Emerging Targets for Oncology & Beyond
Hitting the Bullseye
5/12/2026 - May 13, 2026 ALL TIMES EDT
Cambridge Healthtech Institute’s 5th Annual Emerging Targets for Oncology & Beyond conference spotlights the discovery and validation of novel and previously “undruggable” targets, highlighting the expanding landscape of therapeutic innovation. The program will feature advances in targeted protein degradation strategies, going beyond the surfaceome and proteome, along with AI- and data-driven approaches accelerating target identification and therapeutic design. New this year is a session exploring emerging targets in autoimmune, cardiovascular, and other disease areas beyond oncology, illustrating how breakthroughs in target discovery are transforming drug pipelines across multiple indications.

Sunday, May 10

Recommended Pre-Conference Short Course

SC3: Challenges and Opportunities in Solid Tumor and Autoimmune Disease Therapeutics

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

Tuesday, May 12

Networking Coffee & Dessert Break in the Exhibit Hall with Poster Viewing

Organizer's Opening Remarks

NOVEL TARGETS LANDSCAPE

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

IGSF8, an Emerging Target for Cancer Immunotherapy

Photo of Xiaole Shirley Liu, PhD, CEO, GV20 Therapeutics , CEO , GV20 Therapeutics
Xiaole Shirley Liu, PhD, CEO, GV20 Therapeutics , CEO , GV20 Therapeutics

Despite the exciting clinical benefits of immune checkpoint inhibitors, only a minority of cancer patients respond. We integrate AI, functional genomics, and cancer immunology for drug discovery. Our STEAD platform computationally extracts antibodies from patient tumor RNA-seq profiles and pairs targets with antibodies in silico. Our lead program, GV20-0251, targets novel checkpoint IGSF8 and reached IND in three years, demonstrating favorable safety and promising monotherapy efficacy in metastatic cancer patients.

GPC2 as a Target for Antibody- and Cell-Based Therapies in Childhood Cancers

Photo of Mitchell Ho, PhD, Senior Investigator & Deputy Chief, Laboratory of Molecular Biology; Director, Antibody Engineering Program, National Cancer Institute (NCI), National Institutes of Health (NIH) , Senior Investigator & Deputy Chief , Laboratory of Molecular Biology , National Cancer Institute, National Institutes of Health
Mitchell Ho, PhD, Senior Investigator & Deputy Chief, Laboratory of Molecular Biology; Director, Antibody Engineering Program, National Cancer Institute (NCI), National Institutes of Health (NIH) , Senior Investigator & Deputy Chief , Laboratory of Molecular Biology , National Cancer Institute, National Institutes of Health

Glypican-2 (GPC2), an oncofetal antigen expressed in childhood cancers such as neuroblastoma, is a new target whose inhibition blocks Wnt/ß-catenin signaling and N-Myc activity. I will also describe the engineering of T cells expressing novel antibody–TCR hybrids that incorporate a humanized CT3 Fab specific for GPC2, linked to TCR ? and d chains. These cells outperform CAR T cells in low-antigen tumors and have broad potential for solid tumor therapy.

Refreshment Break in the Exhibit Hall with Poster Viewing

Next-Generation T Cell Receptor Bispecific (TCER) Targeting COL6A3 on the Stroma of Solid Tumors

Photo of Felix Unverdorben, PhD, Associate Director, TCR Discovery and Bispecifics, Immatics Biotechnologies GmbH , Assoc Dir , TCR Discovery and Bispecifics , Immatics GmbH
Felix Unverdorben, PhD, Associate Director, TCR Discovery and Bispecifics, Immatics Biotechnologies GmbH , Assoc Dir , TCR Discovery and Bispecifics , Immatics GmbH

Complementing Immatics’ broad PRAME franchise, the presented T cell receptor bispecific (TCER) is directed against the stromal pHLA target COL6A3 Exon 6, aiming to disrupt the cancer’s protective barrier providing an innovative approach for cancer treatment. TCR potency and specificity are assessed throughout TCR identification and TCER engineering, demonstrating strong thermal stability, high affinity target binding and killing of target positive tumor cell lines.

Breaking the Sweet Silence: TACA Targeting Therapeutics against Tumor-Glycan Shields

Photo of Peter Sondermann, PhD, CEO, Tacalyx GmbH , CEO , Tacalyx GmbH
Peter Sondermann, PhD, CEO, Tacalyx GmbH , CEO , Tacalyx GmbH

Tumor-associated carbohydrate antigens (TACAs) are defined oligosaccharide structures, derived from changes in glycosylation pathways, that are expressed on the surface of cancer cells. Tacalyx develops antibodies against these difficult-to-target structures with the goal of spearheading the next generation of targeted anti-cancer therapeutics and will present its advancements in their current lead programs.

Clean Cancer Targets for Better Therapeutics: Going beyond the Proteome

Photo of Hans H. Wandall, Co-Founder & CSO, Discovery, GO Therapeutics , Co Founder & CSO , Discovery , GO Therapeutics
Hans H. Wandall, Co-Founder & CSO, Discovery, GO Therapeutics , Co Founder & CSO , Discovery , GO Therapeutics

Most targeted cancer therapies rely on proteins overexpressed on tumors, but these are rarely truly cancer-specific, limiting safety and efficacy. GO Therapeutics leverages cancer-selective aberrant O-glycosylation to overcome this limitation. Using high-resolution O-glycoproteomics, we have defined tumor-restricted Tn-glycoepitopes on a large set of targets, including MUC1, MUC4, cMET, LAMP1, and CD44. Antibodies and ADCs directed to these glycoforms show high selectivity, potent in vivo cytotoxicity, negligible normal-tissue reactivity, and favorable cynomolgus toxicology, supporting clinical translation of this glyco-targeting platform.

Close of Day

Recommended Dinner Short Course

SC7: Targeting the Target: Aligning Target and Biologic Format Biology to Achieve Desired Outcomes

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

Wednesday, May 13

Registration Open

PEGS YOUNG SCIENTIST KEYNOTE ALUMNI PANEL

Chairperson’s Remarks

Panel Moderator:

Innovation in Protein Science with Young-Scientist Visionaries

Photo of James A. Wells, PhD, Professor, Departments of Pharmaceutical Chemistry and Cellular & Molecular Pharmacology, University of California, San Francisco , Professor , Departments of Pharmaceutical Chemistry and Cellular & Molecular Pharmacology , University of California San Francisco
James A. Wells, PhD, Professor, Departments of Pharmaceutical Chemistry and Cellular & Molecular Pharmacology, University of California, San Francisco , Professor , Departments of Pharmaceutical Chemistry and Cellular & Molecular Pharmacology , University of California San Francisco

Panelists:

Photo of Kathryn M. Hastie, PhD, Instructor and Director of Antibody Discovery, La Jolla Institute for Immunology , Instructor , Antibody DIscovery , La Jolla Institute for Immunology
Kathryn M. Hastie, PhD, Instructor and Director of Antibody Discovery, La Jolla Institute for Immunology , Instructor , Antibody DIscovery , La Jolla Institute for Immunology
Photo of Jamie B. Spangler, PhD, Associate Professor, Biomedical and Chemical & Biomolecular Engineering, Johns Hopkins University , Associate Professor , Biomedical Engineering and Chemical & Biomolecular Engineering , Johns Hopkins University
Jamie B. Spangler, PhD, Associate Professor, Biomedical and Chemical & Biomolecular Engineering, Johns Hopkins University , Associate Professor , Biomedical Engineering and Chemical & Biomolecular Engineering , Johns Hopkins University
Photo of Kipp Weiskopf, MD, PhD, Head of Antibody Therapeutics and Biologics, Cancer Research Institute, Beth Israel Deaconess Medical Center; Assistant Professor of Medicine & Physician, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School , Head of Antibody Therapeutics and Biologics , Cancer Research Institute , Beth Israel Deaconess Medical Center
Kipp Weiskopf, MD, PhD, Head of Antibody Therapeutics and Biologics, Cancer Research Institute, Beth Israel Deaconess Medical Center; Assistant Professor of Medicine & Physician, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School , Head of Antibody Therapeutics and Biologics , Cancer Research Institute , Beth Israel Deaconess Medical Center
Photo of Timothy A. Whitehead, PhD, Professor, Chemical & Biological Engineering, University of Colorado, Boulder , Professor , Chemical & Biological Engineering , Univ of Colorado Boulder
Timothy A. Whitehead, PhD, Professor, Chemical & Biological Engineering, University of Colorado, Boulder , Professor , Chemical & Biological Engineering , Univ of Colorado Boulder
Photo of Xin Zhou, PhD, Assistant Professor, Biological Chemistry & Molecular Pharmacology, Dana-Farber Cancer Institute, Harvard Medical School , Assistant Professor , Biological Chemistry and Molecular Pharmacology , Harvard Medical School
Xin Zhou, PhD, Assistant Professor, Biological Chemistry & Molecular Pharmacology, Dana-Farber Cancer Institute, Harvard Medical School , Assistant Professor , Biological Chemistry and Molecular Pharmacology , Harvard Medical School

Coffee Break in the Exhibit Hall with Poster Viewing

NOVEL APPROACHES FOR TARGETED PROTEIN DEGRADATION

Chairperson's Remarks

Mitchell Ho, PhD, Senior Investigator & Deputy Chief, Laboratory of Molecular Biology; Director, Antibody Engineering Program, National Cancer Institute (NCI), National Institutes of Health (NIH) , Senior Investigator & Deputy Chief , Laboratory of Molecular Biology , National Cancer Institute, National Institutes of Health

KEYNOTE PRESENTATION: Novel Approaches for Extracellular Targeted Protein Degradation (eTPD)

Photo of James A. Wells, PhD, Professor, Departments of Pharmaceutical Chemistry and Cellular & Molecular Pharmacology, University of California, San Francisco , Professor , Departments of Pharmaceutical Chemistry and Cellular & Molecular Pharmacology , University of California San Francisco
James A. Wells, PhD, Professor, Departments of Pharmaceutical Chemistry and Cellular & Molecular Pharmacology, University of California, San Francisco , Professor , Departments of Pharmaceutical Chemistry and Cellular & Molecular Pharmacology , University of California San Francisco

The cell surface proteome, the surfaceome, is a major hub for cellular communication and a primary source of drug targets, both small molecules and biologics. Inspired by the field intracellular targeted protein degradation (iTPD) that brought us PROTACs and molecular glues, there has been a surge of interest in extracellular targeted protein degradation (eTPD). Our group has been focused on using genetically encoded bi-specific agents to degrade disease-associated membrane and soluble proteins by pirating natural protein degradations systems using AbTACs or KineTACS. Here, I’ll describe novel modalities for eTPD for soluble and membrane bound targets.

Mechanisms and Therapeutic Applications of Antibody-Based Degraders

Photo of Xin Zhou, PhD, Assistant Professor, Biological Chemistry & Molecular Pharmacology, Dana-Farber Cancer Institute, Harvard Medical School , Assistant Professor , Biological Chemistry and Molecular Pharmacology , Harvard Medical School
Xin Zhou, PhD, Assistant Professor, Biological Chemistry & Molecular Pharmacology, Dana-Farber Cancer Institute, Harvard Medical School , Assistant Professor , Biological Chemistry and Molecular Pharmacology , Harvard Medical School

Cell–surface proteins are key regulators of cellular activity, and one promising way to modulate their function is by controlling endocytosis. Under physiological conditions, cells use endocytosis to maintain homeostasis and tune signaling, yet its therapeutic potential is only beginning to be explored. I will discuss how bispecific antibodies can be designed to modularly control membrane protein endocytosis and signalling, their underlying mechanisms, and the potential therapeutic applications of these molecules.

Session Break

AI AND DATA-DRIVEN APPROACHES FOR TARGET DISCOVERY AND THERAPEUTIC DESIGN

Chairperson's Remarks

Horacio G. Nastri, PhD, Vice President, Protein Science and Technology, Incyte Corporation , Vice President , Protein Science and Technology , Incyte Corporation

Panel Moderator:

PANEL DISCUSSION:
Designing for the Inside and Out: How Scaffold Choice Shapes Target Engagement and Drug Performance

Marie-Eve Beaulieu, PhD, Co-Founder & Chief Development Officer, Peptomyc SL , CoFounder & Chief Development Officer , Drug Dev , Peptomyc SL

Panelists:

Bradley M. Lunde, PhD, Associate Director, Adimab LLC , Associate Director , Adimab LLC

Illuminating the Disease Surfaceome: Exploiting Conformational Targets for First-in-Class Cancer Therapies

Photo of Neal Goodwin, PhD, CSO, Research, Immuto Scientific , CSO , Research , Immuto Scientific
Neal Goodwin, PhD, CSO, Research, Immuto Scientific , CSO , Research , Immuto Scientific

Immuto Scientific has advanced technologies that identify cancer-specific protein conformations, called Surface Protein Conformers (SPCs), which expose epitopes that do not present in healthy cells. SPC target discovery through structural proteomics is combined with AI-powered antibody engineering, enabling the development of next-generation biologics, such as ADCs and multispecifics, tailored for precise disease intervention with a much lower risk of off-disease toxicity. The SPC technology provides a key advantage by unlocking previously inaccessible therapeutic targets in challenging indications, like hematologic malignancy and lung cancer.

Mapping Cancer Vulnerabilities through the Cancer Dependency Map

Photo of Francisca Vazquez, PhD, Director, Cancer Dependency Map, Broad Institute , Director , Cancer Dependency Map , Broad Institute
Francisca Vazquez, PhD, Director, Cancer Dependency Map, Broad Institute , Director , Cancer Dependency Map , Broad Institute

DepMap systematically links tumor alterations to gene essentiality, supporting both basic and translational research. Many identified dependencies are now under validation or in clinical trials. Our recent work revealed a synthetic lethal interaction between the PELO/HBS1L and SKI complexes, suggesting PELO as a target in SKI-deficient cancers. DepMap continues to expand incorporating organoid models, new multi-omics data modalities, and new features towards the goal of accelerating precision cancer medicine.

Beyond SSTR and PSMA: Human Data-Driven Discovery and in silico Design of Next-Generation Radiopharmaceutical Therapies

Photo of Hongyoon Choi, PhD, Associate Professor, Nuclear Medicine, Seoul National University Hospital , Assoc Prof , Nuclear Medicine , Seoul National University Hospital
Hongyoon Choi, PhD, Associate Professor, Nuclear Medicine, Seoul National University Hospital , Assoc Prof , Nuclear Medicine , Seoul National University Hospital

Radiopharmaceutical therapy (RPT) is rapidly advancing; however, its progress has been limited by a narrow range of validated targets such as SSTR and PSMA. To achieve further breakthroughs, RPT development must expand toward new target-based programs and a deeper understanding of their mechanisms of action, grounded in translational oncology and multi-omics studies. Emerging methodologies that integrate human tumor atlas data, spatial biology, and AI-driven modeling enable systematic identification of novel targets and optimization of theranostic strategies. Together, these insights provide a scalable, data-driven roadmap for next-generation RPTs that complement existing antibody–drug conjugate and small-molecule modalities.

Developing Highly Specific TCEs against pHLA with Molecular Specificity Mapping

Photo of Marvin Gee, PhD, Co-Founder & Vice President, Target Discovery, 3T Biosciences , Vice President , Target Discovery , 3T Biosciences
Marvin Gee, PhD, Co-Founder & Vice President, Target Discovery, 3T Biosciences , Vice President , Target Discovery , 3T Biosciences

Peptide-HLAs are highly diverse tumor targets normally recognized by patient T cell immune responses. These targets can be discovered and targeted in solid tumors by leveraging off-the-shelf bispecific T cell engager therapies. We utilize an antibody-based discovery platform combined with high-throughput specificity mapping at the molecular resolution to develop highly potent T cell engagers. Preventing off-target specificity can ultimately improve the therapeutic index against pHLA targets.

Ice Cream & Coffee Break in the Exhibit Hall with Poster Viewing

TARGETS FOR AUTOIMMUNE, CARDIOVASCULAR, AND OTHER INDICATIONS OUTSIDE CANCER

Chairperson's Remarks

Marie-Eve Beaulieu, PhD, Co-Founder & Chief Development Officer, Peptomyc SL , CoFounder & Chief Development Officer , Drug Dev , Peptomyc SL

ANTXR1 Antibodies as a Universal Therapy for Cancer and Cardiovascular Disease

Photo of Bradley D. St. Croix, PhD, Head Tumor Angiogenesis, Mouse Cancer Genetics Program, NIH , Head Tumor Angiogenesis , Mouse Cancer Genetics Program , NIH
Bradley D. St. Croix, PhD, Head Tumor Angiogenesis, Mouse Cancer Genetics Program, NIH , Head Tumor Angiogenesis , Mouse Cancer Genetics Program , NIH

Heart disease and cancer—the two leading causes of death—share fibrosis as a common culprit. ANTXR1/TEM8, a pathology-induced protein critical for collagen turnover, drives both tumor growth and heart injury. This lecture shows how ANTXR1-neutralizing antibodies, originally developed for cancer, also protect the heart after myocardial infarction or pressure overload—reversing damage, restoring function, and revealing a shared mechanism linking two of humanity’s deadliest diseases.

Multifunctional Antibody Agonists for the Treatment of Autoimmune Pulmonary Alveolar Proteinosis (Apap)

Photo of Stefan Zielonka, PhD, Professor, Biomolecular Immunotherapy, Technische Universität Darmstadt , Senior Director , Antibody Discovery and Protein Engineering , Merck KGaA
Stefan Zielonka, PhD, Professor, Biomolecular Immunotherapy, Technische Universität Darmstadt , Senior Director , Antibody Discovery and Protein Engineering , Merck KGaA

Autoimmune pulmonary alveolar proteinosis (aPAP) is a rare disorder characterized by myeloid cell dysfunction, excessive pulmonary surfactant accumulation, and impaired innate immunity, primarily resulting from autoantibodies targeting GM-CSF, a cytokine essential for alveolar macrophage function. In this talk, the development of bispecific GM-CSF receptor agonists engineered to resist neutralization by polyclonal autoantibodies will be presented. These novel agonists effectively restore the functional capacity of macrophages and neutrophils, offering a promising therapeutic strategy for overcoming immune dysregulation in aPAP.

Enhanced Inhibition of Ocular Neovascularization with Novel Bispecific Receptor Decoy Proteins that Comprehensively Block Vascular Endothelial Growth Factor Ligands

Photo of Jamie B. Spangler, PhD, Associate Professor, Biomedical and Chemical & Biomolecular Engineering, Johns Hopkins University , Associate Professor , Biomedical Engineering and Chemical & Biomolecular Engineering , Johns Hopkins University
Jamie B. Spangler, PhD, Associate Professor, Biomedical and Chemical & Biomolecular Engineering, Johns Hopkins University , Associate Professor , Biomedical Engineering and Chemical & Biomolecular Engineering , Johns Hopkins University

Ocular neovascularization causes severe vision loss and is driven by the upregulation of vascular endothelial growth factor (VEGF) ligands; hence therapeutics that antagonize VEGF proteins have greatly advanced ocular disease treatment. However, many patients do not show benefit, in part since current therapies do not comprehensively block VEGF activity. We introduce 2 promising bispecific receptor decoy proteins that inhibit all VEGF ligands and significantly reduce ocular neovascularization in animal models.

From Biology to IND: Paradox's Approach to Protein Misfolding Diseases

Photo of Yulong Sun, PhD, Co-Founder & CSO, Paradox Immunotherapeutics , Co-Founder & CSO , Paradox Immunotherapeutics
Yulong Sun, PhD, Co-Founder & CSO, Paradox Immunotherapeutics , Co-Founder & CSO , Paradox Immunotherapeutics

The challenge of protein misfolding disease is targeting the often-scarce misfolded and pathological forms of the protein that cause disease, while avoiding the abundant, healthy form which performs important roles in the body. At Paradox Immunotherapeutics, we utilize a validated, structure-based approach to predict misfolding-specific epitopes to generate antibodies that target pathological proteins for clearance while avoiding the natively-folded species to improve organ function in systemic amyloidosis.

Networking Reception in the Exhibit Hall with Poster Viewing

Close of Emerging Targets in Oncology & Beyond Conference


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