InSysBio to announce a new collaborative project with BeOne Medicines

On January 28, 2026 InSysBio, one of the world’s pioneers of Quantitative Systems Pharmacology (QSP) modeling, reported the extension of collaboration with BeOne Medicines, a global oncology company.

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!

In this project mechanistic translational modeling will be applied to support selection of minimal recommended starting dose and step-up dosing regimen to mitigate cytokine release syndrome and optimize dose-escalation clinical study design.

"Nowadays, trimer-based mechanistic modeling has become an essential tool to guide the selection of an appropriate starting dose for first-in-human trials of T-cell engagers, ensuring the dose is high enough to be effective while avoiding sub-therapeutic levels and multiple dose escalation steps," said Oleg Demin Jr, Head of Oncology and Immuno-Oncology, InSysBio. "InSysBio’s generic QSP model for T-cell engagers mechanistically captures the difference between in vitro and clinical settings, supporting precise preclinical-to-clinical translation. Consequently, it allows prediction of not only the starting dose but also the step-up dosing regimen based on preclinical data."

(Press release, BeOne Medicines, JAN 28, 2026, View Source [SID1234662335])

Calidi Biotherapeutics Provides Corporate Update and Key Value Drivers for 2026

On January 28, 2026 Calidi Biotherapeutics, Inc. (NYSE American: CLDI) ("Calidi" or the "Company"), a biotechnology company pioneering the development of systemically delivered, targeted genetic medicines, reported 2025 successes and provided an update on the Company’s upcoming 2026 milestones.

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!

"I am incredibly proud of the Calidi team for their successful execution of our 2025 priorities," said Eric Poma, PhD, Chief Executive Officer of Calidi. "We demonstrated that in clinically relevant immunocompetent murine models and ex vivo human immune assays, our RedTail platform has been engineered to prevent immune clearance after systemic administration and to only replicate in tumor cells with high specificity. Our data also demonstrate that the platform can effectively express genetic medicines at the tumor site at levels comparable to those achieved with localized dosing while avoiding systemic exposure. This reflects a scalable therapeutic approach that overcomes the limitations of tumor accessibility. I anticipate that we will complete our IND-enabling studies for CLD-401, the lead candidate from the RedTail platform, and submit an IND application by the end of this year."

2025 Accomplishments

First data presented on Calidi’s RedTail platform and selection of first lead candidate, CLD-401. CLD-401 is a tumor-tropic oncolytic virus designed to avoid immune clearance, home to metastatic sites after systemic administration, replicate only in tumors cells, induce an immune priming event at the tumor site, and express high levels of IL-15 superagonist, a potent cytokine that induces NK and T-cell responses to the tumor, in the tumor microenvironment (TME).
Presented new preclinical data surrounding CLD-401 at the American Society for Clinical Oncology (ASCO) (Free ASCO Whitepaper) on May 30, 2025, in Chicago, IL., demonstrating enhanced biological efficacy in clinically relevant immunocompetent tumor models through the delivery of IL-15 superagonist to metastatic tumors sites after systemic administration. Calidi also demonstrated the engineered expression of CD55 on the enveloped virus, allowing the virus to avoid immune clearance and enabling systemic administration. The systemic administration of the CD55-expressing enveloped virus allows Calidi to target metastatic disease patients with high unmet need.
CLD-401 data presented at the Society of Immunotherapy for Cancer (SITC) (Free SITC Whitepaper) on November 7, 2025, in National Harbor, MD., demonstrating that in syngeneic murine models, the RedTail platform is protected from immune clearance after systemic administration and can find and specifically replicate in tumor cells at metastatic sites. The data also demonstrate that the platform can effectively express genetic medicines at the tumor site in concentrations that are similar to what is achievable with localized dosing while avoiding systemic exposure.
Bolstered management team with hiring of new Chief Executive Officer and Chief Medical Officer with deep biopharmaceutical experience
CEO transition to Eric Poma, PhD, bringing more than 25 years of experience in the biopharmaceutical industry, with a strong record of capital fundraising, big pharma collaboration agreements, and clinical program development.
Appointed Guy Travis Clifton, MD, as CMO. Dr. Clifton is a practicing surgical oncologist with over 17 years experience in drug development, early phase and transitional translational trials, and cancer immunotherapy.
Reconstituted Scientific Advisory Board with internationally esteemed advisors to support development of CLD-401 and advance the RedTail platform for the systemic delivery of targeted genetic medicine.
New members added in October 2025:

Mace L. Rothenberg, MD, FACP, former Chief Medical Officer of Pfizer, a physician executive with more than 30 years of experience in drug development, translational research, and risk benefit assessment; and
John Wrangle, MD, MPH, a thoracic oncologist and scientist and expert in translational immunotherapy with extensive experience around IL-15-based treatment in metastatic cancer.
Reduced debt and G&A expenses
Calidi reduced term debt and notes payable (including accrued interest) by $3.1 million in 2025, from $3.8 million at December 31, 2024 to $0.7 million at December 31, 2025 (unaudited).
Calidi reduced G&A expenses by $2.3 million in the first nine months of 2025 compared to the first nine months of 2024, as disclosed in the Company’s Form 10-Q filed with the SEC on November 13, 2025.
2026 Anticipated Milestones

Company intends to file an IND in Q4 2026 for its first RedTail lead candidate, CLD-401, a systemic delivered, targeted genetic medicine engineered to convert tumors into IL-15 superagonist producers.
Phase I study expected to be conducted in a basket of solid tumors, including non-small cell lung cancer, triple-negative breast cancer, and head and neck cancer.
Anticipate streamlined dose escalation study with limited number of doses to be tested.
Initial dose cohort is expected to be in the therapeutic range with the potential for proof-of-concept data early in phase I
Calidi expects to present proof of concept data demonstrating the versatility of RedTail platform to deliver tumor-localized Bi-specific T cell engager (BiTE) alongside T-cell amplifiers.
BiTEs have struggled to drive efficacy in solid tumors because of a lack of activated T-cells in the TME
The RedTail platform allows for the simultaneous delivery of high expression of multiple payloads into TME
BiTE delivery with T-cell amplifiers may overcome the previous limitations of BiTEs in solid tumors
Calidi expects to present proof of concept data for use of RedTail platform into non-oncology indications.
Calidi is exploring new payloads for inflammatory and immune disease
The Company anticipates targeting other cell types via envelope engineering (e.g. CD38, BCMA, etc.)
Calidi expects to leverage selective viral replication in proliferative cells (e.g. activated B cells)

(Press release, Calidi Biotherapeutics, JAN 28, 2026, View Source [SID1234662320])

QureBio Ltd. to Present at ASCO 2026 its Q-1802 Phase II Clinical Results

On January 28, 2026 QureBio Ltd., a clinical-stage biotech company focusing on development of bispecific antibodies and other engineered Biopharmaceuticals for the treatment of cancer, inflammation, and other serious disorders, reported in 2026 ASCO (Free ASCO Whitepaper) abstract submission about the Phase Ib/II Clinical Data of its Q-1802 program.

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 report(Abstract #537934)is titled "PhaseⅠ/Ⅱ Trial of Anti-CLDN18.2/PD-L1 Recombinant Humanized Bispecific Antibody Q-1802 Plus XELOX in Treatment-Naive CLDN18.2-Positive Advanced GC/GEJ"

The authors, Dr. Gong et al, reported the followings : the study enrolled 62 Eligible pts: CLDN18.2-positive (≥40% tumor cells with 2+/3+ membranous staining), HER2-negative, treatment-naïve, histologically confirmed unresectable locally advanced/metastatic GC/GEJ. Pts received Q-1802 (10 mg/kg or 20 mg/kg Q2W) plus standard XELOX.

In this study, no DLT observed; MTD not reached. Most common Q-1802-related ≥3 Grade TEAE: thrombocytopenia (8.1%), followed by neutropenia (6.5%), anemia, WBC decrease, hypokalemia (4.8% each). Rate of Q-1802 permanent discontinuation due to TEAEs: 6.5%; no treatment-related death.

In all 60 efficacy-evaluable pts, ORR, DCR and mPFS were 70.0% (42/60), 98.3% (59/60), and 11.3 months respectively. In the 10 mg/kg cohort, ORR and mPFS were 73.0% (27/37) and 11.3 months in pts with CLDN18.2 high expression; ORR and mPFS were improved to be 81.8% and 12.2 months when CLDN18.2 high expression accompanied with PD-L1 CPS≥5 (N=11).

The authors, Dr. Gong et al, concluded that Q-1802 plus XELOX has manageable safety and promising antitumor activity as first-line therapy for CLDN18.2-positive/HER2-negative advanced GC/GEJ, supporting a phase III trial with Q-1802 10 mg/kg as recommended dose.

Q-1802 phase III trial has recently been approved by Chinese CDE.

About Q-1802

Q-1802, a humanized Claudin18.2/PD-L1 bispecific antibody, kills tumors by both innate immunity such as antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cell-mediated phagocytosis (ADCP) as well as adaptive immunity involving DC and T cells. It is a powerful agent with good safety profile, offering a novel therapeutic opportunity to patients with Claudin18.2 positive solid tumors. Q-1802 exhibits high affinity and selectivity.

(Press release, QureBio, JAN 28, 2026, View Source [SID1234662336])

Defence Therapeutics Expands Collaboration With Canadian Nuclear Laboratories To Accelerate Its Proprietary Radiopharmaceutical Pipeline

On January 28, 2026 Defence Therapeutics Inc. ("Defence" or the "Company"), a publicly traded biotechnology and precision intracellular drug-delivery company, reported the strengthening of its long-standing collaboration with Canadian Nuclear Laboratories ("CNL") as part of its strategic effort to accelerate and expand its proprietary radiopharmaceutical program.

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!

Defence has been collaborating with CNL on the development of an Accum-enhanced radio-immunoconjugate program. This program is now approaching a key inflection point, with critical results being generated by both lab teams on this program that are intended to support candidate selection and enable preparation for first-in-human clinical studies.

Based on the exciting traction on the lead program, Defence and CNL are expanding their collaboration to initiate multiple other radioisotopes programs with Defence’s proprietary Accum, positioning the Company to rapidly expand its radiopharmaceutical portfolio as new data and strategic priorities emerge.

Radiopharmaceutical therapies require precise intracellular and nuclear localization to maximize efficacy, as radioactive payloads must reach the vicinity of the cell nucleus to induce lethal DNA damage. Defence’s proprietary Accum platform enables active intracellular and nuclear transport, unlocking the full therapeutic potential of targeted radiotherapies while increasing potency and reducing toxicity.

"This collaboration with CNL has become a cornerstone of our radiopharmaceutical strategy," said Sébastien Plouffe, CEO of Defence Therapeutics. "Radiopharmaceuticals now represent one of our top development priorities alongside our ADC program. By deepening our work with CNL and leveraging their world-class nuclear science expertise, we are building a strong, proprietary pipeline in this space and accelerating our path toward the clinic."

"We have worked in close collaboration with Defence on their flagship program, delivering the technical results they require, and we are proud to contribute meaningfully to the advancement of their radiopharmaceutical pipeline," said Dr. Monica Regalbuto, Vice-President, Science & Technology at CNL. "We look forward to contributing our nuclear science expertise to help advance these programs toward the clinic."

This collaboration further strengthens Defence’s position in precision intracellular drug delivery and radiopharmaceutical development, supporting its strategy to transform advanced biologics into safer, more effective first-line cancer therapies.

(Press release, Defence Therapeutics, JAN 28, 2026, View Source;utm_medium=rss&utm_campaign=defence-therapeutics-expands-collaboration-with-canadian-nuclear-laboratories-to-accelerate-its-proprietary-radiopharmaceutical-pipeline [SID1234662321])

Caris Life Sciences’ Real-World Data Uncovers Metastatic Breast Cancer Patient Responses and Resistance to Trastuzumab Deruxtecan

On January 28, 2026 Caris Life Sciences (NASDAQ: CAI), a leading, patient-centric, next-generation AI TechBio company and precision medicine pioneer, reported a study in Nature’s npj Breast Cancer journal titled, "Mechanisms of Resistance to Trastuzumab Deruxtecan in Breast Cancer Elucidated by Multiomic Molecular Profiling." Utilizing large-scale real-world clinico-genomic data and Caris’s comprehensive multiomic profiling, Caris scientists uncovered clinically relevant mechanisms of resistance to trastuzumab deruxtecan (T-DXd) in metastatic breast cancer. This study explains why patient responses vary and reveals how resistance can evolve over time.

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!

This study addresses a critical gap in understanding why responses to T–DXd, an antibody–drug conjugate approved by the FDA for HER2-positive and HER2-low metastatic breast cancer, vary widely since resistance to T–DXd is common. By combining large-scale, real-world clinical outcomes with Caris’s comprehensive molecular profiling, including whole exome sequencing (WES) and whole transcriptome sequencing (WTS), the study enables a population-level analysis of resistance pathways across DNA, RNA and protein expression.

"In this large real-world analysis, clinical outcomes allowed Caris to pinpoint molecular features linked to treatment-specific survival," said George W. Sledge, Jr., MD, Caris EVP and Chief Medical Officer. "Post–treatment molecular shifts further reveal biologically plausible routes to acquired resistance, underscoring that RNA–level and multiomic profiling can provide actionable insights beyond standard HER2 classification to better stratify patients and inform therapies."

The study analyzed 2,799 T–DXd–treated breast cancer patients and found that WTS identified ERBB2 (HER2) and ABCC1 as the strongest transcriptomic predictors of T–DXd–specific overall survival. Higher ERBB2 expression correlated with improved outcomes, while higher ABCC1 correlated with poorer outcomes, independent of HER2 category.

ABCC1 further stratified outcomes within HER2–defined subgroups, indicating predictive value beyond standard HER2 testing and post–treatment samples showed increased ABCC1 expression alongside enrichment of mutations in ERBB2, NFE2L2, KEAP1 and TOP1. Consistent with acquired resistance mechanisms. Preclinical models supported a functional, context–dependent role for ABCC1–mediated drug efflux in T–DXd resistance.

"This study demonstrates how population–scale, multiomic real–world data can drive high impact translational discoveries, reinforcing the value proposition for patients and equipping biopharma with actionable insights into resistance biology to guide next–generation drug development," said David Spetzler, MS, PhD, MBA, Caris President.

(Press release, Caris Life Sciences, JAN 28, 2026, View Source [SID1234662337])