PUBLICATIONS

FDA Decisions Expected: March 2025

Your monthly synopsis of new drugs expected to hit the market 

February 14, 2025

Drug pipeline for March 2025

At Prime Therapeutics (Prime), we have positioned ourselves to best prepare our clients to manage new drugs. Our clinical and trade relations teams keep a keen eye on drugs awaiting approval by the United States (U.S.) Food and Drug Administration (FDA).
March 2025: ataluren (Translarna)  
PTC Therapeutics is awaiting the FDA decision for ataluren for the treatment of nonsense mutation Duchenne muscular dystrophy (nmDMD). The FDA granted ataluren Fast Track and Orphan Drug designations. This is the third FDA review for ataluren following Complete Response Letters (CRLs) in 2016 and 2017 requiring additional clinical trial information. Ataluren is a protein restoration therapy that allows the cellular process that synthesizes dystrophin protein to bypass the genetic defect (nonsense mutation) in the dystrophin gene, thereby producing a functional dystrophin protein. The double-blind, Phase 3 Study 041 was conducted to address the CRL deficiencies.¹ In the trial, ataluren was administered orally as 40 mg/kg/day in three divided doses. While ataluren did not demonstrate statistical significance improvement in the primary endpoint of mean 6-Minute Walking Distance (6MWD), it did lead to a significant 21% slowing in the rate of decline in 6MWD in the intent-to-treat (ITT) population (p=0.0248). The greatest benefit was observed among patients with 6MWD of 300 to 400 meters with a reported 30% slowing in rate of decline in 6MWD (p=0.031). If approved, ataluren will provide a new mechanism of action for DMD by circumventing the dystrophin gene errors to produce a functional dystrophin protein. 

For more information, see the ataluren (Translarna) Deep Dive in the January 2025 edition of Prime’s Quarterly Drug Pipeline.  
March 2025: condoliase
Ferring and Seikagaku are awaiting the FDA decision for condoliase for radicular leg pain associated with lumbar disc herniation (LDH). Condoliase is an enzyme administered as a single life-time injection into the intervertebral disc where it degrades central tissue in the disc to relieve pressure on spinal nerves due to disc herniation. Condoliase was evaluated in two double-blind clinical trials in patients who failed to achieve resolution of pain with at least six weeks of conservative therapy (e.g. nonsteroidal anti-inflammatory drugs, physical therapy).² Both trials reported significant improvement from baseline to week 13 in worst leg pain during the past 24 hours averaged over the previous seven days (primary endpoint) as measured by the Visual Analog Scale (least square mean [LSM] difference, -7.5 [p=0.0263] in trial 1 and -15.2 [p=0.001] in trial 2). On Jan. 10, 2025, the FDA’s Anesthetic and Analgesic Drug Products Advisory Committee voted 8–4 in favor that condoliase’s benefits outweighed its risks in patients with radicular leg pain associated with LDH. The panel acknowledged that condoliase would provide a less invasive option compared to surgery, if approved. However, panel members had concerns related safety profile, such as increased risks for severe cutaneous adverse reaction [SCAR], arthritis and disc degeneration. Additional concerns were related to lack of long-term data and the need for special training for clinicians. Panel members recommended a narrower indication, including confirmation of nerve compression, prior failure of six weeks of conservative therapy and post-marketing surveillance.  

For more information, see the condoliase Deep Dive in the January 2025 edition of Prime’s Quarterly Drug Pipeline.  
March 18, 2025: revakinagene taroretcel (NT-501)  
Revakinagene taroretcel, by Neurotech, is a first-in-class treatment that uses encapsulated cell therapy (ECT) technology to promote continuous, long-term delivery of ciliary neurotrophic factor (CNTF) in the eye to reduce photoreceptor cell loss. The FDA granted the product Fast Track and Orphan Drug designations and a Priority Review for the treatment of macular telangiectasia (MacTel) type 2. Revakinagene taroretcel is surgically inserted into the vitreous of the eye during a single outpatient procedure. In clinical trials, it preserved photoreceptors by up to 56% compared to sham at 24 months, as measured by change in ellipsoid zone (EZ) area.³ However, revakinagene taroretcel has not demonstrated a significant improvement in vision. In December 2024, the FDA extended the original decision date by three months. If approved, revakinagene taroretcel will be the first treatment indicated for MacTel type 2. 

For more information, see the revakinagene taroretcel (NT-501)  Deep Dive in the October 2024 edition of Prime’s Quarterly Drug Pipeline.  
March 20, 2025: rivoceranib + camrelizumab 
Elevar is awaiting the FDA decision for rivoceranib for use in combination with camrelizumab, by Jiangsu Hengrui, for the first-line treatment of unresectable or metastatic hepatocellular carcinoma (HCC). Rivoceranib is an oral tyrosine kinase inhibitor (TKI) of vascular endothelial growth factor receptor 2 (VEGFR-2) and camrelizumab is an intravenously (IV)-administered programmed cell death-1 (PD-1)-directed humanized monoclonal antibody. Both products received Orphan Drug designation from the FDA for HCC. In the open-label, Phase 3 CARES-310 trial, treatment with the combination resulted in a significant improvement in the primary endpoints of progression-free survival (PFS) and overall survival (OS) compared to sorafenib (400 mg orally twice daily).⁴ At a median follow-up of 7.8 months, the median PFS was 5.6 months with rivoceranib plus camrelizumab and 3.7 months with sorafenib (p<0.0001). At a median follow-up of 14.5 months, the median OS was 22.1 months with rivoceranib plus camrelizumab and 15.2 months with sorafenib (p<0.0001). This is the second review for rivoceranib plus camrelizumab, following a CRL issued by the FDA in May 2024 citing deficiencies related to a manufacturing site inspection. If approved, the combination of oral rivoceranib plus IV camrelizumab will likely compete with the standard of care regimen of IV atezolizumab (Tecentriq) + IV bevacizumab (Avastin) and the recently approved all-IV regimen of tremelimumab-actl (Imjudo) + durvalumab (Imfinzi).  

For more information, see the rivoceranib + camrelizumab Deep Dive in the January 2024 edition of Prime’s Quarterly Drug Pipeline.  
March 26, 2025: gepotidacin  
Gepotidacin is undergoing a Priority Review by the FDA for the treatment of uncomplicated urinary tract infection (uUTI) in females at least 12 years of age weighing at least 40 kg. The first-in-class triaza-acenaphthylene antibiotic by GlaxoSmithKline also received a Qualified Infectious Disease Product (QIDP) designation for uUTI. Gepotidacin is administered orally twice daily for five days. In the Phase 3 EAGLE-2 and EAGLE-3 trials, gepotidacin was non-inferior to nitrofurantoin, a current standard of care for uUTI.⁵ In EAGLE-2, gepotidacin led to therapeutic success, defined as combined complete symptom resolution and reduction of qualifying uropathogens to <10³ CFU/mL, in 50.6% of patients compared to 47% for nitrofurantoin. In EAGLE-3, therapeutic success was achieved in 58.5% of patients who received gepotidacin compared to 43.6% who received nitrofurantoin. The development of gepotidacin has been funded, in part, using federal funds from the U.S. government. Current oral agents approved to treat uUTI include nitrofurantoin, trimethoprim-sulfamethoxazole, fosfomycin trometamol and pivmecillinam. If approved, gepotidacin will provide an additional option for treating uUTI.
March 27, 2025: diazoxide choline controlled-release (DCCR)
Soleno submitted DCCR to the FDA under a 505(b)(2) new drug application (NDA) for the treatment of Prader-Willi syndrome (PWS) in children at least 4 years of age with hyperphagia. PWS is a rare, genetic disorder with a hallmark feature of excessive hunger (hyperphagia) that can lead to severe obesity. DCCR is a potent activator of the ATP-sensitive potassium (Kᴬᵀᴾ) channel resulting in a decrease in secretion of endogenous appetite stimulatory neuropeptides in the hypothalamus. It received Breakthrough Therapy, Fast Track and Orphan Drug designations as well as a Priority Review by the FDA. In December 2024, the FDA extended the original decision date by three months. DCCR is administered orally once daily. In clinical trials C601 and C602, the DCCR group experienced significant improvements in hyperphagia compared to natural history, as measured by the Hyperphagia Questionnaire for Clinical Trials [HQ-CT]) at 26 weeks (least square mean [LSM] change from baseline, -8.3 points versus -2.5 points, respectively; p<0.001) and 52 weeks (LSM change from baseline, -9.2 points versus -3.4 points, respectively; p<0.001); a reduction in HQ-CT score of at least 7 points was considered clinically meaningful.⁶ If approved, DCCR will be the first pharmacologic agent available to reduce hunger and improve food-related behaviors in patients with PWS.
March 27, 2025: etripamil (Cardamyst) 
Milestone is awaiting the FDA decision for etripamil, a fast-acting, non-dihydropyridine calcium channel blocker (CCB), for the acute conversion of atrioventricular (AV)-nodal-dependent paroxysmal supraventricular tachycardia (PSVT). In the double-blind, placebo-controlled, Phase 3 RAPID trial, participants were trained to recognize onset of PSVT symptoms, attach an electrocardiogram (ECG) monitor and conduct the vagal maneuver. Enrollees were randomized to etripamil or placebo self-administered as an intranasal spray.⁷ The study reported significantly more patients in the etripamil group experienced conversion of a PSVT episode to sinus rhythm within 30 minutes after the first dose compared to patients in the placebo group (64% versus 31%, respectively; p<0.0001). The median time to conversion was 17.2 minutes with etripamil compared to 53.5 minutes with placebo. In a joint guidance by the American College of Cardiology, American Heart Association and Heart Rhythm Society (2015), the agencies advise that self-administered (“pill-in-the-pocket”) off-label use of acute doses of oral beta-blockers or non-dihydropyridine CCBs may be reasonable for ongoing management to terminate acute episodes of PSVT caused by AV-nodal reentrant tachycardia in certain patients. If approved, etripamil will be the first agent specifically indicated in this space and may lead to fewer medical interventions and emergency department visits due to PSVT.

For more information, see the etripamil (Cardamyst) Deep Dive in the January 2025 edition of Prime’s Quarterly Drug Pipeline
March 28, 2025: fitusiran 
Fitusiran is a first-in-class small interference RNA (siRNA) by Sanofi and Alnylam submitted to the FDA for prophylactic treatment of hemophilia A or B in adults and adolescents with or without inhibitors. The agent uses the body’s natural cellular RNA interference mechanisms to increase thrombin generation and rebalance hemostasis. Fitusiran was granted Breakthrough Therapy and Fast Track designations for hemophilia by the FDA. In the open-label, Phase 3 ATLAS-A/B trial, once monthly subcutaneous injections of fitusiran significantly reduced annualized bleeding rate (ABR) compared to on-demand clotting factor concentrates (3.1 versus 31, respectively; p<0.0001), with ABR rates of zero observed for 51% of patients who received fitusiran.⁸ In addition, interim results from the open-label extension ATLAS-OLE trial reported benefit with every-other-month dosing, further reducing treatment burden compared to factor and other non-factor products (excluding gene therapy).⁹ If approved, fitusiran will provide an alternative non-factor prophylactic treatment for patients with hemophilia A or B with or without inhibitors. 

For more information, see the fitusiran Deep Dive in the January 2025 edition of Prime’s Quarterly Drug Pipeline

All brand names are property of their respective owners.
Login Portals
Compliance / Legal
Careers
© 2025 Prime Therapeutics LLC