Thursday, November 9, 2023

IMFINZI (durvalumab) plus bevacizumab met primary endpoint for progression-free survival in liver cancer eligible for embolization in EMERALD-1 Phase III trial

Results from the EMERALD-1 Phase III trial have showcased positive high-level outcomes for AstraZeneca's IMFINZI (durvalumab) when combined with transarterial chemoembolization (TACE) and bevacizumab. 

This combination exhibited a statistically significant and clinically meaningful improvement in the progression-free survival (PFS) primary endpoint compared to TACE alone in patients with hepatocellular carcinoma (HCC) eligible for embolization.

The trial is ongoing, focusing on the secondary endpoint of overall survival (OS).

Hepatocellular carcinoma is the most prevalent form of liver cancer, ranking as the third-leading cause of cancer-related deaths, with approximately 900,000 new cases diagnosed annually worldwide. 

About 20-30% of patients are suitable for embolization, a procedure that restricts blood supply to the tumor while allowing the delivery of chemotherapy or radiation therapy directly to the liver. Despite being the standard of care, most patients undergoing embolization face rapid disease progression or recurrence.

Dr. Riccardo Lencioni, the principal investigator in the trial, emphasized that patients eligible for embolization often experience high rates of progression or recurrence without the opportunity for early intervention using effective systemic therapy.

The results for durvalumab plus bevacizumab suggest a potential shift in treating this challenging disease, demonstrating, for the first time, that adding an immunotherapy combination to TACE significantly enhances progression-free survival.

Susan Galbraith, Executive Vice President, Oncology R&D at AstraZeneca, expressed optimism about the potential of IMFINZI-based treatment in EMERALD-1, anticipating a groundbreaking application of immunotherapy in earlier stages of liver cancer. 

The company looks forward to discussing these findings with regulatory authorities and monitoring the maturation of survival data over time to bring this innovative treatment option to patients.

The safety profiles for IMFINZI and TACE plus bevacizumab were consistent with the known profiles of each medicine, and there were no new safety findings. The detailed data will be presented at an upcoming medical meeting and shared with regulatory authorities.

AstraZeneca is actively engaged in a comprehensive clinical development program to further evaluate IMFINZI across various gastrointestinal cancer settings. 

This includes combinations with bevacizumab in adjuvant HCC (EMERALD-2) and with IMJUDO (tremelimumab-actl), lenvatinib, and TACE in embolization-eligible HCC (EMERALD-3).

Bristol Myers Squibb and Tempus enter into collaboration for AI approaches

 

Tempus, a prominent player in the realm of artificial intelligence and precision medicine, has unveiled a strategic and multi-year research collaboration with Bristol Myers Squibb. 

Together, these two entities are set to embark on a journey aimed at identifying and validating new targets swiftly and with heightened confidence. This process will leverage multimodal datasets, computational methodologies, and patient-derived disease models, particularly in the realm of specific cancer disease areas.

The collaborative efforts of the Tempus and Bristol Myers Squibb teams will initially be concentrated on the crucial task of pinpointing fresh drug targets.

The primary objective is to broaden the spectrum of options for patients who lack effective therapies or have become refractory to existing treatments. This joint endeavor will unfold in a highly collaborative manner, with both teams delving into Tempus' expansive multimodal database to analyze disease cohorts. 

Employing systems biology approaches, they aim to unearth novel targets that can be rapidly and iteratively tested using Tempus' repository of patient-derived organoids.

Tempus, recognized as a technology powerhouse, is at the forefront of advancing precision medicine through the practical application of artificial intelligence in the healthcare domain. 

Boasting one of the world's largest libraries of multimodal data and an operating system that renders this data accessible and practical, Tempus delivers AI-enabled precision medicine solutions to physicians. This not only facilitates personalized patient care but also catalyzes the discovery, development, and delivery of optimal therapeutics.

The overarching goal is to ensure that each patient reaps the benefits of treatments informed by the experiences of those who came before them. 

Tempus achieves this by furnishing physicians with tools that evolve and learn as the company accumulates more data, thereby contributing to a continuous cycle of improvement and innovation in healthcare.

Tuesday, October 17, 2023

TAGRISSO Plus Chemotherapy Granted Priority Review in the US for Patients with EGFR-Mutated Advanced Lung Cancer

 AstraZeneca's supplemental New Drug Application (sNDA) for TAGRISSO (osimertinib) combined with chemotherapy has been accepted and granted Priority Review by the US Food and Drug Administration (FDA).

This application aims to treat adult patients with locally advanced or metastatic epidermal growth factor receptor-mutated (EGFRm) non-small cell lung cancer (NSCLC). The Food and Drug Administration (FDA) grants Priority Review to applications for medicines that have the potential to offer significant improvements over existing options. 

This can be demonstrated through safety or efficacy enhancements, prevention of serious conditions, or improved patient compliance. The anticipated FDA action date for their regulatory decision, known as the Prescription Drug User Fee Act date, is expected during the first quarter of 2024. 

 Each year, approximately 2.2 million people diagnosed with lung cancer globally Out of these, about 70% are diagnosed with advanced NSCLC. In the US and Europe, approximately 10-15% of NSCLC patients 

The basis for the sNDA lies in data from the FLAURA2 Phase III trial, which was presented at the International Association for the Study of Lung Cancer (IASLC) 2023 World Conference on Lung Cancer (WCLC) in a Presidential Symposium. 

In this trial, combining TAGRISSO with chemotherapy demonstrated a 38% reduction in the risk of disease progression or death Median progression-free survival (PFS) was extended by 8.8 months according to investigator assessment. 

Results from blinded independent central review showed a 9.5-month extension in median PFS. The combination showed a clinically meaningful PFS benefit across various subgroups, including patients with central nervous system metastasis 

The trial continues to assess overall survival (OS) as a key secondary endpoint. Regarding safety, the combination of TAGRISSO plus chemotherapy had a generally manageable profile consistent with the established profiles of the individual medicines. 

Adverse event rates were higher in the combination arm due to known chemotherapy-related adverse events. 

In August 2023, TAGRISSO in combination with chemotherapy received Breakthrough Therapy Designation by the FDA for the 1st-line treatment of adult patients with locally advanced or metastatic EGFRm NSCLC. TAGRISSO is already approved as monotherapy in over 100 countries, including the US, EU, China, and Japan, for various indications related to EGFRm NSCLC

Cimeio Therapeutics and University of Pennsylvania Collaborate on CD45 CAR T Cell Therapy for Blood and Bone Marrow Cancers


Cimeio Therapeutics, a key player in the field of innovative therapeutics, has made a significant announcement regarding a preclinical research collaboration with the University of Pennsylvania. 

This collaboration, spearheaded by Saar Gill, M.D., Ph.D., an esteemed associate professor of medicine and a researcher at the University’s Center for Cellular Immunotherapies, aims to delve into the realm of universal immunotherapy. 

The objective is to create a potential treatment modality for various types of blood and bone marrow cancers. The collaboration capitalizes on the unique strengths of both entities, leveraging Cimeio Therapeutics’ proprietary epitope-editing and CD45-targeting technologies in tandem with Penn Medicine’s epitope-editing and CAR T cell technology. 

The ultimate goal is to design a revolutionary CD45-targeting CAR T cell therapy coupled with epitope-edited hematopoietic stem cells (HSCs). CD45, being prominently present on the surface of a wide array of blood cells, is a pivotal receptor that holds promise for the development of a universal CAR T cell therapy tailored for blood cancers. 

Recent breakthroughs in epitope editing have shown promise in effectively editing CD45, a receptor ubiquitously expressed on the surface of both healthy HSCs and numerous blood and bone marrow cancers. 

This collaboration builds upon the research and findings that have demonstrated the potential to shield healthy HSCs while making malignant cells susceptible to a CD45-targeting therapy, presenting a promising avenue for drug development. In addition to this groundbreaking research, Cimeio Therapeutics is advancing its efforts in the field by also working on the development of a CD45-directed antibody-drug conjugate. 

The company has a strategic vision to develop this therapy utilizing CD45 epitope shielded cells, not only for blood cancers but also for autoimmune diseases and various other indications.

Sunday, October 15, 2023

Pfizer's VELSIPITY Granted FDA Approval for Moderate to Severe Active Ulcerative Colitis in Adults

 

Pfizer's VELSIPITY Granted FDA Approval for Moderate to Severe Active Ulcerative Colitis in Adults

Pfizer has received the green light from the U.S. Food and Drug Administration (FDA) for VELSIPITY, also known as etrasimod, an oral medication that is taken once daily and selectively modulates the sphingosine-1-phosphate (S1P) receptor.

 This approval is specifically for adults who are dealing with moderately to severely active ulcerative colitis (UC), a chronic and often disabling condition affecting an estimated 1.25 million individuals in the United States. 

The recommended dose for 2 mg Symptoms of UC can be distressing and encompass chronic diarrhea with blood and mucus, abdominal pain, and a compelling sense of urgency, and the impact goes beyond the physical aspect due to the chronic and unpredictable nature of the symptoms. 

 The FDA approval was grounded on the promising outcomes of the ELEVATE UC Phase 3 registrational program, specifically the ELEVATE UC 52 and ELEVATE UC 12 trials. These trials evaluated the safety and efficacy of VELSIPITY 2 mg once daily in inducing clinical remission among UC patients who had previously failed or were intolerant to at least one conventional treatment, biologic, or Janus kinase (JAK) inhibitor therapy. Notably, a significant proportion of patients in these trials were treatment-naïve to biologic or JAK inhibitor therapy. 

The studies achieved all primary and key secondary efficacy endpoints, while maintaining a safety profile consistent with previous VELSIPITY studies. 

 In the ELEVATE UC 52 trial, patients treated with VELSIPITY demonstrated a substantial increase in clinical remission rates compared to those on placebo at both week 12 and week 52. at both week 12 and week 52 Similarly, in the ELEVATE UC 12 trial, a noteworthy proportion of patients on VELSIPITY achieved clinical remission compared to the placebo group at week 12. These findings, along with positive outcomes in key secondary endpoints like endoscopic improvement and mucosal healing, substantiated the efficacy of VELSIPITY in treating UC. VELSIPITY represents a significant advancement as a once-daily, oral medication that selectively binds with specific S1P receptor subtypes. 

Regulatory applications for VELSIPITY's approval in treating ulcerative colitis have been submitted in various countries worldwide, including Canada, Australia, Mexico, Russia, Switzerland, and Singapore. The European Medicines Agency (EMA) has accepted the Marketing Authorization Application (MAA) for VELSIPITY, and a decision from EMA is expected in the early months of 2024. 

The pivotal ELEVATE UC 52 trial was designed with a 12-week induction period followed by a 40-week maintenance period, employing a randomized, double-blind, placebo-controlled approach. The primary goal was to assess the safety and efficacy of etrasimod 2 mg once daily on clinical remission after both 12 and 52 weeks. The trial demonstrated significant improvements in primary and key secondary endpoints, including endoscopic improvement and mucosal healing. 

 Similarly, the ELEVATE UC 12 trial, which was also randomized and placebo-controlled, focused on assessing the efficacy and safety of etrasimod 2 mg once daily in subjects with moderately to severely active UC. The trial achieved its primary objective, showcasing a noteworthy proportion of patients achieving clinical remission with etrasimod compared to the placebo group at week 12 at week 12 

Importantly, the safety profile of VELSIPITY was consistent across both trials, with common adverse reactions being manageable and not unexpected. These trials affirm that initiating VELSIPITY treatment does not necessitate a complex up-titration regimen, enhancing its practicality and ease of use for patients

Thursday, October 12, 2023

US FDA Approves Encorafenib & Binimetinib for Metastatic Lung Cancer Patients with BRAF V600E Mutation


 

Bayer Unveils Pioneering Cell Therapy Hub, Revolutionizing Global Regenerative Medicine


 

FDA approves encorafenib with binimetinib for metastatic non-small cell lung cancer with a BRAF V600E mutation

On October 11, 2023, the Food and Drug Administration (FDA) granted approval for the use of encorafenib (Braftovi, developed by Array BioPharma Inc., a subsidiary of Pfizer) in combination with binimetinib (Mektovi, also by Array BioPharma Inc.) for adult patients dealing with metastatic non-small cell lung cancer (NSCLC) characterized by a BRAF V600E mutation, as confirmed by an FDA-endorsed diagnostic test.

In conjunction with this approval, the FDA also sanctioned the utilization of FoundationOne CDx (tissue) and FoundationOne Liquid CDx (plasma) as companion diagnostics for encorafenib with binimetinib. It was emphasized that in cases where no mutation is detected in a plasma sample, a test should be conducted on tumor tissue.

The assessment of efficacy encompassed 98 patients grappling with metastatic NSCLC and possessing the BRAF V600E mutation. These patients were enrolled in the PHAROS study (NCT03915951), which was an open-label, multicenter, single-arm study. Notably, patients were not allowed to have had prior exposure to BRAF or MEK inhibitors. Patients received a regimen of encorafenib and binimetinib until either their disease progressed or they experienced unacceptable levels of toxicity.

The primary efficacy outcomes were gauged by the objective response rate (ORR) per RECIST v1.1 and the duration of response (DoR), as evaluated by an independent review committee. Out of 59 patients who were new to treatment, an ORR of 75% was observed (95% CI: 62, 85), with an indeterminate median DoR (NE) (95% CI: 23.1, NE). Among the 39 patients who had previously undergone treatment, an ORR of 46% was noted (95% CI: 30, 63) along with a median DoR of 16.7 months (95% CI: 7.4, NE).

The adverse reactions most frequently reported (≥25%) were fatigue, nausea, diarrhea, musculoskeletal pain, vomiting, abdominal pain, visual impairment, constipation, dyspnea, rash, and cough.

In terms of recommended doses for NSCLC patients positive for the BRAF V600E mutation, the guidance stipulates an oral administration of encorafenib at 450 mg once daily and binimetinib at 45 mg orally twice daily.

This comprehensive review of the application incorporated the Assessment Aid, a voluntary submission from the applicant intended to streamline the FDA's evaluation. Additionally, the application was granted orphan drug designation, underlining the significance and unique status of this therapeutic approach.

Friday, May 5, 2023

Cystic Fibrosis

Cystic fibrosis (CF) is an autosomal recessive genetic disorder that can shorten life expectancy. It is characterized by pulmonary symptoms, including obstructive lung disease that worsens over time, sinusitis, malabsorption due to insufficient pancreatic exocrine function leading to malnutrition, liver disease such as biliary cirrhosis, and CF-related diabetes mellitus (CFRD).

CF was a uniformly fatal disease in childhood when first described in 1938, but today the predicted median survival is 44.4 years.

Over 50% of people with CF are now 18 years and older, evolving the disease from exclusively a childhood illness to one that affects adults.

Newborn screening, improved therapies for lung health and nutrition, and aggressive treatment of respiratory infections and lung transplantation have improved survival rates.

Newer therapies targeting the genetic defect causing the disease, expanding to more age and genetic variants, promise continued improvement in quality of life and overall health.

Epidemiology

CF is a common genetic disorder in Caucasians, with an incidence of 1:3,200. The incidence varies by race/ethnicity, with lower rates in Hispanic, African, and Asian populations.

It is estimated that 1 in 35 Americans are CF carriers, and there are approximately 30,000 affected individuals in the US, with the highest prevalence in North America, Europe, and Australia.

Around 1,000 new cases are diagnosed annually in the US, and the incidence is equal in males and females. Since 2010, all US states screen newborns for CF, resulting in 60% of new diagnoses being made this way

 Pathogenesis

CF is a multisystem disorder caused by genetic variants in the CFTR gene that lead to malfunctioning chloride channels, resulting in the formation of thick and sticky mucus and various health complications. CF also results in abnormal chloride channel function in sweat glands, leading to excessive salt loss.

CF is an autosomal recessive disorder that requires individuals to inherit two deleterious CFTR variants. There are more than 2,000 different CFTR variants reported that are classified into six distinct groups that reflect abnormalities of CFTR protein synthesis, structure and function, with F508del being the most common CF-causing variant.

44.2% of individuals with CF are homozygous for F508del, and the specific CFTR variants an individual carries determine phenotypic severity and organ involvement.

DIAGNOSIS

The CFF published consensus guidelines in 2017 establishing that a diagnosis of CF can be made if an individual has a clinical presentation consistent with the disease i.e. (1) a positive newborn screening; (2) clinical features consistent with CF (the presence of characteristic phenotypes such as of chronic, recurrent sinus and pulmonary disease, nutritional and gastrointestinal abnormalities, urogenital abnormalities in males (e.g., absence of the vas deferens), and/or salt depletion syndromes, or (3) a positive family history of CF and evidence of CFTR dysfunction (e.g., sweat chloride concentration ≥60mmol/L).

 

CF Management

 Therapies to Maintain Optimal Lung Health

CF is marked by the thickening of airway secretions that cause chronic blockage of mucus, inflammation, and recurring infections that lead to persistent harm to the airways and lung tissue damage (bronchiectasis). A chronic cough and production of sputum are symptoms commonly associated with the condition. The management of respiratory symptoms is directed towards preserving lung function and preventing the occurrence of bronchiectasis and destruction of lung parenchyma.

Clearance of Airway Secretions

Airway clearance therapy (ACT) is crucial for maintaining lung health in CF patients, as it helps to remove airway mucus, decreasing bacterial load, and improving gas exchange. Twice daily ACT is recommended for all CF patients, and frequency is increased during pulmonary exacerbations. Common ACT modalities include manual percussion, positive expiratory pressure devices, and high-frequency chest wall oscillation. Exercise is encouraged but not a substitute for ACT. Nebulized agents such as rhDNase and hypertonic saline are often used to thin CF mucus during ACT, but the choice of therapy should be personalized.

Chronic Airway Infections

Aggressive management of chronic airway infections in CF is critical for preserving lung function. It involves frequent respiratory cultures, surveillance for specific pathogens, and eradication therapy when necessary. Nebulized antibiotics can be used as suppressive therapy to prevent colonization and reduce the risk of antibiotic resistance. Other organisms that can impact lung disease are also monitored. CF patients are prone to developing allergic bronchopulmonary aspergillosis, which requires corticosteroid therapy.

Chronic Airway Inflammation

Cystic fibrosis lung disease is caused by infection and inflammation. Corticosteroids are not routinely recommended for CF unless used for another inflammatory comorbidity. Chronic airway inflammation is managed with high-dose ibuprofen or azithromycin. Although ibuprofen has proven benefits, its use is limited by the risk of gastrointestinal bleeding and the need for monitoring serum levels. Azithromycin is typically given orally three times per week and has been shown to improve lung function and reduce PEx. However, screening for mycobacterial infections is recommended before initiating treatment to avoid the development of resistance.

Therapies to Maintain Optimal Nutritional Status

Poor growth is an early manifestation of CF due to decreased intake, malabsorption, and increased metabolic demands. Malnutrition increases morbidity and mortality in CF. The CFF recommends achieving weight-for-length at or above the 50th percentile by 2 years old, and maintaining a body mass index at or above the 50th percentile in children and adolescents aged 2-20 years. Enteral tube feeding should be discussed as an option to optimize nutritional status. Infants should receive breastmilk if possible or standard infant formula, supplemented with table salt. Fecal elastase should be measured to assess pancreatic function, as most individuals with CF have pancreatic insufficiency, leading to malabsorption, bulky stools, malnutrition, and failure to thrive.

 

Pancreatic Enzyme Replacement Therapy

Pancreatic enzyme replacement therapy (PERT) is recommended for those with pancreatic insufficiency or symptoms of malabsorption. The recommended dosage is 2,000-2,500 units/kg of lipase per meal, up to a maximum of 10,000 units/kg/day. Immobilized lipase cartridges may be used for those on continuous enteral tube feeding. Exceeding recommended dosages can result in fibrosing colonopathy. Optimized PERT can help individuals with CF achieve age-appropriate growth.

 

Fat-Soluble Vitamin Replacement Therapy

Pancreatic insufficiency in CF can lead to malabsorption of fat-soluble vitamins A, D, E, and K. Vitamin deficiencies can cause various health problems, including night blindness, ocular xerosis, rickets, osteopenia, osteoporosis, peripheral neuropathy, myopathy, hemolysis, and coagulopathy. To prevent these complications, CF patients should receive supplemental vitamins, and their serum vitamin levels should be monitored annually. The recommended goal for serum 25-hydroxyvitamin D level is at least 30 ng/ml.

 

CFTR Modulator Therapies

CFTR modulators are a major advancement in CF treatment as they directly target the underlying defect in the CFTR protein. Ivacaftor, the first modulator therapy, is a potentiator that helps improve chloride flow through the CFTR protein at the cell surface. Correctors, such as lumacaftor and tezacaftor, help the CFTR protein to form correctly and move to the cell surface. When added to potentiators, correctors improve the amount of protein that reaches the cell surface. Triple-combination therapy, elexacaftor/tezacaftor/ivacaftor (Trikafta), was approved in 2019 and has shown to be highly effective in improving key measures of disease, including lung function and quality of life. Amplifiers are currently under development and are expected to increase the amount of CFTR protein made by a cell.

 

Thursday, May 4, 2023

Vertex Announces U.S. FDA Approval for KALYDECO (ivacaftor) to Treat Eligible Infants With CF Ages 1 Month and Older

On 3 May 2023, Vertex Pharmaceuticals have announced the U.S.Food and Drug Administration (FDA) approved KALYDECO (ivacaftor) for use in children with cystic fibrosis (CF) ages 1 month to less than four months old who have at least one mutation in their cystic fibrosis transmembrane conductance regulator (CFTR) gene that is responsive to KALYDECO based on clinical and/or in vitro assay data.

KALYDECO is already approved in the U.S. and EU for the treatment of CF in patients ages four months and older.
The approval was supported by a cohort in the Phase 3, 24-week, open-label study to evaluate the safety, pharmacokinetics and pharmacodynamics of ivacaftor in subjects with CF who are less than 24 months of age and have an ivacaftor-responsive CFTR mutation.
This cohort demonstrated a safety profile similar to that observed in older children and adults.

FDA programs to expedite drug development

 The FDA has several programs aimed at streamlining and accelerating the development and review of new drugs for the treatment of serious or life-threatening conditions, particularly where there is an unmet medical need.

These expedited programs ensure that treatments for such conditions are available as soon as possible, provided that the benefits of the therapy outweigh its risks, taking into account the severity of the condition and availability of other treatment options.

The programs include:

breakthrough therapy designation,

fast track designation,

accelerated approval, and

priority review

 

Fast Track Designation

Fast track designation is a program designed to accelerate the development and review of drugs that address serious medical conditions with an unmet need.

The designation may be granted based on preclinical data. Sponsors of drugs that receive fast track designation can expect more frequent interactions with the FDA during the drug development process.

Additionally, drugs that have received fast track designation can benefit from rolling review, allowing for the submission of completed sections of the New Drug Application (NDA) on a rolling basis rather than waiting for the entire application to be completed before submission.

 

Accelerated Approval

Accelerated approval program expedites the development and approval of drugs for serious or life-threatening conditions

Allows approval of drugs that show an effect on surrogate or clinical endpoints that can predict clinical benefit

Useful when disease course is lengthy and takes time to measure clinical benefit

Post-marketing trials may be required to confirm clinical benefit, and approval may be withdrawn if trials fail to verify predicted benefit

 

Priority Review

The Program applies to new molecular entity NDAs and original BLAs submitted from Oct 1, 2012, through Sep 30, 2017, including resubmissions following Refuse-to-File actions

For applications filed under the Program, the PDUFA review clock begins after the 60-day filing review period that starts from FDA receipt of the original submission

Any drug, including those with other designations like fast track or breakthrough therapy, can be granted priority review if relevant criteria are met

 

Differences between the criteria for breakthrough therapy designation and fast track designation?

Breakthrough therapy and fast track designation programs aim to accelerate drug development and review for serious or life-threatening conditions.

A breakthrough therapy designation requires preliminary clinical evidence that the drug can substantially improve a significant clinical endpoint over available therapies, while a fast track designation requires nonclinical or clinical data demonstrating the potential to address unmet medical needs for the serious condition.

Wednesday, May 3, 2023

FDA Approves First Respiratory Syncytial Virus (RSV) Vaccine - Arexvy from GSK

On 3 May 2023, the U.S. Food and Drug Administration approved Arexvy, the first respiratory syncytial virus (RSV) vaccine approved for use in the United States.

Arexvy is approved for the prevention of lower respiratory tract disease caused by RSV in individuals 60 years of age and older.

RSV is a highly contagious virus that causes infections of the lungs and breathing passages in individuals of all age groups.

RSV circulation is seasonal, typically starting during the fall and peaking in the winter.

In older adults, RSV is a common cause of lower respiratory tract disease (LRTD), which affects the lungs and can cause life-threatening pneumonia and bronchiolitis (swelling of the small airway passages in the lungs).

According to the U.S.Centers for Disease Control and Prevention, each year in the U.S., RSV leads to approximately 60,000-120,000 hospitalizations and 6,000-10,000 deaths among adults 65 years of age and older.

The safety and effectiveness of Arexvy is based on the FDA’s analysis of data from an ongoing, randomized, placebo-controlled clinical study conducted in the U.S. and internationally in individuals 60 years of age and older.

The main clinical study of Arexvy was designed to assess the safety and effectiveness of a single dose administered to individuals 60 years of age and older.

Participants will remain in the study through three RSV seasons to assess the duration of effectiveness and the safety and effectiveness of repeat vaccination.

Data for a single dose of Arexvy from the first RSV season of the study were available for the FDA’s analysis.

In this study, approximately 12,500 participants have received Arexvy and 12,500 participants have received a placebo.

Among the participants who have received Arexvy and the participants who have received a placebo, the vaccine significantly reduced the risk of developing RSV-associated LRTD by 82.6% and reduced the risk of developing severe RSV-associated LRTD by 94.1%.

Among a subset of these clinical trial participants, the most commonly reported side effects by individuals who received Arexvy were injection site pain, fatigue, muscle pain, headache and joint stiffness/pain.

Among all clinical trial participants, atrial fibrillation within 30 days of vaccination was reported in 10 participants who received Arexvy and 4 participants who received placebo.

The FDA is requiring the company to conduct a postmarketing study to assess the signals of serious risks for Guillain-Barré syndrome and ADEM.

In addition, although not an FDA requirement, the company has committed to assess atrial fibrillation in the postmarketing study

 

Bristol Myers Squibb Receives European Commission Approval for Breyanzi (lisocabtagene maraleucel) for Relapsed or Refractory Large B-cell Lymphoma After One Prior Therapy

Bristol Myers Squibb has received approval from the European Commission for Breyanzi (lisocabtagene maraleucel; liso-cel), a chimeric antigen receptor (CAR) T cell therapy directed towards CD19, for the treatment of adult patients suffering from diffuse large B-cell lymphoma (DLBCL), high grade B-cell lymphoma (HGBCL), primary mediastinal large B-cell lymphoma (PMBCL), and follicular lymphoma grade 3B (FL3B). The treatment is intended for patients who have relapsed within 12 months of completion of, or have shown refractory response to, first-line chemoimmunotherapy.

Approval of Breyanzi was granted after the positive results obtained from the Phase 3 TRANSFORM trial, where the therapy exhibited significant and clinically relevant advancements in the primary endpoint of event-free survival (EFS), and in key secondary endpoints such as complete responses (CR) and progression-free survival (PFS) when compared to standard treatment (which involves salvage immunochemotherapy followed by high-dose chemotherapy and hematopoietic stem cell transplant [HSCT]).  The treatment demonstrated a manageable and well-established safety profile.

In the TRANSFORM study, Breyanzi more than quadrupled median EFS compared to standard therapy (10.1 months vs. 2.3 months) at the time of prespecified interim analysis with a median follow-up of 6.2 months. Results of the primary analysis, with a median follow-up of 17.5 months were consistent with the interim analysis, with median EFS not reached for Breyanzi (95% CI: 9.5-NR) vs. 2.4 months for standard therapy (95% CI: 2.2-4.9).

With Breyanzi, the majority (73.9%) of patients achieved a CR compared to less than half (43.5%) of those who were treated with standard therapy. Median PFS was not reached (95% CI: 12.6-NR) with Breyanzi vs. 6.2 months (95% CI: 4.3-8.6) with standard therapy

The safety profile of Breyanzi is well-established, and in the TRANSFORM study, occurrences of cytokine release syndrome (CRS) and neurologic events were generally low-grade, and mostly resolved quickly with standard protocols and without the use of prophylactic steroids.