Cambridge Healthtech Institute's 6th Annual

Immuno-Oncology Biomarkers

Predictive Biomarkers in Immunotherapy

October 4 - 5, 2021 ALL TIMES EDT

As pharmaceutical and biotechnology companies increase their investment in immuno-oncology programs to facilitate rapid development of novel immunotherapies, there is increasing pressure to discover and validate relevant biomarkers. Cambridge Healthtech Institute’s 6th Annual Immuno-Oncology Biomarkers conference will bring together biomarker experts from industry and academia to address rapid development of predictive and prognostic IO biomarkers, utility of these biomarkers in clinical trials, and their potential as companion diagnostics.

Monday, October 4

7:30 am Registration and Morning Coffee

ADVANCING PRECISION MEDICINE WITH LIQUID BIOPSY

8:25 am

Chairperson's Opening Remarks

Sam Hanash, MD, PhD, Director, Red & Charline McCombs Institute; Evelyn & Sol Rubenstein Distinguished Chair, Cancer Prevention; Professor, Clinical Cancer Prevention-Research, Translational Molecular Pathology, University of Texas MD Anderson Cancer Center
8:30 am

Circulating Exosomes and Their Cargo as Immuno-Oncology Biomarkers

Sam Hanash, MD, PhD, Director, Red & Charline McCombs Institute; Evelyn & Sol Rubenstein Distinguished Chair, Cancer Prevention; Professor, Clinical Cancer Prevention-Research, Translational Molecular Pathology, University of Texas MD Anderson Cancer Center

There is currently substantial interest in liquid biopsy approaches to profile tumors. Profiling of the exosome surfaceome and cargo has yielded immuno-oncology biomarkers that inform about tumor status and potential response to immunotherapy.

9:00 am

Small Extracellular Vesicles (sEV) in Plasma of Cancer Patients as Contributors to Liquid Tumor Biopsy

Theresa L. Whiteside, PhD, Professor, Pathology, Immunology & Otolaryngology, University of Pittsburgh

Exosomes, a subset of small extracellular vesicles (EVs), are emerging as key components of liquid tumor biopsy in cancer and as biomarkers of immune competence. Plasma-derived exosomes can be separated by immune capture into tumor cell-derived and T cell-derived fractions. Each fraction carries information about the tumor or immune competence of the cancer patient, respectively. Simultaneous real-time molecular profiling of the tumor and of the host immune status by plasma-derived exosomes suggest that sEVs are highly promising biomarkers in immuno-oncology.

9:30 am

Liquid Biopsy to Guide Immunotherapy

Catherine Alix-Panabières, PhD, Associate Professor and Director, Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Center of Montpellier, France

The emergence of immunotherapy in oncology requires the discovery, validation and subsequent adoption of robust, sensitive and specific predictive and prognostic biomarkers for daily practice. The use of a liquid biopsy could provide an important complementary or alternative added value to PD-L1 detection in tissue biopsy. In my talk, I will discuss how liquid biopsy could be used in the field of immuno-oncology to predict response or relapse for patients undergoing immune-checkpoint inhibitor therapy.

Michael Brenan, Proteomics Specialist, IsoPlexis
10:15 am Session Break

PREDICTING CLINICAL OUTCOME

10:30 am

Analysis of Peripheral Blood Markers in Patients Receiving Checkpoint Blockade: Pretreatment Characteristics and On-Treatment Pharmacodynamics

Margaret K. Callahan, MD, PhD, Medical Oncologist, Memorial Sloan Kettering Cancer Center

Using data generated from immune profiling of peripheral blood samples collected from ICB-treated patients, we applied a novel supervised statistical learning approach to classify phenotypes and determine their association with survival and treatment response. We will describe how baseline characteristics of T cells in the peripheral blood associate with clinical outcomes.

11:00 am

Activated Natural Killer Cells Predict Poor Clinical Prognosis in High-Risk B- and T-Cell Acute Lymphoblastic Leukemia

Caroline Duault, PharmD, PhD, Research Scientist, Institute for Immunity, Transplantation and Infection, Stanford School of Medicine

B- and T- cell acute lymphoblastic leukemia (B/T-ALL) may be refractory or recur after therapy by suppressing host anti-cancer immune surveillance mediated specifically by natural killer (NK) cells. In order to elucidate the role of NK cells in sustaining ALL regression, we delineated the phenotypic and functional defects in NK cells of high-risk B/T-ALL patients using mass, flow, and in silico cytometry. We found that, compared to normal counterparts, NK cells in B/T-ALL patients exhibit an activated signature characterized by high CD56 and high CD69 expression, production of activated NK-origin cytokines, and calcium signaling. However, we demonstrated that defective maturation of NK cells into cytotoxic effectors prevents NK cells of ALL patients from lysing NK-sensitive targets as efficiently as normal NK cells, despite their activated phenotype. Additionally, we showed that NK cells in ALL are exhausted, which is likely caused by their chronic activation. We found that increased frequencies of activated cytokine-producing NK cells are associated with increased disease severity and independently predict poor clinical outcome in ALL patients. The comprehensive analysis of the NK cell phenotype and functions in ALL patients highlighted the relevance of developing NK cells as diagnostic and prognostic tool to predict clinical outcome and underscore the clinical potential of allogeneic NK infusions to prevent ALL recurrence.


Keith Wharton, MD PhD FCAP, Vice President, Medical Director, Ultivue

Utility of multiplex immunohistochemistry/immunofluorescence (mIHC/IF) highlights the spatial distribution of infiltrating immune cells within the tumor immune microenvironment that allow a detailed characterization of specific cell phenotypes defined by co- or lack of expression of multiple markers that may help in predicting clinical responses and mechanisms of resistance to therapy. In this presentation we will show a unique workflow supporting whole slide imaging of a high-plex mIF and traditional same slide H&E for comprehensive tissue immunophenotyping analysis.

 

12:00 pm Session Break
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. 

BIOMARKER STRATEGIES IN CANCER IMMUNOTHERAPY

12:45 pm

Chairperson's Remarks

Alain Algazi, MD, Associate Professor, Medicine, University of California, San Francisco
12:50 pm

Translational Research Insights Leveraging Predictive Biomarkers of Response to Anti-PD-1 Therapy

 

Terri K. McClanahan, PhD, Executive Director, Molecular Discovery Biologics, Merck Research Labs

Pan-cancer molecular biomarkers of immunotherapy response can identify patients likely to derive benefit from PD-1/PD-L1-directed monotherapy. Clinical genomic data has also illuminated mechanisms of resistance to immunotherapy which can be leveraged to drive rational combinations and novel drug discovery. Data will be presented showing key determinants of response to pembrolizumab across multiple tumor types, as well as emerging resistance signatures, which provide a framework for novel approaches to cancer therapy.

1:20 pm

Dissecting Mechanisms of Resistance to PD-1/PD-L1 Blockade in Bladder Cancer

Matthew D. Galsky, MD, Division of Hematology and Medical Oncology, Professor of Medicine, Icahn School of Medicine at Mount Sinai; Associate Director, Translational Research, Tisch Cancer Institute

Several biomarkers associated with response and resistance to immune checkpoint blockade in urothelial cancer have been described. However, how these different cancer cell, stromal, and immune cell features are interrelated, and which are independently associated with outcomes, are not well defined. Approaches to identify independent features associated with immune checkpoint blockade outcomes for prioritization as biomarkers and therapeutic targets will be described.

Darren Davis, Senior Vice President, Precision for Medicine

Tumor biopsies and advanced multiplex immunofluorescence enable quantification of immune cell subsets including their precise placement within the tumor microenvironment—yet getting samples can put patients at risk. Liquid biopsies are less invasive and enable monitoring of circulating biomarkers. In this presentation, we will show an innovative strategy utilizing liquid biopsies to assess biomarkers in both circulating tumor cells (CTCs) and peripheral immune cells from the same patient sample.

Robert Georgantas, PhD, Senior Vice President of Research and Translational Science, Biodesix

AI and Machine Learning (AI/ML) have enabled the development of diagnostic tests using unbiased -omics level data. In this presentation, we focus on the use of unbiased liquid biopsy-based proteomics for the development of diagnostic tests that predict response of lung cancer patients to immune checkpoint inhibitors (ICIs). Finally, we will introduce methods in translational science to examine the biologic underpinnings that drive test outcomes.

2:35 pm Exhibit Hall with Poster Viewing

INFORMING CLINICAL DEVELOPMENT

3:25 pm

Evaluating Biomarkers of JTX-8064 (Anti-LILRB2/ILT4 mAb) to Inform Clinical Development

Johan Baeck, MD, Senior Vice President, Clinical Development and Medical Affairs, Jounce Therapeutics

JTX-8064 is a LILRB2/ILT4 antagonist mAb that may relieve myeloid cell immuno-suppression by shifting macrophages to an M1-like state and activating T cells. Using histoculture, pharmacodynamic (PD) responses to JTX-8064 can be measured preclinically in a human ex vivo tumor system. Ex vivo evaluation of human tumors identified hypotheses for both predictive and pharmacodynamic markers that can be evaluated in the clinical development of JTX-8064.

3:55 pm

Durable Responses and Immune Activation with Intratumoral Electroporation of pIL-12 Plus Pembrolizumab in Actively Progressing Anti-PD-1 Refractory Advanced Melanoma: KEYNOTE 695 Interim Data

Alain Algazi, MD, Associate Professor, Medicine, University of California, San Francisco

In patients with rigorously defined anti-PD-1 refractory advanced melanoma, we show that increasing intratumoral IL-12 through plasmid electroporation induced deep, durable, systemic responses including regression of visceral lesions with nominal systemic toxicity. This work represents the final stage of translation of our earlier biomarker findings using an approach that adds negligible toxicity to standard-of-care checkpoint inhibitor therapy.

4:25 pm Interactive Discussions

Interactive Discussions are informal, moderated discussions, allowing participants to exchange ideas and experiences and develop future collaborations around a focused topic. Each discussion will be led by a facilitator who keeps the discussion on track and the group engaged. For in-person events, the facilitator will lead from the front of the room while attendees remain seated to promote social distancing. To get the most out of this format, please come prepared to share examples from your work, be a part of a collective, problem-solving session, and participate in active idea sharing. Please visit the Interactive Discussion page for a complete listing of topics and descriptions.

Immuno-Oncology Biomarkers

Tullia C. Bruno, PhD, Assistant Professor, Immunology, University of Pittsburgh & Hillman Cancer Center
  • Predictive biomarkers of response: systemic or local? Or a combination of both?
  • Spatial interrogation of immune cells for improved immunotherapies
  • Moving beyond the T cell: what other components of the TME should we be targeting?​
5:15 pm Close of Day

Tuesday, October 5

8:00 am Registration and Morning Coffee

PREDICTING RESPONSE AND RESISTANCE TO IMMUNOTHERAPY

8:25 am

Chairperson's Remarks

Evisa Gjini, PhD, MBA, Director, Solid Tumors Oncology, Translational Medicine, Bristol-Myers Squibb
8:30 am KEYNOTE PRESENTATION:

Current and Future Methods of Predicting Response to Immunotherapy

David L. Rimm, MD, PhD, Professor, Pathology and Medicine (Oncology); Director, Translational Pathology, Yale University School of Medicine

Companion diagnostics for immunotherapy are not very sensitive or specific, but sufficient to enrich the population of responders to immunotherapy. This session will take a closer look at the only FDA approved methods (IHC and TMB) and then examine some data on potential future methods.

9:00 am

Mechanistic Basis of Response to PD-1 Blockade

Adil Daud, MD, Professor, Hematology/Oncology, University of California, San Francisco; Director, Melanoma Clinical Research, UCSF Helen Diller Family Comprehensive Cancer Center

The use of checkpoint inhibitors for cancer has revolutionized immunotherapy. Certain features of immunotherapy are clear: tissue type, obesity, age, gender, tissue of origin, tumor mutation burden and most importantly the presence of tumor-directed T cells in the tumor microenvironment are critical features. I explore recent publications showing T cell replacement and replenishment as a critical feature and the interesting data from tumor models that informs immunotherapy. 

Anja van de Stolpe, Chief Scientific Officer, Philips Molecular Pathway Diagnostics

Signal transduction pathways control the functional state of the different immune cell types. OncoSignal RT-qPCR tests quantify the activity of signaling pathways in immune cells from blood or tissue samples. Tests can be used to assess activation of the innate and adaptive immune response, enabling use to predict and monitor response to immunotherapy. OncoSignal-based companion diagnostics will be commercialized in a partnership with QIAGEN.

 

10:00 am Exhibit Hall with Poster Viewing
10:40 am

Understanding Mechanisms of Resistance to Therapy in Prostate Cancer

Evisa Gjini, PhD, MBA, Director, Solid Tumors Oncology, Translational Medicine, Bristol-Myers Squibb

Prostate cancer (PCa) is primarily driven by androgen receptor (AR) signaling, with treatment dominated by androgen depravation therapies (ADT). Although the prognosis for low-grade localized PCa is favorable, about half of patients with high-grade localized disease develop metastatic castration resistant PCa (mCRPC). The 5-year survival of mCRPC is only 30%, highlighting an unmet therapeutic need for this subset of patients. PCa is characterized by a low level of tumor infiltrating lymphocytes and is considered a “cold” tumor. Even when CD8+ cells are present in the tumor and stroma, they have been shown to poorly correlate with prognosis, perhaps due to non-active or senescent cell states. Tregs, TAMS, reactive fibroblasts and myofibroblasts, growth factors and cytokines also contribute to the immuno-suppressive environment, tumor invasion, angiogenesis and tumor-stromal cross-talk of PCa. Through an integrative tumor and immunological molecular profiling of primary low- and high-grade treatment-naïve PCa samples, we have identified a subset of high-grade PCa patients who relapse under SOC that may benefit from the addition of adjuvant immune checkpoint blockade (ICB) to SOC to overcome immune suppression. Our analysis suggests that high-grade PCa is characterized by low antigenicity as assessed by loss of MHC-I protein expression and an immunosuppressive microenvironment rich in CAFs, macrophages, T-regs and T-cell exclusion phenotypes. Unlike low-grade, high-grade PCA can have a poor prognosis (within 5 year relapse). However, a subset of high-grade PCa patients that metastasized while on SOC exhibited a biomarker profile that favors the combination of SOC with ICB (lower tumor AR expression, retained tumor MHC-I expression, moderate CD8+ T-cell infiltration and a high IFN-γ RNA signature).

11:10 am

Multimodality Assessment of Melanoma Immunotherapy Response and Resistance

Genevieve Boland, MD, PhD, Assistant Professor, Surgery; Director, Melanoma Surgery Program, Massachusetts General Hospital

Despite progress in melanoma treatment with immunotherapy, many patients develop resistance and succumb to their disease. Analysis of patient-derived tumors and blood samples have allowed us to understand mechanisms of therapeutic response and resistance. We are using multi-omic strategies to identify novel therapeutic targets and biomarkers to assist in risk stratification and disease management.

11:40 am

Use of Machine Learning in Predicting Immune Therapy Response

Iman Osman, MD, Rudolf L. Baer Professor of Dermatology, Professor, Medicine & Urology, New York University Langone Medical Center

Several biomarkers of response to immune checkpoint inhibitors show potential but are not yet scalable to the clinic. We developed a pipeline that integrates deep learning on histology specimens with clinical data to predict ICI response in melanoma. We built a multivariable classifier that predicted response with AUC 0.80. With prospective validation, we believe our approach has potential for integration into clinical practice.

12:10 pm Close of Immuno-Oncology Biomarkers





Preliminary Agenda

Conference Programs