LayerRx Mapping ID
950
Slot System
Featured Buckets
Featured Buckets Admin
Reverse Chronological Sort
Allow Teaser Image
Medscape Lead Concept
5000612

High cost and demand for old cancer drug sparks crisis

Article Type
Changed
Fri, 12/16/2022 - 12:34

 

As a severe shortage drags on and prices soar, transplant centers have been struggling to cope with the paucity and price of fludarabine, a chemotherapy drug that has become an essential component of stem-cell transplants for some blood cancers.

At Oregon Health and Science University, for example, an extensive algorithm now offers guidance through a thicket of alternative options, from adjusting doses and using substitutes to delaying treatment. Meanwhile, some institutions have enlisted ethicists and attorneys to guide their decisions on which patients will have to wait for potentially life-saving treatment.

Even as surgeons turn to alternatives, advocates for transplantation in hematology have warned about the potential for harm.

“This continued fludarabine shortage is forcing centers to use non–[Food and Drug Administration] approved lymphodepleting regimens that may negatively impact the success of a possibly lifesaving CAR-T therapy,” Brenda Sandmaier, MD, president of the Transplantation and Cellular Therapy American Society, and Jeffery Auletta, MD, a senior vice president with the National Marrow Donor, said in a June 30 letter to the FDA. The physicians added that they “request the FDA to take immediate action on this critical shortage. Many centers currently have no ability to purchase fludarabine through their suppliers and have no estimated time frame for return of availability. Other centers are limited to mere weeks of supply, with continued uncertainty of future availability.”

In October, less than 4 months after that letter was sent, one of the manufacturers of fludarabine – Areva Pharmaceuticals – marked up the price of fludarabine to $2,736 per vial, 10-20 times that of two other makers of the drug.
 

In new treatment era, fludarabine remains crucial

In 2015, ASH Clinical News – a publication of the American Society of Hematology – invited a pair of hematologists to discuss whether fludarabine is “dead” as a front-line treatment for chronic lymphocytic leukemia (CLL). “Fludarabine is not dead yet, but the data from those and other long-term trials may be the final nail in its coffin,” said Mitchell Smith, MD, PhD, who was then with Cleveland Clinic and now works for George Washington University.

Seven years later, the role of fludarabine as a long-term chemotherapeutic agent in blood cancer has definitely evolved. Just as oncologists predicted back in 2015, “the use of fludarabine declined for the primary management of CLL and other B cell malignancies, due to the development of targeted therapies such as BTK inhibitors, venetoclax, and other agents,” Memorial Sloan Kettering hematologic oncologist Anthony Mato, MD, said in an interview.

But the drug “remains a critical agent for conditioning the immune system for cellular therapies such as allogeneic stem cell transplantation and CAR-T cells,” Dr. Mato said.

Nirav Shah, MD, a hematologic oncologist at the Medical College of Wisconsin, explained in an interview that “conditioning” in the stem-cell transplant context refers to “wiping out” the immune system, allowing the donor’s stem cells to avoid rejection. “It’s a commonly used drug,” he said, “and shortage was not really a concern that people faced until this year.”
 

 

 

As shortage continues, price hike brings yet another hit

The first reports of fludarabine being in short supply surfaced about a year ago. According to a Nov. 2 update from the American Society of Health-System Pharmacists, five companies now manufacture fludarabine, and all of them report shortages. Areva, which dramatically raised its price, is accepting direct orders. Leucadia and Teva don’t know when the drug will be available; and Fresenius Kabi and Sagent expect availability in early 2023.

Areva, Leucadia, and Teva didn’t provide reasons for their shortages. Fresenius Kabi blamed increased demand, and Sagent pointed to manufacturing delays. Pfizer, another manufacturer, had a tiny market share and stopped making fludarabine in 2020, according to the pharmacist society.

In a May 12 press release, a company called Lannett announced it would take over U.S. distribution of fludarabine for Areva and suggested that the supply shortage would be lucrative: “While total U.S. sales for the 12 months ended March 2022 of Fludarabine Phosphate for injection, USP, 50 mg/2mL were approximately $4.9 million, according to IQVIA, the current market value is believed to be higher due to the recent market disruptions.”

“We were all shocked and outraged when Areva came out with the new, dramatically higher prices,” Bill Greene, PharmD, chief pharmaceutical officer at St. Jude Children’s Research Hospital, said in a recent interview.

In a prior interview, conducted during the summer of 2022, Dr. Greene addressed the topic of hematologic drug shortages. Back then he noted that he was seeking emergency supplies of fludarabine, since all five manufacturers reported having no stock available.

Interviewed again in November 2022, Dr. Greene noted that the hospital “had been able to stay ahead of the need and meet the needs of our patients” through arrangements with Teva and Fresenius Kabi. “In cases of patient need, we certainly are willing to pay a higher product price if that’s what it takes to get it – assuming the product is a quality product.”

The Medical College of Wisconsin’s Dr. Shah said insurers may refuse to cover the higher price, sticking medical institutions with the bill.
 

Alternatives abound, but do they suffice?

There is some good news on the fludarabine shortage front. Areva recently alerted providers that it was releasing fludarabine from non-FDA-approved suppliers with the agency’s permission, and Accord Healthcare said it received permission to sell fludarabine that was marketed in Canada.

Another option – oral fludarabine instead of the standard IV version – remains unavailable in the United States. According to the June letter to the FDA from the American Society for Transplantation and Cellular Therapy and National Marrow Donor Program, it “might be an appropriate alternative” and is available in Europe, Canada and Australia.

The letter warns that “transplant centers have also been forced to move away from fludarabine-based regimens and use alternative drugs such as cladribine or clofarabine, which are both significantly less studied and rely on single-center experience or limited phase II data. ... The limited availability of fludarabine is leading to the use of alternative regimens that are known to be more toxic or understudied alternatives with unknown long-term clinical effects or harms to patients.”

In a November 2022 report published in Transplantation and Cellular Therapy, Dr. Shah and colleagues noted that institutions are adopting strategies such as “(1) pharmacy dose banding and rounding down to save vials, even if a >5% reduction was required; (2) administering all dosing of fludarabine based not on actual body weight but on adjusted body weight; and (3) switching the billing of fludarabine from single-dose vials to billing by dose delivery.”

If the shortage continues, “it becomes necessary for centers to establish algorithms for management now,” they wrote. “Substitution of such agents as bendamustine and cladribine can be considered ... [and] another acceptable solution could be the substitution of clofarabine for fludarabine.”

Still, there are many unanswered questions. “The challenge is that these alternative regimens have not been extensively studied in a large population,” Dr. Shah said. “You have to be more mindful of potential side effects and risks, and the biggest concern is efficacy. Is changing the drug going to be detrimental to a patient’s outcome? To be honest, we don’t know the answer to that.”

Dr. Mato disclosed ties with TG Therapeutics, Pharmacyclics, AbbVie, Acerta, Adaptive Biotechnologies, AstraZeneca, BeiGene, BioPharma, BMS, Curio, Dava, DTRM, Genentech, Genmab, Janssen, Johnson & Johnson, LOXO, Medscape, Nurix, Octapharma, PER, PerView, and Pfizer. Dr. Greene and Dr. Shah have no disclosures.

Publications
Topics
Sections

 

As a severe shortage drags on and prices soar, transplant centers have been struggling to cope with the paucity and price of fludarabine, a chemotherapy drug that has become an essential component of stem-cell transplants for some blood cancers.

At Oregon Health and Science University, for example, an extensive algorithm now offers guidance through a thicket of alternative options, from adjusting doses and using substitutes to delaying treatment. Meanwhile, some institutions have enlisted ethicists and attorneys to guide their decisions on which patients will have to wait for potentially life-saving treatment.

Even as surgeons turn to alternatives, advocates for transplantation in hematology have warned about the potential for harm.

“This continued fludarabine shortage is forcing centers to use non–[Food and Drug Administration] approved lymphodepleting regimens that may negatively impact the success of a possibly lifesaving CAR-T therapy,” Brenda Sandmaier, MD, president of the Transplantation and Cellular Therapy American Society, and Jeffery Auletta, MD, a senior vice president with the National Marrow Donor, said in a June 30 letter to the FDA. The physicians added that they “request the FDA to take immediate action on this critical shortage. Many centers currently have no ability to purchase fludarabine through their suppliers and have no estimated time frame for return of availability. Other centers are limited to mere weeks of supply, with continued uncertainty of future availability.”

In October, less than 4 months after that letter was sent, one of the manufacturers of fludarabine – Areva Pharmaceuticals – marked up the price of fludarabine to $2,736 per vial, 10-20 times that of two other makers of the drug.
 

In new treatment era, fludarabine remains crucial

In 2015, ASH Clinical News – a publication of the American Society of Hematology – invited a pair of hematologists to discuss whether fludarabine is “dead” as a front-line treatment for chronic lymphocytic leukemia (CLL). “Fludarabine is not dead yet, but the data from those and other long-term trials may be the final nail in its coffin,” said Mitchell Smith, MD, PhD, who was then with Cleveland Clinic and now works for George Washington University.

Seven years later, the role of fludarabine as a long-term chemotherapeutic agent in blood cancer has definitely evolved. Just as oncologists predicted back in 2015, “the use of fludarabine declined for the primary management of CLL and other B cell malignancies, due to the development of targeted therapies such as BTK inhibitors, venetoclax, and other agents,” Memorial Sloan Kettering hematologic oncologist Anthony Mato, MD, said in an interview.

But the drug “remains a critical agent for conditioning the immune system for cellular therapies such as allogeneic stem cell transplantation and CAR-T cells,” Dr. Mato said.

Nirav Shah, MD, a hematologic oncologist at the Medical College of Wisconsin, explained in an interview that “conditioning” in the stem-cell transplant context refers to “wiping out” the immune system, allowing the donor’s stem cells to avoid rejection. “It’s a commonly used drug,” he said, “and shortage was not really a concern that people faced until this year.”
 

 

 

As shortage continues, price hike brings yet another hit

The first reports of fludarabine being in short supply surfaced about a year ago. According to a Nov. 2 update from the American Society of Health-System Pharmacists, five companies now manufacture fludarabine, and all of them report shortages. Areva, which dramatically raised its price, is accepting direct orders. Leucadia and Teva don’t know when the drug will be available; and Fresenius Kabi and Sagent expect availability in early 2023.

Areva, Leucadia, and Teva didn’t provide reasons for their shortages. Fresenius Kabi blamed increased demand, and Sagent pointed to manufacturing delays. Pfizer, another manufacturer, had a tiny market share and stopped making fludarabine in 2020, according to the pharmacist society.

In a May 12 press release, a company called Lannett announced it would take over U.S. distribution of fludarabine for Areva and suggested that the supply shortage would be lucrative: “While total U.S. sales for the 12 months ended March 2022 of Fludarabine Phosphate for injection, USP, 50 mg/2mL were approximately $4.9 million, according to IQVIA, the current market value is believed to be higher due to the recent market disruptions.”

“We were all shocked and outraged when Areva came out with the new, dramatically higher prices,” Bill Greene, PharmD, chief pharmaceutical officer at St. Jude Children’s Research Hospital, said in a recent interview.

In a prior interview, conducted during the summer of 2022, Dr. Greene addressed the topic of hematologic drug shortages. Back then he noted that he was seeking emergency supplies of fludarabine, since all five manufacturers reported having no stock available.

Interviewed again in November 2022, Dr. Greene noted that the hospital “had been able to stay ahead of the need and meet the needs of our patients” through arrangements with Teva and Fresenius Kabi. “In cases of patient need, we certainly are willing to pay a higher product price if that’s what it takes to get it – assuming the product is a quality product.”

The Medical College of Wisconsin’s Dr. Shah said insurers may refuse to cover the higher price, sticking medical institutions with the bill.
 

Alternatives abound, but do they suffice?

There is some good news on the fludarabine shortage front. Areva recently alerted providers that it was releasing fludarabine from non-FDA-approved suppliers with the agency’s permission, and Accord Healthcare said it received permission to sell fludarabine that was marketed in Canada.

Another option – oral fludarabine instead of the standard IV version – remains unavailable in the United States. According to the June letter to the FDA from the American Society for Transplantation and Cellular Therapy and National Marrow Donor Program, it “might be an appropriate alternative” and is available in Europe, Canada and Australia.

The letter warns that “transplant centers have also been forced to move away from fludarabine-based regimens and use alternative drugs such as cladribine or clofarabine, which are both significantly less studied and rely on single-center experience or limited phase II data. ... The limited availability of fludarabine is leading to the use of alternative regimens that are known to be more toxic or understudied alternatives with unknown long-term clinical effects or harms to patients.”

In a November 2022 report published in Transplantation and Cellular Therapy, Dr. Shah and colleagues noted that institutions are adopting strategies such as “(1) pharmacy dose banding and rounding down to save vials, even if a >5% reduction was required; (2) administering all dosing of fludarabine based not on actual body weight but on adjusted body weight; and (3) switching the billing of fludarabine from single-dose vials to billing by dose delivery.”

If the shortage continues, “it becomes necessary for centers to establish algorithms for management now,” they wrote. “Substitution of such agents as bendamustine and cladribine can be considered ... [and] another acceptable solution could be the substitution of clofarabine for fludarabine.”

Still, there are many unanswered questions. “The challenge is that these alternative regimens have not been extensively studied in a large population,” Dr. Shah said. “You have to be more mindful of potential side effects and risks, and the biggest concern is efficacy. Is changing the drug going to be detrimental to a patient’s outcome? To be honest, we don’t know the answer to that.”

Dr. Mato disclosed ties with TG Therapeutics, Pharmacyclics, AbbVie, Acerta, Adaptive Biotechnologies, AstraZeneca, BeiGene, BioPharma, BMS, Curio, Dava, DTRM, Genentech, Genmab, Janssen, Johnson & Johnson, LOXO, Medscape, Nurix, Octapharma, PER, PerView, and Pfizer. Dr. Greene and Dr. Shah have no disclosures.

 

As a severe shortage drags on and prices soar, transplant centers have been struggling to cope with the paucity and price of fludarabine, a chemotherapy drug that has become an essential component of stem-cell transplants for some blood cancers.

At Oregon Health and Science University, for example, an extensive algorithm now offers guidance through a thicket of alternative options, from adjusting doses and using substitutes to delaying treatment. Meanwhile, some institutions have enlisted ethicists and attorneys to guide their decisions on which patients will have to wait for potentially life-saving treatment.

Even as surgeons turn to alternatives, advocates for transplantation in hematology have warned about the potential for harm.

“This continued fludarabine shortage is forcing centers to use non–[Food and Drug Administration] approved lymphodepleting regimens that may negatively impact the success of a possibly lifesaving CAR-T therapy,” Brenda Sandmaier, MD, president of the Transplantation and Cellular Therapy American Society, and Jeffery Auletta, MD, a senior vice president with the National Marrow Donor, said in a June 30 letter to the FDA. The physicians added that they “request the FDA to take immediate action on this critical shortage. Many centers currently have no ability to purchase fludarabine through their suppliers and have no estimated time frame for return of availability. Other centers are limited to mere weeks of supply, with continued uncertainty of future availability.”

In October, less than 4 months after that letter was sent, one of the manufacturers of fludarabine – Areva Pharmaceuticals – marked up the price of fludarabine to $2,736 per vial, 10-20 times that of two other makers of the drug.
 

In new treatment era, fludarabine remains crucial

In 2015, ASH Clinical News – a publication of the American Society of Hematology – invited a pair of hematologists to discuss whether fludarabine is “dead” as a front-line treatment for chronic lymphocytic leukemia (CLL). “Fludarabine is not dead yet, but the data from those and other long-term trials may be the final nail in its coffin,” said Mitchell Smith, MD, PhD, who was then with Cleveland Clinic and now works for George Washington University.

Seven years later, the role of fludarabine as a long-term chemotherapeutic agent in blood cancer has definitely evolved. Just as oncologists predicted back in 2015, “the use of fludarabine declined for the primary management of CLL and other B cell malignancies, due to the development of targeted therapies such as BTK inhibitors, venetoclax, and other agents,” Memorial Sloan Kettering hematologic oncologist Anthony Mato, MD, said in an interview.

But the drug “remains a critical agent for conditioning the immune system for cellular therapies such as allogeneic stem cell transplantation and CAR-T cells,” Dr. Mato said.

Nirav Shah, MD, a hematologic oncologist at the Medical College of Wisconsin, explained in an interview that “conditioning” in the stem-cell transplant context refers to “wiping out” the immune system, allowing the donor’s stem cells to avoid rejection. “It’s a commonly used drug,” he said, “and shortage was not really a concern that people faced until this year.”
 

 

 

As shortage continues, price hike brings yet another hit

The first reports of fludarabine being in short supply surfaced about a year ago. According to a Nov. 2 update from the American Society of Health-System Pharmacists, five companies now manufacture fludarabine, and all of them report shortages. Areva, which dramatically raised its price, is accepting direct orders. Leucadia and Teva don’t know when the drug will be available; and Fresenius Kabi and Sagent expect availability in early 2023.

Areva, Leucadia, and Teva didn’t provide reasons for their shortages. Fresenius Kabi blamed increased demand, and Sagent pointed to manufacturing delays. Pfizer, another manufacturer, had a tiny market share and stopped making fludarabine in 2020, according to the pharmacist society.

In a May 12 press release, a company called Lannett announced it would take over U.S. distribution of fludarabine for Areva and suggested that the supply shortage would be lucrative: “While total U.S. sales for the 12 months ended March 2022 of Fludarabine Phosphate for injection, USP, 50 mg/2mL were approximately $4.9 million, according to IQVIA, the current market value is believed to be higher due to the recent market disruptions.”

“We were all shocked and outraged when Areva came out with the new, dramatically higher prices,” Bill Greene, PharmD, chief pharmaceutical officer at St. Jude Children’s Research Hospital, said in a recent interview.

In a prior interview, conducted during the summer of 2022, Dr. Greene addressed the topic of hematologic drug shortages. Back then he noted that he was seeking emergency supplies of fludarabine, since all five manufacturers reported having no stock available.

Interviewed again in November 2022, Dr. Greene noted that the hospital “had been able to stay ahead of the need and meet the needs of our patients” through arrangements with Teva and Fresenius Kabi. “In cases of patient need, we certainly are willing to pay a higher product price if that’s what it takes to get it – assuming the product is a quality product.”

The Medical College of Wisconsin’s Dr. Shah said insurers may refuse to cover the higher price, sticking medical institutions with the bill.
 

Alternatives abound, but do they suffice?

There is some good news on the fludarabine shortage front. Areva recently alerted providers that it was releasing fludarabine from non-FDA-approved suppliers with the agency’s permission, and Accord Healthcare said it received permission to sell fludarabine that was marketed in Canada.

Another option – oral fludarabine instead of the standard IV version – remains unavailable in the United States. According to the June letter to the FDA from the American Society for Transplantation and Cellular Therapy and National Marrow Donor Program, it “might be an appropriate alternative” and is available in Europe, Canada and Australia.

The letter warns that “transplant centers have also been forced to move away from fludarabine-based regimens and use alternative drugs such as cladribine or clofarabine, which are both significantly less studied and rely on single-center experience or limited phase II data. ... The limited availability of fludarabine is leading to the use of alternative regimens that are known to be more toxic or understudied alternatives with unknown long-term clinical effects or harms to patients.”

In a November 2022 report published in Transplantation and Cellular Therapy, Dr. Shah and colleagues noted that institutions are adopting strategies such as “(1) pharmacy dose banding and rounding down to save vials, even if a >5% reduction was required; (2) administering all dosing of fludarabine based not on actual body weight but on adjusted body weight; and (3) switching the billing of fludarabine from single-dose vials to billing by dose delivery.”

If the shortage continues, “it becomes necessary for centers to establish algorithms for management now,” they wrote. “Substitution of such agents as bendamustine and cladribine can be considered ... [and] another acceptable solution could be the substitution of clofarabine for fludarabine.”

Still, there are many unanswered questions. “The challenge is that these alternative regimens have not been extensively studied in a large population,” Dr. Shah said. “You have to be more mindful of potential side effects and risks, and the biggest concern is efficacy. Is changing the drug going to be detrimental to a patient’s outcome? To be honest, we don’t know the answer to that.”

Dr. Mato disclosed ties with TG Therapeutics, Pharmacyclics, AbbVie, Acerta, Adaptive Biotechnologies, AstraZeneca, BeiGene, BioPharma, BMS, Curio, Dava, DTRM, Genentech, Genmab, Janssen, Johnson & Johnson, LOXO, Medscape, Nurix, Octapharma, PER, PerView, and Pfizer. Dr. Greene and Dr. Shah have no disclosures.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Transplant provides no clear survival benefit in real-world MCL study

Article Type
Changed
Fri, 12/16/2022 - 12:34

In younger patients with mantle cell lymphoma treated in U.S. community oncology settings in recent years, use of autologous transplant was not associated with improved survival, results of a large observational study show.

Autologous stem-cell transplant (ASCT) use was not linked overall survival (OS), according to the authors of the retrospective analysis of patients diagnosed with mantle cell lymphoma (MCL) between 2011 and 2021.

This lack of a clear survival benefit with use of ASCT is an “apparent contradiction” with prospective data from earlier clinical trials, authors wrote in the Journal of Clinical Oncology

However, they added, the finding is consistent with several recent registry analyses that also do not support a link between ASCT and overall survival in patients with MCL.

Although these findings are limited by the retrospective nature of the study, the results at least suggest that it is ethical to do research that doesn’t involve ASCT, study author Peter Martin, MD, said in an interview.

Furthermore, emerging data from the randomized TRIANGLE study from the European MCL Network suggest the potential for ASCT to be replaced by maintenance therapy with the Bruton's tyrosine kinase inhibitor ibrutinib, according to Dr. Martin, associate professor with Weill Cornell Medicine, New York.

“There are probably a lot of questions that will come up there, but essentially the barriers to research that do not include ASCT have been moved away, and we can go ahead and study non-ASCT (approaches) in younger patients,” Dr. Martin said.
 

No clear OS benefit

In current guidelines, recommended initial therapy for MCL patients younger than 65 years includes use of high-dose cytarabine-containing chemoimmunotherapy induction, followed by ASCT as consolidation, and then rituximab maintenance, Dr. Martin and coauthors say in their report.

Their primary analysis was based on the Flatiron Health database, which is derived from electronic medical records, mostly in U.S. community oncology practices, according to the report.

The researchers identified 1,274 patients under the age of 65 with a record of first-line treatment for MCL, and of those, 962 (or 76%) were considered eligible for ASCT.

Among ASCT-eligible patients, there was no significant association between receipt of ASCT and OS, with a hazard ratio of 0.86 (95% confidence interval, 0.63-1.18). The 3-year OS was 88% for patients receiving ASCT and similarly, 84% for those who did not, according to authors.

Likewise, there was no association between ASCT and real-world time to next treatment, an endpoint defined as time from start of first-line therapy to subsequent treatment or death, the report says.
 

Findings in perspective

The lack of clear survival benefit with ASCT in this and other recent observational studies may be explained in part by improvements in induction regimens, according to Timothy Fenske, MD, professor in the department of medicine at the Medical College of Wisconsin, Milwaukee.

“As our induction regimens have improved, it is very possible that the benefit for autologous transplantation will become less apparent,” Dr. Fenske said in an interview.

The discussion over ASCT in MCL is expected to evolve further in light of findings from TRIANGLE and EA4151, a randomized phase 3 trial of rituximab with or without ASCT specifically in patients with minimal residual disease (MRD)–negative MCL in first complete remission.

“If that study shows that the MRD-negative patients do not have much benefit from autologous transplantation,” Dr. Fenske said, “I think these studies will all be giving the same message – that autologous transplantation was beneficial back when induction regimens were poor (for example, CHOP without rituximab), but will have much less benefit in patients receiving modern inductions, which by and large will get more patients to be MRD negative.”

However, subgroup analyses of those TRIANGLE will be important, he added, since some patients may still benefit from ASCT, such as younger patients who remain MRD positive, or who have certain other high-risk molecular features.

Dr. Martin reported consulting or advisory roles with Janssen, BeiGene, Karyopharm Therapeutics, Kite/Gilead, Verastem, ADC Therapeutics, Bristol Myers Squibb/Celgene, Epizyme, Merck, MorphoSys, and Takeda. He reported institutional research funding from Karyopharm Therapeutics.

Publications
Topics
Sections

In younger patients with mantle cell lymphoma treated in U.S. community oncology settings in recent years, use of autologous transplant was not associated with improved survival, results of a large observational study show.

Autologous stem-cell transplant (ASCT) use was not linked overall survival (OS), according to the authors of the retrospective analysis of patients diagnosed with mantle cell lymphoma (MCL) between 2011 and 2021.

This lack of a clear survival benefit with use of ASCT is an “apparent contradiction” with prospective data from earlier clinical trials, authors wrote in the Journal of Clinical Oncology

However, they added, the finding is consistent with several recent registry analyses that also do not support a link between ASCT and overall survival in patients with MCL.

Although these findings are limited by the retrospective nature of the study, the results at least suggest that it is ethical to do research that doesn’t involve ASCT, study author Peter Martin, MD, said in an interview.

Furthermore, emerging data from the randomized TRIANGLE study from the European MCL Network suggest the potential for ASCT to be replaced by maintenance therapy with the Bruton's tyrosine kinase inhibitor ibrutinib, according to Dr. Martin, associate professor with Weill Cornell Medicine, New York.

“There are probably a lot of questions that will come up there, but essentially the barriers to research that do not include ASCT have been moved away, and we can go ahead and study non-ASCT (approaches) in younger patients,” Dr. Martin said.
 

No clear OS benefit

In current guidelines, recommended initial therapy for MCL patients younger than 65 years includes use of high-dose cytarabine-containing chemoimmunotherapy induction, followed by ASCT as consolidation, and then rituximab maintenance, Dr. Martin and coauthors say in their report.

Their primary analysis was based on the Flatiron Health database, which is derived from electronic medical records, mostly in U.S. community oncology practices, according to the report.

The researchers identified 1,274 patients under the age of 65 with a record of first-line treatment for MCL, and of those, 962 (or 76%) were considered eligible for ASCT.

Among ASCT-eligible patients, there was no significant association between receipt of ASCT and OS, with a hazard ratio of 0.86 (95% confidence interval, 0.63-1.18). The 3-year OS was 88% for patients receiving ASCT and similarly, 84% for those who did not, according to authors.

Likewise, there was no association between ASCT and real-world time to next treatment, an endpoint defined as time from start of first-line therapy to subsequent treatment or death, the report says.
 

Findings in perspective

The lack of clear survival benefit with ASCT in this and other recent observational studies may be explained in part by improvements in induction regimens, according to Timothy Fenske, MD, professor in the department of medicine at the Medical College of Wisconsin, Milwaukee.

“As our induction regimens have improved, it is very possible that the benefit for autologous transplantation will become less apparent,” Dr. Fenske said in an interview.

The discussion over ASCT in MCL is expected to evolve further in light of findings from TRIANGLE and EA4151, a randomized phase 3 trial of rituximab with or without ASCT specifically in patients with minimal residual disease (MRD)–negative MCL in first complete remission.

“If that study shows that the MRD-negative patients do not have much benefit from autologous transplantation,” Dr. Fenske said, “I think these studies will all be giving the same message – that autologous transplantation was beneficial back when induction regimens were poor (for example, CHOP without rituximab), but will have much less benefit in patients receiving modern inductions, which by and large will get more patients to be MRD negative.”

However, subgroup analyses of those TRIANGLE will be important, he added, since some patients may still benefit from ASCT, such as younger patients who remain MRD positive, or who have certain other high-risk molecular features.

Dr. Martin reported consulting or advisory roles with Janssen, BeiGene, Karyopharm Therapeutics, Kite/Gilead, Verastem, ADC Therapeutics, Bristol Myers Squibb/Celgene, Epizyme, Merck, MorphoSys, and Takeda. He reported institutional research funding from Karyopharm Therapeutics.

In younger patients with mantle cell lymphoma treated in U.S. community oncology settings in recent years, use of autologous transplant was not associated with improved survival, results of a large observational study show.

Autologous stem-cell transplant (ASCT) use was not linked overall survival (OS), according to the authors of the retrospective analysis of patients diagnosed with mantle cell lymphoma (MCL) between 2011 and 2021.

This lack of a clear survival benefit with use of ASCT is an “apparent contradiction” with prospective data from earlier clinical trials, authors wrote in the Journal of Clinical Oncology

However, they added, the finding is consistent with several recent registry analyses that also do not support a link between ASCT and overall survival in patients with MCL.

Although these findings are limited by the retrospective nature of the study, the results at least suggest that it is ethical to do research that doesn’t involve ASCT, study author Peter Martin, MD, said in an interview.

Furthermore, emerging data from the randomized TRIANGLE study from the European MCL Network suggest the potential for ASCT to be replaced by maintenance therapy with the Bruton's tyrosine kinase inhibitor ibrutinib, according to Dr. Martin, associate professor with Weill Cornell Medicine, New York.

“There are probably a lot of questions that will come up there, but essentially the barriers to research that do not include ASCT have been moved away, and we can go ahead and study non-ASCT (approaches) in younger patients,” Dr. Martin said.
 

No clear OS benefit

In current guidelines, recommended initial therapy for MCL patients younger than 65 years includes use of high-dose cytarabine-containing chemoimmunotherapy induction, followed by ASCT as consolidation, and then rituximab maintenance, Dr. Martin and coauthors say in their report.

Their primary analysis was based on the Flatiron Health database, which is derived from electronic medical records, mostly in U.S. community oncology practices, according to the report.

The researchers identified 1,274 patients under the age of 65 with a record of first-line treatment for MCL, and of those, 962 (or 76%) were considered eligible for ASCT.

Among ASCT-eligible patients, there was no significant association between receipt of ASCT and OS, with a hazard ratio of 0.86 (95% confidence interval, 0.63-1.18). The 3-year OS was 88% for patients receiving ASCT and similarly, 84% for those who did not, according to authors.

Likewise, there was no association between ASCT and real-world time to next treatment, an endpoint defined as time from start of first-line therapy to subsequent treatment or death, the report says.
 

Findings in perspective

The lack of clear survival benefit with ASCT in this and other recent observational studies may be explained in part by improvements in induction regimens, according to Timothy Fenske, MD, professor in the department of medicine at the Medical College of Wisconsin, Milwaukee.

“As our induction regimens have improved, it is very possible that the benefit for autologous transplantation will become less apparent,” Dr. Fenske said in an interview.

The discussion over ASCT in MCL is expected to evolve further in light of findings from TRIANGLE and EA4151, a randomized phase 3 trial of rituximab with or without ASCT specifically in patients with minimal residual disease (MRD)–negative MCL in first complete remission.

“If that study shows that the MRD-negative patients do not have much benefit from autologous transplantation,” Dr. Fenske said, “I think these studies will all be giving the same message – that autologous transplantation was beneficial back when induction regimens were poor (for example, CHOP without rituximab), but will have much less benefit in patients receiving modern inductions, which by and large will get more patients to be MRD negative.”

However, subgroup analyses of those TRIANGLE will be important, he added, since some patients may still benefit from ASCT, such as younger patients who remain MRD positive, or who have certain other high-risk molecular features.

Dr. Martin reported consulting or advisory roles with Janssen, BeiGene, Karyopharm Therapeutics, Kite/Gilead, Verastem, ADC Therapeutics, Bristol Myers Squibb/Celgene, Epizyme, Merck, MorphoSys, and Takeda. He reported institutional research funding from Karyopharm Therapeutics.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM THE JOURNAL OF CLINICAL ONCOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Blame MCL, not transplantation, for late effects?

Article Type
Changed
Fri, 12/16/2022 - 12:34

In patients with mantle cell lymphoma, rates of respiratory disease, blood disorders, and infectious diseases do not vary according to the intensity of treatment given, the results of a large retrospective analysis suggested.

The high overall incidence of these late effects, compared with individuals without mantle cell lymphoma (MCL), was similar whether the patients were or were not treated with autologous stem cell transplantation (ASCT), according to authors of the study.

The rate of hospitalization among MCL patients was also high, but again, did not differ between ASCT and non-ASCT subgroups in the study, which included adult patients younger than age 70 with MCL who were treated in Sweden between 2000 and 2014.
 

Late effects independent of ASCT

These findings may have implications for clinicians tempted to avoid intensive first-line treatment including ASCT because it is “demanding” and may cause late effects, study authors wrote in a research article that appeared in Blood Advances.

Dr. Ingrid Grimelius, MD, PhD, professor of oncology, Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden
Dr. Ingrid Glimelius

In fact, the great majority of long-term health care needs in patients with MCL appear to be related to the lymphoma in itself, according to study senior author Ingrid Glimelius, MD, PhD, senior consultant and professor in oncology in the department of immunology, genetics, and pathology at Uppsala University in Sweden.

“You do have to keep your eyes open for complications like blood disorders, infections, and respiratory (disorders),” Dr. Glimelius said in an interview. “But it’s not the transplant that adds to the extra toxicity. So don’t be afraid of giving that, if you think that can prolong your patient’s remission.”
 

Whither transplantation?

While these data may advance the discussion over the relative safety of ASCT, she added, the paradigm is changing to ask a different question: Does the patient need a transplant, or not?

Dr. Glimelius said she was looking forward to results of TRIANGLE, a randomized, open-label, three-arm study initiated by the European MCL Network. This study compares standard first-line treatment including ASCT to the kinase inhibitor ibrutinib, which the U.S. Food and Drug Administration approved in 2013 for patients previously treated for MCL.

In the TRIANGLE study, younger patients with MCL were randomized to the standard first-line treatment, standard treatment plus ibrutinib, or ibrutinib alone.

A preliminary report on the study stated that the current standard is “not superior” to the new ibrutinib-containing regimen without ASCT, though more follow-up is needed.

Full results of the study are expected to be presented at the American Society of Hematology meeting on December 11.

“In my opinion, our data will be practice-changing,” said lead investigator Martin Dreyling, MD, PhD, professor of medicine and head of the lymphoma program at the University of Munich Hospital.
 

Little known about late effects

In the meantime, clinicians may be reassured by the current data from Dr. Glimelius and coauthors, which showed that late effects varied little by treatment choice.

That’s important, Dr. Glimelius said, because even as survival is improving and novel targeted drugs are taking the stage, knowledge about the late effects of MCL remains limited.

Their population-based study included all 620 patients with MCL in the Swedish Lymphoma Register who were 18-69 years of age and diagnosed between 2000 and 2014. Records were found for 620 patients, of whom 247 received high-dose chemotherapy with ASCT.

Compared with healthy individuals with no MCL, the patients with MCL had a high rate of specialist visits and hospital visits, according to the report. The MCL patients also had high risks of infections, respiratory complications, and blood disorders relative to the healthy subjects.
 

 

 

Lack of differences between arms

The key finding of the report, though, is the lack of significant differences in the rate of complications between the ASCT and non–ASCT-treated patients.

Relative to healthy subjects, patients undergoing ASCT and not undergoing ASCT had a higher risk of infections, with hazard ratios of 5.62 (95% confidence interval, 4.20-7.52) and 4.66 (95% CI, 3.62-5.00), respectively.

Relative risks of respiratory complications were also similar, with HRs of 4.38 and 5.26, respectively, and overlapping CIs. Likewise, the risk of blood disorders was not statistically different, with HRs of 9.84 and 5.80, respectively, but again with overlapping CIs.

Outpatient visits, inpatient visits, and bed days were likewise similar between ASCT and non-ASCT arms.

In fact, most patients died of their lymphoma, rather than a treatment complication or another cause of death, the investigators noted in their report.

Dr. Glimelius reported receiving honoraria from Janssen. Coauthors on the paper reported disclosures related to Janssen, Gilead, Celgene, Roche, Acerta. and AbbVie.

Correction, 11/21/22: The photo caption misstated Dr. Ingrid Glimelius' name.

Publications
Topics
Sections

In patients with mantle cell lymphoma, rates of respiratory disease, blood disorders, and infectious diseases do not vary according to the intensity of treatment given, the results of a large retrospective analysis suggested.

The high overall incidence of these late effects, compared with individuals without mantle cell lymphoma (MCL), was similar whether the patients were or were not treated with autologous stem cell transplantation (ASCT), according to authors of the study.

The rate of hospitalization among MCL patients was also high, but again, did not differ between ASCT and non-ASCT subgroups in the study, which included adult patients younger than age 70 with MCL who were treated in Sweden between 2000 and 2014.
 

Late effects independent of ASCT

These findings may have implications for clinicians tempted to avoid intensive first-line treatment including ASCT because it is “demanding” and may cause late effects, study authors wrote in a research article that appeared in Blood Advances.

Dr. Ingrid Grimelius, MD, PhD, professor of oncology, Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden
Dr. Ingrid Glimelius

In fact, the great majority of long-term health care needs in patients with MCL appear to be related to the lymphoma in itself, according to study senior author Ingrid Glimelius, MD, PhD, senior consultant and professor in oncology in the department of immunology, genetics, and pathology at Uppsala University in Sweden.

“You do have to keep your eyes open for complications like blood disorders, infections, and respiratory (disorders),” Dr. Glimelius said in an interview. “But it’s not the transplant that adds to the extra toxicity. So don’t be afraid of giving that, if you think that can prolong your patient’s remission.”
 

Whither transplantation?

While these data may advance the discussion over the relative safety of ASCT, she added, the paradigm is changing to ask a different question: Does the patient need a transplant, or not?

Dr. Glimelius said she was looking forward to results of TRIANGLE, a randomized, open-label, three-arm study initiated by the European MCL Network. This study compares standard first-line treatment including ASCT to the kinase inhibitor ibrutinib, which the U.S. Food and Drug Administration approved in 2013 for patients previously treated for MCL.

In the TRIANGLE study, younger patients with MCL were randomized to the standard first-line treatment, standard treatment plus ibrutinib, or ibrutinib alone.

A preliminary report on the study stated that the current standard is “not superior” to the new ibrutinib-containing regimen without ASCT, though more follow-up is needed.

Full results of the study are expected to be presented at the American Society of Hematology meeting on December 11.

“In my opinion, our data will be practice-changing,” said lead investigator Martin Dreyling, MD, PhD, professor of medicine and head of the lymphoma program at the University of Munich Hospital.
 

Little known about late effects

In the meantime, clinicians may be reassured by the current data from Dr. Glimelius and coauthors, which showed that late effects varied little by treatment choice.

That’s important, Dr. Glimelius said, because even as survival is improving and novel targeted drugs are taking the stage, knowledge about the late effects of MCL remains limited.

Their population-based study included all 620 patients with MCL in the Swedish Lymphoma Register who were 18-69 years of age and diagnosed between 2000 and 2014. Records were found for 620 patients, of whom 247 received high-dose chemotherapy with ASCT.

Compared with healthy individuals with no MCL, the patients with MCL had a high rate of specialist visits and hospital visits, according to the report. The MCL patients also had high risks of infections, respiratory complications, and blood disorders relative to the healthy subjects.
 

 

 

Lack of differences between arms

The key finding of the report, though, is the lack of significant differences in the rate of complications between the ASCT and non–ASCT-treated patients.

Relative to healthy subjects, patients undergoing ASCT and not undergoing ASCT had a higher risk of infections, with hazard ratios of 5.62 (95% confidence interval, 4.20-7.52) and 4.66 (95% CI, 3.62-5.00), respectively.

Relative risks of respiratory complications were also similar, with HRs of 4.38 and 5.26, respectively, and overlapping CIs. Likewise, the risk of blood disorders was not statistically different, with HRs of 9.84 and 5.80, respectively, but again with overlapping CIs.

Outpatient visits, inpatient visits, and bed days were likewise similar between ASCT and non-ASCT arms.

In fact, most patients died of their lymphoma, rather than a treatment complication or another cause of death, the investigators noted in their report.

Dr. Glimelius reported receiving honoraria from Janssen. Coauthors on the paper reported disclosures related to Janssen, Gilead, Celgene, Roche, Acerta. and AbbVie.

Correction, 11/21/22: The photo caption misstated Dr. Ingrid Glimelius' name.

In patients with mantle cell lymphoma, rates of respiratory disease, blood disorders, and infectious diseases do not vary according to the intensity of treatment given, the results of a large retrospective analysis suggested.

The high overall incidence of these late effects, compared with individuals without mantle cell lymphoma (MCL), was similar whether the patients were or were not treated with autologous stem cell transplantation (ASCT), according to authors of the study.

The rate of hospitalization among MCL patients was also high, but again, did not differ between ASCT and non-ASCT subgroups in the study, which included adult patients younger than age 70 with MCL who were treated in Sweden between 2000 and 2014.
 

Late effects independent of ASCT

These findings may have implications for clinicians tempted to avoid intensive first-line treatment including ASCT because it is “demanding” and may cause late effects, study authors wrote in a research article that appeared in Blood Advances.

Dr. Ingrid Grimelius, MD, PhD, professor of oncology, Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden
Dr. Ingrid Glimelius

In fact, the great majority of long-term health care needs in patients with MCL appear to be related to the lymphoma in itself, according to study senior author Ingrid Glimelius, MD, PhD, senior consultant and professor in oncology in the department of immunology, genetics, and pathology at Uppsala University in Sweden.

“You do have to keep your eyes open for complications like blood disorders, infections, and respiratory (disorders),” Dr. Glimelius said in an interview. “But it’s not the transplant that adds to the extra toxicity. So don’t be afraid of giving that, if you think that can prolong your patient’s remission.”
 

Whither transplantation?

While these data may advance the discussion over the relative safety of ASCT, she added, the paradigm is changing to ask a different question: Does the patient need a transplant, or not?

Dr. Glimelius said she was looking forward to results of TRIANGLE, a randomized, open-label, three-arm study initiated by the European MCL Network. This study compares standard first-line treatment including ASCT to the kinase inhibitor ibrutinib, which the U.S. Food and Drug Administration approved in 2013 for patients previously treated for MCL.

In the TRIANGLE study, younger patients with MCL were randomized to the standard first-line treatment, standard treatment plus ibrutinib, or ibrutinib alone.

A preliminary report on the study stated that the current standard is “not superior” to the new ibrutinib-containing regimen without ASCT, though more follow-up is needed.

Full results of the study are expected to be presented at the American Society of Hematology meeting on December 11.

“In my opinion, our data will be practice-changing,” said lead investigator Martin Dreyling, MD, PhD, professor of medicine and head of the lymphoma program at the University of Munich Hospital.
 

Little known about late effects

In the meantime, clinicians may be reassured by the current data from Dr. Glimelius and coauthors, which showed that late effects varied little by treatment choice.

That’s important, Dr. Glimelius said, because even as survival is improving and novel targeted drugs are taking the stage, knowledge about the late effects of MCL remains limited.

Their population-based study included all 620 patients with MCL in the Swedish Lymphoma Register who were 18-69 years of age and diagnosed between 2000 and 2014. Records were found for 620 patients, of whom 247 received high-dose chemotherapy with ASCT.

Compared with healthy individuals with no MCL, the patients with MCL had a high rate of specialist visits and hospital visits, according to the report. The MCL patients also had high risks of infections, respiratory complications, and blood disorders relative to the healthy subjects.
 

 

 

Lack of differences between arms

The key finding of the report, though, is the lack of significant differences in the rate of complications between the ASCT and non–ASCT-treated patients.

Relative to healthy subjects, patients undergoing ASCT and not undergoing ASCT had a higher risk of infections, with hazard ratios of 5.62 (95% confidence interval, 4.20-7.52) and 4.66 (95% CI, 3.62-5.00), respectively.

Relative risks of respiratory complications were also similar, with HRs of 4.38 and 5.26, respectively, and overlapping CIs. Likewise, the risk of blood disorders was not statistically different, with HRs of 9.84 and 5.80, respectively, but again with overlapping CIs.

Outpatient visits, inpatient visits, and bed days were likewise similar between ASCT and non-ASCT arms.

In fact, most patients died of their lymphoma, rather than a treatment complication or another cause of death, the investigators noted in their report.

Dr. Glimelius reported receiving honoraria from Janssen. Coauthors on the paper reported disclosures related to Janssen, Gilead, Celgene, Roche, Acerta. and AbbVie.

Correction, 11/21/22: The photo caption misstated Dr. Ingrid Glimelius' name.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM BLOOD ADVANCES

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Phase 3 trial yields better way to predict MCL outcomes

Article Type
Changed
Fri, 12/16/2022 - 12:34

While assessment of minimal residual disease at a single time point is useful for predicting outcomes in patients treated for mantle cell lymphoma (MCL), multiple assessments over time may offer a more accurate prognostic approach, the results of a recent randomized, phase 3 trial suggest.

Single assessments of minimal residual disease (MRD) effectively prognosticated relapse risks in patients with MCL treated with chemoimmunotherapy and transplantation, according to investigators.

Dr. Simone Ferrero, MD, assistant professor of hematology at the University of Torino, in Torino, Italy
Dr. Simone Ferrero

However, by evaluating MRD status over time, disease progression could be predicted with greater certainty, according to study lead author Simone Ferrero, MD, assistant professor of hematology at the University of Torino (Italy).

“The most important message is that MRD is predictive, and then in particular, if you want to study MRD, you have to do a repeated MRD analysis in order to have a better picture of the disease course and to determine the prognosis of each single patient,” Dr. Ferrero said in an interview.
 

Predictive power of MRD

Piers Blombery, MBBS, hematologist and medical lead of the Molecular Hematology Laboratory at Peter MacCallum Cancer Centre in Melbourne, said these study findings illustrate how the predictive power of MRD is enhanced when it is evaluated longitudinally, over the course of a patient’s disease.

“We need to be prepared to have models of MRD that take into account changes in values across time,” Dr. Blombery said in an interview.

“Conventionally, we have looked at single time points in disease, such as end of induction therapy or post autologous stem cell transplant,” said Dr. Blombery, “but we may need now to have more complex models that can integrate values over multiple time points to guide intervention.”

Dr. Blombery, who was not involved in the study, coauthored an independent commentary on the results that was also published in Blood.
 

Tools to predict relapse

Although initial MCL treatment is often effective, most patients will relapse. Accordingly, there have been considerable efforts to develop tools that identify patients at high risk of relapse, according to Dr. Ferrero and coauthors.

Among these tools is the Mantle Cell Lymphoma International Prognostic Index (MIPI), which risk-stratifies patients on the basis of age, performance status, leukocyte count, and lactate dehydrogenase.

More recently, in several published studies, MRD has been demonstrated a high predictive value in MCL. However, the best way to evaluate MRD has yet to be worked out. There are differences in testing methods, sampling (that is, bone marrow versus peripheral blood), and time points that are evaluated.

In some recent studies, investigators have pooled MRD samples taken at different time points, according to Dr. Ferrero and colleagues. Furthermore, there had been no prior systematic attempts to compare the prognostic performance of MRD data evaluated over time, as compared with MRD at a single time point.
 

Exploiting MRD data

The analysis by Dr. Ferrero and coauthors, which appears in the journal Blood, is a substudy of FIL MCL0208, a recent multicenter, randomized, phase 3 clinical trial that demonstrated the benefit of lenalidomide maintenance over observation following autologous stem cell transplantation (ASCT) among 300 patients with MCL in Italy and Portugal.

In the study, investigators monitored MRD in both peripheral blood and bone marrow at 10 fixed time points throughout treatment (including induction, consolidation, post ASCT, and then every 6 months during maintenance and follow-up), yielding 4,351 individual MRD results.

Individual MRD analyses were prognostic in MCL, according to the investigators, with results that were relevant to disease progression right after ASCT, and were most predictive starting at 6 months after ASCT.

However, the best way to exploit the bulk of MRD information that had been collected, investigators said, was a model that combined regularly updated MRD results with MIPI scores.

This MIPI-adjusted model, based on MRD assessed in bone marrow by real-time quantitative polymerase chain reaction (RQ-PCR), outperformed both MIPI and a MIPI-adjusted single time-point analysis in terms of predicting time to progression.

The predictive power of the MIPI-adjusted bone marrow RQ-PCR analysis was illustrated by an area under the curve of 0.85-0.87, compared with an AUC of just 0.60-0.63 for classic MIPI analysis, 0.62-0.65 for MIPI-adjusted MRD at a post-ASCT time-point analysis, and 0.74-0.77 for MIPI-adjusted MRD at 6 months from transplant.
 

Peripheral blood ‘easier to collect’

Although bone marrow analysis performed best in the single-point MRD evaluations, looking at MRD over time greatly improved the predictive power of the peripheral blood MRD results, yielding an AUC up to 0.81, according to the report.

This enhancement of peripheral blood performance is a finding that may have important clinical implications, according to Dr. Ferrero, considering that peripheral blood monitoring is more practical for long-term, repeated MRD monitoring.

“It is interesting, because peripheral blood is easier to collect than bone marrow,” Dr. Ferrero said. “So if you plan in your analysis to monitor MRD in peripheral blood every 6 months, then you can have a predictive value similar to that of one single point in bone marrow.”

Dr. Ferrero provided disclosures related to Janssen, EUSA Pharma, Gilead, Morphosys, Incyte, Clinigen, Servier, and Gentili. The other authors reported no relevant conflicts of interest.

Publications
Topics
Sections

While assessment of minimal residual disease at a single time point is useful for predicting outcomes in patients treated for mantle cell lymphoma (MCL), multiple assessments over time may offer a more accurate prognostic approach, the results of a recent randomized, phase 3 trial suggest.

Single assessments of minimal residual disease (MRD) effectively prognosticated relapse risks in patients with MCL treated with chemoimmunotherapy and transplantation, according to investigators.

Dr. Simone Ferrero, MD, assistant professor of hematology at the University of Torino, in Torino, Italy
Dr. Simone Ferrero

However, by evaluating MRD status over time, disease progression could be predicted with greater certainty, according to study lead author Simone Ferrero, MD, assistant professor of hematology at the University of Torino (Italy).

“The most important message is that MRD is predictive, and then in particular, if you want to study MRD, you have to do a repeated MRD analysis in order to have a better picture of the disease course and to determine the prognosis of each single patient,” Dr. Ferrero said in an interview.
 

Predictive power of MRD

Piers Blombery, MBBS, hematologist and medical lead of the Molecular Hematology Laboratory at Peter MacCallum Cancer Centre in Melbourne, said these study findings illustrate how the predictive power of MRD is enhanced when it is evaluated longitudinally, over the course of a patient’s disease.

“We need to be prepared to have models of MRD that take into account changes in values across time,” Dr. Blombery said in an interview.

“Conventionally, we have looked at single time points in disease, such as end of induction therapy or post autologous stem cell transplant,” said Dr. Blombery, “but we may need now to have more complex models that can integrate values over multiple time points to guide intervention.”

Dr. Blombery, who was not involved in the study, coauthored an independent commentary on the results that was also published in Blood.
 

Tools to predict relapse

Although initial MCL treatment is often effective, most patients will relapse. Accordingly, there have been considerable efforts to develop tools that identify patients at high risk of relapse, according to Dr. Ferrero and coauthors.

Among these tools is the Mantle Cell Lymphoma International Prognostic Index (MIPI), which risk-stratifies patients on the basis of age, performance status, leukocyte count, and lactate dehydrogenase.

More recently, in several published studies, MRD has been demonstrated a high predictive value in MCL. However, the best way to evaluate MRD has yet to be worked out. There are differences in testing methods, sampling (that is, bone marrow versus peripheral blood), and time points that are evaluated.

In some recent studies, investigators have pooled MRD samples taken at different time points, according to Dr. Ferrero and colleagues. Furthermore, there had been no prior systematic attempts to compare the prognostic performance of MRD data evaluated over time, as compared with MRD at a single time point.
 

Exploiting MRD data

The analysis by Dr. Ferrero and coauthors, which appears in the journal Blood, is a substudy of FIL MCL0208, a recent multicenter, randomized, phase 3 clinical trial that demonstrated the benefit of lenalidomide maintenance over observation following autologous stem cell transplantation (ASCT) among 300 patients with MCL in Italy and Portugal.

In the study, investigators monitored MRD in both peripheral blood and bone marrow at 10 fixed time points throughout treatment (including induction, consolidation, post ASCT, and then every 6 months during maintenance and follow-up), yielding 4,351 individual MRD results.

Individual MRD analyses were prognostic in MCL, according to the investigators, with results that were relevant to disease progression right after ASCT, and were most predictive starting at 6 months after ASCT.

However, the best way to exploit the bulk of MRD information that had been collected, investigators said, was a model that combined regularly updated MRD results with MIPI scores.

This MIPI-adjusted model, based on MRD assessed in bone marrow by real-time quantitative polymerase chain reaction (RQ-PCR), outperformed both MIPI and a MIPI-adjusted single time-point analysis in terms of predicting time to progression.

The predictive power of the MIPI-adjusted bone marrow RQ-PCR analysis was illustrated by an area under the curve of 0.85-0.87, compared with an AUC of just 0.60-0.63 for classic MIPI analysis, 0.62-0.65 for MIPI-adjusted MRD at a post-ASCT time-point analysis, and 0.74-0.77 for MIPI-adjusted MRD at 6 months from transplant.
 

Peripheral blood ‘easier to collect’

Although bone marrow analysis performed best in the single-point MRD evaluations, looking at MRD over time greatly improved the predictive power of the peripheral blood MRD results, yielding an AUC up to 0.81, according to the report.

This enhancement of peripheral blood performance is a finding that may have important clinical implications, according to Dr. Ferrero, considering that peripheral blood monitoring is more practical for long-term, repeated MRD monitoring.

“It is interesting, because peripheral blood is easier to collect than bone marrow,” Dr. Ferrero said. “So if you plan in your analysis to monitor MRD in peripheral blood every 6 months, then you can have a predictive value similar to that of one single point in bone marrow.”

Dr. Ferrero provided disclosures related to Janssen, EUSA Pharma, Gilead, Morphosys, Incyte, Clinigen, Servier, and Gentili. The other authors reported no relevant conflicts of interest.

While assessment of minimal residual disease at a single time point is useful for predicting outcomes in patients treated for mantle cell lymphoma (MCL), multiple assessments over time may offer a more accurate prognostic approach, the results of a recent randomized, phase 3 trial suggest.

Single assessments of minimal residual disease (MRD) effectively prognosticated relapse risks in patients with MCL treated with chemoimmunotherapy and transplantation, according to investigators.

Dr. Simone Ferrero, MD, assistant professor of hematology at the University of Torino, in Torino, Italy
Dr. Simone Ferrero

However, by evaluating MRD status over time, disease progression could be predicted with greater certainty, according to study lead author Simone Ferrero, MD, assistant professor of hematology at the University of Torino (Italy).

“The most important message is that MRD is predictive, and then in particular, if you want to study MRD, you have to do a repeated MRD analysis in order to have a better picture of the disease course and to determine the prognosis of each single patient,” Dr. Ferrero said in an interview.
 

Predictive power of MRD

Piers Blombery, MBBS, hematologist and medical lead of the Molecular Hematology Laboratory at Peter MacCallum Cancer Centre in Melbourne, said these study findings illustrate how the predictive power of MRD is enhanced when it is evaluated longitudinally, over the course of a patient’s disease.

“We need to be prepared to have models of MRD that take into account changes in values across time,” Dr. Blombery said in an interview.

“Conventionally, we have looked at single time points in disease, such as end of induction therapy or post autologous stem cell transplant,” said Dr. Blombery, “but we may need now to have more complex models that can integrate values over multiple time points to guide intervention.”

Dr. Blombery, who was not involved in the study, coauthored an independent commentary on the results that was also published in Blood.
 

Tools to predict relapse

Although initial MCL treatment is often effective, most patients will relapse. Accordingly, there have been considerable efforts to develop tools that identify patients at high risk of relapse, according to Dr. Ferrero and coauthors.

Among these tools is the Mantle Cell Lymphoma International Prognostic Index (MIPI), which risk-stratifies patients on the basis of age, performance status, leukocyte count, and lactate dehydrogenase.

More recently, in several published studies, MRD has been demonstrated a high predictive value in MCL. However, the best way to evaluate MRD has yet to be worked out. There are differences in testing methods, sampling (that is, bone marrow versus peripheral blood), and time points that are evaluated.

In some recent studies, investigators have pooled MRD samples taken at different time points, according to Dr. Ferrero and colleagues. Furthermore, there had been no prior systematic attempts to compare the prognostic performance of MRD data evaluated over time, as compared with MRD at a single time point.
 

Exploiting MRD data

The analysis by Dr. Ferrero and coauthors, which appears in the journal Blood, is a substudy of FIL MCL0208, a recent multicenter, randomized, phase 3 clinical trial that demonstrated the benefit of lenalidomide maintenance over observation following autologous stem cell transplantation (ASCT) among 300 patients with MCL in Italy and Portugal.

In the study, investigators monitored MRD in both peripheral blood and bone marrow at 10 fixed time points throughout treatment (including induction, consolidation, post ASCT, and then every 6 months during maintenance and follow-up), yielding 4,351 individual MRD results.

Individual MRD analyses were prognostic in MCL, according to the investigators, with results that were relevant to disease progression right after ASCT, and were most predictive starting at 6 months after ASCT.

However, the best way to exploit the bulk of MRD information that had been collected, investigators said, was a model that combined regularly updated MRD results with MIPI scores.

This MIPI-adjusted model, based on MRD assessed in bone marrow by real-time quantitative polymerase chain reaction (RQ-PCR), outperformed both MIPI and a MIPI-adjusted single time-point analysis in terms of predicting time to progression.

The predictive power of the MIPI-adjusted bone marrow RQ-PCR analysis was illustrated by an area under the curve of 0.85-0.87, compared with an AUC of just 0.60-0.63 for classic MIPI analysis, 0.62-0.65 for MIPI-adjusted MRD at a post-ASCT time-point analysis, and 0.74-0.77 for MIPI-adjusted MRD at 6 months from transplant.
 

Peripheral blood ‘easier to collect’

Although bone marrow analysis performed best in the single-point MRD evaluations, looking at MRD over time greatly improved the predictive power of the peripheral blood MRD results, yielding an AUC up to 0.81, according to the report.

This enhancement of peripheral blood performance is a finding that may have important clinical implications, according to Dr. Ferrero, considering that peripheral blood monitoring is more practical for long-term, repeated MRD monitoring.

“It is interesting, because peripheral blood is easier to collect than bone marrow,” Dr. Ferrero said. “So if you plan in your analysis to monitor MRD in peripheral blood every 6 months, then you can have a predictive value similar to that of one single point in bone marrow.”

Dr. Ferrero provided disclosures related to Janssen, EUSA Pharma, Gilead, Morphosys, Incyte, Clinigen, Servier, and Gentili. The other authors reported no relevant conflicts of interest.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM BLOOD

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Gloom lifting as MCL treatments evolve

Article Type
Changed
Fri, 12/16/2022 - 12:34

 

An optimistic new review offers guidance about treating mantle cell lymphoma (MCL) in the new era of chimeric antigen receptor (CAR)-T cell therapy and medication choices informed by risk stratification.

Traditionally, MCL has had a notoriously poor prognosis and is still impossible to cure. But survival rates are rising thanks to better treatments, the review authors wrote, and even relapsed/refractory patients have a growing number of options that can potentially give them extra years of life.

“Prognosis has certainly changed in past 10 years. We have been able to have an excellent control of disease, and patients are living longer, even past the 8- or 10-year mark,” Moffit Cancer Center/Memorial Healthcare System hematologist-oncologist Jose Sandoval‐Sus, MD, said in an interview. He is corresponding author of the review, which appeared in the October issue of Current Oncology Reports.

MCL – which affects cells in the mantle zone of lymph nodes – is rare. It usually strikes older men, often presents at an advanced stage, and accounts for 6%-8% of non-Hodgkin lymphomas in the United States.

Prognoses are improving. The review highlights a study released earlier this year that found that median 5-year overall survival has increased from 68.8% (2002-2009) to 81.6% (2010-2015).

Now, the review notes, there are several first-line chemotherapy options that combine agents with rituximab such as rituximab/bendamustine, which “has generally been established as an effective treatment for MCL at first relapse in patients who are bendamustine naive when compared to other chemotherapy agents.”

Other treatments include rituximab, bortezomib, cyclophosphamide, doxorubicin, vincristine, and prednisolone; rituximab, bendamustine and cytarabine; and rituximab, gemcitabine, and oxaliplatin.

“I think of rituximab as a medication of maintenance, either after autologous stem cell transplant or even in patients who have not been through transplant,” Dr. Sandoval‐Sus said. “As maintenance, it really has improved outcomes for these patients.”

But the first step before treatment, he said, is to explore prognostic factors such as alterations on the TP53 gene that “really dictate a lot in terms of the prognosis of patients.” As the review notes, these alterations – either bi-allelic del17p or TP53 mutations – “are associated with poor outcomes after frontline and salvage regimens, including targeted agents such as Burton’s tyrosine kinase inhibitors (BTKis).”

These patients, who make up about 20% of those with MCL, also are most unlikely to benefit from autologous stem cell transplantation, Dr. Sandoval‐Sus said.

What about refracted/relapsed (R/R) cases? BTKis have been a major advance for these patients, he said. However, choosing the best drug can be a challenge. As the review notes, “all approved BTKis for R/R MCL seem to have similar clinical outcomes based on identical mechanism of action, and there are no prospective trials comparing these agents in a head-to-head fashion.”

The authors added that “we wonder if AEs [adverse events] could be decreased by using combinations based on new generation BTKi, but it is still a question that needs to be resolved in the clinical trial arena.”

Stem cell transplants may be an option, the review said, but “in practice the clinical benefit ... is limited to single-center series or small multi-institutional registries with few prospective studies.”

Then there’s CAR-T cell therapy, the game-changer. A type called brexucabtagene autoleucel (Brexu-cel) is now approved in MCL, the review authors wrote, and real-world data “serve as a platform to expand CAR-T therapy to more R/R MCL patients that do not fit the strict inclusion criteria of the studies (e.g., controlled comorbidities and worse performance status)... We strongly recommend early referral of these patients to accredited institutions with ample cellular therapy experience, including high-risk MCL patients (e.g., blastoid/pleomorphic morphology, biallelic del17p, TP53 mutations) so an appropriate bridging strategy and a CAR-T cell roadmap is planned with the patient and caretakers.”

Some researchers are exploring combination treatment with both BTKis and CAR T-cell therapy, “which may be considered for patients with R/R MCL who are naive to both CAR T-cell and BTKi therapy, because combination therapy may increase treatment efficacy,” wrote the authors of another review that appeared in the October issue of Current Oncology Reports. “Based on limited data in patients with CLL, BTKi therapy may be initiated as bridging therapy and continued during lymphodepletion prior to CAR T-cell infusion”

What’s next? Multiple treatments are in the research stage, Dr. Sandoval‐Sus said. “There are a lot of things in development that are really incredible.”

Reversible BTKis, for example, appear to be effective at controlling disease and are well-tolerated, he said. “And we are awaiting the results of clinical trials of targeted therapies.”

For now, he said, the best advice for hematologists is to gain a full understanding of a patient’s MCL, in order to provide the most appropriate treatment. Community oncologists should get at least one second opinion from an academic center or other clinic that treats these kinds of lymphomas, he said, and molecular tests are crucial. A discussion about stem cell transplantation after remission is a good idea, he said, and so is an exploration of clinical trials “from the get-go.”

“In patients who relapse and have high-risk features, they should be started on a BTKi inhibitor for the most part,” he said, “although we need to weigh risks and benefits between the side effects of different BTKi inhibitors. And they should be referred earlier to a CAR T cell therapy center, so they can discuss the benefits and see if they’re an appropriate patient. I think patients are being referred a little bit too late in the second- or third-line setting.”

What about CAR T therapy as a first-line therapy? It’s not FDA-approved, Dr. Sandoval‐Sus said, and “definitely not a standard of care.” But clinical trials are exploring the idea, he said. As for messages to patients, Dr. Sandoval-Sus said he would tell them that MCL is not yet curable, “but the future is very bright.”

Dr. Sandoval-Sus declared advisory board relationships with Seagen, Incyte, Janssen, ADC Therapeutics, TG therapeutics, and Genmab. The other review authors had no disclosures.

Publications
Topics
Sections

 

An optimistic new review offers guidance about treating mantle cell lymphoma (MCL) in the new era of chimeric antigen receptor (CAR)-T cell therapy and medication choices informed by risk stratification.

Traditionally, MCL has had a notoriously poor prognosis and is still impossible to cure. But survival rates are rising thanks to better treatments, the review authors wrote, and even relapsed/refractory patients have a growing number of options that can potentially give them extra years of life.

“Prognosis has certainly changed in past 10 years. We have been able to have an excellent control of disease, and patients are living longer, even past the 8- or 10-year mark,” Moffit Cancer Center/Memorial Healthcare System hematologist-oncologist Jose Sandoval‐Sus, MD, said in an interview. He is corresponding author of the review, which appeared in the October issue of Current Oncology Reports.

MCL – which affects cells in the mantle zone of lymph nodes – is rare. It usually strikes older men, often presents at an advanced stage, and accounts for 6%-8% of non-Hodgkin lymphomas in the United States.

Prognoses are improving. The review highlights a study released earlier this year that found that median 5-year overall survival has increased from 68.8% (2002-2009) to 81.6% (2010-2015).

Now, the review notes, there are several first-line chemotherapy options that combine agents with rituximab such as rituximab/bendamustine, which “has generally been established as an effective treatment for MCL at first relapse in patients who are bendamustine naive when compared to other chemotherapy agents.”

Other treatments include rituximab, bortezomib, cyclophosphamide, doxorubicin, vincristine, and prednisolone; rituximab, bendamustine and cytarabine; and rituximab, gemcitabine, and oxaliplatin.

“I think of rituximab as a medication of maintenance, either after autologous stem cell transplant or even in patients who have not been through transplant,” Dr. Sandoval‐Sus said. “As maintenance, it really has improved outcomes for these patients.”

But the first step before treatment, he said, is to explore prognostic factors such as alterations on the TP53 gene that “really dictate a lot in terms of the prognosis of patients.” As the review notes, these alterations – either bi-allelic del17p or TP53 mutations – “are associated with poor outcomes after frontline and salvage regimens, including targeted agents such as Burton’s tyrosine kinase inhibitors (BTKis).”

These patients, who make up about 20% of those with MCL, also are most unlikely to benefit from autologous stem cell transplantation, Dr. Sandoval‐Sus said.

What about refracted/relapsed (R/R) cases? BTKis have been a major advance for these patients, he said. However, choosing the best drug can be a challenge. As the review notes, “all approved BTKis for R/R MCL seem to have similar clinical outcomes based on identical mechanism of action, and there are no prospective trials comparing these agents in a head-to-head fashion.”

The authors added that “we wonder if AEs [adverse events] could be decreased by using combinations based on new generation BTKi, but it is still a question that needs to be resolved in the clinical trial arena.”

Stem cell transplants may be an option, the review said, but “in practice the clinical benefit ... is limited to single-center series or small multi-institutional registries with few prospective studies.”

Then there’s CAR-T cell therapy, the game-changer. A type called brexucabtagene autoleucel (Brexu-cel) is now approved in MCL, the review authors wrote, and real-world data “serve as a platform to expand CAR-T therapy to more R/R MCL patients that do not fit the strict inclusion criteria of the studies (e.g., controlled comorbidities and worse performance status)... We strongly recommend early referral of these patients to accredited institutions with ample cellular therapy experience, including high-risk MCL patients (e.g., blastoid/pleomorphic morphology, biallelic del17p, TP53 mutations) so an appropriate bridging strategy and a CAR-T cell roadmap is planned with the patient and caretakers.”

Some researchers are exploring combination treatment with both BTKis and CAR T-cell therapy, “which may be considered for patients with R/R MCL who are naive to both CAR T-cell and BTKi therapy, because combination therapy may increase treatment efficacy,” wrote the authors of another review that appeared in the October issue of Current Oncology Reports. “Based on limited data in patients with CLL, BTKi therapy may be initiated as bridging therapy and continued during lymphodepletion prior to CAR T-cell infusion”

What’s next? Multiple treatments are in the research stage, Dr. Sandoval‐Sus said. “There are a lot of things in development that are really incredible.”

Reversible BTKis, for example, appear to be effective at controlling disease and are well-tolerated, he said. “And we are awaiting the results of clinical trials of targeted therapies.”

For now, he said, the best advice for hematologists is to gain a full understanding of a patient’s MCL, in order to provide the most appropriate treatment. Community oncologists should get at least one second opinion from an academic center or other clinic that treats these kinds of lymphomas, he said, and molecular tests are crucial. A discussion about stem cell transplantation after remission is a good idea, he said, and so is an exploration of clinical trials “from the get-go.”

“In patients who relapse and have high-risk features, they should be started on a BTKi inhibitor for the most part,” he said, “although we need to weigh risks and benefits between the side effects of different BTKi inhibitors. And they should be referred earlier to a CAR T cell therapy center, so they can discuss the benefits and see if they’re an appropriate patient. I think patients are being referred a little bit too late in the second- or third-line setting.”

What about CAR T therapy as a first-line therapy? It’s not FDA-approved, Dr. Sandoval‐Sus said, and “definitely not a standard of care.” But clinical trials are exploring the idea, he said. As for messages to patients, Dr. Sandoval-Sus said he would tell them that MCL is not yet curable, “but the future is very bright.”

Dr. Sandoval-Sus declared advisory board relationships with Seagen, Incyte, Janssen, ADC Therapeutics, TG therapeutics, and Genmab. The other review authors had no disclosures.

 

An optimistic new review offers guidance about treating mantle cell lymphoma (MCL) in the new era of chimeric antigen receptor (CAR)-T cell therapy and medication choices informed by risk stratification.

Traditionally, MCL has had a notoriously poor prognosis and is still impossible to cure. But survival rates are rising thanks to better treatments, the review authors wrote, and even relapsed/refractory patients have a growing number of options that can potentially give them extra years of life.

“Prognosis has certainly changed in past 10 years. We have been able to have an excellent control of disease, and patients are living longer, even past the 8- or 10-year mark,” Moffit Cancer Center/Memorial Healthcare System hematologist-oncologist Jose Sandoval‐Sus, MD, said in an interview. He is corresponding author of the review, which appeared in the October issue of Current Oncology Reports.

MCL – which affects cells in the mantle zone of lymph nodes – is rare. It usually strikes older men, often presents at an advanced stage, and accounts for 6%-8% of non-Hodgkin lymphomas in the United States.

Prognoses are improving. The review highlights a study released earlier this year that found that median 5-year overall survival has increased from 68.8% (2002-2009) to 81.6% (2010-2015).

Now, the review notes, there are several first-line chemotherapy options that combine agents with rituximab such as rituximab/bendamustine, which “has generally been established as an effective treatment for MCL at first relapse in patients who are bendamustine naive when compared to other chemotherapy agents.”

Other treatments include rituximab, bortezomib, cyclophosphamide, doxorubicin, vincristine, and prednisolone; rituximab, bendamustine and cytarabine; and rituximab, gemcitabine, and oxaliplatin.

“I think of rituximab as a medication of maintenance, either after autologous stem cell transplant or even in patients who have not been through transplant,” Dr. Sandoval‐Sus said. “As maintenance, it really has improved outcomes for these patients.”

But the first step before treatment, he said, is to explore prognostic factors such as alterations on the TP53 gene that “really dictate a lot in terms of the prognosis of patients.” As the review notes, these alterations – either bi-allelic del17p or TP53 mutations – “are associated with poor outcomes after frontline and salvage regimens, including targeted agents such as Burton’s tyrosine kinase inhibitors (BTKis).”

These patients, who make up about 20% of those with MCL, also are most unlikely to benefit from autologous stem cell transplantation, Dr. Sandoval‐Sus said.

What about refracted/relapsed (R/R) cases? BTKis have been a major advance for these patients, he said. However, choosing the best drug can be a challenge. As the review notes, “all approved BTKis for R/R MCL seem to have similar clinical outcomes based on identical mechanism of action, and there are no prospective trials comparing these agents in a head-to-head fashion.”

The authors added that “we wonder if AEs [adverse events] could be decreased by using combinations based on new generation BTKi, but it is still a question that needs to be resolved in the clinical trial arena.”

Stem cell transplants may be an option, the review said, but “in practice the clinical benefit ... is limited to single-center series or small multi-institutional registries with few prospective studies.”

Then there’s CAR-T cell therapy, the game-changer. A type called brexucabtagene autoleucel (Brexu-cel) is now approved in MCL, the review authors wrote, and real-world data “serve as a platform to expand CAR-T therapy to more R/R MCL patients that do not fit the strict inclusion criteria of the studies (e.g., controlled comorbidities and worse performance status)... We strongly recommend early referral of these patients to accredited institutions with ample cellular therapy experience, including high-risk MCL patients (e.g., blastoid/pleomorphic morphology, biallelic del17p, TP53 mutations) so an appropriate bridging strategy and a CAR-T cell roadmap is planned with the patient and caretakers.”

Some researchers are exploring combination treatment with both BTKis and CAR T-cell therapy, “which may be considered for patients with R/R MCL who are naive to both CAR T-cell and BTKi therapy, because combination therapy may increase treatment efficacy,” wrote the authors of another review that appeared in the October issue of Current Oncology Reports. “Based on limited data in patients with CLL, BTKi therapy may be initiated as bridging therapy and continued during lymphodepletion prior to CAR T-cell infusion”

What’s next? Multiple treatments are in the research stage, Dr. Sandoval‐Sus said. “There are a lot of things in development that are really incredible.”

Reversible BTKis, for example, appear to be effective at controlling disease and are well-tolerated, he said. “And we are awaiting the results of clinical trials of targeted therapies.”

For now, he said, the best advice for hematologists is to gain a full understanding of a patient’s MCL, in order to provide the most appropriate treatment. Community oncologists should get at least one second opinion from an academic center or other clinic that treats these kinds of lymphomas, he said, and molecular tests are crucial. A discussion about stem cell transplantation after remission is a good idea, he said, and so is an exploration of clinical trials “from the get-go.”

“In patients who relapse and have high-risk features, they should be started on a BTKi inhibitor for the most part,” he said, “although we need to weigh risks and benefits between the side effects of different BTKi inhibitors. And they should be referred earlier to a CAR T cell therapy center, so they can discuss the benefits and see if they’re an appropriate patient. I think patients are being referred a little bit too late in the second- or third-line setting.”

What about CAR T therapy as a first-line therapy? It’s not FDA-approved, Dr. Sandoval‐Sus said, and “definitely not a standard of care.” But clinical trials are exploring the idea, he said. As for messages to patients, Dr. Sandoval-Sus said he would tell them that MCL is not yet curable, “but the future is very bright.”

Dr. Sandoval-Sus declared advisory board relationships with Seagen, Incyte, Janssen, ADC Therapeutics, TG therapeutics, and Genmab. The other review authors had no disclosures.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

FDA approved acalabrutinib tablet for MCL, CLL, SLL

Article Type
Changed
Fri, 12/16/2022 - 12:34

The Food and Drug Administration approved AstraZeneca’s new tablet formulation of acalabrutinib (Calquence) for all current indications of the capsule version.

These include adult patients with chronic lymphocytic leukemia, small lymphocytic lymphoma, and relapsed or refractory mantle cell lymphoma.

Approval of the tablet formulation of the selective Bruton tyrosine kinase inhibitor was based on the ELEVATE-PLUS trials, which showed bioequivalence with the capsule. The tablet had the same efficacy and safety profile with the same dosing strength and schedule, AstraZeneca said in a press release.

The benefit of the tablet formulation is that patients with acid reflux and other problems can take it with proton pump inhibitors, antacids, and H2-receptor antagonists, the company noted.

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

The Food and Drug Administration approved AstraZeneca’s new tablet formulation of acalabrutinib (Calquence) for all current indications of the capsule version.

These include adult patients with chronic lymphocytic leukemia, small lymphocytic lymphoma, and relapsed or refractory mantle cell lymphoma.

Approval of the tablet formulation of the selective Bruton tyrosine kinase inhibitor was based on the ELEVATE-PLUS trials, which showed bioequivalence with the capsule. The tablet had the same efficacy and safety profile with the same dosing strength and schedule, AstraZeneca said in a press release.

The benefit of the tablet formulation is that patients with acid reflux and other problems can take it with proton pump inhibitors, antacids, and H2-receptor antagonists, the company noted.

A version of this article first appeared on Medscape.com.

The Food and Drug Administration approved AstraZeneca’s new tablet formulation of acalabrutinib (Calquence) for all current indications of the capsule version.

These include adult patients with chronic lymphocytic leukemia, small lymphocytic lymphoma, and relapsed or refractory mantle cell lymphoma.

Approval of the tablet formulation of the selective Bruton tyrosine kinase inhibitor was based on the ELEVATE-PLUS trials, which showed bioequivalence with the capsule. The tablet had the same efficacy and safety profile with the same dosing strength and schedule, AstraZeneca said in a press release.

The benefit of the tablet formulation is that patients with acid reflux and other problems can take it with proton pump inhibitors, antacids, and H2-receptor antagonists, the company noted.

A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

‘Plethora’ of new MCL treatment options

Article Type
Changed
Fri, 12/16/2022 - 12:34

A “plethora of new agents” has transformed the treatment landscape for relapsed/refractory mantle cell lymphoma (R/RMCL) in recent years, according to an updated literature review that also included a proposed new treatment algorithm and identified areas for further investigation.

Specific research needs include comparative studies of novel treatment combinations like ibrutinib plus venetoclax, which has shown singular promise in clinical trials, and further investigation of emerging immunotherapies like bi-specific T-cell engagers (BiTEs), said review author Mubarak Al-Mansour, MD.

The review article, published online in Clinical Lymphoma, Myeloma & Leukemia, includes a proposed treatment algorithm based on the latest data.

“Since the introduction of [Bruton’s tyrosine kinase] inhibitors, the treatment algorithm and response of R/RMCL patients have dramatically changed. Nevertheless, Bruton's tyrosine kinase resistance is common, which necessitated further investigations to develop novel agents with a more durable response,” explained Dr. Al-Mansour a medical oncologist at Princess Noorah Oncology Center, Jeddah, Saudi Arabia.

Modest clinical activity and tolerability observed with novel agents that targeted B-cell receptor signaling led to investigation of combination strategies in preclinical and early clinical settings, in order to assess whether more durable response rates could be achieved than with single-agent therapy, he said.

“[Of] these combinations, ibrutinib plus venetoclax had the highest response rates in the setting of clinical trials, even in high-risk patients,” Dr. Al-Mansour noted.

Other promising therapies include chimeric antigen receptor (CAR) T-cell therapies (CAR-T) and BiTEs, which “appear to be powerful agents in the therapeutic arsenals of R/RMCL, especially among heavily pretreated patients,” he said, adding, however, that “further investigations are still warranted to assess the clinical activity of CAR-T or BiTEs therapies in combination with other agents.”

Comparative studies also will be needed to assess the relative advantages of various treatment approaches, he said.

These investigations are important given the generally short duration of remission among patients with MCL, which now accounts for between 2% and 6% of all non-Hodgkin lymphoma cases, an incidence that has risen steadily over the past few decades, Dr. Al-Mansour pointed out.

Although many patients achieve an adequate response in the upfront treatment setting, with overall response rates ranging from 60% to 97%, remission is generally short-lived, and the rapid relapses that occur pose a challenge. Additionally, most patients are elderly and have a poor prognosis: Reported progression-free survival in older patients ranges from 2 to 3 years and median overall survival ranges from 28.8 to 52 months, compared with 62 and 139 months, respectively, in young, fit patients, he said.

Furthermore, there is no consensus on the best treatment options in the relapsed/refractory setting, and international guidelines vary widely, he added.

For the current review, Dr. Al-Mansour conducted an online bibliographic search for relevant clinical trial data and meeting abstracts published through the end of March 2022. The data addressed treatment pathways, resistance mechanisms, various approved and investigational agents and treatments used alone or in combination regimens, and stem cell transplant (SCT).

Based on the evidence, Dr. Al-Mansour proposed the following “general algorithm” for the management of R/RMCL:

“Fit patients should be categorized according to their time until disease progression into early (< 24 months) and late (> 24 months) groups. In patients with early progression of the disease, Bruton's tyrosine kinase inhibitors should be offered. Other alternatives should be offered in case of relapse or failure, including CAR-T, [allogeneic-SCT (allo-SCT)], or enrollment in a clinical trial.”

For patients with late disease progression, the algorithm calls for offering Bruton's tyrosine kinase inhibitors, rituximab-bendamustine–based chemotherapy, or rituximab-lenalidomide.

“Other alternatives should be offered in case of relapse or failure, including CAR-T, allo-SCT, or enrollment in a clinical trial. Unfit patients can be offered Bruton's tyrosine kinase inhibitors, considering CAR-T or enrollment in a clinical trial in case of failure.”

Dr. Al-Mansour also noted COVID-19 pandemic–related caveats for the management of R/RMCL.

“Recent epidemiological figures demonstrated that cancer patients are at excessive risk of severe COVID-19. In the case of hematological malignancies, patients are usually on immunosuppressants, which further increase the risk of severe disease and death,” he wrote.

For this reason, and because current treatments consist mainly of targeted agents, which “exert negative effects on patients’ humoral and cell-mediated immunity,” the timing and schedules of treatment regimens should be determined with consideration of COVID-19–related risks, he advised.

Publications
Topics
Sections

A “plethora of new agents” has transformed the treatment landscape for relapsed/refractory mantle cell lymphoma (R/RMCL) in recent years, according to an updated literature review that also included a proposed new treatment algorithm and identified areas for further investigation.

Specific research needs include comparative studies of novel treatment combinations like ibrutinib plus venetoclax, which has shown singular promise in clinical trials, and further investigation of emerging immunotherapies like bi-specific T-cell engagers (BiTEs), said review author Mubarak Al-Mansour, MD.

The review article, published online in Clinical Lymphoma, Myeloma & Leukemia, includes a proposed treatment algorithm based on the latest data.

“Since the introduction of [Bruton’s tyrosine kinase] inhibitors, the treatment algorithm and response of R/RMCL patients have dramatically changed. Nevertheless, Bruton's tyrosine kinase resistance is common, which necessitated further investigations to develop novel agents with a more durable response,” explained Dr. Al-Mansour a medical oncologist at Princess Noorah Oncology Center, Jeddah, Saudi Arabia.

Modest clinical activity and tolerability observed with novel agents that targeted B-cell receptor signaling led to investigation of combination strategies in preclinical and early clinical settings, in order to assess whether more durable response rates could be achieved than with single-agent therapy, he said.

“[Of] these combinations, ibrutinib plus venetoclax had the highest response rates in the setting of clinical trials, even in high-risk patients,” Dr. Al-Mansour noted.

Other promising therapies include chimeric antigen receptor (CAR) T-cell therapies (CAR-T) and BiTEs, which “appear to be powerful agents in the therapeutic arsenals of R/RMCL, especially among heavily pretreated patients,” he said, adding, however, that “further investigations are still warranted to assess the clinical activity of CAR-T or BiTEs therapies in combination with other agents.”

Comparative studies also will be needed to assess the relative advantages of various treatment approaches, he said.

These investigations are important given the generally short duration of remission among patients with MCL, which now accounts for between 2% and 6% of all non-Hodgkin lymphoma cases, an incidence that has risen steadily over the past few decades, Dr. Al-Mansour pointed out.

Although many patients achieve an adequate response in the upfront treatment setting, with overall response rates ranging from 60% to 97%, remission is generally short-lived, and the rapid relapses that occur pose a challenge. Additionally, most patients are elderly and have a poor prognosis: Reported progression-free survival in older patients ranges from 2 to 3 years and median overall survival ranges from 28.8 to 52 months, compared with 62 and 139 months, respectively, in young, fit patients, he said.

Furthermore, there is no consensus on the best treatment options in the relapsed/refractory setting, and international guidelines vary widely, he added.

For the current review, Dr. Al-Mansour conducted an online bibliographic search for relevant clinical trial data and meeting abstracts published through the end of March 2022. The data addressed treatment pathways, resistance mechanisms, various approved and investigational agents and treatments used alone or in combination regimens, and stem cell transplant (SCT).

Based on the evidence, Dr. Al-Mansour proposed the following “general algorithm” for the management of R/RMCL:

“Fit patients should be categorized according to their time until disease progression into early (< 24 months) and late (> 24 months) groups. In patients with early progression of the disease, Bruton's tyrosine kinase inhibitors should be offered. Other alternatives should be offered in case of relapse or failure, including CAR-T, [allogeneic-SCT (allo-SCT)], or enrollment in a clinical trial.”

For patients with late disease progression, the algorithm calls for offering Bruton's tyrosine kinase inhibitors, rituximab-bendamustine–based chemotherapy, or rituximab-lenalidomide.

“Other alternatives should be offered in case of relapse or failure, including CAR-T, allo-SCT, or enrollment in a clinical trial. Unfit patients can be offered Bruton's tyrosine kinase inhibitors, considering CAR-T or enrollment in a clinical trial in case of failure.”

Dr. Al-Mansour also noted COVID-19 pandemic–related caveats for the management of R/RMCL.

“Recent epidemiological figures demonstrated that cancer patients are at excessive risk of severe COVID-19. In the case of hematological malignancies, patients are usually on immunosuppressants, which further increase the risk of severe disease and death,” he wrote.

For this reason, and because current treatments consist mainly of targeted agents, which “exert negative effects on patients’ humoral and cell-mediated immunity,” the timing and schedules of treatment regimens should be determined with consideration of COVID-19–related risks, he advised.

A “plethora of new agents” has transformed the treatment landscape for relapsed/refractory mantle cell lymphoma (R/RMCL) in recent years, according to an updated literature review that also included a proposed new treatment algorithm and identified areas for further investigation.

Specific research needs include comparative studies of novel treatment combinations like ibrutinib plus venetoclax, which has shown singular promise in clinical trials, and further investigation of emerging immunotherapies like bi-specific T-cell engagers (BiTEs), said review author Mubarak Al-Mansour, MD.

The review article, published online in Clinical Lymphoma, Myeloma & Leukemia, includes a proposed treatment algorithm based on the latest data.

“Since the introduction of [Bruton’s tyrosine kinase] inhibitors, the treatment algorithm and response of R/RMCL patients have dramatically changed. Nevertheless, Bruton's tyrosine kinase resistance is common, which necessitated further investigations to develop novel agents with a more durable response,” explained Dr. Al-Mansour a medical oncologist at Princess Noorah Oncology Center, Jeddah, Saudi Arabia.

Modest clinical activity and tolerability observed with novel agents that targeted B-cell receptor signaling led to investigation of combination strategies in preclinical and early clinical settings, in order to assess whether more durable response rates could be achieved than with single-agent therapy, he said.

“[Of] these combinations, ibrutinib plus venetoclax had the highest response rates in the setting of clinical trials, even in high-risk patients,” Dr. Al-Mansour noted.

Other promising therapies include chimeric antigen receptor (CAR) T-cell therapies (CAR-T) and BiTEs, which “appear to be powerful agents in the therapeutic arsenals of R/RMCL, especially among heavily pretreated patients,” he said, adding, however, that “further investigations are still warranted to assess the clinical activity of CAR-T or BiTEs therapies in combination with other agents.”

Comparative studies also will be needed to assess the relative advantages of various treatment approaches, he said.

These investigations are important given the generally short duration of remission among patients with MCL, which now accounts for between 2% and 6% of all non-Hodgkin lymphoma cases, an incidence that has risen steadily over the past few decades, Dr. Al-Mansour pointed out.

Although many patients achieve an adequate response in the upfront treatment setting, with overall response rates ranging from 60% to 97%, remission is generally short-lived, and the rapid relapses that occur pose a challenge. Additionally, most patients are elderly and have a poor prognosis: Reported progression-free survival in older patients ranges from 2 to 3 years and median overall survival ranges from 28.8 to 52 months, compared with 62 and 139 months, respectively, in young, fit patients, he said.

Furthermore, there is no consensus on the best treatment options in the relapsed/refractory setting, and international guidelines vary widely, he added.

For the current review, Dr. Al-Mansour conducted an online bibliographic search for relevant clinical trial data and meeting abstracts published through the end of March 2022. The data addressed treatment pathways, resistance mechanisms, various approved and investigational agents and treatments used alone or in combination regimens, and stem cell transplant (SCT).

Based on the evidence, Dr. Al-Mansour proposed the following “general algorithm” for the management of R/RMCL:

“Fit patients should be categorized according to their time until disease progression into early (< 24 months) and late (> 24 months) groups. In patients with early progression of the disease, Bruton's tyrosine kinase inhibitors should be offered. Other alternatives should be offered in case of relapse or failure, including CAR-T, [allogeneic-SCT (allo-SCT)], or enrollment in a clinical trial.”

For patients with late disease progression, the algorithm calls for offering Bruton's tyrosine kinase inhibitors, rituximab-bendamustine–based chemotherapy, or rituximab-lenalidomide.

“Other alternatives should be offered in case of relapse or failure, including CAR-T, allo-SCT, or enrollment in a clinical trial. Unfit patients can be offered Bruton's tyrosine kinase inhibitors, considering CAR-T or enrollment in a clinical trial in case of failure.”

Dr. Al-Mansour also noted COVID-19 pandemic–related caveats for the management of R/RMCL.

“Recent epidemiological figures demonstrated that cancer patients are at excessive risk of severe COVID-19. In the case of hematological malignancies, patients are usually on immunosuppressants, which further increase the risk of severe disease and death,” he wrote.

For this reason, and because current treatments consist mainly of targeted agents, which “exert negative effects on patients’ humoral and cell-mediated immunity,” the timing and schedules of treatment regimens should be determined with consideration of COVID-19–related risks, he advised.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Noted oncologist ponders death, life, care inequities

Article Type
Changed
Fri, 12/16/2022 - 12:34

Kashyap Patel, MD, followed an unconventional path to becoming a nationally known oncologist. A former news photographer in his native India, Dr. Patel has practiced medicine on three continents.

In 2020, he published a book aimed at cancer specialists and their patients on how to die “with hope and dignity,” titled “Between Life and Death” (Penguin Random House India).

Dr. Kashyap Patel, CEO of Carolina Blood and Cancer Care Associates in Rock Hill, S.C.
Dr. Kashyap Patel

When Dr. Patel, the CEO of Carolina Blood and Cancer Care Associates in Rock Hill, S.C., became president of the Washington-based Community Oncology Alliance 2 years ago, he stepped into a leadership role in community oncology. As an advocate for health care payment reform on Capitol Hill, the South Carolina legislature, and within his own practice, Dr. Patel has long worked to eliminate disparities in U.S. cancer care.

This news organization spoke with Dr. Patel about his unusual career path.
 

Question: Your father had a great influence on you. Can you tell us more about him?

Answer:
My dad was a hermit and a saint. He lost his dad when he was 4 years old and moved to the big city with his cousins. When he was 9 or so, he got a message saying that his mum was very ill. So, he and his cousin raised some money, got a doctor and one of those old, rugged jeeps, and they started driving to the village, but rains had destroyed the road. So, without penicillin, his mum died of pneumonia.

He felt that roads and doctor access were the two big factors that could have saved her life. He eventually became the Superintending Engineer for four districts in Gujarat State, building roads connecting every village, but he never gave up his simplistic, minimalist life.

When I was in elementary school, every other weekend my dad would literally dump me at the Mahatma Gandhi Ashram and come back in 2 hours. So, I’m looking at Gandhi’s cabinets, his pictures, reading about his life. So, my formative years were born in that.
 

Q: I read that you were intending to become an engineer and join the space race. How did your father nudge you toward medicine?

A:
When I was 9 years old, my favorite movie hero died of cancer. To comfort me, my father inserted the idea into my brain: When you grow up, you can become a doctor to cure cancer. So, when I finished high school, I was 24th in the state and had an option to go to the space school in India. On the day when I was going for the interview, I could see tears in my father’s eyes, and he said, You know what, boy? I thought you’re going to become a doctor and cure cancer. So, to honor him, I went to med school instead.

Dr. Kashyap Patel and medical school classmates traveling in India's Gujarat state in 1984.
Courtesty Dr. Kashyap Patel
Dr. Kashyap Patel and medical school classmates traveling in India's Gujarat state in 1984.

Q: I understand that your father also triggered your interest in photography?

A:
I started photographing Kutchi tribal people in 1977, after I bought a camera from a famous architect [Hasmukh Patel], while traveling with my dad. And then my dad bought me a motorcycle, so I started riding myself. From the time I entered med school in 1978 until I finished my residency in 1987, I made several trips following Kutchi migrant families and livestock. They leave their homeland in Kutch [district] during summer in search of grass and water to keep their livestock alive and walk across the state from the desert of Kutch all the way to central Gujarat until monsoon begins. Then they return, only to resume the journey next year. I would catch them along their journey, would talk to them, drink tea and eat millet crepes with them.

Migrant woman in India, photographed by Dr. Patel when he worked as a journalist early in his career.
Courtesy Dr. Kashyap Patel
Migrant woman in India, photographed by Dr. Patel when he worked as a journalist early in his career.

In 1984, between Dr. Patel’s medical school and residency, the Lions Club in his hometown, Ahmedabad, India, sponsored him and three buddies to document people and wildlife in Gujarat state. Traveling by motorcycle, the four friends stayed for free with local families by knocking on doors and explaining that they were medical students. Dr. Patel’s photographs were exhibited by the Lions Club of Ahmedabad and at India’s top art institution, the Lalit Kala gallery.

In the 3rd year of his internal-medicine residency in Bombay (now Mumbai), Dr. Patel approached a national newspaper, The Indian Express, for work. He was immediately sent on assignment to cover a cholera epidemic and filed his story and photographs the following day. He worked as a photojournalist and subeditor for a year.
 

Migrant family in India's Kutch district, photographed by Dr. Patel when he worked as a journalist early in his career.
Courtesy Dr. Kashyap Patel
Migrant family in India's Kutch district, photographed by Dr. Patel when he worked as a journalist early in his career.

Q: Among all your thousands of pictures, do you have a favorite?

A:
There were two photos of Kutchi people that touched me. There was one photo of a lady. All of her worldly belongings were in the picture and a smile on her face showed that we don’t need so many things to be happy. The second photo is of an elderly lady shifting her water pan on her head to a younger family member. And a little girl looks up with a look of curiosity: Will I be doing this when I grow up? We seek so much materialistic happiness. But when you look at the curiosity, smiles, and happiness [in these photos], you realize we could have a lot of happiness in minimalism, as well.

Q: After you finished your residency in Ahmedabad, how did you get started in oncology?

A:
In 1986, Ahmedabad City and Gujarat State did not have structured training programs in oncology, so I went to Bombay [Mumbai], where Dr. B.C. Mehta, a true legend and pioneer in India, had started hematology-oncology training. I was a post-doc research fellow with him for a little over a year but when I started seeing patients, I had to answer to myself, Am I doing everything I can to help these people? I saw that the U.K. had one of the best training programs in hem malignancy, so I started applying. Then something happened that was almost like a miracle.

In April 1992, Dr. Patel was working at the Institute of Kidney Diseases in Ahmedabad. One afternoon, just as the clinic was closing for siesta, a family brought in a young girl. She had drug-induced thrombocytopenia and needed an immediate transfusion. The father offered to sell his wedding ring to pay Dr. Patel if he would supervise the treatment and stay by the girl’s side. Dr. Patel told the man to keep his ring, then he remained in the office with the child. At 4 p.m., the office phone rang. It was Dr. H.K. Parikh, an eminent British physician who was wintering in India and needed to make a medical appointment for his wife. On a normal day, Dr. Patel would have missed the call.

 

 

“This is how I got to meet Dr. Parikh, out of the blue,” said Dr. Patel. “His wife came to the office for 6 weeks and after 6 weeks, he said, You’re a smart guy; you should come to England. That was in April. I sent a resume and all the usual paperwork. On July 16, 1992, at 2 in the morning, I got a call from the U.K. saying, Your job is confirmed. I’m going to fax your appointment through the Royal College of Physicians, and you’re coming to Manchester to work with us. I’d been sponsored by the Overseas Doctors Training Program.

“So, it turns out that if I’d declined to see that patient and declined to stay in my clinic that afternoon, if I’d declined to see this doctor’s wife, I would never have been in the U.K. And that opened up the doors for me. I like that story because I’ve found that standing up for people who do not have a voice, who do not have hope, always leads to what is destined for me.”
 

Q: After working as a registrar in the United Kingdom 4 years, you found yourself in the United States and, once again, had to train as an internist. What was new about U.S. oncology?

A:
I took 3 years to get recertified in Jamaica Hospital in Queens, then became a fellow in hematology-oncology at the Thomas Jefferson in Philadelphia. My U.K. training was all based on hematological malignancy. In the United States, I shifted into solid tumors.

Q: You have a long history of advocating for affordable oncology at the community, state, and federal level, and you recently launched a disparities initiative in your center called NOLA (No One Left Alone). What was the trigger for NOLA?

A:
In the spring of 2020, when we started seeing the COVID surge and the difference in mortality rate between the multiple races, at the same time I saw the AACR [American Association for Cancer Research] 2020 disparity report showing that 34% of cancer deaths are preventable – one in three – if we took care of disparities. The same year, the Community Oncology Alliance asked me to become the president. So, I felt that there is something herding me, leading me, to this position. Eighty percent of cancer patients are treated in community clinics like ours. It put the onus on me to do something.

I learned from Gandhi that I cannot depend on government, I cannot depend on the policy, I have to act myself.

I said, I would not worry about making money, I would rather lose funding on this. So, we started. I read 400+ papers; I spent over 1,000 hours reading about disparities. And I realized that it’s not complicated. There are five pillars to eliminate disparity: access to care for financial reasons, access to biomarker testing or precision medicine, access to social determinants of health, access to cancer screening, and trials. If we focus on these five, we can at least bring that number from 34% to 20%, if not lower.

So, we put that plan in place. I dedicated three employees whose only role is to ensure that not a single patient has to take financial burden from my practice. And we showed it’s doable.

This has now become my mission for the last quarter of my life.
 

 

 

In 2020, Dr. Patel published a book on dying well titled “Between Life and Death.” It’s framed as a series of his conversations with a former patient, Harry Falls. Harry wanted to understand death better, so Dr. Patel narrated five patient stories, drawing the threads together to help Harry face the inevitable. Dr. Patel now uses a similar approach to train clinicians on having meaningful end-of-life conversations with patients.

Dr. Kashyap Patel's book, &amp;quot;Between Life and Death,&amp;quot; published in 2020.
Courtesy Dr. Kashyap Patel
Dr. Kashyap Patel's book, published in 2020.

Q: Why did you feel the need to write a book about dying?

A:
The more I’ve witnessed, the more I’m convinced that there are things that we don’t know about this process, which needs to be explored much more. However, I do feel that there’s a power within all of us to steer the process of leaving this world.

Before I sat down with Harry, I loved to counsel patients, but I didn’t have any structural ideas. It was Harry himself who told me that I now had a simple way to explain dying to a much larger audience.
 

Q: What is your secret for fitting everything into your life?

A:
I’ll tell you, it’s very simple. If I put my soul, heart, mind, actions, and language on the one plane and don’t let my brain and conditioning influence my choices, then I live in the moment. Whenever I let my conditioned mind take all the decisions, those are crooked, because you know, we’re selfish creatures – we can use what we call the convenient lie to hide inconvenient truth. And I try not to do that. I mean, it’s been a journey. It didn’t come overnight. I learned. And I feel that over all these years, the only thing that rewarded me, that opened the door of where I am today, was pure, selfless process, whether it’s the act of talking, speaking, or doing.

Publications
Topics
Sections

Kashyap Patel, MD, followed an unconventional path to becoming a nationally known oncologist. A former news photographer in his native India, Dr. Patel has practiced medicine on three continents.

In 2020, he published a book aimed at cancer specialists and their patients on how to die “with hope and dignity,” titled “Between Life and Death” (Penguin Random House India).

Dr. Kashyap Patel, CEO of Carolina Blood and Cancer Care Associates in Rock Hill, S.C.
Dr. Kashyap Patel

When Dr. Patel, the CEO of Carolina Blood and Cancer Care Associates in Rock Hill, S.C., became president of the Washington-based Community Oncology Alliance 2 years ago, he stepped into a leadership role in community oncology. As an advocate for health care payment reform on Capitol Hill, the South Carolina legislature, and within his own practice, Dr. Patel has long worked to eliminate disparities in U.S. cancer care.

This news organization spoke with Dr. Patel about his unusual career path.
 

Question: Your father had a great influence on you. Can you tell us more about him?

Answer:
My dad was a hermit and a saint. He lost his dad when he was 4 years old and moved to the big city with his cousins. When he was 9 or so, he got a message saying that his mum was very ill. So, he and his cousin raised some money, got a doctor and one of those old, rugged jeeps, and they started driving to the village, but rains had destroyed the road. So, without penicillin, his mum died of pneumonia.

He felt that roads and doctor access were the two big factors that could have saved her life. He eventually became the Superintending Engineer for four districts in Gujarat State, building roads connecting every village, but he never gave up his simplistic, minimalist life.

When I was in elementary school, every other weekend my dad would literally dump me at the Mahatma Gandhi Ashram and come back in 2 hours. So, I’m looking at Gandhi’s cabinets, his pictures, reading about his life. So, my formative years were born in that.
 

Q: I read that you were intending to become an engineer and join the space race. How did your father nudge you toward medicine?

A:
When I was 9 years old, my favorite movie hero died of cancer. To comfort me, my father inserted the idea into my brain: When you grow up, you can become a doctor to cure cancer. So, when I finished high school, I was 24th in the state and had an option to go to the space school in India. On the day when I was going for the interview, I could see tears in my father’s eyes, and he said, You know what, boy? I thought you’re going to become a doctor and cure cancer. So, to honor him, I went to med school instead.

Dr. Kashyap Patel and medical school classmates traveling in India's Gujarat state in 1984.
Courtesty Dr. Kashyap Patel
Dr. Kashyap Patel and medical school classmates traveling in India's Gujarat state in 1984.

Q: I understand that your father also triggered your interest in photography?

A:
I started photographing Kutchi tribal people in 1977, after I bought a camera from a famous architect [Hasmukh Patel], while traveling with my dad. And then my dad bought me a motorcycle, so I started riding myself. From the time I entered med school in 1978 until I finished my residency in 1987, I made several trips following Kutchi migrant families and livestock. They leave their homeland in Kutch [district] during summer in search of grass and water to keep their livestock alive and walk across the state from the desert of Kutch all the way to central Gujarat until monsoon begins. Then they return, only to resume the journey next year. I would catch them along their journey, would talk to them, drink tea and eat millet crepes with them.

Migrant woman in India, photographed by Dr. Patel when he worked as a journalist early in his career.
Courtesy Dr. Kashyap Patel
Migrant woman in India, photographed by Dr. Patel when he worked as a journalist early in his career.

In 1984, between Dr. Patel’s medical school and residency, the Lions Club in his hometown, Ahmedabad, India, sponsored him and three buddies to document people and wildlife in Gujarat state. Traveling by motorcycle, the four friends stayed for free with local families by knocking on doors and explaining that they were medical students. Dr. Patel’s photographs were exhibited by the Lions Club of Ahmedabad and at India’s top art institution, the Lalit Kala gallery.

In the 3rd year of his internal-medicine residency in Bombay (now Mumbai), Dr. Patel approached a national newspaper, The Indian Express, for work. He was immediately sent on assignment to cover a cholera epidemic and filed his story and photographs the following day. He worked as a photojournalist and subeditor for a year.
 

Migrant family in India's Kutch district, photographed by Dr. Patel when he worked as a journalist early in his career.
Courtesy Dr. Kashyap Patel
Migrant family in India's Kutch district, photographed by Dr. Patel when he worked as a journalist early in his career.

Q: Among all your thousands of pictures, do you have a favorite?

A:
There were two photos of Kutchi people that touched me. There was one photo of a lady. All of her worldly belongings were in the picture and a smile on her face showed that we don’t need so many things to be happy. The second photo is of an elderly lady shifting her water pan on her head to a younger family member. And a little girl looks up with a look of curiosity: Will I be doing this when I grow up? We seek so much materialistic happiness. But when you look at the curiosity, smiles, and happiness [in these photos], you realize we could have a lot of happiness in minimalism, as well.

Q: After you finished your residency in Ahmedabad, how did you get started in oncology?

A:
In 1986, Ahmedabad City and Gujarat State did not have structured training programs in oncology, so I went to Bombay [Mumbai], where Dr. B.C. Mehta, a true legend and pioneer in India, had started hematology-oncology training. I was a post-doc research fellow with him for a little over a year but when I started seeing patients, I had to answer to myself, Am I doing everything I can to help these people? I saw that the U.K. had one of the best training programs in hem malignancy, so I started applying. Then something happened that was almost like a miracle.

In April 1992, Dr. Patel was working at the Institute of Kidney Diseases in Ahmedabad. One afternoon, just as the clinic was closing for siesta, a family brought in a young girl. She had drug-induced thrombocytopenia and needed an immediate transfusion. The father offered to sell his wedding ring to pay Dr. Patel if he would supervise the treatment and stay by the girl’s side. Dr. Patel told the man to keep his ring, then he remained in the office with the child. At 4 p.m., the office phone rang. It was Dr. H.K. Parikh, an eminent British physician who was wintering in India and needed to make a medical appointment for his wife. On a normal day, Dr. Patel would have missed the call.

 

 

“This is how I got to meet Dr. Parikh, out of the blue,” said Dr. Patel. “His wife came to the office for 6 weeks and after 6 weeks, he said, You’re a smart guy; you should come to England. That was in April. I sent a resume and all the usual paperwork. On July 16, 1992, at 2 in the morning, I got a call from the U.K. saying, Your job is confirmed. I’m going to fax your appointment through the Royal College of Physicians, and you’re coming to Manchester to work with us. I’d been sponsored by the Overseas Doctors Training Program.

“So, it turns out that if I’d declined to see that patient and declined to stay in my clinic that afternoon, if I’d declined to see this doctor’s wife, I would never have been in the U.K. And that opened up the doors for me. I like that story because I’ve found that standing up for people who do not have a voice, who do not have hope, always leads to what is destined for me.”
 

Q: After working as a registrar in the United Kingdom 4 years, you found yourself in the United States and, once again, had to train as an internist. What was new about U.S. oncology?

A:
I took 3 years to get recertified in Jamaica Hospital in Queens, then became a fellow in hematology-oncology at the Thomas Jefferson in Philadelphia. My U.K. training was all based on hematological malignancy. In the United States, I shifted into solid tumors.

Q: You have a long history of advocating for affordable oncology at the community, state, and federal level, and you recently launched a disparities initiative in your center called NOLA (No One Left Alone). What was the trigger for NOLA?

A:
In the spring of 2020, when we started seeing the COVID surge and the difference in mortality rate between the multiple races, at the same time I saw the AACR [American Association for Cancer Research] 2020 disparity report showing that 34% of cancer deaths are preventable – one in three – if we took care of disparities. The same year, the Community Oncology Alliance asked me to become the president. So, I felt that there is something herding me, leading me, to this position. Eighty percent of cancer patients are treated in community clinics like ours. It put the onus on me to do something.

I learned from Gandhi that I cannot depend on government, I cannot depend on the policy, I have to act myself.

I said, I would not worry about making money, I would rather lose funding on this. So, we started. I read 400+ papers; I spent over 1,000 hours reading about disparities. And I realized that it’s not complicated. There are five pillars to eliminate disparity: access to care for financial reasons, access to biomarker testing or precision medicine, access to social determinants of health, access to cancer screening, and trials. If we focus on these five, we can at least bring that number from 34% to 20%, if not lower.

So, we put that plan in place. I dedicated three employees whose only role is to ensure that not a single patient has to take financial burden from my practice. And we showed it’s doable.

This has now become my mission for the last quarter of my life.
 

 

 

In 2020, Dr. Patel published a book on dying well titled “Between Life and Death.” It’s framed as a series of his conversations with a former patient, Harry Falls. Harry wanted to understand death better, so Dr. Patel narrated five patient stories, drawing the threads together to help Harry face the inevitable. Dr. Patel now uses a similar approach to train clinicians on having meaningful end-of-life conversations with patients.

Dr. Kashyap Patel's book, &amp;quot;Between Life and Death,&amp;quot; published in 2020.
Courtesy Dr. Kashyap Patel
Dr. Kashyap Patel's book, published in 2020.

Q: Why did you feel the need to write a book about dying?

A:
The more I’ve witnessed, the more I’m convinced that there are things that we don’t know about this process, which needs to be explored much more. However, I do feel that there’s a power within all of us to steer the process of leaving this world.

Before I sat down with Harry, I loved to counsel patients, but I didn’t have any structural ideas. It was Harry himself who told me that I now had a simple way to explain dying to a much larger audience.
 

Q: What is your secret for fitting everything into your life?

A:
I’ll tell you, it’s very simple. If I put my soul, heart, mind, actions, and language on the one plane and don’t let my brain and conditioning influence my choices, then I live in the moment. Whenever I let my conditioned mind take all the decisions, those are crooked, because you know, we’re selfish creatures – we can use what we call the convenient lie to hide inconvenient truth. And I try not to do that. I mean, it’s been a journey. It didn’t come overnight. I learned. And I feel that over all these years, the only thing that rewarded me, that opened the door of where I am today, was pure, selfless process, whether it’s the act of talking, speaking, or doing.

Kashyap Patel, MD, followed an unconventional path to becoming a nationally known oncologist. A former news photographer in his native India, Dr. Patel has practiced medicine on three continents.

In 2020, he published a book aimed at cancer specialists and their patients on how to die “with hope and dignity,” titled “Between Life and Death” (Penguin Random House India).

Dr. Kashyap Patel, CEO of Carolina Blood and Cancer Care Associates in Rock Hill, S.C.
Dr. Kashyap Patel

When Dr. Patel, the CEO of Carolina Blood and Cancer Care Associates in Rock Hill, S.C., became president of the Washington-based Community Oncology Alliance 2 years ago, he stepped into a leadership role in community oncology. As an advocate for health care payment reform on Capitol Hill, the South Carolina legislature, and within his own practice, Dr. Patel has long worked to eliminate disparities in U.S. cancer care.

This news organization spoke with Dr. Patel about his unusual career path.
 

Question: Your father had a great influence on you. Can you tell us more about him?

Answer:
My dad was a hermit and a saint. He lost his dad when he was 4 years old and moved to the big city with his cousins. When he was 9 or so, he got a message saying that his mum was very ill. So, he and his cousin raised some money, got a doctor and one of those old, rugged jeeps, and they started driving to the village, but rains had destroyed the road. So, without penicillin, his mum died of pneumonia.

He felt that roads and doctor access were the two big factors that could have saved her life. He eventually became the Superintending Engineer for four districts in Gujarat State, building roads connecting every village, but he never gave up his simplistic, minimalist life.

When I was in elementary school, every other weekend my dad would literally dump me at the Mahatma Gandhi Ashram and come back in 2 hours. So, I’m looking at Gandhi’s cabinets, his pictures, reading about his life. So, my formative years were born in that.
 

Q: I read that you were intending to become an engineer and join the space race. How did your father nudge you toward medicine?

A:
When I was 9 years old, my favorite movie hero died of cancer. To comfort me, my father inserted the idea into my brain: When you grow up, you can become a doctor to cure cancer. So, when I finished high school, I was 24th in the state and had an option to go to the space school in India. On the day when I was going for the interview, I could see tears in my father’s eyes, and he said, You know what, boy? I thought you’re going to become a doctor and cure cancer. So, to honor him, I went to med school instead.

Dr. Kashyap Patel and medical school classmates traveling in India's Gujarat state in 1984.
Courtesty Dr. Kashyap Patel
Dr. Kashyap Patel and medical school classmates traveling in India's Gujarat state in 1984.

Q: I understand that your father also triggered your interest in photography?

A:
I started photographing Kutchi tribal people in 1977, after I bought a camera from a famous architect [Hasmukh Patel], while traveling with my dad. And then my dad bought me a motorcycle, so I started riding myself. From the time I entered med school in 1978 until I finished my residency in 1987, I made several trips following Kutchi migrant families and livestock. They leave their homeland in Kutch [district] during summer in search of grass and water to keep their livestock alive and walk across the state from the desert of Kutch all the way to central Gujarat until monsoon begins. Then they return, only to resume the journey next year. I would catch them along their journey, would talk to them, drink tea and eat millet crepes with them.

Migrant woman in India, photographed by Dr. Patel when he worked as a journalist early in his career.
Courtesy Dr. Kashyap Patel
Migrant woman in India, photographed by Dr. Patel when he worked as a journalist early in his career.

In 1984, between Dr. Patel’s medical school and residency, the Lions Club in his hometown, Ahmedabad, India, sponsored him and three buddies to document people and wildlife in Gujarat state. Traveling by motorcycle, the four friends stayed for free with local families by knocking on doors and explaining that they were medical students. Dr. Patel’s photographs were exhibited by the Lions Club of Ahmedabad and at India’s top art institution, the Lalit Kala gallery.

In the 3rd year of his internal-medicine residency in Bombay (now Mumbai), Dr. Patel approached a national newspaper, The Indian Express, for work. He was immediately sent on assignment to cover a cholera epidemic and filed his story and photographs the following day. He worked as a photojournalist and subeditor for a year.
 

Migrant family in India's Kutch district, photographed by Dr. Patel when he worked as a journalist early in his career.
Courtesy Dr. Kashyap Patel
Migrant family in India's Kutch district, photographed by Dr. Patel when he worked as a journalist early in his career.

Q: Among all your thousands of pictures, do you have a favorite?

A:
There were two photos of Kutchi people that touched me. There was one photo of a lady. All of her worldly belongings were in the picture and a smile on her face showed that we don’t need so many things to be happy. The second photo is of an elderly lady shifting her water pan on her head to a younger family member. And a little girl looks up with a look of curiosity: Will I be doing this when I grow up? We seek so much materialistic happiness. But when you look at the curiosity, smiles, and happiness [in these photos], you realize we could have a lot of happiness in minimalism, as well.

Q: After you finished your residency in Ahmedabad, how did you get started in oncology?

A:
In 1986, Ahmedabad City and Gujarat State did not have structured training programs in oncology, so I went to Bombay [Mumbai], where Dr. B.C. Mehta, a true legend and pioneer in India, had started hematology-oncology training. I was a post-doc research fellow with him for a little over a year but when I started seeing patients, I had to answer to myself, Am I doing everything I can to help these people? I saw that the U.K. had one of the best training programs in hem malignancy, so I started applying. Then something happened that was almost like a miracle.

In April 1992, Dr. Patel was working at the Institute of Kidney Diseases in Ahmedabad. One afternoon, just as the clinic was closing for siesta, a family brought in a young girl. She had drug-induced thrombocytopenia and needed an immediate transfusion. The father offered to sell his wedding ring to pay Dr. Patel if he would supervise the treatment and stay by the girl’s side. Dr. Patel told the man to keep his ring, then he remained in the office with the child. At 4 p.m., the office phone rang. It was Dr. H.K. Parikh, an eminent British physician who was wintering in India and needed to make a medical appointment for his wife. On a normal day, Dr. Patel would have missed the call.

 

 

“This is how I got to meet Dr. Parikh, out of the blue,” said Dr. Patel. “His wife came to the office for 6 weeks and after 6 weeks, he said, You’re a smart guy; you should come to England. That was in April. I sent a resume and all the usual paperwork. On July 16, 1992, at 2 in the morning, I got a call from the U.K. saying, Your job is confirmed. I’m going to fax your appointment through the Royal College of Physicians, and you’re coming to Manchester to work with us. I’d been sponsored by the Overseas Doctors Training Program.

“So, it turns out that if I’d declined to see that patient and declined to stay in my clinic that afternoon, if I’d declined to see this doctor’s wife, I would never have been in the U.K. And that opened up the doors for me. I like that story because I’ve found that standing up for people who do not have a voice, who do not have hope, always leads to what is destined for me.”
 

Q: After working as a registrar in the United Kingdom 4 years, you found yourself in the United States and, once again, had to train as an internist. What was new about U.S. oncology?

A:
I took 3 years to get recertified in Jamaica Hospital in Queens, then became a fellow in hematology-oncology at the Thomas Jefferson in Philadelphia. My U.K. training was all based on hematological malignancy. In the United States, I shifted into solid tumors.

Q: You have a long history of advocating for affordable oncology at the community, state, and federal level, and you recently launched a disparities initiative in your center called NOLA (No One Left Alone). What was the trigger for NOLA?

A:
In the spring of 2020, when we started seeing the COVID surge and the difference in mortality rate between the multiple races, at the same time I saw the AACR [American Association for Cancer Research] 2020 disparity report showing that 34% of cancer deaths are preventable – one in three – if we took care of disparities. The same year, the Community Oncology Alliance asked me to become the president. So, I felt that there is something herding me, leading me, to this position. Eighty percent of cancer patients are treated in community clinics like ours. It put the onus on me to do something.

I learned from Gandhi that I cannot depend on government, I cannot depend on the policy, I have to act myself.

I said, I would not worry about making money, I would rather lose funding on this. So, we started. I read 400+ papers; I spent over 1,000 hours reading about disparities. And I realized that it’s not complicated. There are five pillars to eliminate disparity: access to care for financial reasons, access to biomarker testing or precision medicine, access to social determinants of health, access to cancer screening, and trials. If we focus on these five, we can at least bring that number from 34% to 20%, if not lower.

So, we put that plan in place. I dedicated three employees whose only role is to ensure that not a single patient has to take financial burden from my practice. And we showed it’s doable.

This has now become my mission for the last quarter of my life.
 

 

 

In 2020, Dr. Patel published a book on dying well titled “Between Life and Death.” It’s framed as a series of his conversations with a former patient, Harry Falls. Harry wanted to understand death better, so Dr. Patel narrated five patient stories, drawing the threads together to help Harry face the inevitable. Dr. Patel now uses a similar approach to train clinicians on having meaningful end-of-life conversations with patients.

Dr. Kashyap Patel's book, &amp;quot;Between Life and Death,&amp;quot; published in 2020.
Courtesy Dr. Kashyap Patel
Dr. Kashyap Patel's book, published in 2020.

Q: Why did you feel the need to write a book about dying?

A:
The more I’ve witnessed, the more I’m convinced that there are things that we don’t know about this process, which needs to be explored much more. However, I do feel that there’s a power within all of us to steer the process of leaving this world.

Before I sat down with Harry, I loved to counsel patients, but I didn’t have any structural ideas. It was Harry himself who told me that I now had a simple way to explain dying to a much larger audience.
 

Q: What is your secret for fitting everything into your life?

A:
I’ll tell you, it’s very simple. If I put my soul, heart, mind, actions, and language on the one plane and don’t let my brain and conditioning influence my choices, then I live in the moment. Whenever I let my conditioned mind take all the decisions, those are crooked, because you know, we’re selfish creatures – we can use what we call the convenient lie to hide inconvenient truth. And I try not to do that. I mean, it’s been a journey. It didn’t come overnight. I learned. And I feel that over all these years, the only thing that rewarded me, that opened the door of where I am today, was pure, selfless process, whether it’s the act of talking, speaking, or doing.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

First drug therapy approved for childhood GVHD

Article Type
Changed
Fri, 12/16/2022 - 12:34

Ibrutinib (Imbruvica) is now available for use in children aged 1-12 years who have chronic graft-versus-host disease (cGVHD), which can develop after stem cell transplantation for treatment of a blood cancer.

Specifically, the indication is for pediatric patients with cGVHD who have already been treated with one or more lines of systemic therapy. The manufacturers have also launched a new oral suspension formulation, in addition to capsules and tablets, which were already available.

Ibrutinib is already approved for use in adults with cGVHD.

The drug is also approved for use in several blood cancers, including chronic lymphocytic leukemiamantle cell lymphoma, and Waldenström’s macroglobulinemia. All these approvals are for adult patients.

This is the first pediatric indication for the product and is “incredibly meaningful,” said Gauri Sunkersett, DO, associate medical director at AbbVie, which markets the drug together with Jansen. “As a pediatric oncologist, when my patients describe the physical pain they experience from simply hugging their parents, due to their cGVHD, the importance of researching alternative treatment options in this patient population is further validated.”

These children have already been through a lot, having been diagnosed with a leukemia or lymphoma and then undergoing chemotherapy and/or radiotherapy for a stem cell transplant. Just over half (52%-65%) of children who receive allogeneic transplants go on to develop cGVHD, in which the donor bone marrow or stem cells attack the recipient.

“Imagine going through a transplant and then being told you have a moderate to severe chronic disease that can sometimes also be life-threatening,” commented Paul A. Carpenter, MD, attending physician at Seattle Children’s Hospital. “If these children were between 1 and 12 and didn’t respond to steroid treatment, we didn’t have any rigorously studied treatment options – until now.”

The new indication was approved by the U.S. Food and Drug Administration on the basis of results from the iMAGINE trial, for which Dr. Carpenter was a principal investigator.

The phase 1/2 iMAGINE trial was an open-label, multicenter, single-arm trial conducted with 47 patients (mean age, 13 years; range, 1-19 years) with relapsed/refractory cGVHD who had received at least one prior systemic therapy. Ibrutinib was given at a dose of 420 mg orally once daily to patients aged 12 and older and at a dose of 240 mg/m2 orally once daily to patients who were younger than 12 years.

The overall response rate through week 25 was 60% (confidence interval, 95%, 44%-74%). The median duration of response was 5.3 months (95% CI, 2.8-8.8).

The safety profile was consistent with the established profile for ibrutinib. Observed adverse events in pediatric patients were consistent with those observed in adult patients with moderate to severe cGVHD, the companies noted.

The FDA noted that the most common (≥ 20%) adverse reactions, including laboratory abnormalities, were anemia, musculoskeletal pain, pyrexia, diarrhea, pneumonia, abdominal pain, stomatitis, thrombocytopenia, and headache.

Full prescribing information for ibrutinib is available here.

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

Ibrutinib (Imbruvica) is now available for use in children aged 1-12 years who have chronic graft-versus-host disease (cGVHD), which can develop after stem cell transplantation for treatment of a blood cancer.

Specifically, the indication is for pediatric patients with cGVHD who have already been treated with one or more lines of systemic therapy. The manufacturers have also launched a new oral suspension formulation, in addition to capsules and tablets, which were already available.

Ibrutinib is already approved for use in adults with cGVHD.

The drug is also approved for use in several blood cancers, including chronic lymphocytic leukemiamantle cell lymphoma, and Waldenström’s macroglobulinemia. All these approvals are for adult patients.

This is the first pediatric indication for the product and is “incredibly meaningful,” said Gauri Sunkersett, DO, associate medical director at AbbVie, which markets the drug together with Jansen. “As a pediatric oncologist, when my patients describe the physical pain they experience from simply hugging their parents, due to their cGVHD, the importance of researching alternative treatment options in this patient population is further validated.”

These children have already been through a lot, having been diagnosed with a leukemia or lymphoma and then undergoing chemotherapy and/or radiotherapy for a stem cell transplant. Just over half (52%-65%) of children who receive allogeneic transplants go on to develop cGVHD, in which the donor bone marrow or stem cells attack the recipient.

“Imagine going through a transplant and then being told you have a moderate to severe chronic disease that can sometimes also be life-threatening,” commented Paul A. Carpenter, MD, attending physician at Seattle Children’s Hospital. “If these children were between 1 and 12 and didn’t respond to steroid treatment, we didn’t have any rigorously studied treatment options – until now.”

The new indication was approved by the U.S. Food and Drug Administration on the basis of results from the iMAGINE trial, for which Dr. Carpenter was a principal investigator.

The phase 1/2 iMAGINE trial was an open-label, multicenter, single-arm trial conducted with 47 patients (mean age, 13 years; range, 1-19 years) with relapsed/refractory cGVHD who had received at least one prior systemic therapy. Ibrutinib was given at a dose of 420 mg orally once daily to patients aged 12 and older and at a dose of 240 mg/m2 orally once daily to patients who were younger than 12 years.

The overall response rate through week 25 was 60% (confidence interval, 95%, 44%-74%). The median duration of response was 5.3 months (95% CI, 2.8-8.8).

The safety profile was consistent with the established profile for ibrutinib. Observed adverse events in pediatric patients were consistent with those observed in adult patients with moderate to severe cGVHD, the companies noted.

The FDA noted that the most common (≥ 20%) adverse reactions, including laboratory abnormalities, were anemia, musculoskeletal pain, pyrexia, diarrhea, pneumonia, abdominal pain, stomatitis, thrombocytopenia, and headache.

Full prescribing information for ibrutinib is available here.

A version of this article first appeared on Medscape.com.

Ibrutinib (Imbruvica) is now available for use in children aged 1-12 years who have chronic graft-versus-host disease (cGVHD), which can develop after stem cell transplantation for treatment of a blood cancer.

Specifically, the indication is for pediatric patients with cGVHD who have already been treated with one or more lines of systemic therapy. The manufacturers have also launched a new oral suspension formulation, in addition to capsules and tablets, which were already available.

Ibrutinib is already approved for use in adults with cGVHD.

The drug is also approved for use in several blood cancers, including chronic lymphocytic leukemiamantle cell lymphoma, and Waldenström’s macroglobulinemia. All these approvals are for adult patients.

This is the first pediatric indication for the product and is “incredibly meaningful,” said Gauri Sunkersett, DO, associate medical director at AbbVie, which markets the drug together with Jansen. “As a pediatric oncologist, when my patients describe the physical pain they experience from simply hugging their parents, due to their cGVHD, the importance of researching alternative treatment options in this patient population is further validated.”

These children have already been through a lot, having been diagnosed with a leukemia or lymphoma and then undergoing chemotherapy and/or radiotherapy for a stem cell transplant. Just over half (52%-65%) of children who receive allogeneic transplants go on to develop cGVHD, in which the donor bone marrow or stem cells attack the recipient.

“Imagine going through a transplant and then being told you have a moderate to severe chronic disease that can sometimes also be life-threatening,” commented Paul A. Carpenter, MD, attending physician at Seattle Children’s Hospital. “If these children were between 1 and 12 and didn’t respond to steroid treatment, we didn’t have any rigorously studied treatment options – until now.”

The new indication was approved by the U.S. Food and Drug Administration on the basis of results from the iMAGINE trial, for which Dr. Carpenter was a principal investigator.

The phase 1/2 iMAGINE trial was an open-label, multicenter, single-arm trial conducted with 47 patients (mean age, 13 years; range, 1-19 years) with relapsed/refractory cGVHD who had received at least one prior systemic therapy. Ibrutinib was given at a dose of 420 mg orally once daily to patients aged 12 and older and at a dose of 240 mg/m2 orally once daily to patients who were younger than 12 years.

The overall response rate through week 25 was 60% (confidence interval, 95%, 44%-74%). The median duration of response was 5.3 months (95% CI, 2.8-8.8).

The safety profile was consistent with the established profile for ibrutinib. Observed adverse events in pediatric patients were consistent with those observed in adult patients with moderate to severe cGVHD, the companies noted.

The FDA noted that the most common (≥ 20%) adverse reactions, including laboratory abnormalities, were anemia, musculoskeletal pain, pyrexia, diarrhea, pneumonia, abdominal pain, stomatitis, thrombocytopenia, and headache.

Full prescribing information for ibrutinib is available here.

A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Phase 3 data: Zanubrutinib bests standard CLL treatment

Article Type
Changed
Fri, 12/16/2022 - 12:34

The second-generation selective Bruton tyrosine kinase inhibitor (BTKi) zanubrutinib outperformed the standard treatment bendamustine-rituximab in untreated chronic lymphocytic leukemia (CLL) and small lymphocytic lymphoma (SLL), a new industry-funded phase-3 trial found.

At a median follow-up of 26.2 months, progression to worsening disease or death was much lower in patients with these conditions who took zanubrutinib (Brukinsa), compared with those who took bendamustine-rituximab (hazard ratio. 0.42; 95% confidence interval, 0.28-0.63; P < .00011). The study was published in The Lancet Oncology.

Researchers already knew that ibrutinib, another BTKi, improves progression-free survival, study coauthor Paolo Ghia, MD, PhD, professor of medical oncology at Vita-Salute San Raffaele University, Milan, said in an interview. “Now we confirmed that the same advantage can be seen” in zanubrutinib.

According to Dr. Ghia, bendamustine-rituximab has long been a standard treatment in blood cancers and is considered well tolerated and inexpensive. But BTKis such as first-in-line ibrutinib have shown better results, he said, “and progressively, we are going to abandon bendamustine-rituximab.”

However, ibrutinib causes significant adverse effects such as bleeding, worsening hypertension and arrhythmia, he noted. As a result, second-generation BTKi such as zanubrutinib have entered the picture. The Food and Drug Administration approved it in 2019 for mantle cell lymphoma, and it has since been approved for Waldenström’s macroglobulinemia and marginal zone lymphoma.

In 2021, an interim analysis in a trial of the drug in patients with previously treated CLL, compared with ibrutinib, found that “zanubrutinib was shown to have a superior response rate, an improved PFS, and a lower rate of atrial fibrillation/flutter.”

The drug’s manufacturer, BeiGene, launched the new open-label, multicenter study, in a bid for FDA approval of the drug as a frontline treatment for CLL and SLL. More than 150 hospitals in 14 countries participated in the trial from 2017 to 2019.

The subjects were all adults and at least 65 years old or with comorbidities; None had the genetic trait del(17)(p13.1); 241 were assigned to take zanubrutinib and 238 to bendamustine-rituximab. Another group consisted of 111 patients with CLL and del(17)(p13·1). According to the study authors, these patients are especially difficult to treat.

The vast majority of patients were White (92%-95% depending on group) and male (61%-71%); 90%-92% had CLL.

At follow-up, there was no difference in overall survival between the main zanubrutinib and bendamustine-rituximab groups; 29 (12%) of the 241 patients in the zanubrutinib group and 57 (24%) of 238 patients in the bendamustine-rituximab group had progressed or died (HR, 0.42; 95% CI, 0.27-0.66; P < .00011). Adverse events leading to discontinuation were more common in the bendamustine-rituximab group (14%) versus zanubrutinib (8%).

In the third group, which only received zanubrutinib, 14% of patients died at median follow-up of 30.5 months; 98% of patients had adverse effects, and 5% discontinued treatment.

The researchers wrote that “zanubrutinib showed superior progression­-free survival versus bendamustine-rituximab in older patients or those with comorbidities with untreated CLL, with a low incidence of cardiac arrhythmia. Similar efficacy was observed in patients with del(17p)­–positive disease.”

The study didn’t examine cost; zanubrutinib is quite expensive.

In an interview, hematologist-oncologist Anthony Mato, MD, of Memorial Sloan Kettering Cancer Center in New York said the new study is important although not surprising, since other medications in the same class have shown similar results. Zanubrutinib is an alternative to ibrutinib, although the latter remains “an excellent drug,” he said.

“The era of chemotherapy being a first choice is over,” he said. “We’ve had several randomized studies that show targeted therapies are better tolerated and have better outcomes. We now need to look through the choices to decide which one of these good options are the best for our patients.”

In an interview, hematologist-oncologist Joanna Rhodes, MD, of Northwell Health in Hempstead, N.Y., highlighted the side effect profile of zanubrutinib, noting that it is low and resembles that of other BTKis, making it “another excellent treatment option.”

“We are seeing that bruising, upper respiratory tract infections, diarrhea, and arthralgias are the most common side effects,” she said. “Bleeding also is a common side effect, which is consistent across the class of BTKis, with 5% of patients developing a major bleed. Also, 3% of patients treated with zanubrutinib developed atrial fibrillation, which is consistent with data from other trials. Treatment discontinuation rates were low (8%).”

The study was funded by BeiGene. The authors reported multiple disclosures. Dr. Mato reported research or consulting relationships with BeiGene, AstraZeneca, and AbbVie. Dr. Rhodes reported multiple research or consulting relationships with Abbvie, BeiGene, Genentech, and others.

Publications
Topics
Sections

The second-generation selective Bruton tyrosine kinase inhibitor (BTKi) zanubrutinib outperformed the standard treatment bendamustine-rituximab in untreated chronic lymphocytic leukemia (CLL) and small lymphocytic lymphoma (SLL), a new industry-funded phase-3 trial found.

At a median follow-up of 26.2 months, progression to worsening disease or death was much lower in patients with these conditions who took zanubrutinib (Brukinsa), compared with those who took bendamustine-rituximab (hazard ratio. 0.42; 95% confidence interval, 0.28-0.63; P < .00011). The study was published in The Lancet Oncology.

Researchers already knew that ibrutinib, another BTKi, improves progression-free survival, study coauthor Paolo Ghia, MD, PhD, professor of medical oncology at Vita-Salute San Raffaele University, Milan, said in an interview. “Now we confirmed that the same advantage can be seen” in zanubrutinib.

According to Dr. Ghia, bendamustine-rituximab has long been a standard treatment in blood cancers and is considered well tolerated and inexpensive. But BTKis such as first-in-line ibrutinib have shown better results, he said, “and progressively, we are going to abandon bendamustine-rituximab.”

However, ibrutinib causes significant adverse effects such as bleeding, worsening hypertension and arrhythmia, he noted. As a result, second-generation BTKi such as zanubrutinib have entered the picture. The Food and Drug Administration approved it in 2019 for mantle cell lymphoma, and it has since been approved for Waldenström’s macroglobulinemia and marginal zone lymphoma.

In 2021, an interim analysis in a trial of the drug in patients with previously treated CLL, compared with ibrutinib, found that “zanubrutinib was shown to have a superior response rate, an improved PFS, and a lower rate of atrial fibrillation/flutter.”

The drug’s manufacturer, BeiGene, launched the new open-label, multicenter study, in a bid for FDA approval of the drug as a frontline treatment for CLL and SLL. More than 150 hospitals in 14 countries participated in the trial from 2017 to 2019.

The subjects were all adults and at least 65 years old or with comorbidities; None had the genetic trait del(17)(p13.1); 241 were assigned to take zanubrutinib and 238 to bendamustine-rituximab. Another group consisted of 111 patients with CLL and del(17)(p13·1). According to the study authors, these patients are especially difficult to treat.

The vast majority of patients were White (92%-95% depending on group) and male (61%-71%); 90%-92% had CLL.

At follow-up, there was no difference in overall survival between the main zanubrutinib and bendamustine-rituximab groups; 29 (12%) of the 241 patients in the zanubrutinib group and 57 (24%) of 238 patients in the bendamustine-rituximab group had progressed or died (HR, 0.42; 95% CI, 0.27-0.66; P < .00011). Adverse events leading to discontinuation were more common in the bendamustine-rituximab group (14%) versus zanubrutinib (8%).

In the third group, which only received zanubrutinib, 14% of patients died at median follow-up of 30.5 months; 98% of patients had adverse effects, and 5% discontinued treatment.

The researchers wrote that “zanubrutinib showed superior progression­-free survival versus bendamustine-rituximab in older patients or those with comorbidities with untreated CLL, with a low incidence of cardiac arrhythmia. Similar efficacy was observed in patients with del(17p)­–positive disease.”

The study didn’t examine cost; zanubrutinib is quite expensive.

In an interview, hematologist-oncologist Anthony Mato, MD, of Memorial Sloan Kettering Cancer Center in New York said the new study is important although not surprising, since other medications in the same class have shown similar results. Zanubrutinib is an alternative to ibrutinib, although the latter remains “an excellent drug,” he said.

“The era of chemotherapy being a first choice is over,” he said. “We’ve had several randomized studies that show targeted therapies are better tolerated and have better outcomes. We now need to look through the choices to decide which one of these good options are the best for our patients.”

In an interview, hematologist-oncologist Joanna Rhodes, MD, of Northwell Health in Hempstead, N.Y., highlighted the side effect profile of zanubrutinib, noting that it is low and resembles that of other BTKis, making it “another excellent treatment option.”

“We are seeing that bruising, upper respiratory tract infections, diarrhea, and arthralgias are the most common side effects,” she said. “Bleeding also is a common side effect, which is consistent across the class of BTKis, with 5% of patients developing a major bleed. Also, 3% of patients treated with zanubrutinib developed atrial fibrillation, which is consistent with data from other trials. Treatment discontinuation rates were low (8%).”

The study was funded by BeiGene. The authors reported multiple disclosures. Dr. Mato reported research or consulting relationships with BeiGene, AstraZeneca, and AbbVie. Dr. Rhodes reported multiple research or consulting relationships with Abbvie, BeiGene, Genentech, and others.

The second-generation selective Bruton tyrosine kinase inhibitor (BTKi) zanubrutinib outperformed the standard treatment bendamustine-rituximab in untreated chronic lymphocytic leukemia (CLL) and small lymphocytic lymphoma (SLL), a new industry-funded phase-3 trial found.

At a median follow-up of 26.2 months, progression to worsening disease or death was much lower in patients with these conditions who took zanubrutinib (Brukinsa), compared with those who took bendamustine-rituximab (hazard ratio. 0.42; 95% confidence interval, 0.28-0.63; P < .00011). The study was published in The Lancet Oncology.

Researchers already knew that ibrutinib, another BTKi, improves progression-free survival, study coauthor Paolo Ghia, MD, PhD, professor of medical oncology at Vita-Salute San Raffaele University, Milan, said in an interview. “Now we confirmed that the same advantage can be seen” in zanubrutinib.

According to Dr. Ghia, bendamustine-rituximab has long been a standard treatment in blood cancers and is considered well tolerated and inexpensive. But BTKis such as first-in-line ibrutinib have shown better results, he said, “and progressively, we are going to abandon bendamustine-rituximab.”

However, ibrutinib causes significant adverse effects such as bleeding, worsening hypertension and arrhythmia, he noted. As a result, second-generation BTKi such as zanubrutinib have entered the picture. The Food and Drug Administration approved it in 2019 for mantle cell lymphoma, and it has since been approved for Waldenström’s macroglobulinemia and marginal zone lymphoma.

In 2021, an interim analysis in a trial of the drug in patients with previously treated CLL, compared with ibrutinib, found that “zanubrutinib was shown to have a superior response rate, an improved PFS, and a lower rate of atrial fibrillation/flutter.”

The drug’s manufacturer, BeiGene, launched the new open-label, multicenter study, in a bid for FDA approval of the drug as a frontline treatment for CLL and SLL. More than 150 hospitals in 14 countries participated in the trial from 2017 to 2019.

The subjects were all adults and at least 65 years old or with comorbidities; None had the genetic trait del(17)(p13.1); 241 were assigned to take zanubrutinib and 238 to bendamustine-rituximab. Another group consisted of 111 patients with CLL and del(17)(p13·1). According to the study authors, these patients are especially difficult to treat.

The vast majority of patients were White (92%-95% depending on group) and male (61%-71%); 90%-92% had CLL.

At follow-up, there was no difference in overall survival between the main zanubrutinib and bendamustine-rituximab groups; 29 (12%) of the 241 patients in the zanubrutinib group and 57 (24%) of 238 patients in the bendamustine-rituximab group had progressed or died (HR, 0.42; 95% CI, 0.27-0.66; P < .00011). Adverse events leading to discontinuation were more common in the bendamustine-rituximab group (14%) versus zanubrutinib (8%).

In the third group, which only received zanubrutinib, 14% of patients died at median follow-up of 30.5 months; 98% of patients had adverse effects, and 5% discontinued treatment.

The researchers wrote that “zanubrutinib showed superior progression­-free survival versus bendamustine-rituximab in older patients or those with comorbidities with untreated CLL, with a low incidence of cardiac arrhythmia. Similar efficacy was observed in patients with del(17p)­–positive disease.”

The study didn’t examine cost; zanubrutinib is quite expensive.

In an interview, hematologist-oncologist Anthony Mato, MD, of Memorial Sloan Kettering Cancer Center in New York said the new study is important although not surprising, since other medications in the same class have shown similar results. Zanubrutinib is an alternative to ibrutinib, although the latter remains “an excellent drug,” he said.

“The era of chemotherapy being a first choice is over,” he said. “We’ve had several randomized studies that show targeted therapies are better tolerated and have better outcomes. We now need to look through the choices to decide which one of these good options are the best for our patients.”

In an interview, hematologist-oncologist Joanna Rhodes, MD, of Northwell Health in Hempstead, N.Y., highlighted the side effect profile of zanubrutinib, noting that it is low and resembles that of other BTKis, making it “another excellent treatment option.”

“We are seeing that bruising, upper respiratory tract infections, diarrhea, and arthralgias are the most common side effects,” she said. “Bleeding also is a common side effect, which is consistent across the class of BTKis, with 5% of patients developing a major bleed. Also, 3% of patients treated with zanubrutinib developed atrial fibrillation, which is consistent with data from other trials. Treatment discontinuation rates were low (8%).”

The study was funded by BeiGene. The authors reported multiple disclosures. Dr. Mato reported research or consulting relationships with BeiGene, AstraZeneca, and AbbVie. Dr. Rhodes reported multiple research or consulting relationships with Abbvie, BeiGene, Genentech, and others.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM THE LANCET ONCOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article