關於 cookie 的說明

本網站使用瀏覽器紀錄 (Cookies) 來提供您最好的使用體驗,我們使用的 Cookie 也包括了第三方 Cookie。相關資訊請訪問我們的隱私權與 Cookie 政策。如果您選擇繼續瀏覽或關閉這個提示,便表示您已接受我們的網站使用條款。

搜尋結果Search Result

符合「Clinical Trials/Medical Discoveries」新聞搜尋結果, 共 1475 篇 ,以下為 1 - 24 篇 訂閱此列表,掌握最新動態
Brii Bio Unveils New Data from Its Ongoing Phase 2 ENSURE Study at APASL 2025, Showcasing BRII-179's Unique Potential to Prime and Boost Higher HBsAg Loss Through Target Patient Identification

Preliminary data from Cohort 4 of the ENSURE study supports a novel enrichment strategy to utilize BRII-179 to identify patients who are immune responders and have the potential to achieve higher HBsAg loss at EOT 48-week EOT data from Cohort 1-3 of the ENSURE study clearly suggests the added benefits of elebsiran towards achieving a higher rate of HBsAg loss in combination with PEG-IFNα DURHAM, N.C. and BEIJING, China, March 30, 2025 /PRNewswire/ -- Brii Biosciences Limited ("Brii Bio," or the "Company", stock code: 2137.HK), a biotechnology company developing therapies to improve patient health and choice across diseases with high unmet medical needs, announced new data from its ongoing Phase 2 ENSURE study as a late-breaking oral presentation at the 34th Annual Meeting of Asian Pacific Association for the Study of the Liver (APASL 2025) in Beijing, China. ENSURE (NCT05970289) is a multicenter, open-label Phase 2 study. Cohorts 1-3 were designed to evaluate the contribution of elebsiran, an investigational small interfering ribonucleic acid (siRNA), in combination with pegylated interferon alpha (PEG-IFNα) in participants with chronic HBV infection with baseline hepatitis B surface antigen (HBsAg) of 100-3,000 IU/mL. Participants who completed 9 doses of BRII-179, a recombinant protein-based therapeutic vaccine, in combination with elebsiran (BRII-835) in a previous APAC study BRII-179-835-001 (NCT04749368) were enrolled to Cohort 4 of this study and received elebsiran and PEG-IFNα combination treatment. The design of Cohort 4 as part of this study was based on insight from previous studies that a significant proportion of the chronic HBV patients fail to generate a sufficient immune response after receiving multiple doses of BRII-179, and therefore unlikely to have the immune support to achieve sustainable functional cure. Emerging data from Cohort 4 showed that participants who previously had BRII-179 induced anti-HBs response achieved a substantially higher rate of HBsAg seroclearance than those who did not. At Week 24, more than half of the BRII-179 responders (55.6% [10/18]) achieved HBsAg seroclearance, compared to only 10.0% (1/10) in non-responders. These latest data suggest that BRII-179 can serve as a predictive tool for enriching patients more likely to respond to curative therapies. Additional data from Cohorts 1-3 of the ENSURE study showed that higher end of treatment (EOT) HBsAg loss and seroconversion rates in participants receiving elebsiran in combination with PEG-IFNα than those receiving PEG-IFNα alone.   "The positive Cohort 4 data from the ENSURE study opens new doors for HBV functional cure," said Dr. Grace Lai-Hung Wong, Professor of Gastroenterology and Hepatology at CUHK Medical Data Analytics Centre (MDAC) and Department of Medicine and Therapeutics in Hong Kong SAR, China. "Previous studies have suggested that BRII-179 may offer a unique opportunity to identify CHB patients who are able to elicit the necessary HBsAg antibody response. I believe these new findings provide clear evidence supporting such value proposition and further substantiate the role of BRII-179 in shaping future combination therapies." "We are encouraged that the Cohort 4 from the ENSURE study continue to support our enrichment strategy in developing a functional cure for chronic HBV in target populations," said David Margolis, MD, Chief Medical Officer of Brii Bio. "The results underscore the potential of BRII-179 in identifying patients who are more likely to respond to regimens aimed at functional cure, thereby enhancing functional cure rates in the target population while reducing exposure to costly therapies for those with a lower probability of cure. We are committed to advancing BRII-179 in combination with various modalities through our ongoing studies and collaborations with strategic partners, aiming to deliver higher functional cure rates to 254 million patients worldwide living with chronic HBV infection." Abstract Number: OP0335 Presentation Title: Responders to Prior BRII-179 Treatment Achieved Faster and Higher Rate of HBsAg Seroclearance Following Treatment of Elebsiran and PEG-IFNα in Participants with Chronic Hepatitis B Virus Infection: Preliminary Data from ENSURE Study Presenter: Grace Lai-Hung Wong, MBChB (CUHK), MD (CUHK), FRCP (Lond, Edin), FHKCP, FHKAM (Medicine), Professor of Gastroenterology and Hepatology at CUHK Medical Data Analytics Centre (MDAC) and Department of Medicine and Therapeutics in Hong Kong SAR, China Among the 28 of the 31 participants enrolled in Cohort 4 with baseline HBsAg ≥ 100 IU/mL being analyzed, 18 participants with peak anti-HBs ≥ 10 IU/L induced by prior BRII-179 treatment were defined as BRII-179 responders and 10 participants with peak anti-HBs < 10 IU/L were defined as non-responders. At Week 24 of treatment with elebsiran + PEG-IFNα, 39.3% (11/28) of the Cohort 4 participants achieved HBsAg seroclearance. The rate of HBsAg seroclearance at Week 24 in the BRII-179 responders was 55.6% (10/18), notably higher compared to the non-responders at 10% (1/10). Responders to prior BRII-179 treatment appeared to achieve a faster HBsAg seroclearance compared to BRII-179 naïve participants receiving elebsiran + PEG-IFNα as previously reported. Elebsiran and PEG-IFNα combination therapy was generally safe and well tolerated in participants with virally suppressed chronic HBV infection. Treatment with elebsiran + PEG-IFNα is ongoing for 48 weeks. Abstract Number: LB0009 Presentation Title: Higher end of treatment (EOT) HBsAg loss and seroconversion rates in participants with chronic HBV infection receiving elebsiran (BRII-835) and pegylated interferon alfa-2a (PEG-IFNα) compared to PEG-IFNα alone: Week 48 results from ongoing ENSURE study Presenter: Jidong Jia, M.D., Ph.D., Professor of Medicine at the Liver Research Centre, Beijing Friendship Hospital, Capital Medical University in Beijing, China The rates of HBsAg seroclearance at EOT in elebsiran 200 mg + PEG-IFNα and elebsiran 100 mg + PEG-IFNα cohorts were 26.3% (5/19) and 33.3% (6/18), respectively, notably higher compared to PEG-IFNα alone cohort (5.6%) in participants with baseline HBsAg levels 100-3,000 IU/mL. Greater HBsAg reductions at EOT were observed in elebsiran + PEG-IFNα combination cohorts (mean [SE]: -2.47 [0.28] or -3.01 [0.28] log10 IU/mL in elebsiran 200 mg or 100 mg, respectively) than in PEG-IFNα cohort (-1.02 [0.30] log10 IU/mL). Elebsiran in combination with PEG-IFNα at both 200 mg and 100 mg doses achieved similar HBsAg reductions and seroclearance rates. Elebsiran and PEG-IFNα combination therapy was generally safe and well tolerated in participants with virally suppressed chronic HBV infection. Post-treatment follow-up is ongoing and will continue for 24 weeks after discontinuation of treatment. As part of Brii Bio's unique approach to developing a functional cure for HBV, the Company and its partners are actively progressing multiple combination studies with our differentiated portfolio, including BRII-179, a recombinant protein-based HBV immunotherapeutic being evaluated in multiple combination studies with elebsiran led by Brii Bio; elebsiran being evaluated in combination with PEG-IFNα  in studies led by Brii Bio and tobevibart, an investigational broadly neutralizing monoclonal antibody targeting HBV, being evaluated  in multiple Phase 2 and 3 tobevibart and elebsiran combination studies led by Vir Biotechnology. Key data readouts will be shared in the coming months at the scientific conferences throughout 2025. About Hepatitis B Hepatitis B virus (HBV) infection is one of the world's most significant infectious disease threats with more than 254 million people infected globally.[1] Chronic HBV infection is the leading cause of liver disease and an estimated 820,000 people die of complications from chronic HBV infection each year.[1] HBV is of exceptional concern in China, where 87 million people are chronically infected.[2] About BRII-179 BRII-179 is a novel recombinant protein-based HBV immunotherapeutic candidate that expresses the Pre-S1, Pre-S2, and S HBV surface antigens, and is designed to induce enhanced and broad B-cell and T-cell immunity. In November 2023, the Center for Drug Evaluation (the "CDE") of the National Medical Products Administration (the "NMPA") granted BRII-179 Breakthrough Therapy Designation. About Elebsiran (BRII-835, VIR-2218) Elebsiran is an investigational subcutaneously administered HBV-targeting siRNA designed to degrade hepatitis B virus RNA transcripts and limit the production of hepatitis B surface antigen. It has the potential to have direct antiviral activity against HBV and HDV. It is the first siRNA in the clinic to include Enhanced Stabilization Chemistry Plus technology to enhance stability and minimize off-target activity, which potentially can result in an increased therapeutic index. Brii Bio licensed exclusive rights to develop and commercialize elebsiran for the Greater China territory from Vir Biotechnology, Inc. in 2020. About Brii Bio Brii Biosciences Limited ("Brii Bio", stock code: 2137.HK) is a biotechnology company developing therapies to address major public health challenges where patients experience high unmet medical needs, limited choice and significant social stigmas. With a focus on infectious diseases, the Company is advancing a broad pipeline of unique therapeutic candidates with lead programs against hepatitis B virus (HBV) infection. The Company is led by a visionary and experienced leadership team and has operations in key biotech hubs, including Raleigh-Durham, the San Francisco Bay Area, Beijing and Shanghai. For more information, visit www.briibio.com. [1] World Health Organization. (April 2024). Global hepatitis report 2024: action for access in low- and middle-income countries. World Health Organization. Retrieved from https://www.who.int/publications/i/item/9789240091672 [2] World Health Organization. Hepatitis. World Health Organization. Retrieved from https://www.who.int/china/health-topics/hepatitis#:~:text=There%20are%2087%20million%20people,living%20with%20chronic%20hepatitis%20C.  

文章來源 : PR Newswire 美通社 發表時間 : 瀏覽次數 : 94 加入收藏 :
AnHorn Medicines Announces First Subject Dosed in AH-001 Phase I Clinical Trial in the United States

TAIPEI, March 27, 2025 /PRNewswire/ -- AnHorn Medicines, a pioneering AI-driven new drug discovery company, is pleased to announce that, AH-001, a first-in-class protein degrader for androgenetic alopecia (AGA), has officially entered Phase I clinical trial in the United States. The first subject was dosed this week, marking a major milestone in the development of this innovative therapy. AH-001 represents a groundbreaking advancement in AGA treatment with its novel mechanism of action as a selective protein degrader. Unlike traditional treatments that focus on inhibiting specific enzymes or stimulating hair follicles, AH-001 precisely targets and eliminates key proteins linked to hair loss. This innovative approach has the potential to offer a more effective and long-lasting solution for millions of individuals suffering from AGA worldwide. The global market for AGA treatments is projected to exceed $10 billion by 2030, driven by the growing demand for more effective and sustainable therapies. Current treatments, such as finasteride and minoxidil, have limitations in efficacy and potential side effects, leaving a significant unmet medical need. AH-001's innovative mechanism has the potential to be a game-changer, offering a new level of therapeutic benefit for AGA patients. AnHorn's success in developing AH-001 is driven by its proprietary AI-powered drug discovery platform. By leveraging artificial intelligence, AnHorn has impressively accelerated the identification of promising protein degraders, streamlining the early-stage drug development process. This AI-powered approach has not only improved the efficiency of drug discovery but has also provided valuable insights into targeting complex biological pathways, reinforcing AnHorn's leadership in next-generation therapeutic innovation. With the launch of the Phase I study, AnHorn remains committed to advancing AH-001 through clinical development, aiming to deliver a transformative solution to the market. The company will continue to provide updates as the trial progresses. About AnHorn MedicinesAnHorn is a clinical-stage biotechnology company dedicated to revolutionizing drug discovery through AI-powered innovation. By integrating artificial intelligence with cutting-edge molecular research, AnHorn is pioneering new treatments for conditions with high unmet medical needs. AH-001 is the company's first leading program through its AI-driven platform, exemplifying its mission to create breakthrough therapies that improve patient outcomes.

文章來源 : PR Newswire 美通社 發表時間 : 瀏覽次數 : 292 加入收藏 :
Keymed Biosciences Announces Annual Results of 2024

CHENGDU, China, March 26, 2025 /PRNewswire/ -- March. 25, 2025, Keymed Biosciences Inc. (HKEX: 02162) announced its annual results of 2024, along with a corporate update.  Rapid development of our pipeline products Stapokibart (CM310) (IL-4Rα antibody) Three new drug applications of Stapokibart for the treatment of moderate-to-severe atopic dermatitis (AD) in adults, chronic rhinosinusitis with nasal polyposis (CRSwNP) and seasonal allergic rhinitis have been approved by the NMPA. By the end of 2024, Stapokibart resisted strong sales and recorded revenue of approximately RMB40 million within three and a half months. In September 2024, the new drug application of Stapokibart injection for the treatment of moderate-to-severe atopic dermatitis in adults was approved by the NMPA. The phase III clinical study results indicated that at week 52, the rates of achieving EASI-75 for the Stapokibart group and the placebo-to-Stapokibart group were 92.5% and 88.7%, respectively. The EASI-90 response rates were 77.1% and 65.6%, respectively. The rates of achieving an IGA score of 0 or 1 point with a reduction of ≥ 2 points from baseline were 67.3% and 64.2%, respectively. Additionally, the rates of achieving a weekly average reduction of ≥ 4 points from baseline in the daily PP-NRS score were 67.3% and 60.5%, respectively. Long-term treatment with Stapokibart can consistently improve dermatitis symptoms and quality of life in subjects with moderate-to-severe AD. During the maintenance period, only one subject (0.9%) experienced a relapse. In December 2024, the new drug application of Stapokibart injection for the treatment of chronic rhinosinusitis with nasal polyps was approved by the NMPA. The study results showed that the data from the Phase III clinical trial was positive. Compared to the placebo, Stapokibart significantly reduced nasal polyps (NPS improvement of 2.3 from baseline) and alleviated nasal congestion (NCS improvement of 0.7 from baseline) after 24 weeks. The differences were highly statistically significant (P < 0.0001). Additionally, it effectively relieved rhinosinusitis, restored sense of smell, improved nasal symptoms, and enhanced quality of life. In February 2025, the new drug application of Stapokibart injection for the treatment of seasonal allergic rhinitis was approved by the NMPA. We advanced and completed the data unblinding and statistical analysis for the Phase III clinical study of Stapokibart injection for the treatment of seasonal allergic rhinitis (SAR). The study findings demonstrate that during the pollen season, in comparison with the standard treatment group, which consists of nasal spray hormones combined with antihistamine drugs, the administration of Stapokibart for two weeks effectively controls the typical nasal allergic symptoms of patients, including runny nose, nasal congestion, nasal itching, and sneezing. The least-squares mean (LSMean) of the inter-group difference is -1.3, and its 95% confidence interval (CI) is also -1.3, indicating a highly significant statistical difference (P = 0.0008). This difference far exceeds the minimal clinically important difference (MCID) of 0.23, clearly demonstrating substantial clinical benefits. Moreover, Stapokibart can effectively alleviate ocular allergic symptoms such as eye itching or burning, eye tearing or watering, and eye redness. It comprehensively enhances the quality of life of patients and exhibits excellent safety. In February 2024, we launched a randomized, double-blinded, placebo-controlled Phase III clinical study to evaluate the efficacy and safety of Stapokibart injection in adolescent subjects with moderate-to-severe AD. In May 2024, we initiated a randomized, double-blinded, placebo-controlled Phase III clinical study to evaluate the efficacy and safety of Stapokibart injection in subjects with nodular prurigo. As of the date of this announcement, the patient enrollment for this clinical study is in progress. In June 2024, the long-term efficacy and safety data from the Phase III clinical trial of Stapokibart injection for the treatment of moderate-to-severe AD were presented by way of oral presentation at the European Academy of Allergy and Clinical Immunology (EAACI) Congress 2024. In October 2024, the full text of the 52-week efficacy and safety data of the Phase III clinical study was published in the《Allergy》, the top international journal in allergy and immunology field.   CMG901/AZD0901 (Claudin 18.2 ADC)AstraZeneca has conducted multiple clinical studies regarding CMG901 (AZD0901) for the indications of gastric cancer, pancreatic cancer and biliary tract cancer, including the Phase III clinical trial for 2L+ gastric cancer, Phase II clinical trial for 1L gastric cancer, Phase II clinical trial for 1L pancreatic cancer, and Phase II clinical trial for 2L+ biliary tract cancer. In June 2024, the data from the Phase I clinical study of CMG901 (AZD0901) in the treatment of advanced G/GEJ cancer were presented by way of oral presentation at the American Society of Clinical Oncology (ASCO) Annual Meeting 2024. On 6 January 2025, the data from the Phase I clinical study was released on the international authoritative oncology journal, The Lancet Oncology. The clinical data indicated that the median progression free survival (mPFS) for all 93 patients with Claudin 18.2-high expressing G/GEJ cancer was 4.8 months, and the median overall survival (mOS) was 11.8 months. CM313 (CD38 antibody) In 2024, we initiated and advanced a multi-center, open-label Phase I/II clinical study. In June 2024, a research paper titled "A Novel Anti-CD38 Monoclonal Antibody for Treating Immune Thrombocytopenia" was published in The New England Journal of Medicine. 95.5% of patients achieved a platelet count of ≥50 × 109/L within 8 weeks upon the first acceptance of CM313 infusion, and the durable platelet count response rate (defined as a platelet count of ≥50 × 109/L observed six or more times among the final eight platelet counts) was 63.6%. In July 2024, we completed Phase Ib/IIa clinical study to evaluate the safety, tolerability, pharmacokinetics, pharmacodynamics, immunogenicity and preliminary efficacy of CM313 injection in subjects with SLE. We plan to initiate a Phase II clinical study in the first half of 2025. In 2024, we initiated a randomized, double-blinded, placebo-controlled Phase II clinical study to evaluate the safety, tolerability, pharmacokinetics, pharmacodynamics, immunogenicity, and preliminary efficacy of CM313(SC) injection in subjects with primary immune thrombocytopenia. The first patient was enrolled and dosed in November 2024. We initiated a randomized, double-blinded, placebo-controlled Phase II clinical study to evaluate the safety and efficacy of CM313(SC) injection in subjects with IgA nephropathy in early 2025. As of the date of this announcement, preparations for patient enrollment are underway for this study. CM512 (TSLP x IL-13 bispecific antibody) We have initiated a randomized, double-blinded, single/multiple dose-escalation, placebo-controlled Phase I clinical study to evaluate the safety, tolerability, pharmacokinetics, pharmacokinetics and immunogenicity of CM512 in healthy subjects and patients with moderate-to-severe atopic dermatitis, with enrollment of the first subject completed in September 2024. The overseas Phase I clinical trial evaluating CM512 for the treatment of asthma was initiated in the first quarter of 2025. CM336 (BCMAxCD3 bispecific antibody) Continuously proceeded with a multi-center, open-label Phase I/II clinical study to assess CM336 injection in treating patients with relapsed or refractory multiple myeloma. As of the date of this announcement, the product is currently in the dose-expansion phase of Phase I/II clinical study. In December 2024, the latest data of the phase I/II clinical study of CM336 for relapsed or refractory multiple myeloma was presented as a poster at the 66th American Society of Hematology (ASH) Annual Meeting. CM383 (Aβ protofibrils antibody) As of the date of this announcement, the enrollment of all subjects in the Phase Ia clinical study of CM383 in healthy subjects was completed; In November 2024, the enrollment of the first subject in Phase Ib clinical study of CM383 in patients with mild cognitive dysfunction of Alzheimer's disease origin and mild Alzheimer's disease was completed. CM518D1 (CDH17 ADC) We submitted IND application to CDE, and planned to conduct a multi-center, open-label Phase I/II clinical trial for evaluation of CM518D1 in treatment of patients with advanced solid tumors. CM355/ICP-B02 (CD20 x CD3 bispecific antibody) We are conducting a Phase I/II clinical trial in China to assess the safety, tolerability, PK, and the preliminary anti-tumor activity of CM355 in r/r NHL. Dose escalation of the intravenous infusion formulation ("IV") was completed and the subcutaneous formulation ("SC") is being evaluated. Our preliminary data of both IV and SC formulations have shown good efficacy of CM355 in patients with follicular lymphoma ("FL") and DLBCL. CM350 (GPC3 x CD3 bispecific antibody) Continuously proceeded with a Phase I/II clinical study in 2024 to assess the safety, tolerability, pharmacokinetics, and preliminary efficacy of CM350 in patients with advanced solid tumors. As of the date of this announcement, the product is currently in the dose-escalation of Phase I/II clinical study. CM369/ICP-B05 (CCR8 antibody) We are conducting a Phase I trial to evaluate the safety, tolerability, pharmacokinetic characteristics, and efficacy of CM369 in subjects with advanced solid tumors and relapsed/refractory NHL. Dose escalation of CM369 has been escalated up to 450 mg in solid tumor and 600 mg in NHL, CM369 was well tolerated with no DLTs nor ≥grade3 TRAEs observed. The preliminary results demonstrated a favorable PK profile with sufficient exposure for target coverage and regulatory T-cell depletion. CM380 (GPRC5D×CD3 bispecific antibody) Preclinical studies indicated that CM380 had favorable antitumor effects and was well tolerated. As of the date of this announcement, we are planning to conduct a multi-center, open-label Phase I/II clinical study for evaluation of CM380 in treatment of patients with relapsed or refractory multiple myeloma. Financial and Business Highlights Actively explore diversified BD models to maximize the global value of the pipeline. In July 2024, Chengdu Keymed entered into a license agreement with Belenos Biosciences,Inc. The license agreement grants Belenos the exclusive rights to develop, manufacture,and commercialize the Group's drug candidates CM512 and CM536 globally (excluding the Greater China region). In return, Chengdu Keymed shall receive an upfront and nearterm payment of US$15 million, and iBridge HK shall receive approximately 30.01% of the equity interest in Belenos as consideration. Subject to achievement of certain development, regulatory and commercial milestones, Chengdu Keymed may also receive additional payments up to US$170 million. As of the date of this announcement, Belenos is planning to initiate a Phase I clinical trial evaluating CM512 for the treatment of asthma. In November 2024, Chengdu Keymed and Platina Medicines Ltd ("PML") entered into an exclusive license agreement. The license agreement grants PML the exclusive right to develop, manufacture and commercialize CM336 globally excluding Mainland China, Hong Kong, Macau and Taiwan. In return, the Group shall receive an upfront and near-term payment of US$16 million and a minority equity interest in Ouro Medicines, LLC ("Ouro Medicines") as part of the consideration. Ouro Medicines is the parent company of PML and owns 100% of the equity interest in PML. The Group may also receive additional payments of up to US$610 million subject to achievement of certain clinical, regulatory and commercial milestones and is also entitled to receive tiered royalties on net sales of CM336 and related products from PML. In January 2025, Chengdu Keymed entered into an exclusive out-license agreement with Timberlyne Therapeutics, Inc. The license agreement grants the target company the exclusive right to develop, manufacture and commercialize CM313 globally (excluding Mainland China, Hong Kong, Macau and Taiwan). In return, the Group shall receive an upfront and near-term payment of US$30 million and equity interest of the target company, being the largest shareholder of this company. The Group may also receive additional payments up to US$337.5 million subject to achievement of certain sales and development milestones. The Group is also entitled to receive tiered royalties on net sales from the target company. In January 2025, Chengdu Keymed, Beijing InnoCare and the Joint Venture have entered into the Agreement with Prolium for the development and commercialization of CM355. Under the terms of the Agreement, Prolium will have the exclusive right to develop, register, manufacture, and commercialize CM355 globally in non-oncology indications and in oncology indications outside of Asia. Rapid expansion of workforce and production facilities As of December 31, 2024, we had 1,300 full-time employees in total, including over 240 employees engaging in commercialization and nearly 400 employees engaging in drug discovery and clinical operations. We will continue to recruit talents to meet the growing needs of commercialization, research and development, clinical, production and operation of the Company. As of the date of this announcement, the production capacity of our production base has reached 20,000 litres in total, and all the designs thereof are in compliance with the requirements of cGMP of the NMPA and FDA. Financial highlights As of December 31, 2024, the Company generated a total annual revenue of RMB 430 million, representing a 21% year-on-year increase. This includes about RMB 40 million in sales revenue from Stapokibart over three and a half months and RMB 390 million in licensing revenue from BD agreements. Throughout 2024, the company continued advancing its differentiated pipeline development and clinical research, with R&D expenditures reaching approximately RMB 730 million, a 23% year-on-year increase. The company maintained a strong cash position with total cash on hand of approximately RMB 2.1 billion. Moving forward, Keymed will continue to establish, and refine its technology platforms, rapidly advancing the development of pipeline candidates in China and globally. The company will comprehensively drive the commercialization of approved product and indications. Concurrently, Keymed will proactively explore strategic partnerships worldwide to accelerate global patient access to its drug candidates through diversified BD models. The company plans to further recruit talent in commercial sales, clinical development, and manufacturing, expand cGMP-compliant production capacity, reduce costs, enhance operational efficiency, and remain committed to developing, producing, and commercializing innovative therapies that are globally competitive, high-quality, and affordable for patients worldwide.

文章來源 : PR Newswire 美通社 發表時間 : 瀏覽次數 : 233 加入收藏 :
Juncell Therapeutics to Present Preclinical Data on Innovative Pretreatment Regimen of TIL Therapy at AACR Annual Meeting 2025

SHANGHAI, March 26, 2025 /PRNewswire/ -- Shanghai Juncell Therapeutics Co., Ltd. (Juncell Therapeutics), a clinical-stage biotech developing innovative IL-2-independent Tumor-Infiltrating Lymphocyte (TIL) therapies for cancers, announced that a preclinical study of innovative pretreatment regimens for cell therapy will be presented at the American Association for Cancer Research (AACR) Annual Meeting, taking place April 25 - 30 in Chicago. This study indicated that hydroxychloroquine (HCQ) could significantly increase the tumor-killing effects via elevating the membrane MHC-I protein levels of tumor cells without affecting TILs'proliferation. In addition, its effect on surface PD-L1 expression was not significant compared with IFN-γ. These results provide preclinical evidence for HCQ pretreatment of the adoptive cell therapy of TILs in clinical trials. Details of the poster presentation are below: Abstract Number: 5827Abstract Title: Hydroxychloroquine increases the tumor killing efficiency via elevating the membrane MHC-I protein levels of tumor cellsSession Title: Immunomodulatory Agents and InterventionsLocation: Poster Section 29Poster Board Number: 3Poster Presentation Time: 2:00 PM - 5:00 PM CST, April 29, 2025 About Juncell TherapeuticsJuncell Therapeutics is a biotech dedicated to developing high-quality, accessible TIL cell therapies for the treatment of solid tumors. Juncell Therapeutics has established its proprietary DeepTIL® cell expansion and NovaGMP® gene modification technology platforms, which are designed to address the key challenges of conventional TIL therapies, making TILs "robust, competent, affordable, and accessible." Two clinical-stage TIL therapies have demonstrated promising safety and efficacy in the treatment of ten types of heavily pretreated advanced solid tumors, including lung cancer, triple-negative breast cancer, pancreatic cancer, high-grade glioma, ovarian cancer, endometrial cancer, cervical cancer, head and neck cancer, bile duct cancer and melanoma. 7 of these late-stage tumor patients have achieved complete response, and the longest tumor-free survival time is over 3 years. For more information, please visit:https://www.juncell.com/en https://www.linkedin.com/company/juncell-therapeutics  Juncell Contact:contact@juncell.com

文章來源 : PR Newswire 美通社 發表時間 : 瀏覽次數 : 88 加入收藏 :
Antengene to Present Results From Four Cutting Edge Preclinical Studies at AACR 2025 with Focuses Including AnTenGagerTM TCEs and Synthetic Lethality

SHANGHAI and HONG KONG, March 26, 2025 /PRNewswire/ -- Antengene Corporation Limited ("Antengene", SEHK: 6996.HK), a leading innovative, commercial-stage global biopharmaceutical company dedicated to discovering, developing and commercializing first-in-class and/or best-in-class medicines for hematologic malignancies and solid tumors, today announced that it will release results from four preclinical studies in poster presentations at the 2025 American Association for Cancer Research Annual Meeting (AACR 2025), taking place from April 25th to 30th at the McCormick Place Convention Center, Chicago, the United States. These four posters will feature Antengene's four highly differentiated and high-potential programs, including ATG-201 (CD19 x CD3 TCE) and ATG-042 (MTAPnull-selective PRMT5 Inhibitor), which are poised to enter clinical development in the second half of 2025; ATG-110 (LY6G6D x CD3 TCE), which was developed on the AnTenGagerTM TCE 2.0 platform for the treatment of microsatellite stable colorectal cancer; and a companion diagnostic antibody developed to assess CD24 expression and guide clinical studies of CD24-targeted therapies. Details of the Poster Presentation:ATG-201 (CD19 x CD3 T cell engager)Title: ATG-201, a novel T-cell engager (TCE) effectively depletes B cells with reduced risk of CRS for the treatment of B cell malignancies and B cell related autoimmune diseasesAbstract Number: 7326Session Category: ImmunologySession Title: T Cell Engagers and Novel Antibody-Based TherapiesDate: April 30, 2025Time: 9:00 AM - 12:00 PM (Central Time)10:00 PM, April 30, 2025 - 1:00 AM, May 1, 2025 (Beijing Time)Location: Poster Section 40 Introduction: By depleting autoreactive B cells, CD19-targeted CAR-T have shown early yet promising efficacy in treating patients with B cell-driven autoimmune diseases. However, the clinical application of TCE continues to be greatly hindered by the unfavorable pharmacokinetics and toxicity associated with cytokine release syndrome. To overcome these limitations, Antengene developed ATG-201, a "2+1" CD19 x CD3 TCE, which was evaluated in a series of in vitro studies for binding affinity, T cell dependent cytotoxicity (TDCC) cytokine release, and drug developability. The in vivo anti-lymphoma efficacy and pharmacodynamic effect were evaluated in Raji xenograft model. The tissue resident B cell depletion was assessed in CD34+ hematopoietic stem cells humanized mice. Pharmacokinetic parameters of ATG-201 were evaluated in normal Balb/c mice. Results: Compared to benchmark TCEs, ATG-201 demonstrated stronger naïve B cell depletion activity with much lower cytokine release in vitro. ATG-201 showed potent anti-lymphoma activity in PBMC-humanized subcutaneous Raji xenograft model with significant augment of infiltrated CD8+ T cells in tumor microenvironment and limited proinflammatory cytokines detected in plasma. Single dose ATG-201 completely and deeply depleted B cells in blood, bone marrow and spleen of CD34+ cells humanized mice. ATG-201 demonstrated potent efficacy in mouse systemic lupus erythematosus model, inhibiting the lymph node swelling and auto-antibody producing. Conclusions: ATG-201 demonstrated CD19-dependent CD3 binding and activation, inducing effective B cell depletion in vitro and in vivo with low cytokine release, which provides potential for the treatment of B cell malignancies and B cell related autoimmune diseases. ATG-201 is poised to enter clinical development in the second half of 2025. ATG-042 (MTAPnull-selective PRMT5 Inhibitor)Title: Preclinical characterization of ATG-042, a novel MTAPnull-selective PRMT5 inhibitorAbstract Number: 4230Session Category: Experimental and Molecular TherapeuticsSession Title: HDAC and Methyltransferase InhibitorsDate: April 29, 2025Time: 9:00 AM - 12:00 PM (Central Time)10:00 PM, April 29, 2025 - 1:00 AM, April 30, 2025 (Beijing Time)Location: Poster Section 16 Introduction: Targeting the PRMT5-MTA complex has become a promising strategy for treating MTAPnull cancer in a synthetically lethal manner, avoiding on-target off-tumor hematological toxicity when using first-generation, non-selective PRMT5 inhibitors. Herein, Antengene developed ATG-042, a novel MTAPnull-selective PRMT5 small molecule inhibitor with good brain penetration. In this study, the in vitro activity and MTAP selectivity of ATG-042 were profiled using HCT116 MTAP wild type (wt) cells, HCT116 MTAP knock out (ko) cells and multiple endogenous MTAPnull cell lines. The in vivo efficacy was tested in cell derived xenograft (CDX) mouse models with HCT116 MTAP wt cells, HCT116 MTAP ko cells, LU99 cells (MTAPnull) and U87MG-luc (MTAPnull). The pharmacokinetic and toxicological properties were assessed with corresponding assay methods. Results: ATG-042 showed excellent anti-proliferative activities on multiple endogenous MTAPnull cell lines with IC50 values between 10nM and 100nM. ATG-042 demonstrated high permeability, good metabolic stability, and low risk of drug-drug interaction. In vivo PK study shows that ATG-042 is well absorbed, with a dose-dependent increase in plasma distribution and high oral bioavailability in mice, SD rats and beagle dogs. Furthermore, ATG-042 is brain-penetrable (B/P ratio=51% in mice; KPuubrain=0.73 in rats). ATG-042 showed robust in vivo efficacy in both subcutaneous CDX models (HCT116 -MTAP ko, LU99) and orthotopic CDX model (U87MG-luc) as a single agent. In addition, ATG-042 also exhibited potential synergy in combination with other drugs for antitumor therapy. Conclusions: ATG-042 is an oral MTAPnull-selective PRMT5 inhibitor with potent efficacy against MTAPnull tumor. It also demonstrated good tolerability and brain penetrability. ATG-042 is poised to enter clinical development in the second half of 2025. ATG-110 (LY6G6D x CD3 T cell engager)Title: ATG-110, a novel "2+1" LY6G6D-targeted T-cell engager (TCE) for the treatment of MSS colorectal cancerAbstract Number: 3509Session Category: ImmunologySession Title: T Cell EngagersDate: April 28, 2025Time: 2:00 PM - 5:00 PM (Central Time)3:00 AM - 6:00 AM, April 29, 2025 (Beijing Time)Location: Poster Section 38 Introduction: Colorectal cancer (CRC) is one of the most common cancers worldwide and requires more effective and safer therapies to improve the poor survival outcome, particularly in patients with microsatellite stable (MSS) colorectal cancer, who exhibit primary resistance to immune checkpoint inhibitors and lack effective treatment options. T cell engagers have shown encouraging efficacy in treating hematological malignancies, while exhibiting suboptimal clinical efficacies in solid tumors. The risk of cytokine release syndrome (CRS) remains as a significant challenge clinically. ATG-110 is a novel "2+1" LY6G6D x CD3 TCE developed by Antengene. In this study, ATG-110 was evaluated in a series of preclinical in vitro studies for binding affinity, T cell activation and cytokine release, T cell dependent cytotoxicity (TDCC), and drug developability. The in vivo anti-tumor efficacy was evaluated in PBMC-humanized immunodeficient NDG mice engrafted with LY6G6Dmedium-expression HT55 or LY6G6Dvery low-expression SW480 MSS CRC cells. Results: ATG-110 binds to LY6G6D-positive cell lines, including LY6G6D-overexpression 293T, HT55, LS1034, with the nanomolar grade EC50. The CD3 binding site of ATG-110 is concealed by the LY6G6D Fab arm before binding to LY6G6D, due to the steric hindrance. Therefore, ATG-110 demonstrated limited binding capability to CD3+ cells before LY6G6D crosslinking. It activates T cells and induces cytokine release only in the presence of LY6G6D+ cells. In vitro, ATG-110 resulted in potent T cell dependent cytotoxicity with single-digit pM IC50 values on HT55 and SW480 cells. ATG-110 also showed potent anti-tumor activity in PBMC-humanized SW480 xenograft model and exhibited complete response (CR) in PBMC-humanized HT55 xenograft model. Furthermore, ATG-110 also demonstrated good drug developability. Conclusions: ATG-110 demonstrated LY6G6D-dependent CD3 binding and activation with low risk of CRS. It showed powerful in vitro and in vivo anti-tumor efficacy against colorectal cancer, which warrants further clinical evaluation. ATG1144 (CD24 CDx Antibody)Title: Development of a diagnostic antibody for CD24 targeted therapyAbstract Number: 671Session Category: Clinical ResearchSession Title: Diagnostic Biomarkers 2Date: April 27, 2025Time: 2:00 PM - 5:00 PM (Central Time)3:00 AM - 6:00 AM, April 28, 2025 (Beijing Time)Location: Poster Section 29 Introduction: CD24 has emerged as a promising therapeutic target for anti-cancer treatment. Several clinical trials are being conducted to evaluate the safety and efficacy of CD24-targeted therapies. Here, Antengene developed and characterized an anti-CD24 diagnostic antibody to enhance the screening and selection of patients based on CD24 expression. In this study, the authors described the discovery of the diagnostic antibody, and the evaluation of accuracy, sensitivity (selectivity), specificity, and assay precision of the antibody. Results: Monoclonal antibody clone ATG1144 binds to the hCD24 core peptide in ELISA with an EC50 of 0.06 nM. Distinct membrane staining on human normal esophageal tissue FFPE specimens can also be observed with IHC staining using ATG1144. For accuracy assessment, six CDX and twenty human specimens, comprising both positive and negative specimens (including solid tumors and B-cell non-Hodgkin lymphomas), were validated. Samples exhibiting high, medium, and low CD24 expression levels were evaluated for sensitivity and specificity, and the interpreted results aligned with the reference outcomes. FFPE tissues from three distinct patients were evaluated for assay precision assessment. The TMA IHC staining result revealed that 50-80% of patients with lung, breast, bladder, ovarian, or liver cancer have CD24 expression on tumor cell surface with low expression in the para-cancerous normal tissue. Conclusions: ATG1144 specifically binds to human CD24 with high sensitivity as demonstrated by IHC staining. The development and validation of the method have been finalized using Leica Bond III platforms. These data suggest a potential diagnostic use of ATG1144 for identifying CD24+ patients. About Antengene Antengene Corporation Limited ("Antengene", SEHK: 6996.HK) is a leading commercial-stage R&D-driven global biopharmaceutical company focused on the discovery, development, manufacturing and commercialization of innovative first-in-class/best-in-class therapeutics for the treatment of hematologic malignancies and solid tumors, in realizing its vision of "Treating Patients Beyond Borders". Antengene has built a pipeline of 9 oncology assets at various stages going from clinical to commercial, including 6 with global rights, and 3 with rights for the APAC region. To date, Antengene has obtained 31 investigational new drug (IND) approvals in the U.S. and Asia, and submitted new drug applications (NDAs) in 11 Asia Pacific markets, with the NDA for XPOVIO® (selinexor) already approved in Mainland of China, Taiwan China, Hong Kong China, Macau China, South Korea, Singapore, Malaysia, Thailand, Indonesia and Australia. Forward-looking statements The forward-looking statements made in this article relate only to the events or information as of the date on which the statements are made in this article. Except as required by law, we undertake no obligation to update or revise publicly any forward-looking statements, whether as a result of new information, future events or otherwise, after the date on which the statements are made or to reflect the occurrence of unanticipated events. You should read this article completely and with the understanding that our actual future results or performance may be materially different from what we expect. In this article, statements of, or references to, our intentions or those of any of our Directors or our Company are made as of the date of this article. Any of these intentions may alter in light of future development. For a further discussion of these and other factors that could cause future results to differ materially from any forward-looking statement, please see the other risks and uncertainties described in the Company's Annual Report for the year ended December 31, 2024, and the documents subsequently submitted to the Hong Kong Stock Exchange. For more information, please contact: Investor Contacts: Donald LungE-mail: Donald.Lung@antengene.com  Mobile: +86 18420672158 PR Contacts:Peter Qian  E-mail: Peter.Qian@antengene.com Mobile: +86 13062747000

文章來源 : PR Newswire 美通社 發表時間 : 瀏覽次數 : 375 加入收藏 :
NUS researchers develop microneedle technology to accelerate diabetic wound healing

'Sponge-like' microneedle patches deliver bioactive ingredients and reduce inflammation in slow- and non-healing wounds SINGAPORE, March 26, 2025 /PRNewswire/ -- Diabetic wounds often lead to severe complications that can result in amputations. These chronic and non-healing wounds are marked by persistent inflammation, affecting more than six per cent of the global population. In Singapore, there are about four lower limb amputations daily due to non-healing diabetic wounds. A study focusing on diabetic wounds in Singapore estimated that the gross amputation-related healthcare cost per patient was S$23,000 in 2017. To address this challenge of great national and global importance, researchers from the National University of Singapore (NUS) have developed two microneedle technologies that have shown efficacy in accelerating diabetic wound healing in preclinical models by preserving the functions of proteins called growth factors, and removing undesirable inflammatory compounds. The two novel innovations were developed by a team of scientists led by Assistant Professor Andy Tay from the Department of Biomedical Engineering at the College of Design and Engineering at NUS, and the Institute for Health Innovation and Technology. "Growth factors are important for wound healing because they regulate key cellular functions. However, in diabetic wounds, these growth factors are rapidly broken down by other enzymes known as proteases. This dramatically slows down wound recovery. At the same time, diabetic wounds are characterised by persistently high levels of inflammation," he explained. "We wanted to tackle these two issues by using microneedles for both delivery and extraction. It is minimally invasive, can be fabricated with precision, and allows for the active compounds to be painlessly administered directly into wounds. Microneedle patches are excellent materials for wound healing," he said. The results of the two related studies, which were published online in the scientific journals Biomaterials and Advanced Functional Materials on 4 July 2024 and 24 July 2024 respectively, demonstrate the potential of this innovative approach in treating various skin conditions such as psoriasis or chronic diabetic wounds. Two unique approaches to accelerate wound healing  In the market, hydrogel is used to deliver growth factors to wounds. However, this method is not as effective because the protease-rich environment of chronic wounds rapidly degrades and inactivates the growth factors. This means that the growth factors need to be delivered in high doses repeatedly, which can be costly and time-consuming. In the first approach developed by the NUS research team, instead of delivering the growth factors directly, they first increased the production of growth factors within the wound. They achieved this by developing sucralfate microneedles (SUC-MN) to deliver an important immunomodulatory protein, interleukin-4 (IL-4), to stimulate the production of growth factors in diabetic tissues. IL-4 helps to regulate the immune response and promote tissue regeneration, while sucralfate, a medication commonly used to treat gastrointestinal ulcers, protects growth factors from degradation. The microneedles dissolve in the wound, delivering IL-4 and sucralfate directly to the wound. This localised delivery system minimises systemic side effects, and also avoids secondary damage to delicate, newly formed tissues caused by traditional adhesive dressing that is currently used clinically. The researchers found that SUC-MN significantly accelerated wound healing twice as fast when compared to traditional treatments. First-of-its-kind extractive microneedles to remove pro-inflammatory compounds Although a majority of microneedle technology uses the material for delivery, the NUS team explored the novel use of microneedles to extract undesirable pro-inflammatory proteins and immune cells in the second approach. To do so, the NUS team needed to find a suitable coating material that could act as a sponge to soak up pro-inflammatory compounds, known as chemokines, which are 'messenger' molecules that recruit and trap pro-inflammatory immune cells called monocytes in wound tissues. The research team screened different materials and eventually used heparin-coated porous microneedles (HPMN) to address the issue of persistent inflammation in skin wounds at the source. Based on previous studies, heparin has been found to bind readily to chemokines. The team demonstrated that HPMN could effectively deplete chemokines and monocytes from the wound site, leading to a 50 per cent reduction in tissue inflammation as well as a 90 per cent reduction in wound size by the 14th day of treatment. These initial findings highlight the potential of HPMN as a promising strategy for the treatment of inflammatory skin disorders. The ability of HPMN to remove chemokines and inflammatory cells deep within the skin tissue offers a unique advantage over existing treatments that only target surface-level inflammation. HPMN could be further developed for personalised wound care and tailored treatment of various inflammatory skin conditions such as psoriasis. Next steps The development of SUC-MN and HPMN represents a significant step forward in the field of wound healing and skin disease management. The team intends to conduct further studies to explore the potential of this technology and bring it to market. For extractive microneedles in particular, the team will fabricate microneedles with more controllable pore sizes using advanced technologies, such as 3D printing, and integrate antibacterial properties into the microneedles as clinical non-healing wounds often accompany infections. They are also designing flexible microneedle patches to ensure that they fit well to various tissue shapes. "We are excited about the potential impact of our research and look forward to advancing this technology towards clinical translation. The two approaches developed by our team would provide much-needed relief for patients with diabetic wounds, as well as many patients suffering from skin conditions like atopic dermatitis or psoriasis," said Asst Prof Tay. Read more at: https://news.nus.edu.sg/microneedle-technology-accelerate-diabetic-wound-healing.  

文章來源 : PR Newswire 美通社 發表時間 : 瀏覽次數 : 282 加入收藏 :
2025 年 3 月 31 日 (星期一) 農曆三月初三日
首 頁 我的收藏 搜 尋 新聞發佈