Cambridge Healthtech Institute’s 4th Annual

Bispecific Antibodies for Cancer Immunotherapy

Engineering Next-Generation Biotherapeutics in Immuno-Oncology

October 4 - 5, 2021 ALL TIMES EDT

As the field evolves, the engagement of multiple receptors with bi- or multi-specific biotherapeutics is demonstrating great potential in achieving the efficacy and specificity needed to confront the most challenging cancers. Cambridge Healthtech Institute’s Fourth Annual Bispecific Antibodies for Cancer Immunotherapy conference will showcase advances in targeting strategies, conditional activation, engineering and architecture that will drive this exciting field to a new generation of safe and effective patient treatments.

Monday, October 4

7:30 am Registration and Morning Coffee

BISPECIFIC CAR T CELLS

8:25 am

Chairperson's Remarks

Alison Crawford, PhD, Senior Staff Scientist, Oncology & Angiogenesis, Regeneron Pharmaceuticals
8:30 am

New Shark and Camel Nanobody-Based CAR T Cells Targeting PD-L1 and B7-H3

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

Our laboratory has constructed the VHH and VNAR single domain antibody phage display libraries from camels (Camelus dromedarius) and sharks (Ginglymostoma cirratum). In my talk, I will discuss (i) isolation of camel VHHs and shark VNARs against PD-L1 and B7-H3, (ii) characterization of CAR T cells derived from these single domain antibodies for treating solid tumors including pancreatic cancer, neuroblastoma and hepatocellular carcinoma, and (iii) mechanisms of action related to NFkB and NFAT signaling.

9:00 am

Bispecific Anti-CD20, Anti-CD19 CAR T Cells for Relapsed B Cell Malignancies

Nirav N. Shah, MD, Associate Professor, Hematology, Medical College of Wisconsin

Bispecific anti-CD20, anti-CD19 CAR T cells are being explored as a potential second generation cell therapy treatment for B-cell non-Hodgkin lymphoma. Unique to our program is utilization of a point-of-care CAR T cell manufacturing process with the Prodigy CliniMACS device and exploration of a fresh (non-cryopreserved) infusion of CAR T cells. In this talk, we will review clinical outcomes of this vector, manufacturing platform, benefits of a fresh infusion, and future directions.

9:30 am KEYNOTE PRESENTATION:

Bispecific Antibodies Potently Neutralize SARS-CoV-2 Variants of Concern

Joshua Tan, PhD, Chief, Antibody Biology Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health

The emergence of SARS-CoV-2 variants of concern underscores the need for antibody-based tools that target multiple sites of the spike protein. We isolated 216 monoclonal antibodies and designed several bispecific antibodies that potently neutralize authentic SARS-CoV-2. Notably, two of nine bispecific antibodies neutralized the Alpha, Beta, Gamma and Delta variants and the wild-type virus with comparable potency. Thus, bispecific antibodies represent a promising next-generation countermeasure against SARS-CoV-2 variants of concern.

10:00 am Session Break

COSTIMULATORY BISPECIFICS

10:30 am

Investigating CD3 Bispecific Treatment of Solid Tumors Using PSMAxCD3

Alison Crawford, PhD, Senior Staff Scientist, Oncology & Angiogenesis, Regeneron Pharmaceuticals

We generated a bispecific antibody that targets both the prostate tumor–specific antigen PSMA and CD3 (PMSAxCD3). Mice expressing the human extracellular regions of CD3 and human PSMA were generated to examine antitumor efficacy in the presence of an intact immune system and PSMA expression in normal tissues. Although PSMAxCD3 induced T cell activation and showed antitumor efficacy in mice with low tumor burden, PSMAxCD3 lost efficacy against larger solid tumors. 4-1BB costimulation boosted efficacy against larger tumors, leading to durable antitumor responses.

11:00 am

Immunotherapy Combinations with Bispecific T Cell Engager Molecules Elicit Anti-Solid Tumor Regression in Immune ‘Cold’ Syngeneic Models Refractory to CTLA4/PD1 Checkpoint Blockade

Olivier P. Nolan-Stevaux, PhD, Senior Principal Scientist, Oncology, Amgen Inc.

T cell engager (TCE) molecules that induce T cell-mediated lysis of cancer cells have demonstrated promising anti-solid tumor activity, but the factors that limit the response to these therapies in vivo remain poorly understood. Using immunocompetent mice and surrogate TCE molecules, we demonstrate that pre-treatment tumor T cell density is critical to TCE therapy efficacy, characterize the CD8+ T cell-polarized response to TCE therapy, reveal the dual role of CD4+ T cells in TCE efficacy, and identify therapeutic combinations that cause the regression of poorly T cell-infiltrated cold solid tumors refractory to immune checkpoint blockade therapy.

Jenna Frame, Manager, Scientific Communications & Marketing, Marketing, Biocytogen

Mice expressing human immune checkpoints can serve as powerful preclinical models to evaluate the in vivo efficacy of anti-human antibody candidates for immuno-oncology targets. To ensure immunocompetence, knock-in humanized mouse models are carefully designed and subjected to rigorous validation. This session will highlight the use of humanized CD3E and CD3EDG models for broader targeting of the entire CD3 complex with novel mono- and bispecific antibody therapeutics.

Alex Chenchik, President and Chief Scientific Officer, Cellecta, Inc.

TCR/BCR repertoire profiling holds great potential for understanding disease mechanisms. We introduce a novel technology for profiling of all human TCR and BCR variable regions and phenotypic characterization of immune cells in bulk and at the single-cell level in PBMCs and immune cell fractions. Preliminary data shows that TCR/BCR clonotype analysis combined with targeted expression profiling of immune cells can be applied for large-scale discovery in several immune-responsive model systems. 

CONDITIONAL ACTIVATION

1:05 pm

Direct Control of CAR T Cells through Small Molecule-Regulated Antibodies

Javier Chaparro-Riggers, PhD, Executive Director, BioMedicine Design, Pfizer

We develop conditionally activated CARs to improve the safety profile of CAR T cells. The tumor antigen recognition is directly modulated by an FDA-approved small molecule drug. The resulting CAR T cells demonstrate specific cytotoxicity of tumor cells comparable to that of traditional CARs, but the cytotoxicity is reversibly attenuated by the addition of the small molecule.

1:35 pm

Conditionally Active T Cell Engager Engineered for the Treatment of Solid Tumors

Danielle Dettling, Senior Director, Research and Development, Maverick Therapeutics/Takeda Pharmaceuticals

Maverick has developed a novel recombinant bsAb platform called COBRA (Conditional Bispecific Redirected Activation). COBRAs are engineered to enable targeting of more widely expressed and validated tumor cell surface antigens by focusing T cell engagement within the tumor microenvironment. Maverick has (2) initial leads in clinical development, TAK-186 (EGFR-targeted COBRA) and TAK-280 (B7H3-targeted COBRA), which have the potential to treat a wide variety of tumor indications.  The data presented here will demonstrated the pre-clinical data package on both molecules, with a focus on TAK-280, which encompasses a unique targeting mechanism relative to our initial COBRA TAK-186.  

Juan Liang, Scientist, Research and Development Department, GemPharmatech Co., Ltd.

The humanized mice capable of evaluation of tumor microenvironment in vivo have provided a unique platform for immuno-oncology drug tests.  Genetically engineering mice and human immune system reconstituted mice show different features to facilitate drug evaluation, and model types related to efficacy and toxicity results.  We'll present the pre-clinical model choice based on drug design exemplified by Fc dependent antibody and T cell engager and showing toxicity evaluation data.

2:20 pm Sponsored Presentation (Opportunity Available)
2:35 pm Exhibit Hall with Poster Viewing

SAFETY AND DMPK FOR BISPECIFIC ANTIBODIES

3:25 pm

Anti-CD79b/CD3 T Cell Dependent Bispecific (TDB) for the Treatments of B Cell Malignancies: Impact Antibody Binding Properties on Pharmacokinetics and Pharmacodynamics

Rajbharan Yadav, PhD, Scientist, Development Sciences, Genentech

Preclinical studies demonstrated potency of anti-CD79b/CD3 TDB can be enhanced with increased binding affinity of either the anti-CD79b arm or the anti-CD3 arm. This evaluates the effect of CD3 binding affinities on PK and how it relates to the PD and safety. Anti-cynoCD79b/CD3 TDB was well tolerated, exhibited nonlinear PK consistent with target-mediated clearance (CLTotal range: 42-315 mL/day/kg) and resulted in dose-dependent B cell depletion.

3:55 pm

Development of Bispecific Antibodies against Solid Tumors Using DDS and Molecular Imaging

Masahiro Yasunaga, MD, PhD, Chief, Division of Developmental Therapeutics, National Cancer Center, Japan

We established in vivo imaging to visualize both antibody delivery and T cell migration and observed three phenotypes, (1) good accumulation of both antibodies and T cells, (2) good antibody delivery but suppressed T cell migration, and (3) suppression of T cells and antibodies. In solid tumors, (2) and (3) were predominant. Moreover, exhausted T cells lost proliferative and cytotoxic activities, which could attenuate bispecific antibody (BsAb) efficacy in vivo.  Hence, we are developing BsAb therapy in combination with immunoregulation using DDS and molecular imaging to overcome T cell deserts and exhaustion in the tumor microenvironment.

4:25 pm Close of Day

Tuesday, October 5

8:00 am Registration and Morning Coffee

EMERGING BI- AND MULTI-SPECIFIC PLATFORMS AND FORMATS

8:30 am

PM-IL15: A Novel IL-15-Based Immunocytokine with Unique Tumor Targeting Properties via Glycoepitope Recognition

Anika Jäkel, PhD, Director, Preclinical Pharmacology and Cancer Immunology, Glycotope, Germany

Glycoepitopes are potent targets for tumor therapeutics due to their unique tumor-specificity and broad indication coverage. We developed an IL-15-based immunocytokine (PM-IL15) targeting a conformational induced glycosylation/protein combined epitope (glycoepitope) on MUC1. PM-IL15 is designed to induce anti-tumor responses directly within the tumor microenvironment. It has high potential for the treatment of solid tumors either as monotherapeutic agent or as valuable combination partner for different anti-cancer drugs.

9:00 am

Augmented Bispecific Antibody-Based Anticancer Therapeutics Boost Neutrophil Cytotoxicity

Marjolein van Egmond, PhD, Professor, Oncology and Inflammation, Amsterdam UMC, The Netherlands

IgG mAbs eliminate tumor cells through NK cell-mediated ADCC and macrophage-mediated antibody-dependent phagocytosis. IgG, however, ineffectively recruits neutrophils as effector cells. By contrast, IgA mAbs induce neutrophil migration and activation through the IgA Fc receptor (FcaRI), but do not activate NK cells and have poorer half-life. We combined the agonistic activity of IgG mAbs and FcaRI targeting in a bispecific antibody, and results with this novel molecule will be discussed.

9:30 am Sponsored Presentation (Opportunity Available)
10:00 am Exhibit Hall with Poster Viewing
10:40 am

Tumor-Localizing Bispecific Antibodies

Peter Ellmark, PhD, Vice President, Discovery, Alligator Bioscience AB

Bispecific antibodies have the potential to provide new options in indications where checkpoint inhibitors fail. T cell redirecting CD3 antibodies have paved the way for this class of immunotherapies. A new promising class of tumor associated antigen (TAA) conditionally active agonistic bispecific antibodies that modulates checkpoint co-stimulators, e.g., CD40 and 4-1BB, has recently emerged as a promising approach to enhance T cell priming and activity in immuno-oncology. Alligator develops bispecific antibodies targeting both TAA x CD40/TAA x 4-1BB and data from these programs will be presented.

11:10 am

Molecular Mechanism of HER2 Rapid Internalization and Redirected Trafficking Induced by Anti-HER2 Biparatopic Antibody

Inna Vainshtein, PhD, Senior Director, Bioanalytics, Exelixis

Receptor-mediated internalization is an essential part of MOA for antibody drug conjugates (ADC). A bispecific format of ADC may affect its internalization properties. Contrary to parental arm antibodies a biparatopic anti-HER2 antibody (anti-HER2-Bs), targeting two non-overlapping epitopes on HER2, induced rapid internalization and efficient degradation of receptors. Our study investigated the molecular mechanism of rapid internalization and redirected trafficking providing insights in drug MOA and rational design of bispecific molecules.

11:40 am

CD38-CD3 Bionics in AML Treatment

Flavia Pichiorri, PhD, Associate Professor, Hematologic Malignancies Translational Science, Briskin Center for Multiple Myeloma Research, City of Hope

On the basis of technology developed at City of Hope, we have developed CD38-CD3 Biologics Nested Inside Chains (BIONICS), which comprise a CD38 nanobody linked to an anti-CD3 antibody via a short peptide linker. Our data show that CD38-CD3 BIONICS induces remarkable T cell activation and can eradicate acute myelogenous leukemia (AML) cells not only in vitro but also ex-vivo in autologous patient setting and in vivo animal models.

12:10 pm Close of Bispecific Antibodies for Cancer Immunotherapy





Preliminary Agenda

Conference Programs