diagnosis of MDS with excess blasts, 26.3% had MDS with ring sideroblasts, 20.9% had MDS with multilineage dysplasia, 14.6% had MDS with deletion 5q, and 1.1% had unclassifiable MDS.

The median age of the AML cohort was 68 years. Of this group, 31.7% had a diagnosis of AML not otherwise specified, 29.9% had known cytogenetic alterations, 27.3% had NPM1-mutated AML, and 9.7% had RUNX1-mutated disease.

Samples from all patients underwent tumor whole-genome sequencing and whole-transcriptome sequencing.

The combined sequencing confirmed a diagnosis of AML with recurrent genetic abnormalities in 11% of cases. These patients had disease with distinct gene-expression profiles and favorable prognosis. The sequencing identified combinations of mutations in genes linked with specific AML subtypes.

For example, combinations of mutations in KIT, ZBTB7A, ASXL2, RAD21, CSF3R, and DNM2 were associated with RUNX1-RUNXT1 leukemia, whereas mutations in FLT3, DDX54, WT1, and CALR in promyelocytic leukemia/retinoic acid receptor alpha were associated with promyelocytic leukemia, and KIT and BCORL1 mutations were associated with CBFB-rearranged leukemia.

In addition to rounding up the usual genomic suspects, the investigators also identified combinations that are associated with prognosis. Notably, NPM1 mutations were found in 27.4% of AML and 1% of MDS cases, and these mutations were characterized by four gene-expression signatures that were associated with different combinations of cooperating mutations in cohesin and signaling genes, and with outcome.

They found that patients with co-occurring NPM1 and FLT3 mutations had worse prognosis than those with mutations only in NPM1, whereas patients with NPM1 mutations co-occurring with cohesin gene mutations had better outcomes.

At a briefing prior to her presentation of the data, Dr. Iacobucci explained how her group’s findings might inform treatment, including the possibility of preventing development of AML in patients with MDS.

“What we are doing, in addition to the genomic part, is also establishing a repository of patient-derived xenografts, so in this way we can have the genome information, and we can have the biological material in vivo to test different therapies,” she said.

Dr. Andrew Wei of Alfred Hospital in Melbourne
Benjamin Pena/Medscape
Dr. Andrew H. Wei

In an interview, Andrew H. Wei, MBBS, PhD, from the Alfred Hospital in Melbourne, who was not involved in the genomic study, commented on the role of sequencing in treatment of patients with myeloid malignancies.

“I think the future is that as the leukemia evolves, our therapy will evolve along with it. Furthermore, we now have the potential to measure many of these mutations with much higher sensitivity than just whole-genome sequencing, so we can imagine a future whereby we can track and measure these mutations as they rise in the patient’s bone marrow or blood before the patients becomes sick with florid leukemia, and it gives us the potential to predictably alter our management before they become sick,” he said.

The study was supported by St. Jude Children’s Research Hospital and the Leukemia and Lymphoma Society. Dr. Iacobucci and Dr. Wei reported having no relevant disclosures.

SOURCE: Iacobucci I et al. ASH 2019, Abstract LBA-4.

– A genome-wide study of blood and bone marrow samples from more than 1,300 adults with myeloid disorders has both confirmed the role of known or suspected driver mutations and uncovered new associations that could inform clinical care for patients with acute myeloid leukemia and myelodysplastic syndrome.

Dr. Ilaria Iacobucci, St. Jude Children’s Research Hospital, Memphis, Tenn.
Neil Osterweil/MDedge News
Dr. Ilaria Iacobucci

“Integration of mutational and expression data is important to refine subytpes and constellations of mutations with prognostic significance,” Ilaria Iacobucci, PhD, of St. Jude Children’s Research Hospital in Memphis said during a late-breaking abstract session at the annual meeting of the American Society of Hematology.

Her team conducted an analysis combining full genomic sequencing and gene-expression profiles in blood and bone marrow samples from 598 adults with acute myeloid leukemia (AML) and 706 with myelodysplastic syndrome (MDS).

The goals of the study were to provide “unbiased analysis of AML and MDS by integrated genomic and transcriptome data and clinico-pathologic features and clinical outcome” and to identify and define myeloid leukemia subtypes with diagnostic, prognostic, and therapeutic significance, she said.

The median age of the MDS cohort was 73.2 years (range 23.3-93.1). According to 2016 World Health Organization criteria, 37% had a diagnosis of MDS with excess blasts, 26.3% had MDS with ring sideroblasts, 20.9% had MDS with multilineage dysplasia, 14.6% had MDS with deletion 5q, and 1.1% had unclassifiable MDS.

The median age of the AML cohort was 68 years. Of this group, 31.7% had a diagnosis of AML not otherwise specified, 29.9% had known cytogenetic alterations, 27.3% had NPM1-mutated AML, and 9.7% had RUNX1-mutated disease.

Samples from all patients underwent tumor whole-genome sequencing and whole-transcriptome sequencing.

The combined sequencing confirmed a diagnosis of AML with recurrent genetic abnormalities in 11% of cases. These patients had disease with distinct gene-expression profiles and favorable prognosis. The sequencing identified combinations of mutations in genes linked with specific AML subtypes.

For example, combinations of mutations in KIT, ZBTB7A, ASXL2, RAD21, CSF3R, and DNM2 were associated with RUNX1-RUNXT1 leukemia, whereas mutations in FLT3, DDX54, WT1, and CALR in promyelocytic leukemia/retinoic acid receptor alpha were associated with promyelocytic leukemia, and KIT and BCORL1 mutations were associated with CBFB-rearranged leukemia.

In addition to rounding up the usual genomic suspects, the investigators also identified combinations that are associated with prognosis. Notably, NPM1 mutations were found in 27.4% of AML and 1% of MDS cases, and these mutations were characterized by four gene-expression signatures that were associated with different combinations of cooperating mutations in cohesin and signaling genes, and with outcome.

They found that patients with co-occurring NPM1 and FLT3 mutations had worse prognosis than those with mutations only in NPM1, whereas patients with NPM1 mutations co-occurring with cohesin gene mutations had better outcomes.

At a briefing prior to her presentation of the data, Dr. Iacobucci explained how her group’s findings might inform treatment, including the possibility of preventing development of AML in patients with MDS.

“What we are doing, in addition to the genomic part, is also establishing a repository of patient-derived xenografts, so in this way we can have the genome information, and we can have the biological material in vivo to test different therapies,” she said.

Dr. Andrew Wei of Alfred Hospital in Melbourne
Benjamin Pena/Medscape
Dr. Andrew H. Wei

In an interview, Andrew H. Wei, MBBS, PhD, from the Alfred Hospital in Melbourne, who was not involved in the genomic study, commented on the role of sequencing in treatment of patients with myeloid malignancies.

“I think the future is that as the leukemia evolves, our therapy will evolve along with it. Furthermore, we now have the potential to measure many of these mutations with much higher sensitivity than just whole-genome sequencing, so we can imagine a future whereby we can track and measure these mutations as they rise in the patient’s bone marrow or blood before the patients becomes sick with florid leukemia, and it gives us the potential to predictably alter our management before they become sick,” he said.

The study was supported by St. Jude Children’s Research Hospital and the Leukemia and Lymphoma Society. Dr. Iacobucci and Dr. Wei reported having no relevant disclosures.

SOURCE: Iacobucci I et al. ASH 2019, Abstract LBA-4.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASH 2019

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

High response, survival rates with ponatinib/hCVAD in Ph-positive ALL

Article Type
Changed
Wed, 01/08/2020 - 11:05

– For adults with newly diagnosed acute lymphoblastic leukemia positive for the Philadelphia chromosome (Ph+ALL), the combination of hyper-CVAD chemotherapy and ponatinib is associated with high complete molecular response and 5-year overall survival rates, investigators reported.

Dr. Nicholas J. Short, University of Texas MD Anderson Cancer Center, Houston
Neil Osterweil/MDedge News
Dr. Nicholas J. Short

Long-term follow-up of 86 adults with Ph+ALL treated in the front line with chemotherapy plus ponatinib (Iclusig), a third-generation tyrosine kinase inhibitor (TKI), showed a complete remission (CR) rate of 100%, complete molecular remission (CMR) rate of 86%, and a 5-year overall survival (OS) rate of 74%, reported Nicholas J. Short, MD, from the University of Texas MD Anderson Cancer Center in Houston.

“Although we observed two treatment-related cardiovascular deaths with the original trial design, with almost 50 patients treated since instituting a risk-adapted dosing schedule with lower doses of ponatinib, no additional ponatinib-related deaths have been observed,” he said at the annual meeting of the American Society of Hematology.

The standard of care for adults with Ph+ALL is chemotherapy plus a TKI. With a first- or second-generation TKI plus chemotherapy, reported 5-year OS rates range from 35% to 50%.

“However, relapses are still common, and these are usually driven by the development of new resistance mutations in the ABL gene, particularly the T315I gatekeeper mutation which has been reported in up to 75% of patients at the time of relapse,” he said.

Ponatinib is a pan-BCR-ABL TKI with activity against ALL with T315I mutations, and the combination of this agent with hyper-CVAD chemotherapy (hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone) has been associated with higher response rates than those seen with earlier-generation TKIs, as well as higher levels of minimal residual disease (MRD) negativity, he noted.

Dr. Short and colleagues hypothesized that, compared with the standard of care, hyper-CVAD plus ponatinib would be associated with higher MRD levels, low relapse rates by suppression of T315I subclones, decreased reliance on stem cell transplantation in first remission, and improved long-term survival.

To test this, they treated 86 adults with newly diagnosed Ph+ALL, including those who had undergone one or two previous courses of chemotherapy with a TKI other than ponatinib. The patients had Eastern Cooperative Oncology Group performance status 0-2, adequate organ function, and no clinically significant cardiovascular disease.

The patients underwent eight cycles of hyper-CVAD alternating with high-dose methotrexate/cytarabine approximately every 21 days. The first 37 patients were treated with, ponatinib 45 mg daily for the first 14 days of cycle 1, then continuously for subsequent cycles. Patients with CD20 expression of 20% or greater also received rituximab during the first four cycles. CNS prophylaxis was also administered with 12 doses of intrathecal chemotherapy with alternating methotrexate and cytarabine.

Patients who had a CR received maintenance with ponatinib and vincristine/prednisone monthly for 2 years, followed by ponatinib indefinitely.

Out of concern for vascular toxicity with long-term use of high-dose ponatinib, including the two deaths mentioned before, the protocol was amended after the first 37 patients were treated. The amended protocol reduced ponatinib to 30 mg starting at cycle 2, with further reduction to 15 mg once a CMR (absence of BCR-ABL on polymerase chain reaction) was achieved.

At a median follow-up of 44 months, the event-free survival rates – the primary endpoint – were 71% at 3 years and 68% at 5 years. The 3-year OS rate was 78%, and the 5-year OS rate was 74%.

All patients had complete remission and complete cytogenetic remission as assessed by conventional karyotyping. Additionally, 73 of 85 evaluable patients (86%) achieved a CMR at some point during therapy.

“We had previously reported that achievement of a complete molecular response by 3 months is associated with superior outcomes. Approximately three quarters of patients achieved this milestone,” Dr. Short said.

Grade 3 or greater adverse events of particular concern included transaminase elevations in 29% of patients, elevated bilirubin and pancreatitis in 15% each, and hypertension in 14%.

Four patients had grade 3 or greater venous thromboembolic or arterial events, including the two previously noted deaths from myocardial infarction, both of which occurred prior to the protocol amendment.

At the most recent follow-up, 11 patients had experienced relapse (no CNS-only relapses), and of this group, 5 died and 6 were still alive. Nineteen patients underwent hematopoietic stem cell transplant, and of this group, 13 were still alive and 6 died.

Causes of death in the nine patients who died while in CR included the two myocardial infarction deaths on study, three deaths from sepsis during consolidation, one from lung cancer, one from a head injury after a fall, one from myocardial infarction in a 79-year-old patient 4 years after stopping ponatinib (off study), and one from preexisting congestive heart failure in a 74-year-old patient.

In all, 47 patients were either in ongoing therapy or observation at last follow-up, including three patients who were transitioned to MRD-directed therapy including blinatumomab (Blincyto).

“As a next step, we are now evaluating lower-intensity regimens with ponatinib and blinatumomab in both the frontline and relapsed/refractory settings, with the goals of decreased chemotherapy-related toxicity, increased MRD-negativity rates, further decreased reliance on transplant, and improved long-term outcomes,” Dr. Short said.

The study was sponsored by MD Anderson with support from the National Cancer Institute. Dr. Short reported consulting for AstraZenca, honoraria from Amgen, and consulting and receiving research funding from Takeda Oncology.

SOURCE: Short NJ et al. ASH 2019, Abstract 283.

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

– For adults with newly diagnosed acute lymphoblastic leukemia positive for the Philadelphia chromosome (Ph+ALL), the combination of hyper-CVAD chemotherapy and ponatinib is associated with high complete molecular response and 5-year overall survival rates, investigators reported.

Dr. Nicholas J. Short, University of Texas MD Anderson Cancer Center, Houston
Neil Osterweil/MDedge News
Dr. Nicholas J. Short

Long-term follow-up of 86 adults with Ph+ALL treated in the front line with chemotherapy plus ponatinib (Iclusig), a third-generation tyrosine kinase inhibitor (TKI), showed a complete remission (CR) rate of 100%, complete molecular remission (CMR) rate of 86%, and a 5-year overall survival (OS) rate of 74%, reported Nicholas J. Short, MD, from the University of Texas MD Anderson Cancer Center in Houston.

“Although we observed two treatment-related cardiovascular deaths with the original trial design, with almost 50 patients treated since instituting a risk-adapted dosing schedule with lower doses of ponatinib, no additional ponatinib-related deaths have been observed,” he said at the annual meeting of the American Society of Hematology.

The standard of care for adults with Ph+ALL is chemotherapy plus a TKI. With a first- or second-generation TKI plus chemotherapy, reported 5-year OS rates range from 35% to 50%.

“However, relapses are still common, and these are usually driven by the development of new resistance mutations in the ABL gene, particularly the T315I gatekeeper mutation which has been reported in up to 75% of patients at the time of relapse,” he said.

Ponatinib is a pan-BCR-ABL TKI with activity against ALL with T315I mutations, and the combination of this agent with hyper-CVAD chemotherapy (hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone) has been associated with higher response rates than those seen with earlier-generation TKIs, as well as higher levels of minimal residual disease (MRD) negativity, he noted.

Dr. Short and colleagues hypothesized that, compared with the standard of care, hyper-CVAD plus ponatinib would be associated with higher MRD levels, low relapse rates by suppression of T315I subclones, decreased reliance on stem cell transplantation in first remission, and improved long-term survival.

To test this, they treated 86 adults with newly diagnosed Ph+ALL, including those who had undergone one or two previous courses of chemotherapy with a TKI other than ponatinib. The patients had Eastern Cooperative Oncology Group performance status 0-2, adequate organ function, and no clinically significant cardiovascular disease.

The patients underwent eight cycles of hyper-CVAD alternating with high-dose methotrexate/cytarabine approximately every 21 days. The first 37 patients were treated with, ponatinib 45 mg daily for the first 14 days of cycle 1, then continuously for subsequent cycles. Patients with CD20 expression of 20% or greater also received rituximab during the first four cycles. CNS prophylaxis was also administered with 12 doses of intrathecal chemotherapy with alternating methotrexate and cytarabine.

Patients who had a CR received maintenance with ponatinib and vincristine/prednisone monthly for 2 years, followed by ponatinib indefinitely.

Out of concern for vascular toxicity with long-term use of high-dose ponatinib, including the two deaths mentioned before, the protocol was amended after the first 37 patients were treated. The amended protocol reduced ponatinib to 30 mg starting at cycle 2, with further reduction to 15 mg once a CMR (absence of BCR-ABL on polymerase chain reaction) was achieved.

At a median follow-up of 44 months, the event-free survival rates – the primary endpoint – were 71% at 3 years and 68% at 5 years. The 3-year OS rate was 78%, and the 5-year OS rate was 74%.

All patients had complete remission and complete cytogenetic remission as assessed by conventional karyotyping. Additionally, 73 of 85 evaluable patients (86%) achieved a CMR at some point during therapy.

“We had previously reported that achievement of a complete molecular response by 3 months is associated with superior outcomes. Approximately three quarters of patients achieved this milestone,” Dr. Short said.

Grade 3 or greater adverse events of particular concern included transaminase elevations in 29% of patients, elevated bilirubin and pancreatitis in 15% each, and hypertension in 14%.

Four patients had grade 3 or greater venous thromboembolic or arterial events, including the two previously noted deaths from myocardial infarction, both of which occurred prior to the protocol amendment.

At the most recent follow-up, 11 patients had experienced relapse (no CNS-only relapses), and of this group, 5 died and 6 were still alive. Nineteen patients underwent hematopoietic stem cell transplant, and of this group, 13 were still alive and 6 died.

Causes of death in the nine patients who died while in CR included the two myocardial infarction deaths on study, three deaths from sepsis during consolidation, one from lung cancer, one from a head injury after a fall, one from myocardial infarction in a 79-year-old patient 4 years after stopping ponatinib (off study), and one from preexisting congestive heart failure in a 74-year-old patient.

In all, 47 patients were either in ongoing therapy or observation at last follow-up, including three patients who were transitioned to MRD-directed therapy including blinatumomab (Blincyto).

“As a next step, we are now evaluating lower-intensity regimens with ponatinib and blinatumomab in both the frontline and relapsed/refractory settings, with the goals of decreased chemotherapy-related toxicity, increased MRD-negativity rates, further decreased reliance on transplant, and improved long-term outcomes,” Dr. Short said.

The study was sponsored by MD Anderson with support from the National Cancer Institute. Dr. Short reported consulting for AstraZenca, honoraria from Amgen, and consulting and receiving research funding from Takeda Oncology.

SOURCE: Short NJ et al. ASH 2019, Abstract 283.

– For adults with newly diagnosed acute lymphoblastic leukemia positive for the Philadelphia chromosome (Ph+ALL), the combination of hyper-CVAD chemotherapy and ponatinib is associated with high complete molecular response and 5-year overall survival rates, investigators reported.

Dr. Nicholas J. Short, University of Texas MD Anderson Cancer Center, Houston
Neil Osterweil/MDedge News
Dr. Nicholas J. Short

Long-term follow-up of 86 adults with Ph+ALL treated in the front line with chemotherapy plus ponatinib (Iclusig), a third-generation tyrosine kinase inhibitor (TKI), showed a complete remission (CR) rate of 100%, complete molecular remission (CMR) rate of 86%, and a 5-year overall survival (OS) rate of 74%, reported Nicholas J. Short, MD, from the University of Texas MD Anderson Cancer Center in Houston.

“Although we observed two treatment-related cardiovascular deaths with the original trial design, with almost 50 patients treated since instituting a risk-adapted dosing schedule with lower doses of ponatinib, no additional ponatinib-related deaths have been observed,” he said at the annual meeting of the American Society of Hematology.

The standard of care for adults with Ph+ALL is chemotherapy plus a TKI. With a first- or second-generation TKI plus chemotherapy, reported 5-year OS rates range from 35% to 50%.

“However, relapses are still common, and these are usually driven by the development of new resistance mutations in the ABL gene, particularly the T315I gatekeeper mutation which has been reported in up to 75% of patients at the time of relapse,” he said.

Ponatinib is a pan-BCR-ABL TKI with activity against ALL with T315I mutations, and the combination of this agent with hyper-CVAD chemotherapy (hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone) has been associated with higher response rates than those seen with earlier-generation TKIs, as well as higher levels of minimal residual disease (MRD) negativity, he noted.

Dr. Short and colleagues hypothesized that, compared with the standard of care, hyper-CVAD plus ponatinib would be associated with higher MRD levels, low relapse rates by suppression of T315I subclones, decreased reliance on stem cell transplantation in first remission, and improved long-term survival.

To test this, they treated 86 adults with newly diagnosed Ph+ALL, including those who had undergone one or two previous courses of chemotherapy with a TKI other than ponatinib. The patients had Eastern Cooperative Oncology Group performance status 0-2, adequate organ function, and no clinically significant cardiovascular disease.

The patients underwent eight cycles of hyper-CVAD alternating with high-dose methotrexate/cytarabine approximately every 21 days. The first 37 patients were treated with, ponatinib 45 mg daily for the first 14 days of cycle 1, then continuously for subsequent cycles. Patients with CD20 expression of 20% or greater also received rituximab during the first four cycles. CNS prophylaxis was also administered with 12 doses of intrathecal chemotherapy with alternating methotrexate and cytarabine.

Patients who had a CR received maintenance with ponatinib and vincristine/prednisone monthly for 2 years, followed by ponatinib indefinitely.

Out of concern for vascular toxicity with long-term use of high-dose ponatinib, including the two deaths mentioned before, the protocol was amended after the first 37 patients were treated. The amended protocol reduced ponatinib to 30 mg starting at cycle 2, with further reduction to 15 mg once a CMR (absence of BCR-ABL on polymerase chain reaction) was achieved.

At a median follow-up of 44 months, the event-free survival rates – the primary endpoint – were 71% at 3 years and 68% at 5 years. The 3-year OS rate was 78%, and the 5-year OS rate was 74%.

All patients had complete remission and complete cytogenetic remission as assessed by conventional karyotyping. Additionally, 73 of 85 evaluable patients (86%) achieved a CMR at some point during therapy.

“We had previously reported that achievement of a complete molecular response by 3 months is associated with superior outcomes. Approximately three quarters of patients achieved this milestone,” Dr. Short said.

Grade 3 or greater adverse events of particular concern included transaminase elevations in 29% of patients, elevated bilirubin and pancreatitis in 15% each, and hypertension in 14%.

Four patients had grade 3 or greater venous thromboembolic or arterial events, including the two previously noted deaths from myocardial infarction, both of which occurred prior to the protocol amendment.

At the most recent follow-up, 11 patients had experienced relapse (no CNS-only relapses), and of this group, 5 died and 6 were still alive. Nineteen patients underwent hematopoietic stem cell transplant, and of this group, 13 were still alive and 6 died.

Causes of death in the nine patients who died while in CR included the two myocardial infarction deaths on study, three deaths from sepsis during consolidation, one from lung cancer, one from a head injury after a fall, one from myocardial infarction in a 79-year-old patient 4 years after stopping ponatinib (off study), and one from preexisting congestive heart failure in a 74-year-old patient.

In all, 47 patients were either in ongoing therapy or observation at last follow-up, including three patients who were transitioned to MRD-directed therapy including blinatumomab (Blincyto).

“As a next step, we are now evaluating lower-intensity regimens with ponatinib and blinatumomab in both the frontline and relapsed/refractory settings, with the goals of decreased chemotherapy-related toxicity, increased MRD-negativity rates, further decreased reliance on transplant, and improved long-term outcomes,” Dr. Short said.

The study was sponsored by MD Anderson with support from the National Cancer Institute. Dr. Short reported consulting for AstraZenca, honoraria from Amgen, and consulting and receiving research funding from Takeda Oncology.

SOURCE: Short NJ et al. ASH 2019, Abstract 283.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASH 2019

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

CAR T cells produce complete responses in T-cell malignancies

Article Type
Changed
Mon, 01/13/2020 - 13:35

– Anti-CD5 chimeric antigen receptor (CAR) T cells can produce complete responses (CRs) in patients with relapsed or refractory T-cell malignancies, according to findings from a phase 1 trial.

Dr. LaQuisa C. Hill, Baylor College of Medicine, Houston
Jennifer Smith/MDedge News
Dr. LaQuisa C. Hill

Three of 11 patients achieved a CR after CAR T-cell therapy, and one patient achieved a mixed response that deepened to a CR after transplant. Three responders, all of whom had T-cell lymphoma, were still alive and in CR at last follow-up.

There were no cases of severe cytokine release syndrome (CRS) or severe neurotoxicity, no serious infectious complications, and no nonhematologic grade 4 adverse events in this trial.

LaQuisa C. Hill, MD, of Baylor College of Medicine, Houston, presented these results at the annual meeting of the American Society of Hematology.

“While CD19 CAR T cells have revolutionized the treatment of relapsed/refractory B-cell malignancies, development of CAR T-cell platforms targeting T-cell-driven malignancies have been hindered by three main factors: CAR T-cell fratricide due to shared expression of target antigens leading to impaired expansion, ablation of normal T cells continuing to cause profound immunodeficiency, and the potential of transduced tumor cells providing a means of tumor escape,” Dr. Hill said.

Researchers have theorized that anti-CD5 CAR T cells can overcome these obstacles. In preclinical studies, anti-CD5 CAR T cells eliminated malignant blasts in vitro and in vivo and resulted in “limited and transient” fratricide (Blood. 2015 Aug 20;126[8]:983-92).

With this in mind, Dr. Hill and her colleagues tested CD5.28z CAR T cells in a phase 1 trial (NCT03081910). Eleven patients have been treated thus far – five with T-cell acute lymphoblastic leukemia (T-ALL), three with peripheral T-cell lymphoma (PTCL), two with angioimmunoblastic T-cell lymphoma (AITL), and one with Sézary syndrome.

The patients’ median age at baseline was 62 years (range, 21-71 years), and 63% were men. They had received a median of 5 prior therapies (range, 3-18). Two patients had relapsed after allogeneic hematopoietic stem cell transplant (HSCT), three had relapsed after autologous HSCT, and five were primary refractory.

Patients underwent lymphodepletion with fludarabine and cyclophosphamide, then received CAR T cells at doses of 1 x 107 or 5 x 107.
 

Response

Three lymphoma patients – two with AITL and one with PTCL – were still alive and in CR at last follow-up. The PTCL patient achieved a CR after CAR T-cell therapy and declined a subsequent HSCT. The patient has not received additional therapy and has retained the CR for 7 months.

One AITL patient achieved a CR and declined transplant as well. He relapsed after 7 months but received subsequent therapy and achieved another CR. The other AITL patient had a mixed response to CAR T-cell therapy but proceeded to allogeneic HSCT and achieved a CR that has lasted 9 months.

The remaining three lymphoma patients – two with PTCL and one with Sézary syndrome – progressed and died.

One T-ALL patient achieved a CR lasting 6 weeks, but the patient died while undergoing transplant workup. Two T-ALL patients did not respond to treatment and died. The remaining two patients progressed, and one of them died. The other patient who progressed is still alive and in CR after receiving subsequent therapy.

 

 

Factors associated with response

Dr. Hill said a shortened manufacturing process may be associated with enhanced response, as all responders received CAR T cells produced via a shorter manufacturing process. The shortened process involves freezing cells on day 4-5 post transduction, as opposed to day 7.

“While the numbers are too small to make any definitive conclusions, this seems to correlate with less terminal differentiation, which might improve potency,” Dr. Hill said. “However, additional analyses are ongoing.”

Dr. Hill also pointed out that CAR T-cell expansion was observed in all patients, with higher peak levels observed at the higher dose. In addition, CAR T-cell persistence was durable at both dose levels.

“We have been able to detect the CAR transgene at all follow-up time points, out to 9 months for some patients,” Dr. Hill said. “While limited persistence may play a role in nonresponders, it does not appear to be the only factor.”

Safety

“Surprisingly, no selective ablation of normal T cells has been observed,” Dr. Hill said. “As CAR T cells dwindled [after infusion], we were able to see recovery of normal T cells, all of which expressed normal levels of CD5. This was observed in all patients on study, except for one patient who had prolonged pancytopenia.”

Cytopenias were the most common grade 3/4 adverse events, including neutropenia (n = 8), anemia (n = 7), and thrombocytopenia (n = 5). Other grade 3/4 events included elevated aspartate aminotransferase (n = 2), hypoalbuminemia (n = 1), hyponatremia (n = 1), hypophosphatemia (n = 1), and elevated alanine aminotransferase (n = 1). There were no grade 5 adverse events.

Two patients developed grade 1 CRS, and two had grade 2 CRS. Both patients with grade 2 CRS were treated with tocilizumab, and their symptoms resolved.

One patient developed grade 2 immune effector cell-associated neurotoxicity syndrome, but this resolved with supportive care.

One patient had a central line–associated bloodstream infection (coagulase-negative staphylococci), and one had cytomegalovirus and BK virus reactivation. There were no fungal infections.

“We have demonstrated that CD5 CAR T cells can be manufactured from heavily pretreated patients with T-cell malignancies, and therapy is well tolerated,” Dr. Hill said. “We have seen strong and promising activity in T-cell lymphoma, which we hope to be able to translate to T-ALL as well.”

Dr. Hill said she and her colleagues hope to improve upon these results with a higher dose level of CD5 CAR T cells (1 x 108), which the team plans to start testing soon. The researchers may also investigate other target antigens, such as CD7, as well as the use of donor-derived CAR T cells for patients who have relapsed after allogeneic HSCT.

Dr. Hill said she has no relevant disclosures. Baylor College of Medicine is sponsoring this trial.

SOURCE: Hill L et al. ASH 2019. Abstract 199.

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

– Anti-CD5 chimeric antigen receptor (CAR) T cells can produce complete responses (CRs) in patients with relapsed or refractory T-cell malignancies, according to findings from a phase 1 trial.

Dr. LaQuisa C. Hill, Baylor College of Medicine, Houston
Jennifer Smith/MDedge News
Dr. LaQuisa C. Hill

Three of 11 patients achieved a CR after CAR T-cell therapy, and one patient achieved a mixed response that deepened to a CR after transplant. Three responders, all of whom had T-cell lymphoma, were still alive and in CR at last follow-up.

There were no cases of severe cytokine release syndrome (CRS) or severe neurotoxicity, no serious infectious complications, and no nonhematologic grade 4 adverse events in this trial.

LaQuisa C. Hill, MD, of Baylor College of Medicine, Houston, presented these results at the annual meeting of the American Society of Hematology.

“While CD19 CAR T cells have revolutionized the treatment of relapsed/refractory B-cell malignancies, development of CAR T-cell platforms targeting T-cell-driven malignancies have been hindered by three main factors: CAR T-cell fratricide due to shared expression of target antigens leading to impaired expansion, ablation of normal T cells continuing to cause profound immunodeficiency, and the potential of transduced tumor cells providing a means of tumor escape,” Dr. Hill said.

Researchers have theorized that anti-CD5 CAR T cells can overcome these obstacles. In preclinical studies, anti-CD5 CAR T cells eliminated malignant blasts in vitro and in vivo and resulted in “limited and transient” fratricide (Blood. 2015 Aug 20;126[8]:983-92).

With this in mind, Dr. Hill and her colleagues tested CD5.28z CAR T cells in a phase 1 trial (NCT03081910). Eleven patients have been treated thus far – five with T-cell acute lymphoblastic leukemia (T-ALL), three with peripheral T-cell lymphoma (PTCL), two with angioimmunoblastic T-cell lymphoma (AITL), and one with Sézary syndrome.

The patients’ median age at baseline was 62 years (range, 21-71 years), and 63% were men. They had received a median of 5 prior therapies (range, 3-18). Two patients had relapsed after allogeneic hematopoietic stem cell transplant (HSCT), three had relapsed after autologous HSCT, and five were primary refractory.

Patients underwent lymphodepletion with fludarabine and cyclophosphamide, then received CAR T cells at doses of 1 x 107 or 5 x 107.
 

Response

Three lymphoma patients – two with AITL and one with PTCL – were still alive and in CR at last follow-up. The PTCL patient achieved a CR after CAR T-cell therapy and declined a subsequent HSCT. The patient has not received additional therapy and has retained the CR for 7 months.

One AITL patient achieved a CR and declined transplant as well. He relapsed after 7 months but received subsequent therapy and achieved another CR. The other AITL patient had a mixed response to CAR T-cell therapy but proceeded to allogeneic HSCT and achieved a CR that has lasted 9 months.

The remaining three lymphoma patients – two with PTCL and one with Sézary syndrome – progressed and died.

One T-ALL patient achieved a CR lasting 6 weeks, but the patient died while undergoing transplant workup. Two T-ALL patients did not respond to treatment and died. The remaining two patients progressed, and one of them died. The other patient who progressed is still alive and in CR after receiving subsequent therapy.

 

 

Factors associated with response

Dr. Hill said a shortened manufacturing process may be associated with enhanced response, as all responders received CAR T cells produced via a shorter manufacturing process. The shortened process involves freezing cells on day 4-5 post transduction, as opposed to day 7.

“While the numbers are too small to make any definitive conclusions, this seems to correlate with less terminal differentiation, which might improve potency,” Dr. Hill said. “However, additional analyses are ongoing.”

Dr. Hill also pointed out that CAR T-cell expansion was observed in all patients, with higher peak levels observed at the higher dose. In addition, CAR T-cell persistence was durable at both dose levels.

“We have been able to detect the CAR transgene at all follow-up time points, out to 9 months for some patients,” Dr. Hill said. “While limited persistence may play a role in nonresponders, it does not appear to be the only factor.”

Safety

“Surprisingly, no selective ablation of normal T cells has been observed,” Dr. Hill said. “As CAR T cells dwindled [after infusion], we were able to see recovery of normal T cells, all of which expressed normal levels of CD5. This was observed in all patients on study, except for one patient who had prolonged pancytopenia.”

Cytopenias were the most common grade 3/4 adverse events, including neutropenia (n = 8), anemia (n = 7), and thrombocytopenia (n = 5). Other grade 3/4 events included elevated aspartate aminotransferase (n = 2), hypoalbuminemia (n = 1), hyponatremia (n = 1), hypophosphatemia (n = 1), and elevated alanine aminotransferase (n = 1). There were no grade 5 adverse events.

Two patients developed grade 1 CRS, and two had grade 2 CRS. Both patients with grade 2 CRS were treated with tocilizumab, and their symptoms resolved.

One patient developed grade 2 immune effector cell-associated neurotoxicity syndrome, but this resolved with supportive care.

One patient had a central line–associated bloodstream infection (coagulase-negative staphylococci), and one had cytomegalovirus and BK virus reactivation. There were no fungal infections.

“We have demonstrated that CD5 CAR T cells can be manufactured from heavily pretreated patients with T-cell malignancies, and therapy is well tolerated,” Dr. Hill said. “We have seen strong and promising activity in T-cell lymphoma, which we hope to be able to translate to T-ALL as well.”

Dr. Hill said she and her colleagues hope to improve upon these results with a higher dose level of CD5 CAR T cells (1 x 108), which the team plans to start testing soon. The researchers may also investigate other target antigens, such as CD7, as well as the use of donor-derived CAR T cells for patients who have relapsed after allogeneic HSCT.

Dr. Hill said she has no relevant disclosures. Baylor College of Medicine is sponsoring this trial.

SOURCE: Hill L et al. ASH 2019. Abstract 199.

– Anti-CD5 chimeric antigen receptor (CAR) T cells can produce complete responses (CRs) in patients with relapsed or refractory T-cell malignancies, according to findings from a phase 1 trial.

Dr. LaQuisa C. Hill, Baylor College of Medicine, Houston
Jennifer Smith/MDedge News
Dr. LaQuisa C. Hill

Three of 11 patients achieved a CR after CAR T-cell therapy, and one patient achieved a mixed response that deepened to a CR after transplant. Three responders, all of whom had T-cell lymphoma, were still alive and in CR at last follow-up.

There were no cases of severe cytokine release syndrome (CRS) or severe neurotoxicity, no serious infectious complications, and no nonhematologic grade 4 adverse events in this trial.

LaQuisa C. Hill, MD, of Baylor College of Medicine, Houston, presented these results at the annual meeting of the American Society of Hematology.

“While CD19 CAR T cells have revolutionized the treatment of relapsed/refractory B-cell malignancies, development of CAR T-cell platforms targeting T-cell-driven malignancies have been hindered by three main factors: CAR T-cell fratricide due to shared expression of target antigens leading to impaired expansion, ablation of normal T cells continuing to cause profound immunodeficiency, and the potential of transduced tumor cells providing a means of tumor escape,” Dr. Hill said.

Researchers have theorized that anti-CD5 CAR T cells can overcome these obstacles. In preclinical studies, anti-CD5 CAR T cells eliminated malignant blasts in vitro and in vivo and resulted in “limited and transient” fratricide (Blood. 2015 Aug 20;126[8]:983-92).

With this in mind, Dr. Hill and her colleagues tested CD5.28z CAR T cells in a phase 1 trial (NCT03081910). Eleven patients have been treated thus far – five with T-cell acute lymphoblastic leukemia (T-ALL), three with peripheral T-cell lymphoma (PTCL), two with angioimmunoblastic T-cell lymphoma (AITL), and one with Sézary syndrome.

The patients’ median age at baseline was 62 years (range, 21-71 years), and 63% were men. They had received a median of 5 prior therapies (range, 3-18). Two patients had relapsed after allogeneic hematopoietic stem cell transplant (HSCT), three had relapsed after autologous HSCT, and five were primary refractory.

Patients underwent lymphodepletion with fludarabine and cyclophosphamide, then received CAR T cells at doses of 1 x 107 or 5 x 107.
 

Response

Three lymphoma patients – two with AITL and one with PTCL – were still alive and in CR at last follow-up. The PTCL patient achieved a CR after CAR T-cell therapy and declined a subsequent HSCT. The patient has not received additional therapy and has retained the CR for 7 months.

One AITL patient achieved a CR and declined transplant as well. He relapsed after 7 months but received subsequent therapy and achieved another CR. The other AITL patient had a mixed response to CAR T-cell therapy but proceeded to allogeneic HSCT and achieved a CR that has lasted 9 months.

The remaining three lymphoma patients – two with PTCL and one with Sézary syndrome – progressed and died.

One T-ALL patient achieved a CR lasting 6 weeks, but the patient died while undergoing transplant workup. Two T-ALL patients did not respond to treatment and died. The remaining two patients progressed, and one of them died. The other patient who progressed is still alive and in CR after receiving subsequent therapy.

 

 

Factors associated with response

Dr. Hill said a shortened manufacturing process may be associated with enhanced response, as all responders received CAR T cells produced via a shorter manufacturing process. The shortened process involves freezing cells on day 4-5 post transduction, as opposed to day 7.

“While the numbers are too small to make any definitive conclusions, this seems to correlate with less terminal differentiation, which might improve potency,” Dr. Hill said. “However, additional analyses are ongoing.”

Dr. Hill also pointed out that CAR T-cell expansion was observed in all patients, with higher peak levels observed at the higher dose. In addition, CAR T-cell persistence was durable at both dose levels.

“We have been able to detect the CAR transgene at all follow-up time points, out to 9 months for some patients,” Dr. Hill said. “While limited persistence may play a role in nonresponders, it does not appear to be the only factor.”

Safety

“Surprisingly, no selective ablation of normal T cells has been observed,” Dr. Hill said. “As CAR T cells dwindled [after infusion], we were able to see recovery of normal T cells, all of which expressed normal levels of CD5. This was observed in all patients on study, except for one patient who had prolonged pancytopenia.”

Cytopenias were the most common grade 3/4 adverse events, including neutropenia (n = 8), anemia (n = 7), and thrombocytopenia (n = 5). Other grade 3/4 events included elevated aspartate aminotransferase (n = 2), hypoalbuminemia (n = 1), hyponatremia (n = 1), hypophosphatemia (n = 1), and elevated alanine aminotransferase (n = 1). There were no grade 5 adverse events.

Two patients developed grade 1 CRS, and two had grade 2 CRS. Both patients with grade 2 CRS were treated with tocilizumab, and their symptoms resolved.

One patient developed grade 2 immune effector cell-associated neurotoxicity syndrome, but this resolved with supportive care.

One patient had a central line–associated bloodstream infection (coagulase-negative staphylococci), and one had cytomegalovirus and BK virus reactivation. There were no fungal infections.

“We have demonstrated that CD5 CAR T cells can be manufactured from heavily pretreated patients with T-cell malignancies, and therapy is well tolerated,” Dr. Hill said. “We have seen strong and promising activity in T-cell lymphoma, which we hope to be able to translate to T-ALL as well.”

Dr. Hill said she and her colleagues hope to improve upon these results with a higher dose level of CD5 CAR T cells (1 x 108), which the team plans to start testing soon. The researchers may also investigate other target antigens, such as CD7, as well as the use of donor-derived CAR T cells for patients who have relapsed after allogeneic HSCT.

Dr. Hill said she has no relevant disclosures. Baylor College of Medicine is sponsoring this trial.

SOURCE: Hill L et al. ASH 2019. Abstract 199.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASH 2019

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

CAR T-cell therapy advances in CLL

Article Type
Changed
Thu, 01/12/2023 - 10:44

– Lisocabtagene maraleucel (liso-cel), a CD19-directed chimeric antigen receptor (CAR) T-cell therapy, has demonstrated manageable toxicity and promising clinical activity in the phase 1 portion of a trial enrolling heavily pretreated patients with chronic lymphocytic leukemia/small lymphocytic lymphoma, according to an investigator.

Dr. Tanya Siddiqi of City of Hope National Medical Center in Duarte, Calif.
Andrew D. Bowser/MDedge News
Dr. Tanya Siddiqi

The overall response rate exceeded 80%, and most patients in response at 6 months had maintained that response at the 9-month mark, said Tanya Siddiqi, MD, of City of Hope National Medical Center, Duarte, Calif.

“Clinical responses were rapid, improved with time, and were deep and durable,” Dr. Siddiqi said at the annual meeting of the American Society of Hematology.

These findings have provided justification for conducting the phase 2 portion of the study, which is currently enrolling at the higher of two dose levels evaluated in phase 1, she added.

Dr. Siddiqi reported on a total of 23 patients enrolled in the study, known as TRANSCEND CLL 004. All patients had relapsed/refractory chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), and had received at least two prior therapies, including ibrutinib, while about one-third had failed venetoclax as well.

The median patient age was 66 years, and 83% had high-risk features, according to Dr. Siddiqi, who said patients had received a median of five prior lines of therapy.

Nine patients were treated at dose level 1, or 50 x 106 CAR+ T cells, while 14 were treated at dose level 2, or 100 x 106 CAR+ T cells. Two patients experienced grade 3 or 4 dose-limiting toxicities at the second level, including hypertension in one patient, and encephalopathy, muscle weakness, and tumor lysis syndrome (TLS) in the other.

Cytokine release syndrome (CRS) occurred in 17 patients, though only two cases reached grade 3. Neurologic adverse events were seen in nine patients, of which five were grade 3 or 4.

 

 


Partial or complete responses were noted in 81.5%, or 18 of 22 evaluable patients, including 10 (45.5%) who had complete remission. In the subset of nine patients who had failed both ibrutinib and venetoclax, that overall response rate was a “very impressive” 89% (eight of nine patients), said Dr. Siddiqi, including 67% complete remissions (six patients).

Undetectable minimal residual disease (MRD) was reported in 65% and 75% of patients, depending on the method used to evaluate it.

About two-thirds of the patients had responses by day 30 evaluation, and responses deepened over time in about one-quarter, according to Dr. Siddiqi. Of 12 patients with a response at 6 months, 10 (83%) were still in response at 9 months, and 8 patients have been in response for 12 months or longer, she reported.

Neurologic adverse events seen in the CLL/SLL patients in this study were associated with higher lymph node tumor burden, and increased levels of interleukin(IL)-16 or tumor necrosis factor (TNF), according to further analysis presented by Dr. Siddiqi.

That raises the possibility that IL-16 or TNF may be a “good predictive biomarker” for neurotoxicity, which seems to be driven at least in part by lymphadenopathy. “If there was a way that we could combine the CAR T-cell with something like a novel agent that can shrink the tumor burden quickly, then maybe we can have even less toxicities with these CAR T cells,” Dr. Siddiqi said.

Dr. Siddiqi reported disclosures related to Kite, TG Therapeutics, Celgene, Janssen, Seattle Genetics, AstraZeneca, PCYC, Juno Therapeutics, and BeiGene.

SOURCE: Siddiqi T et al. ASH 2019, Abstract 503.

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

– Lisocabtagene maraleucel (liso-cel), a CD19-directed chimeric antigen receptor (CAR) T-cell therapy, has demonstrated manageable toxicity and promising clinical activity in the phase 1 portion of a trial enrolling heavily pretreated patients with chronic lymphocytic leukemia/small lymphocytic lymphoma, according to an investigator.

Dr. Tanya Siddiqi of City of Hope National Medical Center in Duarte, Calif.
Andrew D. Bowser/MDedge News
Dr. Tanya Siddiqi

The overall response rate exceeded 80%, and most patients in response at 6 months had maintained that response at the 9-month mark, said Tanya Siddiqi, MD, of City of Hope National Medical Center, Duarte, Calif.

“Clinical responses were rapid, improved with time, and were deep and durable,” Dr. Siddiqi said at the annual meeting of the American Society of Hematology.

These findings have provided justification for conducting the phase 2 portion of the study, which is currently enrolling at the higher of two dose levels evaluated in phase 1, she added.

Dr. Siddiqi reported on a total of 23 patients enrolled in the study, known as TRANSCEND CLL 004. All patients had relapsed/refractory chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), and had received at least two prior therapies, including ibrutinib, while about one-third had failed venetoclax as well.

The median patient age was 66 years, and 83% had high-risk features, according to Dr. Siddiqi, who said patients had received a median of five prior lines of therapy.

Nine patients were treated at dose level 1, or 50 x 106 CAR+ T cells, while 14 were treated at dose level 2, or 100 x 106 CAR+ T cells. Two patients experienced grade 3 or 4 dose-limiting toxicities at the second level, including hypertension in one patient, and encephalopathy, muscle weakness, and tumor lysis syndrome (TLS) in the other.

Cytokine release syndrome (CRS) occurred in 17 patients, though only two cases reached grade 3. Neurologic adverse events were seen in nine patients, of which five were grade 3 or 4.

 

 


Partial or complete responses were noted in 81.5%, or 18 of 22 evaluable patients, including 10 (45.5%) who had complete remission. In the subset of nine patients who had failed both ibrutinib and venetoclax, that overall response rate was a “very impressive” 89% (eight of nine patients), said Dr. Siddiqi, including 67% complete remissions (six patients).

Undetectable minimal residual disease (MRD) was reported in 65% and 75% of patients, depending on the method used to evaluate it.

About two-thirds of the patients had responses by day 30 evaluation, and responses deepened over time in about one-quarter, according to Dr. Siddiqi. Of 12 patients with a response at 6 months, 10 (83%) were still in response at 9 months, and 8 patients have been in response for 12 months or longer, she reported.

Neurologic adverse events seen in the CLL/SLL patients in this study were associated with higher lymph node tumor burden, and increased levels of interleukin(IL)-16 or tumor necrosis factor (TNF), according to further analysis presented by Dr. Siddiqi.

That raises the possibility that IL-16 or TNF may be a “good predictive biomarker” for neurotoxicity, which seems to be driven at least in part by lymphadenopathy. “If there was a way that we could combine the CAR T-cell with something like a novel agent that can shrink the tumor burden quickly, then maybe we can have even less toxicities with these CAR T cells,” Dr. Siddiqi said.

Dr. Siddiqi reported disclosures related to Kite, TG Therapeutics, Celgene, Janssen, Seattle Genetics, AstraZeneca, PCYC, Juno Therapeutics, and BeiGene.

SOURCE: Siddiqi T et al. ASH 2019, Abstract 503.

– Lisocabtagene maraleucel (liso-cel), a CD19-directed chimeric antigen receptor (CAR) T-cell therapy, has demonstrated manageable toxicity and promising clinical activity in the phase 1 portion of a trial enrolling heavily pretreated patients with chronic lymphocytic leukemia/small lymphocytic lymphoma, according to an investigator.

Dr. Tanya Siddiqi of City of Hope National Medical Center in Duarte, Calif.
Andrew D. Bowser/MDedge News
Dr. Tanya Siddiqi

The overall response rate exceeded 80%, and most patients in response at 6 months had maintained that response at the 9-month mark, said Tanya Siddiqi, MD, of City of Hope National Medical Center, Duarte, Calif.

“Clinical responses were rapid, improved with time, and were deep and durable,” Dr. Siddiqi said at the annual meeting of the American Society of Hematology.

These findings have provided justification for conducting the phase 2 portion of the study, which is currently enrolling at the higher of two dose levels evaluated in phase 1, she added.

Dr. Siddiqi reported on a total of 23 patients enrolled in the study, known as TRANSCEND CLL 004. All patients had relapsed/refractory chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), and had received at least two prior therapies, including ibrutinib, while about one-third had failed venetoclax as well.

The median patient age was 66 years, and 83% had high-risk features, according to Dr. Siddiqi, who said patients had received a median of five prior lines of therapy.

Nine patients were treated at dose level 1, or 50 x 106 CAR+ T cells, while 14 were treated at dose level 2, or 100 x 106 CAR+ T cells. Two patients experienced grade 3 or 4 dose-limiting toxicities at the second level, including hypertension in one patient, and encephalopathy, muscle weakness, and tumor lysis syndrome (TLS) in the other.

Cytokine release syndrome (CRS) occurred in 17 patients, though only two cases reached grade 3. Neurologic adverse events were seen in nine patients, of which five were grade 3 or 4.

 

 


Partial or complete responses were noted in 81.5%, or 18 of 22 evaluable patients, including 10 (45.5%) who had complete remission. In the subset of nine patients who had failed both ibrutinib and venetoclax, that overall response rate was a “very impressive” 89% (eight of nine patients), said Dr. Siddiqi, including 67% complete remissions (six patients).

Undetectable minimal residual disease (MRD) was reported in 65% and 75% of patients, depending on the method used to evaluate it.

About two-thirds of the patients had responses by day 30 evaluation, and responses deepened over time in about one-quarter, according to Dr. Siddiqi. Of 12 patients with a response at 6 months, 10 (83%) were still in response at 9 months, and 8 patients have been in response for 12 months or longer, she reported.

Neurologic adverse events seen in the CLL/SLL patients in this study were associated with higher lymph node tumor burden, and increased levels of interleukin(IL)-16 or tumor necrosis factor (TNF), according to further analysis presented by Dr. Siddiqi.

That raises the possibility that IL-16 or TNF may be a “good predictive biomarker” for neurotoxicity, which seems to be driven at least in part by lymphadenopathy. “If there was a way that we could combine the CAR T-cell with something like a novel agent that can shrink the tumor burden quickly, then maybe we can have even less toxicities with these CAR T cells,” Dr. Siddiqi said.

Dr. Siddiqi reported disclosures related to Kite, TG Therapeutics, Celgene, Janssen, Seattle Genetics, AstraZeneca, PCYC, Juno Therapeutics, and BeiGene.

SOURCE: Siddiqi T et al. ASH 2019, Abstract 503.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASH 2019

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Inhibitor appears to strengthen anti-BCMA CAR T cells in myeloma patients

Article Type
Changed
Mon, 12/23/2019 - 12:59

– A gamma secretase inhibitor could enhance the efficacy of B-cell maturation antigen (BCMA)-directed chimeric antigen receptor (CAR) T cells in patients with relapsed or refractory multiple myeloma, a phase 1 trial suggests.

Dr. Andrew J. Cowan, University of Washington and Fred Hutchinson Cancer Research Center in Seattle
Jennifer Smith/MDedge News
Dr. Andrew J. Cowan

The inhibitor, JSMD194, increased BCMA expression in all 10 patients studied. All patients responded to anti-BCMA CAR T-cell therapy, including three patients who had previously failed BCMA-directed therapy.


Nine patients remain alive and in response at a median follow-up of 20 weeks, with two patients being followed for more than a year. One patient experienced dose-limiting toxicity and died, which prompted a change to the study’s eligibility criteria.

Andrew J. Cowan, MD, of the University of Washington and Fred Hutchinson Cancer Research Center in Seattle, presented these results at the annual meeting of the American Society of Hematology.

 

 


Dr. Cowan and colleagues previously showed that treatment with a gamma secretase inhibitor increased BCMA expression on tumor cells and improved the efficacy of BCMA-targeted CAR T cells in a mouse model of multiple myeloma. The team also showed that a gamma secretase inhibitor could “markedly” increase the percentage of BCMA-positive tumor cells in myeloma patients (Blood. 2019 Nov 7;134[19]:1585-97).

To expand upon these findings, the researchers began a phase 1 trial of BCMA-directed CAR T cells and the oral gamma secretase inhibitor JSMD194 in patients with relapsed/refractory multiple myeloma.

Ten patients have been treated, five men and five women. The patients’ median age at baseline was 66 years (range, 44-74 years). They received a median of 10 prior therapies (range, 4-23). Nine patients had received at least one autologous stem cell transplant, and one patient had two. One patient underwent allogeneic transplant (as well as autologous transplant).

Three patients had received prior BCMA-directed therapy. Two patients had received BCMA-directed CAR T cells. One of them did not respond, and the other responded but relapsed. The third patient received a BCMA-targeted bispecific T-cell engager and did not respond.

Study treatment

Patients had BCMA expression measured at baseline, then underwent apheresis for CAR T-cell production.

Patients received JSMD194 at 25 mg on days 1, 3, and 5. Then, they received cyclophosphamide at 300 mg and fludarabine at 25 mg for 3 days.

Next, patients received a single CAR T-cell infusion at a dose of 50 x 106 (n = 5), 150 x 106 (n = 3), or 300 x 106 (n = 2). They also received JSMD194 at 25 mg three times a week for 3 weeks.

 

 

Safety

“Nearly all patients had a serious adverse event, which was typically admission to the hospital for neutropenic fever,” Dr. Cowan said.

One patient experienced dose-limiting toxicity and died at day 33. The patient had a disseminating fungal infection, grade 4 cytokine release syndrome (CRS), and neurotoxicity. The patient’s death prompted the researchers to include performance status in the study’s eligibility criteria.

All patients developed CRS. Only the aforementioned patient had grade 4 CRS, and three patients had grade 3 CRS. Six patients experienced neurotoxicity. There were no cases of tumor lysis syndrome.
 

Efficacy

“All patients experienced an increase of cells expressing BCMA,” Dr. Cowan said. “While there was significant variability in BCMA expression at baseline, all cells expressed BCMA after three doses of the gamma secretase inhibitor.”

The median BCMA expression after JSMD194 treatment was 99% (range, 96%-100%), and there was a median 20-fold (range, 8- to 157-fold) increase in BCMA surface density.

The overall response rate was 100%. Two patients achieved a stringent complete response (CR), one achieved a CR, five patients had a very good partial response, and two had a partial response.

The patient with a CR received the 50 x 106 dose of CAR T cells, and the patients with stringent CRs received the 150 x 106 and 300 x 106 doses.

Of the three patients who previously received BCMA-directed therapy, two achieved a very good partial response, and one had a partial response.

Nine of the 10 patients are still alive and in response, with a median follow-up of 20 weeks. The longest follow-up is 444 days.

“To date, all patients have evidence of durable responses,” Dr. Cowan said. “Moreover, all patients had dramatic reductions in involved serum free light chain ... and serum monoclonal proteins.”

Dr. Cowan noted that longer follow-up is needed to assess CAR T-cell persistence and the durability of response.

This trial is sponsored by the Fred Hutchinson Cancer Research Center in collaboration with the National Cancer Institute. Two researchers involved in this work are employees of Juno Therapeutics. Dr. Cowan reported relationships with Juno Therapeutics, Janssen, Celgene, AbbVie, Cellectar, and Sanofi.

SOURCE: Cowan AJ et al. ASH 2019. Abstract 204.

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

– A gamma secretase inhibitor could enhance the efficacy of B-cell maturation antigen (BCMA)-directed chimeric antigen receptor (CAR) T cells in patients with relapsed or refractory multiple myeloma, a phase 1 trial suggests.

Dr. Andrew J. Cowan, University of Washington and Fred Hutchinson Cancer Research Center in Seattle
Jennifer Smith/MDedge News
Dr. Andrew J. Cowan

The inhibitor, JSMD194, increased BCMA expression in all 10 patients studied. All patients responded to anti-BCMA CAR T-cell therapy, including three patients who had previously failed BCMA-directed therapy.


Nine patients remain alive and in response at a median follow-up of 20 weeks, with two patients being followed for more than a year. One patient experienced dose-limiting toxicity and died, which prompted a change to the study’s eligibility criteria.

Andrew J. Cowan, MD, of the University of Washington and Fred Hutchinson Cancer Research Center in Seattle, presented these results at the annual meeting of the American Society of Hematology.

 

 


Dr. Cowan and colleagues previously showed that treatment with a gamma secretase inhibitor increased BCMA expression on tumor cells and improved the efficacy of BCMA-targeted CAR T cells in a mouse model of multiple myeloma. The team also showed that a gamma secretase inhibitor could “markedly” increase the percentage of BCMA-positive tumor cells in myeloma patients (Blood. 2019 Nov 7;134[19]:1585-97).

To expand upon these findings, the researchers began a phase 1 trial of BCMA-directed CAR T cells and the oral gamma secretase inhibitor JSMD194 in patients with relapsed/refractory multiple myeloma.

Ten patients have been treated, five men and five women. The patients’ median age at baseline was 66 years (range, 44-74 years). They received a median of 10 prior therapies (range, 4-23). Nine patients had received at least one autologous stem cell transplant, and one patient had two. One patient underwent allogeneic transplant (as well as autologous transplant).

Three patients had received prior BCMA-directed therapy. Two patients had received BCMA-directed CAR T cells. One of them did not respond, and the other responded but relapsed. The third patient received a BCMA-targeted bispecific T-cell engager and did not respond.

Study treatment

Patients had BCMA expression measured at baseline, then underwent apheresis for CAR T-cell production.

Patients received JSMD194 at 25 mg on days 1, 3, and 5. Then, they received cyclophosphamide at 300 mg and fludarabine at 25 mg for 3 days.

Next, patients received a single CAR T-cell infusion at a dose of 50 x 106 (n = 5), 150 x 106 (n = 3), or 300 x 106 (n = 2). They also received JSMD194 at 25 mg three times a week for 3 weeks.

 

 

Safety

“Nearly all patients had a serious adverse event, which was typically admission to the hospital for neutropenic fever,” Dr. Cowan said.

One patient experienced dose-limiting toxicity and died at day 33. The patient had a disseminating fungal infection, grade 4 cytokine release syndrome (CRS), and neurotoxicity. The patient’s death prompted the researchers to include performance status in the study’s eligibility criteria.

All patients developed CRS. Only the aforementioned patient had grade 4 CRS, and three patients had grade 3 CRS. Six patients experienced neurotoxicity. There were no cases of tumor lysis syndrome.
 

Efficacy

“All patients experienced an increase of cells expressing BCMA,” Dr. Cowan said. “While there was significant variability in BCMA expression at baseline, all cells expressed BCMA after three doses of the gamma secretase inhibitor.”

The median BCMA expression after JSMD194 treatment was 99% (range, 96%-100%), and there was a median 20-fold (range, 8- to 157-fold) increase in BCMA surface density.

The overall response rate was 100%. Two patients achieved a stringent complete response (CR), one achieved a CR, five patients had a very good partial response, and two had a partial response.

The patient with a CR received the 50 x 106 dose of CAR T cells, and the patients with stringent CRs received the 150 x 106 and 300 x 106 doses.

Of the three patients who previously received BCMA-directed therapy, two achieved a very good partial response, and one had a partial response.

Nine of the 10 patients are still alive and in response, with a median follow-up of 20 weeks. The longest follow-up is 444 days.

“To date, all patients have evidence of durable responses,” Dr. Cowan said. “Moreover, all patients had dramatic reductions in involved serum free light chain ... and serum monoclonal proteins.”

Dr. Cowan noted that longer follow-up is needed to assess CAR T-cell persistence and the durability of response.

This trial is sponsored by the Fred Hutchinson Cancer Research Center in collaboration with the National Cancer Institute. Two researchers involved in this work are employees of Juno Therapeutics. Dr. Cowan reported relationships with Juno Therapeutics, Janssen, Celgene, AbbVie, Cellectar, and Sanofi.

SOURCE: Cowan AJ et al. ASH 2019. Abstract 204.

– A gamma secretase inhibitor could enhance the efficacy of B-cell maturation antigen (BCMA)-directed chimeric antigen receptor (CAR) T cells in patients with relapsed or refractory multiple myeloma, a phase 1 trial suggests.

Dr. Andrew J. Cowan, University of Washington and Fred Hutchinson Cancer Research Center in Seattle
Jennifer Smith/MDedge News
Dr. Andrew J. Cowan

The inhibitor, JSMD194, increased BCMA expression in all 10 patients studied. All patients responded to anti-BCMA CAR T-cell therapy, including three patients who had previously failed BCMA-directed therapy.


Nine patients remain alive and in response at a median follow-up of 20 weeks, with two patients being followed for more than a year. One patient experienced dose-limiting toxicity and died, which prompted a change to the study’s eligibility criteria.

Andrew J. Cowan, MD, of the University of Washington and Fred Hutchinson Cancer Research Center in Seattle, presented these results at the annual meeting of the American Society of Hematology.

 

 


Dr. Cowan and colleagues previously showed that treatment with a gamma secretase inhibitor increased BCMA expression on tumor cells and improved the efficacy of BCMA-targeted CAR T cells in a mouse model of multiple myeloma. The team also showed that a gamma secretase inhibitor could “markedly” increase the percentage of BCMA-positive tumor cells in myeloma patients (Blood. 2019 Nov 7;134[19]:1585-97).

To expand upon these findings, the researchers began a phase 1 trial of BCMA-directed CAR T cells and the oral gamma secretase inhibitor JSMD194 in patients with relapsed/refractory multiple myeloma.

Ten patients have been treated, five men and five women. The patients’ median age at baseline was 66 years (range, 44-74 years). They received a median of 10 prior therapies (range, 4-23). Nine patients had received at least one autologous stem cell transplant, and one patient had two. One patient underwent allogeneic transplant (as well as autologous transplant).

Three patients had received prior BCMA-directed therapy. Two patients had received BCMA-directed CAR T cells. One of them did not respond, and the other responded but relapsed. The third patient received a BCMA-targeted bispecific T-cell engager and did not respond.

Study treatment

Patients had BCMA expression measured at baseline, then underwent apheresis for CAR T-cell production.

Patients received JSMD194 at 25 mg on days 1, 3, and 5. Then, they received cyclophosphamide at 300 mg and fludarabine at 25 mg for 3 days.

Next, patients received a single CAR T-cell infusion at a dose of 50 x 106 (n = 5), 150 x 106 (n = 3), or 300 x 106 (n = 2). They also received JSMD194 at 25 mg three times a week for 3 weeks.

 

 

Safety

“Nearly all patients had a serious adverse event, which was typically admission to the hospital for neutropenic fever,” Dr. Cowan said.

One patient experienced dose-limiting toxicity and died at day 33. The patient had a disseminating fungal infection, grade 4 cytokine release syndrome (CRS), and neurotoxicity. The patient’s death prompted the researchers to include performance status in the study’s eligibility criteria.

All patients developed CRS. Only the aforementioned patient had grade 4 CRS, and three patients had grade 3 CRS. Six patients experienced neurotoxicity. There were no cases of tumor lysis syndrome.
 

Efficacy

“All patients experienced an increase of cells expressing BCMA,” Dr. Cowan said. “While there was significant variability in BCMA expression at baseline, all cells expressed BCMA after three doses of the gamma secretase inhibitor.”

The median BCMA expression after JSMD194 treatment was 99% (range, 96%-100%), and there was a median 20-fold (range, 8- to 157-fold) increase in BCMA surface density.

The overall response rate was 100%. Two patients achieved a stringent complete response (CR), one achieved a CR, five patients had a very good partial response, and two had a partial response.

The patient with a CR received the 50 x 106 dose of CAR T cells, and the patients with stringent CRs received the 150 x 106 and 300 x 106 doses.

Of the three patients who previously received BCMA-directed therapy, two achieved a very good partial response, and one had a partial response.

Nine of the 10 patients are still alive and in response, with a median follow-up of 20 weeks. The longest follow-up is 444 days.

“To date, all patients have evidence of durable responses,” Dr. Cowan said. “Moreover, all patients had dramatic reductions in involved serum free light chain ... and serum monoclonal proteins.”

Dr. Cowan noted that longer follow-up is needed to assess CAR T-cell persistence and the durability of response.

This trial is sponsored by the Fred Hutchinson Cancer Research Center in collaboration with the National Cancer Institute. Two researchers involved in this work are employees of Juno Therapeutics. Dr. Cowan reported relationships with Juno Therapeutics, Janssen, Celgene, AbbVie, Cellectar, and Sanofi.

SOURCE: Cowan AJ et al. ASH 2019. Abstract 204.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASH 2019

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

LOXO-305: Next-gen BTK inhibitor safe and effective in B-cell malignancies

Article Type
Changed
Tue, 01/17/2023 - 11:24

– A phase 1 trial of the next-generation Bruton tyrosine kinase (BTK) inhibitor LOXO-305 has demonstrated safety and provided evidence of its efficacy in heavily pretreated patients with B-cell malignancies, including some with acquired resistance to other BTK inhibitors and venetoclax, according to an investigator.

Dr. Anthony R. Mato, Sloan Kettering Cancer Center, New York
Andrew D. Bowser/MDedge News
Dr. Anthony R. Mato

The antitumor activity of this highly selective investigational oral BTK inhibitor was significant in patients with chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL), with a rapid onset of action and resolution of lymphocytosis “consistent with effective BTK target inhibition,” said Anthony R. Mato, MD, of the Center for CLL at Memorial Sloan Kettering Cancer Center in New York.

Responses were also seen in patients with BTK C481 mutations, the primary cause of progressive CLL after BTK inhibitor use, Dr. Mato said at the annual meeting of the American Society of Hematology.

The safety and tolerability profile of LOXO-305 is “consistent with highly selective drug design,” with no evidence of off-target effects, he said. “Collectively, these data demonstrate that BTK remains a highly actionable target despite progression on covalent BTK inhibitors.”

While BTK inhibitors have transformed treatment of B-cell malignancies, resistance remains a major problem, said Dr. Mato, citing 5-year ibrutinib discontinuation rates of 41% in the front line setting and 53.7% in the relapsed/refractory setting.

Key reasons for discontinuation are intolerance, events such as atrial fibrillation and major bleeding, progression of disease, and the appearance of BTK C481 mutations, which prevent covalent BTK inhibitors from achieving effective target inhibition, he said. In contrast, LOXO-305 is designed to non-covalently bind to BTK, regardless of C481 status.

Dr. Mato described results of the phase 1 BRUIN trial, in which 28 adult patients with CLL or B-cell non-Hodgkin lymphomas received once daily oral LOXO-305 at doses ranging from 25 mg to 200 mg. All patients had received at least two lines of prior therapy and had active disease in need of treatment.

For 13 evaluable CLL patients, the overall response rate was 77% (10 patients), Dr. Mato reported. Overall response rates for MCL and other B-cell malignancies were 50%, or three out of six MCL patients and two of four patients with Waldenström macroglobulinemia, diffuse large B-cell lymphoma, or marginal zone lymphoma.

Though only a small subset of CLL patients have had multiple response assessments, the available data suggest that responses “deepen over time” with continued LOXO-305 treatment, Dr. Mato said.

With the median follow-up of 2.7 months, 24 of 28 patients remain on therapy, including all responders. “Some of the responses appear to be quite durable,” Dr. Mato said.

There have been no dose-limiting toxicities, the maximum tolerated dose has not been reached, and there have been no notable adverse events characteristic of covalent BTK inhibitors – namely atrial fibrillation or major bleeding – despite frequent monitoring, according to Dr. Mato.

There were two grade 3 events (leukocytosis and neutropenia), but the remaining treatment-emergent adverse events have been grade 1-2. “Having managed many of these patients, I can tell you that these adverse events were quite manageable,” Dr. Mato told attendees.

BRUIN is a global trial that continues to enroll patients at 18 sites in 3 countries, with a plan in 2020 to incorporate “rational combinations” of agents, according to the investigator.

Dr. Mato reported disclosures related to Gilead, Pharmacyclics, AstraZeneca, AbbVie, Sunesis, Johnson & Johnson, TG Therapeutics, LOXO, DTRM Biopharma, Genentech, Janssen, Acerta, Pharmacyclics, and Celgene.

SOURCE: Mato AR et al. ASH 2019, Abstract 501.

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

– A phase 1 trial of the next-generation Bruton tyrosine kinase (BTK) inhibitor LOXO-305 has demonstrated safety and provided evidence of its efficacy in heavily pretreated patients with B-cell malignancies, including some with acquired resistance to other BTK inhibitors and venetoclax, according to an investigator.

Dr. Anthony R. Mato, Sloan Kettering Cancer Center, New York
Andrew D. Bowser/MDedge News
Dr. Anthony R. Mato

The antitumor activity of this highly selective investigational oral BTK inhibitor was significant in patients with chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL), with a rapid onset of action and resolution of lymphocytosis “consistent with effective BTK target inhibition,” said Anthony R. Mato, MD, of the Center for CLL at Memorial Sloan Kettering Cancer Center in New York.

Responses were also seen in patients with BTK C481 mutations, the primary cause of progressive CLL after BTK inhibitor use, Dr. Mato said at the annual meeting of the American Society of Hematology.

The safety and tolerability profile of LOXO-305 is “consistent with highly selective drug design,” with no evidence of off-target effects, he said. “Collectively, these data demonstrate that BTK remains a highly actionable target despite progression on covalent BTK inhibitors.”

While BTK inhibitors have transformed treatment of B-cell malignancies, resistance remains a major problem, said Dr. Mato, citing 5-year ibrutinib discontinuation rates of 41% in the front line setting and 53.7% in the relapsed/refractory setting.

Key reasons for discontinuation are intolerance, events such as atrial fibrillation and major bleeding, progression of disease, and the appearance of BTK C481 mutations, which prevent covalent BTK inhibitors from achieving effective target inhibition, he said. In contrast, LOXO-305 is designed to non-covalently bind to BTK, regardless of C481 status.

Dr. Mato described results of the phase 1 BRUIN trial, in which 28 adult patients with CLL or B-cell non-Hodgkin lymphomas received once daily oral LOXO-305 at doses ranging from 25 mg to 200 mg. All patients had received at least two lines of prior therapy and had active disease in need of treatment.

For 13 evaluable CLL patients, the overall response rate was 77% (10 patients), Dr. Mato reported. Overall response rates for MCL and other B-cell malignancies were 50%, or three out of six MCL patients and two of four patients with Waldenström macroglobulinemia, diffuse large B-cell lymphoma, or marginal zone lymphoma.

Though only a small subset of CLL patients have had multiple response assessments, the available data suggest that responses “deepen over time” with continued LOXO-305 treatment, Dr. Mato said.

With the median follow-up of 2.7 months, 24 of 28 patients remain on therapy, including all responders. “Some of the responses appear to be quite durable,” Dr. Mato said.

There have been no dose-limiting toxicities, the maximum tolerated dose has not been reached, and there have been no notable adverse events characteristic of covalent BTK inhibitors – namely atrial fibrillation or major bleeding – despite frequent monitoring, according to Dr. Mato.

There were two grade 3 events (leukocytosis and neutropenia), but the remaining treatment-emergent adverse events have been grade 1-2. “Having managed many of these patients, I can tell you that these adverse events were quite manageable,” Dr. Mato told attendees.

BRUIN is a global trial that continues to enroll patients at 18 sites in 3 countries, with a plan in 2020 to incorporate “rational combinations” of agents, according to the investigator.

Dr. Mato reported disclosures related to Gilead, Pharmacyclics, AstraZeneca, AbbVie, Sunesis, Johnson & Johnson, TG Therapeutics, LOXO, DTRM Biopharma, Genentech, Janssen, Acerta, Pharmacyclics, and Celgene.

SOURCE: Mato AR et al. ASH 2019, Abstract 501.

– A phase 1 trial of the next-generation Bruton tyrosine kinase (BTK) inhibitor LOXO-305 has demonstrated safety and provided evidence of its efficacy in heavily pretreated patients with B-cell malignancies, including some with acquired resistance to other BTK inhibitors and venetoclax, according to an investigator.

Dr. Anthony R. Mato, Sloan Kettering Cancer Center, New York
Andrew D. Bowser/MDedge News
Dr. Anthony R. Mato

The antitumor activity of this highly selective investigational oral BTK inhibitor was significant in patients with chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL), with a rapid onset of action and resolution of lymphocytosis “consistent with effective BTK target inhibition,” said Anthony R. Mato, MD, of the Center for CLL at Memorial Sloan Kettering Cancer Center in New York.

Responses were also seen in patients with BTK C481 mutations, the primary cause of progressive CLL after BTK inhibitor use, Dr. Mato said at the annual meeting of the American Society of Hematology.

The safety and tolerability profile of LOXO-305 is “consistent with highly selective drug design,” with no evidence of off-target effects, he said. “Collectively, these data demonstrate that BTK remains a highly actionable target despite progression on covalent BTK inhibitors.”

While BTK inhibitors have transformed treatment of B-cell malignancies, resistance remains a major problem, said Dr. Mato, citing 5-year ibrutinib discontinuation rates of 41% in the front line setting and 53.7% in the relapsed/refractory setting.

Key reasons for discontinuation are intolerance, events such as atrial fibrillation and major bleeding, progression of disease, and the appearance of BTK C481 mutations, which prevent covalent BTK inhibitors from achieving effective target inhibition, he said. In contrast, LOXO-305 is designed to non-covalently bind to BTK, regardless of C481 status.

Dr. Mato described results of the phase 1 BRUIN trial, in which 28 adult patients with CLL or B-cell non-Hodgkin lymphomas received once daily oral LOXO-305 at doses ranging from 25 mg to 200 mg. All patients had received at least two lines of prior therapy and had active disease in need of treatment.

For 13 evaluable CLL patients, the overall response rate was 77% (10 patients), Dr. Mato reported. Overall response rates for MCL and other B-cell malignancies were 50%, or three out of six MCL patients and two of four patients with Waldenström macroglobulinemia, diffuse large B-cell lymphoma, or marginal zone lymphoma.

Though only a small subset of CLL patients have had multiple response assessments, the available data suggest that responses “deepen over time” with continued LOXO-305 treatment, Dr. Mato said.

With the median follow-up of 2.7 months, 24 of 28 patients remain on therapy, including all responders. “Some of the responses appear to be quite durable,” Dr. Mato said.

There have been no dose-limiting toxicities, the maximum tolerated dose has not been reached, and there have been no notable adverse events characteristic of covalent BTK inhibitors – namely atrial fibrillation or major bleeding – despite frequent monitoring, according to Dr. Mato.

There were two grade 3 events (leukocytosis and neutropenia), but the remaining treatment-emergent adverse events have been grade 1-2. “Having managed many of these patients, I can tell you that these adverse events were quite manageable,” Dr. Mato told attendees.

BRUIN is a global trial that continues to enroll patients at 18 sites in 3 countries, with a plan in 2020 to incorporate “rational combinations” of agents, according to the investigator.

Dr. Mato reported disclosures related to Gilead, Pharmacyclics, AstraZeneca, AbbVie, Sunesis, Johnson & Johnson, TG Therapeutics, LOXO, DTRM Biopharma, Genentech, Janssen, Acerta, Pharmacyclics, and Celgene.

SOURCE: Mato AR et al. ASH 2019, Abstract 501.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASH 2019

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Survival data reported from largest CAR T trial in B-cell lymphoma

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

– Updated results from the TRANSCEND NHL trial include survival data with lisocabtagene maraleucel (liso-cel), an anti-CD19 chimeric antigen receptor (CAR) T-cell therapy, in patients with relapsed/refractory B-cell lymphomas.

Dr. Jeremy S. Abramson, of Massachusetts General Hospital in Boston
Jennifer Smith/MDedge News
Dr. Jeremy S. Abramson

The median progression-free survival (PFS) was 6.8 months, and the median overall survival was 21.1 months. PFS results were best among complete responders and among patients with primary mediastinal large B-cell lymphoma or transformed follicular lymphoma.

Jeremy S. Abramson, MD, of Massachusetts General Hospital in Boston, presented these results at the annual meeting of the American Society of Hematology.

“TRANSCEND NHL is the largest clinical study to date of CD19-directed CAR T cells in patients with relapsed/refractory aggressive B-cell lymphoma,” Dr. Abramson said.



The phase 1 trial (NCT02631044) includes 269 patients who received liso-cel. They were diagnosed with transformed follicular lymphoma (22%) or other indolent lymphoma (7%), high-grade B-cell lymphoma (13%), primary mediastinal large B-cell lymphoma (6%), grade 3B follicular lymphoma (1%), or diffuse large B-cell lymphoma not otherwise specified (51%).

At baseline, patients had received a median of three prior systemic therapies (range, one to eight). Some patients had received autologous (33%) or allogeneic (3%) transplant. Many patients were chemotherapy refractory (67%) or had never achieved a complete response to prior therapy (44%).

More than half of patients (59%) received bridging therapy during liso-cel manufacturing. All patients received lymphodepletion with fludarabine and cyclophosphamide, followed by liso-cel at 50 x 106 CAR T cells, 100 x 106 CAR T cells, or 150 x 106 CAR T cells.

Response and survival

The median follow-up was 12.0 months. The overall response rate was 73%, and the complete response rate was 53%.

“Remissions were rapid, with a median of 1 month from CAR T-cell infusion, and durable, with a median duration of response that has not been reached and 55% of patients remaining in response at 1 year,” Dr. Abramson said.

The median PFS was 6.8 months overall, not reached for patients who achieved a complete response, 2.8 months for patients with a partial response, and 1.1 months for patients with stable disease or progressive disease.



The median PFS was not reached for patients with primary mediastinal large B-cell lymphoma or transformed follicular lymphoma, 5.0 months for high-grade B-cell lymphoma, 3.0 months for diffuse large B-cell lymphoma not otherwise specified, and 2.9 months in transformed indolent non-Hodgkin lymphoma.

The median overall survival was 21.1 months overall, not reached for patients who achieved a complete response, 9.0 months for patients who had a partial response, and 5.1 months for patients with stable disease or progressive disease.

Safety

Common treatment-emergent adverse events were neutropenia (63%), anemia (48%), fatigue (44%), nausea (33%), thrombocytopenia (31%), headache (30%), decreased appetite (28%), and diarrhea (26%).

Cytokine release syndrome (CRS) occurred in 42% of patients, and neurologic events occurred in 30%. Grade 3-4 CRS occurred in 2% of patients, and grade 3-4 neurologic events occurred in 10%. There were no cases of grade 5 CRS or neurologic events.



The median time to CRS onset was 5 days, and the median time to onset of neurologic events was 9 days. The median time to resolution of CRS and neurologic events was 5 days and 11 days, respectively.

“The low incidence of severe CRS and neurologic events and their late time of onset support using this product in a large range of patients and in the outpatient setting,” Dr. Abramson said.



There were seven grade 5 treatment-related adverse events, including diffuse alveolar damage, pulmonary hemorrhage, multiple organ dysfunction syndrome, cardiomyopathy, fludarabine leukoencephalopathy, septic shock, and progressive multifocal leukoencephalopathy.

This trial is sponsored by Bristol-Myers Squibb. Dr. Abramson reported relationships with Juno Therapeutics and Celgene, now owned by Bristol-Myers Squibb, and a range of other companies.

SOURCE: Abramson JS et al. ASH 2019, Abstract 241.

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

– Updated results from the TRANSCEND NHL trial include survival data with lisocabtagene maraleucel (liso-cel), an anti-CD19 chimeric antigen receptor (CAR) T-cell therapy, in patients with relapsed/refractory B-cell lymphomas.

Dr. Jeremy S. Abramson, of Massachusetts General Hospital in Boston
Jennifer Smith/MDedge News
Dr. Jeremy S. Abramson

The median progression-free survival (PFS) was 6.8 months, and the median overall survival was 21.1 months. PFS results were best among complete responders and among patients with primary mediastinal large B-cell lymphoma or transformed follicular lymphoma.

Jeremy S. Abramson, MD, of Massachusetts General Hospital in Boston, presented these results at the annual meeting of the American Society of Hematology.

“TRANSCEND NHL is the largest clinical study to date of CD19-directed CAR T cells in patients with relapsed/refractory aggressive B-cell lymphoma,” Dr. Abramson said.



The phase 1 trial (NCT02631044) includes 269 patients who received liso-cel. They were diagnosed with transformed follicular lymphoma (22%) or other indolent lymphoma (7%), high-grade B-cell lymphoma (13%), primary mediastinal large B-cell lymphoma (6%), grade 3B follicular lymphoma (1%), or diffuse large B-cell lymphoma not otherwise specified (51%).

At baseline, patients had received a median of three prior systemic therapies (range, one to eight). Some patients had received autologous (33%) or allogeneic (3%) transplant. Many patients were chemotherapy refractory (67%) or had never achieved a complete response to prior therapy (44%).

More than half of patients (59%) received bridging therapy during liso-cel manufacturing. All patients received lymphodepletion with fludarabine and cyclophosphamide, followed by liso-cel at 50 x 106 CAR T cells, 100 x 106 CAR T cells, or 150 x 106 CAR T cells.

Response and survival

The median follow-up was 12.0 months. The overall response rate was 73%, and the complete response rate was 53%.

“Remissions were rapid, with a median of 1 month from CAR T-cell infusion, and durable, with a median duration of response that has not been reached and 55% of patients remaining in response at 1 year,” Dr. Abramson said.

The median PFS was 6.8 months overall, not reached for patients who achieved a complete response, 2.8 months for patients with a partial response, and 1.1 months for patients with stable disease or progressive disease.



The median PFS was not reached for patients with primary mediastinal large B-cell lymphoma or transformed follicular lymphoma, 5.0 months for high-grade B-cell lymphoma, 3.0 months for diffuse large B-cell lymphoma not otherwise specified, and 2.9 months in transformed indolent non-Hodgkin lymphoma.

The median overall survival was 21.1 months overall, not reached for patients who achieved a complete response, 9.0 months for patients who had a partial response, and 5.1 months for patients with stable disease or progressive disease.

Safety

Common treatment-emergent adverse events were neutropenia (63%), anemia (48%), fatigue (44%), nausea (33%), thrombocytopenia (31%), headache (30%), decreased appetite (28%), and diarrhea (26%).

Cytokine release syndrome (CRS) occurred in 42% of patients, and neurologic events occurred in 30%. Grade 3-4 CRS occurred in 2% of patients, and grade 3-4 neurologic events occurred in 10%. There were no cases of grade 5 CRS or neurologic events.



The median time to CRS onset was 5 days, and the median time to onset of neurologic events was 9 days. The median time to resolution of CRS and neurologic events was 5 days and 11 days, respectively.

“The low incidence of severe CRS and neurologic events and their late time of onset support using this product in a large range of patients and in the outpatient setting,” Dr. Abramson said.



There were seven grade 5 treatment-related adverse events, including diffuse alveolar damage, pulmonary hemorrhage, multiple organ dysfunction syndrome, cardiomyopathy, fludarabine leukoencephalopathy, septic shock, and progressive multifocal leukoencephalopathy.

This trial is sponsored by Bristol-Myers Squibb. Dr. Abramson reported relationships with Juno Therapeutics and Celgene, now owned by Bristol-Myers Squibb, and a range of other companies.

SOURCE: Abramson JS et al. ASH 2019, Abstract 241.

– Updated results from the TRANSCEND NHL trial include survival data with lisocabtagene maraleucel (liso-cel), an anti-CD19 chimeric antigen receptor (CAR) T-cell therapy, in patients with relapsed/refractory B-cell lymphomas.

Dr. Jeremy S. Abramson, of Massachusetts General Hospital in Boston
Jennifer Smith/MDedge News
Dr. Jeremy S. Abramson

The median progression-free survival (PFS) was 6.8 months, and the median overall survival was 21.1 months. PFS results were best among complete responders and among patients with primary mediastinal large B-cell lymphoma or transformed follicular lymphoma.

Jeremy S. Abramson, MD, of Massachusetts General Hospital in Boston, presented these results at the annual meeting of the American Society of Hematology.

“TRANSCEND NHL is the largest clinical study to date of CD19-directed CAR T cells in patients with relapsed/refractory aggressive B-cell lymphoma,” Dr. Abramson said.



The phase 1 trial (NCT02631044) includes 269 patients who received liso-cel. They were diagnosed with transformed follicular lymphoma (22%) or other indolent lymphoma (7%), high-grade B-cell lymphoma (13%), primary mediastinal large B-cell lymphoma (6%), grade 3B follicular lymphoma (1%), or diffuse large B-cell lymphoma not otherwise specified (51%).

At baseline, patients had received a median of three prior systemic therapies (range, one to eight). Some patients had received autologous (33%) or allogeneic (3%) transplant. Many patients were chemotherapy refractory (67%) or had never achieved a complete response to prior therapy (44%).

More than half of patients (59%) received bridging therapy during liso-cel manufacturing. All patients received lymphodepletion with fludarabine and cyclophosphamide, followed by liso-cel at 50 x 106 CAR T cells, 100 x 106 CAR T cells, or 150 x 106 CAR T cells.

Response and survival

The median follow-up was 12.0 months. The overall response rate was 73%, and the complete response rate was 53%.

“Remissions were rapid, with a median of 1 month from CAR T-cell infusion, and durable, with a median duration of response that has not been reached and 55% of patients remaining in response at 1 year,” Dr. Abramson said.

The median PFS was 6.8 months overall, not reached for patients who achieved a complete response, 2.8 months for patients with a partial response, and 1.1 months for patients with stable disease or progressive disease.



The median PFS was not reached for patients with primary mediastinal large B-cell lymphoma or transformed follicular lymphoma, 5.0 months for high-grade B-cell lymphoma, 3.0 months for diffuse large B-cell lymphoma not otherwise specified, and 2.9 months in transformed indolent non-Hodgkin lymphoma.

The median overall survival was 21.1 months overall, not reached for patients who achieved a complete response, 9.0 months for patients who had a partial response, and 5.1 months for patients with stable disease or progressive disease.

Safety

Common treatment-emergent adverse events were neutropenia (63%), anemia (48%), fatigue (44%), nausea (33%), thrombocytopenia (31%), headache (30%), decreased appetite (28%), and diarrhea (26%).

Cytokine release syndrome (CRS) occurred in 42% of patients, and neurologic events occurred in 30%. Grade 3-4 CRS occurred in 2% of patients, and grade 3-4 neurologic events occurred in 10%. There were no cases of grade 5 CRS or neurologic events.



The median time to CRS onset was 5 days, and the median time to onset of neurologic events was 9 days. The median time to resolution of CRS and neurologic events was 5 days and 11 days, respectively.

“The low incidence of severe CRS and neurologic events and their late time of onset support using this product in a large range of patients and in the outpatient setting,” Dr. Abramson said.



There were seven grade 5 treatment-related adverse events, including diffuse alveolar damage, pulmonary hemorrhage, multiple organ dysfunction syndrome, cardiomyopathy, fludarabine leukoencephalopathy, septic shock, and progressive multifocal leukoencephalopathy.

This trial is sponsored by Bristol-Myers Squibb. Dr. Abramson reported relationships with Juno Therapeutics and Celgene, now owned by Bristol-Myers Squibb, and a range of other companies.

SOURCE: Abramson JS et al. ASH 2019, Abstract 241.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASH 2019

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Age, sex, and other factors linked to risk of intracranial hemorrhage in ITP

Article Type
Changed
Tue, 12/17/2019 - 15:56

– A large, retrospective study suggests several factors are associated with an increased risk of intracranial hemorrhage in patients with immune thrombocytopenia.

Mayank Sharma, of the University of Miami
Jennifer Smith/MDedge News
Mayank Sharma

Data on more than 300,000 immune thrombocytopenia (ITP) hospitalizations indicated that older age, male sex, not having private insurance, having a gastrointestinal or “other” bleed, and receiving treatment at a hospital in the western United States, a medium- or large-sized hospital, or an urban teaching hospital were all associated with an increased risk of intracranial hemorrhage (ICH).

Mayank Sharma, of the University of Miami, detailed these findings at the annual meeting of the American Society of Hematology.

Mr. Sharma and colleagues analyzed data from the National Inpatient Sample database from 2007 to 2016. Of the 348,906 ITP hospitalizations included, there were 3,408 (0.98%) cases of ICH.

The overall incidence of ICH was low and remained stable over time, “which is reassuring,” Mr. Sharma said. However, the mortality rate was higher among patients with ICH than among those without it – 26.7% and 3.2%, respectively.

A multivariate analysis showed that female patients had a decreased likelihood of ICH, with an odds ratio of 0.81 (95% confidence interval, 0.68-0.97). Patients with private insurance had a decreased likelihood of ICH as well, with an OR of 0.81 (95% CI, 0.61-1.08).

Conversely, older patients had an increased likelihood of ICH. The OR was 2.23 (95% CI, 1.51-3.31) for patients aged 25-64 years, and the OR was 3.69 (95% CI, 2.34-5.84) for patients aged 65 years and older.

Patients with a gastrointestinal bleed or an other bleed (not including hematuria or epistaxis) had an increased likelihood of ICH. The ORs were 1.60 (95% CI, 1.18-2.16) and 1.69 (95% CI, 1.19-2.42), respectively.

Patients hospitalized in the western United States (OR, 1.62; 95% CI, 1.26-2.08), at a medium-sized hospital (OR, 1.64; 95% CI, 1.08-2.47), at a large hospital (OR, 2.42; 95% CI, 1.65-3.55), or at an urban teaching hospital (OR, 2.73; 95% CI, 1.80-4.13) all had an increased likelihood of ICH.

“Our second objective was to study the factors associated with mortality in ITP patients with ICH,” Mr. Sharma said. “We found female gender and Medicaid, private, or self-pay as primary payers to be associated with a lower mortality in ITP with ICH.

“[A]ge of 25-64 and 65 years and above, coexistence of a GI bleed or other bleed, and admission to a large or urban teaching hospital were associated with a higher mortality,” he added.

Mr. Sharma said the study’s strengths are that it is the most recent study on trends in ITP/ICH hospitalizations, and that it’s a longitudinal assessment of data from a nationally representative database.

The study’s limitations include its retrospective nature and the use of ICD codes, which could lead to inaccuracies. Data on prior therapies and long-term outcomes were not available, and the researchers were unable to differentiate between acute and chronic ITP.

Mr. Sharma said he had no relevant conflicts of interest.
 

SOURCE: Sharma M et al. ASH 2019, Abstract 55.

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

– A large, retrospective study suggests several factors are associated with an increased risk of intracranial hemorrhage in patients with immune thrombocytopenia.

Mayank Sharma, of the University of Miami
Jennifer Smith/MDedge News
Mayank Sharma

Data on more than 300,000 immune thrombocytopenia (ITP) hospitalizations indicated that older age, male sex, not having private insurance, having a gastrointestinal or “other” bleed, and receiving treatment at a hospital in the western United States, a medium- or large-sized hospital, or an urban teaching hospital were all associated with an increased risk of intracranial hemorrhage (ICH).

Mayank Sharma, of the University of Miami, detailed these findings at the annual meeting of the American Society of Hematology.

Mr. Sharma and colleagues analyzed data from the National Inpatient Sample database from 2007 to 2016. Of the 348,906 ITP hospitalizations included, there were 3,408 (0.98%) cases of ICH.

The overall incidence of ICH was low and remained stable over time, “which is reassuring,” Mr. Sharma said. However, the mortality rate was higher among patients with ICH than among those without it – 26.7% and 3.2%, respectively.

A multivariate analysis showed that female patients had a decreased likelihood of ICH, with an odds ratio of 0.81 (95% confidence interval, 0.68-0.97). Patients with private insurance had a decreased likelihood of ICH as well, with an OR of 0.81 (95% CI, 0.61-1.08).

Conversely, older patients had an increased likelihood of ICH. The OR was 2.23 (95% CI, 1.51-3.31) for patients aged 25-64 years, and the OR was 3.69 (95% CI, 2.34-5.84) for patients aged 65 years and older.

Patients with a gastrointestinal bleed or an other bleed (not including hematuria or epistaxis) had an increased likelihood of ICH. The ORs were 1.60 (95% CI, 1.18-2.16) and 1.69 (95% CI, 1.19-2.42), respectively.

Patients hospitalized in the western United States (OR, 1.62; 95% CI, 1.26-2.08), at a medium-sized hospital (OR, 1.64; 95% CI, 1.08-2.47), at a large hospital (OR, 2.42; 95% CI, 1.65-3.55), or at an urban teaching hospital (OR, 2.73; 95% CI, 1.80-4.13) all had an increased likelihood of ICH.

“Our second objective was to study the factors associated with mortality in ITP patients with ICH,” Mr. Sharma said. “We found female gender and Medicaid, private, or self-pay as primary payers to be associated with a lower mortality in ITP with ICH.

“[A]ge of 25-64 and 65 years and above, coexistence of a GI bleed or other bleed, and admission to a large or urban teaching hospital were associated with a higher mortality,” he added.

Mr. Sharma said the study’s strengths are that it is the most recent study on trends in ITP/ICH hospitalizations, and that it’s a longitudinal assessment of data from a nationally representative database.

The study’s limitations include its retrospective nature and the use of ICD codes, which could lead to inaccuracies. Data on prior therapies and long-term outcomes were not available, and the researchers were unable to differentiate between acute and chronic ITP.

Mr. Sharma said he had no relevant conflicts of interest.
 

SOURCE: Sharma M et al. ASH 2019, Abstract 55.

– A large, retrospective study suggests several factors are associated with an increased risk of intracranial hemorrhage in patients with immune thrombocytopenia.

Mayank Sharma, of the University of Miami
Jennifer Smith/MDedge News
Mayank Sharma

Data on more than 300,000 immune thrombocytopenia (ITP) hospitalizations indicated that older age, male sex, not having private insurance, having a gastrointestinal or “other” bleed, and receiving treatment at a hospital in the western United States, a medium- or large-sized hospital, or an urban teaching hospital were all associated with an increased risk of intracranial hemorrhage (ICH).

Mayank Sharma, of the University of Miami, detailed these findings at the annual meeting of the American Society of Hematology.

Mr. Sharma and colleagues analyzed data from the National Inpatient Sample database from 2007 to 2016. Of the 348,906 ITP hospitalizations included, there were 3,408 (0.98%) cases of ICH.

The overall incidence of ICH was low and remained stable over time, “which is reassuring,” Mr. Sharma said. However, the mortality rate was higher among patients with ICH than among those without it – 26.7% and 3.2%, respectively.

A multivariate analysis showed that female patients had a decreased likelihood of ICH, with an odds ratio of 0.81 (95% confidence interval, 0.68-0.97). Patients with private insurance had a decreased likelihood of ICH as well, with an OR of 0.81 (95% CI, 0.61-1.08).

Conversely, older patients had an increased likelihood of ICH. The OR was 2.23 (95% CI, 1.51-3.31) for patients aged 25-64 years, and the OR was 3.69 (95% CI, 2.34-5.84) for patients aged 65 years and older.

Patients with a gastrointestinal bleed or an other bleed (not including hematuria or epistaxis) had an increased likelihood of ICH. The ORs were 1.60 (95% CI, 1.18-2.16) and 1.69 (95% CI, 1.19-2.42), respectively.

Patients hospitalized in the western United States (OR, 1.62; 95% CI, 1.26-2.08), at a medium-sized hospital (OR, 1.64; 95% CI, 1.08-2.47), at a large hospital (OR, 2.42; 95% CI, 1.65-3.55), or at an urban teaching hospital (OR, 2.73; 95% CI, 1.80-4.13) all had an increased likelihood of ICH.

“Our second objective was to study the factors associated with mortality in ITP patients with ICH,” Mr. Sharma said. “We found female gender and Medicaid, private, or self-pay as primary payers to be associated with a lower mortality in ITP with ICH.

“[A]ge of 25-64 and 65 years and above, coexistence of a GI bleed or other bleed, and admission to a large or urban teaching hospital were associated with a higher mortality,” he added.

Mr. Sharma said the study’s strengths are that it is the most recent study on trends in ITP/ICH hospitalizations, and that it’s a longitudinal assessment of data from a nationally representative database.

The study’s limitations include its retrospective nature and the use of ICD codes, which could lead to inaccuracies. Data on prior therapies and long-term outcomes were not available, and the researchers were unable to differentiate between acute and chronic ITP.

Mr. Sharma said he had no relevant conflicts of interest.
 

SOURCE: Sharma M et al. ASH 2019, Abstract 55.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM ASH 2019

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.