FDA approves futibatinib (Lytgobi) for certain biliary tract cancers

Article Type
Changed
Mon, 10/03/2022 - 15:31

The Food and Drug Administration has granted an accelerated approval for a new drug, futibatinib (Lytgobi), to be used for patients with a certain type of biliary tract cancer. Futibatinib was granted priority review and breakthrough designation, the agency said in its announcement.

Futibatinib is indicated for use in adult patients with previously treated, unresectable, locally advanced, or metastatic intrahepatic cholangiocarcinoma harboring fibroblast growth factor receptor 2 (FGFR2) gene fusions or other rearrangements.

The FDA noted that efficacy was evaluated in a multicenter, open-label, single-arm trial (known as TAS-120-101 [NCT02052778]), which involved 103 patients with such tumors. The presence of FGFR2 fusions or other rearrangements was determined using next-generation sequencing.

All the patients in this trial received futibatinib (20 mg orally once daily) until disease progression or unacceptable toxicity.

The overall response rate was 42% (95% confidence interval, 32%-52%), and all of the 43 patients who responded achieved partial responses.

The median duration of response was 9.7 months (95% CI, 7.6-17.1).

The most common adverse reactions that occurred in 20% or more of patients were nail toxicity, musculoskeletal pain, constipation, diarrhea, fatigue, dry mouth, alopecia, stomatitis, abdominal pain, dry skin, arthralgia, dysgeusia, dry eye, nausea, decreased appetite, urinary tract infection, palmar-plantar erythrodysesthesia syndrome, and vomiting.

The manufacturer noted in its announcement that futibatinib covalently binds to FGFR2 and inhibits the signaling pathway. The other approved FGFR inhibitors are reversible ATP-competitive inhibitors.

The company also provided some background information on the cancer.

As a whole, cholangiocarcinoma is an aggressive cancer of the bile ducts. It is diagnosed in approximately 8000 individuals each year in the United States, the company noted.

These cases include both intrahepatic (inside the liver) and extrahepatic (outside the liver) forms of the disease. Approximately 20% of patients diagnosed with cholangiocarcinoma have the intrahepatic form of the disease. Among these 20%, approximately 10%-16% of patients have FGFR2 gene rearrangements, including fusions, which promote tumor proliferation.

Futibatinib is “a key example of the potential of precision medicine in iCCA [intrahepatic cholangiocarcinoma] and represents another advance in the treatment of this rare and challenging disease,” said medical oncologist Lipika Goyal, MD, MPhil, of the Massachusetts General Hospital Cancer Center, Boston, and lead investigator of the pivotal study that supported the approval.

“I am encouraged that treatment options continue to expand and evolve for this disease through the dedicated efforts of many over several years,” she commented in the company’s press release.

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

Publications
Topics
Sections

The Food and Drug Administration has granted an accelerated approval for a new drug, futibatinib (Lytgobi), to be used for patients with a certain type of biliary tract cancer. Futibatinib was granted priority review and breakthrough designation, the agency said in its announcement.

Futibatinib is indicated for use in adult patients with previously treated, unresectable, locally advanced, or metastatic intrahepatic cholangiocarcinoma harboring fibroblast growth factor receptor 2 (FGFR2) gene fusions or other rearrangements.

The FDA noted that efficacy was evaluated in a multicenter, open-label, single-arm trial (known as TAS-120-101 [NCT02052778]), which involved 103 patients with such tumors. The presence of FGFR2 fusions or other rearrangements was determined using next-generation sequencing.

All the patients in this trial received futibatinib (20 mg orally once daily) until disease progression or unacceptable toxicity.

The overall response rate was 42% (95% confidence interval, 32%-52%), and all of the 43 patients who responded achieved partial responses.

The median duration of response was 9.7 months (95% CI, 7.6-17.1).

The most common adverse reactions that occurred in 20% or more of patients were nail toxicity, musculoskeletal pain, constipation, diarrhea, fatigue, dry mouth, alopecia, stomatitis, abdominal pain, dry skin, arthralgia, dysgeusia, dry eye, nausea, decreased appetite, urinary tract infection, palmar-plantar erythrodysesthesia syndrome, and vomiting.

The manufacturer noted in its announcement that futibatinib covalently binds to FGFR2 and inhibits the signaling pathway. The other approved FGFR inhibitors are reversible ATP-competitive inhibitors.

The company also provided some background information on the cancer.

As a whole, cholangiocarcinoma is an aggressive cancer of the bile ducts. It is diagnosed in approximately 8000 individuals each year in the United States, the company noted.

These cases include both intrahepatic (inside the liver) and extrahepatic (outside the liver) forms of the disease. Approximately 20% of patients diagnosed with cholangiocarcinoma have the intrahepatic form of the disease. Among these 20%, approximately 10%-16% of patients have FGFR2 gene rearrangements, including fusions, which promote tumor proliferation.

Futibatinib is “a key example of the potential of precision medicine in iCCA [intrahepatic cholangiocarcinoma] and represents another advance in the treatment of this rare and challenging disease,” said medical oncologist Lipika Goyal, MD, MPhil, of the Massachusetts General Hospital Cancer Center, Boston, and lead investigator of the pivotal study that supported the approval.

“I am encouraged that treatment options continue to expand and evolve for this disease through the dedicated efforts of many over several years,” she commented in the company’s press release.

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

The Food and Drug Administration has granted an accelerated approval for a new drug, futibatinib (Lytgobi), to be used for patients with a certain type of biliary tract cancer. Futibatinib was granted priority review and breakthrough designation, the agency said in its announcement.

Futibatinib is indicated for use in adult patients with previously treated, unresectable, locally advanced, or metastatic intrahepatic cholangiocarcinoma harboring fibroblast growth factor receptor 2 (FGFR2) gene fusions or other rearrangements.

The FDA noted that efficacy was evaluated in a multicenter, open-label, single-arm trial (known as TAS-120-101 [NCT02052778]), which involved 103 patients with such tumors. The presence of FGFR2 fusions or other rearrangements was determined using next-generation sequencing.

All the patients in this trial received futibatinib (20 mg orally once daily) until disease progression or unacceptable toxicity.

The overall response rate was 42% (95% confidence interval, 32%-52%), and all of the 43 patients who responded achieved partial responses.

The median duration of response was 9.7 months (95% CI, 7.6-17.1).

The most common adverse reactions that occurred in 20% or more of patients were nail toxicity, musculoskeletal pain, constipation, diarrhea, fatigue, dry mouth, alopecia, stomatitis, abdominal pain, dry skin, arthralgia, dysgeusia, dry eye, nausea, decreased appetite, urinary tract infection, palmar-plantar erythrodysesthesia syndrome, and vomiting.

The manufacturer noted in its announcement that futibatinib covalently binds to FGFR2 and inhibits the signaling pathway. The other approved FGFR inhibitors are reversible ATP-competitive inhibitors.

The company also provided some background information on the cancer.

As a whole, cholangiocarcinoma is an aggressive cancer of the bile ducts. It is diagnosed in approximately 8000 individuals each year in the United States, the company noted.

These cases include both intrahepatic (inside the liver) and extrahepatic (outside the liver) forms of the disease. Approximately 20% of patients diagnosed with cholangiocarcinoma have the intrahepatic form of the disease. Among these 20%, approximately 10%-16% of patients have FGFR2 gene rearrangements, including fusions, which promote tumor proliferation.

Futibatinib is “a key example of the potential of precision medicine in iCCA [intrahepatic cholangiocarcinoma] and represents another advance in the treatment of this rare and challenging disease,” said medical oncologist Lipika Goyal, MD, MPhil, of the Massachusetts General Hospital Cancer Center, Boston, and lead investigator of the pivotal study that supported the approval.

“I am encouraged that treatment options continue to expand and evolve for this disease through the dedicated efforts of many over several years,” she commented in the company’s press release.

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

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

Oncologists’ wealth and debt: COVID had little impact

Article Type
Changed
Fri, 08/19/2022 - 09:56

 

2021 bought welcome relief to oncologists, whose incomes generally rose as practices reopened after COVID-19 restrictions were lifted and patients ventured out again, concludes the latest Medscape Oncologist Wealth & Debt Report 2022.

Comparing the findings with those in the larger Medscape Physician Wealth & Debt Report 2022, which surveyed more than 13,000 physicians in 29 specialties, the findings for oncologists show how they compare with those who chose other paths in medicine.

Oncologists’ income rose, on average, by 2% in the past year and now stands at an average of $411,000 annually, up from $403,000 in the 2021 report.

This puts oncologists in the top third of specialties, with plastic surgeons again in the top slot (with average income of $576,000 in 2022).

One-fifth (20%) of oncologists surveyed reported a family worth of more than $5 million, which represents substantial family wealth, the report comments.

However, 22% of oncologists reported that their family net worth was less than $500,000, and another 10% estimated that it to fall between $500,000 and $1 million.

For comparison, the average U.S. family net worth is about $749,000, according to data from the Federal Reserve.
 

Most live ‘within their means’

Most oncologists (94%) and also most (94%) of all of the physicians surveyed said that they live within or below their means.

How does one do this? Just paying off credit cards each month and contributing enough to a 401(k) account to receive an employer match does not meet this standard, said Joel Greenwald MD, CFP, a wealth management advisor for physicians. To live within or below your means, you also need to be saving at least 20% toward retirement, pay down student loans, contribute to your kids’ college savings, and set aside rainy day cash, he explained.

When physicians were asked about their favorite cost-cutting tactics, replies included bringing lunch to work, keeping a car for 15 years, and carrying out their own household maintenance and repairs. One doctor described a “24-hour rule” when it comes to shopping: “Revisit the desired purchase after 24 hours to see if it’s still desired.”

But how well do these tactics go down with ‘the other half’ and the rest of the household? Two-thirds (66%) of oncologists, and a similar proportion of all physicians, said that they argue with their significant other about spending. This appears to be high in comparison with the finding from a recent survey that across the United States, about one in four couples (25%) argue about money at least once a month.

Regarding spending, the top expense among oncologists was for childcare (16%), private tuition for offspring (14%), mortgage on a second home (14%), college tuition for offspring (14%), and a car lease (12%).

Around 17% of oncologists reported that they are still paying off their own college or medical school loans. For this statistic, they are about in the middle of all specialties.

The report notes that freeing oneself from medical school debt is very costly. Physicians in the United States pay an average of $356,000-$440,000, about half of which is interest.
 

Little change over 2021

The COVID pandemic had much less of an impact on physicians than it had on the general population when it comes to keeping up with payments, and most physicians were not affected. Only 3% of oncologists said they fell behind with payments for mortgage; 6% fell behind with payments for other bills.

In comparison, nearly half (46%) of Americans missed one or more payments of rent or mortgage because of COVID, according to a 2021 industry survey.

Over the past year, most oncologists (70%) did not change their spending habits, and only 11% cut expenses by deferring or refinancing loans. Also, most oncologists (75%) avoided major financial loses. Only 8% reported financial losses because of problems at their medical practice.

However, a slightly higher percentage of oncologists reported a stock or company investment that had turned sour in 2022 (37%) in comparison with 2021 (28%).

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

Publications
Topics
Sections

 

2021 bought welcome relief to oncologists, whose incomes generally rose as practices reopened after COVID-19 restrictions were lifted and patients ventured out again, concludes the latest Medscape Oncologist Wealth & Debt Report 2022.

Comparing the findings with those in the larger Medscape Physician Wealth & Debt Report 2022, which surveyed more than 13,000 physicians in 29 specialties, the findings for oncologists show how they compare with those who chose other paths in medicine.

Oncologists’ income rose, on average, by 2% in the past year and now stands at an average of $411,000 annually, up from $403,000 in the 2021 report.

This puts oncologists in the top third of specialties, with plastic surgeons again in the top slot (with average income of $576,000 in 2022).

One-fifth (20%) of oncologists surveyed reported a family worth of more than $5 million, which represents substantial family wealth, the report comments.

However, 22% of oncologists reported that their family net worth was less than $500,000, and another 10% estimated that it to fall between $500,000 and $1 million.

For comparison, the average U.S. family net worth is about $749,000, according to data from the Federal Reserve.
 

Most live ‘within their means’

Most oncologists (94%) and also most (94%) of all of the physicians surveyed said that they live within or below their means.

How does one do this? Just paying off credit cards each month and contributing enough to a 401(k) account to receive an employer match does not meet this standard, said Joel Greenwald MD, CFP, a wealth management advisor for physicians. To live within or below your means, you also need to be saving at least 20% toward retirement, pay down student loans, contribute to your kids’ college savings, and set aside rainy day cash, he explained.

When physicians were asked about their favorite cost-cutting tactics, replies included bringing lunch to work, keeping a car for 15 years, and carrying out their own household maintenance and repairs. One doctor described a “24-hour rule” when it comes to shopping: “Revisit the desired purchase after 24 hours to see if it’s still desired.”

But how well do these tactics go down with ‘the other half’ and the rest of the household? Two-thirds (66%) of oncologists, and a similar proportion of all physicians, said that they argue with their significant other about spending. This appears to be high in comparison with the finding from a recent survey that across the United States, about one in four couples (25%) argue about money at least once a month.

Regarding spending, the top expense among oncologists was for childcare (16%), private tuition for offspring (14%), mortgage on a second home (14%), college tuition for offspring (14%), and a car lease (12%).

Around 17% of oncologists reported that they are still paying off their own college or medical school loans. For this statistic, they are about in the middle of all specialties.

The report notes that freeing oneself from medical school debt is very costly. Physicians in the United States pay an average of $356,000-$440,000, about half of which is interest.
 

Little change over 2021

The COVID pandemic had much less of an impact on physicians than it had on the general population when it comes to keeping up with payments, and most physicians were not affected. Only 3% of oncologists said they fell behind with payments for mortgage; 6% fell behind with payments for other bills.

In comparison, nearly half (46%) of Americans missed one or more payments of rent or mortgage because of COVID, according to a 2021 industry survey.

Over the past year, most oncologists (70%) did not change their spending habits, and only 11% cut expenses by deferring or refinancing loans. Also, most oncologists (75%) avoided major financial loses. Only 8% reported financial losses because of problems at their medical practice.

However, a slightly higher percentage of oncologists reported a stock or company investment that had turned sour in 2022 (37%) in comparison with 2021 (28%).

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

 

2021 bought welcome relief to oncologists, whose incomes generally rose as practices reopened after COVID-19 restrictions were lifted and patients ventured out again, concludes the latest Medscape Oncologist Wealth & Debt Report 2022.

Comparing the findings with those in the larger Medscape Physician Wealth & Debt Report 2022, which surveyed more than 13,000 physicians in 29 specialties, the findings for oncologists show how they compare with those who chose other paths in medicine.

Oncologists’ income rose, on average, by 2% in the past year and now stands at an average of $411,000 annually, up from $403,000 in the 2021 report.

This puts oncologists in the top third of specialties, with plastic surgeons again in the top slot (with average income of $576,000 in 2022).

One-fifth (20%) of oncologists surveyed reported a family worth of more than $5 million, which represents substantial family wealth, the report comments.

However, 22% of oncologists reported that their family net worth was less than $500,000, and another 10% estimated that it to fall between $500,000 and $1 million.

For comparison, the average U.S. family net worth is about $749,000, according to data from the Federal Reserve.
 

Most live ‘within their means’

Most oncologists (94%) and also most (94%) of all of the physicians surveyed said that they live within or below their means.

How does one do this? Just paying off credit cards each month and contributing enough to a 401(k) account to receive an employer match does not meet this standard, said Joel Greenwald MD, CFP, a wealth management advisor for physicians. To live within or below your means, you also need to be saving at least 20% toward retirement, pay down student loans, contribute to your kids’ college savings, and set aside rainy day cash, he explained.

When physicians were asked about their favorite cost-cutting tactics, replies included bringing lunch to work, keeping a car for 15 years, and carrying out their own household maintenance and repairs. One doctor described a “24-hour rule” when it comes to shopping: “Revisit the desired purchase after 24 hours to see if it’s still desired.”

But how well do these tactics go down with ‘the other half’ and the rest of the household? Two-thirds (66%) of oncologists, and a similar proportion of all physicians, said that they argue with their significant other about spending. This appears to be high in comparison with the finding from a recent survey that across the United States, about one in four couples (25%) argue about money at least once a month.

Regarding spending, the top expense among oncologists was for childcare (16%), private tuition for offspring (14%), mortgage on a second home (14%), college tuition for offspring (14%), and a car lease (12%).

Around 17% of oncologists reported that they are still paying off their own college or medical school loans. For this statistic, they are about in the middle of all specialties.

The report notes that freeing oneself from medical school debt is very costly. Physicians in the United States pay an average of $356,000-$440,000, about half of which is interest.
 

Little change over 2021

The COVID pandemic had much less of an impact on physicians than it had on the general population when it comes to keeping up with payments, and most physicians were not affected. Only 3% of oncologists said they fell behind with payments for mortgage; 6% fell behind with payments for other bills.

In comparison, nearly half (46%) of Americans missed one or more payments of rent or mortgage because of COVID, according to a 2021 industry survey.

Over the past year, most oncologists (70%) did not change their spending habits, and only 11% cut expenses by deferring or refinancing loans. Also, most oncologists (75%) avoided major financial loses. Only 8% reported financial losses because of problems at their medical practice.

However, a slightly higher percentage of oncologists reported a stock or company investment that had turned sour in 2022 (37%) in comparison with 2021 (28%).

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

FDA approves Enhertu (trastuzumab deruxtecan) for HER2 lung cancer

Article Type
Changed
Tue, 08/16/2022 - 09:03

Patients with lung cancer now have another treatment option: If their tumors are found to carry HER2 mutations, they can now be treated with trastuzumab deruxtecan (Enhertu), a drug that specifically targets that defect.

This product is already approved for used in HER2-positive breast cancer and gastric cancer. Now it has become the first product approved by the U.S. Food and Drug Administration for use in HER2-positive lung cancer.


The FDA also approved companion diagnostic tests to detect HER2 mutations: Life Technologies Corporation’s Oncomine Dx Target Test for use in lung tissue and Guardant Health’s Guardant360 CDx for use on plasma samples. The agency notes that if no mutation is detected in a plasma specimen, the tumor tissue should be tested.

Specifically, the new indication is used in patients with unresectable or metastatic non–small cell lung cancer (NSCLC) whose tumors have activating HER2 (ERBB2) mutations, as detected by an FDA-approved test, and who have already received a prior systemic therapy.

About 3% of nonsquamous NSCLC tumors carry mutations in the HER2 gene, and they are associated with female sex, never-smokers, and a poor prognosis.

“HER2 mutant non–small cell lung cancer is an aggressive form of disease, which commonly affects young patients who have faced limited treatment options and a poor prognosis to date,” said Dave Fredrickson, executive vice-president of the oncology business unit at AstraZeneca.

The new approval “provides these patients with the opportunity to benefit from a targeted therapy and highlights the importance of testing for predictive markers, including HER2 in lung cancer, at the time of diagnosis to ensure patients receive the most appropriate treatment for their specific disease,” he commented in a company press release.

This is an accelerated approval, based on overall response rate data from the DESTINY-Lung02 phase 2 trial, the company noted. An interim efficacy analysis in a prespecified patient cohort showed that trastuzumab deruxtecan (at 5.4 mg/kg) demonstrated a confirmed overall response rate of 57.7% (n = 52; 95% confidence interval, 43.2%-71.3%) in patients with HER2-mutant unresectable or metastatic nonsquamous NSCLC who had received one prior systemic therapy as assessed by blinded independent central review. Complete responses were seen in 1.9% of patients (n = 1) and partial responses in 55.8% of patients (n = 29), with a median duration of response of 8.7 months (95% CI, 7.1-NE).

The FDA noted that for the 52 patients in the primary efficacy population of the DESTINY-Lung02 trial, the median age was 58 years (range, 30-78 years); 69% were female; and 79% were Asian, 12% were White, and 10% were of other races.

Clinical data welcomed by experts

Clinical data are already available from the DESTINY-Lung 01 trial, and the results were welcomed enthusiastically by experts when they were published in the New England Journal of Medicine earlier this year.

“These results establish the new standard of care for patients with NSCLC harboring HER2 mutations,” Antonio Passaro, MD, PhD, from the European Institute of Oncology IRCCS, Milan, and Solange Peters, MD, PhD, from Lausanne (Switzerland) University Hospital, wrote in an accompanying editorial.

This trial involved 91 patients, all treated with trastuzumab deruxtecan (at 6.4 mg/kg of body weight every 3 weeks). The median duration of treatment was 6.9 months, and the median follow-up was 13.1 months.

The results showed a 55% centrally confirmed objective response, and median duration of response was 9.3 months.

In addition, the investigators reported a median progression-free survival of 8.2 months and a median overall survival of almost 18 months, both of which they described as “encouraging” in this patient population.

However, the results also highlighted a problem with the drug in this patient population. Notably, 26% of patients experienced interstitial lung disease, which resulted in death in two patients. The drug was also withdrawn in 16 patients and interrupted in 8 patients because of this adverse event.

Editorialists Dr. Passaro and Dr. Peters described this finding as “a concern” and note that “the incidence of interstitial lung disease is significantly higher among patients with lung cancer than among those with breast or gastric cancers, which may indicate a role of smoking-related damage.”

They also highlighted the need for an “investigation of the clinical efficacy of a reduced dose of trastuzumab deruxtecan,” and so the dose was reduced for the DESTINY-Lung02 trial.

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

Publications
Topics
Sections

Patients with lung cancer now have another treatment option: If their tumors are found to carry HER2 mutations, they can now be treated with trastuzumab deruxtecan (Enhertu), a drug that specifically targets that defect.

This product is already approved for used in HER2-positive breast cancer and gastric cancer. Now it has become the first product approved by the U.S. Food and Drug Administration for use in HER2-positive lung cancer.


The FDA also approved companion diagnostic tests to detect HER2 mutations: Life Technologies Corporation’s Oncomine Dx Target Test for use in lung tissue and Guardant Health’s Guardant360 CDx for use on plasma samples. The agency notes that if no mutation is detected in a plasma specimen, the tumor tissue should be tested.

Specifically, the new indication is used in patients with unresectable or metastatic non–small cell lung cancer (NSCLC) whose tumors have activating HER2 (ERBB2) mutations, as detected by an FDA-approved test, and who have already received a prior systemic therapy.

About 3% of nonsquamous NSCLC tumors carry mutations in the HER2 gene, and they are associated with female sex, never-smokers, and a poor prognosis.

“HER2 mutant non–small cell lung cancer is an aggressive form of disease, which commonly affects young patients who have faced limited treatment options and a poor prognosis to date,” said Dave Fredrickson, executive vice-president of the oncology business unit at AstraZeneca.

The new approval “provides these patients with the opportunity to benefit from a targeted therapy and highlights the importance of testing for predictive markers, including HER2 in lung cancer, at the time of diagnosis to ensure patients receive the most appropriate treatment for their specific disease,” he commented in a company press release.

This is an accelerated approval, based on overall response rate data from the DESTINY-Lung02 phase 2 trial, the company noted. An interim efficacy analysis in a prespecified patient cohort showed that trastuzumab deruxtecan (at 5.4 mg/kg) demonstrated a confirmed overall response rate of 57.7% (n = 52; 95% confidence interval, 43.2%-71.3%) in patients with HER2-mutant unresectable or metastatic nonsquamous NSCLC who had received one prior systemic therapy as assessed by blinded independent central review. Complete responses were seen in 1.9% of patients (n = 1) and partial responses in 55.8% of patients (n = 29), with a median duration of response of 8.7 months (95% CI, 7.1-NE).

The FDA noted that for the 52 patients in the primary efficacy population of the DESTINY-Lung02 trial, the median age was 58 years (range, 30-78 years); 69% were female; and 79% were Asian, 12% were White, and 10% were of other races.

Clinical data welcomed by experts

Clinical data are already available from the DESTINY-Lung 01 trial, and the results were welcomed enthusiastically by experts when they were published in the New England Journal of Medicine earlier this year.

“These results establish the new standard of care for patients with NSCLC harboring HER2 mutations,” Antonio Passaro, MD, PhD, from the European Institute of Oncology IRCCS, Milan, and Solange Peters, MD, PhD, from Lausanne (Switzerland) University Hospital, wrote in an accompanying editorial.

This trial involved 91 patients, all treated with trastuzumab deruxtecan (at 6.4 mg/kg of body weight every 3 weeks). The median duration of treatment was 6.9 months, and the median follow-up was 13.1 months.

The results showed a 55% centrally confirmed objective response, and median duration of response was 9.3 months.

In addition, the investigators reported a median progression-free survival of 8.2 months and a median overall survival of almost 18 months, both of which they described as “encouraging” in this patient population.

However, the results also highlighted a problem with the drug in this patient population. Notably, 26% of patients experienced interstitial lung disease, which resulted in death in two patients. The drug was also withdrawn in 16 patients and interrupted in 8 patients because of this adverse event.

Editorialists Dr. Passaro and Dr. Peters described this finding as “a concern” and note that “the incidence of interstitial lung disease is significantly higher among patients with lung cancer than among those with breast or gastric cancers, which may indicate a role of smoking-related damage.”

They also highlighted the need for an “investigation of the clinical efficacy of a reduced dose of trastuzumab deruxtecan,” and so the dose was reduced for the DESTINY-Lung02 trial.

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

Patients with lung cancer now have another treatment option: If their tumors are found to carry HER2 mutations, they can now be treated with trastuzumab deruxtecan (Enhertu), a drug that specifically targets that defect.

This product is already approved for used in HER2-positive breast cancer and gastric cancer. Now it has become the first product approved by the U.S. Food and Drug Administration for use in HER2-positive lung cancer.


The FDA also approved companion diagnostic tests to detect HER2 mutations: Life Technologies Corporation’s Oncomine Dx Target Test for use in lung tissue and Guardant Health’s Guardant360 CDx for use on plasma samples. The agency notes that if no mutation is detected in a plasma specimen, the tumor tissue should be tested.

Specifically, the new indication is used in patients with unresectable or metastatic non–small cell lung cancer (NSCLC) whose tumors have activating HER2 (ERBB2) mutations, as detected by an FDA-approved test, and who have already received a prior systemic therapy.

About 3% of nonsquamous NSCLC tumors carry mutations in the HER2 gene, and they are associated with female sex, never-smokers, and a poor prognosis.

“HER2 mutant non–small cell lung cancer is an aggressive form of disease, which commonly affects young patients who have faced limited treatment options and a poor prognosis to date,” said Dave Fredrickson, executive vice-president of the oncology business unit at AstraZeneca.

The new approval “provides these patients with the opportunity to benefit from a targeted therapy and highlights the importance of testing for predictive markers, including HER2 in lung cancer, at the time of diagnosis to ensure patients receive the most appropriate treatment for their specific disease,” he commented in a company press release.

This is an accelerated approval, based on overall response rate data from the DESTINY-Lung02 phase 2 trial, the company noted. An interim efficacy analysis in a prespecified patient cohort showed that trastuzumab deruxtecan (at 5.4 mg/kg) demonstrated a confirmed overall response rate of 57.7% (n = 52; 95% confidence interval, 43.2%-71.3%) in patients with HER2-mutant unresectable or metastatic nonsquamous NSCLC who had received one prior systemic therapy as assessed by blinded independent central review. Complete responses were seen in 1.9% of patients (n = 1) and partial responses in 55.8% of patients (n = 29), with a median duration of response of 8.7 months (95% CI, 7.1-NE).

The FDA noted that for the 52 patients in the primary efficacy population of the DESTINY-Lung02 trial, the median age was 58 years (range, 30-78 years); 69% were female; and 79% were Asian, 12% were White, and 10% were of other races.

Clinical data welcomed by experts

Clinical data are already available from the DESTINY-Lung 01 trial, and the results were welcomed enthusiastically by experts when they were published in the New England Journal of Medicine earlier this year.

“These results establish the new standard of care for patients with NSCLC harboring HER2 mutations,” Antonio Passaro, MD, PhD, from the European Institute of Oncology IRCCS, Milan, and Solange Peters, MD, PhD, from Lausanne (Switzerland) University Hospital, wrote in an accompanying editorial.

This trial involved 91 patients, all treated with trastuzumab deruxtecan (at 6.4 mg/kg of body weight every 3 weeks). The median duration of treatment was 6.9 months, and the median follow-up was 13.1 months.

The results showed a 55% centrally confirmed objective response, and median duration of response was 9.3 months.

In addition, the investigators reported a median progression-free survival of 8.2 months and a median overall survival of almost 18 months, both of which they described as “encouraging” in this patient population.

However, the results also highlighted a problem with the drug in this patient population. Notably, 26% of patients experienced interstitial lung disease, which resulted in death in two patients. The drug was also withdrawn in 16 patients and interrupted in 8 patients because of this adverse event.

Editorialists Dr. Passaro and Dr. Peters described this finding as “a concern” and note that “the incidence of interstitial lung disease is significantly higher among patients with lung cancer than among those with breast or gastric cancers, which may indicate a role of smoking-related damage.”

They also highlighted the need for an “investigation of the clinical efficacy of a reduced dose of trastuzumab deruxtecan,” and so the dose was reduced for the DESTINY-Lung02 trial.

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

Oncologists flock to Chicago for ASCO, after 2 years online

Article Type
Changed
Mon, 06/06/2022 - 10:17

The biggest cancer conference in the world is back in person after 2 years online during the COVID pandemic. And it appears many are eager to attend the American Society of Clinical Oncology annual meeting in person now that they can.

By early May, ASCO already had 30,000 registrations, of which 80% were in person – there were 27,000 hotel reservations.

“That’s almost identical to where we were in terms of numbers in 2019 at the same point in time,” Julie Gralow, MD, chief medical officer at ASCO, said in an interview.

These figures, which are from May 11, are likely to increase. In past years, there has been an upswing in registrations right before the meeting starts.

The annual meeting begins on Friday, June 3, and runs until Tuesday, June 7. It will be held in Chicago, yet again, in the vast McCormick Place, sections of which were transformed into field hospital wards when the pandemic hit in 2020.

But the meeting will also continue to be transmitted virtually, as it has been for the past 2 years, for those not attending in person.

“I do think that the hybrid model will move forward,” Dr. Gralow said. “We can get a lot of attendees, especially from very distant places, who can’t travel, or can’t easily travel, and we have learned how to make that experience better for them as well.”

Attendees can also change their minds if, for example, rising numbers of COVID cases as the meeting nears put them off traveling. “We are allowing people to change to virtual. So I think there may be a little bit of that, depending on what happens to COVID in different parts of the world,” Dr. Gralow commented.

For those who do attend, the organization is “doing the best that we can to keep people safe,” said Dr. Gralow, who was previously a professor of global health and is now a breast medical oncologist and clinical trialist.

To attend in person, ASCO is mandating proof of vaccination (which in the United States means two doses of the COVID vaccine). “If you prove in advance that you are vaccinated, we will send you your badge, so you don’t have to stand in line,” she added.

“As far as masks go, we are saying right now that we are complying with Chicago’s rules, which mean there is no mandatory indoor masking,” she continued. “We are recommending masking because this is a group of physicians who treat immunocompromised patients. So we are recommending that.”

This stance has gotten some push-back on Twitter from both physicians and patient advocates, with some surprised that masking is not mandatory.

“I know that ‘mask-optional’ meetings mean most will omit masks; I literally just saw this at my last meeting as one of the few masked MDs,” commented radiation oncologist Fumiko Ladd Chino, MD. She appealed to the organizers with a plea: “There’s still time to change #ASCO22 policies. We’re in it for patient health.”

Patient advocate Manju George, MVSc, PhD, a rectal cancer survivor, was also campaigning for a change in policy by setting up a letter that others could sign, adding that “ASCO leadership is being flooded with pleads from concerned HCPs.”

When asked whether it was considering a change in mask policy, ASCO replied: “As far as health and safety go, the protocols we’ve put in place meet or exceed current [World Health Organization, [Centers for Disease Control and Prevention, and city of Chicago guidelines. ASCO is also closely coordinating with both the city and the convention center and we are actively monitoring local conditions.”

“To protect the health and safety of all meeting attendees, our protocols require attendees to be fully vaccinated and self-test negative for COVID-19 within 48 hours prior to their arrival at the meeting. In addition, we expect all attendees to be masked when indoors and are encouraging regular self-testing. We fully expect members of our community to do their part to help keep everyone safe, and we’re making it easy for attendees to comply with our policies by providing medical-grade masks as well as both rapid antigen and [polymerase chain reaction] COVID-19 tests,” the organization said.

There will also be a notification system so attendees can select how they identify for closeness, with red meaning stand back, no hugs, no handshakes; yellow signifying something more intermediate; and green signaling the person is okay with contact with a handshake or a hug. This system has already been used during smaller ASCO subspecialty meetings earlier this year, and feedback from delegates was positive, Dr. Gralow commented.
 

 

 

Advancing equitable care

The theme of the 2022 meeting, chosen by ASCO President Everett Vokes, MD, is advancing equitable cancer care through innovation.

It builds on the theme of equity from 2021, chosen by previous president Lori Pierce, MD, which was “Equity: Every Patient. Every Day. Everywhere.”

Some of this relates to disparities in equity, commented Dr. Gralow. This is the focus of a premeeting press briefing on May 26 that will highlight a few abstracts that focus on disparities and what can be done to address them. One study (abstract 6511) focuses on telemedicine, which was increasingly used during the pandemic, but the results show not all U.S. patient populations could access the specialty care they needed in this way.
 

De-escalation of therapy

De-escalation of therapy is another theme running through the meeting.

“There are some cancers where we have achieved such good outcomes that it is time to look at de-escalating therapy because we know that we are probably way overtreating a component of those patients. ... So we are looking at whether we can find subpopulations where we can back off on therapy,” commented Dr. Gralow.

One example is the LUMINA trial in breast cancer (abstract LBA501), which looked at omitting radiotherapy after surgery. “In standard practice we have already been doing this, not based on solid data, but based on an accumulation of retrospective analyses and similar evidence,” commented Dr. Gralow. This trial tested the approach prospectively, lowered the age range of patients, and better defined which patients were likely to benefit.

Another example is the DYNAMIC trial in colorectal cancer (abstract LBA-100), which looks at omitting chemotherapy based on levels of circulating tumor DNA after surgery. These patients had stage 2 disease and generally do very well with surgery and adjuvant chemotherapy, Dr. Gralow stated. This trial aims to find the subset of patients who could do just as well without the chemotherapy; it may also identify those patients at the other end of the scale, who perhaps need a bit more treatment, she added.
 

Spotlight on innovation

The focus on innovation includes exploring drugs developed outside the United States. One example is nimotuzumab, which is already approved in China for use in nasopharyngeal cancer but is also being explored in other cancer types. At ASCO, data will be presented in patients with KRAS wild-type pancreatic cancer (abstract 4011). This study, like the other trials with nimotuzumab, was conducted in China.

This brings up an important point about the data the Food and Drug Administration requires for new drug approvals, commented Dr. Gralow.

She noted that the FDA recently rejected an application for sintilimab, a drug also developed in China, on the basis that all trial data submitted for approval were from China. The agency said it would like to see multiregional clinical trials and trials that reflect the U.S. cancer population.
 

Advice for attendees

A large trial in a rare cancer promises to establish a new standard of care, where previously a number of different regimens have been used in various parts of the world, and even at different hospitals within the same country. These are the results from an international trial in children and adolescents/young adults with Ewing’s sarcoma (abstract LBA-02). “I have been told by experts in the field that these results will change practice ... [and] will have a global impact,” commented Dr. Gralow.

In addition to the scientific sessions that will see new data, there are a number of educational sessions that will tackle tricky issues that clinicians sometimes face. “Microaggressions, Bias, and Equity in the Workplace” will be discussed in one session, while another promises, “Strategies to Address Moral Distress in Clinicians: What Should We Do When We Don’t Know What to Do?”

There is also a special session featuring the “Cancer Groundshot: Addressing the Global and National Inequities in Cancer Care.” This is a move spearheaded by Bishal Gyawali, MD, PhD, from Brigham and Women’s Hospital, Boston, who was reacting to the lofty goals of the presidential Cancer Moonshot, including the aim of “ending cancer as we know it.” In a blog post in 2016 he suggested “forget the moon; let’s get back to blood and flesh reality on the ground ... [and] research that can be immediately applied to every global community.” He recounts the journey from ‘Blog Post to ASCO Session’ in a recent commentary.

Dr. Gyawali also has some advice for those attending the ASCO annual meeting: Reach out to people you respect, trust that connections will happen, scrutinize the data, listen critically for jargon, and perhaps most importantly, have fun.

“There’s more to life than your job,” he wrote. “Don’t stress. Think about the bigger picture. Think about your patients. And remember, life is beautiful, even when it feels like it isn’t.”

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

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

The biggest cancer conference in the world is back in person after 2 years online during the COVID pandemic. And it appears many are eager to attend the American Society of Clinical Oncology annual meeting in person now that they can.

By early May, ASCO already had 30,000 registrations, of which 80% were in person – there were 27,000 hotel reservations.

“That’s almost identical to where we were in terms of numbers in 2019 at the same point in time,” Julie Gralow, MD, chief medical officer at ASCO, said in an interview.

These figures, which are from May 11, are likely to increase. In past years, there has been an upswing in registrations right before the meeting starts.

The annual meeting begins on Friday, June 3, and runs until Tuesday, June 7. It will be held in Chicago, yet again, in the vast McCormick Place, sections of which were transformed into field hospital wards when the pandemic hit in 2020.

But the meeting will also continue to be transmitted virtually, as it has been for the past 2 years, for those not attending in person.

“I do think that the hybrid model will move forward,” Dr. Gralow said. “We can get a lot of attendees, especially from very distant places, who can’t travel, or can’t easily travel, and we have learned how to make that experience better for them as well.”

Attendees can also change their minds if, for example, rising numbers of COVID cases as the meeting nears put them off traveling. “We are allowing people to change to virtual. So I think there may be a little bit of that, depending on what happens to COVID in different parts of the world,” Dr. Gralow commented.

For those who do attend, the organization is “doing the best that we can to keep people safe,” said Dr. Gralow, who was previously a professor of global health and is now a breast medical oncologist and clinical trialist.

To attend in person, ASCO is mandating proof of vaccination (which in the United States means two doses of the COVID vaccine). “If you prove in advance that you are vaccinated, we will send you your badge, so you don’t have to stand in line,” she added.

“As far as masks go, we are saying right now that we are complying with Chicago’s rules, which mean there is no mandatory indoor masking,” she continued. “We are recommending masking because this is a group of physicians who treat immunocompromised patients. So we are recommending that.”

This stance has gotten some push-back on Twitter from both physicians and patient advocates, with some surprised that masking is not mandatory.

“I know that ‘mask-optional’ meetings mean most will omit masks; I literally just saw this at my last meeting as one of the few masked MDs,” commented radiation oncologist Fumiko Ladd Chino, MD. She appealed to the organizers with a plea: “There’s still time to change #ASCO22 policies. We’re in it for patient health.”

Patient advocate Manju George, MVSc, PhD, a rectal cancer survivor, was also campaigning for a change in policy by setting up a letter that others could sign, adding that “ASCO leadership is being flooded with pleads from concerned HCPs.”

When asked whether it was considering a change in mask policy, ASCO replied: “As far as health and safety go, the protocols we’ve put in place meet or exceed current [World Health Organization, [Centers for Disease Control and Prevention, and city of Chicago guidelines. ASCO is also closely coordinating with both the city and the convention center and we are actively monitoring local conditions.”

“To protect the health and safety of all meeting attendees, our protocols require attendees to be fully vaccinated and self-test negative for COVID-19 within 48 hours prior to their arrival at the meeting. In addition, we expect all attendees to be masked when indoors and are encouraging regular self-testing. We fully expect members of our community to do their part to help keep everyone safe, and we’re making it easy for attendees to comply with our policies by providing medical-grade masks as well as both rapid antigen and [polymerase chain reaction] COVID-19 tests,” the organization said.

There will also be a notification system so attendees can select how they identify for closeness, with red meaning stand back, no hugs, no handshakes; yellow signifying something more intermediate; and green signaling the person is okay with contact with a handshake or a hug. This system has already been used during smaller ASCO subspecialty meetings earlier this year, and feedback from delegates was positive, Dr. Gralow commented.
 

 

 

Advancing equitable care

The theme of the 2022 meeting, chosen by ASCO President Everett Vokes, MD, is advancing equitable cancer care through innovation.

It builds on the theme of equity from 2021, chosen by previous president Lori Pierce, MD, which was “Equity: Every Patient. Every Day. Everywhere.”

Some of this relates to disparities in equity, commented Dr. Gralow. This is the focus of a premeeting press briefing on May 26 that will highlight a few abstracts that focus on disparities and what can be done to address them. One study (abstract 6511) focuses on telemedicine, which was increasingly used during the pandemic, but the results show not all U.S. patient populations could access the specialty care they needed in this way.
 

De-escalation of therapy

De-escalation of therapy is another theme running through the meeting.

“There are some cancers where we have achieved such good outcomes that it is time to look at de-escalating therapy because we know that we are probably way overtreating a component of those patients. ... So we are looking at whether we can find subpopulations where we can back off on therapy,” commented Dr. Gralow.

One example is the LUMINA trial in breast cancer (abstract LBA501), which looked at omitting radiotherapy after surgery. “In standard practice we have already been doing this, not based on solid data, but based on an accumulation of retrospective analyses and similar evidence,” commented Dr. Gralow. This trial tested the approach prospectively, lowered the age range of patients, and better defined which patients were likely to benefit.

Another example is the DYNAMIC trial in colorectal cancer (abstract LBA-100), which looks at omitting chemotherapy based on levels of circulating tumor DNA after surgery. These patients had stage 2 disease and generally do very well with surgery and adjuvant chemotherapy, Dr. Gralow stated. This trial aims to find the subset of patients who could do just as well without the chemotherapy; it may also identify those patients at the other end of the scale, who perhaps need a bit more treatment, she added.
 

Spotlight on innovation

The focus on innovation includes exploring drugs developed outside the United States. One example is nimotuzumab, which is already approved in China for use in nasopharyngeal cancer but is also being explored in other cancer types. At ASCO, data will be presented in patients with KRAS wild-type pancreatic cancer (abstract 4011). This study, like the other trials with nimotuzumab, was conducted in China.

This brings up an important point about the data the Food and Drug Administration requires for new drug approvals, commented Dr. Gralow.

She noted that the FDA recently rejected an application for sintilimab, a drug also developed in China, on the basis that all trial data submitted for approval were from China. The agency said it would like to see multiregional clinical trials and trials that reflect the U.S. cancer population.
 

Advice for attendees

A large trial in a rare cancer promises to establish a new standard of care, where previously a number of different regimens have been used in various parts of the world, and even at different hospitals within the same country. These are the results from an international trial in children and adolescents/young adults with Ewing’s sarcoma (abstract LBA-02). “I have been told by experts in the field that these results will change practice ... [and] will have a global impact,” commented Dr. Gralow.

In addition to the scientific sessions that will see new data, there are a number of educational sessions that will tackle tricky issues that clinicians sometimes face. “Microaggressions, Bias, and Equity in the Workplace” will be discussed in one session, while another promises, “Strategies to Address Moral Distress in Clinicians: What Should We Do When We Don’t Know What to Do?”

There is also a special session featuring the “Cancer Groundshot: Addressing the Global and National Inequities in Cancer Care.” This is a move spearheaded by Bishal Gyawali, MD, PhD, from Brigham and Women’s Hospital, Boston, who was reacting to the lofty goals of the presidential Cancer Moonshot, including the aim of “ending cancer as we know it.” In a blog post in 2016 he suggested “forget the moon; let’s get back to blood and flesh reality on the ground ... [and] research that can be immediately applied to every global community.” He recounts the journey from ‘Blog Post to ASCO Session’ in a recent commentary.

Dr. Gyawali also has some advice for those attending the ASCO annual meeting: Reach out to people you respect, trust that connections will happen, scrutinize the data, listen critically for jargon, and perhaps most importantly, have fun.

“There’s more to life than your job,” he wrote. “Don’t stress. Think about the bigger picture. Think about your patients. And remember, life is beautiful, even when it feels like it isn’t.”

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

The biggest cancer conference in the world is back in person after 2 years online during the COVID pandemic. And it appears many are eager to attend the American Society of Clinical Oncology annual meeting in person now that they can.

By early May, ASCO already had 30,000 registrations, of which 80% were in person – there were 27,000 hotel reservations.

“That’s almost identical to where we were in terms of numbers in 2019 at the same point in time,” Julie Gralow, MD, chief medical officer at ASCO, said in an interview.

These figures, which are from May 11, are likely to increase. In past years, there has been an upswing in registrations right before the meeting starts.

The annual meeting begins on Friday, June 3, and runs until Tuesday, June 7. It will be held in Chicago, yet again, in the vast McCormick Place, sections of which were transformed into field hospital wards when the pandemic hit in 2020.

But the meeting will also continue to be transmitted virtually, as it has been for the past 2 years, for those not attending in person.

“I do think that the hybrid model will move forward,” Dr. Gralow said. “We can get a lot of attendees, especially from very distant places, who can’t travel, or can’t easily travel, and we have learned how to make that experience better for them as well.”

Attendees can also change their minds if, for example, rising numbers of COVID cases as the meeting nears put them off traveling. “We are allowing people to change to virtual. So I think there may be a little bit of that, depending on what happens to COVID in different parts of the world,” Dr. Gralow commented.

For those who do attend, the organization is “doing the best that we can to keep people safe,” said Dr. Gralow, who was previously a professor of global health and is now a breast medical oncologist and clinical trialist.

To attend in person, ASCO is mandating proof of vaccination (which in the United States means two doses of the COVID vaccine). “If you prove in advance that you are vaccinated, we will send you your badge, so you don’t have to stand in line,” she added.

“As far as masks go, we are saying right now that we are complying with Chicago’s rules, which mean there is no mandatory indoor masking,” she continued. “We are recommending masking because this is a group of physicians who treat immunocompromised patients. So we are recommending that.”

This stance has gotten some push-back on Twitter from both physicians and patient advocates, with some surprised that masking is not mandatory.

“I know that ‘mask-optional’ meetings mean most will omit masks; I literally just saw this at my last meeting as one of the few masked MDs,” commented radiation oncologist Fumiko Ladd Chino, MD. She appealed to the organizers with a plea: “There’s still time to change #ASCO22 policies. We’re in it for patient health.”

Patient advocate Manju George, MVSc, PhD, a rectal cancer survivor, was also campaigning for a change in policy by setting up a letter that others could sign, adding that “ASCO leadership is being flooded with pleads from concerned HCPs.”

When asked whether it was considering a change in mask policy, ASCO replied: “As far as health and safety go, the protocols we’ve put in place meet or exceed current [World Health Organization, [Centers for Disease Control and Prevention, and city of Chicago guidelines. ASCO is also closely coordinating with both the city and the convention center and we are actively monitoring local conditions.”

“To protect the health and safety of all meeting attendees, our protocols require attendees to be fully vaccinated and self-test negative for COVID-19 within 48 hours prior to their arrival at the meeting. In addition, we expect all attendees to be masked when indoors and are encouraging regular self-testing. We fully expect members of our community to do their part to help keep everyone safe, and we’re making it easy for attendees to comply with our policies by providing medical-grade masks as well as both rapid antigen and [polymerase chain reaction] COVID-19 tests,” the organization said.

There will also be a notification system so attendees can select how they identify for closeness, with red meaning stand back, no hugs, no handshakes; yellow signifying something more intermediate; and green signaling the person is okay with contact with a handshake or a hug. This system has already been used during smaller ASCO subspecialty meetings earlier this year, and feedback from delegates was positive, Dr. Gralow commented.
 

 

 

Advancing equitable care

The theme of the 2022 meeting, chosen by ASCO President Everett Vokes, MD, is advancing equitable cancer care through innovation.

It builds on the theme of equity from 2021, chosen by previous president Lori Pierce, MD, which was “Equity: Every Patient. Every Day. Everywhere.”

Some of this relates to disparities in equity, commented Dr. Gralow. This is the focus of a premeeting press briefing on May 26 that will highlight a few abstracts that focus on disparities and what can be done to address them. One study (abstract 6511) focuses on telemedicine, which was increasingly used during the pandemic, but the results show not all U.S. patient populations could access the specialty care they needed in this way.
 

De-escalation of therapy

De-escalation of therapy is another theme running through the meeting.

“There are some cancers where we have achieved such good outcomes that it is time to look at de-escalating therapy because we know that we are probably way overtreating a component of those patients. ... So we are looking at whether we can find subpopulations where we can back off on therapy,” commented Dr. Gralow.

One example is the LUMINA trial in breast cancer (abstract LBA501), which looked at omitting radiotherapy after surgery. “In standard practice we have already been doing this, not based on solid data, but based on an accumulation of retrospective analyses and similar evidence,” commented Dr. Gralow. This trial tested the approach prospectively, lowered the age range of patients, and better defined which patients were likely to benefit.

Another example is the DYNAMIC trial in colorectal cancer (abstract LBA-100), which looks at omitting chemotherapy based on levels of circulating tumor DNA after surgery. These patients had stage 2 disease and generally do very well with surgery and adjuvant chemotherapy, Dr. Gralow stated. This trial aims to find the subset of patients who could do just as well without the chemotherapy; it may also identify those patients at the other end of the scale, who perhaps need a bit more treatment, she added.
 

Spotlight on innovation

The focus on innovation includes exploring drugs developed outside the United States. One example is nimotuzumab, which is already approved in China for use in nasopharyngeal cancer but is also being explored in other cancer types. At ASCO, data will be presented in patients with KRAS wild-type pancreatic cancer (abstract 4011). This study, like the other trials with nimotuzumab, was conducted in China.

This brings up an important point about the data the Food and Drug Administration requires for new drug approvals, commented Dr. Gralow.

She noted that the FDA recently rejected an application for sintilimab, a drug also developed in China, on the basis that all trial data submitted for approval were from China. The agency said it would like to see multiregional clinical trials and trials that reflect the U.S. cancer population.
 

Advice for attendees

A large trial in a rare cancer promises to establish a new standard of care, where previously a number of different regimens have been used in various parts of the world, and even at different hospitals within the same country. These are the results from an international trial in children and adolescents/young adults with Ewing’s sarcoma (abstract LBA-02). “I have been told by experts in the field that these results will change practice ... [and] will have a global impact,” commented Dr. Gralow.

In addition to the scientific sessions that will see new data, there are a number of educational sessions that will tackle tricky issues that clinicians sometimes face. “Microaggressions, Bias, and Equity in the Workplace” will be discussed in one session, while another promises, “Strategies to Address Moral Distress in Clinicians: What Should We Do When We Don’t Know What to Do?”

There is also a special session featuring the “Cancer Groundshot: Addressing the Global and National Inequities in Cancer Care.” This is a move spearheaded by Bishal Gyawali, MD, PhD, from Brigham and Women’s Hospital, Boston, who was reacting to the lofty goals of the presidential Cancer Moonshot, including the aim of “ending cancer as we know it.” In a blog post in 2016 he suggested “forget the moon; let’s get back to blood and flesh reality on the ground ... [and] research that can be immediately applied to every global community.” He recounts the journey from ‘Blog Post to ASCO Session’ in a recent commentary.

Dr. Gyawali also has some advice for those attending the ASCO annual meeting: Reach out to people you respect, trust that connections will happen, scrutinize the data, listen critically for jargon, and perhaps most importantly, have fun.

“There’s more to life than your job,” he wrote. “Don’t stress. Think about the bigger picture. Think about your patients. And remember, life is beautiful, even when it feels like it isn’t.”

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

Immunotherapy now first line for esophageal cancer

Article Type
Changed
Thu, 12/15/2022 - 14:31

Immunotherapy with nivolumab (Opdivo) is now approved in the United States for first-line use in the treatment of unresectable advanced or metastatic esophageal squamous cell carcinoma (ESCC).

The new approval for the drug, a programmed cell death–ligand-1 inhibitor, is for use in this patient population regardless of PD-L1 status.

The indication also specifies that nivolumab is to be used together with chemotherapy (with a fluoropyrimidine- and platinum-containing regimen) or in combination with ipilimumab (Yervoy), an immunotherapy with a different mechanism of action.

“Today’s approvals bring two first-line immunotherapy-based treatment options at once ... to newly diagnosed patients with unresectable advanced or metastatic ESCC,” commented Adam Lenkowsky, a senior vice president at Bristol-Myers Squibb, which makes both nivolumab and ipilimumab.

The approval of the new indication by the Food and Drug Administration was based on improved survival shown in the phase 3 CheckMate-648 trial, which involved nearly 1,000 patients. The trial had three arms and compared nivolumab plus chemotherapy (n = 321) and nivolumab plus ipilimumab (n = 324) with chemotherapy alone (n = 324).

The results showed improved survival with both nivolumab combinations compared with chemotherapy (fluorouracil and cisplatin) alone. Overall survival was improved both in all randomized patients (a secondary endpoint) and in patients whose tumors expressed PD-L1 (≥ 1%), the primary endpoint.

For the combination of nivolumab plus chemotherapy, median overall survival was 13.2 versus 10.7 months, compared with chemotherapy alone in all randomized patients, and 15.4 versus 9.1 months in patients whose tumors express PD-L1 (≥ 1%).

For the combination of nivolumab plus ipilimumab, median overall survival was 12.8 versus 10.7 months with chemotherapy alone in all randomized patients and 13.7 versus 9.1 months in patients whose tumors express PD-L1 (≥ 1%).

However, progression-free survival did not reach statistical significance in any group.

“Unresectable advanced or metastatic ESCC is a challenging disease, and there’s a need for additional treatment options that may extend survival in the first-line setting,” commented Jaffer A. Ajani, MD, professor of gastrointestinal medical oncology at the University of Texas MD Anderson Cancer Center, Houston. He was also the lead U.S. investigator for CheckMate-648 and, in a company press release, said the “two nivolumab-based combinations showed a survival benefit compared to chemotherapy alone, offering new treatment options regardless of PD-L1 status.”

Results from the trial were presented at the 2021 annual meeting of the American Society of Clinical Oncology. At that time, trial investigator Ian Chau, MD, a consultant medical oncologist at the Royal Marsden Hospital in Sutton, England, told attendees that “nivolumab plus chemotherapy and nivolumab plus ipilimumab each represent a new potential first-line standard of care for patients with advanced ESCC.”

Commenting on that presentation, Samuel J. Klempner, MD, a gastrointestinal medical oncologist at the Massachusetts General Hospital Cancer Center, Boston, noted that the “prospect of a chemo-free regimen for advanced ESCC with the well-studied combination of ipilimumab and nivolumab would represent a welcome addition to our treatment armamentarium.”
 

No new safety signals

Dr. Chau noted there were no new safety signals with either of the immunotherapies.

Nivolumab and/or chemotherapy were discontinued in 39% of patients and delayed in 71% of patients for an adverse reaction.

Nivolumab and/or ipilimumab were discontinued in 23% of patients and delayed in 46% of patients for an adverse reaction.

The manufacturer cautioned that immunotherapy with nivolumab with or without ipilimumab has been associated with severe and fatal immune-mediated adverse reactions including pneumonitis, colitis, hepatitis and hepatotoxicity, endocrinopathies, nephritis and renal dysfunction, dermatologic adverse reactions, and infusion-related reactions.

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

Publications
Topics
Sections

Immunotherapy with nivolumab (Opdivo) is now approved in the United States for first-line use in the treatment of unresectable advanced or metastatic esophageal squamous cell carcinoma (ESCC).

The new approval for the drug, a programmed cell death–ligand-1 inhibitor, is for use in this patient population regardless of PD-L1 status.

The indication also specifies that nivolumab is to be used together with chemotherapy (with a fluoropyrimidine- and platinum-containing regimen) or in combination with ipilimumab (Yervoy), an immunotherapy with a different mechanism of action.

“Today’s approvals bring two first-line immunotherapy-based treatment options at once ... to newly diagnosed patients with unresectable advanced or metastatic ESCC,” commented Adam Lenkowsky, a senior vice president at Bristol-Myers Squibb, which makes both nivolumab and ipilimumab.

The approval of the new indication by the Food and Drug Administration was based on improved survival shown in the phase 3 CheckMate-648 trial, which involved nearly 1,000 patients. The trial had three arms and compared nivolumab plus chemotherapy (n = 321) and nivolumab plus ipilimumab (n = 324) with chemotherapy alone (n = 324).

The results showed improved survival with both nivolumab combinations compared with chemotherapy (fluorouracil and cisplatin) alone. Overall survival was improved both in all randomized patients (a secondary endpoint) and in patients whose tumors expressed PD-L1 (≥ 1%), the primary endpoint.

For the combination of nivolumab plus chemotherapy, median overall survival was 13.2 versus 10.7 months, compared with chemotherapy alone in all randomized patients, and 15.4 versus 9.1 months in patients whose tumors express PD-L1 (≥ 1%).

For the combination of nivolumab plus ipilimumab, median overall survival was 12.8 versus 10.7 months with chemotherapy alone in all randomized patients and 13.7 versus 9.1 months in patients whose tumors express PD-L1 (≥ 1%).

However, progression-free survival did not reach statistical significance in any group.

“Unresectable advanced or metastatic ESCC is a challenging disease, and there’s a need for additional treatment options that may extend survival in the first-line setting,” commented Jaffer A. Ajani, MD, professor of gastrointestinal medical oncology at the University of Texas MD Anderson Cancer Center, Houston. He was also the lead U.S. investigator for CheckMate-648 and, in a company press release, said the “two nivolumab-based combinations showed a survival benefit compared to chemotherapy alone, offering new treatment options regardless of PD-L1 status.”

Results from the trial were presented at the 2021 annual meeting of the American Society of Clinical Oncology. At that time, trial investigator Ian Chau, MD, a consultant medical oncologist at the Royal Marsden Hospital in Sutton, England, told attendees that “nivolumab plus chemotherapy and nivolumab plus ipilimumab each represent a new potential first-line standard of care for patients with advanced ESCC.”

Commenting on that presentation, Samuel J. Klempner, MD, a gastrointestinal medical oncologist at the Massachusetts General Hospital Cancer Center, Boston, noted that the “prospect of a chemo-free regimen for advanced ESCC with the well-studied combination of ipilimumab and nivolumab would represent a welcome addition to our treatment armamentarium.”
 

No new safety signals

Dr. Chau noted there were no new safety signals with either of the immunotherapies.

Nivolumab and/or chemotherapy were discontinued in 39% of patients and delayed in 71% of patients for an adverse reaction.

Nivolumab and/or ipilimumab were discontinued in 23% of patients and delayed in 46% of patients for an adverse reaction.

The manufacturer cautioned that immunotherapy with nivolumab with or without ipilimumab has been associated with severe and fatal immune-mediated adverse reactions including pneumonitis, colitis, hepatitis and hepatotoxicity, endocrinopathies, nephritis and renal dysfunction, dermatologic adverse reactions, and infusion-related reactions.

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

Immunotherapy with nivolumab (Opdivo) is now approved in the United States for first-line use in the treatment of unresectable advanced or metastatic esophageal squamous cell carcinoma (ESCC).

The new approval for the drug, a programmed cell death–ligand-1 inhibitor, is for use in this patient population regardless of PD-L1 status.

The indication also specifies that nivolumab is to be used together with chemotherapy (with a fluoropyrimidine- and platinum-containing regimen) or in combination with ipilimumab (Yervoy), an immunotherapy with a different mechanism of action.

“Today’s approvals bring two first-line immunotherapy-based treatment options at once ... to newly diagnosed patients with unresectable advanced or metastatic ESCC,” commented Adam Lenkowsky, a senior vice president at Bristol-Myers Squibb, which makes both nivolumab and ipilimumab.

The approval of the new indication by the Food and Drug Administration was based on improved survival shown in the phase 3 CheckMate-648 trial, which involved nearly 1,000 patients. The trial had three arms and compared nivolumab plus chemotherapy (n = 321) and nivolumab plus ipilimumab (n = 324) with chemotherapy alone (n = 324).

The results showed improved survival with both nivolumab combinations compared with chemotherapy (fluorouracil and cisplatin) alone. Overall survival was improved both in all randomized patients (a secondary endpoint) and in patients whose tumors expressed PD-L1 (≥ 1%), the primary endpoint.

For the combination of nivolumab plus chemotherapy, median overall survival was 13.2 versus 10.7 months, compared with chemotherapy alone in all randomized patients, and 15.4 versus 9.1 months in patients whose tumors express PD-L1 (≥ 1%).

For the combination of nivolumab plus ipilimumab, median overall survival was 12.8 versus 10.7 months with chemotherapy alone in all randomized patients and 13.7 versus 9.1 months in patients whose tumors express PD-L1 (≥ 1%).

However, progression-free survival did not reach statistical significance in any group.

“Unresectable advanced or metastatic ESCC is a challenging disease, and there’s a need for additional treatment options that may extend survival in the first-line setting,” commented Jaffer A. Ajani, MD, professor of gastrointestinal medical oncology at the University of Texas MD Anderson Cancer Center, Houston. He was also the lead U.S. investigator for CheckMate-648 and, in a company press release, said the “two nivolumab-based combinations showed a survival benefit compared to chemotherapy alone, offering new treatment options regardless of PD-L1 status.”

Results from the trial were presented at the 2021 annual meeting of the American Society of Clinical Oncology. At that time, trial investigator Ian Chau, MD, a consultant medical oncologist at the Royal Marsden Hospital in Sutton, England, told attendees that “nivolumab plus chemotherapy and nivolumab plus ipilimumab each represent a new potential first-line standard of care for patients with advanced ESCC.”

Commenting on that presentation, Samuel J. Klempner, MD, a gastrointestinal medical oncologist at the Massachusetts General Hospital Cancer Center, Boston, noted that the “prospect of a chemo-free regimen for advanced ESCC with the well-studied combination of ipilimumab and nivolumab would represent a welcome addition to our treatment armamentarium.”
 

No new safety signals

Dr. Chau noted there were no new safety signals with either of the immunotherapies.

Nivolumab and/or chemotherapy were discontinued in 39% of patients and delayed in 71% of patients for an adverse reaction.

Nivolumab and/or ipilimumab were discontinued in 23% of patients and delayed in 46% of patients for an adverse reaction.

The manufacturer cautioned that immunotherapy with nivolumab with or without ipilimumab has been associated with severe and fatal immune-mediated adverse reactions including pneumonitis, colitis, hepatitis and hepatotoxicity, endocrinopathies, nephritis and renal dysfunction, dermatologic adverse reactions, and infusion-related reactions.

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

FDA approves first drug for myelofibrosis with thrombocytopenia

Article Type
Changed
Wed, 03/02/2022 - 08:02

 

The Food and Drug Administration has granted accelerated approval for a new drug for the treatment of myelofibrosis, the first specifically for patients with low platelet counts.

Pacritinib (Vonjo, CTI BioPharma) is indicated for use in the treatment of adults with intermediate- or high-risk primary or secondary (post–polycythemia vera or post–essential thrombocythemia) myelofibrosis with a platelet count below 50 × 109/L.

Pacritinib is a novel oral kinase inhibitor with specificity for activity against Janus associated kinase 2 (JAK2) and IRAK1, without inhibiting JAK1. The recommended dosage is 200 mg orally twice daily.

In the United States, there are approximately 21,000 patients with myelofibrosis, notes the manufacturer. About one-third develop severe thrombocytopenia.

“Myelofibrosis with severe thrombocytopenia, defined as blood platelet counts below 50 × 109/L, has been shown to result in poor survival outcomes coupled with debilitating symptoms. Limited treatment options have rendered this disease as an area of urgent unmet medical need,” said John Mascarenhas, MD, associate professor, medicine, hematology, and medical oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York.

“I am pleased to see that a new, efficacious, and safe treatment option is now available for these patients,” he said in a company press release.

Dr. Mascarenhas was the lead investigator of the phase 3 PERSIST-2 trial that was the basis for the approval. Results from the trial were published in 2018 in JAMA Oncology and reported in detail at the time by this news organization.

Authors of an accompanying editorial noted the trial was truncated after the FDA imposed a clinical hold on pacritinib in February 2016 after reports from an earlier trial, PERSIST-1, of patient deaths related to cardiac failure and arrest as well as intracranial hemorrhage. The clinical hold was lifted in January 2017 after the manufacturer provided the FDA with more mature data.

Despite the truncation, the PERSIST-2 trial provided sufficient data to obtain accelerated approval for the drug. The study compared pacritinib with best available therapy (BAT).

In the cohort of patients treated with pacritinib 200 mg twice daily, 29% of patients had a reduction in spleen volume of at least 35% compared with 3% of patients receiving BAT, which included ruxolitinib.

The company is now expected to demonstrate clinical benefit in a confirmatory trial and has the PACIFICA trial underway. Results are expected in mid-2025.

The most common adverse reactions (reported by ≥ 20% of patients) were diarrhea, thrombocytopenia, nausea, anemia, and peripheral edema. The most frequent serious adverse reactions (≥ 3%) were anemia, thrombocytopenia, pneumonia, cardiac failure, disease progression, pyrexia, and squamous cell carcinoma of the skin.

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

Publications
Topics
Sections

 

The Food and Drug Administration has granted accelerated approval for a new drug for the treatment of myelofibrosis, the first specifically for patients with low platelet counts.

Pacritinib (Vonjo, CTI BioPharma) is indicated for use in the treatment of adults with intermediate- or high-risk primary or secondary (post–polycythemia vera or post–essential thrombocythemia) myelofibrosis with a platelet count below 50 × 109/L.

Pacritinib is a novel oral kinase inhibitor with specificity for activity against Janus associated kinase 2 (JAK2) and IRAK1, without inhibiting JAK1. The recommended dosage is 200 mg orally twice daily.

In the United States, there are approximately 21,000 patients with myelofibrosis, notes the manufacturer. About one-third develop severe thrombocytopenia.

“Myelofibrosis with severe thrombocytopenia, defined as blood platelet counts below 50 × 109/L, has been shown to result in poor survival outcomes coupled with debilitating symptoms. Limited treatment options have rendered this disease as an area of urgent unmet medical need,” said John Mascarenhas, MD, associate professor, medicine, hematology, and medical oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York.

“I am pleased to see that a new, efficacious, and safe treatment option is now available for these patients,” he said in a company press release.

Dr. Mascarenhas was the lead investigator of the phase 3 PERSIST-2 trial that was the basis for the approval. Results from the trial were published in 2018 in JAMA Oncology and reported in detail at the time by this news organization.

Authors of an accompanying editorial noted the trial was truncated after the FDA imposed a clinical hold on pacritinib in February 2016 after reports from an earlier trial, PERSIST-1, of patient deaths related to cardiac failure and arrest as well as intracranial hemorrhage. The clinical hold was lifted in January 2017 after the manufacturer provided the FDA with more mature data.

Despite the truncation, the PERSIST-2 trial provided sufficient data to obtain accelerated approval for the drug. The study compared pacritinib with best available therapy (BAT).

In the cohort of patients treated with pacritinib 200 mg twice daily, 29% of patients had a reduction in spleen volume of at least 35% compared with 3% of patients receiving BAT, which included ruxolitinib.

The company is now expected to demonstrate clinical benefit in a confirmatory trial and has the PACIFICA trial underway. Results are expected in mid-2025.

The most common adverse reactions (reported by ≥ 20% of patients) were diarrhea, thrombocytopenia, nausea, anemia, and peripheral edema. The most frequent serious adverse reactions (≥ 3%) were anemia, thrombocytopenia, pneumonia, cardiac failure, disease progression, pyrexia, and squamous cell carcinoma of the skin.

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

 

The Food and Drug Administration has granted accelerated approval for a new drug for the treatment of myelofibrosis, the first specifically for patients with low platelet counts.

Pacritinib (Vonjo, CTI BioPharma) is indicated for use in the treatment of adults with intermediate- or high-risk primary or secondary (post–polycythemia vera or post–essential thrombocythemia) myelofibrosis with a platelet count below 50 × 109/L.

Pacritinib is a novel oral kinase inhibitor with specificity for activity against Janus associated kinase 2 (JAK2) and IRAK1, without inhibiting JAK1. The recommended dosage is 200 mg orally twice daily.

In the United States, there are approximately 21,000 patients with myelofibrosis, notes the manufacturer. About one-third develop severe thrombocytopenia.

“Myelofibrosis with severe thrombocytopenia, defined as blood platelet counts below 50 × 109/L, has been shown to result in poor survival outcomes coupled with debilitating symptoms. Limited treatment options have rendered this disease as an area of urgent unmet medical need,” said John Mascarenhas, MD, associate professor, medicine, hematology, and medical oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York.

“I am pleased to see that a new, efficacious, and safe treatment option is now available for these patients,” he said in a company press release.

Dr. Mascarenhas was the lead investigator of the phase 3 PERSIST-2 trial that was the basis for the approval. Results from the trial were published in 2018 in JAMA Oncology and reported in detail at the time by this news organization.

Authors of an accompanying editorial noted the trial was truncated after the FDA imposed a clinical hold on pacritinib in February 2016 after reports from an earlier trial, PERSIST-1, of patient deaths related to cardiac failure and arrest as well as intracranial hemorrhage. The clinical hold was lifted in January 2017 after the manufacturer provided the FDA with more mature data.

Despite the truncation, the PERSIST-2 trial provided sufficient data to obtain accelerated approval for the drug. The study compared pacritinib with best available therapy (BAT).

In the cohort of patients treated with pacritinib 200 mg twice daily, 29% of patients had a reduction in spleen volume of at least 35% compared with 3% of patients receiving BAT, which included ruxolitinib.

The company is now expected to demonstrate clinical benefit in a confirmatory trial and has the PACIFICA trial underway. Results are expected in mid-2025.

The most common adverse reactions (reported by ≥ 20% of patients) were diarrhea, thrombocytopenia, nausea, anemia, and peripheral edema. The most frequent serious adverse reactions (≥ 3%) were anemia, thrombocytopenia, pneumonia, cardiac failure, disease progression, pyrexia, and squamous cell carcinoma of the skin.

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

FDA approves new CAR T-cell treatment for multiple myeloma

Article Type
Changed
Wed, 03/02/2022 - 08:04

A new treatment option for patients with refractory/relapsed multiple myeloma who have already tried four or more therapies has been approved by the U.S. Food and Drug Administration.

The product, ciltacabtagene autoleucel (cilta-cel), will be marketed as Carvykti by Janssen and Legend Biotech. It is a chimeric antigen receptor (CAR) T-cell therapy directed against B-cell maturation antigen (BCMA), which is a new target for therapies for multiple myeloma.

There are already two other therapies on the market that target BCMA – another CAR T cell, idecabtagene vicleucel (Abecma), which was approved by the FDA in March 2021, and a drug conjugate, belantamab mafodotin (Blenrep), which was approved in August 2020.

The approval of cilta-cel was based on clinical data from the CARTITUDE-1 study, which were initially presented in December 2020 at the annual meeting of the American Society of Hematology, as reported at the time by this news organization.

The trial involved 97 patients with relapsed/refractory multiple myeloma who had already received a median of six previous treatments (range, three to 18), including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 monoclonal antibody.

“The treatment journey for the majority of patients living with multiple myeloma is a relentless cycle of remission and relapse, with fewer patients achieving a deep response as they progress through later lines of therapy,” commented Sundar Jagannath, MBBS, professor of medicine, hematology, and medical oncology at Mount Sinai, who was a principal investigator on the pivotal study.

“That is why I have been really excited about the results from the CARTITUDE-1 study, which has demonstrated that cilta-cel can provide deep and durable responses and long-term treatment-free intervals, even in this heavily pretreated multiple myeloma patient population,” he said.

“Today’s approval of Carvykti helps address a great unmet need for these patients,” he commented in a press release from the manufacturer.

Like other CAR T-cell therapies, ciltacabtagene autoleucel is a one-time treatment. It involves collecting blood from the patient, extracting T cells, genetically engineering them, then transfusing them back to the patient, who in the meantime has undergone conditioning.

The results from CARTITUDE-1 show that this one-time treatment resulted in deep and durable responses.

The overall response rate was 98%, and the majority of patients (78%) achieved a stringent complete response, in which physicians are unable to observe any signs or symptoms of disease via imaging or other tests after treatment.

At a median of 18 months’ follow-up, the median duration of response was 21.8 months.

“The responses in the CARTITUDE-1 study showed durability over time and resulted in the majority of heavily pretreated patients achieving deep responses after 18-month follow-up,” commented Mr. Jagannath.

“The approval of cilta-cel provides physicians an immunotherapy treatment option that offers patients an opportunity to be free from anti-myeloma therapies for a period of time,” he added.

As with other CAR T-cell therapies, there were serious side effects, and these products are available only through restricted programs under a risk evaluation and mitigation strategy.

The product information for Cartykti includes a boxed warning that mentions cytokine release syndrome (CRS), immune effector cell–associated neurotoxicity syndrome, parkinsonism, Guillain-Barré syndrome, hemophagocytic lymphohistiocytosis/macrophage activation syndrome, and prolonged and/or recurrent cytopenias.

The most common adverse reactions (reported in greater than or equal to 20% of patients) are pyrexia, CRS, hypogammaglobulinemia, hypotension, musculoskeletal pain, fatigue, infections–pathogens unspecified, cough, chills, diarrhea, nausea, encephalopathy, decreased appetite, upper respiratory tract infection, headache, tachycardia, dizziness, dyspnea, edema, viral infections, coagulopathy, constipation, and vomiting.

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

Publications
Topics
Sections

A new treatment option for patients with refractory/relapsed multiple myeloma who have already tried four or more therapies has been approved by the U.S. Food and Drug Administration.

The product, ciltacabtagene autoleucel (cilta-cel), will be marketed as Carvykti by Janssen and Legend Biotech. It is a chimeric antigen receptor (CAR) T-cell therapy directed against B-cell maturation antigen (BCMA), which is a new target for therapies for multiple myeloma.

There are already two other therapies on the market that target BCMA – another CAR T cell, idecabtagene vicleucel (Abecma), which was approved by the FDA in March 2021, and a drug conjugate, belantamab mafodotin (Blenrep), which was approved in August 2020.

The approval of cilta-cel was based on clinical data from the CARTITUDE-1 study, which were initially presented in December 2020 at the annual meeting of the American Society of Hematology, as reported at the time by this news organization.

The trial involved 97 patients with relapsed/refractory multiple myeloma who had already received a median of six previous treatments (range, three to 18), including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 monoclonal antibody.

“The treatment journey for the majority of patients living with multiple myeloma is a relentless cycle of remission and relapse, with fewer patients achieving a deep response as they progress through later lines of therapy,” commented Sundar Jagannath, MBBS, professor of medicine, hematology, and medical oncology at Mount Sinai, who was a principal investigator on the pivotal study.

“That is why I have been really excited about the results from the CARTITUDE-1 study, which has demonstrated that cilta-cel can provide deep and durable responses and long-term treatment-free intervals, even in this heavily pretreated multiple myeloma patient population,” he said.

“Today’s approval of Carvykti helps address a great unmet need for these patients,” he commented in a press release from the manufacturer.

Like other CAR T-cell therapies, ciltacabtagene autoleucel is a one-time treatment. It involves collecting blood from the patient, extracting T cells, genetically engineering them, then transfusing them back to the patient, who in the meantime has undergone conditioning.

The results from CARTITUDE-1 show that this one-time treatment resulted in deep and durable responses.

The overall response rate was 98%, and the majority of patients (78%) achieved a stringent complete response, in which physicians are unable to observe any signs or symptoms of disease via imaging or other tests after treatment.

At a median of 18 months’ follow-up, the median duration of response was 21.8 months.

“The responses in the CARTITUDE-1 study showed durability over time and resulted in the majority of heavily pretreated patients achieving deep responses after 18-month follow-up,” commented Mr. Jagannath.

“The approval of cilta-cel provides physicians an immunotherapy treatment option that offers patients an opportunity to be free from anti-myeloma therapies for a period of time,” he added.

As with other CAR T-cell therapies, there were serious side effects, and these products are available only through restricted programs under a risk evaluation and mitigation strategy.

The product information for Cartykti includes a boxed warning that mentions cytokine release syndrome (CRS), immune effector cell–associated neurotoxicity syndrome, parkinsonism, Guillain-Barré syndrome, hemophagocytic lymphohistiocytosis/macrophage activation syndrome, and prolonged and/or recurrent cytopenias.

The most common adverse reactions (reported in greater than or equal to 20% of patients) are pyrexia, CRS, hypogammaglobulinemia, hypotension, musculoskeletal pain, fatigue, infections–pathogens unspecified, cough, chills, diarrhea, nausea, encephalopathy, decreased appetite, upper respiratory tract infection, headache, tachycardia, dizziness, dyspnea, edema, viral infections, coagulopathy, constipation, and vomiting.

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

A new treatment option for patients with refractory/relapsed multiple myeloma who have already tried four or more therapies has been approved by the U.S. Food and Drug Administration.

The product, ciltacabtagene autoleucel (cilta-cel), will be marketed as Carvykti by Janssen and Legend Biotech. It is a chimeric antigen receptor (CAR) T-cell therapy directed against B-cell maturation antigen (BCMA), which is a new target for therapies for multiple myeloma.

There are already two other therapies on the market that target BCMA – another CAR T cell, idecabtagene vicleucel (Abecma), which was approved by the FDA in March 2021, and a drug conjugate, belantamab mafodotin (Blenrep), which was approved in August 2020.

The approval of cilta-cel was based on clinical data from the CARTITUDE-1 study, which were initially presented in December 2020 at the annual meeting of the American Society of Hematology, as reported at the time by this news organization.

The trial involved 97 patients with relapsed/refractory multiple myeloma who had already received a median of six previous treatments (range, three to 18), including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 monoclonal antibody.

“The treatment journey for the majority of patients living with multiple myeloma is a relentless cycle of remission and relapse, with fewer patients achieving a deep response as they progress through later lines of therapy,” commented Sundar Jagannath, MBBS, professor of medicine, hematology, and medical oncology at Mount Sinai, who was a principal investigator on the pivotal study.

“That is why I have been really excited about the results from the CARTITUDE-1 study, which has demonstrated that cilta-cel can provide deep and durable responses and long-term treatment-free intervals, even in this heavily pretreated multiple myeloma patient population,” he said.

“Today’s approval of Carvykti helps address a great unmet need for these patients,” he commented in a press release from the manufacturer.

Like other CAR T-cell therapies, ciltacabtagene autoleucel is a one-time treatment. It involves collecting blood from the patient, extracting T cells, genetically engineering them, then transfusing them back to the patient, who in the meantime has undergone conditioning.

The results from CARTITUDE-1 show that this one-time treatment resulted in deep and durable responses.

The overall response rate was 98%, and the majority of patients (78%) achieved a stringent complete response, in which physicians are unable to observe any signs or symptoms of disease via imaging or other tests after treatment.

At a median of 18 months’ follow-up, the median duration of response was 21.8 months.

“The responses in the CARTITUDE-1 study showed durability over time and resulted in the majority of heavily pretreated patients achieving deep responses after 18-month follow-up,” commented Mr. Jagannath.

“The approval of cilta-cel provides physicians an immunotherapy treatment option that offers patients an opportunity to be free from anti-myeloma therapies for a period of time,” he added.

As with other CAR T-cell therapies, there were serious side effects, and these products are available only through restricted programs under a risk evaluation and mitigation strategy.

The product information for Cartykti includes a boxed warning that mentions cytokine release syndrome (CRS), immune effector cell–associated neurotoxicity syndrome, parkinsonism, Guillain-Barré syndrome, hemophagocytic lymphohistiocytosis/macrophage activation syndrome, and prolonged and/or recurrent cytopenias.

The most common adverse reactions (reported in greater than or equal to 20% of patients) are pyrexia, CRS, hypogammaglobulinemia, hypotension, musculoskeletal pain, fatigue, infections–pathogens unspecified, cough, chills, diarrhea, nausea, encephalopathy, decreased appetite, upper respiratory tract infection, headache, tachycardia, dizziness, dyspnea, edema, viral infections, coagulopathy, constipation, and vomiting.

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

FDA approves first drug for rare inherited anemia

Article Type
Changed
Tue, 02/22/2022 - 10:35

A new drug that is both the first in its class and the first disease-modifying agent for hemolytic anemia in adults with pyruvate kinase (PK) deficiency has been approved by the Food and Drug Administration.

The new drug, mitapivat (Pyrukynd, Agios), was approved on the basis of clinical trials that showed that it significantly improved hemolysis and anemia in patients with PK deficiency.


PK deficiency is rare. In clinical practice, its frequency is approximately 3-9 cases per 1 million people, the FDA noted. However, PK deficiency likely is misdiagnosed or undiagnosed, making it difficult to determine its frequency in the general population

PK deficiency is an inherited disorder that causes premature red blood cell destruction, leading to anemia, the agency explained in its announcement. Symptoms of PK deficiency range in severity and include fatigue, unusually pale skin, jaundice, shortness of breath, and a fast heart rate. Patients can also develop an enlarged spleen, can have too much iron in their blood from repeated blood transfusions, and can develop gallstones.

“Pyrukynd is the first approved therapy for PK deficiency and marks an important milestone for these patients, who may face tremendous challenges and debilitating symptoms throughout the course of this lifelong disease,” said Rachael Grace, MD, pediatric hematologist and director of hematology clinical research at Boston Children’s Hospital.

She was an investigator in the clinical trials that led to the approval. In a statement from the manufacturer, she added that “partnering with Agios and the PK deficiency community to improve understanding of the natural history of this rare disease and bring a new medicine to patients has been an honor, and I look forward to additional collaboration in the future.”

Clinical data

Clinical data that formed the basis of the approval came from two trials, one of which was a randomized, placebo-controlled trial, and the other a single-arm study, the FDA noted. In these studies, patients received up to 50 mg of mitapivat orally twice daily after an initial dose titration period

The randomized trial involved 80 adults with PK deficiency who were not having regular blood transfusions. They were allocated to receive either mitapivat or placebo and were followed for an average of 24 weeks. The primary endpoint was the number of patients who achieved a hemoglobin response (defined as a 1.5 g/dL or greater increase in hemoglobin concentration that was sustained at two or more scheduled assessments). At the end of the study, 40% of participants who received mitapivat had a hemoglobin response, compared with no participants who received placebo.

The single-arm study involved 27 adults with PK deficiency who were receiving regular blood transfusions. They took mitapivat for an average of 40 weeks. In this study, the primary endpoint was the reduction in transfusion burden, defined as at least a 33% reduction in the number of red blood cell units transfused during the last 24 weeks of treatment, compared with the historical transfusion burden on the individual participant (standardized to 24 weeks). The results show that 33% of participants who received mitapivat met this reduction in transfusion burden; 22% of participants did not require any transfusions over the last 24 weeks of treatment.

The most common side effects reported were decreases in estrone and estradiol in men, increased urate level, back pain, and joint stiffness. The effects of estrone and estradiol could not be reliably assessed in women because of normal changes in these hormone levels during the menstrual cycle and use of hormonal contraception.

The FDA warns of drug interactions that could necessitate dose adjustments, and also that abruptly stopping mitapivat could worsen premature red blood cell destruction.

The agency noted that this application received orphan drug designation, fast track designation, and priority review.

Agios is offering access programs aimed at reducing or eliminating patient out-of-pocket costs. Further details are on the myAgios patient support services program.

The company also noted that mitapivat is awaiting approval in the European Union. A decision is expected before the end of 2022.

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

Publications
Topics
Sections

A new drug that is both the first in its class and the first disease-modifying agent for hemolytic anemia in adults with pyruvate kinase (PK) deficiency has been approved by the Food and Drug Administration.

The new drug, mitapivat (Pyrukynd, Agios), was approved on the basis of clinical trials that showed that it significantly improved hemolysis and anemia in patients with PK deficiency.


PK deficiency is rare. In clinical practice, its frequency is approximately 3-9 cases per 1 million people, the FDA noted. However, PK deficiency likely is misdiagnosed or undiagnosed, making it difficult to determine its frequency in the general population

PK deficiency is an inherited disorder that causes premature red blood cell destruction, leading to anemia, the agency explained in its announcement. Symptoms of PK deficiency range in severity and include fatigue, unusually pale skin, jaundice, shortness of breath, and a fast heart rate. Patients can also develop an enlarged spleen, can have too much iron in their blood from repeated blood transfusions, and can develop gallstones.

“Pyrukynd is the first approved therapy for PK deficiency and marks an important milestone for these patients, who may face tremendous challenges and debilitating symptoms throughout the course of this lifelong disease,” said Rachael Grace, MD, pediatric hematologist and director of hematology clinical research at Boston Children’s Hospital.

She was an investigator in the clinical trials that led to the approval. In a statement from the manufacturer, she added that “partnering with Agios and the PK deficiency community to improve understanding of the natural history of this rare disease and bring a new medicine to patients has been an honor, and I look forward to additional collaboration in the future.”

Clinical data

Clinical data that formed the basis of the approval came from two trials, one of which was a randomized, placebo-controlled trial, and the other a single-arm study, the FDA noted. In these studies, patients received up to 50 mg of mitapivat orally twice daily after an initial dose titration period

The randomized trial involved 80 adults with PK deficiency who were not having regular blood transfusions. They were allocated to receive either mitapivat or placebo and were followed for an average of 24 weeks. The primary endpoint was the number of patients who achieved a hemoglobin response (defined as a 1.5 g/dL or greater increase in hemoglobin concentration that was sustained at two or more scheduled assessments). At the end of the study, 40% of participants who received mitapivat had a hemoglobin response, compared with no participants who received placebo.

The single-arm study involved 27 adults with PK deficiency who were receiving regular blood transfusions. They took mitapivat for an average of 40 weeks. In this study, the primary endpoint was the reduction in transfusion burden, defined as at least a 33% reduction in the number of red blood cell units transfused during the last 24 weeks of treatment, compared with the historical transfusion burden on the individual participant (standardized to 24 weeks). The results show that 33% of participants who received mitapivat met this reduction in transfusion burden; 22% of participants did not require any transfusions over the last 24 weeks of treatment.

The most common side effects reported were decreases in estrone and estradiol in men, increased urate level, back pain, and joint stiffness. The effects of estrone and estradiol could not be reliably assessed in women because of normal changes in these hormone levels during the menstrual cycle and use of hormonal contraception.

The FDA warns of drug interactions that could necessitate dose adjustments, and also that abruptly stopping mitapivat could worsen premature red blood cell destruction.

The agency noted that this application received orphan drug designation, fast track designation, and priority review.

Agios is offering access programs aimed at reducing or eliminating patient out-of-pocket costs. Further details are on the myAgios patient support services program.

The company also noted that mitapivat is awaiting approval in the European Union. A decision is expected before the end of 2022.

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

A new drug that is both the first in its class and the first disease-modifying agent for hemolytic anemia in adults with pyruvate kinase (PK) deficiency has been approved by the Food and Drug Administration.

The new drug, mitapivat (Pyrukynd, Agios), was approved on the basis of clinical trials that showed that it significantly improved hemolysis and anemia in patients with PK deficiency.


PK deficiency is rare. In clinical practice, its frequency is approximately 3-9 cases per 1 million people, the FDA noted. However, PK deficiency likely is misdiagnosed or undiagnosed, making it difficult to determine its frequency in the general population

PK deficiency is an inherited disorder that causes premature red blood cell destruction, leading to anemia, the agency explained in its announcement. Symptoms of PK deficiency range in severity and include fatigue, unusually pale skin, jaundice, shortness of breath, and a fast heart rate. Patients can also develop an enlarged spleen, can have too much iron in their blood from repeated blood transfusions, and can develop gallstones.

“Pyrukynd is the first approved therapy for PK deficiency and marks an important milestone for these patients, who may face tremendous challenges and debilitating symptoms throughout the course of this lifelong disease,” said Rachael Grace, MD, pediatric hematologist and director of hematology clinical research at Boston Children’s Hospital.

She was an investigator in the clinical trials that led to the approval. In a statement from the manufacturer, she added that “partnering with Agios and the PK deficiency community to improve understanding of the natural history of this rare disease and bring a new medicine to patients has been an honor, and I look forward to additional collaboration in the future.”

Clinical data

Clinical data that formed the basis of the approval came from two trials, one of which was a randomized, placebo-controlled trial, and the other a single-arm study, the FDA noted. In these studies, patients received up to 50 mg of mitapivat orally twice daily after an initial dose titration period

The randomized trial involved 80 adults with PK deficiency who were not having regular blood transfusions. They were allocated to receive either mitapivat or placebo and were followed for an average of 24 weeks. The primary endpoint was the number of patients who achieved a hemoglobin response (defined as a 1.5 g/dL or greater increase in hemoglobin concentration that was sustained at two or more scheduled assessments). At the end of the study, 40% of participants who received mitapivat had a hemoglobin response, compared with no participants who received placebo.

The single-arm study involved 27 adults with PK deficiency who were receiving regular blood transfusions. They took mitapivat for an average of 40 weeks. In this study, the primary endpoint was the reduction in transfusion burden, defined as at least a 33% reduction in the number of red blood cell units transfused during the last 24 weeks of treatment, compared with the historical transfusion burden on the individual participant (standardized to 24 weeks). The results show that 33% of participants who received mitapivat met this reduction in transfusion burden; 22% of participants did not require any transfusions over the last 24 weeks of treatment.

The most common side effects reported were decreases in estrone and estradiol in men, increased urate level, back pain, and joint stiffness. The effects of estrone and estradiol could not be reliably assessed in women because of normal changes in these hormone levels during the menstrual cycle and use of hormonal contraception.

The FDA warns of drug interactions that could necessitate dose adjustments, and also that abruptly stopping mitapivat could worsen premature red blood cell destruction.

The agency noted that this application received orphan drug designation, fast track designation, and priority review.

Agios is offering access programs aimed at reducing or eliminating patient out-of-pocket costs. Further details are on the myAgios patient support services program.

The company also noted that mitapivat is awaiting approval in the European Union. A decision is expected before the end of 2022.

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

FDA approves first-ever drug for cold agglutinin disease

Article Type
Changed
Tue, 02/08/2022 - 09:18

A new drug has become the first and only treatment for people with cold agglutinin disease (CAD) that is approved by the U.S. Food and Drug Administration.

CAD is a rare autoimmune hemolytic anemia, affecting about 5,000 people in the United States. It is caused by antibodies binding to the surface of red blood cells, which starts a process that causes the body’s immune system to mistakenly attack healthy red blood cells and cause their rupture (hemolysis). This can lead to severe anemia, which is often treated by blood transfusions.

The new product is sutimlimab-jome (Enjaymo), and it works by inhibiting the destruction of red blood cells and so decreases the need for blood transfusions.

“For people living with cold agglutinin disease, it is as if their body’s immune system is waging a war on itself. The relentless destruction of healthy red blood cells is a daily, silent reality for people with CAD. For the first time, we have a treatment that targets complement-mediated hemolysis, which is the underlying cause of the red blood cell destruction in many CAD patients,” commented Catherine Broome, MD, associate professor of medicine at Georgetown University Lombardi Comprehensive Cancer Center, in a company press release.

Dr. Broome was the principal investigator in the CARDINAL study, which was the basis for the new approval. In this pivotal study, patients treated with sutimlimab had an improvement in anemia as measured by hemoglobin (Hgb) and bilirubin levels, she commented in the company statement.

The CARDINAL study was a 26-week open-label, single-arm phase 3 study conducted in 24 patients with CAD who had recent history of blood transfusion.

The primary efficacy endpoint was a composite defined as the proportion of patients who achieved normalization of Hgb level greater than or equal to 12 g/dL or demonstrated an increase from baseline in Hgb level greater than or equal to 2 g/dL at the treatment assessment time point (mean value from weeks 23, 25, and 26) and no blood transfusion from weeks 5 through 26.

More than half of the patients (13 of 24, 54%) met the composite primary endpoint criteria, with 17 of 24 (71%) patients remaining transfusion-free after week 5. Most patients (22 of 24, 92%) did not use other CAD-related treatments.

For the secondary measures on disease process, patients enrolled in the trial experienced a mean increase in Hgb level of 2.29 g/dL at week 3 and 3.18 g/dL at the 26-week treatment assessment time points (increasing from the mean baseline level of 8.6 g/dL). In addition, there was a mean reduction in bilirubin levels (n = 14) of -2.23 mg/dL from a mean baseline level of 3.23 mg/dL.

The most common adverse reactions occurring in 10% or more of patients were respiratory tract infection, viral infection, diarrhea, dyspepsia, cough, arthralgia, arthritis, and peripheral edema. Serious adverse reactions were reported in 3 of 24 (13%) patients, and these included streptococcal sepsis and staphylococcal wound infection (n = 1), arthralgia (n = 1), and respiratory tract infection (n = 1).

None of the adverse reactions led to discontinuation of the drug, the company noted. Dosage interruptions due to an adverse reaction occurred in 4 of 24 (17%) patients who received the drug.

The recommended dose is based on body weight (6,500 mg for people 39-75 kg and 7,500 mg for people greater than 75 kg). The drug is administered intravenously weekly for the first 2 weeks with administration every 2 weeks thereafter.

Full prescribing information is available here.

The product is a humanized monoclonal antibody that is designed to selectively target and inhibit C1s in the classical complement pathway, which is part of the innate immune system. By blocking C1s, Enjaymo inhibits the activation of the complement cascade in the immune system and inhibits C1-activated hemolysis in CAD to prevent the abnormal destruction of healthy red blood cells. Enjaymo does not inhibit the lectin and alternative pathways, the company noted.

In the U.S., Enjaymo received FDA breakthrough therapy and orphan drug designation, as well as priority review. The product is awaiting approval in Europe and Japan.

Sanofi says the product is expected to be available in the United States in the coming weeks, with a list price, or wholesale acquisition cost, of $1,800 per vial. Actual costs to patients are generally anticipated to be lower, as the list price does not reflect insurance coverage, copay support, or financial assistance from patient support programs. The company offers support for eligible patients on 1-833-223-2428.

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

Publications
Topics
Sections

A new drug has become the first and only treatment for people with cold agglutinin disease (CAD) that is approved by the U.S. Food and Drug Administration.

CAD is a rare autoimmune hemolytic anemia, affecting about 5,000 people in the United States. It is caused by antibodies binding to the surface of red blood cells, which starts a process that causes the body’s immune system to mistakenly attack healthy red blood cells and cause their rupture (hemolysis). This can lead to severe anemia, which is often treated by blood transfusions.

The new product is sutimlimab-jome (Enjaymo), and it works by inhibiting the destruction of red blood cells and so decreases the need for blood transfusions.

“For people living with cold agglutinin disease, it is as if their body’s immune system is waging a war on itself. The relentless destruction of healthy red blood cells is a daily, silent reality for people with CAD. For the first time, we have a treatment that targets complement-mediated hemolysis, which is the underlying cause of the red blood cell destruction in many CAD patients,” commented Catherine Broome, MD, associate professor of medicine at Georgetown University Lombardi Comprehensive Cancer Center, in a company press release.

Dr. Broome was the principal investigator in the CARDINAL study, which was the basis for the new approval. In this pivotal study, patients treated with sutimlimab had an improvement in anemia as measured by hemoglobin (Hgb) and bilirubin levels, she commented in the company statement.

The CARDINAL study was a 26-week open-label, single-arm phase 3 study conducted in 24 patients with CAD who had recent history of blood transfusion.

The primary efficacy endpoint was a composite defined as the proportion of patients who achieved normalization of Hgb level greater than or equal to 12 g/dL or demonstrated an increase from baseline in Hgb level greater than or equal to 2 g/dL at the treatment assessment time point (mean value from weeks 23, 25, and 26) and no blood transfusion from weeks 5 through 26.

More than half of the patients (13 of 24, 54%) met the composite primary endpoint criteria, with 17 of 24 (71%) patients remaining transfusion-free after week 5. Most patients (22 of 24, 92%) did not use other CAD-related treatments.

For the secondary measures on disease process, patients enrolled in the trial experienced a mean increase in Hgb level of 2.29 g/dL at week 3 and 3.18 g/dL at the 26-week treatment assessment time points (increasing from the mean baseline level of 8.6 g/dL). In addition, there was a mean reduction in bilirubin levels (n = 14) of -2.23 mg/dL from a mean baseline level of 3.23 mg/dL.

The most common adverse reactions occurring in 10% or more of patients were respiratory tract infection, viral infection, diarrhea, dyspepsia, cough, arthralgia, arthritis, and peripheral edema. Serious adverse reactions were reported in 3 of 24 (13%) patients, and these included streptococcal sepsis and staphylococcal wound infection (n = 1), arthralgia (n = 1), and respiratory tract infection (n = 1).

None of the adverse reactions led to discontinuation of the drug, the company noted. Dosage interruptions due to an adverse reaction occurred in 4 of 24 (17%) patients who received the drug.

The recommended dose is based on body weight (6,500 mg for people 39-75 kg and 7,500 mg for people greater than 75 kg). The drug is administered intravenously weekly for the first 2 weeks with administration every 2 weeks thereafter.

Full prescribing information is available here.

The product is a humanized monoclonal antibody that is designed to selectively target and inhibit C1s in the classical complement pathway, which is part of the innate immune system. By blocking C1s, Enjaymo inhibits the activation of the complement cascade in the immune system and inhibits C1-activated hemolysis in CAD to prevent the abnormal destruction of healthy red blood cells. Enjaymo does not inhibit the lectin and alternative pathways, the company noted.

In the U.S., Enjaymo received FDA breakthrough therapy and orphan drug designation, as well as priority review. The product is awaiting approval in Europe and Japan.

Sanofi says the product is expected to be available in the United States in the coming weeks, with a list price, or wholesale acquisition cost, of $1,800 per vial. Actual costs to patients are generally anticipated to be lower, as the list price does not reflect insurance coverage, copay support, or financial assistance from patient support programs. The company offers support for eligible patients on 1-833-223-2428.

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

A new drug has become the first and only treatment for people with cold agglutinin disease (CAD) that is approved by the U.S. Food and Drug Administration.

CAD is a rare autoimmune hemolytic anemia, affecting about 5,000 people in the United States. It is caused by antibodies binding to the surface of red blood cells, which starts a process that causes the body’s immune system to mistakenly attack healthy red blood cells and cause their rupture (hemolysis). This can lead to severe anemia, which is often treated by blood transfusions.

The new product is sutimlimab-jome (Enjaymo), and it works by inhibiting the destruction of red blood cells and so decreases the need for blood transfusions.

“For people living with cold agglutinin disease, it is as if their body’s immune system is waging a war on itself. The relentless destruction of healthy red blood cells is a daily, silent reality for people with CAD. For the first time, we have a treatment that targets complement-mediated hemolysis, which is the underlying cause of the red blood cell destruction in many CAD patients,” commented Catherine Broome, MD, associate professor of medicine at Georgetown University Lombardi Comprehensive Cancer Center, in a company press release.

Dr. Broome was the principal investigator in the CARDINAL study, which was the basis for the new approval. In this pivotal study, patients treated with sutimlimab had an improvement in anemia as measured by hemoglobin (Hgb) and bilirubin levels, she commented in the company statement.

The CARDINAL study was a 26-week open-label, single-arm phase 3 study conducted in 24 patients with CAD who had recent history of blood transfusion.

The primary efficacy endpoint was a composite defined as the proportion of patients who achieved normalization of Hgb level greater than or equal to 12 g/dL or demonstrated an increase from baseline in Hgb level greater than or equal to 2 g/dL at the treatment assessment time point (mean value from weeks 23, 25, and 26) and no blood transfusion from weeks 5 through 26.

More than half of the patients (13 of 24, 54%) met the composite primary endpoint criteria, with 17 of 24 (71%) patients remaining transfusion-free after week 5. Most patients (22 of 24, 92%) did not use other CAD-related treatments.

For the secondary measures on disease process, patients enrolled in the trial experienced a mean increase in Hgb level of 2.29 g/dL at week 3 and 3.18 g/dL at the 26-week treatment assessment time points (increasing from the mean baseline level of 8.6 g/dL). In addition, there was a mean reduction in bilirubin levels (n = 14) of -2.23 mg/dL from a mean baseline level of 3.23 mg/dL.

The most common adverse reactions occurring in 10% or more of patients were respiratory tract infection, viral infection, diarrhea, dyspepsia, cough, arthralgia, arthritis, and peripheral edema. Serious adverse reactions were reported in 3 of 24 (13%) patients, and these included streptococcal sepsis and staphylococcal wound infection (n = 1), arthralgia (n = 1), and respiratory tract infection (n = 1).

None of the adverse reactions led to discontinuation of the drug, the company noted. Dosage interruptions due to an adverse reaction occurred in 4 of 24 (17%) patients who received the drug.

The recommended dose is based on body weight (6,500 mg for people 39-75 kg and 7,500 mg for people greater than 75 kg). The drug is administered intravenously weekly for the first 2 weeks with administration every 2 weeks thereafter.

Full prescribing information is available here.

The product is a humanized monoclonal antibody that is designed to selectively target and inhibit C1s in the classical complement pathway, which is part of the innate immune system. By blocking C1s, Enjaymo inhibits the activation of the complement cascade in the immune system and inhibits C1-activated hemolysis in CAD to prevent the abnormal destruction of healthy red blood cells. Enjaymo does not inhibit the lectin and alternative pathways, the company noted.

In the U.S., Enjaymo received FDA breakthrough therapy and orphan drug designation, as well as priority review. The product is awaiting approval in Europe and Japan.

Sanofi says the product is expected to be available in the United States in the coming weeks, with a list price, or wholesale acquisition cost, of $1,800 per vial. Actual costs to patients are generally anticipated to be lower, as the list price does not reflect insurance coverage, copay support, or financial assistance from patient support programs. The company offers support for eligible patients on 1-833-223-2428.

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