SELLAS Life Sciences Group to Host Virtual R&D Day on October 29, 2025: Advancing Novel Therapies in Acute Myeloid Leukemia (AML): An Overview of the Ongoing Phase 3 REGAL Trial of Galinpepimut-S (GPS) and SLS009 Program Update

On September 30, 2025 SELLAS Life Sciences Group, Inc. (NASDAQ: SLS) ("SELLAS" or the "Company"), a late-stage clinical biopharmaceutical company focused on the development of novel therapies for a broad range of cancer indications, reported that it will host a virtual R&D Day on Wednesday, October 29, 2025 at 10:00 AM ET featuring key opinion leaders (KOLs), alongside company management, to discuss the unmet medical need and evolving treatment landscape for acute myeloid leukemia (AML) (Press release, Sellas Life Sciences, SEP 30, 2025, View Source [SID1234656352]).

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

The event will feature an overview of the ongoing Phase 3 REGAL trial of GPS (results expected by year-end) and a discussion of the unmet needs of AML patients in complete second remission (CR2). SELLAS will also present an update of SLS009, its highly selective cyclin-dependent kinase 9 (CDK9) inhibitor, highlighting recently reported Phase 2 data and plans for a newly diagnosed and frontline AML study anticipated to begin in the first quarter of 2026.

A live Q&A session will follow the formal presentations.

About Omer Jamy, MD
Omer Jamy, MD is Assistant Professor of Medicine at the University of Alabama (UAB) in the Division of Hematology and Oncology and Associate Scientist, Experimental Therapeutics at the O’Neal Comprehensive Cancer Center. He serves as principal investigator of the Phase 3 REGAL study at UAB, one of the trial’s highest enrolling sites, and leads several clinical trials in addition to REGAL, focusing on AML, chronic myelogenous leukemia, and allogeneic stem cell transplantation. Dr. Jamy completed his internal medicine residency at the University of Tennessee in Memphis followed by fellowship training at UAB in hematology/oncology, bone marrow transplantation and cellular therapy.

About Panagiotis Tsirigotis, MD, PhD
Panagiotis Tsirigotis, MD, PhD is Professor of Hematology at the National and Kapodistrian University of Athens, School of Medicine in Athens, Greece, and Scientific Director of the Transplantation Program of the Hematology Unit since 2010. He is an investigator in the Phase 3 REGAL trial and has enrolled the highest number of patients in the study. His clinical work focuses on AML with particular emphasis on cellular therapies and hematopoietic cell transplantation. Dr. Tsirigotis’ research focuses on the application of immunotherapy methods to prevent leukemia relapse after allogeneic transplantation, such as the administration of donor lymphocytes, as well as on mechanisms of immune escape in hematologic malignancies. He is the president of Acute Leukemia Working Party of the Hellenic Society of Hematology, and Vice President of the Hellenic Transplant Organization.

About Philip Amrein, MD
Philip Amrein, MD is Assistant Professor of Medicine at Harvard Medical School and a physician at the Massachusetts General Hospital (MGH), where he is part of the Cancer Center, Leukemia, Cellular Immunotherapy, and Hematology/Oncology departments. Dr. Amrein specializes in treating adults with acute and chronic leukemias, myelodysplasia, and myeloproliferative neoplasms, and leads numerous clinical trials exploring novel treatment approaches. Dr. Amrein has conducted research with SLS009 and is an expert in cyclin-dependent kinases (CDK).

About Sharif Khan, MD
Sharif Khan, MD is a hematologist at Bon Secours Health System in Greenville, SC. He is a highly rated specialist in indications such as AML, myeloproliferative neoplasms, multiple myeloma, non-Hodgkin lymphoma, and bone marrow transplantation. In addition to clinical care, Dr. Khan is a researcher for cutting-edge, breakthrough therapies and started a CAR T-Cell program at Bon Secours Health System. Dr. Khan serves as an investigator in both the REGAL trial of GPS and the SLS009 clinical program.

Star Therapeutics Announces Oversubscribed $125 Million Series D Financing

On September 30, 2025 Star Therapeutics, a clinical stage biotechnology company discovering and developing best-in-class antibodies for bleeding disorders and other diseases, reported an oversubscribed $125 million Series D financing (Press release, Star Therapeutics, SEP 30, 2025, View Source [SID1234656350]). The financing round was co-led by Sanofi Ventures and Viking Global Investors with participation from both existing and other new investors, including Janus Henderson Investors, Frazier Life Sciences and GordonMD Global Investments. Existing investors include Agent Capital, Blue Owl Capital, Catalio Capital Management, Cormorant Asset Management, New Leaf Venture Partners, NYBC Ventures, OrbiMed, Qatar Investment Authority (QIA), RA Capital Management, Redmile Group, Sofinnova Investments, Soleus Capital and Westlake BioPartners.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

Proceeds from the Series D financing will support the continued advancement of Star’s pipeline including its lead program, VGA039, a first-in-class monoclonal antibody that targets Protein S to restore balance in blood clotting. VGA039 is being developed as a universal hemostatic therapy for multiple bleeding disorders, starting with von Willebrand disease (VWD). Star has initiated a pivotal Phase 3 clinical trial of VGA039 in patients with all types of VWD, designed to evaluate VGA039 administered once monthly via subcutaneous injection.

"We’re making significant progress across our pipeline, highlighted by the recent initiation of our Phase 3 trial of VGA039 to prevent bleeding in people with all types of VWD, the most common inherited bleeding disorder, with more than 50,000 patients diagnosed and treated in the U.S.," said Adam Rosenthal, Ph.D., CEO and Founder of Star Therapeutics. "VGA039 has the potential to be transformative for VWD patients and could meaningfully reduce the treatment burden, given its once monthly subcutaneous dosing regimen, in comparison to factor replacement prophylaxis which requires two to three IV infusions per week. This new funding from leading life sciences investors reinforces the urgency and impact of our mission: to bring life-changing therapies to people living with serious bleeding disorders and other diseases in hematology and immunology."

In conjunction with the financing, Jason Hafler, Ph.D., from Sanofi Ventures will join Star’s board of directors, and Maneka Mirchandaney of Viking Global Investors will join as a board observer.

"At Sanofi Ventures, we’re proud to bring meaningful experience in supporting innovative therapies for bleeding disorders and remain deeply committed to exploring best-in-class therapies to better meet the needs of patients. VWD represents a compelling opportunity, with a sizable patient population and significant unmet need," said Jason Hafler, Ph.D., Managing Director at Sanofi Ventures. "With VGA039 advancing into Phase 3 for VWD, Star is well-positioned to deliver both substantial patient impact and long-term value. We’re proud to support a team with a strong track record and a bold vision for transforming care in VWD and beyond."

About VGA039
VGA039 is a monoclonal antibody therapy with a novel mechanism of action that targets Protein S, thereby restoring balance to the blood clotting process. VGA039 has potential to be a universal hemostatic therapy that can treat numerous bleeding disorders, starting with VWD. As a subcutaneously self-administered antibody therapy with a convenient once monthly dosing regimen, VGA039 has the potential to dramatically reduce treatment burden for patients. VGA039 has received Fast Track and orphan drug designations from the United States Food and Drug Administration (FDA).

Interim positive data from VIVID-2, a Phase 1 single ascending dose study of VGA039 in patients with VWD, were previously reported at the Annual Meeting of the American Society of Hematology (ASH) (Free ASH Whitepaper) in December 2024. A Phase 1/2 multidose study, VIVID-3, is ongoing (NCT05776069). VGA039 has advanced into a Phase 3 study, VIVID-6 (NCT07115004), a global single arm cross-over study designed to investigate the safety and efficacy of subcutaneous administration of VGA039 as prophylaxis for bleeding in patients with every type of VWD. For additional information on VIVID-6, including how to enroll, please visit the website here.

About von Willebrand disease
Von Willebrand disease (VWD) is the most common inherited bleeding disorder in which the blood does not clot properly, caused by absent or defective von Willebrand factor (VWF). VWD patients may experience excessive bleeding with variability in severity and frequency, negatively impacting their daily lives. Current therapies for VWD prophylaxis include factor replacement therapies requiring multiple intravenous (IV) infusions every week. More than 50,000 people in the United States are estimated to be receiving treatment for VWD.

Soligenix Announces Expansion of European Medical Advisory Board for Cutaneous T-Cell Lymphoma

On September 30, 2025 Soligenix, Inc. (Nasdaq: SNGX) (Soligenix or the Company), a late-stage biopharmaceutical company focused on developing and commercializing products to treat rare diseases where there is an unmet medical need, reported the expansion of its European Medical Advisory Board (MAB) to provide additional medical/clinical strategic guidance to the Company as it advances its confirmatory Phase 3 multicenter, double-blind, placebo-controlled study evaluating the safety and efficacy of HyBryte (synthetic hypericin) in the treatment of cutaneous T-cell lymphoma (CTCL) patients with early-stage disease (Press release, Soligenix, SEP 30, 2025, View Source [SID1234656349]). This confirmatory, 18-week study is expected to enroll approximately 80 patients and is targeted to report top-line results in the second half of 2026.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"We are excited to expand our European MAB to include Drs. Scarisbrick and Vermeer who are each esteemed dermatologists that will bring valuable guidance to our program," stated Christopher J. Schaber, PhD, President and Chief Executive Officer of Soligenix. "We consider expert European involvement a necessary component for the development and approval of HyBryte in both the European Union (EU) and the United Kingdom (UK). As such, it is a privilege to expand the MAB to five members that are highly-respected leaders in their field and committed to working with us as we advance HyBryte towards commercialization. We look forward to working with the MAB and reporting top-line results for the confirmatory Phase 3 study in 2026."

Comprised of internationally renowned physicians with extensive experience in treating and running clinical research trials in CTCL, this esteemed expanded MAB will play an important advisory role in the conduct and interpretation of the upcoming Phase 3 clinical study and the associated regulatory interactions with health authorities. The MAB will provide expert feedback, input, and guidance on clinical strategies and their implementation, as well as on other critical items, such as health economics and reimbursement, to assist Soligenix in meeting the needs of patients suffering from CTCL.

European MAB Members

Martine Bagot, MD, PhD – France

Martine Bagot is Professor and Head of the Department of Dermatology at the Hôpital Saint Louis in Paris, France and co-directs the INSERM Unit 976 Human Immunology, Pathophysiology and Immunotherapy. Dr. Bagot also chairs the French Group for the Study of Cutaneous Lymphomas. Dr. Bagot has co-authored more than 750 peer-reviewed publications, most of which are in the complementary fields of dermatology, immunology, and oncology. Main publications include numerous clinical trials in dermato-oncology. Dr. Bagot is involved with major European international societies such as the International Society for Cutaneous Lymphoma (ISCL), The European Society for Dermatological Research (ESDR), and the European Organisation for Research and Treatment of Cancer (EORTC) Cutaneous Lymphoma Task Force, where she previously served as its President and is currently a steering committee member.

Pietro Quaglino, MD – Italy

Pietro Quaglino is Associate Professor of Dermatology at the Department of Medical Sciences, University of Turin Medical School, Italy. His clinical and research activities focus on melanoma, cutaneous lymphoma, and immune dermatology. He is the principal investigator of several clinical trials in melanoma and cutaneous lymphoma. He is board member of the GIPMe (Gruppo Italiano Polidisciplinare sul Melanoma), board member and treasurer of the IMI (Italian Melanoma Intergroup), past chairman and current steering committee member of the EORTC Cutaneous Lymphoma Task Force, and member of the Board Directors of the ISCL. He has published more than 160 peer-reviewed scientific papers. Dr. Quaglino is Assistant Editor of the Giornale Italiano di Dermatologia e Venereologia, reviewer of international journals on dermatology and referee for ANVUR (National Agency for the Evaluation of Universities and Research Institutes).

Pablo Luis Ortiz-Romero, MD, PhD – Spain

Pablo Luis Ortiz-Romero is Professor of Dermatology and Head of the Dermatology Department at Hospital Universitario 12 de Octubre, Spain. He is a distinguished dermatologist and researcher specializing in cutaneous lymphomas, particularly mycosis fungoides and Sézary syndrome forms of CTCL. His experience is extensive, having contributed to over 200 publications and numerous clinical studies in the field. Dr. Ortiz-Romero is affiliated with the ISCL and has served on its Board of Directors. His research includes innovative treatments for CTCL, and he is one of the leading physicians focused on novel treatments for CTCL in Spain. He served as the Secretary General of the EORTC Cutaneous Lymphoma Tumor Group and is currently a member of its steering committee.

Julia Scarisbrick, MBhons, ChB, FRCP, MD – United Kingdom

Julia Scarisbrick leads the Specialist Cutaneous Lymphoma Service at University Hospital Birmingham, UK. She holds an honorary Chair as Professor for the Institute of Immunology and Immunotherapy and Dermatology Co-Lead for Dermatology Research Group, Clinical Sciences, University of Birmingham. Prof. Scarisbrick is an internationally renowned dermatologist in the UK specializing in clinical and molecular research involving cutaneous lymphomas with over 150 publications as well as a number of book chapters to her name. She is the President of the ISCL, Past Chairperson of the EORTC Cutaneous Lymphoma Group, the European Director for Cutaneous Lymphoma International Consortium, the Chairperson and Trustee for the UK Photopheresis Society and Treasurer and Trustee for the UK Cutaneous Lymphoma Group (Past Chair).

Maarten H. Vermeer, MD, PhD – The Netherlands

Maarten H. Vermeer is the Head of the Department of Dermatology of the Leiden University Medical Center (LUMC). Dr. Vermeer has been researching the pathogenesis and treatment of cutaneous lymphomas for more than 25 years. He has more than 175 scientific publications to his name. Recently, his research activities have concentrated on clinicopathologic studies, genomic analysis of genetic and epigenetic alterations in cutaneous lymphoma tumor cells as well as international collaborative studies to develop diagnostic markers and standardize flowcytometry in cutaneous lymphomas. Dr. Vermeer served on the board of the European Society Dermatological Research, and the EORTC Cutaneous Lymphoma Working Group and chaired the board of the ISCL.

About HyBryte

HyBryte (research name SGX301) is a novel, first-in-class, photodynamic therapy utilizing safe, visible light for activation. The active ingredient in HyBryte is synthetic hypericin, a potent photosensitizer that is topically applied to skin lesions that is taken up by the malignant T-cells, and then activated by safe, visible light approximately 24 hours later. The use of visible light in the red-yellow spectrum has the advantage of penetrating more deeply into the skin (much more so than ultraviolet light) and therefore potentially treating deeper skin disease and thicker plaques and lesions. This treatment approach avoids the risk of secondary malignancies (including melanoma) inherent with the frequently employed DNA-damaging drugs and other phototherapy that are dependent on ultraviolet exposure. Combined with photoactivation, hypericin has demonstrated significant anti-proliferative effects on activated normal human lymphoid cells and inhibited growth of malignant T-cells isolated from CTCL patients. In a published Phase 2 clinical study in CTCL, patients experienced a statistically significant (p=0.04) improvement with topical hypericin treatment whereas the placebo was ineffective. HyBryte has received orphan drug and fast track designations from the FDA, as well as orphan designation from the European Medicines Agency (EMA).

The published Phase 3 FLASH trial enrolled a total of 169 patients (166 evaluable) with Stage IA, IB or IIA CTCL. The trial consisted of three treatment cycles. Treatments were administered twice weekly for the first 6 weeks and treatment response was determined at the end of the 8th week of each cycle. In the first double-blind treatment cycle (Cycle 1), 116 patients received HyBryte treatment (0.25% synthetic hypericin) and 50 received placebo treatment of their index lesions. A total of 16% of the patients receiving HyBryte achieved at least a 50% reduction in their lesions (graded using a standard measurement of dermatologic lesions, the CAILS score) compared to only 4% of patients in the placebo group at 8 weeks (p=0.04) during the first treatment cycle (primary endpoint). HyBryte treatment in this cycle was safe and well tolerated.

In the second open-label treatment cycle (Cycle 2), all patients received HyBryte treatment of their index lesions. Evaluation of 155 patients in this cycle (110 receiving 12 weeks of HyBryte treatment and 45 receiving 6 weeks of placebo treatment followed by 6 weeks of HyBryte treatment), demonstrated that the response rate among the 12-week treatment group was 40% (p<0.0001 vs the placebo treatment rate in Cycle 1). Comparison of the 12-week and 6-week treatment responses also revealed a statistically significant improvement (p<0.0001) between the two timepoints, indicating that continued treatment results in better outcomes. HyBryte continued to be safe and well tolerated. Additional analyses also indicated that HyBryte is equally effective in treating both plaque (response 42%, p<0.0001 relative to placebo treatment in Cycle 1) and patch (response 37%, p=0.0009 relative to placebo treatment in Cycle 1) lesions of CTCL, a particularly relevant finding given the historical difficulty in treating plaque lesions in particular.

The third (optional) treatment cycle (Cycle 3) was focused on safety and all patients could elect to receive HyBryte treatment of all their lesions. Of note, 66% of patients elected to continue with this optional compassionate use / safety cycle of the study. Of the subset of patients that received HyBryte throughout all 3 cycles of treatment, 49% of them demonstrated a positive treatment response (p<0.0001 vs patients receiving placebo in Cycle 1). Moreover, in a subset of patients evaluated in this cycle, it was demonstrated that HyBryte is not systemically available, consistent with the general safety of this topical product observed to date. At the end of Cycle 3, HyBryte continued to be well tolerated despite extended and increased use of the product to treat multiple lesions.

Overall safety of HyBryte is a critical attribute of this treatment and was monitored throughout the three treatment cycles (Cycles 1, 2 and 3) and the 6-month follow-up period. HyBryte’s mechanism of action is not associated with DNA damage, making it a safer alternative than currently available therapies, all of which are associated with significant, and sometimes fatal, side effects. Predominantly these include the risk of melanoma and other malignancies, as well as the risk of significant skin damage and premature skin aging. Currently available treatments are only approved in the context of previous treatment failure with other modalities and there is no approved front-line therapy available. Within this landscape, treatment of CTCL is strongly motivated by the safety risk of each product. HyBryte potentially represents the safest available efficacious treatment for CTCL. With very limited systemic absorption, a compound that is not mutagenic and a light source that is not carcinogenic, there is no evidence to date of any potential safety issues.

Following the first Phase 3 study of HyBryte for the treatment of CTCL, the FDA and the EMA indicated that they would require a second successful Phase 3 trial to support marketing approval. With agreement from the EMA on the key design components, the second, confirmatory study, called FLASH2, is ongoing. This study is a randomized, double-blind, placebo-controlled, multicenter study that will enroll approximately 80 subjects with early-stage CTCL. The FLASH2 study replicates the double-blind, placebo-controlled design used in the first successful Phase 3 FLASH study that consisted of three 6-week treatment cycles (18 weeks total), with the primary efficacy assessment occurring at the end of the initial 6-week double-blind, placebo-controlled treatment cycle (Cycle 1). However, this second study extends the double-blind, placebo-controlled assessment to 18 weeks of continuous treatment (no "between-Cycle" treatment breaks) with the primary endpoint assessment occurring at the end of the 18-week timepoint. In the first Phase 3 study, a treatment response of 49% (p<0.0001 vs patients receiving placebo in Cycle 1) was observed in patients completing 18 weeks (3 cycles) of therapy. In this second study, all important clinical study design components remain the same as in the first FLASH study, including the primary endpoint and key inclusion-exclusion criteria. The extended treatment for a continuous 18 weeks in a single cycle is expected to statistically demonstrate HyBryte’s increased effect over a more prolonged, "real world" treatment course. Given the extensive engagement with the CTCL community, the esteemed Medical Advisory Board and the previous trial experience with this disease, accelerated enrollment in support of this study is anticipated, including the potential to enroll previously identified and treated HyBryte patients from the FLASH study. Discussions with the FDA on an appropriate study design remain ongoing. While collaborative, the agency has expressed a preference for a longer duration comparative study over a placebo-controlled trial. Given the shorter time to potential commercial revenue and the similar trial design to the first FLASH study afforded by the EMA accepted protocol, this study is being initiated. At the same time, discussions with the FDA will continue on potential modifications to the development path to adequately address their feedback.

Additional supportive studies have demonstrated the utility of longer treatment times with a 75% response rate after 18 weeks treatment (Study RW-HPN-MF-01), the lack of significant systemic exposure to hypericin after topical application (Study HPN-CTCL-02) and its relative efficacy and tolerability compared to Valchlor (Study HPN-CTCL-04).

In addition, the FDA awarded an Orphan Products Development grant to support the investigator-initiated study evaluation of HyBryte for expanded treatment in patients with early-stage CTCL, including in the home use setting. The grant, totaling $2.6 million over 4 years, was awarded to the University of Pennsylvania that was a leading enroller in the Phase 3 FLASH study.

About Cutaneous T-Cell Lymphoma (CTCL)

CTCL is a class of non-Hodgkin’s lymphoma (NHL), a type of cancer of the white blood cells that are an integral part of the immune system. Unlike most NHLs which generally involve B-cell lymphocytes (involved in producing antibodies), CTCL is caused by an expansion of malignant T-cell lymphocytes (involved in cell-mediated immunity) normally programmed to migrate to the skin. These malignant cells migrate to the skin where they form various lesions, typically beginning as patches and may progress to raised plaques and tumors. Mortality is related to the stage of CTCL, with median survival generally ranging from about 12 years in the early stages to only 2.5 years when the disease has advanced. There is currently no cure for CTCL. Typically, CTCL lesions are treated and regress but usually return either in the same part of the body or in new areas.

CTCL constitutes a rare group of NHLs, occurring in about 4% of the more than 1.7 million individuals living with the disease in the U.S. and Europe (European Union and United Kingdom). It is estimated, based upon review of historic published studies and reports and an interpolation of data on the incidence of CTCL that it affects approximately 31,000 individuals in the U.S. (based on SEER data, with approximately 3,200 new cases seen annually) and approximately 38,000 individuals in Europe (based on ECIS prevalence estimates, with approximately 3,800 new cases annually).

Labcorp Announces Collaboration with Roche to Advance Digital Pathology Capabilities

On September 30, 2025 Labcorp (NYSE: LH), a global leader of innovative and comprehensive laboratory services, reported a collaboration with Roche to implement the company’s FDA-cleared VENTANA DP 600 and DP 200 slide scanners (Press release, LabCorp, SEP 30, 2025, View Source [SID1234656346]). The investment enables pathologists to diagnose patients using digital images and supports future artificial intelligence (AI) integration.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"Labcorp is committed to advancing its diagnostic capabilities through the adoption of innovative technologies," said Marcia Eisenberg, Ph.D., Labcorp’s chief scientific officer. "Integrating digital slide scanning into our pathology workflow will help improve accuracy, streamline operations and enable future AI-driven insights."

Labcorp’s initiative comes at a critical time for the field of pathology. The U.S. faces a projected shortage of nearly 5,700 pathologists by 2030, while the demand for diagnostic services continues to grow due to an aging population and the increasing incidence of cancer and other diseases.

The digital pathology workflow converts glass slides into high-resolution digital images that can be instantly viewed using a conventional PC workstation (rather than a microscope) and analyzed and shared from any location, eliminating reliance on physical slides and enabling pathologists to operate across different sites.

Key Features and Benefits of Digital Pathology:

Improved access to expertise: Enables remote sharing of digital slides, eliminating reliance on physical slides and enabling rapid consultations with specialists, regardless of location.
Enhanced efficiency: Streamlines workflows by eliminating the need to handle, transport or retrieve physical slides.
Reduced cost and risk: Digital storage eliminates transit costs and minimizes the risk of slide damage or loss of irreplaceable tissue samples.
AI integration: Establishes a foundation for AI tools that can improve consistency, accuracy and efficiency in pathology workflows.
Labcorp’s investment builds on the company’s existing relationship with Roche and its digital pathology portfolio for the company’s Oncology business.

"We look forward to expanding our collaboration with Labcorp to advance the digital transformation of their pathology labs," said Michael Rivers, life cycle leader for digital pathology at Roche Tissue Diagnostics. "This collaboration, which combines Labcorp’s strength in diagnostic services with Roche’s expertise in digital pathology solutions, holds the potential to create new opportunities for highly efficient, timely and effective patient care."

This latest collaboration builds on Labcorp’s commitment to expanding digital capabilities and delivering innovative solutions across its Diagnostics and Biopharma Laboratory Services (BLS) business segments. Labcorp teams use digital pathology technology globally to support clinical trials, enabling more efficient data sharing, centralized review and faster decision-making.

KYORIN and Hinge Bio Enter into a Collaboration and License Agreement of HB2198 in Japan for SLE and other diseases

On September 30, 2025 KYORIN Pharmaceutical Co., Ltd. reported that it has entered into an exclusive license agreement with Hinge Bio Inc. (BURLINGAME, CA, CEO:Harold E. "Barry" Selick, "Hinge Bio") on September 30, 2025, for HB2198, new drug candidate for systemic lupus erythematosus (SLE), discovered by Hinge Bio (Press release, Kyorin, SEP 30, 2025, View Source [SID1234656345]).

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

Under the terms of this agreement, KYORIN has obtained the exclusive rights in Japan to develop and commercialize HB2198 for the treatment of SLE and other diseases. KYORIN will make an upfront payment of USD 10 million to Hinge Bio. Hinge Bio is also eligible to receive milestone payments of up to USD 95 million for the SLE indication upon the progress of development, approval and the achievement of certain net sales. For indications other than SLE, and separate payment will be made. In addition, royalties based on net sales will be provided to Hinge Bio, and KYORIN will bear a portion of the clinical development costs incurred by Hinge Bio.

Autoimmune diseases such as SLE are caused by an aberrant immune system that mistakenly identifies the body’s own tissues as foreign, leading to excessive immune responses that cause chronic inflammation and organ damage. Many patients cannot get their symptoms under control with existing treatments and suffer from conditions such as joint pain, skin problems, kidney damage, and central nervous system issues.

HB2198 is a humanized bispecific antibody discovered using Hinge Bio’s proprietary GEM-DIMER technology platform. It aims to "reset" the immune system by simultaneously targeting both CD19 and CD20 molecules on the surface of B cells, which play a major role in the development of autoimmune diseases, and by rapidly and deeply depleting B cells in both the circulating blood and lymphoid tissues. Preclinical in vivo studies have demonstrated HB2198’s deep and rapid B cell depletion activity, and it is expected to provide an excellent new treatment option for patients suffering from autoimmune diseases. A Phase 1 clinical trial for patients with SLE is currently being prepared in the United States.

KYORIN aims to create high-value new drugs that meet medical needs and has prioritized "Expand development pipeline" under its medium-term business plan "Vision 110 —Stage1—". With this agreement, we are committed to advancing the development of new treatment options for patients as soon as possible, contributing to people’s health.

The impact on the consolidated earnings forecast for the fiscal year ending March 2026 is expected to include the upfront payment to Hinge Bio under this agreement, which will be recorded as SG&A expenses (R&D expenses) for the current fiscal year.