Antibodies for Cancer Therapy
Driving Breakthrough Therapies
5/11/2026 - May 12, 2026 ALL TIMES EDT
Cambridge Healthtech Institute’s 16th Annual Antibodies for Cancer Therapy conference delves into the rapidly advancing field of antibody engineering and its pivotal role in transforming cancer treatment. This year’s program spotlights the latest innovations in T cell engagers, ADCs, bispecifics, and emerging scaffold designs that expand the boundaries of therapeutic potential. Sessions will explore comparisons of TCEs with ADCs and CAR-Ts, strategies to enhance antigen selectivity and anti-tumor activity, and novel dual-targeting and conditional or masking approaches aimed at improving specificity and minimizing toxicity. Additional discussions will address the evolution beyond traditional IgG formats into IgE, IgA, and other next-generation antibody modalities, underscoring the continuing leadership of antibody-based therapies in next-generation oncology.

Sunday, May 10

Recommended Pre-Conference Short Course

SC5: Safety & Efficacy of Bispecifics and ADCs

*Separate registration required. See short course page for details.

Monday, May 11

Registration and Morning Coffee

Organizer's Opening Remarks

COMPARING AND COMBINING TCEs, ADCs, CAR Ts AND RADIOIMMUNOTHERAPY

Chairperson's Remarks

Daniel A. Vallera, PhD, Lion Scholar; Director, Section on Molecular Cancer Therapeutics; Professor, Therapeutic Radiology, University of Minnesota Masonic Cancer Center , Lion Scholar and Professor; Director, Section on Molecular Cancer Therapeutics; Professor , Therapeutic Radiology , University of Minnesota Masonic Cancer Center

KEYNOTE PRESENTATION: Comparing TCEs, ADCs, and CAR T Cell Therapy: What Have We Learned So Far?

Photo of Patrick Baeuerle, PhD, Chief Scientific Advisor, Cullinan Therapeutics, Inc. , Chief Scientific Advisor , Cullinan Therapeutics, Inc.
Patrick Baeuerle, PhD, Chief Scientific Advisor, Cullinan Therapeutics, Inc. , Chief Scientific Advisor , Cullinan Therapeutics, Inc.

T cell-engaging antibodies (TCEs) are bispecific, antibody-based adaptor proteins that connect any kind of cytotoxic T cells with select target cells for redirected lysis. Over the last three years, TCEs have seen an unparalleled surge in approvals as a standalone therapy. A total of twelve TCEs are now approved that very effectively treat hematological as well as solid tumor indications. I will review all learnings from the twelve approved TCEs.

Optimal Method of Targeting TRBC Alleles in T Cell Malignancies: Comparing CAR Ts and ADCs

Photo of Suman Paul, MBBS, PhD, Assistant Professor, Oncology, Johns Hopkins University , Assistant Professor , Oncology , Johns Hopkins University
Suman Paul, MBBS, PhD, Assistant Professor, Oncology, Johns Hopkins University , Assistant Professor , Oncology , Johns Hopkins University

T cell cancers are often fatal, necessitating the generation of novel therapies. Targeting TRBC1 can kill T cell cancers while preserving sufficient healthy T cells to maintain immunity. However, the first-in-human clinical trial of anti-TRBC1 CAR T cells reported a low response rate and unexplained loss of anti-TRBC1 CAR T cells. Our study shows how the CAR T cells are lost due to killing by the patient’s normal T cells. We further show that the generation of TRBC1-targeting ADCs bypasses this limitation and may produce superior responses in patients with T cell cancers.

Radioimmunotherapy: Engineered Antibody Formats, Fusion Proteins, and Combination Therapy

Photo of Anna M. Wu, PhD, Chair and Professor, Immunology & Theranostics, Center for Theranostic Studies, City of Hope , Chair and Professor , Immunology & Theranostics , Beckman Research Institute of City of Hope
Anna M. Wu, PhD, Chair and Professor, Immunology & Theranostics, Center for Theranostic Studies, City of Hope , Chair and Professor , Immunology & Theranostics , Beckman Research Institute of City of Hope

The development of antibodies for radiopharmaceutical therapy continues apace, with recent progress in engineered antibody formats and fusion proteins adding versatility to treatment approaches. Combination therapies are likely to provide the greatest efficacy, and prospects for combining radioimmunotherapy with modalities including external beam therapy and immunotherapies will be described. Clinical examples include targeting CD25 and CD38 in hematologic malignancies and CEA in colorectal and other adenocarcinomas.

Networking Coffee Break

MASKING AND CONDITIONAL ANTIBODIES—IMPROVING SPECIFICITY AND REDUCING TOXICITY

INDUCER Technology—The Next Generation of T Cell Engager Design

Photo of William Winston, PhD, Senior Vice President, Research, Werewolf Therapeutics , SVP , Research , Werewolf Therapeutics
William Winston, PhD, Senior Vice President, Research, Werewolf Therapeutics , SVP , Research , Werewolf Therapeutics

Challenges for TCE molecules targeting solid tumors include cytokine release syndrome and on-target/off-tumor toxicity due to systemic activity. Our INDUCER platform addresses these issues through conditional activation in the tumor microenvironment. Unlike existing prodrug TCEs, our approach uses a novel and differentiated anti-CD3 masking strategy that renders INDUCER molecules inactive systemically until regaining full function exclusively within the tumor microenvironment.

Coupling Tumor-Specific Payload Delivery with a Novel Target for Immune Engagement

Photo of John Burg, PhD, Senior Director, Protein Sciences, Pheast Therapeutics , Sr. Director, Protein Sciences , Protein Sciences , Pheast Therapeutics
John Burg, PhD, Senior Director, Protein Sciences, Pheast Therapeutics , Sr. Director, Protein Sciences , Protein Sciences , Pheast Therapeutics

Achieving specificity and efficacy remain key challenges in immuno-oncology. We have developed a bispecific antibody-drug conjugate (ADC), integrating a tumor targeting arm with a functional arm that enhances immune engagement by a novel mechanism. This strategy enables selective payload delivery while amplifying the immune response in the tumor microenvironment. Preclinical studies demonstrate potent in vitro and in vivo activity.

Session Break

Session Break

DUAL-TARGETING STRATEGY

Chairperson's Remarks

William Winston, PhD, Senior Vice President, Research, Werewolf Therapeutics , SVP , Research , Werewolf Therapeutics

Next-Generation 4-1BB T Cell Engaging Bispecific Antibody (Grabody T) Demonstrated Clinical Activity and Safety Profile

Photo of Sang Hoon Lee, PhD, CEO & Founder, ABL Bio Inc. , CEO & Founder , ABL Bio Inc
Sang Hoon Lee, PhD, CEO & Founder, ABL Bio Inc. , CEO & Founder , ABL Bio Inc

Ragistomig (ABL503/TJ-L14B) is a bispecific antibody combining PD-L1 checkpoint pathway with 4-1BB agonistic activity to overcome the current limitation of PD-(L)1 therapy and 4-1BB related toxicity. ABL503 is full length anti-PD-L1 mAb (Fc-silenced human IgG1) fused with scFv of anti-4-1BB engaging mAb. Givastomig (ABL111/TJ033721) is a bispecific antibody designed to target tumors with a wide range of Claudin 18.2 (CLDN18.2) expression and engage 4-1BB through a unique conditional activation mechanism at the tumor sites to avoid systemic toxicities.

Dual-Ig: A Novel Next-Generation T Cell Engager Targeting Both CD3 and CD137

Photo of Hiroaki Nagano, PhD, Pharmacology Researcher, Discovery Pharmacology, Research Division, Chugai Pharmaceutical, Co. Ltd. , Pharmacology Researcher , Discovery Pharmacology, Research Division , Chugai Pharmaceutical, Co. Ltd
Hiroaki Nagano, PhD, Pharmacology Researcher, Discovery Pharmacology, Research Division, Chugai Pharmaceutical, Co. Ltd. , Pharmacology Researcher , Discovery Pharmacology, Research Division , Chugai Pharmaceutical, Co. Ltd

TA/CD3 bispecific antibody is a potent approach in cancer treatment, but its efficacy is limited to tumors with less T cell infiltration. Dual-Ig binds to both CD3 and CD137 in the same Fab non-simultaneously, providing Signal 1 and Signal 2 to overcome this limitation while preventing off-target killing of TA-negative cells. We present the mechanism underlying this non-simultaneously binding and demonstrate advantages over conventional bispecific antibodies.

Simultaneous Targeting of Critical Immune Checkpoints and Activator by a Novel Multifunctional Fusion Protein for Cancer Therapy

Photo of Xiaodong Xiao, PhD, CEO, Jecho Laboratories, Inc. , CEO , Jecho Laboratories, Inc.
Xiaodong Xiao, PhD, CEO, Jecho Laboratories, Inc. , CEO , Jecho Laboratories, Inc.

We report a novel fusion protein, V5, for cancer immunotherapy. V5 contains a structure-guided, enhanced PD-1 ectodomain and a CD80 domain. The synergy of PD-1 blockade and T cell activation through CD80 led to persistent inhibition of various tumors in animal models, including colorectal, liver, pancreatic, and cholangiocarcinoma cancers, regardless of PD-L1 expression or resistance to conventional PD-1 blockade. V5 treatment induced effective immune memory. V5 also activates tumor-specific T cells via APCs and PD-L2 signaling. Preclinical studies in cynomolgus monkeys showed a promising safety profile, supporting V5 in human studies.

Networking Coffee & Refreshment Break

Transition to Plenary Keynote Session

PLENARY KEYNOTE

Plenary Keynote Introduction

Photo of Mahiuddin Ahmed, PhD, President and CSO, VITRUVIAE , President and CSO , VITRUVIAE
Mahiuddin Ahmed, PhD, President and CSO, VITRUVIAE , President and CSO , VITRUVIAE

CARs 2026: New Models and New Runways

Photo of Michel Sadelain, MD, PhD, Director, Columbia University Initiative in Cell Engineering and Therapy (CICET); Director, Cell Therapy Initiative, Herbert Irving Comprehensive Cancer Center; Professor of Medicine, Columbia University Irving Medical Center , Stephen & Barbara Friedman Chair & Director , Center for Cell Engineering , Memorial Sloan Kettering Cancer Centre
Michel Sadelain, MD, PhD, Director, Columbia University Initiative in Cell Engineering and Therapy (CICET); Director, Cell Therapy Initiative, Herbert Irving Comprehensive Cancer Center; Professor of Medicine, Columbia University Irving Medical Center , Stephen & Barbara Friedman Chair & Director , Center for Cell Engineering , Memorial Sloan Kettering Cancer Centre

T cell engineering holds great promise for the treatment of cancers and other pathologies. The original chimeric antigen receptor (CAR) prototypes targeting CD19 are now giving way to further refined receptors endowed with greater sensitivity and combinatorial possibilities. Emerging new targets and engineering tools augur favorably for broadening the use of CAR therapies.

YOUNG SCIENTIST KEYNOTE

Deep Learning-Based Binder Design to Probe Biology

Photo of Martin Pacesa, PhD, Assistant Professor, Pharmacology, University of Zurich , Assistant Professor , Department of Pharmacology , University of Zurich
Martin Pacesa, PhD, Assistant Professor, Pharmacology, University of Zurich , Assistant Professor , Department of Pharmacology , University of Zurich

Protein-protein interactions are central to biology and drug discovery, yet traditional antibody generation is slow and costly. BindCraft is an open-source, automated computational pipeline for de novo protein binder design that routinely yields nanomolar binders with 10-100% experimental success, without high-throughput screening or maturation. We illustrate applications to peptides, cell-surface receptors, allergens, and gene editors, and outline how deep learning workflows can accelerate next-generation therapeutics, diagnostics, and bioprocessing.


  • What are the advantages/drawbacks of minibinders?
  • Are there "unbindable" protein sites?
  • Are natural amino acid building blocks enough for drug development?
  • What therapeutic properties should deep learning models account for?

Welcome Reception in the Exhibit Hall with Poster Viewing

Close of Day

Tuesday, May 12

Registration and Morning Coffee

IMPROVING T CELL ENGAGER ANTIGEN SELECTIVITY AND ANTI-TUMOR ACTIVITY

Chairperson's Remarks

David Cole, Head of Research, Accession Therapeutics Inc.; Honorary Professor, Cardiff University , Head of Research , Research , Accession Therapeutics Inc.

CBX250, a Novel Cathepsin G Peptide-HLA-Targeting T Cell Engager that Exhibits High Tumor Antigen Selectivity and Potent Antileukemic Activity in Vivo

Photo of Scott Chunhua Shi, PhD, Associate Director Institute & Head of Biological Discovery, ORBIT Therapeutic Discovery, MD Anderson Cancer Center , Institute Assoc Dir, Head of Biological Discovery , ORBIT Therapeutic Discovery Division , MD Anderson Cancer Ctr
Scott Chunhua Shi, PhD, Associate Director Institute & Head of Biological Discovery, ORBIT Therapeutic Discovery, MD Anderson Cancer Center , Institute Assoc Dir, Head of Biological Discovery , ORBIT Therapeutic Discovery Division , MD Anderson Cancer Ctr

TCRm represent a promising modality for tumor-therapeutics. We have developed a robust TCRm discovery pipeline that enabled identification of the CTSG peptide–HLA-A*02 complex from AML blasts, leading to the clinical candidate CBX250 (in collaboration with Crossbow Therapeutics). CBX250 demonstrates potent in vitro and in vivo efficacy without detectable cross-reactivity, and US Phase I trials are ongoing. A first-in-class TCRm×CD3 targeting CCNB1 is also in IND-enabling studies to benefit more patients.

Addressing Solid-Tumor Heterogeneity: TROCEPT-Mediated Activation of a Universal Bispecific T Cell Engager via IV Delivery

Photo of David Cole, Head of Research, Accession Therapeutics Inc.; Honorary Professor, Cardiff University , Head of Research , Research , Accession Therapeutics Inc.
David Cole, Head of Research, Accession Therapeutics Inc.; Honorary Professor, Cardiff University , Head of Research , Research , Accession Therapeutics Inc.

Tumors are very heterogenous and include immunosuppressive cell types, limiting the ability of current therapies to target all cells in the tumor with high potency. TROCEPT is a novel immuno-virotherapy that only targets cancer cells, and turns them into drug factories. We have used this technology to deliver a novel universal bispecific T cell engager, that can target all cancer cells, only inside the tumor.

Combining CD3 and CD28 T Cell Engagers for Enhanced Anti-Tumor Activity

Photo of Gregory L. Moore, PhD, Executive Director, Protein Engineering, Xencor, Inc. , Exec. Director , Protein Engineering , Xencor Inc.
Gregory L. Moore, PhD, Executive Director, Protein Engineering, Xencor, Inc. , Exec. Director , Protein Engineering , Xencor Inc.

CD3 T cell engagers activate T cells through Signal 1, but solid tumors generally lack the costimulatory signals required for full activation, potentially leading to anergy and reduced efficacy. Tumor-targeted CD28 bispecific antibodies that strictly depend on concurrent Signal 1 for costimulatory activity represent a key advancement in immunotherapy. Adding targeted costimulation enhances cytokine production, proliferation, survival, restores activity in restimulation settings, and drives stronger anti-tumor responses in preclinical models.

Coffee Break in the Exhibit Hall with Poster Viewing

NOVEL FORMATS AND ALTERNATIVE APPROACHES

Overcoming the Tumor Penetration Challenge: Nanofitin-Based Drug Conjugates for Deep and Efficient Tumor Engagement

Photo of Mathieu Cinier, PhD, CSO, Affilogic , Scientific Director & CSO , Affilogic
Mathieu Cinier, PhD, CSO, Affilogic , Scientific Director & CSO , Affilogic

Treating cancer requires balancing cytotoxicity, immune activation, and safety to eradicate malignant cells while sparing healthy tissue. Nanofitin-based drug conjugates offer a powerful solution by combining antibody-like high-affinity and specificity with the deep tumor penetration of small scaffolds. Their rapid systemic clearance minimizes off-target toxicity while ensuring efficient delivery of potent payloads such as MMAE. In preclinical models, weekly dosing of an MMAE-Nanofitin conjugate achieved complete tumor growth inhibition. This approach overcomes diffusion and resistance barriers by reaching tumor cells beyond vascularized regions, paving the way for next-generation targeted cancer therapeutics.

Beyond IgG: The Therapeutic Potential of IgE and IgA Antibodies and Their Derivatives

Photo of Kevin FitzGerald, PhD, CSO, Epsilogen Ltd. , CSO , Epsilogen Ltd
Kevin FitzGerald, PhD, CSO, Epsilogen Ltd. , CSO , Epsilogen Ltd

Epsilogen was founded to develop therapeutic IgE antibodies. Through acquisition of TigaTx in March 2025, Epsilogen is now also developing therapeutic IgA. Epsilogen's lead antibody, MOv18 IgE, targets folate receptor alpha and has completed a phase 1 clinical trial. It is currently in a phase Ib trial treating platinum-resistant ovarian cancer patients. Epsilogen also has IgE and IgA antibodies in pre-clinical studies and it has a number of proprietary platforms including bispecific IgE and hybrid antibodies that combine IgE or IgA with IgG functionality.

Session Break

Close of Antibodies for Cancer Therapy Conference

Recommended Dinner Short Course

SC6: Developability of Bispecific Antibodies

*Separate registration required. See short course page for details.


For more details on the conference, please contact:

Mimi Langley

Executive Director, Conferences

Cambridge Healthtech Institute

Email: mlangley@healthtech.com

 

For sponsorship information, please contact:

Companies A-K

Jason Gerardi

Sr. Manager, Business Development

Cambridge Healthtech Institute

Phone: 781-972-5452

Email: jgerardi@healthtech.com

 

Companies L-Z

Ashley Parsons

Manager, Business Development

Cambridge Healthtech Institute

Phone: 781-972-1340

Email: ashleyparsons@healthtech.com


Register

View By:


Premier Sponsors

FairJourneyBiologics GenScript-CRO Integral-Molecular_NEW   Nona_Biosciences_NEW