logo
Global phase III trials demonstrate that nerandomilast slowed lung function decline in IPF and PPF, with similar discontinuation rates to placebo

Global phase III trials demonstrate that nerandomilast slowed lung function decline in IPF and PPF, with similar discontinuation rates to placebo

Yahoo19-05-2025

Nerandomilast, an investigational agent, met the primary endpoint of both phase III trials, FIBRONEER™-IPF and FIBRONEER™-ILD, significantly reducing the decline in forced vital capacity (FVC) [mL] by absolute change from baseline at week 52, compared to placebo.1,2
Nerandomilast's safety and tolerability profile was consistent across both trials, with similar rates of permanent treatment discontinuation to placebo.1,2
In both trials the composite key secondary endpoint (time to first acute IPF/ILD exacerbation, first hospitalization for respiratory cause, or death) was not met. However, in the FIBRONEER™-ILD trial, death occurred in a numerically smaller proportion of patients treated with nerandomilast than placebo.1,2
New drug applications for nerandomilast in idiopathic pulmonary fibrosis (IPF) and progressive pulmonary fibrosis (PPF) have been submitted in the US, China and the EU, with other geographies to follow.
Boehringer Ingelheim announced today detailed findings from the Phase III FIBRONEER™-IPF and FIBRONEER™-ILD trials. These studies evaluated nerandomilast, an investigational oral, preferential inhibitor of phosphodiesterase 4B (PDE4B), in patients with idiopathic pulmonary fibrosis (IPF) and progressive pulmonary fibrosis (PPF), respectively, with and without background antifibrotic therapy. Results were published in the New England Journal of Medicine and also presented as a late-breaking abstract at the American Thoracic Society (ATS) 2025 International Conference.
Nerandomilast is an investigational agent and has not been approved for use; its efficacy and safety has not been established.
Both trials met the primary endpoint at both doses, 9 mg and 18 mg, as measured by a reduction in the absolute change from baseline in forced vital capacity (FVC) [mL] decline at week 52 versus placebo.1,2 FVC is a measure of lung function.3
"After several challenges in the scientific community to bring forward new clinical data, IPF and PPF continue to take a devastating toll on patients,' said Toby Maher, M.D., Ph.D., Professor of Clinical Medicine, Keck School of Medicine, USC Los Angeles. 'Having two phase III trials meet the primary endpoint is a major breakthrough for the scientific community, highlighting nerandomilast's potential to have a meaningful impact on patients' unmet needs, being studied as mono therapy or in combination with current treatments.'
Across both trials, similar rates of permanent treatment discontinuation to placebo were observed: In FIBRONEER™-IPF, adverse events led to permanent discontinuation of the trial regimen in 14.0% of the nerandomilast 18 mg group, 11.7% of the nerandomilast 9 mg group, and 10.7% of the placebo group.1,2 In FIBRONEER™-ILD, adverse events led to permanent discontinuation of the trial regimen in 10.0% of the nerandomilast 18 mg group, 8.1% in the nerandomilast 9 mg group, and 10.2% in the placebo group.2 In both trials there were no imbalances between the nerandomilast and placebo groups with respect to adverse events of special interest such as vasculitis, depression, suicidality, or drug induced liver injury.1,2
'Idiopathic pulmonary fibrosis and progressive pulmonary fibrosis are devastating conditions, with one in two people dying within five years of IPF diagnosis. Despite this stark reality, ongoing research may provide new possibilities for patients, as there remains a need for additional therapies,' said Shashank Deshpande, Head of Human Pharma and Member of the Board of Managing Directors at Boehringer Ingelheim. 'The latest efficacy, safety and tolerability data on nerandomilast points to its potential in addressing the needs of those impacted by IPF and PPF.'
FIBRONEER™-IPF (NCT05321069) was a Phase III, double-blind, randomized, placebo-controlled trial evaluating the efficacy and safety of nerandomilast over at least 52 weeks in patients with IPF. A total of 1,177 patients across 36 countries were randomly assigned 1:1:1 to receive nerandomilast 9 mg twice-daily (n=392), nerandomilast 18 mg twice-daily (n=392), or placebo twice-daily (n=393). Randomization was stratified by use of background antifibrotic therapy, with 77.7% receiving nintedanib or pirfenidone at enrollment.
Key efficacy results at 52 weeks, as published in the New England Journal of Medicine, include:
Placebo
Nerandomilast 9 mg twice-daily
Nerandomilast 18 mg twice-daily
Primary Endpoint (adjusted mean change from baseline in FVC)
All patients
−183.5 mL (n=391)
(95% confidence interval [CI]: -210.9 to -156.1)
−138.6 mL (n=390)
(95% CI: -165.6 to -111.6)
−114.7 mL (n=392)
(95% CI: -141.8 to - 87.5)
Subgroups of Primary Endpoint (adjusted mean change from baseline in FVC)*
*Trial was not powered to show a difference for these subgroups
No background antifibrotic therapy
−148.7 mL (n=87)
−70.4 mL (n=86)
−79.2 mL (n=87)
Background nintedanib
−191.6 mL (n=172)
−130.7 mL (n=184)
−118.5 mL (n=178)
Background pirfenidone
−197.0 mL (n=132)
−201.8 mL (n=120)
−133.7 mL (n=127)The composite key secondary endpoint (time to first acute IPF exacerbation, first hospitalization for respiratory cause, or death) was not met.
The most frequent adverse event was diarrhea, reported in 16.0% of the placebo group, 31.1% of the nerandomilast 9 mg group, and 41.3% of the nerandomilast 18 mg group. The adverse event that most frequently led to discontinuation of the trial regimen was diarrhea, with 1.8% in the nerandomilast 9 mg group, 6.1%, in the nerandomilast 18 mg group, and in 0.5% in the placebo group discontinuing treatment. Other adverse events were balanced across treatment groups, specifically showing no imbalances between the nerandomilast and placebo groups in adverse events of special interest such as vasculitis, depression, suicidality, or drug-induced liver injury. Adverse events led to treatment discontinuation more frequently among patients taking background antifibrotic therapy across treatment groups. Serious adverse events occurred in 33%, 31%, and 30% of patients treated with placebo, nerandomilast 9 mg, and nerandomilast 18 mg, respectively. Adverse events were fatal in 5%, 4%, and 2% of patients treated with placebo, nerandomilast 9 mg, and nerandomilast 18 mg, respectively.
FIBRONEER™-ILD (NCT05321082) was a Phase III, double-blind, randomized, placebo-controlled trial evaluating the efficacy and safety of nerandomilast over at least 52 weeks in patients with PPF. A total of 1,176 patients across 44 countries were randomly assigned 1:1:1 to receive nerandomilast 9 mg twice-daily (n=393), nerandomilast 18 mg twice-daily (n=391), or placebo twice-daily (n=392). Randomization was stratified by use of background antifibrotic therapy, with 43.5% receiving nintedanib.
Key efficacy results at 52 weeks, as published in the New England Journal of Medicine, include:
Placebo
Nerandomilast 9 mg twice-daily
Nerandomilast 18 mg twice-daily
Primary Endpoint (adjusted mean change from baseline in FVC)
All patients
−165.8 mL (n=391)
(95% CI: -190.5 to -141.0)
−84.6 mL (n=390)
(95% CI: -109.6 to -59.7)
−98.6 mL (n=390)
(95% CI: -123.7 to -73.4)
Subgroups of Primary Endpoint (adjusted mean change from baseline in FVC)*
*Trial was not powered to show a difference for these subgroups
No background
−154.1 mL (n=222)
(95% CI: -187.1 to -121.2)
−82.3 mL (n=217)
(95% CI: -115.9 to -48.8)
−95.2 mL (n=220)
(95% CI: -128.6 to -61.9)
Background nintedanib†
−180.9 mL (n=169)
(95% CI: -218.6 to -143.2)
−87.8 mL (n=173)
(95% CI: -125.4 to -50.2)
−102.9 mL (n=170)
(95% CI: -141.2 to -64.5)
†2 patients in this group took pirfenidone rather than nintedanib. These were classified as protocol deviations, but the data from these patients were analyzed as part of the background nintedanib group.
Nerandomilast was not statistically significant for the composite key secondary endpoint (time to first acute exacerbation, hospitalization for a respiratory cause, or death), but there were numerically fewer deaths in both treatment groups: 9 mg group (n=33/8.4%), 18 mg group (n=24/6.1%), versus placebo (n=50/12.8%).
The safety and tolerability profile of nerandomilast was largely consistent with FIBRONEER™-IPF. The most frequent adverse event was diarrhea, reported in 24.7% of the placebo group, 29.5% of the nerandomilast 9 mg twice daily group, and 36.6% of the nerandomilast 18 mg twice daily group over 52 weeks. There were no imbalances between the nerandomilast and placebo groups in adverse events of special interest such as vasculitis, depression, or suicidality. The adverse events that most frequently led to treatment discontinuation were 'condition aggravated' (i.e., worsening of pulmonary fibrosis) which led to discontinuation in 1.5% in the nerandomilast 9 mg group, 1.0%, in the nerandomilast 18 mg group, and in 3.1% in the placebo group, and diarrhea, which led to discontinuation in 1.3%, 2.6%, and 0.5% of these treatment groups, respectively. The incidence of adverse events that led to treatment discontinuation was generally similar among patients taking background nintedanib therapy and patients not taking background nintedanib therapy. Serious adverse events occurred in 35.2%, 31.8%, and 33.2% of patients treated with placebo, nerandomilast 9 mg, and nerandomilast 18 mg, respectively. Adverse events were fatal in 5.1%, 3.6%, and 2.0% of patients treated with placebo, nerandomilast 9 mg, and nerandomilast 18 mg, respectively.
Nerandomilast (BI 1015550) is an investigational orally administered preferential inhibitor of phosphodiesterase 4B (PDE4B) that is being studied as a potential treatment for IPF and PPF.1,2
Nerandomilast was granted Breakthrough Therapy Designation by the U.S. Food and Drug Administration (FDA) for the treatment of IPF in February 2022 and for the treatment of PPF in April 2025. The U.S. FDA recently granted priority review to the New Drug Application (NDA) for nerandomilast in IPF, with an anticipated action date in the fourth quarter of 2025. An NDA for nerandomilast in PPF has also been filed with the FDA. Regulatory submissions for nerandomilast in IPF and PPF are also under review in China and the EU, with filings in other geographies to follow.
IPF is one of the more common progressive fibrosing interstitial lung diseases (ILDs).4 In IPF, the root cause of pulmonary fibrosis is not known. Symptoms of IPF include breathlessness during activity, a dry and persistent cough, fatigue and weakness.5 The disease primarily affects patients over the age of 50 and affects more men than women.5
Patients with certain types of non-IPF fibrosing ILD may also develop a progressive phenotype known as PPF. In PPF, there is often a known underlying disease (e.g. rheumatoid arthritis or systemic sclerosis) cause for the ILD.6 In ILDs other than IPF, PPF is defined by worsening respiratory symptoms, physiological evidence of disease progression and radiological evidence of disease progression.6
The prevalence of IPF and PPF varies by region. It is estimated that up to 3.6 million people could be affected by IPF worldwide and up to 5.6 million by PPF.7,8
Boehringer Ingelheim is a biopharmaceutical company active in both human and animal health. As one of the industry's top investors in Research and Development, the company focuses on developing innovative therapies in areas of high unmet medical need. Independent since its foundation in 1885, Boehringer takes a long-term perspective, embedding sustainability along the entire value chain. More than 54,500 employees serve over 130 markets to build a healthier, more sustainable, and equitable tomorrow. Learn more at http://www.boehringer-ingelheim.com/uk (UK) or www.boehringer-ingelheim.com (rest of world).
1Richeldi L, et al. (2025) Nerandomilast in patients with idiopathic pulmonary fibrosis. In: NEJM. 2025.
2Maher TM, et al. (2025) Nerandomilast in patients with progressive pulmonary fibrosis. In: NEJM. 2025.
3Twisk JWR et al. (1998). Tracking of lung function parameters and the longitudinal relationship with lifestyle. European Respiratory Journal. 12(3):627–634.
4Sauleda J, et al. Idiopathic Pulmonary Fibrosis: Epidemiology, Natural History, Phenotypes. Med Sci (Basel). 2018 Nov 29;6(4):110. doi: 10.3390/medsci6040110. PMID: 30501130; PMCID: PMC6313500.
5European Lung Foundation (2023) IPF - Idiopathic Pulmonary Fibrosis. Accessed April 2025. Available at: https://europeanlung.org/en/information-hub/factsheets/ipf-idiopathic-pulmonary-fibrosis/.
6Podolanczuk AJ, Fernández Peréz ER. Identification, course, and management of progressive pulmonary fibrosis. Am J Manag Care. 2024 Oct;30(7 Suppl):S122-S130. doi: 10.37765/ajmc.2024.89634. PMID: 39495032.
7Maher TM, Bendstrup E, Dron L, Langley J, Smith G, Khalid JM, Patel H, Kreuter M. Global incidence and prevalence of idiopathic pulmonary fibrosis. Respir Res. 2021 Jul 7;22(1):197. doi: 10.1186/s12931-021-01791-z. PMID: 34233665; PMCID: PMC8261998.
8Cottin V, Teague R, Nicholson L, Langham S, Baldwin M. The Burden of Progressive-Fibrosing Interstitial Lung Diseases. Front Med (Lausanne). 2022 Feb 1;9:799912. Doi: 10.3389/fmed.2022.799912. PMID: 35178411; PMCID: PMC8843847. Assuming a world population of 8bn

Orange background

Try Our AI Features

Explore what Daily8 AI can do for you:

Comments

No comments yet...

Related Articles

Vertex Presents Positive Data for Zimislecel in Type 1 Diabetes at the American Diabetes Association 85th Scientific Sessions
Vertex Presents Positive Data for Zimislecel in Type 1 Diabetes at the American Diabetes Association 85th Scientific Sessions

Yahoo

timean hour ago

  • Yahoo

Vertex Presents Positive Data for Zimislecel in Type 1 Diabetes at the American Diabetes Association 85th Scientific Sessions

– Results from the study continue to demonstrate the transformative potential of zimislecel with consistent and durable patient benefit – – All 12 patients with at least one year of follow-up who received a full dose of zimislecel as a single infusion achieved ADA-recommended target HbA1c levels <7% and >70% time-in-range (70-180 mg/dL), and 10/12 patients were insulin free – – Data presented at ADA simultaneously published in the New England Journal of Medicine – – Vertex to host investor webcast tonight, June 20, 2025, at 7:15 p.m. CT / 8:15 p.m. ET – BOSTON, June 20, 2025--(BUSINESS WIRE)--Vertex Pharmaceuticals Incorporated (Nasdaq: VRTX) today announced simultaneous presentation and publication of updated data from the Phase 1/2 portion of the Phase 1/2/3 FORWARD-101 clinical trial of zimislecel (VX-880), an investigational stem cell-derived, fully differentiated islet cell therapy, in people with type 1 diabetes (T1D) with impaired hypoglycemic awareness and severe hypoglycemic events (SHEs). The data were featured in an oral presentation at the American Diabetes Association (ADA) annual conference in Chicago as part of the symposium, "Innovation and Progress in Stem Cell-Derived Islet-Cell Replacement Therapy," from 6:15-6:30 p.m. CT (abstract 2025-A-1921) and published online by the New England Journal of Medicine. The data are from 12 patients who received the full dose of zimislecel as a single infusion and were followed for at least one year, as of October 2024. Results from the study to date continue to demonstrate the transformative potential of zimislecel with consistent and durable patient benefit with longer follow-up. All 12 participants: Demonstrated engraftment with glucose-responsive endogenous C-peptide production, which was durable through one year of follow-up. Achieved the ADA targets of HbA1c <7% and time in range of >70%. Were free of SHEs from day 90 onwards. Had a reduction in exogenous insulin use (mean reduction in daily insulin dose: 92%). 10/12 (83%) no longer required exogenous insulin at Month 12. Achieved the Phase 1/2 primary endpoint of elimination of SHEs with HbA1c <7%. Zimislecel continues to be generally well tolerated. Most adverse events (AEs) were mild or moderate, and there were no serious AEs related to zimislecel treatment. As previously reported, two patient deaths occurred, both unrelated to treatment with zimislecel. The safety profile is generally consistent with the immunosuppressive regimen used in the study, the infusion procedure, and complications from long-standing diabetes. "These data on the first fully differentiated, stem cell-derived, off-the-shelf islet cell therapy continue to be unprecedented. The magnitude, consistency and durability of the results from all 12 patients with more than one year of follow-up reinforce the transformative potential of zimislecel for people living with T1D complicated by severe hypoglycemia," said Carmen Bozic, M.D., Executive Vice President, Global Medicines Development and Medical Affairs, and Chief Medical Officer at Vertex. "We are excited to complete enrollment and dosing in the Phase 1/2/3 Program and look forward to potential regulatory submissions next year." "It's remarkable to see 12 out of 12 patients with baseline HbA1c above 7% and multiple severe hypoglycemic events reach consensus targets for glycemic control by both HbA1c and time in range as well as elimination of severe hypoglycemic events," said Michael R. Rickels, M.D., M.S., Medical Director, Pancreatic Islet Cell Transplant Program, Willard and Rhoda Ware Professor in Diabetes and Metabolic Diseases, Presenting Author and Steering Committee Co-Chair for the zimislecel clinical program, and author on the New England Journal of Medicine paper. "As I think about my patients and the unmet need in the type 1 diabetes community, the results we've seen so far for restoring endogenous insulin secretion with a stem cell-derived islet therapy bring me hope and confidence for a transformative treatment option for individuals with type 1 diabetes in the not-so-distant future." About Type 1 DiabetesT1D results from the autoimmune destruction of insulin-producing beta cells in pancreatic islets. Insulin deficiency results in hyperglycemia and can lead to acute life-threatening complications such as diabetic ketoacidosis. People with T1D are reliant on lifelong treatment with exogenous insulin that requires careful monitoring of blood glucose levels. Even with the availability of advanced exogenous insulin delivery and glucose monitoring systems, people with T1D can have periods of very low and very high blood sugar levels. Exogenous insulin has a narrow therapeutic range and carries an inherent risk of causing low blood sugar levels or hypoglycemic events, which can potentially result in arrhythmias, seizures, coma and even death. Due to the limitations and complexities of exogenous insulin treatment, it can be difficult for people with T1D to achieve and maintain good glucose control. Exposure to prolonged periods of high blood glucose levels, or hyperglycemia, can lead to long-term complications such as nerve damage, kidney disease/failure, eye disease (including vision loss), cardiovascular disease, stroke and even death. HbA1c is a measure of average blood glucose over the most recent ~2-3 months, and the consensus guidance is to maintain an HbA1c of <7% to reduce the risk of long-term complications; only ~1 in 4 people with T1D globally meet this clinical target. Current standards of care do not address the underlying cause of the disease and leave people with T1D susceptible to both hypo- and hyperglycemia and their associated morbidity and mortality. There is no cure for T1D. About ZimislecelZimislecel (VX-880) is an investigational allogeneic stem cell-derived, fully differentiated, insulin-producing islet cell therapy manufactured using proprietary technology. Zimislecel is being evaluated for patients who have T1D with impaired hypoglycemic awareness and severe hypoglycemia. Zimislecel has the potential to restore the body's ability to regulate glucose levels by restoring pancreatic islet cell function, including glucose-responsive insulin production. Zimislecel is delivered by an infusion into the hepatic portal vein and requires chronic immunosuppressive therapy to protect the islet cells from immune rejection. The zimislecel trial has expanded to additional sites that are currently active and enrolling in the U.S., Canada and Europe. Zimislecel has been granted Regenerative Medicine Advanced Therapy (RMAT) and Fast Track designations from the U.S. Food and Drug Administration, Priority Medicines (PRIME) designation from the European Medicines Agency (EMA), and has secured an Innovation Passport under the Innovative Licensing and Access Pathway (ILAP) from the UK Medicines and Healthcare products Regulatory Agency (MHRA). Zimislecel is investigational and has not been approved by health authorities globally. About VertexVertex is a global biotechnology company that invests in scientific innovation to create transformative medicines for people with serious diseases and conditions. The company has approved therapies for cystic fibrosis, sickle cell disease, transfusion-dependent beta thalassemia and acute pain, and it continues to advance clinical and research programs in these areas. Vertex also has a robust clinical pipeline of investigational therapies across a range of modalities in other serious diseases where it has deep insight into causal human biology, including neuropathic pain, APOL1-mediated kidney disease, IgA nephropathy, primary membranous nephropathy, autosomal dominant polycystic kidney disease, type 1 diabetes and myotonic dystrophy type 1. Vertex was founded in 1989 and has its global headquarters in Boston, with international headquarters in London. Additionally, the company has research and development sites and commercial offices in North America, Europe, Australia, Latin America and the Middle East. Vertex is consistently recognized as one of the industry's top places to work, including 15 consecutive years on Science magazine's Top Employers list and one of Fortune's 100 Best Companies to Work For. For company updates and to learn more about Vertex's history of innovation, visit or follow us on LinkedIn, Facebook, Instagram, YouTube and X. Special Note Regarding Forward-Looking StatementsThis press release contains forward-looking statements as defined in the Private Securities Litigation Reform Act of 1995, as amended, including, without limitation, (i) statements by Carmen Bozic, M.D., and Michael R. Rickels, M.D., M.S., in this press release, (ii) plans, expectations for, and the potential benefits of zimislecel, (iii) expectations for the Phase 1/2/3 clinical trial for zimislecel, including expectations for the trial to complete enrollment and dosing, and (iv) plans for potential regulatory submissions next year. While Vertex believes the forward-looking statements contained in this press release are accurate, these forward-looking statements represent the company's beliefs only as of the date of this press release and there are a number of risks and uncertainties that could cause actual events or results to differ materially from those expressed or implied by such forward-looking statements. Those risks and uncertainties include, among other things, that data from a limited number of patients may not be indicative of final clinical trial results, that data from the company's research and development programs may not support registration or further development of its potential medicines in a timely manner, or at all, due to safety, efficacy, that timelines for regulatory submissions may be longer than anticipated, and other risks listed under the heading "Risk Factors" in Vertex's most recent annual report and subsequent quarterly reports filed with the Securities and Exchange Commission at and available through the company's website at You should not place undue reliance on these statements, or the scientific data presented. Vertex disclaims any obligation to update the information contained in this press release as new information becomes available. (VRTX-GEN) Investor Event and Webcast Vertex will host an investor event on Friday, June 20, 2025, at 7:15 p.m. CT/8:15 p.m. ET, in Chicago, to discuss the positive zimislecel data in type 1 diabetes. A live webcast of the presentation and Q&A portions can be accessed through the Investor Relations section of Vertex's website at An archived webcast will be available on the company's website. View source version on Contacts Vertex Pharmaceuticals IncorporatedInvestors: InvestorInfo@ Media: mediainfo@ orInternational: +44 20 3204 5275

EMA Recommends Rezdiffra for MASH Liver Fibrosis
EMA Recommends Rezdiffra for MASH Liver Fibrosis

Medscape

time8 hours ago

  • Medscape

EMA Recommends Rezdiffra for MASH Liver Fibrosis

The European Medicines Agency (EMA) has recommended conditional marketing authorization for Rezdiffra (resmetirom) for adults with noncirrhotic metabolic dysfunction-associated steatohepatitis (MASH) with moderate-to-advanced liver fibrosis. MASH was formerly known as nonalcoholic fatty liver disease (NASH). There are currently no authorized treatments for MASH in the EU, making Rezdiffra a potential first. MASH is a serious liver disease that occurs when fat accumulates in the liver, causing inflammation. Left untreated, it can lead to cirrhosis and cancer. Symptoms may only present in more advanced stages of the condition, at which point they may include discomfort or pain in the upper right abdomen, muscle weakness or loss, and swelling in the abdomen or legs. Estimates suggest that up to 5% of people in Europe have MASH and that around 25% have metabolic dysfunction‐associated steatotic liver disease (MASLD), the condition that precedes MASH. MASLD was previously known as nonalcoholic fatty liver disease (NAFLD). The active substance of Rezdiffra is resmetirom, which is a partial agonist of the thyroid hormone receptor-beta. It works by promoting lipophagy and hepatic fatty acid beta-oxidation to reduce liver fat, inflammation, and liver fibrosis. The EMA's decision comes after interim results from a pivotal, ongoing phase 3 trial including 966 adults with biopsy-confirmed MASH with varying stages of fibrosis. Patients were randomly assigned treatment on a 1:1:1 ratio to receive once-daily resmetirom at 80 mg or 100 mg, or a placebo. The primary endpoints were MASH resolution at week 52 with no worsening of fibrosis, and a reduction in fibrosis by at least one stage alongside no worsening of NAFLD activity score. MASH resolution included a reduction in the NAFLD activity score by ≥ 2 points. Scores range from 0 to 8; higher scores indicate more severe disease. After 12 months, 30% of patients in the 100 mg resmetirom group and 26% of those in the 80 mg group achieved MASH resolution with no worsening fibrosis compared with 10% in the placebo group. Meanwhile, 26% of patients in the 100 mg resmetirom group and 24% of patients in the 80 mg group experienced fibrosis improvement by at least one stage with no worsening of NAFLD activity score compared with 14% in the placebo group. The most frequent side effects were diarrhea, nausea, itching, and pruritus. Diarrhea and nausea were more frequent among those taking resmetirom than those taking the placebo. The rate of serious adverse events was similar across all groups and ranged from 10.9% to 12.7%. Rezdiffra will be available as 60 mg, 80 mg, and 100 mg film-coated tablets. It should be taken alongside diet and exercise. The opinion adopted by the EMA will now go to the European Commission to await a decision on EU-wide marketing authorization. In the meantime, the EMA has required Rezdiffra's applicant to complete both the pivotal and another ongoing trial to provide further data regarding the drug's efficacy. This conditional approval comes as the EMA perceives that the benefits to patients from immediate availability outweigh the risk inherent in incomplete data. Detailed recommendations for using Rezdiffra will be described in the summary of product characteristics, which will be published on the EMA website in all official European Union languages.

EMA Reviews Alcohol Dependency Drug After French Concerns
EMA Reviews Alcohol Dependency Drug After French Concerns

Medscape

time10 hours ago

  • Medscape

EMA Reviews Alcohol Dependency Drug After French Concerns

The Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency (EMA) has announced it will review the use of medicines containing sodium oxybate, a drug used in narcolepsy, in people with alcohol dependency. The review comes at the request of the French Medicines Agency and has been initiated under Article 31 of Directive 2001/83/EC. This procedure is used to address concerns over the safety or benefit-risk balance of a medicine or a class of medicines; to resolve disagreements between member states on issues related to the authorization of medicines; or to give an opinion on an issue of Europe-wide interest so that the agency can make a recommendation for a harmonized position across the EU. The European Commission, any member state, or the company that markets the medicine can initiate a referral. In a referral, the European Medicines Agency (EMA) is requested to conduct a scientific assessment of a particular medicine or class of medicines on behalf of the European Union. It involves the matter being referred to the Pharmacovigilance Risk Assessment Committee (PRAC) after due notice to the EMA, all member states, and the European Commission. Narcolepsy Indication Sodium oxybate (Xyrem) is the sodium salt of gamma-hydroxybutyric acid. As an oral solution (500 mg/mL), it is indicated for the treatment of narcolepsy with cataplexy in adult patients, adolescents, and children from the age of 7 years. The drug is a central nervous system depressant believed to act by attaching to brain cell receptor molecules to promote delta (slow) brain waves and nighttime sleep. When taken at bedtime, Xyrem increases sleep depth and length, reducing the number of sleeping periods during the day and improving symptoms of narcolepsy. Because of the drug's potential for abuse, it can only be obtained with a special prescription, and treatment should be initiated and maintained under the guidance of a doctor with experience in treating sleep disorders. Doctors are advised to check for a history of or susceptibility to drug abuse before treatment, and to monitor for misuse and abuse during treatment. Approved for Alcohol Abuse in Austria and Italy The drug has also been suggested to reduce pain and improve function in fibromyalgia, but the EMA said the data were insufficient to support its use for this in the European population. However, sodium oxybate is also approved in Italy and in Austria, under the name Alcover, to treat alcohol dependence and alcohol withdrawal syndrome. It is this use that the review will examine. The EMA has confirmed the positive benefit-risk profile of the treatment as prescribed in Italy and Austria. France, however, despite having a high prevalence of alcohol problems, has resisted its use for this purpose. Trials have suggested that sodium oxybate can rapidly alleviate symptoms of alcohol withdrawal syndrome and was at least as effective as diazepam and clomethiazole. It can also be used in the maintenance of abstinence in alcohol-dependent patients, in whom it is reportedly as effective as naltrexone or disulfiram. Because existing approved medications are only modestly effective, there is a pressing need for effective treatments with an alternative mode of action, particularly in patients with very high consumption levels. Sodium oxybate also has a favorable safety profile. However, studies in alcohol use disorder have been limited and further investigations have been recommended.

DOWNLOAD THE APP

Get Started Now: Download the App

Ready to dive into a world of global content with local flavor? Download Daily8 app today from your preferred app store and start exploring.
app-storeplay-store