Abemaciclib becomes first CDK inhibitor to clinch single-agent approval for breast cancer

Article Type
Changed
Thu, 12/15/2022 - 17:48

The fall 2017 approval by the US Food and Drug Administration (FDA) of abemaciclib made it the third cyclin-dependent kinase (CDK) inhibitor approved for the treatment of hormone receptor (HR)-positive breast cancer, and the first to receive an approved indication as monotherapy in that setting. Abemaciclib is a small-molecule inhibitor of the CDK4 and CDK6 proteins, which are key gatekeepers of the cell cycle and frequently dysregulated in HR-positive breast cancer. On the basis of the randomized, placebo-controlled, multicenter phase 3 MONARCH-2 trial, it was approved in combination with fulvestrant for the treatment of women with HR-positive, HER2-negative advanced or metastatic breast cancer who had progressed during endocrine therapy.1

A total of 669 women aged 18 years and older, with any menopausal status, an Eastern Cooperative Oncology Group (ECOG) Performance Status of 0 or 1, measurable disease per Response Evaluation Criteria in Solid Tumors (RECIST, version 1.1) or nonmeasurable bone-only disease, were enrolled. Patients had progressed during neoadjuvant or adjuvant endocrine therapy, within 12 months of adjuvant endocrine therapy, or during frontline endocrine treatment for metastatic disease.

Those who had received more than 1 endocrine therapy or any prior chemotherapy for metastatic breast cancer or prior treatment with everolimus or CDK4/6 inhibitors, as well as those with the presence of visceral crisis or evidence or history of central nervous system (CNS) metastases, were excluded from the study.

Patients were randomized 2:1 to receive 150 mg abemaciclib or placebo, both in combination with 500 mg fulvestrant. The initial dose of abemaciclib was 200 mg, but this was amended to 150mg after enrollment of the first 178 patients to alleviate diarrhea-related toxicity concerns. Randomization was stratified according to metastatic site (visceral, bone only, or other) and endocrine therapy resistance (primary or secondary).

Tumors were measured by computed tomography (CT) and magnetic-resonance imaging (MRI) according to RECIST-1.1 within 28 days before random assignment, every 8 weeks for the first year, every 12 weeks thereafter, and then within 2 weeks of clinical progression. Bone scintigraphy was also performed at baseline and then every 6th cycle starting with cycle 7. Hematologic and blood chemistry laboratory tests were performed centrally on days 1 and 15 of the first cycle and day 1 of all remaining cycles.

The primary endpoint was progression-free survival (PFS); median PFS was 16.4 months in the abemaciclib arm, compared with 9.3 months in the placebo arm in the intent-to-treat population (hazard ratio [HR], 0.553;P < .0000001), translating to a 45% reduction in the risk of disease progression or death with the combination. Objective response rate in the 2 groups among patients with measurable disease was 48.1% and 21.3%, respectively, which included a complete response rate of 3.5% in the abemaciclib arm. The median duration of response was not yet reached in the study group, compared with 25.6 months for placebo. Overall survival data were not yet mature.

The agency also approved abemaciclib as monotherapy for women and men with HR-positive, HER2-negative advanced or metastatic breast cancer with disease progression following endocrine therapy and prior chemotherapy in the metastatic setting. That approval was based on data from the single-arm MONARCH-1 trial of 132 patients who received 200 mg abemaciclib twice daily on a continuous schedule.2

Patients had adequate organ function, measurable disease per RECIST-1.1, and an ECOG performance status of 0 or 1. Patients must have progressed on or after previous endocrine therapy and have received prior treatment with at least 2 chemotherapy regimens, at least 1 of them, but no more than 2, having been administered in the metastatic setting. Exclusion criteria included prior receipt of a CDK inhibitor, major surgery within 14 days of the start of the study, and CNS metastases.

Tumor assessments were performed by CT or MRI according to RECIST-1.1 within the 4 weeks prior to the first dose of study drug and then subsequently at every other cycle. Responses were confirmed at least 4 weeks after the initial observation. The overall response rate was 19.7%, made up completely of partial responses. Median duration of response was 8.6 months, median PFS was 6 months and median OS was 17.7 months.
 

Adverse events

The most common adverse events experienced with the combination of abemaciclib and fulvestrant were neutropenia (23.6%) and diarrhea (13.4%). The rate of grade 4 neutropenia was higher in the combination arm (2.9% vs 0.4%) and there were 3 deaths with the combination that were linked to treatment-related AEs. In the monotherapy trial, abemaciclib treatment most commonly caused diarrhea (90.2%), fatigue (65.2%), nausea (64.4%), decreased appetite (45.5%), and abdominal pain (38.6%). Grade 3 diarrhea and fatigue occurred in 19.7% and 12.9% of patients, respectively. Serious AEs occurred in 24.2% of patients and AEs led to treatment discontinuation in 7.6% of patients.

 

 

Warnings and precautions

Abemaciclib is marketed as Verzenio by Eli Lilly and Company. Warnings and precautions relating to diarrhea, neutropenia, hepatotoxicity, venous thromboembolism (VTE), and embryofetal toxicity are detailed in the prescribing information. In the event of diarrhea, patients should be treated with antidiarrheal therapy and should increase oral fluids and notify their health care provider. Treatment should be interrupted for grade 3 or 4 diarrhea and then resumed at a lower dose upon return to grade 1.

To guard against neutropenia, complete blood counts should be performed prior to starting therapy, every 2 weeks for the first 2 months, monthly for the subsequent 2 months, and then as clinically indicated. Treatment should be interrupted or delayed or the dose reduced for grade 3 or 4 neutropenia and patients should report episodes of fever.

Liver function tests should be performed before starting abemaciclib, every 2 weeks for the first 2 months, monthly for the next 2 months, and then as clinically indicated. For patients who develop persistent or recurrent grade 2, 3 or 4 hepatic transaminase elevation, dose interruption, reduction, discontinuation, or delay should be considered.

Patients should be monitored for signs and symptoms of VTE and pulmonary embolism, and treated appropriately. Pregnant women should be advised of the potential risk to a fetus, and those of reproductive potential should be counselled on the importance of using effective contraception during treatment and for at least 3 weeks after the last dose.3

References

1. Sledge Jr GW, Toi M, Neven P, et al. MONARCH 2: Abemaciclib in combination with fulvestrant in women with HR+/HER2- advanced breast cancer who had progressed while receiving endocrine therapy. J Clin Oncol. 2017;35(25):2875-2884.
2. Dickler MN, Tolaney SM, Rugo HS, et al. MONARCH 1, a phase 2 study of abemaciclib, a CDK4 and CDK6 inhibitor, as a single agent, in patients with refractory HR+/HER2- metastatic breast cancer. Clin Cancer Res. http://clincancerres.aacrjournals.org/content/early/2017/05/20/1078-0432.CCR-17-0754. Published online first on May 22, 2017. Accessed January 19, 2018.
3. Verzenio (abemaciclib) tablets, for oral use. Prescribing information. Eli Lilly and Co. http://uspl.lilly.com/verzenio/verzenio.html#pi. September 2017. Accessed November 20, 2017.

Article PDF
Author and Disclosure Information

Edited by Jame Abraham, MD, FACP; report prepared by Jane de Lartigue, PhD

Issue
The Journal of Community and Supportive Oncology - 16(1)
Publications
Topics
Page Number
e2-e4
Sections
Author and Disclosure Information

Edited by Jame Abraham, MD, FACP; report prepared by Jane de Lartigue, PhD

Author and Disclosure Information

Edited by Jame Abraham, MD, FACP; report prepared by Jane de Lartigue, PhD

Article PDF
Article PDF

The fall 2017 approval by the US Food and Drug Administration (FDA) of abemaciclib made it the third cyclin-dependent kinase (CDK) inhibitor approved for the treatment of hormone receptor (HR)-positive breast cancer, and the first to receive an approved indication as monotherapy in that setting. Abemaciclib is a small-molecule inhibitor of the CDK4 and CDK6 proteins, which are key gatekeepers of the cell cycle and frequently dysregulated in HR-positive breast cancer. On the basis of the randomized, placebo-controlled, multicenter phase 3 MONARCH-2 trial, it was approved in combination with fulvestrant for the treatment of women with HR-positive, HER2-negative advanced or metastatic breast cancer who had progressed during endocrine therapy.1

A total of 669 women aged 18 years and older, with any menopausal status, an Eastern Cooperative Oncology Group (ECOG) Performance Status of 0 or 1, measurable disease per Response Evaluation Criteria in Solid Tumors (RECIST, version 1.1) or nonmeasurable bone-only disease, were enrolled. Patients had progressed during neoadjuvant or adjuvant endocrine therapy, within 12 months of adjuvant endocrine therapy, or during frontline endocrine treatment for metastatic disease.

Those who had received more than 1 endocrine therapy or any prior chemotherapy for metastatic breast cancer or prior treatment with everolimus or CDK4/6 inhibitors, as well as those with the presence of visceral crisis or evidence or history of central nervous system (CNS) metastases, were excluded from the study.

Patients were randomized 2:1 to receive 150 mg abemaciclib or placebo, both in combination with 500 mg fulvestrant. The initial dose of abemaciclib was 200 mg, but this was amended to 150mg after enrollment of the first 178 patients to alleviate diarrhea-related toxicity concerns. Randomization was stratified according to metastatic site (visceral, bone only, or other) and endocrine therapy resistance (primary or secondary).

Tumors were measured by computed tomography (CT) and magnetic-resonance imaging (MRI) according to RECIST-1.1 within 28 days before random assignment, every 8 weeks for the first year, every 12 weeks thereafter, and then within 2 weeks of clinical progression. Bone scintigraphy was also performed at baseline and then every 6th cycle starting with cycle 7. Hematologic and blood chemistry laboratory tests were performed centrally on days 1 and 15 of the first cycle and day 1 of all remaining cycles.

The primary endpoint was progression-free survival (PFS); median PFS was 16.4 months in the abemaciclib arm, compared with 9.3 months in the placebo arm in the intent-to-treat population (hazard ratio [HR], 0.553;P < .0000001), translating to a 45% reduction in the risk of disease progression or death with the combination. Objective response rate in the 2 groups among patients with measurable disease was 48.1% and 21.3%, respectively, which included a complete response rate of 3.5% in the abemaciclib arm. The median duration of response was not yet reached in the study group, compared with 25.6 months for placebo. Overall survival data were not yet mature.

The agency also approved abemaciclib as monotherapy for women and men with HR-positive, HER2-negative advanced or metastatic breast cancer with disease progression following endocrine therapy and prior chemotherapy in the metastatic setting. That approval was based on data from the single-arm MONARCH-1 trial of 132 patients who received 200 mg abemaciclib twice daily on a continuous schedule.2

Patients had adequate organ function, measurable disease per RECIST-1.1, and an ECOG performance status of 0 or 1. Patients must have progressed on or after previous endocrine therapy and have received prior treatment with at least 2 chemotherapy regimens, at least 1 of them, but no more than 2, having been administered in the metastatic setting. Exclusion criteria included prior receipt of a CDK inhibitor, major surgery within 14 days of the start of the study, and CNS metastases.

Tumor assessments were performed by CT or MRI according to RECIST-1.1 within the 4 weeks prior to the first dose of study drug and then subsequently at every other cycle. Responses were confirmed at least 4 weeks after the initial observation. The overall response rate was 19.7%, made up completely of partial responses. Median duration of response was 8.6 months, median PFS was 6 months and median OS was 17.7 months.
 

Adverse events

The most common adverse events experienced with the combination of abemaciclib and fulvestrant were neutropenia (23.6%) and diarrhea (13.4%). The rate of grade 4 neutropenia was higher in the combination arm (2.9% vs 0.4%) and there were 3 deaths with the combination that were linked to treatment-related AEs. In the monotherapy trial, abemaciclib treatment most commonly caused diarrhea (90.2%), fatigue (65.2%), nausea (64.4%), decreased appetite (45.5%), and abdominal pain (38.6%). Grade 3 diarrhea and fatigue occurred in 19.7% and 12.9% of patients, respectively. Serious AEs occurred in 24.2% of patients and AEs led to treatment discontinuation in 7.6% of patients.

 

 

Warnings and precautions

Abemaciclib is marketed as Verzenio by Eli Lilly and Company. Warnings and precautions relating to diarrhea, neutropenia, hepatotoxicity, venous thromboembolism (VTE), and embryofetal toxicity are detailed in the prescribing information. In the event of diarrhea, patients should be treated with antidiarrheal therapy and should increase oral fluids and notify their health care provider. Treatment should be interrupted for grade 3 or 4 diarrhea and then resumed at a lower dose upon return to grade 1.

To guard against neutropenia, complete blood counts should be performed prior to starting therapy, every 2 weeks for the first 2 months, monthly for the subsequent 2 months, and then as clinically indicated. Treatment should be interrupted or delayed or the dose reduced for grade 3 or 4 neutropenia and patients should report episodes of fever.

Liver function tests should be performed before starting abemaciclib, every 2 weeks for the first 2 months, monthly for the next 2 months, and then as clinically indicated. For patients who develop persistent or recurrent grade 2, 3 or 4 hepatic transaminase elevation, dose interruption, reduction, discontinuation, or delay should be considered.

Patients should be monitored for signs and symptoms of VTE and pulmonary embolism, and treated appropriately. Pregnant women should be advised of the potential risk to a fetus, and those of reproductive potential should be counselled on the importance of using effective contraception during treatment and for at least 3 weeks after the last dose.3

The fall 2017 approval by the US Food and Drug Administration (FDA) of abemaciclib made it the third cyclin-dependent kinase (CDK) inhibitor approved for the treatment of hormone receptor (HR)-positive breast cancer, and the first to receive an approved indication as monotherapy in that setting. Abemaciclib is a small-molecule inhibitor of the CDK4 and CDK6 proteins, which are key gatekeepers of the cell cycle and frequently dysregulated in HR-positive breast cancer. On the basis of the randomized, placebo-controlled, multicenter phase 3 MONARCH-2 trial, it was approved in combination with fulvestrant for the treatment of women with HR-positive, HER2-negative advanced or metastatic breast cancer who had progressed during endocrine therapy.1

A total of 669 women aged 18 years and older, with any menopausal status, an Eastern Cooperative Oncology Group (ECOG) Performance Status of 0 or 1, measurable disease per Response Evaluation Criteria in Solid Tumors (RECIST, version 1.1) or nonmeasurable bone-only disease, were enrolled. Patients had progressed during neoadjuvant or adjuvant endocrine therapy, within 12 months of adjuvant endocrine therapy, or during frontline endocrine treatment for metastatic disease.

Those who had received more than 1 endocrine therapy or any prior chemotherapy for metastatic breast cancer or prior treatment with everolimus or CDK4/6 inhibitors, as well as those with the presence of visceral crisis or evidence or history of central nervous system (CNS) metastases, were excluded from the study.

Patients were randomized 2:1 to receive 150 mg abemaciclib or placebo, both in combination with 500 mg fulvestrant. The initial dose of abemaciclib was 200 mg, but this was amended to 150mg after enrollment of the first 178 patients to alleviate diarrhea-related toxicity concerns. Randomization was stratified according to metastatic site (visceral, bone only, or other) and endocrine therapy resistance (primary or secondary).

Tumors were measured by computed tomography (CT) and magnetic-resonance imaging (MRI) according to RECIST-1.1 within 28 days before random assignment, every 8 weeks for the first year, every 12 weeks thereafter, and then within 2 weeks of clinical progression. Bone scintigraphy was also performed at baseline and then every 6th cycle starting with cycle 7. Hematologic and blood chemistry laboratory tests were performed centrally on days 1 and 15 of the first cycle and day 1 of all remaining cycles.

The primary endpoint was progression-free survival (PFS); median PFS was 16.4 months in the abemaciclib arm, compared with 9.3 months in the placebo arm in the intent-to-treat population (hazard ratio [HR], 0.553;P < .0000001), translating to a 45% reduction in the risk of disease progression or death with the combination. Objective response rate in the 2 groups among patients with measurable disease was 48.1% and 21.3%, respectively, which included a complete response rate of 3.5% in the abemaciclib arm. The median duration of response was not yet reached in the study group, compared with 25.6 months for placebo. Overall survival data were not yet mature.

The agency also approved abemaciclib as monotherapy for women and men with HR-positive, HER2-negative advanced or metastatic breast cancer with disease progression following endocrine therapy and prior chemotherapy in the metastatic setting. That approval was based on data from the single-arm MONARCH-1 trial of 132 patients who received 200 mg abemaciclib twice daily on a continuous schedule.2

Patients had adequate organ function, measurable disease per RECIST-1.1, and an ECOG performance status of 0 or 1. Patients must have progressed on or after previous endocrine therapy and have received prior treatment with at least 2 chemotherapy regimens, at least 1 of them, but no more than 2, having been administered in the metastatic setting. Exclusion criteria included prior receipt of a CDK inhibitor, major surgery within 14 days of the start of the study, and CNS metastases.

Tumor assessments were performed by CT or MRI according to RECIST-1.1 within the 4 weeks prior to the first dose of study drug and then subsequently at every other cycle. Responses were confirmed at least 4 weeks after the initial observation. The overall response rate was 19.7%, made up completely of partial responses. Median duration of response was 8.6 months, median PFS was 6 months and median OS was 17.7 months.
 

Adverse events

The most common adverse events experienced with the combination of abemaciclib and fulvestrant were neutropenia (23.6%) and diarrhea (13.4%). The rate of grade 4 neutropenia was higher in the combination arm (2.9% vs 0.4%) and there were 3 deaths with the combination that were linked to treatment-related AEs. In the monotherapy trial, abemaciclib treatment most commonly caused diarrhea (90.2%), fatigue (65.2%), nausea (64.4%), decreased appetite (45.5%), and abdominal pain (38.6%). Grade 3 diarrhea and fatigue occurred in 19.7% and 12.9% of patients, respectively. Serious AEs occurred in 24.2% of patients and AEs led to treatment discontinuation in 7.6% of patients.

 

 

Warnings and precautions

Abemaciclib is marketed as Verzenio by Eli Lilly and Company. Warnings and precautions relating to diarrhea, neutropenia, hepatotoxicity, venous thromboembolism (VTE), and embryofetal toxicity are detailed in the prescribing information. In the event of diarrhea, patients should be treated with antidiarrheal therapy and should increase oral fluids and notify their health care provider. Treatment should be interrupted for grade 3 or 4 diarrhea and then resumed at a lower dose upon return to grade 1.

To guard against neutropenia, complete blood counts should be performed prior to starting therapy, every 2 weeks for the first 2 months, monthly for the subsequent 2 months, and then as clinically indicated. Treatment should be interrupted or delayed or the dose reduced for grade 3 or 4 neutropenia and patients should report episodes of fever.

Liver function tests should be performed before starting abemaciclib, every 2 weeks for the first 2 months, monthly for the next 2 months, and then as clinically indicated. For patients who develop persistent or recurrent grade 2, 3 or 4 hepatic transaminase elevation, dose interruption, reduction, discontinuation, or delay should be considered.

Patients should be monitored for signs and symptoms of VTE and pulmonary embolism, and treated appropriately. Pregnant women should be advised of the potential risk to a fetus, and those of reproductive potential should be counselled on the importance of using effective contraception during treatment and for at least 3 weeks after the last dose.3

References

1. Sledge Jr GW, Toi M, Neven P, et al. MONARCH 2: Abemaciclib in combination with fulvestrant in women with HR+/HER2- advanced breast cancer who had progressed while receiving endocrine therapy. J Clin Oncol. 2017;35(25):2875-2884.
2. Dickler MN, Tolaney SM, Rugo HS, et al. MONARCH 1, a phase 2 study of abemaciclib, a CDK4 and CDK6 inhibitor, as a single agent, in patients with refractory HR+/HER2- metastatic breast cancer. Clin Cancer Res. http://clincancerres.aacrjournals.org/content/early/2017/05/20/1078-0432.CCR-17-0754. Published online first on May 22, 2017. Accessed January 19, 2018.
3. Verzenio (abemaciclib) tablets, for oral use. Prescribing information. Eli Lilly and Co. http://uspl.lilly.com/verzenio/verzenio.html#pi. September 2017. Accessed November 20, 2017.

References

1. Sledge Jr GW, Toi M, Neven P, et al. MONARCH 2: Abemaciclib in combination with fulvestrant in women with HR+/HER2- advanced breast cancer who had progressed while receiving endocrine therapy. J Clin Oncol. 2017;35(25):2875-2884.
2. Dickler MN, Tolaney SM, Rugo HS, et al. MONARCH 1, a phase 2 study of abemaciclib, a CDK4 and CDK6 inhibitor, as a single agent, in patients with refractory HR+/HER2- metastatic breast cancer. Clin Cancer Res. http://clincancerres.aacrjournals.org/content/early/2017/05/20/1078-0432.CCR-17-0754. Published online first on May 22, 2017. Accessed January 19, 2018.
3. Verzenio (abemaciclib) tablets, for oral use. Prescribing information. Eli Lilly and Co. http://uspl.lilly.com/verzenio/verzenio.html#pi. September 2017. Accessed November 20, 2017.

Issue
The Journal of Community and Supportive Oncology - 16(1)
Issue
The Journal of Community and Supportive Oncology - 16(1)
Page Number
e2-e4
Page Number
e2-e4
Publications
Publications
Topics
Article Type
Sections
Citation Override
JCSO February 2018;16(1):e2-e4
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media

Uridine triacetate provides antidote for 5-fluorouracil overdose and toxicity

Article Type
Changed
Fri, 01/04/2019 - 11:13
Display Headline
Uridine triacetate provides antidote for 5-fluorouracil overdose and toxicity

Fluorouracil or 5-fluorouracil (5-FU) is an effective cytotoxic drug that is incorporated into various chemotherapeutic regimens for the treatment of numerous tumor types, but its clinical utility is limited by its narrow therapeutic index and the risk of overdose and serious toxic effects. Until recently, these outcomes were managed with supportive care, but the approval of uridine triacetate provides an antidote to reverse 5-FU-associated toxicity, to prevent death and potentially allow some patients to resume chemotherapy.

 

Click on the PDF icon at the top of this introduction to read the full article. 

 

Article PDF
Issue
The Journal of Community and Supportive Oncology - 14(8)
Publications
Topics
Page Number
332-333
Legacy Keywords
fluorouracil, 5-FU, cytotoxic, chemotherapy, overdose, toxic effects, uridine triacetate
Article PDF
Article PDF

Fluorouracil or 5-fluorouracil (5-FU) is an effective cytotoxic drug that is incorporated into various chemotherapeutic regimens for the treatment of numerous tumor types, but its clinical utility is limited by its narrow therapeutic index and the risk of overdose and serious toxic effects. Until recently, these outcomes were managed with supportive care, but the approval of uridine triacetate provides an antidote to reverse 5-FU-associated toxicity, to prevent death and potentially allow some patients to resume chemotherapy.

 

Click on the PDF icon at the top of this introduction to read the full article. 

 

Fluorouracil or 5-fluorouracil (5-FU) is an effective cytotoxic drug that is incorporated into various chemotherapeutic regimens for the treatment of numerous tumor types, but its clinical utility is limited by its narrow therapeutic index and the risk of overdose and serious toxic effects. Until recently, these outcomes were managed with supportive care, but the approval of uridine triacetate provides an antidote to reverse 5-FU-associated toxicity, to prevent death and potentially allow some patients to resume chemotherapy.

 

Click on the PDF icon at the top of this introduction to read the full article. 

 

Issue
The Journal of Community and Supportive Oncology - 14(8)
Issue
The Journal of Community and Supportive Oncology - 14(8)
Page Number
332-333
Page Number
332-333
Publications
Publications
Topics
Article Type
Display Headline
Uridine triacetate provides antidote for 5-fluorouracil overdose and toxicity
Display Headline
Uridine triacetate provides antidote for 5-fluorouracil overdose and toxicity
Legacy Keywords
fluorouracil, 5-FU, cytotoxic, chemotherapy, overdose, toxic effects, uridine triacetate
Legacy Keywords
fluorouracil, 5-FU, cytotoxic, chemotherapy, overdose, toxic effects, uridine triacetate
Citation Override
JCSO 2016;14(8):332-33
Disallow All Ads
Alternative CME
Article PDF Media

Trabectedin expands treatment options for some forms of advanced soft tissue sarcoma

Article Type
Changed
Fri, 01/04/2019 - 11:12
Display Headline
Trabectedin expands treatment options for some forms of advanced soft tissue sarcoma
In the fall of 2015, the United States joined the growing list of countries in which trabectedin, a novel form of chemotherapy, is approved for the treatment of certain types of advanced soft tissue sarcoma. The drug, a synthetic derivative of a compound originally isolated from a sea squirt, has a complex mechanism of action that distinguishes it from other cytotoxic drugs, allowing it to target both the tumor and its microenvironment. Based on the results of a randomized, open-label, multicenter phase 3 clinical trial, trabectedin received regulatory approval for the treatment of unresectable or metastatic liposarcoma or leiomyosarcoma.

 

Click on the PDF icon at the top of this introduction to read the full article. 

 

Article PDF
Issue
The Journal of Community and Supportive Oncology - 14(5)
Publications
Topics
Page Number
189-191
Legacy Keywords
trabectedin, soft tissue sarcoma, ET743-SAR-3007, liposarcoma, leiomyosarcoma, dacarbazine
Sections
Article PDF
Article PDF
In the fall of 2015, the United States joined the growing list of countries in which trabectedin, a novel form of chemotherapy, is approved for the treatment of certain types of advanced soft tissue sarcoma. The drug, a synthetic derivative of a compound originally isolated from a sea squirt, has a complex mechanism of action that distinguishes it from other cytotoxic drugs, allowing it to target both the tumor and its microenvironment. Based on the results of a randomized, open-label, multicenter phase 3 clinical trial, trabectedin received regulatory approval for the treatment of unresectable or metastatic liposarcoma or leiomyosarcoma.

 

Click on the PDF icon at the top of this introduction to read the full article. 

 

In the fall of 2015, the United States joined the growing list of countries in which trabectedin, a novel form of chemotherapy, is approved for the treatment of certain types of advanced soft tissue sarcoma. The drug, a synthetic derivative of a compound originally isolated from a sea squirt, has a complex mechanism of action that distinguishes it from other cytotoxic drugs, allowing it to target both the tumor and its microenvironment. Based on the results of a randomized, open-label, multicenter phase 3 clinical trial, trabectedin received regulatory approval for the treatment of unresectable or metastatic liposarcoma or leiomyosarcoma.

 

Click on the PDF icon at the top of this introduction to read the full article. 

 

Issue
The Journal of Community and Supportive Oncology - 14(5)
Issue
The Journal of Community and Supportive Oncology - 14(5)
Page Number
189-191
Page Number
189-191
Publications
Publications
Topics
Article Type
Display Headline
Trabectedin expands treatment options for some forms of advanced soft tissue sarcoma
Display Headline
Trabectedin expands treatment options for some forms of advanced soft tissue sarcoma
Legacy Keywords
trabectedin, soft tissue sarcoma, ET743-SAR-3007, liposarcoma, leiomyosarcoma, dacarbazine
Legacy Keywords
trabectedin, soft tissue sarcoma, ET743-SAR-3007, liposarcoma, leiomyosarcoma, dacarbazine
Sections
Citation Override
JCSO 2016;14:189-19
Disallow All Ads
Alternative CME
Article PDF Media

More success for immunotherapy with nivolumab approval for metastatic RCC

Article Type
Changed
Fri, 01/04/2019 - 11:12
Display Headline
More success for immunotherapy with nivolumab approval for metastatic RCC
Following its recent success in non–small-cell lung cancer (NSCLC), the immunotherapeutic agent nivolumab received approval from the US Food and Drug Administration last fall for the treatment of advanced clear-cell renal-cell carcinoma (RCC) after prior antiangiogenic therapy. Nivolumab, an immune checkpoint inhibitor, is a fully human immunoglobulin G4 monoclonal antibody that binds to the programmed cell death 1 (PD-1) receptor on the surface of T cells, prevents their inactivation by tumor cells overexpressing PD-1 ligands, and helps to reinstate the anti-tumor immune response.
 
Click on the PDF icon at the top of this introduction to read the full article.  
 
Article PDF
Issue
The Journal of Community and Supportive Oncology - 14(4)
Publications
Topics
Page Number
138-140
Legacy Keywords
renal cell carcinoma, RCC, immunotherapy, nivolumab, CheckMate-025, programmed death cell

Sections
Article PDF
Article PDF
Following its recent success in non–small-cell lung cancer (NSCLC), the immunotherapeutic agent nivolumab received approval from the US Food and Drug Administration last fall for the treatment of advanced clear-cell renal-cell carcinoma (RCC) after prior antiangiogenic therapy. Nivolumab, an immune checkpoint inhibitor, is a fully human immunoglobulin G4 monoclonal antibody that binds to the programmed cell death 1 (PD-1) receptor on the surface of T cells, prevents their inactivation by tumor cells overexpressing PD-1 ligands, and helps to reinstate the anti-tumor immune response.
 
Click on the PDF icon at the top of this introduction to read the full article.  
 
Following its recent success in non–small-cell lung cancer (NSCLC), the immunotherapeutic agent nivolumab received approval from the US Food and Drug Administration last fall for the treatment of advanced clear-cell renal-cell carcinoma (RCC) after prior antiangiogenic therapy. Nivolumab, an immune checkpoint inhibitor, is a fully human immunoglobulin G4 monoclonal antibody that binds to the programmed cell death 1 (PD-1) receptor on the surface of T cells, prevents their inactivation by tumor cells overexpressing PD-1 ligands, and helps to reinstate the anti-tumor immune response.
 
Click on the PDF icon at the top of this introduction to read the full article.  
 
Issue
The Journal of Community and Supportive Oncology - 14(4)
Issue
The Journal of Community and Supportive Oncology - 14(4)
Page Number
138-140
Page Number
138-140
Publications
Publications
Topics
Article Type
Display Headline
More success for immunotherapy with nivolumab approval for metastatic RCC
Display Headline
More success for immunotherapy with nivolumab approval for metastatic RCC
Legacy Keywords
renal cell carcinoma, RCC, immunotherapy, nivolumab, CheckMate-025, programmed death cell

Legacy Keywords
renal cell carcinoma, RCC, immunotherapy, nivolumab, CheckMate-025, programmed death cell

Sections
Citation Override
JCSO 2016;14:138-14
Disallow All Ads
Alternative CME
Article PDF Media

Encapsulated irinotecan provides novel option for hard-to-treat pancreatic cancer

Article Type
Changed
Wed, 05/26/2021 - 13:55

In the fall of 2015, the US Food and Drug Administration approved the addition of a novel, much-needed treatment option for patients with metastatic pancreatic cancer, a particularly hard-to-treat form of the disease, in the second-line setting following progression on gemcitabine- based chemotherapy.1 MM-398 is a modified version of the chemotherapeutic agent irinotecan, in which the drug is encapsulated in a nanoliposomal construct that is designed to improve delivery to the tumor and enhance anti-tumor efficacy while minimizing side effects in the rest of the body.

 

Click on the PDF icon at the top of this introduction to read the full article.

 

Article PDF
Author and Disclosure Information

Edited by Jame Abraham, MD; report prepared by Jane de Lartigue, PhD

Issue
The Journal of Community and Supportive Oncology - 14(3)
Publications
Topics
Page Number
91-93
Legacy Keywords
irinotecan, pancreatic cancer, NAPOLI-1 trial, fluorouracil, leucovorin
Sections
Author and Disclosure Information

Edited by Jame Abraham, MD; report prepared by Jane de Lartigue, PhD

Author and Disclosure Information

Edited by Jame Abraham, MD; report prepared by Jane de Lartigue, PhD

Article PDF
Article PDF

In the fall of 2015, the US Food and Drug Administration approved the addition of a novel, much-needed treatment option for patients with metastatic pancreatic cancer, a particularly hard-to-treat form of the disease, in the second-line setting following progression on gemcitabine- based chemotherapy.1 MM-398 is a modified version of the chemotherapeutic agent irinotecan, in which the drug is encapsulated in a nanoliposomal construct that is designed to improve delivery to the tumor and enhance anti-tumor efficacy while minimizing side effects in the rest of the body.

 

Click on the PDF icon at the top of this introduction to read the full article.

 

In the fall of 2015, the US Food and Drug Administration approved the addition of a novel, much-needed treatment option for patients with metastatic pancreatic cancer, a particularly hard-to-treat form of the disease, in the second-line setting following progression on gemcitabine- based chemotherapy.1 MM-398 is a modified version of the chemotherapeutic agent irinotecan, in which the drug is encapsulated in a nanoliposomal construct that is designed to improve delivery to the tumor and enhance anti-tumor efficacy while minimizing side effects in the rest of the body.

 

Click on the PDF icon at the top of this introduction to read the full article.

 

Issue
The Journal of Community and Supportive Oncology - 14(3)
Issue
The Journal of Community and Supportive Oncology - 14(3)
Page Number
91-93
Page Number
91-93
Publications
Publications
Topics
Article Type
Legacy Keywords
irinotecan, pancreatic cancer, NAPOLI-1 trial, fluorouracil, leucovorin
Legacy Keywords
irinotecan, pancreatic cancer, NAPOLI-1 trial, fluorouracil, leucovorin
Sections
Citation Override
JCSO 2017;14(3):91-93
Disallow All Ads
Alternative CME
Article PDF Media

Second targeted combination regime approved for metastatic melanoma

Article Type
Changed
Fri, 01/04/2019 - 11:12
Display Headline
Second targeted combination regime approved for metastatic melanoma
Researchers are increasingly looking to rational combinations of drugs to meet the challenge of resistance to molecularly targeted anticancer therapies. The treatment of metastatic melanoma was significantly advanced with the development of BRAF inhibitors, but is still limited by the short-lived nature of the responses to these drugs and the almost universal occurrence of tumor regrowth. More durable responses could be produced by targeting multiple points in the mitogen-activated protein kinase (MAPK) signaling pathway of which BRAF is a central component. In the fall of 2015, the MEK inhibitor cobimetinib and BRAF inhibitor vemurafenib became the second such combination to receive regulatory approval in the United States following positive results from the international, phase 3, randomized coBRIM trial, which demonstrated the superiority of the combination over single-agent BRAF inhibitor therapy. It also marked the first US approval for cobimetinib.1 

 

Click on the PDF icon at the top of this introduction to read the full article.

 

Article PDF
Issue
The Journal of Community and Supportive Oncology - 14(2)
Publications
Topics
Page Number
51-53
Legacy Keywords
melanoma, cobimetinib, vemurafenib, molecularly targeted anticancer therapies, metastatic melanoma, BRAF inhibitors, mitogen-activated protein kinase, MAPK, MEK inhibitor
Sections
Article PDF
Article PDF
Researchers are increasingly looking to rational combinations of drugs to meet the challenge of resistance to molecularly targeted anticancer therapies. The treatment of metastatic melanoma was significantly advanced with the development of BRAF inhibitors, but is still limited by the short-lived nature of the responses to these drugs and the almost universal occurrence of tumor regrowth. More durable responses could be produced by targeting multiple points in the mitogen-activated protein kinase (MAPK) signaling pathway of which BRAF is a central component. In the fall of 2015, the MEK inhibitor cobimetinib and BRAF inhibitor vemurafenib became the second such combination to receive regulatory approval in the United States following positive results from the international, phase 3, randomized coBRIM trial, which demonstrated the superiority of the combination over single-agent BRAF inhibitor therapy. It also marked the first US approval for cobimetinib.1 

 

Click on the PDF icon at the top of this introduction to read the full article.

 

Researchers are increasingly looking to rational combinations of drugs to meet the challenge of resistance to molecularly targeted anticancer therapies. The treatment of metastatic melanoma was significantly advanced with the development of BRAF inhibitors, but is still limited by the short-lived nature of the responses to these drugs and the almost universal occurrence of tumor regrowth. More durable responses could be produced by targeting multiple points in the mitogen-activated protein kinase (MAPK) signaling pathway of which BRAF is a central component. In the fall of 2015, the MEK inhibitor cobimetinib and BRAF inhibitor vemurafenib became the second such combination to receive regulatory approval in the United States following positive results from the international, phase 3, randomized coBRIM trial, which demonstrated the superiority of the combination over single-agent BRAF inhibitor therapy. It also marked the first US approval for cobimetinib.1 

 

Click on the PDF icon at the top of this introduction to read the full article.

 

Issue
The Journal of Community and Supportive Oncology - 14(2)
Issue
The Journal of Community and Supportive Oncology - 14(2)
Page Number
51-53
Page Number
51-53
Publications
Publications
Topics
Article Type
Display Headline
Second targeted combination regime approved for metastatic melanoma
Display Headline
Second targeted combination regime approved for metastatic melanoma
Legacy Keywords
melanoma, cobimetinib, vemurafenib, molecularly targeted anticancer therapies, metastatic melanoma, BRAF inhibitors, mitogen-activated protein kinase, MAPK, MEK inhibitor
Legacy Keywords
melanoma, cobimetinib, vemurafenib, molecularly targeted anticancer therapies, metastatic melanoma, BRAF inhibitors, mitogen-activated protein kinase, MAPK, MEK inhibitor
Sections
Citation Override
JCSO 2016;14:51-53
Disallow All Ads
Alternative CME
Article PDF Media

Idarucizumab given the nod as the first specific antidote for an oral anticoagulant

Article Type
Changed
Fri, 01/04/2019 - 11:12
Display Headline
Idarucizumab given the nod as the first specific antidote for an oral anticoagulant
In the fall of 2015, idarucizumab became the first drug-specific antidote to an oral anticoagulant to receive regulatory approval from the US Food and Drug Administration. Anticoagulants are designed to inhibit the formation of blood clots and prevent thrombotic disorders, but they can increase bleeding risk in patients who experience trauma or illness or who undergo invasive surgical procedures. For this reason, traditional anticoagulants have antidotes that reverse their therapeutic effects, but newer oral anticoagulants lack specific reversal agents and that has proven a barrier to their use in patients with higher bleeding risk.

Click on the PDF icon at the top of this introduction to read the full article.

 

Article PDF
Issue
The Journal of Community and Supportive Oncology - 14(1)
Publications
Topics
Page Number
8-10
Legacy Keywords
venous thromboembolism, VTE, deep venous thrombosis, pulmonary embolism, idarucizumab, humanized monoclonal antibody fragment, reversal agent, thrombin, direct oral anticoagulants, DOACs, dabigatran, rivaroxaban, apixaban, edoxaban, andexanet alfa
Sections
Article PDF
Article PDF
In the fall of 2015, idarucizumab became the first drug-specific antidote to an oral anticoagulant to receive regulatory approval from the US Food and Drug Administration. Anticoagulants are designed to inhibit the formation of blood clots and prevent thrombotic disorders, but they can increase bleeding risk in patients who experience trauma or illness or who undergo invasive surgical procedures. For this reason, traditional anticoagulants have antidotes that reverse their therapeutic effects, but newer oral anticoagulants lack specific reversal agents and that has proven a barrier to their use in patients with higher bleeding risk.

Click on the PDF icon at the top of this introduction to read the full article.

 

In the fall of 2015, idarucizumab became the first drug-specific antidote to an oral anticoagulant to receive regulatory approval from the US Food and Drug Administration. Anticoagulants are designed to inhibit the formation of blood clots and prevent thrombotic disorders, but they can increase bleeding risk in patients who experience trauma or illness or who undergo invasive surgical procedures. For this reason, traditional anticoagulants have antidotes that reverse their therapeutic effects, but newer oral anticoagulants lack specific reversal agents and that has proven a barrier to their use in patients with higher bleeding risk.

Click on the PDF icon at the top of this introduction to read the full article.

 

Issue
The Journal of Community and Supportive Oncology - 14(1)
Issue
The Journal of Community and Supportive Oncology - 14(1)
Page Number
8-10
Page Number
8-10
Publications
Publications
Topics
Article Type
Display Headline
Idarucizumab given the nod as the first specific antidote for an oral anticoagulant
Display Headline
Idarucizumab given the nod as the first specific antidote for an oral anticoagulant
Legacy Keywords
venous thromboembolism, VTE, deep venous thrombosis, pulmonary embolism, idarucizumab, humanized monoclonal antibody fragment, reversal agent, thrombin, direct oral anticoagulants, DOACs, dabigatran, rivaroxaban, apixaban, edoxaban, andexanet alfa
Legacy Keywords
venous thromboembolism, VTE, deep venous thrombosis, pulmonary embolism, idarucizumab, humanized monoclonal antibody fragment, reversal agent, thrombin, direct oral anticoagulants, DOACs, dabigatran, rivaroxaban, apixaban, edoxaban, andexanet alfa
Sections
Citation Override
JCSO 2016;14:8-1
Disallow All Ads
Alternative CME
Article PDF Media

Filgrastim-sndz debuts as the first biosimilar approved in United States

Article Type
Changed
Fri, 01/04/2019 - 11:11
Display Headline
Filgrastim-sndz debuts as the first biosimilar approved in United States
In March this year, filgrastim-sndz became the first biosimilar to receive approval from the US Food and Drug Administration for use in the United States. Biosimilars are biological products that show comparable quality, efficacy, and safety to a reference drug that is already approved. In this case, filgrastim-sndz, by Sandoz, a Novartis company, is a biosimilar of Amgen’s filgrastim, the reference drug or originator, which is already licensed in the United States for 5 indications, including for cancer patients undergoing treatments that deplete white blood cells, as well as those preparing for autologous peripheral blood stem-cell transplant, or those with severe, chronic neutropenia.1
 
Click on the PDF icon at the top of this introduction to read the full article.
 
Article PDF
Issue
The Journal of Community and Supportive Oncology - 13(12)
Publications
Topics
Page Number
420-422
Legacy Keywords
Biosimilars, filgrastim, figrastim-sndz, granulocyte-colony stimulating factor, G-CSF
Sections
Article PDF
Article PDF
In March this year, filgrastim-sndz became the first biosimilar to receive approval from the US Food and Drug Administration for use in the United States. Biosimilars are biological products that show comparable quality, efficacy, and safety to a reference drug that is already approved. In this case, filgrastim-sndz, by Sandoz, a Novartis company, is a biosimilar of Amgen’s filgrastim, the reference drug or originator, which is already licensed in the United States for 5 indications, including for cancer patients undergoing treatments that deplete white blood cells, as well as those preparing for autologous peripheral blood stem-cell transplant, or those with severe, chronic neutropenia.1
 
Click on the PDF icon at the top of this introduction to read the full article.
 
In March this year, filgrastim-sndz became the first biosimilar to receive approval from the US Food and Drug Administration for use in the United States. Biosimilars are biological products that show comparable quality, efficacy, and safety to a reference drug that is already approved. In this case, filgrastim-sndz, by Sandoz, a Novartis company, is a biosimilar of Amgen’s filgrastim, the reference drug or originator, which is already licensed in the United States for 5 indications, including for cancer patients undergoing treatments that deplete white blood cells, as well as those preparing for autologous peripheral blood stem-cell transplant, or those with severe, chronic neutropenia.1
 
Click on the PDF icon at the top of this introduction to read the full article.
 
Issue
The Journal of Community and Supportive Oncology - 13(12)
Issue
The Journal of Community and Supportive Oncology - 13(12)
Page Number
420-422
Page Number
420-422
Publications
Publications
Topics
Article Type
Display Headline
Filgrastim-sndz debuts as the first biosimilar approved in United States
Display Headline
Filgrastim-sndz debuts as the first biosimilar approved in United States
Legacy Keywords
Biosimilars, filgrastim, figrastim-sndz, granulocyte-colony stimulating factor, G-CSF
Legacy Keywords
Biosimilars, filgrastim, figrastim-sndz, granulocyte-colony stimulating factor, G-CSF
Sections
Citation Override
JCSO 2015;13:420-422
Disallow All Ads
Alternative CME
Article PDF Media

Approval reinstates gefitinib as a therapy for lung cancer

Article Type
Changed
Fri, 01/04/2019 - 11:11
Display Headline
Approval reinstates gefitinib as a therapy for lung cancer
This past summer, the United States joined more than 60 countries worldwide in approving the epidermal growth factor receptor (EGFR) inhibitor gefitinib for the treatment of patients with non-small-cell lung cancer (NSCLC) who harbor certain EGFR mutations. The approval marked “restoration of fortune” for the drug, which originally received accelerated approval from the US Food and Drug Administration (FDA) in 2003 for the treatment of advanced NSCLC after progression on platinum doublet chemotherapy and docetaxel but was voluntarily withdrawn from the market after subsequent confirmatory randomized trials failed to verify a survival benefit.

 

Click on the PDF icon at the top of this introduction to read the full article.

 

Article PDF
Issue
The Journal of Community and Supportive Oncology - 13(11)
Publications
Topics
Page Number
385-387
Legacy Keywords
non-small-cell lung cancer, NSCLC, EGFR, epidermal growth factor receptor, ISEL study, gefitinibtyrosine kinase inhibitor, TKI
Sections
Article PDF
Article PDF
This past summer, the United States joined more than 60 countries worldwide in approving the epidermal growth factor receptor (EGFR) inhibitor gefitinib for the treatment of patients with non-small-cell lung cancer (NSCLC) who harbor certain EGFR mutations. The approval marked “restoration of fortune” for the drug, which originally received accelerated approval from the US Food and Drug Administration (FDA) in 2003 for the treatment of advanced NSCLC after progression on platinum doublet chemotherapy and docetaxel but was voluntarily withdrawn from the market after subsequent confirmatory randomized trials failed to verify a survival benefit.

 

Click on the PDF icon at the top of this introduction to read the full article.

 

This past summer, the United States joined more than 60 countries worldwide in approving the epidermal growth factor receptor (EGFR) inhibitor gefitinib for the treatment of patients with non-small-cell lung cancer (NSCLC) who harbor certain EGFR mutations. The approval marked “restoration of fortune” for the drug, which originally received accelerated approval from the US Food and Drug Administration (FDA) in 2003 for the treatment of advanced NSCLC after progression on platinum doublet chemotherapy and docetaxel but was voluntarily withdrawn from the market after subsequent confirmatory randomized trials failed to verify a survival benefit.

 

Click on the PDF icon at the top of this introduction to read the full article.

 

Issue
The Journal of Community and Supportive Oncology - 13(11)
Issue
The Journal of Community and Supportive Oncology - 13(11)
Page Number
385-387
Page Number
385-387
Publications
Publications
Topics
Article Type
Display Headline
Approval reinstates gefitinib as a therapy for lung cancer
Display Headline
Approval reinstates gefitinib as a therapy for lung cancer
Legacy Keywords
non-small-cell lung cancer, NSCLC, EGFR, epidermal growth factor receptor, ISEL study, gefitinibtyrosine kinase inhibitor, TKI
Legacy Keywords
non-small-cell lung cancer, NSCLC, EGFR, epidermal growth factor receptor, ISEL study, gefitinibtyrosine kinase inhibitor, TKI
Sections
Citation Override
.JCSO 2015;13:385-387
Disallow All Ads
Alternative CME
Article PDF Media

Dinutuximab combination therapy becomes first approval for high-risk neuroblastoma

Article Type
Changed
Wed, 05/26/2021 - 13:56
Display Headline
Dinutuximab combination therapy becomes first approval for high-risk neuroblastoma
The approval of dinutuximab by the US Food and Drug Administration marks the third approval for a pediatric cancer and the first for patients with high-risk neuroblastoma.1 Dinutuximab is an immunotherapeutic agent; a monoclonal antibody (mAb) targeting a glycolipid that is highly expressed on the surface of neuroblastoma cells.

The mAb was approved in combination with the cytokines granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin-2 (IL-2), and the oral retinoid isotretinoin (RA). The approval was based on a pivotal, phase 3, multicenter, open-label, randomized trial conducted by the Children’s Oncology Group between October 2001 and January 2009 that was stopped early after the combination demonstrated superiority over standard therapy with respect to event-free survival (EFS).2

Two hundred and twenty-six patients (mostly pediatric patients, though age up to 31 years at diagnosis was allowed) with high-risk neuroblastoma were enrolled based on the following criteria: age of ≤31 years at diagnosis; completion of induction therapy, autologous stem-cell transplant (SCT), and radiation therapy, with autologous SCT performed within 9 months of initiation of induction therapy; achievement of at least partial response prior to autologous SCT; enrollment between 50-100 days after final autologous SCT; absence of progressive disease; adequate organ function; life expectancy of at least 2 months; and prior enrollment in the COG biology study (ANBL00B1). An additional 25 patients with biopsy-proven residual disease after autologous SCT were also enrolled, but were nonrandomly assigned to the immunotherapy arm and were excluded from the primary outcome analysis. Patients with systemic infections or a requirement for concomitant systemic corticosteroids or immunosuppressant usage were ineligible. 

 

Click on the PDF icon at the top of this introduction to read the full article.

 

Article PDF
Issue
The Journal of Community and Supportive Oncology - 13(10)
Publications
Topics
Page Number
344-346
Legacy Keywords
dinutuximab, neuroblastoma, immunotherapeutic agent, monoclonal antibody, mAb, cytokine, granulocyte macrophage colony-stimulating factor, GM-CSF, interleukin-2, IL-2, retinoid isotretinoin, RA, glycolipid disialoganglioside, GD2
Sections
Article PDF
Article PDF
The approval of dinutuximab by the US Food and Drug Administration marks the third approval for a pediatric cancer and the first for patients with high-risk neuroblastoma.1 Dinutuximab is an immunotherapeutic agent; a monoclonal antibody (mAb) targeting a glycolipid that is highly expressed on the surface of neuroblastoma cells.

The mAb was approved in combination with the cytokines granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin-2 (IL-2), and the oral retinoid isotretinoin (RA). The approval was based on a pivotal, phase 3, multicenter, open-label, randomized trial conducted by the Children’s Oncology Group between October 2001 and January 2009 that was stopped early after the combination demonstrated superiority over standard therapy with respect to event-free survival (EFS).2

Two hundred and twenty-six patients (mostly pediatric patients, though age up to 31 years at diagnosis was allowed) with high-risk neuroblastoma were enrolled based on the following criteria: age of ≤31 years at diagnosis; completion of induction therapy, autologous stem-cell transplant (SCT), and radiation therapy, with autologous SCT performed within 9 months of initiation of induction therapy; achievement of at least partial response prior to autologous SCT; enrollment between 50-100 days after final autologous SCT; absence of progressive disease; adequate organ function; life expectancy of at least 2 months; and prior enrollment in the COG biology study (ANBL00B1). An additional 25 patients with biopsy-proven residual disease after autologous SCT were also enrolled, but were nonrandomly assigned to the immunotherapy arm and were excluded from the primary outcome analysis. Patients with systemic infections or a requirement for concomitant systemic corticosteroids or immunosuppressant usage were ineligible. 

 

Click on the PDF icon at the top of this introduction to read the full article.

 

The approval of dinutuximab by the US Food and Drug Administration marks the third approval for a pediatric cancer and the first for patients with high-risk neuroblastoma.1 Dinutuximab is an immunotherapeutic agent; a monoclonal antibody (mAb) targeting a glycolipid that is highly expressed on the surface of neuroblastoma cells.

The mAb was approved in combination with the cytokines granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin-2 (IL-2), and the oral retinoid isotretinoin (RA). The approval was based on a pivotal, phase 3, multicenter, open-label, randomized trial conducted by the Children’s Oncology Group between October 2001 and January 2009 that was stopped early after the combination demonstrated superiority over standard therapy with respect to event-free survival (EFS).2

Two hundred and twenty-six patients (mostly pediatric patients, though age up to 31 years at diagnosis was allowed) with high-risk neuroblastoma were enrolled based on the following criteria: age of ≤31 years at diagnosis; completion of induction therapy, autologous stem-cell transplant (SCT), and radiation therapy, with autologous SCT performed within 9 months of initiation of induction therapy; achievement of at least partial response prior to autologous SCT; enrollment between 50-100 days after final autologous SCT; absence of progressive disease; adequate organ function; life expectancy of at least 2 months; and prior enrollment in the COG biology study (ANBL00B1). An additional 25 patients with biopsy-proven residual disease after autologous SCT were also enrolled, but were nonrandomly assigned to the immunotherapy arm and were excluded from the primary outcome analysis. Patients with systemic infections or a requirement for concomitant systemic corticosteroids or immunosuppressant usage were ineligible. 

 

Click on the PDF icon at the top of this introduction to read the full article.

 

Issue
The Journal of Community and Supportive Oncology - 13(10)
Issue
The Journal of Community and Supportive Oncology - 13(10)
Page Number
344-346
Page Number
344-346
Publications
Publications
Topics
Article Type
Display Headline
Dinutuximab combination therapy becomes first approval for high-risk neuroblastoma
Display Headline
Dinutuximab combination therapy becomes first approval for high-risk neuroblastoma
Legacy Keywords
dinutuximab, neuroblastoma, immunotherapeutic agent, monoclonal antibody, mAb, cytokine, granulocyte macrophage colony-stimulating factor, GM-CSF, interleukin-2, IL-2, retinoid isotretinoin, RA, glycolipid disialoganglioside, GD2
Legacy Keywords
dinutuximab, neuroblastoma, immunotherapeutic agent, monoclonal antibody, mAb, cytokine, granulocyte macrophage colony-stimulating factor, GM-CSF, interleukin-2, IL-2, retinoid isotretinoin, RA, glycolipid disialoganglioside, GD2
Sections
Citation Override
JCSO 2015;13:344-346
Disallow All Ads
Alternative CME
Article PDF Media