Cambridge Healthtech Institute's 6th Annual

Preclinical and Translational Immuno-Oncology

Predictive Preclinical Models and Translational Strategies for Cancer Immunotherapy

October 5 - 6, 2021 ALL TIMES EDT

The recent advancements in immunotherapies, such as immune checkpoint modulators, bispecific antibodies, and adoptive T cell transfer, are shifting the way cancer patients are treated. Rapid development of novel immuno-oncology programs is creating the need for predictive preclinical models and translational strategies to understand combination immunotherapy, study responses and resistance to cancer immunotherapy, and identify novel biomarkers and targets. Please join Cambridge Healthtech Institute’s 6th Annual Preclinical and Translational Immuno-Oncology meeting and learn about new clinically relevant models for screening IO therapies and translational strategies to support clinical IO programs.

Tuesday, October 5

12:30 pm Registration

KEYNOTE PRESENTATION

Oliver Jonas, Scientific Founder, Kibur Medical; and Assistant Professor in Radiology, Harvard Medical School, Kibur Medical
1:30 pm KEYNOTE PRESENTATION:

Precision Medicine in Immuno-Oncology: Selecting for Monotherapy, Understanding Resistance Biology, and Informing Combination Therapies

Roy Baynes, MD, PhD, Senior Vice President & Head, Global Clinical Development; CMO, Merck, Sharp, and Dohme

Precision medicine tools have enabled PD-1 antibody therapy to transform cancer care in a number of major tumor types. They have also provided insights to resistance biology and potential logical choices for combination therapies. Combination therapies with PD-1 antibodies as the backbone are beginning to impact many therapeutic choices.

PK/PD MODELS FOR IMMUNO-ONCOLOGY

2:00 pm

Evaluating Strategies for Overcoming Rituximab Resistance Using a Quantitative Systems Pharmacology Model of Antibody-Dependent Cell-Mediated Cytotoxicity and Phagocytosis

Dean Bottino, PhD, Senior Scientific Director, Clinical Translational Modeling & Simulation Leader, Takeda

Given that antibody dependent cell mediated cytotoxicity (ADCC) and phagocytosis (ADCP) are thought to be the major mechanisms of action of Rituximab (R), increasing the activation levels of natural killer (NK) and macrophage (MP) cells may be one strategy for overcoming R resistance. We have developed and calibrated to ex-vivo literature data a quantitative systems pharmacology (QSP) model of ADCC/ADCP to interrogate which mechanisms of R resistance could be overcome by increased NK or MP activation, and how much effector cell activation would be required to overcome a given degree and mechanism of R resistance. This ADCC/ADCP model was then incorporated into an in-house QSP model built with ex-vivo and clinical data to support the development of a Takeda investigational drug which is being developed to restore R sensitivity in an R-resistant patient population.

2:30 pm

Leveraging PK/PD Modeling to Speed Up the Path to Proof-of-Concept in Immuno-Oncology

Arijit Chakravarty, PhD, CEO, Fractal Therapeutics

Most preclinical in vivo efficacy models supporting drug discovery and development in immuno-oncology have not been proven to be translationally predictive. Successfully treating a preclinical immuno-oncology model does not predict for success in the clinic, in contrast to the situation for traditional oncology. This lack of translational relevance represents a significant (and often underappreciated) challenge for drug development in this field. In the absence of translational efficacy models, how do you rationally plan and execute a preclinical-to-clinical translation strategy? In this talk, we will discuss how to make that happen, relying in particular on ex vivo assays using human whole blood, PK/PD modeling for markers that are directly downstream of drug action (such as target occupancy), and Bayesian trial design approaches. This talk, which provides practical guidance for drug developers in immuno-oncology, will help attendees in the design of robust science-driven strategies for establishing clinical proof of concept for their assets.

Oliver Jonas, Scientific Founder, Kibur Medical; and Assistant Professor in Radiology, Harvard Medical School, Kibur Medical

The implantable microdevice (IMD) is a translational tool that allows us to systematically screen many individual and combination therapies in a small cohort of study animals.

 

This session explains how IMDs work, and how we use them to facilitate the collection of robust data for numerous applications, including: 

  • Combination screening
  • Testing of compounds that have no PK or toxicology data
  • Candidate comparison
  • Specific biology, e.g., candidate interaction with TME
  • Clinical translation
3:30 pm Exhibit Hall with Poster Viewing
4:10 pm Transition to Plenary Keynote

PLENARY KEYNOTE SESSION: ADVANCING PRECISION IMMUNO-ONCOLOGY

4:20 pm

Plenary Keynote Introduction

Benjamin G. Neel, MD, PhD, Professor, Medicine, NYU Grossman School of Medicine; Director, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health
4:25 pm

The Rapid Evolution of Precision IO: The Future Role of Biomarkers in IO Development and Clinical Utilization

Zhen Su, MD, MBA, CEO, Marengo Therapeutics

As we turn the corner into a new decade, scientific and technological advances are helping to achieve a more precision approach for immunotherapy. While PD-L1 staining offers enrichment, mutation burden, microsatellite instability, and other immune biomarkers may be able to advance the precision approach. Evolution and real-world adaptation of testing tools, such as companion diagnostic capabilities, will become the new focal point to advance patient care.

4:55 pm

Lessons Learned from BCMA and CD19 CAR T Trials

Kristen M. Hege, MD, Senior Vice President, Early Clinical Development, Hematology/Oncology and Cell Therapy, Bristol-Myers Squibb

Dr. Hege will discuss drawing correlations between patient and product characteristics and clinical outcomes for iterative improvement; identifying variables associated with CAR T expansion and persistence; understanding mechanisms and predictors of non-response, durable response and tumor escape; and new directions for cell therapy.

5:25 pm Immuno-Oncology Summit Connects

Explore new products and services in our Exhibit Hall, engage with poster presenters, schedule 1-on-1 meetings, and build your research community during this open networking period.

6:25 pm Close of Day

Wednesday, October 6

8:00 am Registration and Morning Coffee

MODELS AND STRATEGIES FOR CANCER IMMUNOTHERAPY

8:25 am

Chairperson's Remarks

Russell Jenkins, MD, PhD, Center for Cancer Research, Massachusetts General Hospital
8:30 am

Mouse Models to Test Therapeutic Bispecific Antibodies

Gavin Thurston, PhD, Senior Vice President, Oncology Research, Regeneron Pharmaceuticals
9:00 am

Novel Preclinical Models to Evaluate Immune Oncology Therapeutics

Vish Muthusamy, PhD, Executive Director, Center for Precision Cancer Modeling, Yale School of Medicine

We have developed syngeneic, engineered mouse lines which, when implanted into the left ventricle, result in disseminated metastases that can be visualized by bioluminescent imaging at very early stages of establishment. This model can be used in testing efficacy of immune therapies targeting metastatic tumors. In combination with sub-cutaneously implanted non-luminescent tumor cell line counterpart, we have successfully demonstrated treatment of primary tumor with experimental immune therapy agents resulting in abscopal effects on deep-seated metastases. We have also developed an in vitro patient-derived explant (PDE) model preserving the tumor immune microenvironment including the infiltrating immune cells in a viable and functional state for greater than a month. We have successfully demonstrated proof of concept and observe complete elimination of tumor cells following in vitro treatment with an immune checkpoint inhibitor. This platform could also eventually be used to perform real time multiplexed testing to enable personalized immuno-oncology therapy.

9:30 am

Overcoming Resistance to Anti-BCMA Bispecific Antibody in a Syngeneic Mouse Model of Multiple Myeloma through Combination Therapy

Marta Chesi, PhD, Associate Professor, Medicine, Mayo Clinic

Anti-BCMA bispecific antibodies are very effective against relapse/refractory myeloma. However, patients inevitably relapse. Using the immunocompetent Vk*MYC mouse model of myeloma, we found that tumor burden and T cell exhaustion limit the efficacy of anti-BCMA/CD3 antibody. The addition of an IMiD only transiently deepened the response, but further exacerbated T cell exhaustion. Surprisingly, concurrent treatment with anti-BCMA/CD3 and cyclophosphamide proved very effective, prevented exhaustion and provided immunological protection against tumor reoccurrence.

Jason Ptacek, Associate Director, Research Services, Ionpath

Dr. Ptacek will provide an overview of how high-definition spatial proteomics, enabled by Multiplexed Ion Beam Imaging (MIBI), can be used to explore the tumor microenvironment with unprecedented depth. Examples of quantitative single-cell phenotype mapping in tissue analysis applications and the ability to generate actionable insights from this highly multiplexed, spatial proteomic data will be presented.

10:30 am Exhibit Hall & Last Chance for Poster Viewing

ORGANOID AND SPHEROID MODELS

11:20 am

Exploiting Genetically Defined, Organoid-Based Models of High-Grade Serous Ovarian Cancer

Benjamin G. Neel, MD, PhD, Professor, Medicine, NYU Grossman School of Medicine; Director, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health

Current therapies, including conventional chemotherapy, targeted therapy, and immune therapy are typically tested in different types of preclinical models. Chemotherapy and targeted therapy are often analyzed using cancer cell lines and xenografts, patient-derived xenografts and more recently, patient-derived organoids. These models have the genetics of the human disease but lack a functional immune system/tumor microenvironment (TME). Immune-therapies have been tested primarily in "syngeneic tumor models" of unclear cell-of-origin and "irrelevant" genetics. Using mouse fallopian tube organoids, we engineered a suite of new, genetically defined models of high-grade serous ovarian cancer. I will discuss the use of these models to derive new combination therapies for this deadly disease.

11:50 am

In vitro Models for Cancer Immunity and Immunotherapy

Shay Soker, PhD, Professor, Wake Forest Institute for Regenerative Medicine; Director, Wake Forest Organoid Research Center

3D human tissue organoids replicate native tissue structure and function and can be studied in vitro for several weeks to allow intensive investigations. We have used human tissue organoids to study cancer progression, metastasis and response to therapy. Patient-specific tumor organoids, consisting of tumor and stromal cells could replicate the patient’s own treatment response. We have recently improved the tumor organoids’ capabilities by adding immune cells to create immune-responsive tumor organoids.

12:20 pm

Ex vivo Profiling of Novel Cancer Therapies Using Organotypic Tumor Spheroids

Russell Jenkins, MD, PhD, Center for Cancer Research, Massachusetts General Hospital

Preclinical models that translate to human immunity are needed to assess novel cancer immunotherapy combinations. New approaches and model systems are needed to deprioritize ineffective strategies and to better understand mechanisms of response and resistance to promising approaches. To address this need, we have developed a system for ex vivo profiling of novel immunotherapy combinations using 3D microfluidic culture of murine- and patient-derived organotypic tumor spheroids (MDOTS/PDOTS). 

12:50 pm Enjoy Lunch on Your Own

MECHANISMS AND TARGETS IN IMMUNO-ONCOLOGY

1:50 pm

Chairperson's Remarks

Katherine Seidl, PhD, Head, Oncology Drug Discovery Unit, Takeda Pharmaceuticals International Co.
1:55 pm

Decoding Immune Signatures to Guide Therapeutic Design

 

Katherine Seidl, PhD, Head, Oncology Drug Discovery Unit, Takeda Pharmaceuticals International Co.

Innovative immuno-oncology therapeutics require focus on various underexplored mechanisms in the cancer immunity cycle. We are building a framework to identify the key determinant mechanisms and parameters that can be used to link patients with therapies. Allogeneic cell therapy platforms will be discussed in this context as an example with application to other therapeutic approaches. Attributes regarding patient population, cell platform, targets, and armoring options are taken into account.

2:25 pm

ImmunoPerturbomics: New Targets, Mechanisms and Modalities in Immuno-Oncology

Shashank Patel, PhD, Director, R&D Immunology, NextCure

CRISPR-Cas9 cell engineering is making great strides in driving novel immuno-medicines to the clinic. This talk will focus on how this technology is uncovering the mechanisms of immune resistance to checkpoint therapies and identifying next-generation IO targets and modalities.

2:55 pm Session Break
3:10 pm

Neoantigens and Inhibigens Selected with the ATLAS Bioassay Have Differential Impacts on Tumor Immunotherapy: Preclinical and Clinical Results

Jessica Baker Flechtner, PhD, CSO, Genocea Biosciences
3:40 pm

Preclinical Development of an Anti-PD-1-GITR-L Agonist Bispecific for Immuno-Oncology

Hamsell M. Alvarez, PhD, Associate Director, Oncology Discovery, AbbVie

Since PD-1 and GITR are co-expressed on recently activated antigen-specific T cells and on memory cells, a bispecific that targets both specificities is warranted. Anti-PD-1-GITR-L is a bispecific molecule that overcomes immune escape to PD-(L)1 blockade by enhancing and sustaining the activation, proliferation, and memory differentiation of primed antigen-specific T cells by inducing a PD-1 mediated GITR clustering. The MoA is independent of FcγR-mediated cross-linking, and ADCC-mediated Treg depletion.

4:10 pm

CD24 Signaling through Macrophage Siglec-10 Is a Target for Cancer Immunotherapy by Macrophages

Amira Barkal, MD, PhD, Resident Physician, Internal Medicine, Brigham & Women’s Hospital, Harvard Medical School

Cancer cells evade clearance by macrophages through the overexpression of anti-phagocytic surface proteins called ‘don’t eat me’ signals. Here we report that CD24 is the dominant ‘don’t eat me’ signal in multiple solid tumors. In preclinical models, CD24 blockade leads to a macrophage-dependent reduction in tumor growth and extension of survival. Collectively, these data support the therapeutic potential for targeting CD24, and demonstrate the importance of assessing the global expression of innate immune checkpoints in order to inform therapeutic strategies with broad, durable responses.

4:40 pm Close of Summit





Preliminary Agenda

Conference Programs