FDA issues warning about counterfeit BiCNU

Article Type
Changed
Fri, 01/04/2019 - 09:51
Display Headline
FDA issues warning about counterfeit BiCNU

A counterfeit version of BiCNU has been detected in foreign countries, the Food and Drug Administration reports.

BiCNU is approved to treat different types of brain cancer, multiple myeloma, and lymphoma (Hodgkin’s and non-Hodgkin’s), manufactured by Emcure Pharmaceuticals, and distributed by Heritage Pharmaceuticals.

There has been no counterfeit BiCNU detected in the United States, but the FDA encourages health care professionals to diligently inspect BiCNU vials before administering the drug to patients.

“While the [National Drug Code] on the outer package of the authentic and counterfeit version might match, the best way to distinguish a counterfeit is to look at the BiCNU vial inside the packaging,” the FDA reported in a written statement, which includes a list of the counterfeit lots.

To report sales solicitation of suspect drugs call the FDA’s office of criminal investigations at 800-551-3989 or e-mail DrugSupplyChainIntegrity@fda.hhs.gov. To report adverse events related to suspect medications, submit a report online at www.fda.gov/medwatch/report.htm.

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

References

Author and Disclosure Information

Publications
Topics
Author and Disclosure Information

Author and Disclosure Information

A counterfeit version of BiCNU has been detected in foreign countries, the Food and Drug Administration reports.

BiCNU is approved to treat different types of brain cancer, multiple myeloma, and lymphoma (Hodgkin’s and non-Hodgkin’s), manufactured by Emcure Pharmaceuticals, and distributed by Heritage Pharmaceuticals.

There has been no counterfeit BiCNU detected in the United States, but the FDA encourages health care professionals to diligently inspect BiCNU vials before administering the drug to patients.

“While the [National Drug Code] on the outer package of the authentic and counterfeit version might match, the best way to distinguish a counterfeit is to look at the BiCNU vial inside the packaging,” the FDA reported in a written statement, which includes a list of the counterfeit lots.

To report sales solicitation of suspect drugs call the FDA’s office of criminal investigations at 800-551-3989 or e-mail DrugSupplyChainIntegrity@fda.hhs.gov. To report adverse events related to suspect medications, submit a report online at www.fda.gov/medwatch/report.htm.

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

A counterfeit version of BiCNU has been detected in foreign countries, the Food and Drug Administration reports.

BiCNU is approved to treat different types of brain cancer, multiple myeloma, and lymphoma (Hodgkin’s and non-Hodgkin’s), manufactured by Emcure Pharmaceuticals, and distributed by Heritage Pharmaceuticals.

There has been no counterfeit BiCNU detected in the United States, but the FDA encourages health care professionals to diligently inspect BiCNU vials before administering the drug to patients.

“While the [National Drug Code] on the outer package of the authentic and counterfeit version might match, the best way to distinguish a counterfeit is to look at the BiCNU vial inside the packaging,” the FDA reported in a written statement, which includes a list of the counterfeit lots.

To report sales solicitation of suspect drugs call the FDA’s office of criminal investigations at 800-551-3989 or e-mail DrugSupplyChainIntegrity@fda.hhs.gov. To report adverse events related to suspect medications, submit a report online at www.fda.gov/medwatch/report.htm.

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

References

References

Publications
Publications
Topics
Article Type
Display Headline
FDA issues warning about counterfeit BiCNU
Display Headline
FDA issues warning about counterfeit BiCNU
Article Source

PURLs Copyright

Inside the Article

New dose-determining approach applied to pembrolizumab

Article Type
Changed
Fri, 01/04/2019 - 13:15
Display Headline
New dose-determining approach applied to pembrolizumab

A 2-mg/kg dosage of pembrolizumab given to adult patients with non–small-cell lung cancer (NSCLC) every 3 weeks is optimal to both reduce tumor size and avoid treatment-related adverse events, according to a new method of analysis.

Current methods for determining the maximum tolerated dose (MTD) based on dose-limiting toxicities may be outmoded for targeted therapies and immunotherapies, where the biologically effective dose (BED) may be much lower than the MTD.

“Using the MTD rather than the BED could expose patients to a higher dose than that necessary to achieve clinical effect and may increase toxicity, which could lower overall clinical effectiveness. Therefore, dose determination in oncology should use a multifactorial approach that includes not only clinical data from the first treatment cycle but extended clinical data, preclinical models, pharmacokinetics, pharmacodynamics, and integrated modeling and simulation,” said Dr. Manash Chatterjee of Merck, and associates (Ann Oncol. 2016 April 26. doi: 10.1093/annonc/mdw174).

Dr. Chatterjee and associates analyzed data from KEYNOTE-001 using various statistical and mathematical modeling techniques to determine the lowest effective drug dosage of pembrolizumab, a highly selective humanized IgG4 monoclonal antibody that targets the programmed death-1 receptor, for treatment of patients with NSCLC.

Patients enrolled in the KEYNOTE-001 trial received pembrolizumab at doses of 2 mg/kg every 3 weeks, 10 mg/kg every 3 weeks, or 10 mg/kg every 2 weeks until disease progression or intolerable toxicity, or until the patient or investigator decided to stop treatment. Sixty-one patients were treated with pembrolizumab at 2 mg/kg every 3 weeks, 287 patients were treated at 10 mg/kg every 3 weeks, and 202 patients were treated at 10 mg/kg every 2 weeks. PD-1 expression was assessed from biopsy samples using a prototype assay.

Decreases from baseline tumor size were observed for 67% of patients with known PD-L1 expression treated with pembrolizumab at 2 mg/kg every 3 weeks, 66% for patients treated at 10 mg/kg every 3 weeks, and 63% for patients treated at 10 mg/kg every 2 weeks.

Data on occurrence and severity of adverse events were collected throughout the study and up to 60 days after treatment was discontinued. Adverse events were graded according to the National Cancer Institute’s Common Terminology Criteria for Adverse Events (version 4.0). Treatment-related adverse events were reported for 47% of patients treated at 2 mg/kg every 3 weeks. The frequency and severity of adverse events were similar for all dosages, the investigators reported.

Exposure-efficacy was evaluated by way of exploratory regression analyses followed by nonlinear mixed effects modeling. The model found no significant difference in the exposure-efficacy relationship based on dosage. PD-L1 expression and EGFR mutation were the only two factors that explained a significant portion of interindividual variability in tumor size decrease.

Exposure-safety was evaluated via logistic regression modeling where safety was defined as the frequency of adverse events. The model revealed that there was no significant relationship between adverse events and exposure to pembrolizumab or dosage. Overall duration of treatment was the only factor that was significantly related to adverse events.

“These results support the use of a 2-mg/kg Q3W dosage in patients with previously treated, advanced NSCLC,” the investigators concluded.

jcraig@frontlinemedcom.com

On Twitter@jess_craig94

References

Author and Disclosure Information

Publications
Topics
Author and Disclosure Information

Author and Disclosure Information

A 2-mg/kg dosage of pembrolizumab given to adult patients with non–small-cell lung cancer (NSCLC) every 3 weeks is optimal to both reduce tumor size and avoid treatment-related adverse events, according to a new method of analysis.

Current methods for determining the maximum tolerated dose (MTD) based on dose-limiting toxicities may be outmoded for targeted therapies and immunotherapies, where the biologically effective dose (BED) may be much lower than the MTD.

“Using the MTD rather than the BED could expose patients to a higher dose than that necessary to achieve clinical effect and may increase toxicity, which could lower overall clinical effectiveness. Therefore, dose determination in oncology should use a multifactorial approach that includes not only clinical data from the first treatment cycle but extended clinical data, preclinical models, pharmacokinetics, pharmacodynamics, and integrated modeling and simulation,” said Dr. Manash Chatterjee of Merck, and associates (Ann Oncol. 2016 April 26. doi: 10.1093/annonc/mdw174).

Dr. Chatterjee and associates analyzed data from KEYNOTE-001 using various statistical and mathematical modeling techniques to determine the lowest effective drug dosage of pembrolizumab, a highly selective humanized IgG4 monoclonal antibody that targets the programmed death-1 receptor, for treatment of patients with NSCLC.

Patients enrolled in the KEYNOTE-001 trial received pembrolizumab at doses of 2 mg/kg every 3 weeks, 10 mg/kg every 3 weeks, or 10 mg/kg every 2 weeks until disease progression or intolerable toxicity, or until the patient or investigator decided to stop treatment. Sixty-one patients were treated with pembrolizumab at 2 mg/kg every 3 weeks, 287 patients were treated at 10 mg/kg every 3 weeks, and 202 patients were treated at 10 mg/kg every 2 weeks. PD-1 expression was assessed from biopsy samples using a prototype assay.

Decreases from baseline tumor size were observed for 67% of patients with known PD-L1 expression treated with pembrolizumab at 2 mg/kg every 3 weeks, 66% for patients treated at 10 mg/kg every 3 weeks, and 63% for patients treated at 10 mg/kg every 2 weeks.

Data on occurrence and severity of adverse events were collected throughout the study and up to 60 days after treatment was discontinued. Adverse events were graded according to the National Cancer Institute’s Common Terminology Criteria for Adverse Events (version 4.0). Treatment-related adverse events were reported for 47% of patients treated at 2 mg/kg every 3 weeks. The frequency and severity of adverse events were similar for all dosages, the investigators reported.

Exposure-efficacy was evaluated by way of exploratory regression analyses followed by nonlinear mixed effects modeling. The model found no significant difference in the exposure-efficacy relationship based on dosage. PD-L1 expression and EGFR mutation were the only two factors that explained a significant portion of interindividual variability in tumor size decrease.

Exposure-safety was evaluated via logistic regression modeling where safety was defined as the frequency of adverse events. The model revealed that there was no significant relationship between adverse events and exposure to pembrolizumab or dosage. Overall duration of treatment was the only factor that was significantly related to adverse events.

“These results support the use of a 2-mg/kg Q3W dosage in patients with previously treated, advanced NSCLC,” the investigators concluded.

jcraig@frontlinemedcom.com

On Twitter@jess_craig94

A 2-mg/kg dosage of pembrolizumab given to adult patients with non–small-cell lung cancer (NSCLC) every 3 weeks is optimal to both reduce tumor size and avoid treatment-related adverse events, according to a new method of analysis.

Current methods for determining the maximum tolerated dose (MTD) based on dose-limiting toxicities may be outmoded for targeted therapies and immunotherapies, where the biologically effective dose (BED) may be much lower than the MTD.

“Using the MTD rather than the BED could expose patients to a higher dose than that necessary to achieve clinical effect and may increase toxicity, which could lower overall clinical effectiveness. Therefore, dose determination in oncology should use a multifactorial approach that includes not only clinical data from the first treatment cycle but extended clinical data, preclinical models, pharmacokinetics, pharmacodynamics, and integrated modeling and simulation,” said Dr. Manash Chatterjee of Merck, and associates (Ann Oncol. 2016 April 26. doi: 10.1093/annonc/mdw174).

Dr. Chatterjee and associates analyzed data from KEYNOTE-001 using various statistical and mathematical modeling techniques to determine the lowest effective drug dosage of pembrolizumab, a highly selective humanized IgG4 monoclonal antibody that targets the programmed death-1 receptor, for treatment of patients with NSCLC.

Patients enrolled in the KEYNOTE-001 trial received pembrolizumab at doses of 2 mg/kg every 3 weeks, 10 mg/kg every 3 weeks, or 10 mg/kg every 2 weeks until disease progression or intolerable toxicity, or until the patient or investigator decided to stop treatment. Sixty-one patients were treated with pembrolizumab at 2 mg/kg every 3 weeks, 287 patients were treated at 10 mg/kg every 3 weeks, and 202 patients were treated at 10 mg/kg every 2 weeks. PD-1 expression was assessed from biopsy samples using a prototype assay.

Decreases from baseline tumor size were observed for 67% of patients with known PD-L1 expression treated with pembrolizumab at 2 mg/kg every 3 weeks, 66% for patients treated at 10 mg/kg every 3 weeks, and 63% for patients treated at 10 mg/kg every 2 weeks.

Data on occurrence and severity of adverse events were collected throughout the study and up to 60 days after treatment was discontinued. Adverse events were graded according to the National Cancer Institute’s Common Terminology Criteria for Adverse Events (version 4.0). Treatment-related adverse events were reported for 47% of patients treated at 2 mg/kg every 3 weeks. The frequency and severity of adverse events were similar for all dosages, the investigators reported.

Exposure-efficacy was evaluated by way of exploratory regression analyses followed by nonlinear mixed effects modeling. The model found no significant difference in the exposure-efficacy relationship based on dosage. PD-L1 expression and EGFR mutation were the only two factors that explained a significant portion of interindividual variability in tumor size decrease.

Exposure-safety was evaluated via logistic regression modeling where safety was defined as the frequency of adverse events. The model revealed that there was no significant relationship between adverse events and exposure to pembrolizumab or dosage. Overall duration of treatment was the only factor that was significantly related to adverse events.

“These results support the use of a 2-mg/kg Q3W dosage in patients with previously treated, advanced NSCLC,” the investigators concluded.

jcraig@frontlinemedcom.com

On Twitter@jess_craig94

References

References

Publications
Publications
Topics
Article Type
Display Headline
New dose-determining approach applied to pembrolizumab
Display Headline
New dose-determining approach applied to pembrolizumab
Article Source

FROM ANNALS OF ONCOLOGY

PURLs Copyright

Inside the Article

Vitals

Key clinical point: A 2-mg/kg dosage of pembrolizumab given to adult NSCLC patients every 3 weeks is optimal to both reduce tumor size and avoid treatment-related adverse events.

Major finding: Mathematical models of exposure-efficacy and exposure-safety revealed no significant relationships between pembrolizumab dosage and tumor size decrease or treatment-related adverse events.

Data source: KEYNOTE-001, a multicenter, open-label, phase Ib trial of 550 patients with advanced NSCLC. The study included multiple expansion cohorts.

Disclosures: The study was supported by Merck. Six investigators reported receiving grant support, personal fees, or nonfinancial support from several companies. Thirteen investigators are full-time employees of Merck and may hold stock options in the company. The remaining four investigators reported having no financial disclosures.

CIMAvax comes to U.S. after years of development in Cuba

Article Type
Changed
Fri, 01/18/2019 - 15:55
Display Headline
CIMAvax comes to U.S. after years of development in Cuba

The Molecular Immunology Center is a campus of concrete and stone buildings with dark-tinted windows set against palm trees and pink tropical flowers. The buildings are imposing and seem to stretch endlessly along the flat terrain of Havana, Cuba. The research institute, like most buildings in Cuba, is just a few minutes’ walk to the Atlantic Ocean coastline. Inside the research and development building, investigators are working on creating 18 new cancer immunotherapy treatments from vaccines to monoclonal antibodies to synthetic cytokines. A few buildings down, in an almost 50,000–square foot manufacturing plant, a positive pressure air gradient system whirs, and stirred tank bioreactors clank in continuous motion. It is here that the CIMAvax-EGF, a therapeutic vaccine used to treat non–small-cell lung cancer (NSCLC), is manufactured, formulated, and packaged into 40-L sterile bulk bags.

Shown is the old city of Havana.
©Konstik/Thinkstock
Shown is the old city of Havana.

The Molecular Immunology Center, also called the Center of Molecular Immunology or Centro de Immunologia Molecular, was founded in the early 1990’s and the research that eventually led to CIMAvax-EGF began almost immediately, spurred by Cuba’s high rate of lung cancer–related deaths (at the time the second leading cause of death and the leading cause of cancer mortality in the country).

The vaccine works by stimulating a patient’s own immune system to make antibodies against epidermal growth factor (EGF), which depletes circulating EGF levels and prevents EGF from binding to its receptor. “The epidermal growth factor receptor (EGFR) is a well-known oncogene. Its overactivation can induce malignant transformation of a normal cell, signaling inhibition of apoptosis, cell proliferation, angiogenesis, metastasis, and tumor-induced proinflammatory and immunosuppressive processes,” wrote Dr. Pedro Rodríguez and his associates at the Molecular Immunology Center in Cuba (MEDICC Rev. 2010;12:17-23).

“The EGFR signaling and transduction pathway can be efficiently interrupted by EGF deprivation, direct specific mAb [monoclonal antibody] receptor inhibition, or low-molecular-weight molecules competing intracellularly with adenosine triphosphate for the receptor’s tyrosine kinase activity site, with negative repercussions on cell proliferation and, consequently, on tumor development. Inducing EGF deprivation by active immunotherapy is an emerging concept developed by Cuban researchers that involves manipulating an individual’s immune response to release its own effector antibodies against EGF, thereby reducing tumor size or preventing its progression,” the investigators wrote.

Early preclinical studies for vaccine formulation began in Cuba in 1992. Over the next few years, researchers perfected the vaccine’s formula: EGF along with two immunopotentiating molecules, an adjuvant called Montanide ISA 51 and the virulent protein P64k from the microbial organism Neisseria meningitidis (Ann Oncol. 1998;9:431-5; Ann Oncol. 2003;14:461-6).

Together, the adjuvant and the virulent protein activate the immune system while the overabundance of EGF in the body directs antibodies to be made specifically against EGF and not virulent protein P64k. Although some antibodies bind the EGF introduced by the vaccine, other antibodies target cancerous cells where EGFR is overexpressed.

NSCLC was selected “because of its frequency and because EGFR is overexpressed in tissues during development and progression of lung neoplasms,” reported the investigators.

Subsequent clinical trials focused on fine-tuning administration route, dose, dosing interval, optimal combinations with other established therapies, and reducing vaccine-related side effects, which include fever, chills, nausea, headache, asthenia, and tremor.

Researchers found that administering the vaccine in “high but fractioned dose[s] in multiple anatomical sites (such as the 2 deltoid and 2 gluteal regions), thereby bringing the EGF vaccine closer to regional lymph nodes and synergizing the immune response” resulted in the best patient outcomes. In addition, significantly better patient outcomes were achieved when the vaccine was administered before and after chemotherapy.

By 1995, Cuban researchers had successfully completed five separate phase I/II clinical trials and one phase II clinical trial.

Pooling the results from the various clinical trials, non–small-cell lung cancer patients who received the vaccine and who were under the age of 60, lived an average of 4-6 months longer than patients who did not receive the vaccine or were older than 60.

By 2009 “more than 700 patients [had] received the vaccine over the years, many of them in seven clinical trials in Cuba, Canada, and the U.K.,” reported an online Cuban newspaper, Cuba Headlines.

It wasn’t until September 2011 that Cuban researchers officially began distributing the vaccine to Cuban citizens.

“The drug could turn the cancer into a manageable, chronic disease by generating antibodies against the proteins, which triggered the uncontrolled cell proliferation,” Dr. Gisela Gonzalez of the Center of Molecular Immunology said in an interview in 2009 following the official launch of the vaccine.

Today, ongoing clinical trials continue in Cuba and are also underway (at various stages) in several other countries, including the United States. The vaccine still has a long way to go before it can be hailed as a true “cancer cure,” a sentiment that many American publications expressed and one that the researchers at Roswell Park Cancer Institute who are leading the U.S. clinical trials are vehemently trying to quell. Nonetheless, the now global reach of CIMAvax may mark a promising paradigm shift in the field of oncology.

 

 

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

References

Author and Disclosure Information

Publications
Topics
Author and Disclosure Information

Author and Disclosure Information

The Molecular Immunology Center is a campus of concrete and stone buildings with dark-tinted windows set against palm trees and pink tropical flowers. The buildings are imposing and seem to stretch endlessly along the flat terrain of Havana, Cuba. The research institute, like most buildings in Cuba, is just a few minutes’ walk to the Atlantic Ocean coastline. Inside the research and development building, investigators are working on creating 18 new cancer immunotherapy treatments from vaccines to monoclonal antibodies to synthetic cytokines. A few buildings down, in an almost 50,000–square foot manufacturing plant, a positive pressure air gradient system whirs, and stirred tank bioreactors clank in continuous motion. It is here that the CIMAvax-EGF, a therapeutic vaccine used to treat non–small-cell lung cancer (NSCLC), is manufactured, formulated, and packaged into 40-L sterile bulk bags.

Shown is the old city of Havana.
©Konstik/Thinkstock
Shown is the old city of Havana.

The Molecular Immunology Center, also called the Center of Molecular Immunology or Centro de Immunologia Molecular, was founded in the early 1990’s and the research that eventually led to CIMAvax-EGF began almost immediately, spurred by Cuba’s high rate of lung cancer–related deaths (at the time the second leading cause of death and the leading cause of cancer mortality in the country).

The vaccine works by stimulating a patient’s own immune system to make antibodies against epidermal growth factor (EGF), which depletes circulating EGF levels and prevents EGF from binding to its receptor. “The epidermal growth factor receptor (EGFR) is a well-known oncogene. Its overactivation can induce malignant transformation of a normal cell, signaling inhibition of apoptosis, cell proliferation, angiogenesis, metastasis, and tumor-induced proinflammatory and immunosuppressive processes,” wrote Dr. Pedro Rodríguez and his associates at the Molecular Immunology Center in Cuba (MEDICC Rev. 2010;12:17-23).

“The EGFR signaling and transduction pathway can be efficiently interrupted by EGF deprivation, direct specific mAb [monoclonal antibody] receptor inhibition, or low-molecular-weight molecules competing intracellularly with adenosine triphosphate for the receptor’s tyrosine kinase activity site, with negative repercussions on cell proliferation and, consequently, on tumor development. Inducing EGF deprivation by active immunotherapy is an emerging concept developed by Cuban researchers that involves manipulating an individual’s immune response to release its own effector antibodies against EGF, thereby reducing tumor size or preventing its progression,” the investigators wrote.

Early preclinical studies for vaccine formulation began in Cuba in 1992. Over the next few years, researchers perfected the vaccine’s formula: EGF along with two immunopotentiating molecules, an adjuvant called Montanide ISA 51 and the virulent protein P64k from the microbial organism Neisseria meningitidis (Ann Oncol. 1998;9:431-5; Ann Oncol. 2003;14:461-6).

Together, the adjuvant and the virulent protein activate the immune system while the overabundance of EGF in the body directs antibodies to be made specifically against EGF and not virulent protein P64k. Although some antibodies bind the EGF introduced by the vaccine, other antibodies target cancerous cells where EGFR is overexpressed.

NSCLC was selected “because of its frequency and because EGFR is overexpressed in tissues during development and progression of lung neoplasms,” reported the investigators.

Subsequent clinical trials focused on fine-tuning administration route, dose, dosing interval, optimal combinations with other established therapies, and reducing vaccine-related side effects, which include fever, chills, nausea, headache, asthenia, and tremor.

Researchers found that administering the vaccine in “high but fractioned dose[s] in multiple anatomical sites (such as the 2 deltoid and 2 gluteal regions), thereby bringing the EGF vaccine closer to regional lymph nodes and synergizing the immune response” resulted in the best patient outcomes. In addition, significantly better patient outcomes were achieved when the vaccine was administered before and after chemotherapy.

By 1995, Cuban researchers had successfully completed five separate phase I/II clinical trials and one phase II clinical trial.

Pooling the results from the various clinical trials, non–small-cell lung cancer patients who received the vaccine and who were under the age of 60, lived an average of 4-6 months longer than patients who did not receive the vaccine or were older than 60.

By 2009 “more than 700 patients [had] received the vaccine over the years, many of them in seven clinical trials in Cuba, Canada, and the U.K.,” reported an online Cuban newspaper, Cuba Headlines.

It wasn’t until September 2011 that Cuban researchers officially began distributing the vaccine to Cuban citizens.

“The drug could turn the cancer into a manageable, chronic disease by generating antibodies against the proteins, which triggered the uncontrolled cell proliferation,” Dr. Gisela Gonzalez of the Center of Molecular Immunology said in an interview in 2009 following the official launch of the vaccine.

Today, ongoing clinical trials continue in Cuba and are also underway (at various stages) in several other countries, including the United States. The vaccine still has a long way to go before it can be hailed as a true “cancer cure,” a sentiment that many American publications expressed and one that the researchers at Roswell Park Cancer Institute who are leading the U.S. clinical trials are vehemently trying to quell. Nonetheless, the now global reach of CIMAvax may mark a promising paradigm shift in the field of oncology.

 

 

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

The Molecular Immunology Center is a campus of concrete and stone buildings with dark-tinted windows set against palm trees and pink tropical flowers. The buildings are imposing and seem to stretch endlessly along the flat terrain of Havana, Cuba. The research institute, like most buildings in Cuba, is just a few minutes’ walk to the Atlantic Ocean coastline. Inside the research and development building, investigators are working on creating 18 new cancer immunotherapy treatments from vaccines to monoclonal antibodies to synthetic cytokines. A few buildings down, in an almost 50,000–square foot manufacturing plant, a positive pressure air gradient system whirs, and stirred tank bioreactors clank in continuous motion. It is here that the CIMAvax-EGF, a therapeutic vaccine used to treat non–small-cell lung cancer (NSCLC), is manufactured, formulated, and packaged into 40-L sterile bulk bags.

Shown is the old city of Havana.
©Konstik/Thinkstock
Shown is the old city of Havana.

The Molecular Immunology Center, also called the Center of Molecular Immunology or Centro de Immunologia Molecular, was founded in the early 1990’s and the research that eventually led to CIMAvax-EGF began almost immediately, spurred by Cuba’s high rate of lung cancer–related deaths (at the time the second leading cause of death and the leading cause of cancer mortality in the country).

The vaccine works by stimulating a patient’s own immune system to make antibodies against epidermal growth factor (EGF), which depletes circulating EGF levels and prevents EGF from binding to its receptor. “The epidermal growth factor receptor (EGFR) is a well-known oncogene. Its overactivation can induce malignant transformation of a normal cell, signaling inhibition of apoptosis, cell proliferation, angiogenesis, metastasis, and tumor-induced proinflammatory and immunosuppressive processes,” wrote Dr. Pedro Rodríguez and his associates at the Molecular Immunology Center in Cuba (MEDICC Rev. 2010;12:17-23).

“The EGFR signaling and transduction pathway can be efficiently interrupted by EGF deprivation, direct specific mAb [monoclonal antibody] receptor inhibition, or low-molecular-weight molecules competing intracellularly with adenosine triphosphate for the receptor’s tyrosine kinase activity site, with negative repercussions on cell proliferation and, consequently, on tumor development. Inducing EGF deprivation by active immunotherapy is an emerging concept developed by Cuban researchers that involves manipulating an individual’s immune response to release its own effector antibodies against EGF, thereby reducing tumor size or preventing its progression,” the investigators wrote.

Early preclinical studies for vaccine formulation began in Cuba in 1992. Over the next few years, researchers perfected the vaccine’s formula: EGF along with two immunopotentiating molecules, an adjuvant called Montanide ISA 51 and the virulent protein P64k from the microbial organism Neisseria meningitidis (Ann Oncol. 1998;9:431-5; Ann Oncol. 2003;14:461-6).

Together, the adjuvant and the virulent protein activate the immune system while the overabundance of EGF in the body directs antibodies to be made specifically against EGF and not virulent protein P64k. Although some antibodies bind the EGF introduced by the vaccine, other antibodies target cancerous cells where EGFR is overexpressed.

NSCLC was selected “because of its frequency and because EGFR is overexpressed in tissues during development and progression of lung neoplasms,” reported the investigators.

Subsequent clinical trials focused on fine-tuning administration route, dose, dosing interval, optimal combinations with other established therapies, and reducing vaccine-related side effects, which include fever, chills, nausea, headache, asthenia, and tremor.

Researchers found that administering the vaccine in “high but fractioned dose[s] in multiple anatomical sites (such as the 2 deltoid and 2 gluteal regions), thereby bringing the EGF vaccine closer to regional lymph nodes and synergizing the immune response” resulted in the best patient outcomes. In addition, significantly better patient outcomes were achieved when the vaccine was administered before and after chemotherapy.

By 1995, Cuban researchers had successfully completed five separate phase I/II clinical trials and one phase II clinical trial.

Pooling the results from the various clinical trials, non–small-cell lung cancer patients who received the vaccine and who were under the age of 60, lived an average of 4-6 months longer than patients who did not receive the vaccine or were older than 60.

By 2009 “more than 700 patients [had] received the vaccine over the years, many of them in seven clinical trials in Cuba, Canada, and the U.K.,” reported an online Cuban newspaper, Cuba Headlines.

It wasn’t until September 2011 that Cuban researchers officially began distributing the vaccine to Cuban citizens.

“The drug could turn the cancer into a manageable, chronic disease by generating antibodies against the proteins, which triggered the uncontrolled cell proliferation,” Dr. Gisela Gonzalez of the Center of Molecular Immunology said in an interview in 2009 following the official launch of the vaccine.

Today, ongoing clinical trials continue in Cuba and are also underway (at various stages) in several other countries, including the United States. The vaccine still has a long way to go before it can be hailed as a true “cancer cure,” a sentiment that many American publications expressed and one that the researchers at Roswell Park Cancer Institute who are leading the U.S. clinical trials are vehemently trying to quell. Nonetheless, the now global reach of CIMAvax may mark a promising paradigm shift in the field of oncology.

 

 

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

References

References

Publications
Publications
Topics
Article Type
Display Headline
CIMAvax comes to U.S. after years of development in Cuba
Display Headline
CIMAvax comes to U.S. after years of development in Cuba
Article Source

PURLs Copyright

Inside the Article

CAR T-cell start-up launched

Article Type
Changed
Fri, 12/16/2022 - 11:37
Display Headline
CAR T-cell start-up launched

Dr. Siddhartha Mukherjee has partnered with Puretech Health to launch a new biotechnology and immuno-oncology company to broaden the use of chimeric antigen receptor (CAR) T-cell therapy. Dr. Mukherjee, a Columbia University researcher, hematologist, oncologist, and Pulitzer Prize–winning author of “The Emperor of All Maladies: A Biography of Cancer,” (New York: Scribner, a division of Simon & Schuster, 2011) is licensing his CAR T-cell technology to the joint venture, called Vor BioPharma.

Vor BioPharma will focus on advancing and expanding CAR T-cell therapy, a relatively new cancer treatment where T cells are first collected from a patient’s blood and then genetically engineered to produce CAR proteins on their surface. The CAR proteins are designed to bind specific antigens found on the patient’s cancer cells. These genetically engineered T cells are grown in a laboratory and then infused into the patient. As of now, CAR T-cell therapy is primarily used to treat B-cell leukemias and other chronic lymphocytic leukemia.

“We continue to make great strides in developing new ways to treat cancer using the body’s immune system,” said Dr. Mukherjee in a written statement announcing the partnership. “The positive clinical response researchers have achieved with CAR T-cell therapies in B-cell leukemias has led to great interest within the oncology community and is something we hope to achieve in other cancers over time,” he said.

“CAR T-cell therapies have shown remarkable progress in the clinic, yet their applicability beyond a small subset of cancers is currently very limited,” said Dr. Sanjiv Sam Gambhir of Stanford University and a member of the Vor Scientific Advisory Board. “This technology seeks to address bottlenecks that prevent CAR T-cell therapy from becoming more broadly useful in treating cancers outside of B-cell cancers.”

Other Vor BioPharma employees and Scientific Advisory Board members include Dr. Joseph Bolen, former President and Chief Scientific Officer of Moderna Therapeutics; Dr. Dan Littman of the Howard Hughes Medical Institute; and Dr. Derrick Rossi of Harvard University.

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

References

Author and Disclosure Information

Publications
Topics
Author and Disclosure Information

Author and Disclosure Information

Dr. Siddhartha Mukherjee has partnered with Puretech Health to launch a new biotechnology and immuno-oncology company to broaden the use of chimeric antigen receptor (CAR) T-cell therapy. Dr. Mukherjee, a Columbia University researcher, hematologist, oncologist, and Pulitzer Prize–winning author of “The Emperor of All Maladies: A Biography of Cancer,” (New York: Scribner, a division of Simon & Schuster, 2011) is licensing his CAR T-cell technology to the joint venture, called Vor BioPharma.

Vor BioPharma will focus on advancing and expanding CAR T-cell therapy, a relatively new cancer treatment where T cells are first collected from a patient’s blood and then genetically engineered to produce CAR proteins on their surface. The CAR proteins are designed to bind specific antigens found on the patient’s cancer cells. These genetically engineered T cells are grown in a laboratory and then infused into the patient. As of now, CAR T-cell therapy is primarily used to treat B-cell leukemias and other chronic lymphocytic leukemia.

“We continue to make great strides in developing new ways to treat cancer using the body’s immune system,” said Dr. Mukherjee in a written statement announcing the partnership. “The positive clinical response researchers have achieved with CAR T-cell therapies in B-cell leukemias has led to great interest within the oncology community and is something we hope to achieve in other cancers over time,” he said.

“CAR T-cell therapies have shown remarkable progress in the clinic, yet their applicability beyond a small subset of cancers is currently very limited,” said Dr. Sanjiv Sam Gambhir of Stanford University and a member of the Vor Scientific Advisory Board. “This technology seeks to address bottlenecks that prevent CAR T-cell therapy from becoming more broadly useful in treating cancers outside of B-cell cancers.”

Other Vor BioPharma employees and Scientific Advisory Board members include Dr. Joseph Bolen, former President and Chief Scientific Officer of Moderna Therapeutics; Dr. Dan Littman of the Howard Hughes Medical Institute; and Dr. Derrick Rossi of Harvard University.

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

Dr. Siddhartha Mukherjee has partnered with Puretech Health to launch a new biotechnology and immuno-oncology company to broaden the use of chimeric antigen receptor (CAR) T-cell therapy. Dr. Mukherjee, a Columbia University researcher, hematologist, oncologist, and Pulitzer Prize–winning author of “The Emperor of All Maladies: A Biography of Cancer,” (New York: Scribner, a division of Simon & Schuster, 2011) is licensing his CAR T-cell technology to the joint venture, called Vor BioPharma.

Vor BioPharma will focus on advancing and expanding CAR T-cell therapy, a relatively new cancer treatment where T cells are first collected from a patient’s blood and then genetically engineered to produce CAR proteins on their surface. The CAR proteins are designed to bind specific antigens found on the patient’s cancer cells. These genetically engineered T cells are grown in a laboratory and then infused into the patient. As of now, CAR T-cell therapy is primarily used to treat B-cell leukemias and other chronic lymphocytic leukemia.

“We continue to make great strides in developing new ways to treat cancer using the body’s immune system,” said Dr. Mukherjee in a written statement announcing the partnership. “The positive clinical response researchers have achieved with CAR T-cell therapies in B-cell leukemias has led to great interest within the oncology community and is something we hope to achieve in other cancers over time,” he said.

“CAR T-cell therapies have shown remarkable progress in the clinic, yet their applicability beyond a small subset of cancers is currently very limited,” said Dr. Sanjiv Sam Gambhir of Stanford University and a member of the Vor Scientific Advisory Board. “This technology seeks to address bottlenecks that prevent CAR T-cell therapy from becoming more broadly useful in treating cancers outside of B-cell cancers.”

Other Vor BioPharma employees and Scientific Advisory Board members include Dr. Joseph Bolen, former President and Chief Scientific Officer of Moderna Therapeutics; Dr. Dan Littman of the Howard Hughes Medical Institute; and Dr. Derrick Rossi of Harvard University.

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

References

References

Publications
Publications
Topics
Article Type
Display Headline
CAR T-cell start-up launched
Display Headline
CAR T-cell start-up launched
Article Source

PURLs Copyright

Inside the Article

Chemoradiotherapy, erlotinib gave no survival boost to advanced pancreatic cancer patients

Progress but not precision
Article Type
Changed
Wed, 05/26/2021 - 13:54
Display Headline
Chemoradiotherapy, erlotinib gave no survival boost to advanced pancreatic cancer patients

Chemoradiotherapy does not improve overall or progression-free survival in patients with advanced pancreatic cancer, compared with chemotherapy alone. Gemcitabine plus erlotinib also does not improve overall or progression-free survival when compared with patients who received only gemcitabine.

“This open-label, randomized clinical trial showed no survival benefit of chemoradiotherapy compared with chemotherapy in patients with locally advanced pancreatic cancer... the addition of erlotinib to gemcitabine, despite excellent adherence (92%), failed to improve survival and yet was associated with increased grade 3 hematologic, digestive, and skin toxicities,” wrote Dr. Pascal Hammel of Beaujon Hospital, France, and associates (JAMA. 2016 May 3. doi: 10.1001/jama.2016.4324).

“This suggests that in patients with locally advanced pancreatic cancer, more efficient systemic treatments are needed to treat any early micrometastatic spread and to downstage tumors,” they said.

Investigators enrolled 449 patients with advanced pancreatic cancer in the international, LAP07 phase III trial; 442 met the demographic criteria and were randomly divided into two groups, 223 of which received gemcitabine and 219 of which received gemcitabine plus erlotinib, during the first of two randomization steps. Patients in both groups received their designated drug regime intravenously for three weeks followed by a one week resting period for a total of four cycles.

During step one, 135 of the 223 patients who received gemcitabine, and 134 of the 219 patients who received gemcitabine plus erlotinib, survived the 16-week period progression free and were eligible for step two randomization; 136 patients were then randomly selected to receive chemotherapy and 133 patients were randomly selected to receive chemoradiotherapy.

After the two randomization steps, 68 patients received gemcitabine with chemotherapy, 68 patients received gemcitabine plus erlotinib with chemotherapy, 67 patients received gemcitabine with chemoradiotherapy, and 66 patients received gemcitabine plus erlotinib with chemoradiotherapy.

By the end of the clinical trial, 379 patients had died and 385 had experienced tumor progression. There was no significant difference in overall survival between patients receiving gemcitabine or gemcitabine plus erlotinib (hazard ratio, 1.19; 95% confidence interval, 0.97-1.45; P = .09), and there was no significant difference in progression-free survival (HR, 1.12; 95% CI, 0.92-1.36; P = .26). Patients who received erlotinib were at a significantly elevated risk for experiencing anemia, neutropenia, diarrhea, and acneiform rash when compared with patients who did not receive erlotinib.

There was no significant difference in overall survival between patients receiving chemotherapy or chemoradiotherapy (HR, 1.03; 95% CI, 0.79-1.34; P = .83), and there was no significant difference in progression-free survival (HR, .78; 95% CI, 0.61-1.01; P = .06).

There was also no significant difference in survival when first-step randomization status was combined with second-randomization status (P = .24).

This study was supported by Roche and the French National Institute of Cancer. Dr. Hammel reported receiving consulting fees from Celgene. Seven of the other thirteen investigators reported receiving personal fees, nonfinancial support, grant support, personal fees, or honoraria from Amgen, Merck Serono, Eli Lilly, Roche, Celgene, Sanofi, Novartis, Integragen, Eisai, Invectys, or Nestle.

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

References

Body

Clinical trials that find no difference between groups never garner as much excitement as trials with positive findings. However, clear negative results chart the path forward by informing the design of next-generation studies and hastening retirement of ineffective therapies.

The results of the LAP07 trial are persuasive that contemporary chemoradiation does not add a survival advantage to chemotherapy alone. However, the heterogeneity in response to both chemotherapy and radiation is unclear. What if any features distinguished tumors resistant to treatment and, conversely, those that responded? What features distinguish tumors with propensity to spread locally versus diffusely? Tumors in the former category stand to benefit from regionally focused treatment such as chemoradiation.

The LAP07 trial is progress, but it does not achieve the goal of precision medicine.

The LAP07 trial contributes important new information to help guide treatment decisions for patients with locally advanced pancreas cancer. Ideally, future pancreatic cancer trials will identify molecular markers that better predict responsiveness to specific treatments including radiation and will allow for more focused approaches to treatment selection. In the meantime, chemoradiation need not constitute an essential component of the therapeutic backbone.

Dr. Deborah Schrag is at the Dana Farber Cancer Institute in Boston. Dr. Schrag made these remarks in an editorial accompanying Dr. Hammel’s report (JAMA. 2016 May 3. doi: 10.1001/jama.2016.4284), and she reported having no conflict of interest disclosures.

Author and Disclosure Information

Publications
Topics
Legacy Keywords
pancreatic cancer chemotherapy chemoradiotherapy
Author and Disclosure Information

Author and Disclosure Information

Body

Clinical trials that find no difference between groups never garner as much excitement as trials with positive findings. However, clear negative results chart the path forward by informing the design of next-generation studies and hastening retirement of ineffective therapies.

The results of the LAP07 trial are persuasive that contemporary chemoradiation does not add a survival advantage to chemotherapy alone. However, the heterogeneity in response to both chemotherapy and radiation is unclear. What if any features distinguished tumors resistant to treatment and, conversely, those that responded? What features distinguish tumors with propensity to spread locally versus diffusely? Tumors in the former category stand to benefit from regionally focused treatment such as chemoradiation.

The LAP07 trial is progress, but it does not achieve the goal of precision medicine.

The LAP07 trial contributes important new information to help guide treatment decisions for patients with locally advanced pancreas cancer. Ideally, future pancreatic cancer trials will identify molecular markers that better predict responsiveness to specific treatments including radiation and will allow for more focused approaches to treatment selection. In the meantime, chemoradiation need not constitute an essential component of the therapeutic backbone.

Dr. Deborah Schrag is at the Dana Farber Cancer Institute in Boston. Dr. Schrag made these remarks in an editorial accompanying Dr. Hammel’s report (JAMA. 2016 May 3. doi: 10.1001/jama.2016.4284), and she reported having no conflict of interest disclosures.

Body

Clinical trials that find no difference between groups never garner as much excitement as trials with positive findings. However, clear negative results chart the path forward by informing the design of next-generation studies and hastening retirement of ineffective therapies.

The results of the LAP07 trial are persuasive that contemporary chemoradiation does not add a survival advantage to chemotherapy alone. However, the heterogeneity in response to both chemotherapy and radiation is unclear. What if any features distinguished tumors resistant to treatment and, conversely, those that responded? What features distinguish tumors with propensity to spread locally versus diffusely? Tumors in the former category stand to benefit from regionally focused treatment such as chemoradiation.

The LAP07 trial is progress, but it does not achieve the goal of precision medicine.

The LAP07 trial contributes important new information to help guide treatment decisions for patients with locally advanced pancreas cancer. Ideally, future pancreatic cancer trials will identify molecular markers that better predict responsiveness to specific treatments including radiation and will allow for more focused approaches to treatment selection. In the meantime, chemoradiation need not constitute an essential component of the therapeutic backbone.

Dr. Deborah Schrag is at the Dana Farber Cancer Institute in Boston. Dr. Schrag made these remarks in an editorial accompanying Dr. Hammel’s report (JAMA. 2016 May 3. doi: 10.1001/jama.2016.4284), and she reported having no conflict of interest disclosures.

Title
Progress but not precision
Progress but not precision

Chemoradiotherapy does not improve overall or progression-free survival in patients with advanced pancreatic cancer, compared with chemotherapy alone. Gemcitabine plus erlotinib also does not improve overall or progression-free survival when compared with patients who received only gemcitabine.

“This open-label, randomized clinical trial showed no survival benefit of chemoradiotherapy compared with chemotherapy in patients with locally advanced pancreatic cancer... the addition of erlotinib to gemcitabine, despite excellent adherence (92%), failed to improve survival and yet was associated with increased grade 3 hematologic, digestive, and skin toxicities,” wrote Dr. Pascal Hammel of Beaujon Hospital, France, and associates (JAMA. 2016 May 3. doi: 10.1001/jama.2016.4324).

“This suggests that in patients with locally advanced pancreatic cancer, more efficient systemic treatments are needed to treat any early micrometastatic spread and to downstage tumors,” they said.

Investigators enrolled 449 patients with advanced pancreatic cancer in the international, LAP07 phase III trial; 442 met the demographic criteria and were randomly divided into two groups, 223 of which received gemcitabine and 219 of which received gemcitabine plus erlotinib, during the first of two randomization steps. Patients in both groups received their designated drug regime intravenously for three weeks followed by a one week resting period for a total of four cycles.

During step one, 135 of the 223 patients who received gemcitabine, and 134 of the 219 patients who received gemcitabine plus erlotinib, survived the 16-week period progression free and were eligible for step two randomization; 136 patients were then randomly selected to receive chemotherapy and 133 patients were randomly selected to receive chemoradiotherapy.

After the two randomization steps, 68 patients received gemcitabine with chemotherapy, 68 patients received gemcitabine plus erlotinib with chemotherapy, 67 patients received gemcitabine with chemoradiotherapy, and 66 patients received gemcitabine plus erlotinib with chemoradiotherapy.

By the end of the clinical trial, 379 patients had died and 385 had experienced tumor progression. There was no significant difference in overall survival between patients receiving gemcitabine or gemcitabine plus erlotinib (hazard ratio, 1.19; 95% confidence interval, 0.97-1.45; P = .09), and there was no significant difference in progression-free survival (HR, 1.12; 95% CI, 0.92-1.36; P = .26). Patients who received erlotinib were at a significantly elevated risk for experiencing anemia, neutropenia, diarrhea, and acneiform rash when compared with patients who did not receive erlotinib.

There was no significant difference in overall survival between patients receiving chemotherapy or chemoradiotherapy (HR, 1.03; 95% CI, 0.79-1.34; P = .83), and there was no significant difference in progression-free survival (HR, .78; 95% CI, 0.61-1.01; P = .06).

There was also no significant difference in survival when first-step randomization status was combined with second-randomization status (P = .24).

This study was supported by Roche and the French National Institute of Cancer. Dr. Hammel reported receiving consulting fees from Celgene. Seven of the other thirteen investigators reported receiving personal fees, nonfinancial support, grant support, personal fees, or honoraria from Amgen, Merck Serono, Eli Lilly, Roche, Celgene, Sanofi, Novartis, Integragen, Eisai, Invectys, or Nestle.

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

Chemoradiotherapy does not improve overall or progression-free survival in patients with advanced pancreatic cancer, compared with chemotherapy alone. Gemcitabine plus erlotinib also does not improve overall or progression-free survival when compared with patients who received only gemcitabine.

“This open-label, randomized clinical trial showed no survival benefit of chemoradiotherapy compared with chemotherapy in patients with locally advanced pancreatic cancer... the addition of erlotinib to gemcitabine, despite excellent adherence (92%), failed to improve survival and yet was associated with increased grade 3 hematologic, digestive, and skin toxicities,” wrote Dr. Pascal Hammel of Beaujon Hospital, France, and associates (JAMA. 2016 May 3. doi: 10.1001/jama.2016.4324).

“This suggests that in patients with locally advanced pancreatic cancer, more efficient systemic treatments are needed to treat any early micrometastatic spread and to downstage tumors,” they said.

Investigators enrolled 449 patients with advanced pancreatic cancer in the international, LAP07 phase III trial; 442 met the demographic criteria and were randomly divided into two groups, 223 of which received gemcitabine and 219 of which received gemcitabine plus erlotinib, during the first of two randomization steps. Patients in both groups received their designated drug regime intravenously for three weeks followed by a one week resting period for a total of four cycles.

During step one, 135 of the 223 patients who received gemcitabine, and 134 of the 219 patients who received gemcitabine plus erlotinib, survived the 16-week period progression free and were eligible for step two randomization; 136 patients were then randomly selected to receive chemotherapy and 133 patients were randomly selected to receive chemoradiotherapy.

After the two randomization steps, 68 patients received gemcitabine with chemotherapy, 68 patients received gemcitabine plus erlotinib with chemotherapy, 67 patients received gemcitabine with chemoradiotherapy, and 66 patients received gemcitabine plus erlotinib with chemoradiotherapy.

By the end of the clinical trial, 379 patients had died and 385 had experienced tumor progression. There was no significant difference in overall survival between patients receiving gemcitabine or gemcitabine plus erlotinib (hazard ratio, 1.19; 95% confidence interval, 0.97-1.45; P = .09), and there was no significant difference in progression-free survival (HR, 1.12; 95% CI, 0.92-1.36; P = .26). Patients who received erlotinib were at a significantly elevated risk for experiencing anemia, neutropenia, diarrhea, and acneiform rash when compared with patients who did not receive erlotinib.

There was no significant difference in overall survival between patients receiving chemotherapy or chemoradiotherapy (HR, 1.03; 95% CI, 0.79-1.34; P = .83), and there was no significant difference in progression-free survival (HR, .78; 95% CI, 0.61-1.01; P = .06).

There was also no significant difference in survival when first-step randomization status was combined with second-randomization status (P = .24).

This study was supported by Roche and the French National Institute of Cancer. Dr. Hammel reported receiving consulting fees from Celgene. Seven of the other thirteen investigators reported receiving personal fees, nonfinancial support, grant support, personal fees, or honoraria from Amgen, Merck Serono, Eli Lilly, Roche, Celgene, Sanofi, Novartis, Integragen, Eisai, Invectys, or Nestle.

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

References

References

Publications
Publications
Topics
Article Type
Display Headline
Chemoradiotherapy, erlotinib gave no survival boost to advanced pancreatic cancer patients
Display Headline
Chemoradiotherapy, erlotinib gave no survival boost to advanced pancreatic cancer patients
Legacy Keywords
pancreatic cancer chemotherapy chemoradiotherapy
Legacy Keywords
pancreatic cancer chemotherapy chemoradiotherapy
Article Source

FROM JAMA

PURLs Copyright

Inside the Article

Vitals

Key clinical point: Compared with chemotherapy, chemoradiotherapy did not improve survival outcomes in patients with advanced pancreatic cancer. Supplementing gemcitabine with erlotinib also did not improve survival outcomes.

Major finding: There was no significant difference in overall survival between patients receiving gemcitabine or gemcitabine plus erlotinib (HR, 1.19; 95% CI, 0.97-1.45; P = .09) nor was there a significant difference in progression-free survival (HR, 1.12; 95% CI, 0.92-1.36; P = .26). There was also no significant difference in overall survival between patients receiving chemotherapy or chemoradiotherapy (HR, 1.03; 95% CI, 0.79-1.34; P = .83), and there was no significant difference in progression-free survival (HR, 0.78; 95% CI, 0.61-1.01; P = .06).

Data source: An international, multicenter, open-label, unblinded, randomized phase III clinical trial involving 449 patients with advanced pancreatic cancer.

Disclosures: This study was supported by Roche and the French National Institute of Cancer. Dr. Hammel reported receiving consulting fees from Celgene. Dr. Hammel’s associates reported receiving personal fees, nonfinancial support, grant support, personal fees, or honoraria from Amgen, Merck Serono, Lilly, Roche, Celgene, Sanofi, Novartis, Integragen, Eisai, Invectys, and Nestle.

FDA grants priority review of olaratumab for advanced sarcoma

Article Type
Changed
Fri, 01/04/2019 - 13:15
Display Headline
FDA grants priority review of olaratumab for advanced sarcoma

The Food and Drug Administration has granted priority review of olaratumab, in combination with doxorubicin, for the treatment of patients with advanced soft tissue sarcoma who unsuccessfully underwent prior radiotherapy or surgery for their cancer.

Olaratumab is a human IgG1 monoclonal antibody that directly targets tumor cells by disrupting the platelet-derived growth factor receptor alpha, a receptor that is believed to play a role in tumor growth and progression.

“We are hopeful that, if approved, olaratumab will provide a meaningful addition to the limited treatment options for this rare and difficult-to-treat disease,” Dr. Richard Gaynor, senior vice president of product development and medical affairs for Lilly Oncology, the maker of the drug, said in a written statement issued by company.

The biologics license application submission for olaratumab was based on a phase II clinical trial of 129 patients with metastatic or unresectable soft tissue sarcoma. Sixty-four patients were assigned to the treatment group and received both doxorubicin and olaratumab. Patients in this group continued to receive olaratumab through observed disease progression. Sixty-five patients were assigned to the control group and received only doxorubicin until disease progression was first observed. Patients who received olaratumab in addition to doxorubicin experienced a longer median progression-free survival, compared with patients who only received doxorubicin (6.6 months vs. 4.1 months; stratified hazard ratio, 0.672; 95% confidence interval, 0.442 to 1.021; P = .0615). The results of the phase II clinical trial were reported at the 2015 American Society of Clinical Oncology annual meeting and the 2015 Connective Tissue Oncology Society annual meeting.

The FDA previously granted olaratumab breakthrough therapy, fast track, and orphan drug designation.

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

References

Author and Disclosure Information

Publications
Topics
Author and Disclosure Information

Author and Disclosure Information

The Food and Drug Administration has granted priority review of olaratumab, in combination with doxorubicin, for the treatment of patients with advanced soft tissue sarcoma who unsuccessfully underwent prior radiotherapy or surgery for their cancer.

Olaratumab is a human IgG1 monoclonal antibody that directly targets tumor cells by disrupting the platelet-derived growth factor receptor alpha, a receptor that is believed to play a role in tumor growth and progression.

“We are hopeful that, if approved, olaratumab will provide a meaningful addition to the limited treatment options for this rare and difficult-to-treat disease,” Dr. Richard Gaynor, senior vice president of product development and medical affairs for Lilly Oncology, the maker of the drug, said in a written statement issued by company.

The biologics license application submission for olaratumab was based on a phase II clinical trial of 129 patients with metastatic or unresectable soft tissue sarcoma. Sixty-four patients were assigned to the treatment group and received both doxorubicin and olaratumab. Patients in this group continued to receive olaratumab through observed disease progression. Sixty-five patients were assigned to the control group and received only doxorubicin until disease progression was first observed. Patients who received olaratumab in addition to doxorubicin experienced a longer median progression-free survival, compared with patients who only received doxorubicin (6.6 months vs. 4.1 months; stratified hazard ratio, 0.672; 95% confidence interval, 0.442 to 1.021; P = .0615). The results of the phase II clinical trial were reported at the 2015 American Society of Clinical Oncology annual meeting and the 2015 Connective Tissue Oncology Society annual meeting.

The FDA previously granted olaratumab breakthrough therapy, fast track, and orphan drug designation.

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

The Food and Drug Administration has granted priority review of olaratumab, in combination with doxorubicin, for the treatment of patients with advanced soft tissue sarcoma who unsuccessfully underwent prior radiotherapy or surgery for their cancer.

Olaratumab is a human IgG1 monoclonal antibody that directly targets tumor cells by disrupting the platelet-derived growth factor receptor alpha, a receptor that is believed to play a role in tumor growth and progression.

“We are hopeful that, if approved, olaratumab will provide a meaningful addition to the limited treatment options for this rare and difficult-to-treat disease,” Dr. Richard Gaynor, senior vice president of product development and medical affairs for Lilly Oncology, the maker of the drug, said in a written statement issued by company.

The biologics license application submission for olaratumab was based on a phase II clinical trial of 129 patients with metastatic or unresectable soft tissue sarcoma. Sixty-four patients were assigned to the treatment group and received both doxorubicin and olaratumab. Patients in this group continued to receive olaratumab through observed disease progression. Sixty-five patients were assigned to the control group and received only doxorubicin until disease progression was first observed. Patients who received olaratumab in addition to doxorubicin experienced a longer median progression-free survival, compared with patients who only received doxorubicin (6.6 months vs. 4.1 months; stratified hazard ratio, 0.672; 95% confidence interval, 0.442 to 1.021; P = .0615). The results of the phase II clinical trial were reported at the 2015 American Society of Clinical Oncology annual meeting and the 2015 Connective Tissue Oncology Society annual meeting.

The FDA previously granted olaratumab breakthrough therapy, fast track, and orphan drug designation.

jcraig@frontlinemedcom.com

On Twitter @jess_craig94

References

References

Publications
Publications
Topics
Article Type
Display Headline
FDA grants priority review of olaratumab for advanced sarcoma
Display Headline
FDA grants priority review of olaratumab for advanced sarcoma
Article Source

PURLs Copyright

Inside the Article

Racial disparities found in early-stage, young-onset CRC survival

Article Type
Changed
Wed, 05/26/2021 - 13:54
Display Headline
Racial disparities found in early-stage, young-onset CRC survival

Racial disparities in survival rates among young patients exist for those diagnosed with both early-stage and late-state colorectal cancer (CRC), investigators have found.

A review of Surveillance, Epidemiology, and End Results (SEER) data on young-onset CRC incidence and mortality rates showed that overall and cancer-specific survival rates were significantly lower for non-Hispanic black (NHB) patients when compared with non-Hispanic white (NHW) and Hispanic patients, reported Dr. Andreana Holowatyi of Wayne State University, Detroit, and her associates.

“Study of patients with young-onset CRC offers an opportunity to examine differences in cancer-specific survival by race, minimizing the potential impact that routine CRC screening among individuals 50 years of age and older might have on CRC-related outcomes,” the authors wrote (J Clin Oncol. 2016 May 2. doi: 10.1200/JCO.2015.65.0994).

The authors reviewed SEER data on 28,145 men and women aged 20-49 years at the time of CRC diagnosis. The sample included 19,497 NHWs, 4,384 NHBs, and 4,264 Hispanic patients and excluded patients who were diagnosed with cancer upon autopsy, patients who survived less than 2 months post diagnosis, and patients who were diagnosed with histopathologic subtypes other than adenocarcinoma. Patients diagnosed with any other type of cancer prior to their CRC diagnoses were excluded from survival analysis.

Stratification of patients by tumor stage and site showed that NHB patients were at an increased risk of cancer-specific death from colon cancers diagnosed at every stage, with the greatest differences observed among those with stage II cancers (hazard ratio, 1.69; 95% confidence interval, 1.33-2.14; P less than .001).

NHB patients were more likely to have CRCs of the proximal colon, compared with NHW and Hispanic patients (39.9% vs. 30.3% vs. 30.7% respectively; P less than .001) and more likely to have more advanced stage cancer. In addition, the NHB subpopulation included a significantly higher percentage of women, compared with NHW and Hispanic patients (50.3% vs. 45.8% vs. 46.3%, P less than .001).

In Cox proportional hazard models controlling for age, sex, race, county-level poverty, tumor stage, surgical intervention, and radiation therapy, Dr. Holowatyi and her associates found that NHB patients had significantly higher risk of cancer-specific death in colon cancers (HR, 1.35; 95% CI 1.26 to 1.45; P less than .001) and rectum/rectosigmoid junction cancers (HR, 1.51; 95% CI 1.37 to 1.68; P less than .001), compared with NHWs. Similar findings were reported for risk of overall death.

“Further studies of the clinical and molecular characteristics of young-onset CRCs are needed to explore potential tumor/treatment interactions associated with racial differences in survival and to refine clinical algorithms for CRC treatment and early detection,” the authors wrote.

This study was supported by funding from the Cancer Biology Graduate Program at Wayne State University, the National Cancer Institute, and an Epidemiology Core and National Institutes of Health Center grant. The authors did not have any disclosures to report.

jcraig@frontlinemedcom.com

References

Click for Credit Link
Author and Disclosure Information

Publications
Topics
Click for Credit Link
Click for Credit Link
Author and Disclosure Information

Author and Disclosure Information

Racial disparities in survival rates among young patients exist for those diagnosed with both early-stage and late-state colorectal cancer (CRC), investigators have found.

A review of Surveillance, Epidemiology, and End Results (SEER) data on young-onset CRC incidence and mortality rates showed that overall and cancer-specific survival rates were significantly lower for non-Hispanic black (NHB) patients when compared with non-Hispanic white (NHW) and Hispanic patients, reported Dr. Andreana Holowatyi of Wayne State University, Detroit, and her associates.

“Study of patients with young-onset CRC offers an opportunity to examine differences in cancer-specific survival by race, minimizing the potential impact that routine CRC screening among individuals 50 years of age and older might have on CRC-related outcomes,” the authors wrote (J Clin Oncol. 2016 May 2. doi: 10.1200/JCO.2015.65.0994).

The authors reviewed SEER data on 28,145 men and women aged 20-49 years at the time of CRC diagnosis. The sample included 19,497 NHWs, 4,384 NHBs, and 4,264 Hispanic patients and excluded patients who were diagnosed with cancer upon autopsy, patients who survived less than 2 months post diagnosis, and patients who were diagnosed with histopathologic subtypes other than adenocarcinoma. Patients diagnosed with any other type of cancer prior to their CRC diagnoses were excluded from survival analysis.

Stratification of patients by tumor stage and site showed that NHB patients were at an increased risk of cancer-specific death from colon cancers diagnosed at every stage, with the greatest differences observed among those with stage II cancers (hazard ratio, 1.69; 95% confidence interval, 1.33-2.14; P less than .001).

NHB patients were more likely to have CRCs of the proximal colon, compared with NHW and Hispanic patients (39.9% vs. 30.3% vs. 30.7% respectively; P less than .001) and more likely to have more advanced stage cancer. In addition, the NHB subpopulation included a significantly higher percentage of women, compared with NHW and Hispanic patients (50.3% vs. 45.8% vs. 46.3%, P less than .001).

In Cox proportional hazard models controlling for age, sex, race, county-level poverty, tumor stage, surgical intervention, and radiation therapy, Dr. Holowatyi and her associates found that NHB patients had significantly higher risk of cancer-specific death in colon cancers (HR, 1.35; 95% CI 1.26 to 1.45; P less than .001) and rectum/rectosigmoid junction cancers (HR, 1.51; 95% CI 1.37 to 1.68; P less than .001), compared with NHWs. Similar findings were reported for risk of overall death.

“Further studies of the clinical and molecular characteristics of young-onset CRCs are needed to explore potential tumor/treatment interactions associated with racial differences in survival and to refine clinical algorithms for CRC treatment and early detection,” the authors wrote.

This study was supported by funding from the Cancer Biology Graduate Program at Wayne State University, the National Cancer Institute, and an Epidemiology Core and National Institutes of Health Center grant. The authors did not have any disclosures to report.

jcraig@frontlinemedcom.com

Racial disparities in survival rates among young patients exist for those diagnosed with both early-stage and late-state colorectal cancer (CRC), investigators have found.

A review of Surveillance, Epidemiology, and End Results (SEER) data on young-onset CRC incidence and mortality rates showed that overall and cancer-specific survival rates were significantly lower for non-Hispanic black (NHB) patients when compared with non-Hispanic white (NHW) and Hispanic patients, reported Dr. Andreana Holowatyi of Wayne State University, Detroit, and her associates.

“Study of patients with young-onset CRC offers an opportunity to examine differences in cancer-specific survival by race, minimizing the potential impact that routine CRC screening among individuals 50 years of age and older might have on CRC-related outcomes,” the authors wrote (J Clin Oncol. 2016 May 2. doi: 10.1200/JCO.2015.65.0994).

The authors reviewed SEER data on 28,145 men and women aged 20-49 years at the time of CRC diagnosis. The sample included 19,497 NHWs, 4,384 NHBs, and 4,264 Hispanic patients and excluded patients who were diagnosed with cancer upon autopsy, patients who survived less than 2 months post diagnosis, and patients who were diagnosed with histopathologic subtypes other than adenocarcinoma. Patients diagnosed with any other type of cancer prior to their CRC diagnoses were excluded from survival analysis.

Stratification of patients by tumor stage and site showed that NHB patients were at an increased risk of cancer-specific death from colon cancers diagnosed at every stage, with the greatest differences observed among those with stage II cancers (hazard ratio, 1.69; 95% confidence interval, 1.33-2.14; P less than .001).

NHB patients were more likely to have CRCs of the proximal colon, compared with NHW and Hispanic patients (39.9% vs. 30.3% vs. 30.7% respectively; P less than .001) and more likely to have more advanced stage cancer. In addition, the NHB subpopulation included a significantly higher percentage of women, compared with NHW and Hispanic patients (50.3% vs. 45.8% vs. 46.3%, P less than .001).

In Cox proportional hazard models controlling for age, sex, race, county-level poverty, tumor stage, surgical intervention, and radiation therapy, Dr. Holowatyi and her associates found that NHB patients had significantly higher risk of cancer-specific death in colon cancers (HR, 1.35; 95% CI 1.26 to 1.45; P less than .001) and rectum/rectosigmoid junction cancers (HR, 1.51; 95% CI 1.37 to 1.68; P less than .001), compared with NHWs. Similar findings were reported for risk of overall death.

“Further studies of the clinical and molecular characteristics of young-onset CRCs are needed to explore potential tumor/treatment interactions associated with racial differences in survival and to refine clinical algorithms for CRC treatment and early detection,” the authors wrote.

This study was supported by funding from the Cancer Biology Graduate Program at Wayne State University, the National Cancer Institute, and an Epidemiology Core and National Institutes of Health Center grant. The authors did not have any disclosures to report.

jcraig@frontlinemedcom.com

References

References

Publications
Publications
Topics
Article Type
Display Headline
Racial disparities found in early-stage, young-onset CRC survival
Display Headline
Racial disparities found in early-stage, young-onset CRC survival
Click for Credit Status
Active
Article Source

FROM THE JOURNAL OF CLINICAL ONCOLOGY

PURLs Copyright

Inside the Article

Vitals

Key clinical point: Among those with young-onset CRC, non-Hispanic black patients have lower survival rates than do non-Hispanic white and Hispanic patients.

Major finding: Non-Hispanic black patients had significantly higher risk of cancer-specific death in colon cancer (HR, 1.35; 95% CI, 1.26 to 1.45; P less than .001) and rectum/rectosigmoid junction cancers (HR, 1.51; 95% CI, 1.37 to 1.68; P less than .001).

Data source: A retrospective study of SEER incidence and mortality data on 28,145 patients with young-onset colorectal cancer.

Disclosures: This study was supported by funding from the Cancer Biology Graduate Program at Wayne State University, the National Cancer Institute, and an Epidemiology Core and National Institutes of Health Center grant. None of the authors reported having any financial disclosures.