Cambridge Healthtech Institute's 3rd Annual

Safety and Efficacy of Bispecific Antibodies, ADCs, and Combination Therapies

Enhancing Safety and Creating Synergies with Novel Therapeutic Modalities

11 November 2025 ALL TIMES WET (GMT/UTC)


Novel bispecific and multispecific antibodies, antibody-drug conjugates (ADCs), and combination treatments open the door to addressing a broader set of clinical indications. Alongside these developments come new challenges in optimizing safety and efficacy of these next-generation therapeutics. At PEGS Europe, Cambridge Healthtech Institute’s Third Annual Safety and Efficacy of Bispecific Antibodies, ADCs, and Combination Therapies program will highlight novel therapeutic candidates and how researchers are mitigating toxicity, managing combination therapies, reducing off-target effects, and addressing challenges in treatment of solid tumors. This meeting will complement topics in bispecific antibody discovery and engineering for meeting unmet clinical needs.

Recommended Short Course*
Monday, 4 November, 14:00 – 17:00
SC3: Developability of Bispecific Antibodies
*Separate registration required. See short courses page for details. All short courses take place in-person only.





Tuesday, 11 November

Registration and Morning Coffee

SAFETY AND EFFICACY OF BISPECIFICS AND ADCs

Chairperson's Remarks

Rakesh Dixit, PhD, DABT, CEO & President, Bionavigen Oncology, LLC; CSO, TMAB Therapeutics, Regio Biosciences , CEO & President , Bionavigen Oncology, LLC and Regio Biosciences

KEYNOTE PRESENTATION:
Safety of Bispecifics, ADCs, and Combination Therapies

Photo of Rakesh Dixit, PhD, DABT, CEO & President, Bionavigen Oncology, LLC; CSO, TMAB Therapeutics, Regio Biosciences , CEO & President , Bionavigen Oncology, LLC and Regio Biosciences
Rakesh Dixit, PhD, DABT, CEO & President, Bionavigen Oncology, LLC; CSO, TMAB Therapeutics, Regio Biosciences , CEO & President , Bionavigen Oncology, LLC and Regio Biosciences

The safety and efficacy of bispecific antibodies, ADCs, and combination therapies represent significant advancements in oncology. Each modality offers unique mechanisms to target cancer cells more effectively while aiming to reduce collateral damage to normal tissues. However, their use entails careful consideration of potential toxicities and requires robust management strategies to ensure patient safety. These therapies offer significant promise for improving treatment outcomes and restoring hope for cancer patients. This presentation will highlight developments in bispecific antibodies that reduce the risk of cytokine release syndrome (CRS), more stable ADCs with advanced linker chemistry, and groundbreaking combination strategies that balance efficacy with minimised side effects. Join us in exploring these transformative solutions for the future of cancer care.

Cancer Therapy with Bispecific Antibodies Directed to CD3 and CD28: Two Targets, Two Signals

Photo of Martin Pflügler, PhD, CEO, TWYCE GmbH , CEO , TWYCE GmbH
Martin Pflügler, PhD, CEO, TWYCE GmbH , CEO , TWYCE GmbH

Efficacy of bispecific antibodies (bsAbs) in solid tumours is limited by (i) lack of accessibility of the tumour site for immune effector cells, (ii) lack of sufficiently tumour-specific target antigens, and (iii) lack of costimulatory “signal 2” that enables thorough and long-lasting T cell activation. We plan to overcome these limitations by a combination of functionally interrelated bsAbs that target two different antigens expressed on both tumour cells and the tumour microenvironment/tumour vasculature and stimulate CD3 and CD28 on T cells.

Beyond ADCs: Cancerlysins—Bispecific Antibodies That Selectively Eradicate Cancer Cells by Inducing Apoptosis

Photo of Victor S. Goldmacher, PhD, CSO, R&D, ImmuVia , CSO , R&D , ImmuVia, Inc.
Victor S. Goldmacher, PhD, CSO, R&D, ImmuVia , CSO , R&D , ImmuVia, Inc.

IMV-M is a bispecific antibody that co-targets MUC16 and death receptor 5 (DR5) to induce tumour-selective apoptosis. MUC16 is overexpressed in non-small cell lung cancer, ovarian cancer, and pancreatic adenocarcinoma, while its expression in normal tissues is minimal. DR5 is broadly expressed across these tumour types. IMV-M has demonstrated potent antitumour activity in xenograft models, favorable safety in non-human primates, and is CMC-ready.

Grand Opening Coffee Break in the Exhibit Hall with Poster Viewing

Emerging Immunogenicity Challenges for Next-Generation Biotherapeutics

Photo of Andreas Hollenstein, PhD, Principal Scientist, Immunosafety, Roche , Sr. Principal Scientist , Immunosafety , Roche pRED RICB
Andreas Hollenstein, PhD, Principal Scientist, Immunosafety, Roche , Sr. Principal Scientist , Immunosafety , Roche pRED RICB

Since the beginning of immunotherapy, the limits for higher efficiency have been pushed. This shift did not come without tradeoffs for safety and immunogenicity. This presentation showcases several examples of drug enhancing approaches that also can have a profound impact on immunogenicity. Understanding the underlying mechanisms can help to avoid immunogenic transformation and lead to the development of better drugs for patients.

Structure-Aided Design and Engineering of an FGFR1c x KLB Multispecific Altibody Agonist for MASH

Photo of Yang Shen, PhD, Executive Director of Antibody Engineering, Bispecifics, Regeneron , Executive Director of Antibody Engineering , Bispecifics & Antibody Engineering , Regeneron
Yang Shen, PhD, Executive Director of Antibody Engineering, Bispecifics, Regeneron , Executive Director of Antibody Engineering , Bispecifics & Antibody Engineering , Regeneron

Bispecific antibody has already evolved as a well-validated therapeutic modality with multiple regulatory approvals mostly in oncology setting over the past few years. Multispecific antibody targeting more epitopes on the same or different targets has recently emerged as a novel modality with unique advantages to afford better potency via binding to additional epitopes, broader target space by engaging more challenging receptor complexes and higher specificity from restricting activity to only overlapping target expression pattern. FGF21 is a master coordinator for fatty acid homeostasis between liver and adipose tissue. Developing FGF21 mimetics has been one of a few therapeutic strategies currently being tested to treat MASH (metabolic dysfunction-associated steatohepatitis) clinically. Harnessing structure-aided design, we have engineered a multispecific AltibodyTM agonist mimicking FGF21 ligand function by activating FGFR1c only in the presence of its coreceptor KLB. Format-based stepwise optimization further enhances its agonist activity. Our study illuminates that a combination of factors, such as IgG subclass, linker length and building block, can contribute to the improved agonism, highlighting the challenges and complexity of engineering multispecific agonist antibody. Our FGFR1c x KLB multispecific Altibody offers an attractive alternative strategy to the current Fc-FGF21 fusion tested in clinic with superior PK, lower immunogenicity and similar efficacy in preclinical settings.

Luncheon in the Exhibit Hall with Poster Viewing

APPROACHES TO ADDRESS SAFETY AND EFFICACY OF T CELL ENGAGERS

Chairperson's Remarks

Javier Chaparro-Riggers, PhD, Executive Director, BioMedicine Design, Pfizer Inc. , Executive Director , Biomedicine Design , Pfizer

Engineering Approaches to Address Safety and Efficacy Challenges of T Cell Engagers

Photo of Javier Chaparro-Riggers, PhD, Executive Director, BioMedicine Design, Pfizer Inc. , Executive Director , Biomedicine Design , Pfizer
Javier Chaparro-Riggers, PhD, Executive Director, BioMedicine Design, Pfizer Inc. , Executive Director , Biomedicine Design , Pfizer

Allogeneic CAR T cells can overcome limitations associated with autologous cancer therapies, providing immediate access to standardised, affordable batches of CAR T with improved efficacy. We systematically interrogated genes providing resistance to allogeneic rejection using in vivo genome-wide CRISPR KO in T cells. We identified Fas and B2m and demonstrated using base editing in human CAR T cells that TCR/FAS-inactivation outperforms TCR/B2M-deletion in the resistance to both T and NK cell-mediated allo-rejection.

Machine Learning-Guided Design of Logic-Gated and Avidity-Driven T Cell Engagers for Solid Malignancies

Photo of Ryan Henrici, MD, PhD, Vice President, Discovery Medicine, BigHat Biosciences , Vice President , Discovery Medicine , BigHat Biosciences
Ryan Henrici, MD, PhD, Vice President, Discovery Medicine, BigHat Biosciences , Vice President , Discovery Medicine , BigHat Biosciences

Broad application of T cell engagers to patients with solid tumors is limited by the availability of antigens that discriminate malignant from non-malignant tissues. We show that T cell engagers can be readily built for classic solid tumor targets with a lab-in-the-loop approach to AI/ML-guided antibody design, efficiently clearing tumors without established on-target toxicities. We provide examples of avidity-gated and Boolean logic-gated T cell engagers in diverse malignancies.

Cancer Immunotherapy Using Bispecific γδ-T Cell Engagers

Photo of Hans van der Vliet, MD, PhD, Professor, Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam , Professor, Medical Oncology , Amsterdam UMC, Cancer Center Amsterdam
Hans van der Vliet, MD, PhD, Professor, Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam , Professor, Medical Oncology , Amsterdam UMC, Cancer Center Amsterdam

Vγ9Vδ2-T cells constitute a relatively homogeneous population of pro-inflammatory immune effector cells. This presentation will focus on the preclinical and early clinical development of bispecific Vγ9Vδ2-T cell engagers as a novel approach for cancer immunotherapy.

AZD6750: Design and Efficacy of a CD8-Guided IL-2 that Enhances CD8+ T Cell Function

Photo of Matthew Elder, PhD, Director, Project Lead, Immuno-Oncology Team, AstraZeneca , Director , Immuno-Oncology Team , AstraZeneca
Matthew Elder, PhD, Director, Project Lead, Immuno-Oncology Team, AstraZeneca , Director , Immuno-Oncology Team , AstraZeneca

IL-2 is a clinically validated immunotherapy. However, toxicity limits its use and despite years of research, the optimal approach to deliver IL-2 has yet to be achieved. AZD6750 applies cis-guiding to deliver a modified IL-2 mutein preferentially to CD8+ T cells. This approach potentially limits the side effects of Aldesleukin, typically attributed to the broad effects of IL-2R signalling, which should result in a safer, more effective IL-2.

Refreshment Break in the Exhibit Hall with Poster Viewing

EMERGING THERAPEUTICS

Targeting Transferrin Receptor to Transport Antisense Oligonucleotides across the Blood-Brain Barrier

Photo of Padma Akkapeddi, PhD, Senior Scientist, Antibody Discovery & Protein Engineering, Denali Therapeutics, Inc. , Senior Scientist , Antibody Discovery & Protein Engineering , Denali Therapeutics Inc
Padma Akkapeddi, PhD, Senior Scientist, Antibody Discovery & Protein Engineering, Denali Therapeutics, Inc. , Senior Scientist , Antibody Discovery & Protein Engineering , Denali Therapeutics Inc

The blood-brain barrier (BBB) restricts the effective delivery of protein therapeutics to the central nervous system (CNS). To address this, we have developed an engineered Fc BBB transport vehicle (TV) that binds a highly expressed brain endothelial cell target and exploits receptor-mediated transcytosis to facilitate biotherapeutic delivery to the CNS. TVs were engineered to bind the apical domain of the human transferring receptor (hTfR) using directed evolution and without the use of amino acid insertions, deletions, or unnatural appendages. The TV platform is a highly modular CNS delivery platform that readily accommodates diverse antibody architectures, including bispecific formats, protein fusions and oligonucleotides.

In this work, we describe the development of a new TV platform that enables transport of ASOs into the brain, called “Oligonucleotide Transport Vehicle” (OTV). Systemically delivered OTV drives significant, cumulative, and sustained knockdown of the target gene expression across multiple CNS regions and all major cell types. When compared with other clinically relevant ASO delivery routes, such as a high affinity TfR antibody or direct ASO delivery to the CSF, systemic OTV delivery also results in more uniform ASO biodistribution and target knockdown. Our data supports systemically delivered OTV as a potential therapeutic oligonucleotide-delivery platform for neurological disorders.

Advancing the Cancer-Targeting Radio-Antibody Drug Conjugate 177Lu-AKIR001 to Clinical Trials

Photo of Marika Nestor, PhD, Professor, Immunology, Genetics, and Pathology, Uppsala University; CEO, Akiram Therapeutics , Professor , Immunology, Genetics, and Pathology , Uppsala University
Marika Nestor, PhD, Professor, Immunology, Genetics, and Pathology, Uppsala University; CEO, Akiram Therapeutics , Professor , Immunology, Genetics, and Pathology , Uppsala University

This talk explores the development of 177Lu-AKIR001, a novel radio-antibody drug conjugate (RADC) for targeted cancer therapy. By integrating antibody engineering with radionuclide technology, this approach combines precise tumour targeting with localised radiation delivery. Preclinical findings demonstrate strong efficacy and safety, supporting the ongoing clinical trial. The presentation will discuss the design, challenges, and translational potential of RADCs, bridging protein engineering and radiopharmaceutical innovation.

POSTER HIGHLIGHT:
AZD1163: First-in-Class Anti-PAD2/4 Bi-Specific Antibody to Inhibit Autoantigen Formation in Rheumatoid Arthritis

Photo of Juan Bonfante, PhD, Senior Scientist, Biologics Engineering, AstraZeneca , Senior Scientist , Biologics Engineering , AstraZeneca
Juan Bonfante, PhD, Senior Scientist, Biologics Engineering, AstraZeneca , Senior Scientist , Biologics Engineering , AstraZeneca

Rheumatoid arthritis is a chronic autoimmune disease marked by joint inflammation and destruction. Citrullinated proteins, produced by PAD2 and PAD4 enzymes, drive autoantibody generation and joint damage. AZD1163, a novel bispecific monoclonal antibody, potently inhibits human PAD2 and PAD4. It shows high-affinity binding, preserves neutrophil function, avoids cytokine release, and is well tolerated. Favourable human pharmacokinetics support AZD1163’s continued clinical development.

Attend Concurrent Session

Welcome Reception in the Exhibit Hall with Poster Viewing

Close of Safety and Efficacy of Bispecific Antibodies, ADCs, and Combination Therapies Conference


For more details on the conference, please contact:

Iris Goldman
Conference Producer
Cambridge Healthtech Institute
Email: igoldman@healthtech.com

For sponsorship information, please contact:

Companies A-K
Jason Gerardi
Sr. Manager, Business Development
Cambridge Healthtech Institute
Phone: (+1) 781-972-5452
Email: jgerardi@healthtech.com

Companies L-Z
Ashley Parsons
Manager, Business Development
Cambridge Healthtech Institute
Phone: (+1) 781-972-1340
Email: ashleyparsons@healthtech.com