Meeting ID
5340-17
Series ID
2017
Display Conference Events In Series

MDS genetic analysis identifies allogeneic HSCT candidates

Article Type
Changed
Tue, 07/21/2020 - 14:18

 

– Genetic mutation analysis of patients with myelodysplastic syndrome (MDS) may have a useful role in routine practice based on recent reports that showed clear links between certain gene mutations and the outcomes of patients who underwent allogeneic hematopoietic stem cell transplantation (HSCT).

Two reports published in 2017 helped strengthen the case for routine mutation analysis in distinguishing patients with MDS or myeloproliferative neoplasms (MDN) who are very likely to have just a brief response to allogeneic HSCT from similar patients who seem likely to have several years of overall survival following transplantation.

Dr. Amer M. Zeidan of Yale University, New Haven, Conn.
Mitchel L. Zoler/Frontline Medical News
Dr. Amer M. Zeidan
When patients have markers for poor outcomes, “consider alternatives to allogeneic HSCT,” such as enrolling them in a trial, Amer M. Zeidan, MBBS, advised at a conference held by Imedex. “For other patients, allogeneic HSCT is reasonable, especially [for] younger patients,” those less than 40 years old, said Dr. Zeidan, a hematologist/oncologist at Yale University in New Haven, Conn.

Allogeneic HSCT is the only potentially curative procedure for patients with MDS or MDN. Although an increasing number of these patients undergo transplantation, clinicians need to choose the patients they select for the treatment carefully. “Molecular testing is playing an increasing role in selecting the best candidates,” Dr. Zeidan said.

The largest reported genetic study of allogeneic HSCT in MDS patients involved 1,514 patients entered into a U.S.-based dataset during 2005-2015. Testing identified at least one mutation in 1,196 (79%) of these patients.

Analysis of data from these patients found a disparate pattern of posttransplant survival that appeared to link with gene mutations and other risk factors. The highest risk patients were those with a mutation in their TP53 gene, found in 289 patients (19% of the 1,514 tested) who had a median overall survival (OS) of 0.7 years and a 3-year OS of 20% (New Engl J Med. 2017 Feb 9;376[6]:536-47).

Among patients without a TP53 mutation, OS depended on age, with the best survival seen among patients less than 40 years old. Patients in this subgroup who also had no other high-risk features – no therapy-related MDS, a platelet level of at least 30 x 109 at the time of transplantation, and bone marrow blasts less than 15% at diagnosis – had the best OS, 82% at 3-years of follow-up. The studied cohort included 116 patients (8%) who fell into this low-risk, best-outcome category, the optimal population for receiving an allogeneic HSCT, Dr. Zeidan said. Another 98 patients (6%) who had at least one of these high risk feature had a median OS of 2.6 years and a 3-year OS of 49%.

Additional gene mutations further subdivided the older patients in the study, those at least 40 years old, into various risk subgroups. Older patients with a mutation in a ras-pathway gene had a 0.9 year median OS and a 3-year OS of 30%. This subgroup included 129 patients (9%). Among older patients with no mutation in the ras-pathway gene, mutations in the JAK2 gene also linked with worse survival, a median OS of 0.5 years and a 3-year OS of 28% of a subgroup with 28 patients (2%). The largest subgroup in the study was older patients with no mutations in the TP53, JAK2, or ras-pathway genes, a subgroup with 854 patients (56%), who had a median OS of 2.3 years and a 3-year OS of 46%.

The second recent report was a Japanese study of 797 MDS patients who underwent genetic testing and received an allogeneic HSCT through the Japan Marrow Donor Program. The investigators found identifiable mutations in 617 patients (77%) and documented that patients with a TP53 or ras-pathway mutation had a “dismal prognosis” when associated with a complex karyotype and myelodysplastic or myeloproliferative neoplasms. However, among patients with a mutated TP53 gene or complex karyotype alone, long-term survival following transplantation appeared possible (Blood. 2017. doi: org/10.1182/blood-2016-12-754796.

Two smaller, earlier studies (J Clin Oncol. 2014 Sept 1;32[25]:2691-8; J Clin Oncol. 2016 Oct 20;34[30]:2627-37) also implicated mutations in the TET2, DNMT3A, ASXL1, and RUNX1 genes as identifying MDS patients with worse OS following allogeneic HSCT, Dr. Zeidan noted, but the combination of a TP53 gene mutation and a complex karyotype appears to confer the worst prognosis of all. Patients with mutations in more than one of these genes fared much worse than those with single mutations.

Dr. Zeidan had no relevant disclosures.

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

 

– Genetic mutation analysis of patients with myelodysplastic syndrome (MDS) may have a useful role in routine practice based on recent reports that showed clear links between certain gene mutations and the outcomes of patients who underwent allogeneic hematopoietic stem cell transplantation (HSCT).

Two reports published in 2017 helped strengthen the case for routine mutation analysis in distinguishing patients with MDS or myeloproliferative neoplasms (MDN) who are very likely to have just a brief response to allogeneic HSCT from similar patients who seem likely to have several years of overall survival following transplantation.

Dr. Amer M. Zeidan of Yale University, New Haven, Conn.
Mitchel L. Zoler/Frontline Medical News
Dr. Amer M. Zeidan
When patients have markers for poor outcomes, “consider alternatives to allogeneic HSCT,” such as enrolling them in a trial, Amer M. Zeidan, MBBS, advised at a conference held by Imedex. “For other patients, allogeneic HSCT is reasonable, especially [for] younger patients,” those less than 40 years old, said Dr. Zeidan, a hematologist/oncologist at Yale University in New Haven, Conn.

Allogeneic HSCT is the only potentially curative procedure for patients with MDS or MDN. Although an increasing number of these patients undergo transplantation, clinicians need to choose the patients they select for the treatment carefully. “Molecular testing is playing an increasing role in selecting the best candidates,” Dr. Zeidan said.

The largest reported genetic study of allogeneic HSCT in MDS patients involved 1,514 patients entered into a U.S.-based dataset during 2005-2015. Testing identified at least one mutation in 1,196 (79%) of these patients.

Analysis of data from these patients found a disparate pattern of posttransplant survival that appeared to link with gene mutations and other risk factors. The highest risk patients were those with a mutation in their TP53 gene, found in 289 patients (19% of the 1,514 tested) who had a median overall survival (OS) of 0.7 years and a 3-year OS of 20% (New Engl J Med. 2017 Feb 9;376[6]:536-47).

Among patients without a TP53 mutation, OS depended on age, with the best survival seen among patients less than 40 years old. Patients in this subgroup who also had no other high-risk features – no therapy-related MDS, a platelet level of at least 30 x 109 at the time of transplantation, and bone marrow blasts less than 15% at diagnosis – had the best OS, 82% at 3-years of follow-up. The studied cohort included 116 patients (8%) who fell into this low-risk, best-outcome category, the optimal population for receiving an allogeneic HSCT, Dr. Zeidan said. Another 98 patients (6%) who had at least one of these high risk feature had a median OS of 2.6 years and a 3-year OS of 49%.

Additional gene mutations further subdivided the older patients in the study, those at least 40 years old, into various risk subgroups. Older patients with a mutation in a ras-pathway gene had a 0.9 year median OS and a 3-year OS of 30%. This subgroup included 129 patients (9%). Among older patients with no mutation in the ras-pathway gene, mutations in the JAK2 gene also linked with worse survival, a median OS of 0.5 years and a 3-year OS of 28% of a subgroup with 28 patients (2%). The largest subgroup in the study was older patients with no mutations in the TP53, JAK2, or ras-pathway genes, a subgroup with 854 patients (56%), who had a median OS of 2.3 years and a 3-year OS of 46%.

The second recent report was a Japanese study of 797 MDS patients who underwent genetic testing and received an allogeneic HSCT through the Japan Marrow Donor Program. The investigators found identifiable mutations in 617 patients (77%) and documented that patients with a TP53 or ras-pathway mutation had a “dismal prognosis” when associated with a complex karyotype and myelodysplastic or myeloproliferative neoplasms. However, among patients with a mutated TP53 gene or complex karyotype alone, long-term survival following transplantation appeared possible (Blood. 2017. doi: org/10.1182/blood-2016-12-754796.

Two smaller, earlier studies (J Clin Oncol. 2014 Sept 1;32[25]:2691-8; J Clin Oncol. 2016 Oct 20;34[30]:2627-37) also implicated mutations in the TET2, DNMT3A, ASXL1, and RUNX1 genes as identifying MDS patients with worse OS following allogeneic HSCT, Dr. Zeidan noted, but the combination of a TP53 gene mutation and a complex karyotype appears to confer the worst prognosis of all. Patients with mutations in more than one of these genes fared much worse than those with single mutations.

Dr. Zeidan had no relevant disclosures.

 

– Genetic mutation analysis of patients with myelodysplastic syndrome (MDS) may have a useful role in routine practice based on recent reports that showed clear links between certain gene mutations and the outcomes of patients who underwent allogeneic hematopoietic stem cell transplantation (HSCT).

Two reports published in 2017 helped strengthen the case for routine mutation analysis in distinguishing patients with MDS or myeloproliferative neoplasms (MDN) who are very likely to have just a brief response to allogeneic HSCT from similar patients who seem likely to have several years of overall survival following transplantation.

Dr. Amer M. Zeidan of Yale University, New Haven, Conn.
Mitchel L. Zoler/Frontline Medical News
Dr. Amer M. Zeidan
When patients have markers for poor outcomes, “consider alternatives to allogeneic HSCT,” such as enrolling them in a trial, Amer M. Zeidan, MBBS, advised at a conference held by Imedex. “For other patients, allogeneic HSCT is reasonable, especially [for] younger patients,” those less than 40 years old, said Dr. Zeidan, a hematologist/oncologist at Yale University in New Haven, Conn.

Allogeneic HSCT is the only potentially curative procedure for patients with MDS or MDN. Although an increasing number of these patients undergo transplantation, clinicians need to choose the patients they select for the treatment carefully. “Molecular testing is playing an increasing role in selecting the best candidates,” Dr. Zeidan said.

The largest reported genetic study of allogeneic HSCT in MDS patients involved 1,514 patients entered into a U.S.-based dataset during 2005-2015. Testing identified at least one mutation in 1,196 (79%) of these patients.

Analysis of data from these patients found a disparate pattern of posttransplant survival that appeared to link with gene mutations and other risk factors. The highest risk patients were those with a mutation in their TP53 gene, found in 289 patients (19% of the 1,514 tested) who had a median overall survival (OS) of 0.7 years and a 3-year OS of 20% (New Engl J Med. 2017 Feb 9;376[6]:536-47).

Among patients without a TP53 mutation, OS depended on age, with the best survival seen among patients less than 40 years old. Patients in this subgroup who also had no other high-risk features – no therapy-related MDS, a platelet level of at least 30 x 109 at the time of transplantation, and bone marrow blasts less than 15% at diagnosis – had the best OS, 82% at 3-years of follow-up. The studied cohort included 116 patients (8%) who fell into this low-risk, best-outcome category, the optimal population for receiving an allogeneic HSCT, Dr. Zeidan said. Another 98 patients (6%) who had at least one of these high risk feature had a median OS of 2.6 years and a 3-year OS of 49%.

Additional gene mutations further subdivided the older patients in the study, those at least 40 years old, into various risk subgroups. Older patients with a mutation in a ras-pathway gene had a 0.9 year median OS and a 3-year OS of 30%. This subgroup included 129 patients (9%). Among older patients with no mutation in the ras-pathway gene, mutations in the JAK2 gene also linked with worse survival, a median OS of 0.5 years and a 3-year OS of 28% of a subgroup with 28 patients (2%). The largest subgroup in the study was older patients with no mutations in the TP53, JAK2, or ras-pathway genes, a subgroup with 854 patients (56%), who had a median OS of 2.3 years and a 3-year OS of 46%.

The second recent report was a Japanese study of 797 MDS patients who underwent genetic testing and received an allogeneic HSCT through the Japan Marrow Donor Program. The investigators found identifiable mutations in 617 patients (77%) and documented that patients with a TP53 or ras-pathway mutation had a “dismal prognosis” when associated with a complex karyotype and myelodysplastic or myeloproliferative neoplasms. However, among patients with a mutated TP53 gene or complex karyotype alone, long-term survival following transplantation appeared possible (Blood. 2017. doi: org/10.1182/blood-2016-12-754796.

Two smaller, earlier studies (J Clin Oncol. 2014 Sept 1;32[25]:2691-8; J Clin Oncol. 2016 Oct 20;34[30]:2627-37) also implicated mutations in the TET2, DNMT3A, ASXL1, and RUNX1 genes as identifying MDS patients with worse OS following allogeneic HSCT, Dr. Zeidan noted, but the combination of a TP53 gene mutation and a complex karyotype appears to confer the worst prognosis of all. Patients with mutations in more than one of these genes fared much worse than those with single mutations.

Dr. Zeidan had no relevant disclosures.

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

Targeted drugs transform CLL management

Article Type
Changed
Thu, 01/12/2023 - 10:46

 

NEW YORK– New, targeted treatments, especially ibrutinib (Imbruvica), have sharply shifted prognosis for patients with chronic lymphocytic leukemia (CLL) and raised new issues for managing these patients now that they survive years longer.

“Ibrutinib has produced a profound change in survival” of patients with CLL, Timothy G. Call, MD, a hematologist/oncologist at the Mayo Clinic in Rochester, Minn., said at a conference held by Imedex. It “has changed the playing field.” No other new agent so far “has produced the same level of progression-free survival in CLL.”

Dr. Timothy G. Call
Mitchel L. Zoler/Frontline Medical News
Dr. Timothy G. Call
The consequence has been a steep increase in prevalence of CLL as patients live longer, producing new issues in patient monitoring and the costs of treatment both for individual patients and for society.

An analysis published in early 2017 projected a greater than 50% jump in U.S. patients living with CLL from 2011, before the advent of targeted oral drugs, to 2025, when the study predicted that there will be nearly 200,000 U.S. patients living with CLL (J Clin Oncol. 2017 January;35[2]:166-74). With targeted drugs like ibrutinib and idelalisib (Zydelig) costing about $130,000 per patient each year, the projected cost for managing the U.S. CLL population is on track to rise to more than $5 billion by 2025, a nearly sixfold increase, compared with CLL patient expenditures in 2011, according to this analysis.

The impact of the higher cost of treatment is already being felt more acutely by many patients because of recent cuts in assistance from the Patient Access Network, which helps patients with copays but recently had to put a lid on CLL assistance availability when its funding availability hit a wall, Dr. Call said.

On the clinical side, there are new considerations triggered by greater patient longevity. “As we make patients live longer with CLL, we need to double down on the diagnosis and treatment of its complications,” such as watching for development of secondary cancers, Dr. Call said in an interview. This stems from the reduced immunosurveillance in patients with CLL and their resulting increased susceptibility to developing environmentally-triggered malignancies like lung and skin cancers. Other long-term implications of impaired immunosurveillance include increased infection susceptibility, an ongoing risk for Richter’s or Hodgkin lymphoma transformation, and a risk for autoimmune complications, such as red blood cell aplasia and transfusion-associated graft versus host disease.

Patients with CLL can also be susceptible to complications from long-term use of the targeted drug they’re on. The new targeted agents can trigger bruising and bleeding, diarrhea, rash, fatigue, muscle and joint aches, and arrhythmia, he noted.

Potential adverse effects, specifically from ibrutinib, include a 3% risk for a major bleed, a 10% rate of new-onset atrial fibrillation, and a 20% risk for new hypertension, Dr. Call said. Before starting ibrutinib, patients should undergo screening for hepatitis B virus infection and receive prophylaxis against herpes zoster activation with acyclovir or valacyclovir. If the patient starts with a CD4 cell count below 200 cells/mm3, it might be prudent to prophylax the patient against Pneumocystis jirovecii pneumonia.

However, even if a toxicity develops on ibrutinib, Dr. Call recommended reducing the dosage rather than discontinuing the drug. “I rarely see a loss in response from a reduced dosage of ibrutinib,” he said.

Because ibrutinib is primarily metabolized via the liver enzyme cytochrome P450 3A (CYP3A), other drugs that enhance or reduce the activity of this enzyme produce significant changes in ibrutinib levels. The Food and Drug Administration considers ibrutinib a “sensitive substrate” for fluctuations in CYP3A activity. Strong CYP3A inhibitors include clarithromycin, ketoconazole, and various anti-HIV medications; moderate CYP3A4 inhibitors include ciprofloxacin, and verapamil; and inducers of CYP3A include phenytoin and rifampin. A more complete list of the drugs that inhibit or induce CYP3A activity can be found at the FDA website: https://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/DrugInteractionsLabeling/ucm093664.htm#table2-2.

Dr. Call presented the combined experience from several U.S. Mayo Clinic centers for 118 patients treated with ibrutinib after the drug received FDA marketing approval in November 2013. The clinicians identified 75 patients (64%) who were on a concurrent medication that could potentially increase the risk for ibrutinib toxicity and 4 patients (3%) who were on a concurrent drug with the potential to reduce ibrutinib efficacy (Leukemia Lymphoma, 2017;58[6]:1376-83).

“We don’t change the dosage of ibrutinib when the patient is on a CYP3A inducer, but, if the patient is getting a CYP3A inhibitor, we change that to another drug or reduce the ibrutinib dosage,” Dr. Call said.

Dr. Call had no disclosures.

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

 

NEW YORK– New, targeted treatments, especially ibrutinib (Imbruvica), have sharply shifted prognosis for patients with chronic lymphocytic leukemia (CLL) and raised new issues for managing these patients now that they survive years longer.

“Ibrutinib has produced a profound change in survival” of patients with CLL, Timothy G. Call, MD, a hematologist/oncologist at the Mayo Clinic in Rochester, Minn., said at a conference held by Imedex. It “has changed the playing field.” No other new agent so far “has produced the same level of progression-free survival in CLL.”

Dr. Timothy G. Call
Mitchel L. Zoler/Frontline Medical News
Dr. Timothy G. Call
The consequence has been a steep increase in prevalence of CLL as patients live longer, producing new issues in patient monitoring and the costs of treatment both for individual patients and for society.

An analysis published in early 2017 projected a greater than 50% jump in U.S. patients living with CLL from 2011, before the advent of targeted oral drugs, to 2025, when the study predicted that there will be nearly 200,000 U.S. patients living with CLL (J Clin Oncol. 2017 January;35[2]:166-74). With targeted drugs like ibrutinib and idelalisib (Zydelig) costing about $130,000 per patient each year, the projected cost for managing the U.S. CLL population is on track to rise to more than $5 billion by 2025, a nearly sixfold increase, compared with CLL patient expenditures in 2011, according to this analysis.

The impact of the higher cost of treatment is already being felt more acutely by many patients because of recent cuts in assistance from the Patient Access Network, which helps patients with copays but recently had to put a lid on CLL assistance availability when its funding availability hit a wall, Dr. Call said.

On the clinical side, there are new considerations triggered by greater patient longevity. “As we make patients live longer with CLL, we need to double down on the diagnosis and treatment of its complications,” such as watching for development of secondary cancers, Dr. Call said in an interview. This stems from the reduced immunosurveillance in patients with CLL and their resulting increased susceptibility to developing environmentally-triggered malignancies like lung and skin cancers. Other long-term implications of impaired immunosurveillance include increased infection susceptibility, an ongoing risk for Richter’s or Hodgkin lymphoma transformation, and a risk for autoimmune complications, such as red blood cell aplasia and transfusion-associated graft versus host disease.

Patients with CLL can also be susceptible to complications from long-term use of the targeted drug they’re on. The new targeted agents can trigger bruising and bleeding, diarrhea, rash, fatigue, muscle and joint aches, and arrhythmia, he noted.

Potential adverse effects, specifically from ibrutinib, include a 3% risk for a major bleed, a 10% rate of new-onset atrial fibrillation, and a 20% risk for new hypertension, Dr. Call said. Before starting ibrutinib, patients should undergo screening for hepatitis B virus infection and receive prophylaxis against herpes zoster activation with acyclovir or valacyclovir. If the patient starts with a CD4 cell count below 200 cells/mm3, it might be prudent to prophylax the patient against Pneumocystis jirovecii pneumonia.

However, even if a toxicity develops on ibrutinib, Dr. Call recommended reducing the dosage rather than discontinuing the drug. “I rarely see a loss in response from a reduced dosage of ibrutinib,” he said.

Because ibrutinib is primarily metabolized via the liver enzyme cytochrome P450 3A (CYP3A), other drugs that enhance or reduce the activity of this enzyme produce significant changes in ibrutinib levels. The Food and Drug Administration considers ibrutinib a “sensitive substrate” for fluctuations in CYP3A activity. Strong CYP3A inhibitors include clarithromycin, ketoconazole, and various anti-HIV medications; moderate CYP3A4 inhibitors include ciprofloxacin, and verapamil; and inducers of CYP3A include phenytoin and rifampin. A more complete list of the drugs that inhibit or induce CYP3A activity can be found at the FDA website: https://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/DrugInteractionsLabeling/ucm093664.htm#table2-2.

Dr. Call presented the combined experience from several U.S. Mayo Clinic centers for 118 patients treated with ibrutinib after the drug received FDA marketing approval in November 2013. The clinicians identified 75 patients (64%) who were on a concurrent medication that could potentially increase the risk for ibrutinib toxicity and 4 patients (3%) who were on a concurrent drug with the potential to reduce ibrutinib efficacy (Leukemia Lymphoma, 2017;58[6]:1376-83).

“We don’t change the dosage of ibrutinib when the patient is on a CYP3A inducer, but, if the patient is getting a CYP3A inhibitor, we change that to another drug or reduce the ibrutinib dosage,” Dr. Call said.

Dr. Call had no disclosures.

 

NEW YORK– New, targeted treatments, especially ibrutinib (Imbruvica), have sharply shifted prognosis for patients with chronic lymphocytic leukemia (CLL) and raised new issues for managing these patients now that they survive years longer.

“Ibrutinib has produced a profound change in survival” of patients with CLL, Timothy G. Call, MD, a hematologist/oncologist at the Mayo Clinic in Rochester, Minn., said at a conference held by Imedex. It “has changed the playing field.” No other new agent so far “has produced the same level of progression-free survival in CLL.”

Dr. Timothy G. Call
Mitchel L. Zoler/Frontline Medical News
Dr. Timothy G. Call
The consequence has been a steep increase in prevalence of CLL as patients live longer, producing new issues in patient monitoring and the costs of treatment both for individual patients and for society.

An analysis published in early 2017 projected a greater than 50% jump in U.S. patients living with CLL from 2011, before the advent of targeted oral drugs, to 2025, when the study predicted that there will be nearly 200,000 U.S. patients living with CLL (J Clin Oncol. 2017 January;35[2]:166-74). With targeted drugs like ibrutinib and idelalisib (Zydelig) costing about $130,000 per patient each year, the projected cost for managing the U.S. CLL population is on track to rise to more than $5 billion by 2025, a nearly sixfold increase, compared with CLL patient expenditures in 2011, according to this analysis.

The impact of the higher cost of treatment is already being felt more acutely by many patients because of recent cuts in assistance from the Patient Access Network, which helps patients with copays but recently had to put a lid on CLL assistance availability when its funding availability hit a wall, Dr. Call said.

On the clinical side, there are new considerations triggered by greater patient longevity. “As we make patients live longer with CLL, we need to double down on the diagnosis and treatment of its complications,” such as watching for development of secondary cancers, Dr. Call said in an interview. This stems from the reduced immunosurveillance in patients with CLL and their resulting increased susceptibility to developing environmentally-triggered malignancies like lung and skin cancers. Other long-term implications of impaired immunosurveillance include increased infection susceptibility, an ongoing risk for Richter’s or Hodgkin lymphoma transformation, and a risk for autoimmune complications, such as red blood cell aplasia and transfusion-associated graft versus host disease.

Patients with CLL can also be susceptible to complications from long-term use of the targeted drug they’re on. The new targeted agents can trigger bruising and bleeding, diarrhea, rash, fatigue, muscle and joint aches, and arrhythmia, he noted.

Potential adverse effects, specifically from ibrutinib, include a 3% risk for a major bleed, a 10% rate of new-onset atrial fibrillation, and a 20% risk for new hypertension, Dr. Call said. Before starting ibrutinib, patients should undergo screening for hepatitis B virus infection and receive prophylaxis against herpes zoster activation with acyclovir or valacyclovir. If the patient starts with a CD4 cell count below 200 cells/mm3, it might be prudent to prophylax the patient against Pneumocystis jirovecii pneumonia.

However, even if a toxicity develops on ibrutinib, Dr. Call recommended reducing the dosage rather than discontinuing the drug. “I rarely see a loss in response from a reduced dosage of ibrutinib,” he said.

Because ibrutinib is primarily metabolized via the liver enzyme cytochrome P450 3A (CYP3A), other drugs that enhance or reduce the activity of this enzyme produce significant changes in ibrutinib levels. The Food and Drug Administration considers ibrutinib a “sensitive substrate” for fluctuations in CYP3A activity. Strong CYP3A inhibitors include clarithromycin, ketoconazole, and various anti-HIV medications; moderate CYP3A4 inhibitors include ciprofloxacin, and verapamil; and inducers of CYP3A include phenytoin and rifampin. A more complete list of the drugs that inhibit or induce CYP3A activity can be found at the FDA website: https://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/DrugInteractionsLabeling/ucm093664.htm#table2-2.

Dr. Call presented the combined experience from several U.S. Mayo Clinic centers for 118 patients treated with ibrutinib after the drug received FDA marketing approval in November 2013. The clinicians identified 75 patients (64%) who were on a concurrent medication that could potentially increase the risk for ibrutinib toxicity and 4 patients (3%) who were on a concurrent drug with the potential to reduce ibrutinib efficacy (Leukemia Lymphoma, 2017;58[6]:1376-83).

“We don’t change the dosage of ibrutinib when the patient is on a CYP3A inducer, but, if the patient is getting a CYP3A inhibitor, we change that to another drug or reduce the ibrutinib dosage,” Dr. Call said.

Dr. Call had no disclosures.

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

Lenalidomide maintains posttransplant remissions in myeloma

Article Type
Changed
Tue, 07/21/2020 - 14:18

 

– Recent trial results have shown the importance of treating patients with multiple myeloma with the immunomodulator lenalidomide for maintaining negative minimal residual disease and sustained complete responses following autologous stem cell transplantation, experts said at a conference held by Imedex.

“How do you keep the therapy pedal to the metal over time? Lenalidomide is easy to deliver, convenient, and improves progression-free survival and overall survival,” said Paul G. Richardson, MD, professor of medicine at Harvard University and clinical program leader of the Multiple Myeloma Center at the Dana-Farber Cancer Institute in Boston. “Lenalidomide maintenance is standard of care. It provides a platform on which you can reliably add new agents for maintenance of remission following autologous stem cell transplantation,” in patients with multiple myeloma.

Mitchel L. Zoler/Frontline Medical News
Dr. Paul G. Richardson
Dr. Richardson said that some of the best evidence for lenalidomide’s efficacy in posttransplant patients came in a 2012 report from a phase III trial that compared lenalidomide (Revlimid) maintenance with placebo in 460 patients following induction. The results showed that maintenance with 5-15 mg/day of lenalidomide reduced the rate of either progressive disease or death by 63% (hazard ratio, 0.37; 95% confidence interval, 0.26-0.53), compared with placebo, during a median follow-up of 18 months (N Engl J Med. 2012 May 10;366[19]:1770-81).

In a meta-analysis of three randomized, controlled trials with 1,208 patients with multiple myeloma who had undergone induction and autologous stem cell transplant, the overall mortality rate dropped by 0.75 (95% CI, 0.63-0.90) among patients maintained on lenalidomide, compared with placebo (J Clin Oncol. 2016;34:suppl;abstract 8001).

A third study that Dr. Richardson cited showed the difficulty of improving on lenalidomide. The BMT CTN 0702 (Stem Cell Transplant With Lenalidomide Maintenance in Patients With Multiple Myeloma) trial randomized 758 multiple myeloma patients to three different autologous stem cell transplant regimens, each followed by lenalidomide maintenance. One arm followed the transplant with four cycles of consolidation therapy with lenalidomide, dexamethasone, and bortezomib (Velcade); one arm used two tandem transplantations; and the third arm used a single transplantation. All three arms had similar rates of progression-free survival and overall survival during follow-up (Blood. 2016 Dec 6;LBA-1). The results showed that “lenalidomide maintenance is an equalizer,” Dr. Richardson said.

Dr. Suzanne Lentzsch
Mitchel L. Zoler/Frontline Medical News
Dr. Suzanne Lentzsch
While agreeing that lenalidomide is a key treatment following autologous stem cell transplant in patients with multiple myeloma, Suzanne Lentzsch, MD, highlighted recent, limited evidence that showed that the best outcomes occurred in posttransplant patients who achieved “deep and sustained,” complete responses on prolonged treatment with a combination consolidation regimen.

She cited a U.S., multicenter, phase II study that followed 66 newly diagnosed patients with multiple myeloma who proceeded through induction, autologous stem cell transplantation, and multiple cycles of consolidation therapy with a regimen of lenalidomide, the proteasome inhibitor carfilzomib (Kyprolis), and dexamethasone. Fifty patients went through 18 cycles of this consolidation regimen and showed an 84% rate of stringent complete response, “the first time we’ve seen such results,” said Dr. Lentzsch, director of the Multiple Myeloma and Amyloidosis Program at Columbia University in New York. “This translated into an excellent” progression-free survival of 86% after 3 years and an overall survival of 95% after 3 years (Blood. 2016 Dec 5;Abstract 675). “I’m pretty impressed” by the results, she added.

An advantage of the combined regimen used in this study is that it is “relatively well tolerated,” as well as effective for keeping patients in remission, Dr. Lentzsch said. However, she highlighted that this was a small study, so its treatment implications are limited for the time being. “For high-risk patients, use the full combination. For everyone else, we need to wait for results from a randomized, controlled trial,” she advised.

Dr. Richardson has been a consultant to Celgene, the company that markets lenalidomide, and is also a consultant to Genmab, Janssen, Novartis, Oncopeptides, and Takeda and has received research funding from Celgene and Takeda. Dr. Lentzsch has been a consultant to Amgen, Bristol-Myers Squibb, and Caelum Biosciences.

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

 

– Recent trial results have shown the importance of treating patients with multiple myeloma with the immunomodulator lenalidomide for maintaining negative minimal residual disease and sustained complete responses following autologous stem cell transplantation, experts said at a conference held by Imedex.

“How do you keep the therapy pedal to the metal over time? Lenalidomide is easy to deliver, convenient, and improves progression-free survival and overall survival,” said Paul G. Richardson, MD, professor of medicine at Harvard University and clinical program leader of the Multiple Myeloma Center at the Dana-Farber Cancer Institute in Boston. “Lenalidomide maintenance is standard of care. It provides a platform on which you can reliably add new agents for maintenance of remission following autologous stem cell transplantation,” in patients with multiple myeloma.

Mitchel L. Zoler/Frontline Medical News
Dr. Paul G. Richardson
Dr. Richardson said that some of the best evidence for lenalidomide’s efficacy in posttransplant patients came in a 2012 report from a phase III trial that compared lenalidomide (Revlimid) maintenance with placebo in 460 patients following induction. The results showed that maintenance with 5-15 mg/day of lenalidomide reduced the rate of either progressive disease or death by 63% (hazard ratio, 0.37; 95% confidence interval, 0.26-0.53), compared with placebo, during a median follow-up of 18 months (N Engl J Med. 2012 May 10;366[19]:1770-81).

In a meta-analysis of three randomized, controlled trials with 1,208 patients with multiple myeloma who had undergone induction and autologous stem cell transplant, the overall mortality rate dropped by 0.75 (95% CI, 0.63-0.90) among patients maintained on lenalidomide, compared with placebo (J Clin Oncol. 2016;34:suppl;abstract 8001).

A third study that Dr. Richardson cited showed the difficulty of improving on lenalidomide. The BMT CTN 0702 (Stem Cell Transplant With Lenalidomide Maintenance in Patients With Multiple Myeloma) trial randomized 758 multiple myeloma patients to three different autologous stem cell transplant regimens, each followed by lenalidomide maintenance. One arm followed the transplant with four cycles of consolidation therapy with lenalidomide, dexamethasone, and bortezomib (Velcade); one arm used two tandem transplantations; and the third arm used a single transplantation. All three arms had similar rates of progression-free survival and overall survival during follow-up (Blood. 2016 Dec 6;LBA-1). The results showed that “lenalidomide maintenance is an equalizer,” Dr. Richardson said.

Dr. Suzanne Lentzsch
Mitchel L. Zoler/Frontline Medical News
Dr. Suzanne Lentzsch
While agreeing that lenalidomide is a key treatment following autologous stem cell transplant in patients with multiple myeloma, Suzanne Lentzsch, MD, highlighted recent, limited evidence that showed that the best outcomes occurred in posttransplant patients who achieved “deep and sustained,” complete responses on prolonged treatment with a combination consolidation regimen.

She cited a U.S., multicenter, phase II study that followed 66 newly diagnosed patients with multiple myeloma who proceeded through induction, autologous stem cell transplantation, and multiple cycles of consolidation therapy with a regimen of lenalidomide, the proteasome inhibitor carfilzomib (Kyprolis), and dexamethasone. Fifty patients went through 18 cycles of this consolidation regimen and showed an 84% rate of stringent complete response, “the first time we’ve seen such results,” said Dr. Lentzsch, director of the Multiple Myeloma and Amyloidosis Program at Columbia University in New York. “This translated into an excellent” progression-free survival of 86% after 3 years and an overall survival of 95% after 3 years (Blood. 2016 Dec 5;Abstract 675). “I’m pretty impressed” by the results, she added.

An advantage of the combined regimen used in this study is that it is “relatively well tolerated,” as well as effective for keeping patients in remission, Dr. Lentzsch said. However, she highlighted that this was a small study, so its treatment implications are limited for the time being. “For high-risk patients, use the full combination. For everyone else, we need to wait for results from a randomized, controlled trial,” she advised.

Dr. Richardson has been a consultant to Celgene, the company that markets lenalidomide, and is also a consultant to Genmab, Janssen, Novartis, Oncopeptides, and Takeda and has received research funding from Celgene and Takeda. Dr. Lentzsch has been a consultant to Amgen, Bristol-Myers Squibb, and Caelum Biosciences.

 

– Recent trial results have shown the importance of treating patients with multiple myeloma with the immunomodulator lenalidomide for maintaining negative minimal residual disease and sustained complete responses following autologous stem cell transplantation, experts said at a conference held by Imedex.

“How do you keep the therapy pedal to the metal over time? Lenalidomide is easy to deliver, convenient, and improves progression-free survival and overall survival,” said Paul G. Richardson, MD, professor of medicine at Harvard University and clinical program leader of the Multiple Myeloma Center at the Dana-Farber Cancer Institute in Boston. “Lenalidomide maintenance is standard of care. It provides a platform on which you can reliably add new agents for maintenance of remission following autologous stem cell transplantation,” in patients with multiple myeloma.

Mitchel L. Zoler/Frontline Medical News
Dr. Paul G. Richardson
Dr. Richardson said that some of the best evidence for lenalidomide’s efficacy in posttransplant patients came in a 2012 report from a phase III trial that compared lenalidomide (Revlimid) maintenance with placebo in 460 patients following induction. The results showed that maintenance with 5-15 mg/day of lenalidomide reduced the rate of either progressive disease or death by 63% (hazard ratio, 0.37; 95% confidence interval, 0.26-0.53), compared with placebo, during a median follow-up of 18 months (N Engl J Med. 2012 May 10;366[19]:1770-81).

In a meta-analysis of three randomized, controlled trials with 1,208 patients with multiple myeloma who had undergone induction and autologous stem cell transplant, the overall mortality rate dropped by 0.75 (95% CI, 0.63-0.90) among patients maintained on lenalidomide, compared with placebo (J Clin Oncol. 2016;34:suppl;abstract 8001).

A third study that Dr. Richardson cited showed the difficulty of improving on lenalidomide. The BMT CTN 0702 (Stem Cell Transplant With Lenalidomide Maintenance in Patients With Multiple Myeloma) trial randomized 758 multiple myeloma patients to three different autologous stem cell transplant regimens, each followed by lenalidomide maintenance. One arm followed the transplant with four cycles of consolidation therapy with lenalidomide, dexamethasone, and bortezomib (Velcade); one arm used two tandem transplantations; and the third arm used a single transplantation. All three arms had similar rates of progression-free survival and overall survival during follow-up (Blood. 2016 Dec 6;LBA-1). The results showed that “lenalidomide maintenance is an equalizer,” Dr. Richardson said.

Dr. Suzanne Lentzsch
Mitchel L. Zoler/Frontline Medical News
Dr. Suzanne Lentzsch
While agreeing that lenalidomide is a key treatment following autologous stem cell transplant in patients with multiple myeloma, Suzanne Lentzsch, MD, highlighted recent, limited evidence that showed that the best outcomes occurred in posttransplant patients who achieved “deep and sustained,” complete responses on prolonged treatment with a combination consolidation regimen.

She cited a U.S., multicenter, phase II study that followed 66 newly diagnosed patients with multiple myeloma who proceeded through induction, autologous stem cell transplantation, and multiple cycles of consolidation therapy with a regimen of lenalidomide, the proteasome inhibitor carfilzomib (Kyprolis), and dexamethasone. Fifty patients went through 18 cycles of this consolidation regimen and showed an 84% rate of stringent complete response, “the first time we’ve seen such results,” said Dr. Lentzsch, director of the Multiple Myeloma and Amyloidosis Program at Columbia University in New York. “This translated into an excellent” progression-free survival of 86% after 3 years and an overall survival of 95% after 3 years (Blood. 2016 Dec 5;Abstract 675). “I’m pretty impressed” by the results, she added.

An advantage of the combined regimen used in this study is that it is “relatively well tolerated,” as well as effective for keeping patients in remission, Dr. Lentzsch said. However, she highlighted that this was a small study, so its treatment implications are limited for the time being. “For high-risk patients, use the full combination. For everyone else, we need to wait for results from a randomized, controlled trial,” she advised.

Dr. Richardson has been a consultant to Celgene, the company that markets lenalidomide, and is also a consultant to Genmab, Janssen, Novartis, Oncopeptides, and Takeda and has received research funding from Celgene and Takeda. Dr. Lentzsch has been a consultant to Amgen, Bristol-Myers Squibb, and Caelum Biosciences.

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

Newly diagnosed mantle cell lymphoma is ‘one of the hardest consultations’

Article Type
Changed
Tue, 01/17/2023 - 11:25

 

– Relatively young patients with newly diagnosed, average-risk mantle cell lymphoma who go into remission on induction therapy face a difficult choice on their next management step: undergo immediate autologous stem cell transplantation or defer the stem cell transplant and continue on maintenance therapy.

The choice is especially difficult because both are currently considered reasonable options and each choice has certain attractions and downsides, experts highlighted in discussing this fork-in-the-road decision patients face.

Dr. Timothy S. Fenske, Medical College of Wisconsin
Mitchel L. Zoler/Frontline Medical News
Dr. Timothy S. Fenske


Immediate autologous stem cell transplantation (ASCT) has a good chance to allow the patient to remain treatment free and in remission for as long as about 10 years, but it involves intensive upfront treatment for 6-9 months, during which the patient will likely not be able to work or carry on many usual activities. Deferring the transplant with maintenance therapy puts off this life-disrupting initial period of intensive therapy for what may be several years, but relapse on maintenance therapy is inevitable and once it happens the patient may not have as successful an outcome from an ASCT. It also means several years of ongoing drug therapy with a maintenance regimen.

“I tell my fellows that patients with newly diagnosed mantle cell lymphoma [are] one of the hardest consultations because, unlike most other lymphomas, there is no established standard therapy but a range of options,” Timothy S. Fenske, MD, said at the conference held by Imedex. “I go through the pros and cons with patients, and it comes down to the patient’s perceived quality of life and their lifestyle.”

“It’s a very difficult decision [for patients] because we don’t have the data we’d like to have,” observed Peter Martin, MD, director of the clinical research program in lymphoma at Weill Cornell Medicine, New York.

“There is a lot of upfront toxicity with transplantation, with 6-9 months out of work in my experience. Patients often tell me that they can’t afford to do that; they’ll lose their employment insurance and won’t be able to pay for replacement insurance. But then they will hopefully go 6-10 years without more treatment, which is a real benefit. With less intensive upfront treatment they have a chance for similar overall survival, but they’ll need more ongoing treatment. It’s pretty complicated and challenging” for patients to make a decision, he said. “It depends a lot on where patients are in their lives and what they are willing to accept,” Dr. Martin said.

In general, Dr. Martin took a more skeptical view of ASCT than Dr. Fenske. “There is no evidence that ASCT cures patients or prolongs their survival. It improves progression-free survival, but not necessarily overall survival,” Dr. Martin noted.

In fact, a report in December 2016 at the American Society of Hematology annual meeting (Blood. 2016 Dec 5;abstract 1095) suggested that “biology is the primary driver of outcomes in mantle cell lymphoma, not treatment,” said Dr. Martin. The results from a limited number of patients enrolled in the Nordic mantle cell lymphoma trials provided good but preliminary evidence that “if you have good biology it doesn’t matter what the treatment is, you will do well,” he explained.

Dr. Peter Martin of Weill Cornell Medicine
Mitchel L. Zoler/Frontline Medical News
Dr. Peter Martin


In fact, a report in December 2016 at the American Society of Hematology annual meeting (Blood. 2016 Dec 5;abstract 1095) suggested that “biology is the primary driver of outcomes in mantle cell lymphoma, not treatment,” said Dr. Martin. The results from a limited number of patients enrolled in the Nordic mantle cell lymphoma trials provided good but preliminary evidence that “less intense therapy works just as well” as more intense therapy, as long as the patient has a favorable genetic profile, he explained.

In contrast, Dr. Fenske put a much more positive spin on more intensive treatment upfront with ASCT.

“There is not much debate that you get longer progression-free survival with the more intensive approach. The question is, does progression-free survival matter in mantle cell lymphoma? I argue that it does because relapse in patients with mantle cell lymphoma is no picnic. What you can expect in patients with relapsed or refractory mantle cell lymphoma is a progression-free survival of about 1-2 years, and an overall survival of about 2-3 years,” said Dr. Fenske, head of the section of bone marrow transplant and hematologic malignancies at the Medical College of Wisconsin in Milwaukee.


As an example of the poor prognosis of relapsed or refractory mantle cell lymphoma patients Dr. Fenske cited a review he coauthored of 97 patients treated with ibrutinib (Imbruvica. Their median duration of response was 17 months and median progression-free survival was 15 months. Once ibrutinib treatment failure occurred their median overall survival was less than 3 months. (Hematol Oncol. 2017 Jan 8.doi:10.1002/hon.2380).

“It’s easy to get carried away” when patients temporarily respond to a drug like ibrutinib or other new agents with a degree of efficacy for lymphomas, Dr. Fenske said, but these transient responses “don’t solve the problem. The patient is headed for trouble,” usually within a couple of years.

“I would argue that, especially for younger patients, the goal is to try to achieve the longest first remission, and that means an ASCT.” Dr. Fenske admitted that this strategy won’t work for very-high-risk patients, but for these patients no good treatment options currently exist.

He also stressed that research is just beginning to explore using measurement of negative minimal residual disease to identify patients with the best outcomes following initial induction treatment. It is possible that patients with undetectable minimal residual disease can avoid immediate ASCT and instead receive maintenance therapy, a hypothesis slated for testing in a randomized trial, he said.

Dr. Martin has been a consultant to Celgene, Gilead, Janssen, Novartis, Pharmacyclics, and Verastem. Dr. Fenske has been a consultant to Abbvie, Celgene, Pharmacyclics, Sanofi, and Seattle Genetics.

 

 

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

 

– Relatively young patients with newly diagnosed, average-risk mantle cell lymphoma who go into remission on induction therapy face a difficult choice on their next management step: undergo immediate autologous stem cell transplantation or defer the stem cell transplant and continue on maintenance therapy.

The choice is especially difficult because both are currently considered reasonable options and each choice has certain attractions and downsides, experts highlighted in discussing this fork-in-the-road decision patients face.

Dr. Timothy S. Fenske, Medical College of Wisconsin
Mitchel L. Zoler/Frontline Medical News
Dr. Timothy S. Fenske


Immediate autologous stem cell transplantation (ASCT) has a good chance to allow the patient to remain treatment free and in remission for as long as about 10 years, but it involves intensive upfront treatment for 6-9 months, during which the patient will likely not be able to work or carry on many usual activities. Deferring the transplant with maintenance therapy puts off this life-disrupting initial period of intensive therapy for what may be several years, but relapse on maintenance therapy is inevitable and once it happens the patient may not have as successful an outcome from an ASCT. It also means several years of ongoing drug therapy with a maintenance regimen.

“I tell my fellows that patients with newly diagnosed mantle cell lymphoma [are] one of the hardest consultations because, unlike most other lymphomas, there is no established standard therapy but a range of options,” Timothy S. Fenske, MD, said at the conference held by Imedex. “I go through the pros and cons with patients, and it comes down to the patient’s perceived quality of life and their lifestyle.”

“It’s a very difficult decision [for patients] because we don’t have the data we’d like to have,” observed Peter Martin, MD, director of the clinical research program in lymphoma at Weill Cornell Medicine, New York.

“There is a lot of upfront toxicity with transplantation, with 6-9 months out of work in my experience. Patients often tell me that they can’t afford to do that; they’ll lose their employment insurance and won’t be able to pay for replacement insurance. But then they will hopefully go 6-10 years without more treatment, which is a real benefit. With less intensive upfront treatment they have a chance for similar overall survival, but they’ll need more ongoing treatment. It’s pretty complicated and challenging” for patients to make a decision, he said. “It depends a lot on where patients are in their lives and what they are willing to accept,” Dr. Martin said.

In general, Dr. Martin took a more skeptical view of ASCT than Dr. Fenske. “There is no evidence that ASCT cures patients or prolongs their survival. It improves progression-free survival, but not necessarily overall survival,” Dr. Martin noted.

In fact, a report in December 2016 at the American Society of Hematology annual meeting (Blood. 2016 Dec 5;abstract 1095) suggested that “biology is the primary driver of outcomes in mantle cell lymphoma, not treatment,” said Dr. Martin. The results from a limited number of patients enrolled in the Nordic mantle cell lymphoma trials provided good but preliminary evidence that “if you have good biology it doesn’t matter what the treatment is, you will do well,” he explained.

Dr. Peter Martin of Weill Cornell Medicine
Mitchel L. Zoler/Frontline Medical News
Dr. Peter Martin


In fact, a report in December 2016 at the American Society of Hematology annual meeting (Blood. 2016 Dec 5;abstract 1095) suggested that “biology is the primary driver of outcomes in mantle cell lymphoma, not treatment,” said Dr. Martin. The results from a limited number of patients enrolled in the Nordic mantle cell lymphoma trials provided good but preliminary evidence that “less intense therapy works just as well” as more intense therapy, as long as the patient has a favorable genetic profile, he explained.

In contrast, Dr. Fenske put a much more positive spin on more intensive treatment upfront with ASCT.

“There is not much debate that you get longer progression-free survival with the more intensive approach. The question is, does progression-free survival matter in mantle cell lymphoma? I argue that it does because relapse in patients with mantle cell lymphoma is no picnic. What you can expect in patients with relapsed or refractory mantle cell lymphoma is a progression-free survival of about 1-2 years, and an overall survival of about 2-3 years,” said Dr. Fenske, head of the section of bone marrow transplant and hematologic malignancies at the Medical College of Wisconsin in Milwaukee.


As an example of the poor prognosis of relapsed or refractory mantle cell lymphoma patients Dr. Fenske cited a review he coauthored of 97 patients treated with ibrutinib (Imbruvica. Their median duration of response was 17 months and median progression-free survival was 15 months. Once ibrutinib treatment failure occurred their median overall survival was less than 3 months. (Hematol Oncol. 2017 Jan 8.doi:10.1002/hon.2380).

“It’s easy to get carried away” when patients temporarily respond to a drug like ibrutinib or other new agents with a degree of efficacy for lymphomas, Dr. Fenske said, but these transient responses “don’t solve the problem. The patient is headed for trouble,” usually within a couple of years.

“I would argue that, especially for younger patients, the goal is to try to achieve the longest first remission, and that means an ASCT.” Dr. Fenske admitted that this strategy won’t work for very-high-risk patients, but for these patients no good treatment options currently exist.

He also stressed that research is just beginning to explore using measurement of negative minimal residual disease to identify patients with the best outcomes following initial induction treatment. It is possible that patients with undetectable minimal residual disease can avoid immediate ASCT and instead receive maintenance therapy, a hypothesis slated for testing in a randomized trial, he said.

Dr. Martin has been a consultant to Celgene, Gilead, Janssen, Novartis, Pharmacyclics, and Verastem. Dr. Fenske has been a consultant to Abbvie, Celgene, Pharmacyclics, Sanofi, and Seattle Genetics.

 

 

 

– Relatively young patients with newly diagnosed, average-risk mantle cell lymphoma who go into remission on induction therapy face a difficult choice on their next management step: undergo immediate autologous stem cell transplantation or defer the stem cell transplant and continue on maintenance therapy.

The choice is especially difficult because both are currently considered reasonable options and each choice has certain attractions and downsides, experts highlighted in discussing this fork-in-the-road decision patients face.

Dr. Timothy S. Fenske, Medical College of Wisconsin
Mitchel L. Zoler/Frontline Medical News
Dr. Timothy S. Fenske


Immediate autologous stem cell transplantation (ASCT) has a good chance to allow the patient to remain treatment free and in remission for as long as about 10 years, but it involves intensive upfront treatment for 6-9 months, during which the patient will likely not be able to work or carry on many usual activities. Deferring the transplant with maintenance therapy puts off this life-disrupting initial period of intensive therapy for what may be several years, but relapse on maintenance therapy is inevitable and once it happens the patient may not have as successful an outcome from an ASCT. It also means several years of ongoing drug therapy with a maintenance regimen.

“I tell my fellows that patients with newly diagnosed mantle cell lymphoma [are] one of the hardest consultations because, unlike most other lymphomas, there is no established standard therapy but a range of options,” Timothy S. Fenske, MD, said at the conference held by Imedex. “I go through the pros and cons with patients, and it comes down to the patient’s perceived quality of life and their lifestyle.”

“It’s a very difficult decision [for patients] because we don’t have the data we’d like to have,” observed Peter Martin, MD, director of the clinical research program in lymphoma at Weill Cornell Medicine, New York.

“There is a lot of upfront toxicity with transplantation, with 6-9 months out of work in my experience. Patients often tell me that they can’t afford to do that; they’ll lose their employment insurance and won’t be able to pay for replacement insurance. But then they will hopefully go 6-10 years without more treatment, which is a real benefit. With less intensive upfront treatment they have a chance for similar overall survival, but they’ll need more ongoing treatment. It’s pretty complicated and challenging” for patients to make a decision, he said. “It depends a lot on where patients are in their lives and what they are willing to accept,” Dr. Martin said.

In general, Dr. Martin took a more skeptical view of ASCT than Dr. Fenske. “There is no evidence that ASCT cures patients or prolongs their survival. It improves progression-free survival, but not necessarily overall survival,” Dr. Martin noted.

In fact, a report in December 2016 at the American Society of Hematology annual meeting (Blood. 2016 Dec 5;abstract 1095) suggested that “biology is the primary driver of outcomes in mantle cell lymphoma, not treatment,” said Dr. Martin. The results from a limited number of patients enrolled in the Nordic mantle cell lymphoma trials provided good but preliminary evidence that “if you have good biology it doesn’t matter what the treatment is, you will do well,” he explained.

Dr. Peter Martin of Weill Cornell Medicine
Mitchel L. Zoler/Frontline Medical News
Dr. Peter Martin


In fact, a report in December 2016 at the American Society of Hematology annual meeting (Blood. 2016 Dec 5;abstract 1095) suggested that “biology is the primary driver of outcomes in mantle cell lymphoma, not treatment,” said Dr. Martin. The results from a limited number of patients enrolled in the Nordic mantle cell lymphoma trials provided good but preliminary evidence that “less intense therapy works just as well” as more intense therapy, as long as the patient has a favorable genetic profile, he explained.

In contrast, Dr. Fenske put a much more positive spin on more intensive treatment upfront with ASCT.

“There is not much debate that you get longer progression-free survival with the more intensive approach. The question is, does progression-free survival matter in mantle cell lymphoma? I argue that it does because relapse in patients with mantle cell lymphoma is no picnic. What you can expect in patients with relapsed or refractory mantle cell lymphoma is a progression-free survival of about 1-2 years, and an overall survival of about 2-3 years,” said Dr. Fenske, head of the section of bone marrow transplant and hematologic malignancies at the Medical College of Wisconsin in Milwaukee.


As an example of the poor prognosis of relapsed or refractory mantle cell lymphoma patients Dr. Fenske cited a review he coauthored of 97 patients treated with ibrutinib (Imbruvica. Their median duration of response was 17 months and median progression-free survival was 15 months. Once ibrutinib treatment failure occurred their median overall survival was less than 3 months. (Hematol Oncol. 2017 Jan 8.doi:10.1002/hon.2380).

“It’s easy to get carried away” when patients temporarily respond to a drug like ibrutinib or other new agents with a degree of efficacy for lymphomas, Dr. Fenske said, but these transient responses “don’t solve the problem. The patient is headed for trouble,” usually within a couple of years.

“I would argue that, especially for younger patients, the goal is to try to achieve the longest first remission, and that means an ASCT.” Dr. Fenske admitted that this strategy won’t work for very-high-risk patients, but for these patients no good treatment options currently exist.

He also stressed that research is just beginning to explore using measurement of negative minimal residual disease to identify patients with the best outcomes following initial induction treatment. It is possible that patients with undetectable minimal residual disease can avoid immediate ASCT and instead receive maintenance therapy, a hypothesis slated for testing in a randomized trial, he said.

Dr. Martin has been a consultant to Celgene, Gilead, Janssen, Novartis, Pharmacyclics, and Verastem. Dr. Fenske has been a consultant to Abbvie, Celgene, Pharmacyclics, Sanofi, and Seattle Genetics.

 

 

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

EXPERT ANALYSIS FROM A MEETING ON HEMATOLOGIC MALIGNANCIES

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

Thousands expected to get CAR T-cells by 2018

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

 

– Should the first chimeric antigen receptor (CAR) T-cell treatment receive Food and Drug Administration approval for refractory, aggressive non-Hodgkin lymphoma, possibly before the end of 2017, several thousand U.S. patients will be potential candidates for the treatment, Jeremy S. Abramson, MD, predicted at a conference held by Imedex.

Dr. Abramson has led studies using a CAR T cell that differs from the one furthest along in development. He based his predicted timetable for an approved biologics license of the process, with which he can engineer patient-specific T cells that are under development by Kite Pharma, on the “remarkable” level of complete responses the intervention produced in a pivotal phase II study.

Mitchel L. Zoler/Frontline Medical News
Dr. Jeremy S. Abramson
Results from the ZUMA-1 study, first reported in December 2016 at the American Society of Hematology annual meeting, showed superior responses in 101 patients who received infusions of the modified and expanded CAR T cells, a process that Kite calls axicabtagene ciloleucel. Comparatively, the existing chemotherapy treatment results for the types of non-Hodgkin lymphoma patients who were enrolled in the trial were bleak. Of the treated patients, 77 had a diffuse large B-cell lymphoma, and 24 had primary mediastinal B-cell lymphoma or transformed follicular lymphoma.

“Seeing a complete response rate of close to 40% that is sustained for more than 3 months and a complete response rate of close to a third at 6 months is light years beyond what is now available for patients” with these chemotherapy refractory B-cell lymphomas, said Dr. Abramson, clinical director of the Center for Lymphoma at Massachusetts General Hospital in Boston. “This is the first treatment to show a significant impact on large B-cell lymphoma, and that makes me optimistic” that the FDA will grant license approval later in 2017, he said in an interview.

Dr. Abramson acknowledged that some patients developed grade 3 or 4 cytokine-release syndrome and neurologic events, but the events were reversible and manageable if treated with the immunosuppressant tocilizumab (Actemra) or a corticosteroid.

At the end of March, Kite reported the completion of its FDA application, and, on April 2, the ZUMA-1 investigators presented an updated report on their results, with a 36% complete response rate across all enrolled patients at 6 month follow-up and a 39% complete response rate in all patients out to a median follow-up of 8.7 months.

Annually, in U.S. practice, perhaps 3,000 new patients with diffuse large B-cell lymphoma would meet the enrollment criteria for ZUMA-1, Dr. Abramson estimated. Once axicabtagene ciloleucel becomes commercially available in the United States, several thousand U.S. patients might initially seek the treatment.
 

Dr. Abramson has been a consultant to Kite Pharma and to AbbVie, Genentech, Gilead, and Seattle Genetics.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event
Related Articles

 

– Should the first chimeric antigen receptor (CAR) T-cell treatment receive Food and Drug Administration approval for refractory, aggressive non-Hodgkin lymphoma, possibly before the end of 2017, several thousand U.S. patients will be potential candidates for the treatment, Jeremy S. Abramson, MD, predicted at a conference held by Imedex.

Dr. Abramson has led studies using a CAR T cell that differs from the one furthest along in development. He based his predicted timetable for an approved biologics license of the process, with which he can engineer patient-specific T cells that are under development by Kite Pharma, on the “remarkable” level of complete responses the intervention produced in a pivotal phase II study.

Mitchel L. Zoler/Frontline Medical News
Dr. Jeremy S. Abramson
Results from the ZUMA-1 study, first reported in December 2016 at the American Society of Hematology annual meeting, showed superior responses in 101 patients who received infusions of the modified and expanded CAR T cells, a process that Kite calls axicabtagene ciloleucel. Comparatively, the existing chemotherapy treatment results for the types of non-Hodgkin lymphoma patients who were enrolled in the trial were bleak. Of the treated patients, 77 had a diffuse large B-cell lymphoma, and 24 had primary mediastinal B-cell lymphoma or transformed follicular lymphoma.

“Seeing a complete response rate of close to 40% that is sustained for more than 3 months and a complete response rate of close to a third at 6 months is light years beyond what is now available for patients” with these chemotherapy refractory B-cell lymphomas, said Dr. Abramson, clinical director of the Center for Lymphoma at Massachusetts General Hospital in Boston. “This is the first treatment to show a significant impact on large B-cell lymphoma, and that makes me optimistic” that the FDA will grant license approval later in 2017, he said in an interview.

Dr. Abramson acknowledged that some patients developed grade 3 or 4 cytokine-release syndrome and neurologic events, but the events were reversible and manageable if treated with the immunosuppressant tocilizumab (Actemra) or a corticosteroid.

At the end of March, Kite reported the completion of its FDA application, and, on April 2, the ZUMA-1 investigators presented an updated report on their results, with a 36% complete response rate across all enrolled patients at 6 month follow-up and a 39% complete response rate in all patients out to a median follow-up of 8.7 months.

Annually, in U.S. practice, perhaps 3,000 new patients with diffuse large B-cell lymphoma would meet the enrollment criteria for ZUMA-1, Dr. Abramson estimated. Once axicabtagene ciloleucel becomes commercially available in the United States, several thousand U.S. patients might initially seek the treatment.
 

Dr. Abramson has been a consultant to Kite Pharma and to AbbVie, Genentech, Gilead, and Seattle Genetics.

 

– Should the first chimeric antigen receptor (CAR) T-cell treatment receive Food and Drug Administration approval for refractory, aggressive non-Hodgkin lymphoma, possibly before the end of 2017, several thousand U.S. patients will be potential candidates for the treatment, Jeremy S. Abramson, MD, predicted at a conference held by Imedex.

Dr. Abramson has led studies using a CAR T cell that differs from the one furthest along in development. He based his predicted timetable for an approved biologics license of the process, with which he can engineer patient-specific T cells that are under development by Kite Pharma, on the “remarkable” level of complete responses the intervention produced in a pivotal phase II study.

Mitchel L. Zoler/Frontline Medical News
Dr. Jeremy S. Abramson
Results from the ZUMA-1 study, first reported in December 2016 at the American Society of Hematology annual meeting, showed superior responses in 101 patients who received infusions of the modified and expanded CAR T cells, a process that Kite calls axicabtagene ciloleucel. Comparatively, the existing chemotherapy treatment results for the types of non-Hodgkin lymphoma patients who were enrolled in the trial were bleak. Of the treated patients, 77 had a diffuse large B-cell lymphoma, and 24 had primary mediastinal B-cell lymphoma or transformed follicular lymphoma.

“Seeing a complete response rate of close to 40% that is sustained for more than 3 months and a complete response rate of close to a third at 6 months is light years beyond what is now available for patients” with these chemotherapy refractory B-cell lymphomas, said Dr. Abramson, clinical director of the Center for Lymphoma at Massachusetts General Hospital in Boston. “This is the first treatment to show a significant impact on large B-cell lymphoma, and that makes me optimistic” that the FDA will grant license approval later in 2017, he said in an interview.

Dr. Abramson acknowledged that some patients developed grade 3 or 4 cytokine-release syndrome and neurologic events, but the events were reversible and manageable if treated with the immunosuppressant tocilizumab (Actemra) or a corticosteroid.

At the end of March, Kite reported the completion of its FDA application, and, on April 2, the ZUMA-1 investigators presented an updated report on their results, with a 36% complete response rate across all enrolled patients at 6 month follow-up and a 39% complete response rate in all patients out to a median follow-up of 8.7 months.

Annually, in U.S. practice, perhaps 3,000 new patients with diffuse large B-cell lymphoma would meet the enrollment criteria for ZUMA-1, Dr. Abramson estimated. Once axicabtagene ciloleucel becomes commercially available in the United States, several thousand U.S. patients might initially seek the treatment.
 

Dr. Abramson has been a consultant to Kite Pharma and to AbbVie, Genentech, Gilead, and Seattle Genetics.

Publications
Publications
Topics
Article Type
Sections
Article Source

EXPERT ANALYSIS FROM A MEETING ON HEMATOLOGIC MALIGNANCIES

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

Obinutuzumab vs. rituximab weighed as follicular lymphoma therapy

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

 

– Does obinutuzumab have a leg up over rituximab for treating follicular lymphoma?

A strict reading of the efficacy records of the two anti-CD20 antibodies when they went head-to-head suggests that obinutuzumab (Gazyva) edged out rituximab (Rituxan), but a broader view leaves the door open for rituximab as a still viable option depending on a patient’s status and priorities, experts said at the conference held by Imedex.

Dr. John P. Leonard
Dr. John P. Leonard
For the roughly 80% of patients with follicular lymphoma (indolent non-Hodgkin lymphoma) who die with their disease rather than from it, management needs to focus on quality of life issues as much as on efficacy, said John P. Leonard, MD, professor of medicine at Weill Cornell Medicine, New York.

While conceding that quality of life correlates with progression-free survival (PFS), he stressed that it also correlates with treatment toxicities, treatment duration, and disease-related side effects.

Trial results have indicated that patients with newly diagnosed follicular lymphoma are reasonably treated with rituximab alone, or with rituximab plus bendamustine, without need for maintenance therapy, Dr. Leonard said.

In contrast, GALLIUM, a phase III trial that compared rituximab against obinutuzumab, used a maintenance phase of monotherapy with each of these two drugs following an induction phase when each of the drugs was combined with chemotherapy.

“If you use this approach [tested in GALLIUM] you need to use maintenance therapy,” and it was in GALLIUM that the most dramatic efficacy advantage for obinutuzumab over rituximab appeared, in the form of longer PFS although, so far, without demonstrated advantage in overall survival. The GALLIUM results, reported in December 2016 at the American Society of Hematology meeting, showed a 3-year PFS rate of 80% among patients treated with obinutuzumab and 73% among those treated with rituximab, a hazard ratio of 0.66 in favor of obinutuzumab that was statistically significant (P = .001) for the study’s primary endpoint (Blood. 2016 Dec 4;abstract 6).

“If you follow this study, you commit the patient to maintenance. We need to talk with patients about the pros and cons of maintenance, the pros and cons of chemotherapy, and the pros and cons of single agent therapy” with one of these anti-CD20 antibodies, Dr. Leonard said. “Right now, I think it’s unclear which antibody is best,” he concluded

Dr. Jonathan W. Friedberg
Dr. Jonathan W. Friedberg
On the other hand, for Jonathan W. Friedberg, MD, the PFS advantage from obinutuzumab in GALLIUM trumps other concerns. “Based upon the GALLIUM data for symptomatic, aggressive follicular lymphoma obinutuzumab plus chemotherapy is the best option to achieve prolonged PFS,” said Dr. Friedberg, professor of medicine and director of the Wilmot Cancer Institute at the University of Rochester (N.Y.).

To further buttress the case for obinutuzumab, he also cited the higher response rate among relapsed patients when single-agent obinutuzumab went against single-agent rituximab (J Clin Oncol. 2015 Oct 20:33[30]:3467-74), and the overall survival advantage that obinutuzumab gave patients when combined with bendamustine in patients refractory to rituximab (Blood. 2016 Dec 5; abstract 615).

Agreeing that the design of GALLIUM focused on combining an anti-CD20 antibody with chemotherapy, Dr. Friedberg acknowledged that, as initial therapy, “using rituximab monotherapy is very reasonable for many patients. I divide [follicular lymphoma] patients into those who are very symptomatic” (for example, those with hydronephrosis) “and need chemotherapy, and those who are not that symptomatic for whom single-agent rituximab is very reasonable,” he said in an interview.

Tumor aggressiveness is another way to identify patients who need chemotherapy plus an antibody, he added. “If the patient is not symptomatic, I generally first observe them, and if the growth is slow, you can sometimes intervene with rituximab alone, but, if the growth is fast, you also need chemotherapy,” Dr. Friedberg said.

Cost may soon become another consideration now that the U.S. patent on rituximab has expired leading to the ongoing development of several biosimilar versions of the antibody. If biosimilar formulations of rituximab soon appear on the U.S. market and if they result in a significant drop in drug price, it would introduce yet another significant variable. “Presuming biosimilar rituximab lowers the cost, that would be another important treatment decision,” he said.

Dr. Friedberg has been a consultant to Bayer. Dr. Leonard has been a consultant to 13 drug companies. Neither disclosed a relationship with the companies that market obinutuzumab or rituximab.
 

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event
Related Articles

 

– Does obinutuzumab have a leg up over rituximab for treating follicular lymphoma?

A strict reading of the efficacy records of the two anti-CD20 antibodies when they went head-to-head suggests that obinutuzumab (Gazyva) edged out rituximab (Rituxan), but a broader view leaves the door open for rituximab as a still viable option depending on a patient’s status and priorities, experts said at the conference held by Imedex.

Dr. John P. Leonard
Dr. John P. Leonard
For the roughly 80% of patients with follicular lymphoma (indolent non-Hodgkin lymphoma) who die with their disease rather than from it, management needs to focus on quality of life issues as much as on efficacy, said John P. Leonard, MD, professor of medicine at Weill Cornell Medicine, New York.

While conceding that quality of life correlates with progression-free survival (PFS), he stressed that it also correlates with treatment toxicities, treatment duration, and disease-related side effects.

Trial results have indicated that patients with newly diagnosed follicular lymphoma are reasonably treated with rituximab alone, or with rituximab plus bendamustine, without need for maintenance therapy, Dr. Leonard said.

In contrast, GALLIUM, a phase III trial that compared rituximab against obinutuzumab, used a maintenance phase of monotherapy with each of these two drugs following an induction phase when each of the drugs was combined with chemotherapy.

“If you use this approach [tested in GALLIUM] you need to use maintenance therapy,” and it was in GALLIUM that the most dramatic efficacy advantage for obinutuzumab over rituximab appeared, in the form of longer PFS although, so far, without demonstrated advantage in overall survival. The GALLIUM results, reported in December 2016 at the American Society of Hematology meeting, showed a 3-year PFS rate of 80% among patients treated with obinutuzumab and 73% among those treated with rituximab, a hazard ratio of 0.66 in favor of obinutuzumab that was statistically significant (P = .001) for the study’s primary endpoint (Blood. 2016 Dec 4;abstract 6).

“If you follow this study, you commit the patient to maintenance. We need to talk with patients about the pros and cons of maintenance, the pros and cons of chemotherapy, and the pros and cons of single agent therapy” with one of these anti-CD20 antibodies, Dr. Leonard said. “Right now, I think it’s unclear which antibody is best,” he concluded

Dr. Jonathan W. Friedberg
Dr. Jonathan W. Friedberg
On the other hand, for Jonathan W. Friedberg, MD, the PFS advantage from obinutuzumab in GALLIUM trumps other concerns. “Based upon the GALLIUM data for symptomatic, aggressive follicular lymphoma obinutuzumab plus chemotherapy is the best option to achieve prolonged PFS,” said Dr. Friedberg, professor of medicine and director of the Wilmot Cancer Institute at the University of Rochester (N.Y.).

To further buttress the case for obinutuzumab, he also cited the higher response rate among relapsed patients when single-agent obinutuzumab went against single-agent rituximab (J Clin Oncol. 2015 Oct 20:33[30]:3467-74), and the overall survival advantage that obinutuzumab gave patients when combined with bendamustine in patients refractory to rituximab (Blood. 2016 Dec 5; abstract 615).

Agreeing that the design of GALLIUM focused on combining an anti-CD20 antibody with chemotherapy, Dr. Friedberg acknowledged that, as initial therapy, “using rituximab monotherapy is very reasonable for many patients. I divide [follicular lymphoma] patients into those who are very symptomatic” (for example, those with hydronephrosis) “and need chemotherapy, and those who are not that symptomatic for whom single-agent rituximab is very reasonable,” he said in an interview.

Tumor aggressiveness is another way to identify patients who need chemotherapy plus an antibody, he added. “If the patient is not symptomatic, I generally first observe them, and if the growth is slow, you can sometimes intervene with rituximab alone, but, if the growth is fast, you also need chemotherapy,” Dr. Friedberg said.

Cost may soon become another consideration now that the U.S. patent on rituximab has expired leading to the ongoing development of several biosimilar versions of the antibody. If biosimilar formulations of rituximab soon appear on the U.S. market and if they result in a significant drop in drug price, it would introduce yet another significant variable. “Presuming biosimilar rituximab lowers the cost, that would be another important treatment decision,” he said.

Dr. Friedberg has been a consultant to Bayer. Dr. Leonard has been a consultant to 13 drug companies. Neither disclosed a relationship with the companies that market obinutuzumab or rituximab.
 

 

– Does obinutuzumab have a leg up over rituximab for treating follicular lymphoma?

A strict reading of the efficacy records of the two anti-CD20 antibodies when they went head-to-head suggests that obinutuzumab (Gazyva) edged out rituximab (Rituxan), but a broader view leaves the door open for rituximab as a still viable option depending on a patient’s status and priorities, experts said at the conference held by Imedex.

Dr. John P. Leonard
Dr. John P. Leonard
For the roughly 80% of patients with follicular lymphoma (indolent non-Hodgkin lymphoma) who die with their disease rather than from it, management needs to focus on quality of life issues as much as on efficacy, said John P. Leonard, MD, professor of medicine at Weill Cornell Medicine, New York.

While conceding that quality of life correlates with progression-free survival (PFS), he stressed that it also correlates with treatment toxicities, treatment duration, and disease-related side effects.

Trial results have indicated that patients with newly diagnosed follicular lymphoma are reasonably treated with rituximab alone, or with rituximab plus bendamustine, without need for maintenance therapy, Dr. Leonard said.

In contrast, GALLIUM, a phase III trial that compared rituximab against obinutuzumab, used a maintenance phase of monotherapy with each of these two drugs following an induction phase when each of the drugs was combined with chemotherapy.

“If you use this approach [tested in GALLIUM] you need to use maintenance therapy,” and it was in GALLIUM that the most dramatic efficacy advantage for obinutuzumab over rituximab appeared, in the form of longer PFS although, so far, without demonstrated advantage in overall survival. The GALLIUM results, reported in December 2016 at the American Society of Hematology meeting, showed a 3-year PFS rate of 80% among patients treated with obinutuzumab and 73% among those treated with rituximab, a hazard ratio of 0.66 in favor of obinutuzumab that was statistically significant (P = .001) for the study’s primary endpoint (Blood. 2016 Dec 4;abstract 6).

“If you follow this study, you commit the patient to maintenance. We need to talk with patients about the pros and cons of maintenance, the pros and cons of chemotherapy, and the pros and cons of single agent therapy” with one of these anti-CD20 antibodies, Dr. Leonard said. “Right now, I think it’s unclear which antibody is best,” he concluded

Dr. Jonathan W. Friedberg
Dr. Jonathan W. Friedberg
On the other hand, for Jonathan W. Friedberg, MD, the PFS advantage from obinutuzumab in GALLIUM trumps other concerns. “Based upon the GALLIUM data for symptomatic, aggressive follicular lymphoma obinutuzumab plus chemotherapy is the best option to achieve prolonged PFS,” said Dr. Friedberg, professor of medicine and director of the Wilmot Cancer Institute at the University of Rochester (N.Y.).

To further buttress the case for obinutuzumab, he also cited the higher response rate among relapsed patients when single-agent obinutuzumab went against single-agent rituximab (J Clin Oncol. 2015 Oct 20:33[30]:3467-74), and the overall survival advantage that obinutuzumab gave patients when combined with bendamustine in patients refractory to rituximab (Blood. 2016 Dec 5; abstract 615).

Agreeing that the design of GALLIUM focused on combining an anti-CD20 antibody with chemotherapy, Dr. Friedberg acknowledged that, as initial therapy, “using rituximab monotherapy is very reasonable for many patients. I divide [follicular lymphoma] patients into those who are very symptomatic” (for example, those with hydronephrosis) “and need chemotherapy, and those who are not that symptomatic for whom single-agent rituximab is very reasonable,” he said in an interview.

Tumor aggressiveness is another way to identify patients who need chemotherapy plus an antibody, he added. “If the patient is not symptomatic, I generally first observe them, and if the growth is slow, you can sometimes intervene with rituximab alone, but, if the growth is fast, you also need chemotherapy,” Dr. Friedberg said.

Cost may soon become another consideration now that the U.S. patent on rituximab has expired leading to the ongoing development of several biosimilar versions of the antibody. If biosimilar formulations of rituximab soon appear on the U.S. market and if they result in a significant drop in drug price, it would introduce yet another significant variable. “Presuming biosimilar rituximab lowers the cost, that would be another important treatment decision,” he said.

Dr. Friedberg has been a consultant to Bayer. Dr. Leonard has been a consultant to 13 drug companies. Neither disclosed a relationship with the companies that market obinutuzumab or rituximab.
 

Publications
Publications
Topics
Article Type
Sections
Article Source

EXPERT ANALYSIS FROM A MEETING ON HEMATOLOGIC MALIGNANCIES

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

Expanded drug combinations produce best myeloma induction

Article Type
Changed
Tue, 07/21/2020 - 14:18

 

– Optimal induction therapy for patients with multiple myeloma requires all the therapeutic tools currently available for combination therapy, which means using four agents followed by autologous stem cell transplantation, Paul G. Richardson, MD, said at a conference held by Imedex.

He suggested that a rational combination regimen for induction therapy of multiple myeloma would include a second- or third-generation immunomodulator such as lenalidomide (Revlimid) or pomalidomide (Pomalyst); a proteasome inhibitor such as bortezomib (Velcade), carfilzomib (Kyprolis), or ixazomib (Ninlaro); plus standard dexamethasone to form a contemporary backbone regimen for inducing remission in patients with multiple myeloma.

Mitchel L. Zoler/Frontline Medical News
Dr. Paul G. Richardson (left) and Dr. Ruben Niesvizky
Dr. Richardson advocated for the addition of an anti-CD38 monoclonal antibody such as daratumumab (Darzalex), as well as follow-up management with an autologous stem cell transplant, a total package of treatments that has not yet been tested in a reported trial.

“Therapeutic parsimony is not recommended. You need a combination” of all available drug classes, suggested Dr. Richardson, professor of medicine at Harvard Medical School and clinical program leader of the Multiple Myeloma Center at the Dana-Farber Cancer Institute in Boston.

“It isn’t rational to limit the treatment choices. We need to bring them all together,” he said. The “most rational” combination melds an immunomodulator, proteasome inhibitor, plus a monoclonal antibody that he called a “true game changer. Add the antibody on top of the three-drug platform” of an immunomodulator, proteasome inhibitor, and dexamethasone. Dr. Richardson conceded, however, that a concern with such combinations is how to manage the toxicity that would likely result.

He cited several recent examples of demonstrated superior efficacy in the form of more complete responses from combined regimens, followed by autologous stem cell transplantation.

For example, a recent report from a French-led group compared the efficacy of a combination of an immunomodulator, proteasome inhibitor, and dexamethasone against the same combination, followed by autologous stem cell transplantation (N Engl J Med. 2017 Apr 6;376[14]:1311-20). The combined regimen plus transplant resulted in a 59% complete response rate, “the best response rate we’ve ever seen” in multiple myeloma, Dr. Richardson noted, compared with a 48% complete response rate in patients who did not undergo a stem cell transplant.

Another speaker at the meeting, Ruben Niesvizky, MD, suggested a more cautious approach to using the monoclonal antibody daratumumab for induction. He cited the published experience in adding the antibody to pared-down backbone therapy in the setting of relapsed or relapsed and refractory disease, such as a proteasome inhibitor plus dexamethasone (N Engl J Med. 2016 Aug 25;375[8]:754-66) or an immunomodulator plus dexamethasone (N Engl J Med. 2016 Oct 6;375[14]:1319-31).

Adding a monoclonal antibody such as daratumumab to combination therapy is the “wave of the future,” said Dr. Niesvizky, professor of medicine and director of the Multiple Myeloma Center at New York Presbyterian Hospital/Weill Cornell Medical Center. It provides treatment that reduces disease-related complications and achieves effective and extended disease control with improved overall survival, while being well tolerated and facilitating stem cell collection.

Dr. Richardson has been a consultant to Celgene, Genmab, Janssen, Novartis, Oncopeptides AB, and Takeda and has received research funding from Celgene and Takeda. Dr. Niesvizky has been a consultant to Amgen, Bristol-Myers Squibb, Celgene, Janssen, and Takeda and has received research support from Amgen, Bristol-Myers Squibb, Celgene, and Takeda.

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

 

– Optimal induction therapy for patients with multiple myeloma requires all the therapeutic tools currently available for combination therapy, which means using four agents followed by autologous stem cell transplantation, Paul G. Richardson, MD, said at a conference held by Imedex.

He suggested that a rational combination regimen for induction therapy of multiple myeloma would include a second- or third-generation immunomodulator such as lenalidomide (Revlimid) or pomalidomide (Pomalyst); a proteasome inhibitor such as bortezomib (Velcade), carfilzomib (Kyprolis), or ixazomib (Ninlaro); plus standard dexamethasone to form a contemporary backbone regimen for inducing remission in patients with multiple myeloma.

Mitchel L. Zoler/Frontline Medical News
Dr. Paul G. Richardson (left) and Dr. Ruben Niesvizky
Dr. Richardson advocated for the addition of an anti-CD38 monoclonal antibody such as daratumumab (Darzalex), as well as follow-up management with an autologous stem cell transplant, a total package of treatments that has not yet been tested in a reported trial.

“Therapeutic parsimony is not recommended. You need a combination” of all available drug classes, suggested Dr. Richardson, professor of medicine at Harvard Medical School and clinical program leader of the Multiple Myeloma Center at the Dana-Farber Cancer Institute in Boston.

“It isn’t rational to limit the treatment choices. We need to bring them all together,” he said. The “most rational” combination melds an immunomodulator, proteasome inhibitor, plus a monoclonal antibody that he called a “true game changer. Add the antibody on top of the three-drug platform” of an immunomodulator, proteasome inhibitor, and dexamethasone. Dr. Richardson conceded, however, that a concern with such combinations is how to manage the toxicity that would likely result.

He cited several recent examples of demonstrated superior efficacy in the form of more complete responses from combined regimens, followed by autologous stem cell transplantation.

For example, a recent report from a French-led group compared the efficacy of a combination of an immunomodulator, proteasome inhibitor, and dexamethasone against the same combination, followed by autologous stem cell transplantation (N Engl J Med. 2017 Apr 6;376[14]:1311-20). The combined regimen plus transplant resulted in a 59% complete response rate, “the best response rate we’ve ever seen” in multiple myeloma, Dr. Richardson noted, compared with a 48% complete response rate in patients who did not undergo a stem cell transplant.

Another speaker at the meeting, Ruben Niesvizky, MD, suggested a more cautious approach to using the monoclonal antibody daratumumab for induction. He cited the published experience in adding the antibody to pared-down backbone therapy in the setting of relapsed or relapsed and refractory disease, such as a proteasome inhibitor plus dexamethasone (N Engl J Med. 2016 Aug 25;375[8]:754-66) or an immunomodulator plus dexamethasone (N Engl J Med. 2016 Oct 6;375[14]:1319-31).

Adding a monoclonal antibody such as daratumumab to combination therapy is the “wave of the future,” said Dr. Niesvizky, professor of medicine and director of the Multiple Myeloma Center at New York Presbyterian Hospital/Weill Cornell Medical Center. It provides treatment that reduces disease-related complications and achieves effective and extended disease control with improved overall survival, while being well tolerated and facilitating stem cell collection.

Dr. Richardson has been a consultant to Celgene, Genmab, Janssen, Novartis, Oncopeptides AB, and Takeda and has received research funding from Celgene and Takeda. Dr. Niesvizky has been a consultant to Amgen, Bristol-Myers Squibb, Celgene, Janssen, and Takeda and has received research support from Amgen, Bristol-Myers Squibb, Celgene, and Takeda.

 

– Optimal induction therapy for patients with multiple myeloma requires all the therapeutic tools currently available for combination therapy, which means using four agents followed by autologous stem cell transplantation, Paul G. Richardson, MD, said at a conference held by Imedex.

He suggested that a rational combination regimen for induction therapy of multiple myeloma would include a second- or third-generation immunomodulator such as lenalidomide (Revlimid) or pomalidomide (Pomalyst); a proteasome inhibitor such as bortezomib (Velcade), carfilzomib (Kyprolis), or ixazomib (Ninlaro); plus standard dexamethasone to form a contemporary backbone regimen for inducing remission in patients with multiple myeloma.

Mitchel L. Zoler/Frontline Medical News
Dr. Paul G. Richardson (left) and Dr. Ruben Niesvizky
Dr. Richardson advocated for the addition of an anti-CD38 monoclonal antibody such as daratumumab (Darzalex), as well as follow-up management with an autologous stem cell transplant, a total package of treatments that has not yet been tested in a reported trial.

“Therapeutic parsimony is not recommended. You need a combination” of all available drug classes, suggested Dr. Richardson, professor of medicine at Harvard Medical School and clinical program leader of the Multiple Myeloma Center at the Dana-Farber Cancer Institute in Boston.

“It isn’t rational to limit the treatment choices. We need to bring them all together,” he said. The “most rational” combination melds an immunomodulator, proteasome inhibitor, plus a monoclonal antibody that he called a “true game changer. Add the antibody on top of the three-drug platform” of an immunomodulator, proteasome inhibitor, and dexamethasone. Dr. Richardson conceded, however, that a concern with such combinations is how to manage the toxicity that would likely result.

He cited several recent examples of demonstrated superior efficacy in the form of more complete responses from combined regimens, followed by autologous stem cell transplantation.

For example, a recent report from a French-led group compared the efficacy of a combination of an immunomodulator, proteasome inhibitor, and dexamethasone against the same combination, followed by autologous stem cell transplantation (N Engl J Med. 2017 Apr 6;376[14]:1311-20). The combined regimen plus transplant resulted in a 59% complete response rate, “the best response rate we’ve ever seen” in multiple myeloma, Dr. Richardson noted, compared with a 48% complete response rate in patients who did not undergo a stem cell transplant.

Another speaker at the meeting, Ruben Niesvizky, MD, suggested a more cautious approach to using the monoclonal antibody daratumumab for induction. He cited the published experience in adding the antibody to pared-down backbone therapy in the setting of relapsed or relapsed and refractory disease, such as a proteasome inhibitor plus dexamethasone (N Engl J Med. 2016 Aug 25;375[8]:754-66) or an immunomodulator plus dexamethasone (N Engl J Med. 2016 Oct 6;375[14]:1319-31).

Adding a monoclonal antibody such as daratumumab to combination therapy is the “wave of the future,” said Dr. Niesvizky, professor of medicine and director of the Multiple Myeloma Center at New York Presbyterian Hospital/Weill Cornell Medical Center. It provides treatment that reduces disease-related complications and achieves effective and extended disease control with improved overall survival, while being well tolerated and facilitating stem cell collection.

Dr. Richardson has been a consultant to Celgene, Genmab, Janssen, Novartis, Oncopeptides AB, and Takeda and has received research funding from Celgene and Takeda. Dr. Niesvizky has been a consultant to Amgen, Bristol-Myers Squibb, Celgene, Janssen, and Takeda and has received research support from Amgen, Bristol-Myers Squibb, Celgene, and Takeda.

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

EXPERT ANALYSIS FROM A MEETING ON HEMATOLOGIC MALIGNANCIES

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
CME ID
135809

VIDEO: Blinatumomab, inotuzumab reshape relapsed ALL treatment

Article Type
Changed
Tue, 07/21/2020 - 14:18

 

NEW YORK– A pair of new monoclonal antibodies have dramatically changed treatment for patients with acute lymphoblastic leukemia to prepare them for a stem cell transplant, Daniel J. DeAngelo, MD, said at a conference held by Imedex.

“We don’t use standard chemotherapy for reinduction anymore; we use blinatumomab or inotuzumab,” said Dr. DeAngelo, a hematologist oncologist at Dana-Farber Cancer Institute in Boston.

Blinatumomab (Blincyto), approved by the Food and Drug Administration in 2014, has produced “exceptional” response rates, becoming “standard of care” for patients with relapsed acute lymphoblastic leukemia (ALL) that does not have a Philadelphia chromosome, Dr. DeAngelo said in a video interview.

Approved based on results from a phase II study, blinatumomab’s efficacy and safety were recently further delineated in results from the first phase III trial (N Engl J Med. 2017 Mar 2;376[9]:836-74), with 376 treated patients. In that trial, blinatumomab more than doubled the complete remission rate, compared with control patients (34% vs. 16%), and nearly doubled median overall survival – 7.7 months with blinatumomab, compared with 4.0 months for control patients treated with standard chemotherapy.

These findings “further substantiated” blinatumomab’s role, he said.

Blinatumomab’s big limitations are certain adverse effects and the logistics of its dosing. The major adverse effect is “cytokine release syndrome,” which manifests as fever, low blood pressure, and neurologic toxicities that can range from tremors to encephalopathy and seizure. These are manageable by close observation of patients by experienced nurses, Dr. DeAngelo said.

Dosing involves 4 weeks of continuous infusion, starting with 10 days done entirely in the hospital, with the remaining 18 days with patients going home but needing to return every 48 hours to have their infusion bag changed. “Depending on how far the patient lives from the clinic, it can be a logistical challenge,” he said.

A second new antibody he has used on many patients is inotuzumab, which was accepted for review for approval by the FDA in February 2017, with action expected by August.

Dr. DeAngelo served as a coinvestigator in a phase III trial reported in 2016 with 218 evaluable patients. In that trial, investigators reported an 81% complete remission rate with inotuzumab treatment, compared with a 29% among control patients on chemotherapy (N Engl J Med. 2016 Aug 25;375[8]:740-53).

Inotuzumab was effective against patients with Philadelphia chromosome positive ALL, but it will not work for the roughly 5%-10% of ALL patients who lack CD-22 expression in their B-cell ALL.

Inotuzumab is easier to administer than blinatumomab, requiring a once a week infusion, and causes little immediate toxicity – although thrombocytopenia and liver-function abnormalities can occur with continued use, and the risk of veno-occlusive disease is increased when patients later receive a stem cell transplant, Dr. DeAngelo said.

“It’s nice to have options” when choosing antibody-based treatment, he said. Blinatumomab is a good choice for patients with a lower tumor burden – either patients with early relapse or with minimal residual disease – while inotuzumab works better for patients with more bulky disease, as well as those who are not able to accommodate the logistic demands of blinatumomab infusions.

Dr. DeAngelo also highlighted several trials now underway that are testing the efficacy of both antibodies when used as part of first-line treatment.

Dr. DeAngelo has been a consultant to Amgen, the company that markets blinatumomab (Blincyto); to Pfizer, the company developing inotuzumab; and to Ariad, InCyte, and Novartis.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

NEW YORK– A pair of new monoclonal antibodies have dramatically changed treatment for patients with acute lymphoblastic leukemia to prepare them for a stem cell transplant, Daniel J. DeAngelo, MD, said at a conference held by Imedex.

“We don’t use standard chemotherapy for reinduction anymore; we use blinatumomab or inotuzumab,” said Dr. DeAngelo, a hematologist oncologist at Dana-Farber Cancer Institute in Boston.

Blinatumomab (Blincyto), approved by the Food and Drug Administration in 2014, has produced “exceptional” response rates, becoming “standard of care” for patients with relapsed acute lymphoblastic leukemia (ALL) that does not have a Philadelphia chromosome, Dr. DeAngelo said in a video interview.

Approved based on results from a phase II study, blinatumomab’s efficacy and safety were recently further delineated in results from the first phase III trial (N Engl J Med. 2017 Mar 2;376[9]:836-74), with 376 treated patients. In that trial, blinatumomab more than doubled the complete remission rate, compared with control patients (34% vs. 16%), and nearly doubled median overall survival – 7.7 months with blinatumomab, compared with 4.0 months for control patients treated with standard chemotherapy.

These findings “further substantiated” blinatumomab’s role, he said.

Blinatumomab’s big limitations are certain adverse effects and the logistics of its dosing. The major adverse effect is “cytokine release syndrome,” which manifests as fever, low blood pressure, and neurologic toxicities that can range from tremors to encephalopathy and seizure. These are manageable by close observation of patients by experienced nurses, Dr. DeAngelo said.

Dosing involves 4 weeks of continuous infusion, starting with 10 days done entirely in the hospital, with the remaining 18 days with patients going home but needing to return every 48 hours to have their infusion bag changed. “Depending on how far the patient lives from the clinic, it can be a logistical challenge,” he said.

A second new antibody he has used on many patients is inotuzumab, which was accepted for review for approval by the FDA in February 2017, with action expected by August.

Dr. DeAngelo served as a coinvestigator in a phase III trial reported in 2016 with 218 evaluable patients. In that trial, investigators reported an 81% complete remission rate with inotuzumab treatment, compared with a 29% among control patients on chemotherapy (N Engl J Med. 2016 Aug 25;375[8]:740-53).

Inotuzumab was effective against patients with Philadelphia chromosome positive ALL, but it will not work for the roughly 5%-10% of ALL patients who lack CD-22 expression in their B-cell ALL.

Inotuzumab is easier to administer than blinatumomab, requiring a once a week infusion, and causes little immediate toxicity – although thrombocytopenia and liver-function abnormalities can occur with continued use, and the risk of veno-occlusive disease is increased when patients later receive a stem cell transplant, Dr. DeAngelo said.

“It’s nice to have options” when choosing antibody-based treatment, he said. Blinatumomab is a good choice for patients with a lower tumor burden – either patients with early relapse or with minimal residual disease – while inotuzumab works better for patients with more bulky disease, as well as those who are not able to accommodate the logistic demands of blinatumomab infusions.

Dr. DeAngelo also highlighted several trials now underway that are testing the efficacy of both antibodies when used as part of first-line treatment.

Dr. DeAngelo has been a consultant to Amgen, the company that markets blinatumomab (Blincyto); to Pfizer, the company developing inotuzumab; and to Ariad, InCyte, and Novartis.

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

 

NEW YORK– A pair of new monoclonal antibodies have dramatically changed treatment for patients with acute lymphoblastic leukemia to prepare them for a stem cell transplant, Daniel J. DeAngelo, MD, said at a conference held by Imedex.

“We don’t use standard chemotherapy for reinduction anymore; we use blinatumomab or inotuzumab,” said Dr. DeAngelo, a hematologist oncologist at Dana-Farber Cancer Institute in Boston.

Blinatumomab (Blincyto), approved by the Food and Drug Administration in 2014, has produced “exceptional” response rates, becoming “standard of care” for patients with relapsed acute lymphoblastic leukemia (ALL) that does not have a Philadelphia chromosome, Dr. DeAngelo said in a video interview.

Approved based on results from a phase II study, blinatumomab’s efficacy and safety were recently further delineated in results from the first phase III trial (N Engl J Med. 2017 Mar 2;376[9]:836-74), with 376 treated patients. In that trial, blinatumomab more than doubled the complete remission rate, compared with control patients (34% vs. 16%), and nearly doubled median overall survival – 7.7 months with blinatumomab, compared with 4.0 months for control patients treated with standard chemotherapy.

These findings “further substantiated” blinatumomab’s role, he said.

Blinatumomab’s big limitations are certain adverse effects and the logistics of its dosing. The major adverse effect is “cytokine release syndrome,” which manifests as fever, low blood pressure, and neurologic toxicities that can range from tremors to encephalopathy and seizure. These are manageable by close observation of patients by experienced nurses, Dr. DeAngelo said.

Dosing involves 4 weeks of continuous infusion, starting with 10 days done entirely in the hospital, with the remaining 18 days with patients going home but needing to return every 48 hours to have their infusion bag changed. “Depending on how far the patient lives from the clinic, it can be a logistical challenge,” he said.

A second new antibody he has used on many patients is inotuzumab, which was accepted for review for approval by the FDA in February 2017, with action expected by August.

Dr. DeAngelo served as a coinvestigator in a phase III trial reported in 2016 with 218 evaluable patients. In that trial, investigators reported an 81% complete remission rate with inotuzumab treatment, compared with a 29% among control patients on chemotherapy (N Engl J Med. 2016 Aug 25;375[8]:740-53).

Inotuzumab was effective against patients with Philadelphia chromosome positive ALL, but it will not work for the roughly 5%-10% of ALL patients who lack CD-22 expression in their B-cell ALL.

Inotuzumab is easier to administer than blinatumomab, requiring a once a week infusion, and causes little immediate toxicity – although thrombocytopenia and liver-function abnormalities can occur with continued use, and the risk of veno-occlusive disease is increased when patients later receive a stem cell transplant, Dr. DeAngelo said.

“It’s nice to have options” when choosing antibody-based treatment, he said. Blinatumomab is a good choice for patients with a lower tumor burden – either patients with early relapse or with minimal residual disease – while inotuzumab works better for patients with more bulky disease, as well as those who are not able to accommodate the logistic demands of blinatumomab infusions.

Dr. DeAngelo also highlighted several trials now underway that are testing the efficacy of both antibodies when used as part of first-line treatment.

Dr. DeAngelo has been a consultant to Amgen, the company that markets blinatumomab (Blincyto); to Pfizer, the company developing inotuzumab; and to Ariad, InCyte, and Novartis.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
Publications
Publications
Topics
Article Type
Sections
Article Source

EXPERT ANALYSIS FROM A MEETING ON HEMATOLOGIC MALIGNANCIES

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

VIDEO: Careful TKI hiatus makes CML pregnancy possible

Article Type
Changed
Tue, 07/21/2020 - 14:18

 

– The success that tyrosine kinase inhibitors have had in prolonging life and producing deep hematologic and molecular remissions in patients with chronic myeloid leukemia has led to an unexpected bonus for young women living with the disease: an opportunity to safely become pregnant and mother a child.

The approach is not yet routine and poses a level of risk to both the mother and fetus, especially because tyrosine kinase inhibitors (TKIs) are teratogenic. But with careful planning, close gestational monitoring, and with support from skilled obstetricians, the scenario of a successful pregnancy in women with chronic myeloid leukemia (CML) has now played out several dozen times at a handful of U.S. centers, Mrinal S. Patnaik, MD, said in a talk at the conference held by Imedex.

“We make it clear that this is experimental and is associated with risk, and we share the data [from case reports]; but if the woman wants to go forward,” a protocol now exists “to successfully get them to pregnancy,” said Dr. Patnaik, a hematologist oncologist at the Mayo Clinic in Rochester, Minn.

At Mayo alone, upwards of 20 women with CML have been successfully shepherded through pregnancy, he said in a video interview.

The prospect for a planned pregnancy is reserved for women with their CML well controlled for at least 2 years using a TKI, most often imatinib (Gleevec). In addition to being under complete hematologic control, the candidate patient must also show a deep molecular response, which means a blood level of the BRC-ABL tyrosine kinase that drives CML at least 4 or 4.5 logs (10,000-50,000-fold) below pretreatment levels or molecularly undetectable.

The patient then monitors her ovulatory cycle and stops her medication at the time of ovulation, attempts conception, and then monitors whether pregnancy has actually started. If it has, she needs to stay off her TKI regimen through at least the first 18 weeks of gestation, although an even longer drug holiday is preferred. If not, she resumes the medication and repeats the process later if she wants.

Once the women is pregnant and remains off her TKI regimen Dr. Patnaik and his associates closely follow the woman for signs of a molecular or hematologic relapse, although the latter are unusual. If a resurgence of CML stem cells occurs, the woman receives treatment with pegylated interferon-alpha, which is safe during pregnancy. When possible, TKI treatment remains on hold into the breast-feeding period.

During pregnancy and delivery, the patient requires careful and regular follow-up by a maternal-fetal medicine specialist and has an ongoing risk for high platelet counts causing placental blood clots, fetuses that are small for gestational age, preterm labor, premature rupture of membranes, and other complications.

“These are manageable with good obstetrical care,” Dr. Patnaik said. “We have developed a good system to work out the obstetrical complications.

“By and large we can be successful, but it requires a lot of monitoring and a lot of patient compliance with regular follow-ups,” he stressed.

In a video interview at the meeting, Dr. Patnaik discussed the approach he takes with his patients.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– The success that tyrosine kinase inhibitors have had in prolonging life and producing deep hematologic and molecular remissions in patients with chronic myeloid leukemia has led to an unexpected bonus for young women living with the disease: an opportunity to safely become pregnant and mother a child.

The approach is not yet routine and poses a level of risk to both the mother and fetus, especially because tyrosine kinase inhibitors (TKIs) are teratogenic. But with careful planning, close gestational monitoring, and with support from skilled obstetricians, the scenario of a successful pregnancy in women with chronic myeloid leukemia (CML) has now played out several dozen times at a handful of U.S. centers, Mrinal S. Patnaik, MD, said in a talk at the conference held by Imedex.

“We make it clear that this is experimental and is associated with risk, and we share the data [from case reports]; but if the woman wants to go forward,” a protocol now exists “to successfully get them to pregnancy,” said Dr. Patnaik, a hematologist oncologist at the Mayo Clinic in Rochester, Minn.

At Mayo alone, upwards of 20 women with CML have been successfully shepherded through pregnancy, he said in a video interview.

The prospect for a planned pregnancy is reserved for women with their CML well controlled for at least 2 years using a TKI, most often imatinib (Gleevec). In addition to being under complete hematologic control, the candidate patient must also show a deep molecular response, which means a blood level of the BRC-ABL tyrosine kinase that drives CML at least 4 or 4.5 logs (10,000-50,000-fold) below pretreatment levels or molecularly undetectable.

The patient then monitors her ovulatory cycle and stops her medication at the time of ovulation, attempts conception, and then monitors whether pregnancy has actually started. If it has, she needs to stay off her TKI regimen through at least the first 18 weeks of gestation, although an even longer drug holiday is preferred. If not, she resumes the medication and repeats the process later if she wants.

Once the women is pregnant and remains off her TKI regimen Dr. Patnaik and his associates closely follow the woman for signs of a molecular or hematologic relapse, although the latter are unusual. If a resurgence of CML stem cells occurs, the woman receives treatment with pegylated interferon-alpha, which is safe during pregnancy. When possible, TKI treatment remains on hold into the breast-feeding period.

During pregnancy and delivery, the patient requires careful and regular follow-up by a maternal-fetal medicine specialist and has an ongoing risk for high platelet counts causing placental blood clots, fetuses that are small for gestational age, preterm labor, premature rupture of membranes, and other complications.

“These are manageable with good obstetrical care,” Dr. Patnaik said. “We have developed a good system to work out the obstetrical complications.

“By and large we can be successful, but it requires a lot of monitoring and a lot of patient compliance with regular follow-ups,” he stressed.

In a video interview at the meeting, Dr. Patnaik discussed the approach he takes with his patients.

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

 

– The success that tyrosine kinase inhibitors have had in prolonging life and producing deep hematologic and molecular remissions in patients with chronic myeloid leukemia has led to an unexpected bonus for young women living with the disease: an opportunity to safely become pregnant and mother a child.

The approach is not yet routine and poses a level of risk to both the mother and fetus, especially because tyrosine kinase inhibitors (TKIs) are teratogenic. But with careful planning, close gestational monitoring, and with support from skilled obstetricians, the scenario of a successful pregnancy in women with chronic myeloid leukemia (CML) has now played out several dozen times at a handful of U.S. centers, Mrinal S. Patnaik, MD, said in a talk at the conference held by Imedex.

“We make it clear that this is experimental and is associated with risk, and we share the data [from case reports]; but if the woman wants to go forward,” a protocol now exists “to successfully get them to pregnancy,” said Dr. Patnaik, a hematologist oncologist at the Mayo Clinic in Rochester, Minn.

At Mayo alone, upwards of 20 women with CML have been successfully shepherded through pregnancy, he said in a video interview.

The prospect for a planned pregnancy is reserved for women with their CML well controlled for at least 2 years using a TKI, most often imatinib (Gleevec). In addition to being under complete hematologic control, the candidate patient must also show a deep molecular response, which means a blood level of the BRC-ABL tyrosine kinase that drives CML at least 4 or 4.5 logs (10,000-50,000-fold) below pretreatment levels or molecularly undetectable.

The patient then monitors her ovulatory cycle and stops her medication at the time of ovulation, attempts conception, and then monitors whether pregnancy has actually started. If it has, she needs to stay off her TKI regimen through at least the first 18 weeks of gestation, although an even longer drug holiday is preferred. If not, she resumes the medication and repeats the process later if she wants.

Once the women is pregnant and remains off her TKI regimen Dr. Patnaik and his associates closely follow the woman for signs of a molecular or hematologic relapse, although the latter are unusual. If a resurgence of CML stem cells occurs, the woman receives treatment with pegylated interferon-alpha, which is safe during pregnancy. When possible, TKI treatment remains on hold into the breast-feeding period.

During pregnancy and delivery, the patient requires careful and regular follow-up by a maternal-fetal medicine specialist and has an ongoing risk for high platelet counts causing placental blood clots, fetuses that are small for gestational age, preterm labor, premature rupture of membranes, and other complications.

“These are manageable with good obstetrical care,” Dr. Patnaik said. “We have developed a good system to work out the obstetrical complications.

“By and large we can be successful, but it requires a lot of monitoring and a lot of patient compliance with regular follow-ups,” he stressed.

In a video interview at the meeting, Dr. Patnaik discussed the approach he takes with his patients.

The video associated with this article is no longer available on this site. Please view all of our videos on the MDedge YouTube channel
Publications
Publications
Topics
Article Type
Sections
Article Source

EXPERT ANALYSIS FROM A MEETING ON HEMATOLOGIC MALIGNANCIES

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