LayerRx Mapping ID
428
Slot System
Featured Buckets
Featured Buckets Admin
Medscape Lead Concept
63214

Novel agent to be studied in relapsed/refractory AML

Article Type
Changed
Fri, 01/04/2019 - 10:12


The first clinical use of PCM-075, a PLK1 (polo-like kinase 1) inhibitor, will be in adults with relapsed/refractory acute myeloid leukemia in a phase 1b/2 trial, Trovagene announced.

The aim of the phase 1 portion of the trial is to find out whether PCM-075 given orally daily for 5 consecutive days every 28 days is safe and tolerable in such patients or in those AML patients who are ineligible for intensive induction therapy. The researchers are also trying to determine the maximum tolerated dose of PCM-075 or recommended phase 2 dose of PCM-075 in combination with decitabine and/or PCM-075 in combination with low-dose cytarabine.

The primary outcomes of the phase 1 portion of the trial are the number of participants with dose-limiting toxicity and adverse events from baseline out to 30 days after the last dose of PCM-075, up to 27 months. The primary outcome of phase 2 , called PCM-075 in Combination With Either Low-Dose Cytarabine or Decitabine in Adult Patients With Acute Myeloid Leukemia, will be the rate of complete response plus complete response with incomplete blood count recovery out to 27 months.

The PLK1 enzyme is overexpressed in multiple hematologic and solid tumor cancers, and studies have shown that inhibition of polo-like kinases can lead to tumor cell death, Trovagene said in its statement.

Bill Welch, CEO of Trovagene, added that “PCM-075 is the first highly PLK1-selective competitive inhibitor administered orally to enter clinical trials with potential activity in both hematologic and solid tumor cancers.”

Publications
Topics
Sections


The first clinical use of PCM-075, a PLK1 (polo-like kinase 1) inhibitor, will be in adults with relapsed/refractory acute myeloid leukemia in a phase 1b/2 trial, Trovagene announced.

The aim of the phase 1 portion of the trial is to find out whether PCM-075 given orally daily for 5 consecutive days every 28 days is safe and tolerable in such patients or in those AML patients who are ineligible for intensive induction therapy. The researchers are also trying to determine the maximum tolerated dose of PCM-075 or recommended phase 2 dose of PCM-075 in combination with decitabine and/or PCM-075 in combination with low-dose cytarabine.

The primary outcomes of the phase 1 portion of the trial are the number of participants with dose-limiting toxicity and adverse events from baseline out to 30 days after the last dose of PCM-075, up to 27 months. The primary outcome of phase 2 , called PCM-075 in Combination With Either Low-Dose Cytarabine or Decitabine in Adult Patients With Acute Myeloid Leukemia, will be the rate of complete response plus complete response with incomplete blood count recovery out to 27 months.

The PLK1 enzyme is overexpressed in multiple hematologic and solid tumor cancers, and studies have shown that inhibition of polo-like kinases can lead to tumor cell death, Trovagene said in its statement.

Bill Welch, CEO of Trovagene, added that “PCM-075 is the first highly PLK1-selective competitive inhibitor administered orally to enter clinical trials with potential activity in both hematologic and solid tumor cancers.”


The first clinical use of PCM-075, a PLK1 (polo-like kinase 1) inhibitor, will be in adults with relapsed/refractory acute myeloid leukemia in a phase 1b/2 trial, Trovagene announced.

The aim of the phase 1 portion of the trial is to find out whether PCM-075 given orally daily for 5 consecutive days every 28 days is safe and tolerable in such patients or in those AML patients who are ineligible for intensive induction therapy. The researchers are also trying to determine the maximum tolerated dose of PCM-075 or recommended phase 2 dose of PCM-075 in combination with decitabine and/or PCM-075 in combination with low-dose cytarabine.

The primary outcomes of the phase 1 portion of the trial are the number of participants with dose-limiting toxicity and adverse events from baseline out to 30 days after the last dose of PCM-075, up to 27 months. The primary outcome of phase 2 , called PCM-075 in Combination With Either Low-Dose Cytarabine or Decitabine in Adult Patients With Acute Myeloid Leukemia, will be the rate of complete response plus complete response with incomplete blood count recovery out to 27 months.

The PLK1 enzyme is overexpressed in multiple hematologic and solid tumor cancers, and studies have shown that inhibition of polo-like kinases can lead to tumor cell death, Trovagene said in its statement.

Bill Welch, CEO of Trovagene, added that “PCM-075 is the first highly PLK1-selective competitive inhibitor administered orally to enter clinical trials with potential activity in both hematologic and solid tumor cancers.”

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default

Start with fitness when deciding on treatment for elderly AML patients

Article Type
Changed
Fri, 01/04/2019 - 10:12

 

When evaluating older patients with acute myeloid leukemia for treatment, start with their fitness levels.

ML is a disease of older adults, and with increasing age comes higher treatment-related mortality, lower complete remission rates, higher relapse risk, and shorter overall survival. So it may not be surprising that fewer than half of U.S. patients with newly diagnosed acute myeloid leukemia over age 65 receive any chemotherapy at all, wrote Li-Wen Huang, MD, and Rebecca L. Olin, MD, of the University of California, San Francisco.

Dr. Li-Wen Huang
Prognostic models and comprehensive geriatric assessments, however, can help in evaluating whether intensive chemotherapy suitable is for each patient, the authors reported (J Geriatr Oncol. 2017;8[6]:417-20).

Fitness is key: Older patients considered fit for intensive chemotherapy should receive standard induction therapy, and reduced-intensity allogeneic stem cell transplantation should then be considered. Patients considered unfit for intensive therapy, on the other hand, should receive hypomethylating agents.

Several new therapeutic agents have shown promising results either by improving intensive chemotherapy (CPX-351), by improving upon lower-intensity therapy (venetoclax, antibody drug conjugates), or by targeting somatic mutations (FLT3 inhibitors and others), the investigators concluded.

Dr. Huang reported no conflicts. Dr. Olin has received research funding from Daiichi Sankyo, Astellas, and Genentech.

Publications
Topics
Sections

 

When evaluating older patients with acute myeloid leukemia for treatment, start with their fitness levels.

ML is a disease of older adults, and with increasing age comes higher treatment-related mortality, lower complete remission rates, higher relapse risk, and shorter overall survival. So it may not be surprising that fewer than half of U.S. patients with newly diagnosed acute myeloid leukemia over age 65 receive any chemotherapy at all, wrote Li-Wen Huang, MD, and Rebecca L. Olin, MD, of the University of California, San Francisco.

Dr. Li-Wen Huang
Prognostic models and comprehensive geriatric assessments, however, can help in evaluating whether intensive chemotherapy suitable is for each patient, the authors reported (J Geriatr Oncol. 2017;8[6]:417-20).

Fitness is key: Older patients considered fit for intensive chemotherapy should receive standard induction therapy, and reduced-intensity allogeneic stem cell transplantation should then be considered. Patients considered unfit for intensive therapy, on the other hand, should receive hypomethylating agents.

Several new therapeutic agents have shown promising results either by improving intensive chemotherapy (CPX-351), by improving upon lower-intensity therapy (venetoclax, antibody drug conjugates), or by targeting somatic mutations (FLT3 inhibitors and others), the investigators concluded.

Dr. Huang reported no conflicts. Dr. Olin has received research funding from Daiichi Sankyo, Astellas, and Genentech.

 

When evaluating older patients with acute myeloid leukemia for treatment, start with their fitness levels.

ML is a disease of older adults, and with increasing age comes higher treatment-related mortality, lower complete remission rates, higher relapse risk, and shorter overall survival. So it may not be surprising that fewer than half of U.S. patients with newly diagnosed acute myeloid leukemia over age 65 receive any chemotherapy at all, wrote Li-Wen Huang, MD, and Rebecca L. Olin, MD, of the University of California, San Francisco.

Dr. Li-Wen Huang
Prognostic models and comprehensive geriatric assessments, however, can help in evaluating whether intensive chemotherapy suitable is for each patient, the authors reported (J Geriatr Oncol. 2017;8[6]:417-20).

Fitness is key: Older patients considered fit for intensive chemotherapy should receive standard induction therapy, and reduced-intensity allogeneic stem cell transplantation should then be considered. Patients considered unfit for intensive therapy, on the other hand, should receive hypomethylating agents.

Several new therapeutic agents have shown promising results either by improving intensive chemotherapy (CPX-351), by improving upon lower-intensity therapy (venetoclax, antibody drug conjugates), or by targeting somatic mutations (FLT3 inhibitors and others), the investigators concluded.

Dr. Huang reported no conflicts. Dr. Olin has received research funding from Daiichi Sankyo, Astellas, and Genentech.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM THE JOURNAL OF GERIATRIC ONCOLOGY

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

Team discovers mechanism of resistance in AML

Article Type
Changed
Sat, 11/18/2017 - 00:01
Display Headline
Team discovers mechanism of resistance in AML

Children’s Research Hospital
John Schuetz, PhD Photo from St. Jude

Researchers say they have uncovered a target to overcome drug resistance in acute myeloid leukemia (AML).

The team discovered how a linkage between 2 proteins enables AML cells to resist chemotherapy and showed that disrupting the linkage could render the cells vulnerable to treatment.

The researchers believe their discovery could lead to drugs to enhance chemotherapy in patients with AML and other cancers.

John Schuetz, PhD, of St. Jude Children’s Research Hospital in Memphis, Tennessee, and his colleagues described this research in Nature Communications.

The team launched their experiments based on previous findings that a protein called ABCC4 was greatly elevated in aggressive cases of AML.

Dr Schuetz and his colleagues searched for other proteins that might interact with ABCC4 and enable its function. The team’s screening of candidate proteins yielded one, MPP1, which was also greatly increased in AML.

The researchers found the 2 proteins are connected, and the connection enables cells to assume the characteristics of highly proliferative leukemia cells.

These experiments involved genetically altering hematopoietic progenitor cells to have high MPP1 and ABCC4 levels. The cells were grown in culture and then replated to see if they would continue to grow, as such self-renewal is a hallmark of leukemia cells.

The researchers found that serial regrowth depended on the cells having high levels of both ABCC4 and MPP1.

“Typically, if you take normal progenitors and you replate, you could do that one time, maybe twice,” Dr Schuetz said. “But our big surprise was that overexpressing MPP1—analogous to what you would see in leukemia—allows those progenitors to self-renew, to be replated over and over, to form new colonies.”

The experiments also revealed that MPP1 and ABCC4 functioned at the cell membrane, where they could play a role in the machinery that would rid the leukemia cells of chemotherapy drugs.

“When we disrupted their interaction, ABCC4 moved off the membrane and the cells became more sensitive to drugs used in AML—drugs that are pumped out of the cell by ABCC4,” Dr Schuetz said.

By screening thousands of compounds, the researchers identified some that could disrupt the ABCC4-MPP1 connection. One, called Antimycin-A, reversed drug resistance in AML cell lines and in cells from AML patients.

Antimycin-A is too toxic to be used in chemotherapy, but the researchers believe identification of the compound should aid the search for other, less-toxic drugs to disrupt the ABCC4-MPP1 interaction.

The team’s findings could also enable clinicians to identify AML patients with high levels of ABCC4 and MPP1. In such patients, drugs that disrupt ABCC4-MPP1 might enhance the effectiveness of standard chemotherapy, Dr Schuetz said.

He also noted that other cancers, including breast and colon cancer and medulloblastoma, show high levels of both ABCC4 and MPP1. Chemotherapy for those cancers might also be enhanced by drugs that disrupt ABCC4-MPP1.

Publications
Topics

Children’s Research Hospital
John Schuetz, PhD Photo from St. Jude

Researchers say they have uncovered a target to overcome drug resistance in acute myeloid leukemia (AML).

The team discovered how a linkage between 2 proteins enables AML cells to resist chemotherapy and showed that disrupting the linkage could render the cells vulnerable to treatment.

The researchers believe their discovery could lead to drugs to enhance chemotherapy in patients with AML and other cancers.

John Schuetz, PhD, of St. Jude Children’s Research Hospital in Memphis, Tennessee, and his colleagues described this research in Nature Communications.

The team launched their experiments based on previous findings that a protein called ABCC4 was greatly elevated in aggressive cases of AML.

Dr Schuetz and his colleagues searched for other proteins that might interact with ABCC4 and enable its function. The team’s screening of candidate proteins yielded one, MPP1, which was also greatly increased in AML.

The researchers found the 2 proteins are connected, and the connection enables cells to assume the characteristics of highly proliferative leukemia cells.

These experiments involved genetically altering hematopoietic progenitor cells to have high MPP1 and ABCC4 levels. The cells were grown in culture and then replated to see if they would continue to grow, as such self-renewal is a hallmark of leukemia cells.

The researchers found that serial regrowth depended on the cells having high levels of both ABCC4 and MPP1.

“Typically, if you take normal progenitors and you replate, you could do that one time, maybe twice,” Dr Schuetz said. “But our big surprise was that overexpressing MPP1—analogous to what you would see in leukemia—allows those progenitors to self-renew, to be replated over and over, to form new colonies.”

The experiments also revealed that MPP1 and ABCC4 functioned at the cell membrane, where they could play a role in the machinery that would rid the leukemia cells of chemotherapy drugs.

“When we disrupted their interaction, ABCC4 moved off the membrane and the cells became more sensitive to drugs used in AML—drugs that are pumped out of the cell by ABCC4,” Dr Schuetz said.

By screening thousands of compounds, the researchers identified some that could disrupt the ABCC4-MPP1 connection. One, called Antimycin-A, reversed drug resistance in AML cell lines and in cells from AML patients.

Antimycin-A is too toxic to be used in chemotherapy, but the researchers believe identification of the compound should aid the search for other, less-toxic drugs to disrupt the ABCC4-MPP1 interaction.

The team’s findings could also enable clinicians to identify AML patients with high levels of ABCC4 and MPP1. In such patients, drugs that disrupt ABCC4-MPP1 might enhance the effectiveness of standard chemotherapy, Dr Schuetz said.

He also noted that other cancers, including breast and colon cancer and medulloblastoma, show high levels of both ABCC4 and MPP1. Chemotherapy for those cancers might also be enhanced by drugs that disrupt ABCC4-MPP1.

Children’s Research Hospital
John Schuetz, PhD Photo from St. Jude

Researchers say they have uncovered a target to overcome drug resistance in acute myeloid leukemia (AML).

The team discovered how a linkage between 2 proteins enables AML cells to resist chemotherapy and showed that disrupting the linkage could render the cells vulnerable to treatment.

The researchers believe their discovery could lead to drugs to enhance chemotherapy in patients with AML and other cancers.

John Schuetz, PhD, of St. Jude Children’s Research Hospital in Memphis, Tennessee, and his colleagues described this research in Nature Communications.

The team launched their experiments based on previous findings that a protein called ABCC4 was greatly elevated in aggressive cases of AML.

Dr Schuetz and his colleagues searched for other proteins that might interact with ABCC4 and enable its function. The team’s screening of candidate proteins yielded one, MPP1, which was also greatly increased in AML.

The researchers found the 2 proteins are connected, and the connection enables cells to assume the characteristics of highly proliferative leukemia cells.

These experiments involved genetically altering hematopoietic progenitor cells to have high MPP1 and ABCC4 levels. The cells were grown in culture and then replated to see if they would continue to grow, as such self-renewal is a hallmark of leukemia cells.

The researchers found that serial regrowth depended on the cells having high levels of both ABCC4 and MPP1.

“Typically, if you take normal progenitors and you replate, you could do that one time, maybe twice,” Dr Schuetz said. “But our big surprise was that overexpressing MPP1—analogous to what you would see in leukemia—allows those progenitors to self-renew, to be replated over and over, to form new colonies.”

The experiments also revealed that MPP1 and ABCC4 functioned at the cell membrane, where they could play a role in the machinery that would rid the leukemia cells of chemotherapy drugs.

“When we disrupted their interaction, ABCC4 moved off the membrane and the cells became more sensitive to drugs used in AML—drugs that are pumped out of the cell by ABCC4,” Dr Schuetz said.

By screening thousands of compounds, the researchers identified some that could disrupt the ABCC4-MPP1 connection. One, called Antimycin-A, reversed drug resistance in AML cell lines and in cells from AML patients.

Antimycin-A is too toxic to be used in chemotherapy, but the researchers believe identification of the compound should aid the search for other, less-toxic drugs to disrupt the ABCC4-MPP1 interaction.

The team’s findings could also enable clinicians to identify AML patients with high levels of ABCC4 and MPP1. In such patients, drugs that disrupt ABCC4-MPP1 might enhance the effectiveness of standard chemotherapy, Dr Schuetz said.

He also noted that other cancers, including breast and colon cancer and medulloblastoma, show high levels of both ABCC4 and MPP1. Chemotherapy for those cancers might also be enhanced by drugs that disrupt ABCC4-MPP1.

Publications
Publications
Topics
Article Type
Display Headline
Team discovers mechanism of resistance in AML
Display Headline
Team discovers mechanism of resistance in AML
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Rigosertib produces better OS in MDS than tAML

Article Type
Changed
Thu, 11/16/2017 - 00:04
Display Headline
Rigosertib produces better OS in MDS than tAML

myelodysplastic syndrome
Micrograph showing

Rigosertib has demonstrated activity and tolerability in patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia transformed from MDS (tAML), according to researchers.

In a phase 1/2 study, rigosertib produced responses in a quarter of MDS/tAML patients and enabled stable disease in another quarter.

Overall survival (OS) was about a year longer for responders than for non-responders.

MDS patients were more likely to respond to rigosertib and therefore enjoyed longer OS than tAML patients.

Overall, rigosertib was considered well-tolerated. There were no treatment-related deaths, though 18% of patients experienced treatment-related serious adverse events (AEs).

Lewis Silverman, MD, of Icahn School of Medicine at Mount Sinai in New York, New York, and his colleagues described these results in Leukemia Research.

The study was sponsored by Onconova Therapeutics, Inc., the company developing rigosertib.

Rigosertib is an inhibitor of Ras-effector pathways that interacts with the Ras binding domains common to several signaling proteins, including Raf and PI3 kinase.

Dr Silverman and his colleagues tested intravenous rigosertib in a dose-escalation, phase 1/2 study of 22 patients. Patients had tAML (n=13), high-risk MDS (n=6), intermediate-2-risk MDS (n=2), or chronic myelomonocytic leukemia (n=1).

All patients had relapsed or were refractory to standard therapy and had no approved options for second-line therapies. The patients’ median age was 78 (range, 59-84), and 90% were male.

Patients received 3- to 7-day continuous infusions of rigosertib at doses ranging from 650 mg/m2/day to 1700 mg/m2/day in 14-day cycles.

The mean number of treatment cycles was 5.6 ± 5.8 (range, 1-23). The maximum tolerated dose of rigosertib was 1700 mg/m2/day, and the recommended phase 2 dose was 1375 mg/m2/day.

Safety

All patients had at least 1 AE. The most common AEs of any grade were fatigue (n=16, 73%), diarrhea (n=12, 55%), pyrexia (n=12, 55%), dyspnea (n=11, 50%), insomnia (n=11, 50%), anemia (n=10, 46%), constipation (n=9, 41%), nausea (n=9, 41%), cough (n=9, 41%), and decreased appetite (n=9, 41%).

The most common grade 3 or higher AEs were anemia (n=9, 41%), thrombocytopenia (n=5, 23%), pneumonia (n=5, 23%), hypoglycemia (n=4, 18%), hyponatremia (n=4, 18%), and hypophosphatemia (n=4, 18%).

Four patients (18%) had treatment-related serious AEs. This included hematuria and pollakiuria (n=1), dysuria and pollakiuria (n=1), asthenia (n=1), and dyspnea (n=1). Thirteen patients (59%) stopped treatment due to AEs.

Ten patients, who remained on study from 1 to 19 months, died within 30 days of stopping rigosertib. There were no treatment-related deaths.

Efficacy

Nineteen patients were evaluable for efficacy.

Five patients responded to treatment. Four patients with MDS had a marrow complete response, and 1 with tAML had a marrow partial response. Two of the patients with marrow complete response also had hematologic improvements.

Five patients had stable disease, 3 with MDS and 2 with tAML.

The median OS was 15.7 months for responders and 2.0 months for non-responders (P=0.0070). The median OS was 12.0 months for MDS patients and 2.0 months for tAML patients (P<0.0001).

“The publication of results from this historical study provides support of the relationship between bone marrow blast response and improvement in overall survival in this group of patients with MDS and acute myeloid leukemia for whom no FDA-approved treatments are currently available,” said Ramesh Kumar, president and chief executive officer of Onconova Therapeutics, Inc.

He added that these data are “fundamental to the rationale” of ongoing studies of rigosertib in high-risk MDS patients.

Publications
Topics

myelodysplastic syndrome
Micrograph showing

Rigosertib has demonstrated activity and tolerability in patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia transformed from MDS (tAML), according to researchers.

In a phase 1/2 study, rigosertib produced responses in a quarter of MDS/tAML patients and enabled stable disease in another quarter.

Overall survival (OS) was about a year longer for responders than for non-responders.

MDS patients were more likely to respond to rigosertib and therefore enjoyed longer OS than tAML patients.

Overall, rigosertib was considered well-tolerated. There were no treatment-related deaths, though 18% of patients experienced treatment-related serious adverse events (AEs).

Lewis Silverman, MD, of Icahn School of Medicine at Mount Sinai in New York, New York, and his colleagues described these results in Leukemia Research.

The study was sponsored by Onconova Therapeutics, Inc., the company developing rigosertib.

Rigosertib is an inhibitor of Ras-effector pathways that interacts with the Ras binding domains common to several signaling proteins, including Raf and PI3 kinase.

Dr Silverman and his colleagues tested intravenous rigosertib in a dose-escalation, phase 1/2 study of 22 patients. Patients had tAML (n=13), high-risk MDS (n=6), intermediate-2-risk MDS (n=2), or chronic myelomonocytic leukemia (n=1).

All patients had relapsed or were refractory to standard therapy and had no approved options for second-line therapies. The patients’ median age was 78 (range, 59-84), and 90% were male.

Patients received 3- to 7-day continuous infusions of rigosertib at doses ranging from 650 mg/m2/day to 1700 mg/m2/day in 14-day cycles.

The mean number of treatment cycles was 5.6 ± 5.8 (range, 1-23). The maximum tolerated dose of rigosertib was 1700 mg/m2/day, and the recommended phase 2 dose was 1375 mg/m2/day.

Safety

All patients had at least 1 AE. The most common AEs of any grade were fatigue (n=16, 73%), diarrhea (n=12, 55%), pyrexia (n=12, 55%), dyspnea (n=11, 50%), insomnia (n=11, 50%), anemia (n=10, 46%), constipation (n=9, 41%), nausea (n=9, 41%), cough (n=9, 41%), and decreased appetite (n=9, 41%).

The most common grade 3 or higher AEs were anemia (n=9, 41%), thrombocytopenia (n=5, 23%), pneumonia (n=5, 23%), hypoglycemia (n=4, 18%), hyponatremia (n=4, 18%), and hypophosphatemia (n=4, 18%).

Four patients (18%) had treatment-related serious AEs. This included hematuria and pollakiuria (n=1), dysuria and pollakiuria (n=1), asthenia (n=1), and dyspnea (n=1). Thirteen patients (59%) stopped treatment due to AEs.

Ten patients, who remained on study from 1 to 19 months, died within 30 days of stopping rigosertib. There were no treatment-related deaths.

Efficacy

Nineteen patients were evaluable for efficacy.

Five patients responded to treatment. Four patients with MDS had a marrow complete response, and 1 with tAML had a marrow partial response. Two of the patients with marrow complete response also had hematologic improvements.

Five patients had stable disease, 3 with MDS and 2 with tAML.

The median OS was 15.7 months for responders and 2.0 months for non-responders (P=0.0070). The median OS was 12.0 months for MDS patients and 2.0 months for tAML patients (P<0.0001).

“The publication of results from this historical study provides support of the relationship between bone marrow blast response and improvement in overall survival in this group of patients with MDS and acute myeloid leukemia for whom no FDA-approved treatments are currently available,” said Ramesh Kumar, president and chief executive officer of Onconova Therapeutics, Inc.

He added that these data are “fundamental to the rationale” of ongoing studies of rigosertib in high-risk MDS patients.

myelodysplastic syndrome
Micrograph showing

Rigosertib has demonstrated activity and tolerability in patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia transformed from MDS (tAML), according to researchers.

In a phase 1/2 study, rigosertib produced responses in a quarter of MDS/tAML patients and enabled stable disease in another quarter.

Overall survival (OS) was about a year longer for responders than for non-responders.

MDS patients were more likely to respond to rigosertib and therefore enjoyed longer OS than tAML patients.

Overall, rigosertib was considered well-tolerated. There were no treatment-related deaths, though 18% of patients experienced treatment-related serious adverse events (AEs).

Lewis Silverman, MD, of Icahn School of Medicine at Mount Sinai in New York, New York, and his colleagues described these results in Leukemia Research.

The study was sponsored by Onconova Therapeutics, Inc., the company developing rigosertib.

Rigosertib is an inhibitor of Ras-effector pathways that interacts with the Ras binding domains common to several signaling proteins, including Raf and PI3 kinase.

Dr Silverman and his colleagues tested intravenous rigosertib in a dose-escalation, phase 1/2 study of 22 patients. Patients had tAML (n=13), high-risk MDS (n=6), intermediate-2-risk MDS (n=2), or chronic myelomonocytic leukemia (n=1).

All patients had relapsed or were refractory to standard therapy and had no approved options for second-line therapies. The patients’ median age was 78 (range, 59-84), and 90% were male.

Patients received 3- to 7-day continuous infusions of rigosertib at doses ranging from 650 mg/m2/day to 1700 mg/m2/day in 14-day cycles.

The mean number of treatment cycles was 5.6 ± 5.8 (range, 1-23). The maximum tolerated dose of rigosertib was 1700 mg/m2/day, and the recommended phase 2 dose was 1375 mg/m2/day.

Safety

All patients had at least 1 AE. The most common AEs of any grade were fatigue (n=16, 73%), diarrhea (n=12, 55%), pyrexia (n=12, 55%), dyspnea (n=11, 50%), insomnia (n=11, 50%), anemia (n=10, 46%), constipation (n=9, 41%), nausea (n=9, 41%), cough (n=9, 41%), and decreased appetite (n=9, 41%).

The most common grade 3 or higher AEs were anemia (n=9, 41%), thrombocytopenia (n=5, 23%), pneumonia (n=5, 23%), hypoglycemia (n=4, 18%), hyponatremia (n=4, 18%), and hypophosphatemia (n=4, 18%).

Four patients (18%) had treatment-related serious AEs. This included hematuria and pollakiuria (n=1), dysuria and pollakiuria (n=1), asthenia (n=1), and dyspnea (n=1). Thirteen patients (59%) stopped treatment due to AEs.

Ten patients, who remained on study from 1 to 19 months, died within 30 days of stopping rigosertib. There were no treatment-related deaths.

Efficacy

Nineteen patients were evaluable for efficacy.

Five patients responded to treatment. Four patients with MDS had a marrow complete response, and 1 with tAML had a marrow partial response. Two of the patients with marrow complete response also had hematologic improvements.

Five patients had stable disease, 3 with MDS and 2 with tAML.

The median OS was 15.7 months for responders and 2.0 months for non-responders (P=0.0070). The median OS was 12.0 months for MDS patients and 2.0 months for tAML patients (P<0.0001).

“The publication of results from this historical study provides support of the relationship between bone marrow blast response and improvement in overall survival in this group of patients with MDS and acute myeloid leukemia for whom no FDA-approved treatments are currently available,” said Ramesh Kumar, president and chief executive officer of Onconova Therapeutics, Inc.

He added that these data are “fundamental to the rationale” of ongoing studies of rigosertib in high-risk MDS patients.

Publications
Publications
Topics
Article Type
Display Headline
Rigosertib produces better OS in MDS than tAML
Display Headline
Rigosertib produces better OS in MDS than tAML
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Generic azacitidine approved in Canada

Article Type
Changed
Thu, 11/16/2017 - 00:01
Display Headline
Generic azacitidine approved in Canada

acute myeloid leukemia
Micrograph showing

Health Canada has approved Dr. Reddy’s Laboratories Ltd.’s Azacitidine for Injection 100 mg/vial, a bioequivalent generic version of VIDAZA® (azacitidine for injection).

The generic drug is approved for the same indications as VIDAZA®.

This includes treating adults with intermediate-2 or high-risk myelodysplastic syndromes (according to the International Prognostic Scoring System) who are not eligible for hematopoietic stem cell transplant.

It also includes treating adults who have acute myeloid leukemia with 20% to 30% blasts and multi-lineage dysplasia (according to World Health Organization classification) who are not eligible for hematopoietic stem cell transplant.

“The approval and launch of Azacitidine for Injection is an important milestone for Dr. Reddy’s in Canada,” said Vinod Ramachandran, PhD, country manager, Dr. Reddy’s Canada.

“The launch of the first generic azacitidine for injection is another step in our long-term commitment to bring more cost-effective options to Canadian patients.”

Publications
Topics

acute myeloid leukemia
Micrograph showing

Health Canada has approved Dr. Reddy’s Laboratories Ltd.’s Azacitidine for Injection 100 mg/vial, a bioequivalent generic version of VIDAZA® (azacitidine for injection).

The generic drug is approved for the same indications as VIDAZA®.

This includes treating adults with intermediate-2 or high-risk myelodysplastic syndromes (according to the International Prognostic Scoring System) who are not eligible for hematopoietic stem cell transplant.

It also includes treating adults who have acute myeloid leukemia with 20% to 30% blasts and multi-lineage dysplasia (according to World Health Organization classification) who are not eligible for hematopoietic stem cell transplant.

“The approval and launch of Azacitidine for Injection is an important milestone for Dr. Reddy’s in Canada,” said Vinod Ramachandran, PhD, country manager, Dr. Reddy’s Canada.

“The launch of the first generic azacitidine for injection is another step in our long-term commitment to bring more cost-effective options to Canadian patients.”

acute myeloid leukemia
Micrograph showing

Health Canada has approved Dr. Reddy’s Laboratories Ltd.’s Azacitidine for Injection 100 mg/vial, a bioequivalent generic version of VIDAZA® (azacitidine for injection).

The generic drug is approved for the same indications as VIDAZA®.

This includes treating adults with intermediate-2 or high-risk myelodysplastic syndromes (according to the International Prognostic Scoring System) who are not eligible for hematopoietic stem cell transplant.

It also includes treating adults who have acute myeloid leukemia with 20% to 30% blasts and multi-lineage dysplasia (according to World Health Organization classification) who are not eligible for hematopoietic stem cell transplant.

“The approval and launch of Azacitidine for Injection is an important milestone for Dr. Reddy’s in Canada,” said Vinod Ramachandran, PhD, country manager, Dr. Reddy’s Canada.

“The launch of the first generic azacitidine for injection is another step in our long-term commitment to bring more cost-effective options to Canadian patients.”

Publications
Publications
Topics
Article Type
Display Headline
Generic azacitidine approved in Canada
Display Headline
Generic azacitidine approved in Canada
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

AML candidate drug back in the pipeline

Article Type
Changed
Fri, 01/04/2019 - 10:12

 

The Food and Drug Administration has given the biopharmaceutical company Cellectis permission to resume phase 1 trials of UCART123, a gene-edited T-cell investigational drug that targets CD123, as a potential treatment for acute myeloid leukemia (AML) and blastic plasmacytoid dendritic cell neoplasm (BPDCN), according to a press release from the company.

UCART123 is the first allogeneic, “off-the-shelf” gene-edited chimeric antigen receptor (CAR) T-cell product candidate that the FDA has approved for clinical trials. The agency had placed a clinical hold on phase 1 trials of the gene-edited CAR T-cell drug on Sept. 4, following a patient death in the BPDCN clinical study. In order to proceed with the trials, Cellectis agreed to several changes in the study protocols.

The changes include decreasing the dose of the UCART123 therapy to 6.25x104 cells/kg and lowering the dose of the lympho-depleting regimen of cyclophosphamide to 750 mg/m2 per day over 3 days with a maximum daily dose of 1.33 g. Additionally, there can be no uncontrolled infection after receipt of the lympho-depleting preconditioning regimen. Patients must be afebrile at the start of treatment, off all but a replacement dose of corticosteroids, and have no organ dysfunction. Plus, the next three patients treated in each study must be under age 65.

There’s also a condition that patient enrollments be staggered by at least 28 days.

The drug sponsor is working with investigators and each clinical site to obtain the Institutional Review Board’s approval of the revised protocols.

The hold followed the death of a 78-year-old man with relapsed/refractory BPDCN with 30% blasts in his bone marrow and cutaneous lesions. The first dose of UCART123 at 6.25x105 cells/kg was administered without complication, but at day 5 the patient began experiencing side effects, including cytokine release syndrome and a lung infection. At day 8, the cytokine release syndrome had worsened and the patient had also developed capillary leak syndrome. He died on day 9 of the study.

In the AML phase 1 study, a 58-year-old woman with AML and 84% blasts in her bone marrow received the same dose of UCART123. She also developed cytokine release syndrome and capillary leak syndrome but both resolved with treatment.

Both patients also received the same preconditioning treatment: 30 mg/m2 per day fludarabine for 4 days and 1g/m2 per day cyclophosphamide for 3 days.

Publications
Topics
Sections

 

The Food and Drug Administration has given the biopharmaceutical company Cellectis permission to resume phase 1 trials of UCART123, a gene-edited T-cell investigational drug that targets CD123, as a potential treatment for acute myeloid leukemia (AML) and blastic plasmacytoid dendritic cell neoplasm (BPDCN), according to a press release from the company.

UCART123 is the first allogeneic, “off-the-shelf” gene-edited chimeric antigen receptor (CAR) T-cell product candidate that the FDA has approved for clinical trials. The agency had placed a clinical hold on phase 1 trials of the gene-edited CAR T-cell drug on Sept. 4, following a patient death in the BPDCN clinical study. In order to proceed with the trials, Cellectis agreed to several changes in the study protocols.

The changes include decreasing the dose of the UCART123 therapy to 6.25x104 cells/kg and lowering the dose of the lympho-depleting regimen of cyclophosphamide to 750 mg/m2 per day over 3 days with a maximum daily dose of 1.33 g. Additionally, there can be no uncontrolled infection after receipt of the lympho-depleting preconditioning regimen. Patients must be afebrile at the start of treatment, off all but a replacement dose of corticosteroids, and have no organ dysfunction. Plus, the next three patients treated in each study must be under age 65.

There’s also a condition that patient enrollments be staggered by at least 28 days.

The drug sponsor is working with investigators and each clinical site to obtain the Institutional Review Board’s approval of the revised protocols.

The hold followed the death of a 78-year-old man with relapsed/refractory BPDCN with 30% blasts in his bone marrow and cutaneous lesions. The first dose of UCART123 at 6.25x105 cells/kg was administered without complication, but at day 5 the patient began experiencing side effects, including cytokine release syndrome and a lung infection. At day 8, the cytokine release syndrome had worsened and the patient had also developed capillary leak syndrome. He died on day 9 of the study.

In the AML phase 1 study, a 58-year-old woman with AML and 84% blasts in her bone marrow received the same dose of UCART123. She also developed cytokine release syndrome and capillary leak syndrome but both resolved with treatment.

Both patients also received the same preconditioning treatment: 30 mg/m2 per day fludarabine for 4 days and 1g/m2 per day cyclophosphamide for 3 days.

 

The Food and Drug Administration has given the biopharmaceutical company Cellectis permission to resume phase 1 trials of UCART123, a gene-edited T-cell investigational drug that targets CD123, as a potential treatment for acute myeloid leukemia (AML) and blastic plasmacytoid dendritic cell neoplasm (BPDCN), according to a press release from the company.

UCART123 is the first allogeneic, “off-the-shelf” gene-edited chimeric antigen receptor (CAR) T-cell product candidate that the FDA has approved for clinical trials. The agency had placed a clinical hold on phase 1 trials of the gene-edited CAR T-cell drug on Sept. 4, following a patient death in the BPDCN clinical study. In order to proceed with the trials, Cellectis agreed to several changes in the study protocols.

The changes include decreasing the dose of the UCART123 therapy to 6.25x104 cells/kg and lowering the dose of the lympho-depleting regimen of cyclophosphamide to 750 mg/m2 per day over 3 days with a maximum daily dose of 1.33 g. Additionally, there can be no uncontrolled infection after receipt of the lympho-depleting preconditioning regimen. Patients must be afebrile at the start of treatment, off all but a replacement dose of corticosteroids, and have no organ dysfunction. Plus, the next three patients treated in each study must be under age 65.

There’s also a condition that patient enrollments be staggered by at least 28 days.

The drug sponsor is working with investigators and each clinical site to obtain the Institutional Review Board’s approval of the revised protocols.

The hold followed the death of a 78-year-old man with relapsed/refractory BPDCN with 30% blasts in his bone marrow and cutaneous lesions. The first dose of UCART123 at 6.25x105 cells/kg was administered without complication, but at day 5 the patient began experiencing side effects, including cytokine release syndrome and a lung infection. At day 8, the cytokine release syndrome had worsened and the patient had also developed capillary leak syndrome. He died on day 9 of the study.

In the AML phase 1 study, a 58-year-old woman with AML and 84% blasts in her bone marrow received the same dose of UCART123. She also developed cytokine release syndrome and capillary leak syndrome but both resolved with treatment.

Both patients also received the same preconditioning treatment: 30 mg/m2 per day fludarabine for 4 days and 1g/m2 per day cyclophosphamide for 3 days.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default

EMA grants accelerated assessment to drug for AML

Article Type
Changed
Thu, 11/09/2017 - 00:02
Display Headline
EMA grants accelerated assessment to drug for AML

Image by Lance Liotta
AML cells

The European Medicines Agency’s Committee for Medicinal Products for Human Use has granted accelerated assessment to a marketing authorization application (MAA) for CPX-351 (Vyxeos™), a fixed-ratio combination of cytarabine and daunorubicin inside a lipid vesicle.

The MAA is for CPX-351 to treat adults with high-risk acute myeloid leukemia (AML), defined as therapy-related AML or AML with myelodysplasia-related changes.

Accelerated assessment is designed to reduce the review timeline for products of major interest for public health and therapeutic innovation.

“If approved, Vyxeos will become the first new chemotherapy treatment option specifically for European patients with therapy-related AML or AML with myelodysplasia-related changes,” said Karen Smith, MD, PhD, executive vice president, research and development and chief medical officer at Jazz Pharmaceuticals, the company developing and marketing CPX-351.

The MAA for CPX-351 is supported by clinical data from 5 studies, including a phase 3 study. Results from this study were presented at the 2016 ASCO Annual Meeting.

In this study, researchers compared CPX-351 to cytarabine and daunorubicin (7+3) in 309 patients, ages 60 to 75, with newly diagnosed, therapy-related AML or AML with myelodysplasia-related changes.

The complete response rate was 38% in the CPX-351 arm and 26% in the 7+3 arm (P=0.036).

The rate of hematopoietic stem cell transplant was 34% in the CPX-351 arm and 25% in the 7+3 arm.

The median overall survival was 9.6 months in the CPX-351 arm and 5.9 months in the 7+3 arm (P=0.005).

All-cause 30-day mortality was 6% in the CPX-351 arm and 11% in the 7+3 arm. Sixty-day mortality was 14% and 21%, respectively.

Six percent of patients in both arms had a fatal adverse event (AE) on treatment or within 30 days of therapy that was not in the setting of progressive disease.

The rate of AEs that led to discontinuation was 18% in the CPX-351 arm and 13% in the 7+3 arm. AEs leading to discontinuation in the CPX-351 arm included prolonged cytopenias, infection, cardiotoxicity, respiratory failure, hemorrhage, renal insufficiency, colitis, and generalized medical deterioration.

The most common AEs (incidence ≥ 25%) in the CPX-351 arm were hemorrhagic events, febrile neutropenia, rash, edema, nausea, mucositis, diarrhea, constipation, musculoskeletal pain, fatigue, abdominal pain, dyspnea, headache, cough, decreased appetite, arrhythmia, pneumonia, bacteremia, chills, sleep disorders, and vomiting.

The most common serious AEs (incidence ≥ 5%) in the CPX-351 arm were dyspnea, myocardial toxicity, sepsis, pneumonia, febrile neutropenia, bacteremia, and hemorrhage.

Publications
Topics

Image by Lance Liotta
AML cells

The European Medicines Agency’s Committee for Medicinal Products for Human Use has granted accelerated assessment to a marketing authorization application (MAA) for CPX-351 (Vyxeos™), a fixed-ratio combination of cytarabine and daunorubicin inside a lipid vesicle.

The MAA is for CPX-351 to treat adults with high-risk acute myeloid leukemia (AML), defined as therapy-related AML or AML with myelodysplasia-related changes.

Accelerated assessment is designed to reduce the review timeline for products of major interest for public health and therapeutic innovation.

“If approved, Vyxeos will become the first new chemotherapy treatment option specifically for European patients with therapy-related AML or AML with myelodysplasia-related changes,” said Karen Smith, MD, PhD, executive vice president, research and development and chief medical officer at Jazz Pharmaceuticals, the company developing and marketing CPX-351.

The MAA for CPX-351 is supported by clinical data from 5 studies, including a phase 3 study. Results from this study were presented at the 2016 ASCO Annual Meeting.

In this study, researchers compared CPX-351 to cytarabine and daunorubicin (7+3) in 309 patients, ages 60 to 75, with newly diagnosed, therapy-related AML or AML with myelodysplasia-related changes.

The complete response rate was 38% in the CPX-351 arm and 26% in the 7+3 arm (P=0.036).

The rate of hematopoietic stem cell transplant was 34% in the CPX-351 arm and 25% in the 7+3 arm.

The median overall survival was 9.6 months in the CPX-351 arm and 5.9 months in the 7+3 arm (P=0.005).

All-cause 30-day mortality was 6% in the CPX-351 arm and 11% in the 7+3 arm. Sixty-day mortality was 14% and 21%, respectively.

Six percent of patients in both arms had a fatal adverse event (AE) on treatment or within 30 days of therapy that was not in the setting of progressive disease.

The rate of AEs that led to discontinuation was 18% in the CPX-351 arm and 13% in the 7+3 arm. AEs leading to discontinuation in the CPX-351 arm included prolonged cytopenias, infection, cardiotoxicity, respiratory failure, hemorrhage, renal insufficiency, colitis, and generalized medical deterioration.

The most common AEs (incidence ≥ 25%) in the CPX-351 arm were hemorrhagic events, febrile neutropenia, rash, edema, nausea, mucositis, diarrhea, constipation, musculoskeletal pain, fatigue, abdominal pain, dyspnea, headache, cough, decreased appetite, arrhythmia, pneumonia, bacteremia, chills, sleep disorders, and vomiting.

The most common serious AEs (incidence ≥ 5%) in the CPX-351 arm were dyspnea, myocardial toxicity, sepsis, pneumonia, febrile neutropenia, bacteremia, and hemorrhage.

Image by Lance Liotta
AML cells

The European Medicines Agency’s Committee for Medicinal Products for Human Use has granted accelerated assessment to a marketing authorization application (MAA) for CPX-351 (Vyxeos™), a fixed-ratio combination of cytarabine and daunorubicin inside a lipid vesicle.

The MAA is for CPX-351 to treat adults with high-risk acute myeloid leukemia (AML), defined as therapy-related AML or AML with myelodysplasia-related changes.

Accelerated assessment is designed to reduce the review timeline for products of major interest for public health and therapeutic innovation.

“If approved, Vyxeos will become the first new chemotherapy treatment option specifically for European patients with therapy-related AML or AML with myelodysplasia-related changes,” said Karen Smith, MD, PhD, executive vice president, research and development and chief medical officer at Jazz Pharmaceuticals, the company developing and marketing CPX-351.

The MAA for CPX-351 is supported by clinical data from 5 studies, including a phase 3 study. Results from this study were presented at the 2016 ASCO Annual Meeting.

In this study, researchers compared CPX-351 to cytarabine and daunorubicin (7+3) in 309 patients, ages 60 to 75, with newly diagnosed, therapy-related AML or AML with myelodysplasia-related changes.

The complete response rate was 38% in the CPX-351 arm and 26% in the 7+3 arm (P=0.036).

The rate of hematopoietic stem cell transplant was 34% in the CPX-351 arm and 25% in the 7+3 arm.

The median overall survival was 9.6 months in the CPX-351 arm and 5.9 months in the 7+3 arm (P=0.005).

All-cause 30-day mortality was 6% in the CPX-351 arm and 11% in the 7+3 arm. Sixty-day mortality was 14% and 21%, respectively.

Six percent of patients in both arms had a fatal adverse event (AE) on treatment or within 30 days of therapy that was not in the setting of progressive disease.

The rate of AEs that led to discontinuation was 18% in the CPX-351 arm and 13% in the 7+3 arm. AEs leading to discontinuation in the CPX-351 arm included prolonged cytopenias, infection, cardiotoxicity, respiratory failure, hemorrhage, renal insufficiency, colitis, and generalized medical deterioration.

The most common AEs (incidence ≥ 25%) in the CPX-351 arm were hemorrhagic events, febrile neutropenia, rash, edema, nausea, mucositis, diarrhea, constipation, musculoskeletal pain, fatigue, abdominal pain, dyspnea, headache, cough, decreased appetite, arrhythmia, pneumonia, bacteremia, chills, sleep disorders, and vomiting.

The most common serious AEs (incidence ≥ 5%) in the CPX-351 arm were dyspnea, myocardial toxicity, sepsis, pneumonia, febrile neutropenia, bacteremia, and hemorrhage.

Publications
Publications
Topics
Article Type
Display Headline
EMA grants accelerated assessment to drug for AML
Display Headline
EMA grants accelerated assessment to drug for AML
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

FDA lifts hold on trials of universal CAR T-cell therapy

Article Type
Changed
Wed, 11/08/2017 - 00:01
Display Headline
FDA lifts hold on trials of universal CAR T-cell therapy

Image from NIAID
T cells

The US Food and Drug Administration (FDA) has lifted the full clinical hold on 2 phase 1 studies of UCART123, an allogeneic chimeric antigen receptor (CAR) T-cell therapy targeting CD123.

One of these studies was designed for patients with acute myeloid leukemia (AML), and the other was designed for patients with blastic plasmacytoid dendritic cell neoplasm (BPDCN).

The hold meant no new subjects could be enrolled in either trial, and there could be no further dosing of subjects who were already enrolled.

The hold was placed in September because the first patient treated in the BPDCN trial died. The patient developed grade 2 cytokine release syndrome (CRS) and a grade 3 lung infection. This was followed by grade 4 capillary leak syndrome and grade 5 CRS.

The first patient treated in the AML trial also developed grade 4 capillary leak syndrome and grade 3 CRS, but both resolved.

Now, the FDA has lifted the hold on the trials because Cellectis, the company developing UCART123, agreed to implement the following main revisions to phase 1 UCART123 protocols:

  • Decrease the cohort dose level to 6.25 x 104 UCART123 cells/kg
  • Decrease the cyclophosphamide dose of the lymphodepleting regimen to 750 mg/m²/day over 3 days, with a maximum daily dose of 1.33 grams
  • Include specific criteria at Day 0, the day of UCART123 infusion, such as no new uncontrolled infection after receipt of lymphodepletion, afebrile, off all but replacement dose of corticosteroids, and no organ dysfunction since eligibility screening
  • Ensure the next 3 patients to be treated in each protocol will be under the age of 65
  • Ensure that enrollment will be staggered across the UCART123 protocols; at least 28 days should elapse between the enrollments of 2 patients across the 2 studies.

Cellectis is currently working with investigators and clinical sites to obtain internal review board approval on the revised protocols and resume patient enrollment.

Publications
Topics

Image from NIAID
T cells

The US Food and Drug Administration (FDA) has lifted the full clinical hold on 2 phase 1 studies of UCART123, an allogeneic chimeric antigen receptor (CAR) T-cell therapy targeting CD123.

One of these studies was designed for patients with acute myeloid leukemia (AML), and the other was designed for patients with blastic plasmacytoid dendritic cell neoplasm (BPDCN).

The hold meant no new subjects could be enrolled in either trial, and there could be no further dosing of subjects who were already enrolled.

The hold was placed in September because the first patient treated in the BPDCN trial died. The patient developed grade 2 cytokine release syndrome (CRS) and a grade 3 lung infection. This was followed by grade 4 capillary leak syndrome and grade 5 CRS.

The first patient treated in the AML trial also developed grade 4 capillary leak syndrome and grade 3 CRS, but both resolved.

Now, the FDA has lifted the hold on the trials because Cellectis, the company developing UCART123, agreed to implement the following main revisions to phase 1 UCART123 protocols:

  • Decrease the cohort dose level to 6.25 x 104 UCART123 cells/kg
  • Decrease the cyclophosphamide dose of the lymphodepleting regimen to 750 mg/m²/day over 3 days, with a maximum daily dose of 1.33 grams
  • Include specific criteria at Day 0, the day of UCART123 infusion, such as no new uncontrolled infection after receipt of lymphodepletion, afebrile, off all but replacement dose of corticosteroids, and no organ dysfunction since eligibility screening
  • Ensure the next 3 patients to be treated in each protocol will be under the age of 65
  • Ensure that enrollment will be staggered across the UCART123 protocols; at least 28 days should elapse between the enrollments of 2 patients across the 2 studies.

Cellectis is currently working with investigators and clinical sites to obtain internal review board approval on the revised protocols and resume patient enrollment.

Image from NIAID
T cells

The US Food and Drug Administration (FDA) has lifted the full clinical hold on 2 phase 1 studies of UCART123, an allogeneic chimeric antigen receptor (CAR) T-cell therapy targeting CD123.

One of these studies was designed for patients with acute myeloid leukemia (AML), and the other was designed for patients with blastic plasmacytoid dendritic cell neoplasm (BPDCN).

The hold meant no new subjects could be enrolled in either trial, and there could be no further dosing of subjects who were already enrolled.

The hold was placed in September because the first patient treated in the BPDCN trial died. The patient developed grade 2 cytokine release syndrome (CRS) and a grade 3 lung infection. This was followed by grade 4 capillary leak syndrome and grade 5 CRS.

The first patient treated in the AML trial also developed grade 4 capillary leak syndrome and grade 3 CRS, but both resolved.

Now, the FDA has lifted the hold on the trials because Cellectis, the company developing UCART123, agreed to implement the following main revisions to phase 1 UCART123 protocols:

  • Decrease the cohort dose level to 6.25 x 104 UCART123 cells/kg
  • Decrease the cyclophosphamide dose of the lymphodepleting regimen to 750 mg/m²/day over 3 days, with a maximum daily dose of 1.33 grams
  • Include specific criteria at Day 0, the day of UCART123 infusion, such as no new uncontrolled infection after receipt of lymphodepletion, afebrile, off all but replacement dose of corticosteroids, and no organ dysfunction since eligibility screening
  • Ensure the next 3 patients to be treated in each protocol will be under the age of 65
  • Ensure that enrollment will be staggered across the UCART123 protocols; at least 28 days should elapse between the enrollments of 2 patients across the 2 studies.

Cellectis is currently working with investigators and clinical sites to obtain internal review board approval on the revised protocols and resume patient enrollment.

Publications
Publications
Topics
Article Type
Display Headline
FDA lifts hold on trials of universal CAR T-cell therapy
Display Headline
FDA lifts hold on trials of universal CAR T-cell therapy
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Cancer drug costs increasing despite competition

Article Type
Changed
Mon, 11/06/2017 - 00:01
Display Headline
Cancer drug costs increasing despite competition

Photo by Bill Branson
Vials of drugs

Cancer drug costs in the US increase substantially after launch, regardless of competition, according to a study published in the Journal of Clinical Oncology.*

Researchers studied 24 cancer drugs approved over the last 20 years and found a mean cumulative cost increase of about 37%, or 19% when adjusted for inflation.

Among drugs approved to treat hematologic malignancies, the greatest inflation-adjusted price increases were for arsenic trioxide (57%), nelarabine (55%), and rituximab (49%).

The lowest inflation-adjusted price increases were for ofatumumab (8%), clofarabine (8%), and liposomal vincristine (18%).

For this study, Daniel A. Goldstein, MD, of Emory University in Atlanta, Georgia, and his colleagues measured the monthly price trajectories of 24 cancer drugs approved by the US Food and Drug Administration. This included 10 drugs approved to treat hematologic malignancies between 1997 and 2011.

To account for discounts and rebates, the researchers used the average sales prices published by the Centers for Medicare and Medicaid Services and adjusted to general and health-related inflation rates. For each drug, the researchers calculated the cumulative and annual drug cost changes.

Results

The mean follow-up was 8 years. The mean cumulative cost increase for all 24 drugs was +36.5% (95% CI, 24.7% to 48.3%).

The general inflation-adjusted increase was +19.1% (95% CI, 11.0% to 27.2%), and the health-related inflation-adjusted increase was +8.4% (95% CI, 1.4% to 15.4%).

Only 1 of the 24 drugs studied had a price decrease over time. That drug is ziv-aflibercept, which was approved to treat metastatic colorectal cancer in 2012.

Ziv-aflibercept was launched with an annual price exceeding $110,000. After public outcry, the drug’s manufacturer, Sanofi, cut the price in half. By the end of the study’s follow-up period in 2017, the cost of ziv-aflibercept had decreased 13% (inflation-adjusted decrease of 15%, health-related inflation-adjusted decrease of 20%).

Cost changes for the drugs approved to treat hematologic malignancies are listed in the following table.

Drug (indication, approval date, years of follow-up)

Mean monthly cost at launch Mean annual cost change (SD) Cumulative cost change General and health-related inflation-adjusted change, respectively

Arsenic trioxide (APL, 2000, 12)  $11,455 +6% (4) +95% +57%, +39%
Bendamustine (CLL, NHL, 2008, 8)  $6924 +5% (5) +50% +32%, +21%
Bortezomib (MM, MCL, 2003, 12)  $5490 +4% (3) +63% +31%, +16%
Brentuximab (lymphoma, 2011, 4)  $19,482 +8% (0.1) +35% +29%, +22%
Clofarabine (ALL, 2004, 11)  $56,486 +3% (3) +31% +8%, -4%
Liposomal vincristine (ALL, 2012, 3)  $34,602 +8% (0.5) +21% +18%, +14%
Nelarabine (ALL, lymphoma, 2005, 10)  $18,513 +6% (2) +83% +55%, +39%
Ofatumumab (CLL, 2009, 6)  $4538 +3% (2) +17% +8%, -0.5%
Pralatrexate (lymphoma, 2009, 6)  $31,684 +6% (4) +43% +31%, +21%
Rituximab (NHL, CLL, 1997, 12)  $4111 +5% (0.5) +85% +49%, +32%

Abbreviations: ALL, acute lymphoblastic leukemia; APL, acute promyelocytic leukemia; CLL, chronic lymphocytic leukemia; MCL, mantle cell lymphoma; MM, multiple myeloma; NHL, non-Hodgkin lymphoma; SD, standard deviation.

The researchers noted that there was a steady increase in drug costs over the study period, regardless of whether a drug was granted a new supplemental indication, the drug had a new off-label indication, or a competitor drug was approved.

The only variable that was significantly associated with price change was the amount of time that had elapsed from a drug’s launch.

This association was significant in models in which the researchers used prices adjusted to inflation (P=0.002) and health-related inflation (P=0.023). However, it was not significant when the researchers used the actual drug price (P=0.085).

*Data in the abstract differ from data in the body of the JCO paper. This article includes data from the body of the JCO paper.

Publications
Topics

Photo by Bill Branson
Vials of drugs

Cancer drug costs in the US increase substantially after launch, regardless of competition, according to a study published in the Journal of Clinical Oncology.*

Researchers studied 24 cancer drugs approved over the last 20 years and found a mean cumulative cost increase of about 37%, or 19% when adjusted for inflation.

Among drugs approved to treat hematologic malignancies, the greatest inflation-adjusted price increases were for arsenic trioxide (57%), nelarabine (55%), and rituximab (49%).

The lowest inflation-adjusted price increases were for ofatumumab (8%), clofarabine (8%), and liposomal vincristine (18%).

For this study, Daniel A. Goldstein, MD, of Emory University in Atlanta, Georgia, and his colleagues measured the monthly price trajectories of 24 cancer drugs approved by the US Food and Drug Administration. This included 10 drugs approved to treat hematologic malignancies between 1997 and 2011.

To account for discounts and rebates, the researchers used the average sales prices published by the Centers for Medicare and Medicaid Services and adjusted to general and health-related inflation rates. For each drug, the researchers calculated the cumulative and annual drug cost changes.

Results

The mean follow-up was 8 years. The mean cumulative cost increase for all 24 drugs was +36.5% (95% CI, 24.7% to 48.3%).

The general inflation-adjusted increase was +19.1% (95% CI, 11.0% to 27.2%), and the health-related inflation-adjusted increase was +8.4% (95% CI, 1.4% to 15.4%).

Only 1 of the 24 drugs studied had a price decrease over time. That drug is ziv-aflibercept, which was approved to treat metastatic colorectal cancer in 2012.

Ziv-aflibercept was launched with an annual price exceeding $110,000. After public outcry, the drug’s manufacturer, Sanofi, cut the price in half. By the end of the study’s follow-up period in 2017, the cost of ziv-aflibercept had decreased 13% (inflation-adjusted decrease of 15%, health-related inflation-adjusted decrease of 20%).

Cost changes for the drugs approved to treat hematologic malignancies are listed in the following table.

Drug (indication, approval date, years of follow-up)

Mean monthly cost at launch Mean annual cost change (SD) Cumulative cost change General and health-related inflation-adjusted change, respectively

Arsenic trioxide (APL, 2000, 12)  $11,455 +6% (4) +95% +57%, +39%
Bendamustine (CLL, NHL, 2008, 8)  $6924 +5% (5) +50% +32%, +21%
Bortezomib (MM, MCL, 2003, 12)  $5490 +4% (3) +63% +31%, +16%
Brentuximab (lymphoma, 2011, 4)  $19,482 +8% (0.1) +35% +29%, +22%
Clofarabine (ALL, 2004, 11)  $56,486 +3% (3) +31% +8%, -4%
Liposomal vincristine (ALL, 2012, 3)  $34,602 +8% (0.5) +21% +18%, +14%
Nelarabine (ALL, lymphoma, 2005, 10)  $18,513 +6% (2) +83% +55%, +39%
Ofatumumab (CLL, 2009, 6)  $4538 +3% (2) +17% +8%, -0.5%
Pralatrexate (lymphoma, 2009, 6)  $31,684 +6% (4) +43% +31%, +21%
Rituximab (NHL, CLL, 1997, 12)  $4111 +5% (0.5) +85% +49%, +32%

Abbreviations: ALL, acute lymphoblastic leukemia; APL, acute promyelocytic leukemia; CLL, chronic lymphocytic leukemia; MCL, mantle cell lymphoma; MM, multiple myeloma; NHL, non-Hodgkin lymphoma; SD, standard deviation.

The researchers noted that there was a steady increase in drug costs over the study period, regardless of whether a drug was granted a new supplemental indication, the drug had a new off-label indication, or a competitor drug was approved.

The only variable that was significantly associated with price change was the amount of time that had elapsed from a drug’s launch.

This association was significant in models in which the researchers used prices adjusted to inflation (P=0.002) and health-related inflation (P=0.023). However, it was not significant when the researchers used the actual drug price (P=0.085).

*Data in the abstract differ from data in the body of the JCO paper. This article includes data from the body of the JCO paper.

Photo by Bill Branson
Vials of drugs

Cancer drug costs in the US increase substantially after launch, regardless of competition, according to a study published in the Journal of Clinical Oncology.*

Researchers studied 24 cancer drugs approved over the last 20 years and found a mean cumulative cost increase of about 37%, or 19% when adjusted for inflation.

Among drugs approved to treat hematologic malignancies, the greatest inflation-adjusted price increases were for arsenic trioxide (57%), nelarabine (55%), and rituximab (49%).

The lowest inflation-adjusted price increases were for ofatumumab (8%), clofarabine (8%), and liposomal vincristine (18%).

For this study, Daniel A. Goldstein, MD, of Emory University in Atlanta, Georgia, and his colleagues measured the monthly price trajectories of 24 cancer drugs approved by the US Food and Drug Administration. This included 10 drugs approved to treat hematologic malignancies between 1997 and 2011.

To account for discounts and rebates, the researchers used the average sales prices published by the Centers for Medicare and Medicaid Services and adjusted to general and health-related inflation rates. For each drug, the researchers calculated the cumulative and annual drug cost changes.

Results

The mean follow-up was 8 years. The mean cumulative cost increase for all 24 drugs was +36.5% (95% CI, 24.7% to 48.3%).

The general inflation-adjusted increase was +19.1% (95% CI, 11.0% to 27.2%), and the health-related inflation-adjusted increase was +8.4% (95% CI, 1.4% to 15.4%).

Only 1 of the 24 drugs studied had a price decrease over time. That drug is ziv-aflibercept, which was approved to treat metastatic colorectal cancer in 2012.

Ziv-aflibercept was launched with an annual price exceeding $110,000. After public outcry, the drug’s manufacturer, Sanofi, cut the price in half. By the end of the study’s follow-up period in 2017, the cost of ziv-aflibercept had decreased 13% (inflation-adjusted decrease of 15%, health-related inflation-adjusted decrease of 20%).

Cost changes for the drugs approved to treat hematologic malignancies are listed in the following table.

Drug (indication, approval date, years of follow-up)

Mean monthly cost at launch Mean annual cost change (SD) Cumulative cost change General and health-related inflation-adjusted change, respectively

Arsenic trioxide (APL, 2000, 12)  $11,455 +6% (4) +95% +57%, +39%
Bendamustine (CLL, NHL, 2008, 8)  $6924 +5% (5) +50% +32%, +21%
Bortezomib (MM, MCL, 2003, 12)  $5490 +4% (3) +63% +31%, +16%
Brentuximab (lymphoma, 2011, 4)  $19,482 +8% (0.1) +35% +29%, +22%
Clofarabine (ALL, 2004, 11)  $56,486 +3% (3) +31% +8%, -4%
Liposomal vincristine (ALL, 2012, 3)  $34,602 +8% (0.5) +21% +18%, +14%
Nelarabine (ALL, lymphoma, 2005, 10)  $18,513 +6% (2) +83% +55%, +39%
Ofatumumab (CLL, 2009, 6)  $4538 +3% (2) +17% +8%, -0.5%
Pralatrexate (lymphoma, 2009, 6)  $31,684 +6% (4) +43% +31%, +21%
Rituximab (NHL, CLL, 1997, 12)  $4111 +5% (0.5) +85% +49%, +32%

Abbreviations: ALL, acute lymphoblastic leukemia; APL, acute promyelocytic leukemia; CLL, chronic lymphocytic leukemia; MCL, mantle cell lymphoma; MM, multiple myeloma; NHL, non-Hodgkin lymphoma; SD, standard deviation.

The researchers noted that there was a steady increase in drug costs over the study period, regardless of whether a drug was granted a new supplemental indication, the drug had a new off-label indication, or a competitor drug was approved.

The only variable that was significantly associated with price change was the amount of time that had elapsed from a drug’s launch.

This association was significant in models in which the researchers used prices adjusted to inflation (P=0.002) and health-related inflation (P=0.023). However, it was not significant when the researchers used the actual drug price (P=0.085).

*Data in the abstract differ from data in the body of the JCO paper. This article includes data from the body of the JCO paper.

Publications
Publications
Topics
Article Type
Display Headline
Cancer drug costs increasing despite competition
Display Headline
Cancer drug costs increasing despite competition
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Adequately nourished AML patients have survival advantage

Article Type
Changed
Fri, 01/04/2019 - 10:11

 

Good nutritional status can extend the lives of patients with acute myeloid leukemia going into induction chemotherapy, according to a retrospective study of 95 adult AML patients.

Those with good nutritional status had significantly shorter hospital stays than did undernourished patients. Furthermore, they had greater 12-month survival, compared with undernourished patients.

“Assessment of nutritional status is essential because undernutrition in this population is common,” Elise Deluche, MD, and her coinvestigators wrote (Nutrition. 2017 Sep;41:120-5). They assessed the nutritional status of 95 consecutive adult AML patients admitted to Limoges (France) University Hospital during 2009-2014 and followed their nutritional status for 12 months.Patients were considered undernourished if they had lost more than 5% of their weight, and had a body mass index (BMI) of under 18.5 kg/m2 if less than 70 years of age, or under 21 kg/m2 if aged at least 70 years.

Fourteen patients (15%) were undernourished at admission. That proportion grew after chemotherapy induction to 17 patients (18%), but there were no significant differences from admission in BMI, weight, or albumin.

The adequately nourished patients had significantly worse nutritional status at discharge than admission, with a significantly lower median weight (P =.02), BMI (P = .04), and albumin levels (P = .0002), compared with their admission values.

Importantly, the well nourished patients had shorter hospital stays than their undernourished counterparts, at 31 days, compared with 39 days (P = .03). Furthermore, their 12-month survival was greater, at 89.9%, than that of the undernourished patient, at 58.3% (P = .002).

After chemotherapy induction, 64 patients (67%) were in complete remission: 57 (70%) in the adequately nourished and 7 (50%) in the undernourished group, a nonsignificant difference.

This is the first study to look solely at patients with AML, Dr. DeLuche and her coinvestigators said, as previous nutritional studies have also included patients with acute lymphoblastic leukemia, and it “confirmed that the length of hospitalization was shorter for patients without undernutrition.” They added that their study included a more accurate representation of AML patients with a median patient age of 58 years, much older than the range of 28-41 found in other studies. A quarter of the patients in Dr. DeLuche’s study were over age 65.

“[Existing] screening tools should be improved and adapted to the specific situation of induction chemotherapy for monitoring nutritional status during hospitalization,” they concluded.

The study received no outside funding, and the investigators had no conflicts of interest.

Publications
Topics
Sections

 

Good nutritional status can extend the lives of patients with acute myeloid leukemia going into induction chemotherapy, according to a retrospective study of 95 adult AML patients.

Those with good nutritional status had significantly shorter hospital stays than did undernourished patients. Furthermore, they had greater 12-month survival, compared with undernourished patients.

“Assessment of nutritional status is essential because undernutrition in this population is common,” Elise Deluche, MD, and her coinvestigators wrote (Nutrition. 2017 Sep;41:120-5). They assessed the nutritional status of 95 consecutive adult AML patients admitted to Limoges (France) University Hospital during 2009-2014 and followed their nutritional status for 12 months.Patients were considered undernourished if they had lost more than 5% of their weight, and had a body mass index (BMI) of under 18.5 kg/m2 if less than 70 years of age, or under 21 kg/m2 if aged at least 70 years.

Fourteen patients (15%) were undernourished at admission. That proportion grew after chemotherapy induction to 17 patients (18%), but there were no significant differences from admission in BMI, weight, or albumin.

The adequately nourished patients had significantly worse nutritional status at discharge than admission, with a significantly lower median weight (P =.02), BMI (P = .04), and albumin levels (P = .0002), compared with their admission values.

Importantly, the well nourished patients had shorter hospital stays than their undernourished counterparts, at 31 days, compared with 39 days (P = .03). Furthermore, their 12-month survival was greater, at 89.9%, than that of the undernourished patient, at 58.3% (P = .002).

After chemotherapy induction, 64 patients (67%) were in complete remission: 57 (70%) in the adequately nourished and 7 (50%) in the undernourished group, a nonsignificant difference.

This is the first study to look solely at patients with AML, Dr. DeLuche and her coinvestigators said, as previous nutritional studies have also included patients with acute lymphoblastic leukemia, and it “confirmed that the length of hospitalization was shorter for patients without undernutrition.” They added that their study included a more accurate representation of AML patients with a median patient age of 58 years, much older than the range of 28-41 found in other studies. A quarter of the patients in Dr. DeLuche’s study were over age 65.

“[Existing] screening tools should be improved and adapted to the specific situation of induction chemotherapy for monitoring nutritional status during hospitalization,” they concluded.

The study received no outside funding, and the investigators had no conflicts of interest.

 

Good nutritional status can extend the lives of patients with acute myeloid leukemia going into induction chemotherapy, according to a retrospective study of 95 adult AML patients.

Those with good nutritional status had significantly shorter hospital stays than did undernourished patients. Furthermore, they had greater 12-month survival, compared with undernourished patients.

“Assessment of nutritional status is essential because undernutrition in this population is common,” Elise Deluche, MD, and her coinvestigators wrote (Nutrition. 2017 Sep;41:120-5). They assessed the nutritional status of 95 consecutive adult AML patients admitted to Limoges (France) University Hospital during 2009-2014 and followed their nutritional status for 12 months.Patients were considered undernourished if they had lost more than 5% of their weight, and had a body mass index (BMI) of under 18.5 kg/m2 if less than 70 years of age, or under 21 kg/m2 if aged at least 70 years.

Fourteen patients (15%) were undernourished at admission. That proportion grew after chemotherapy induction to 17 patients (18%), but there were no significant differences from admission in BMI, weight, or albumin.

The adequately nourished patients had significantly worse nutritional status at discharge than admission, with a significantly lower median weight (P =.02), BMI (P = .04), and albumin levels (P = .0002), compared with their admission values.

Importantly, the well nourished patients had shorter hospital stays than their undernourished counterparts, at 31 days, compared with 39 days (P = .03). Furthermore, their 12-month survival was greater, at 89.9%, than that of the undernourished patient, at 58.3% (P = .002).

After chemotherapy induction, 64 patients (67%) were in complete remission: 57 (70%) in the adequately nourished and 7 (50%) in the undernourished group, a nonsignificant difference.

This is the first study to look solely at patients with AML, Dr. DeLuche and her coinvestigators said, as previous nutritional studies have also included patients with acute lymphoblastic leukemia, and it “confirmed that the length of hospitalization was shorter for patients without undernutrition.” They added that their study included a more accurate representation of AML patients with a median patient age of 58 years, much older than the range of 28-41 found in other studies. A quarter of the patients in Dr. DeLuche’s study were over age 65.

“[Existing] screening tools should be improved and adapted to the specific situation of induction chemotherapy for monitoring nutritional status during hospitalization,” they concluded.

The study received no outside funding, and the investigators had no conflicts of interest.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM NUTRITION

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

 

Key clinical point: Adequately nourished acute myeloid leukemia patients have shorter hospital stays and longer survival after chemotherapy.

Major finding: AML patients who were adequately nourished going into induction chemotherapy had significantly shorter hospital stays (31 days versus 39 days) and greater 12-month survival than did those who were undernourished (89.9% versus 58.3%).

Data source: A study of 95 consecutive AML patients admitted to a single center and assessed for nutritional status before and after induction chemotherapy.

Disclosures: The study received no outside funding, and the investigators had no conflicts of interest.

Disqus Comments
Default