Immuno-Oncology Xchange
East Coast 2021
May 25 & 27
Welcome to hubXchange’s virtual East Coast Immuno-Oncology Xchange 2021, bringing together executives from pharma and biotech to address and find solutions to the key issues faced in developing immuno-oncology therapies.
Discussion topics will cover Immune Biomarkers & Translational Research, Preclinical Research, Clinical Development, Cell Therapies and Next-Generation Therapies.
Take advantage of this unique highly interactive meeting format designed for maximum engagement, collaboration and networking with your peers.
Immune Biomarkers & Translational Research
- Exploring the dynamic nature of the TME
- The need for new biomarker discovery and development
- Harnessing the ability to derive more meaningful information from precious tissue samples
- Novel strategies for advanced image analysis
Vice President, Digital Health Strategies
Ultivue
Florian Leiss is VP Digital Health Strategies at Ultivue. He is responsible for digital and data-driven offerings and image analysis. Florian has been working for the 10+ years in software development for healthcare to make AI-powered CDx in oncology a reality, to support radiologists with routine reporting, or to empower pathologists with image analysis at AstraZeneca, Smart Reporting and Definiens. He holds PhDs in neuroscience and philosophy.
- Few pre-clinical models available to test predictive biomarker hypotheses early during drug discovery
- Default is often to use the target itself as the best predictive biomarker, is it the best choice?
- Frequency of target expression can be derived from multiple sources, but there is no clear link between target expression and potential for response
- Extensive clinical experience is still required to identify best predictive biomarkers and their appropriate cut-offs
Senior Director, Global Head, Clinical Biomarker Strategy
EMD Serono
Isabelle has extensive experience and expertise in IO drug discovery and development with current focus in clinical biomarkers and companion diagnostics. Isabelle was in charge of developing the small molecule IO pipeline at Bristol Myers Squibb in 2014/2015. She has since joined EMD Serono as Global Head of Clinical Biomarker Strategy where she leads a Team which supports IO, Oncology and Immunology products in the pipeline. She has also led the clinical biomarker and patient selection strategy and is co-author on several publications for a bi-functional IO molecule, bintrafusp alfa, under development at EMD Serono.
- How do we optimize early stage clinical study design for exploratory biomarker development?
- How do we take advantage of peripheral immune components (autoantibodies, exosomes, T cell markers, etc.) to understand the tumor immune micro-environment?
- Current diagnostic criteria in cancer immunotherapy doesn’t accurately identify subjects likely to respond. How do we use what’s available develop rational combination therapies with other immunotherapies or chemotherapies?
Director, Translational Biology
Macrogenics
Patrick Kaminker is an industry scientist that received his degree in Pharmacology from the University of Kentucky. After focusing his postdoctoral studies on the intersection of telomeres with aging and cancer in Dr. Judy Campisi’s lab at the Lawrence Berkeley National Labs he joined the Buck Institute for Age Research. Dr. Kaminker then spent several years at Celera and Human Genome Sciences in the pursuit of novel targeted cancer therapies. He took an interest in translational research while at Human Genome Sciences while looking for biomarkers associated with TRAIL agonists and how a certain population had shown durable anti-tumor activity associated with certain immune signatures. Since that time, he has been focused on the translational aspects of multiple classes of therapeutics including: epigenetic modifiers, antibody-drug conjugates, and recently cancer immunotherapies and currently runs the translational group at Macrogenics.
1:00 – 1:30pm
The need to better understand the tumour microenvironment (TME) dictates the characterization of the cell types involved, the roles they play and how they respond to treatment. To develop better cancer immunotherapies, in vitro primary immune cell bioassays offer an early assessment of their effects on the various players of the TME.
Cytotoxic cells are key players in the anti-tumour immune response. CD8+ T cells, also known as cytotoxic T lymphocytes, recognize and kill cells presenting antigens bound to MHC class I molecules, such as neoantigen present on the tumour cells.
Activation of CD8+ T cells requires at least two signals from antigen presenting cells: binding of their T cell receptor (TCR) to the appropriate peptide presented on MHC I and co-stimulation by B7 binding to CD28. However, tumour environment developed different mechanisms to reduce this immune cell killing such as checkpoint inhibitors expression. In that case, the immune system will be dampened. A second type of cytotoxic cells, named Natural killer cells, are an innate immune cell type that plays an important role in anti-cancer immunity. NK cells target cells lacking MHC I expression, including cancer cells that have lost expression of MHC I. NK cells induce apoptosis in cancer cells via release of perforin and granzymes from granules, like CD8+ T cells. But here too, NK cells can be broken down by the tumour cells, notably by the PD1/PDL1 interaction.
Increasing their efficacy as a therapeutic strategy has made the development of new therapeutics enhancing their anti-tumour response a priority. Those new therapeutics can have several shapes. The Antibody-dependent cell-mediated cytotoxicity (ADCC) activity of the IgG1 isotypes is one of the mechanisms that can be applied by immunoglobulin-based therapeutics. Another way to increase the killing of cancer cells is to target tumour antigens expressed by those cells using for example bispecific molecules that will enhance on the other side the T cell activation.
Using primary immune cells, in vitro bioassays were developed to better screen the potential effect of new therapeutics on immune cell killing activity. Their ability to increase or induce a cytotoxic activity and facilitate the anti-tumour immune response can as a result be assessed early in the drug development process.
1:00 – 1:30pm
Use of various genomics techniques, including NGS, to identify and characterize biomarkers relies on an efficient extraction of nucleic acid from biological samples. Poor or inefficient extractions from rare or limited samples, such as tumor biopsies or small cell populations, can prohibit the number or types of assays that can be performed on each sample, and therefore limit the information obtained from each sample. Most conventional technologies for the extraction and purification of nucleic acid for genomics analyses rely on the principle of solid phase extraction, an inefficient multistep workflow that carries a risk of nucleic acid loss or damage at several steps.
The Ionic® Purification System enables a simple, automated workflow to extract and purify nucleic acids with dramatically increased yields and quality from a wide range of sample types, including formalin-fixed paraffin-embedded (FFPE) tissues and low numbers of cultured or sorted cells. Nucleic acids are isolated in their natural, native form using the company’s core isotachophoresis technology. Since isolation occurs without binding and stripping nucleic acid from a fixed surface, nucleic acid loss and fragmentation are minimized, and purification induced bias is eliminated.
1:40 – 2:40pm
- How do we assess the response to an IO therapy within a patient? What is the best timing? What tissues are the best to assess?
- How do we understand how an IO drug works and which patients would benefit most form that drug?
- How can we show that an IO drug differentiate from SOC and other IO drugs to ensure maximal impact for patients?
- How can we test and confirm IO drug mechanisms of action before going into the clinic when the models are challenging?
Head, Immuno-Oncology Biology & Translation
Takeda Oncology
Katherine Seidl is currently Head of Immuno-Oncology Biology and Translation at Takeda Oncology. Prior to this she was the VP of Immunology, Research and Development, at SQZ Biotech. Katherine earned a Ph.D. in Genetics from Stanford University and completed her postdoctoral training at Harvard Medical School. A 15+ year career in drug discovery and development followed including Director of Immunotherapy at bluebird bio, Cambridge, MA. Katherine has worked at Novartis Institute of Biomedical Research in Switzerland and Cambridge, MA as a Group Leader in the departments of Oncology and Autoimmune, Transplantation and Inflammation. Previous experience in pre-clinical and translational research include Diamed, Wyeth, and Pfizer.
Preclinical Research
Biomarker discovery – need for speed, automation and throughput in screenings for gene and protein biomarkers?
- How do you best screen and study the interplay between tumor and immune response?
- Importance of high throughput assays for biomarker discovery, especially in pre-clinical
- What aspects of primary and secondary screening assays are important?
Global Product Manager
Thermo Fisher Scientific
Stefan Jellbauer is a Global Product Manager at Thermo Fisher Scientific supporting the QuantiGene gene expression platform and Luminex instruments. After 10 years in academic research, Stefan joined Affymetrix/eBioscience, where he worked in the roles of Field Application Scientist and Technical Specialist with Affymetrix and Thermo Fisher Scientific. He continued his work in cancer diagnostic research with Farcast Biosciences as Technical liaison supporting BioPharma business development for ex-vivo human tumor platform. Stefan re-joined the QuantiGene team at Thermo Fisher Scientific as a Product Manager.
Stefan received his Ph.D. in Biology (Medical Microbiology/Immunology) from Ludwig Maximilian’s University of Munich (Germany), focusing on tumor vaccination. He completed his post-doctoral work at the University of California, Irvine studying mucosal immunology and host-pathogen interactions.
Defining optimal in vivo models to identify clinically relevant immunotherapies and investigate their efficacy in combination with IO and non-IO agents
- Can syngeneic mouse models provide data on effectiveness of IO combination approaches that is translatable into clinical success or are more advanced models a must have?
- How do we best model checkpoint inhibitor resistance and the immune-excluded tumor microenvironment in mice?
- What other approaches and systems can we leverage to complement in vivo efficacy data to maximize translatability of mouse data to human settings?
- Optimizing timing and sequencing of combination approaches: what are the limitations of mouse models and how do we overcome them?
Head of Pharmacology
Jounce Therapeutics
Monica joined Jounce Therapeutics in 2017, bringing over 15 years of experience in developing and interrogating complex mouse models of hematological and solid malignancies. She has a deep understanding of cell biology and tumor immunology, which she leverages to advance clinically translatable approaches to treat cancer. At Jounce, Monica leads a multidisciplinary team, whose expertise spans several functional areas, from in vivo pharmacology, to 3D model development to histopathology.
Monica obtained her PhD in Molecular Genetics from the International School for Advanced Studies in Trieste, Italy, and completed post-doctoral studies at Boston Children’s Hospital/Harvard Medical School. She co-authored over 40 peer-reviewed publications, and was the recipient of several awards, including a Leukemia and Lymphoma Society Special Fellow Award and the V Foundation Scholar Award, and two NIH/SBIR research grants.
9:10 – 10:10pm
- Overcoming safety challenges with xCD3 bispecifics and CAR-T
- Cytokine Release/CRS and strategies to overcome these
- On target/off tumor toxicity
- Increasing efficacy in solid tumors
- Combination treatments including checkpoint inhibitors, Costimulatory combinations, etc
- The challenges of modeling these pre-clinically
Associate Director, Immuno-Oncology
Regeneron
Alison Crawford is an Associate Direct in immuno-oncology at Regeneron Pharmaceuticals, Inc. She has >18 years of immunology research experience, including > 9 years in the pharmaceutical industry. At Regeneron Pharmaceuticals, her team is responsible for IND-enabling studies as well as mechanistic studies using bispecific antibodies for oncology indications. She led the in vivo pre-clinical research efforts on REGN4018 (MUC16xCD3) and REGN5668 (MUC16xCD28) to advance the antibodies through to IND submission. Alison completed her BSc in Immunology from Glasgow University before being admitted to the Wellcome Trust Ph.D. program at Edinburgh University where she focused on T cell memory. Her post-doctoral work at the University of Pennsylvania examined T cell exhaustion during chronic viral infection and the use of checkpoint blockade to alleviate this exhaustion.
1:00 – 1:30pm
The Euretos AI Platform leverages AI technologies, such as machine reading and learning, to create data driven disease insights into systems biological, molecular mechanisms that drive phenotypes, disease pathology, toxicity and drug response. In this session we will explain how some of the world’s leading biopharma companies use the Euretos platform in developing therapies based on checkpoint inhibitors, monoclonal & bispecific antibodies and antibody drug conjugates.
1:40 – 2:40pm
- Is the therapeutic and diagnostic potential of EVs in Immuno-oncology sufficient to warrant the investment needed to overcome technical and biological challenges?
- Do EVs show greater potential as biomarkers and IO therapeutic targets than their use in drug delivery and EV cargo applications?
- Is there a need at this stage for harmonisation of standard operating procedures in EV research?
Chief Medical Officer
Synexa Life Sciences
Justin Devine co-founded Synexa Life Sciences in 2003 and is the current serving Chief Medical Officer of Synexa Life Sciences. Justin is a physician and has further degree qualifications in both immunology and pharmacology.
His primary focus is to understand and meet both the challenges and objectives in the drug development industry by designing biomarker strategies to bring real insight to the challenges of clinical development.
He is particularly passionate about improving the drug development process by bringing innovative approaches to early phase research, including new ideas in translational medicine, bioinformatics and artificial intelligence.
Clinical Development
- When taking on a multi-omic approach (e.i. a combo of NGS, mIHC, RNAish, WES, etc.) to understanding the TME within a study, what are the key considerations and risks associated with such initiatives?
- What are the current challenges faced when integrating and interpretating multi-omic data sets in translational/clinical IO research studies?
- What strategies, processes, and/or tools are currently being utilized to help facilitate the success and efficiency of such research studies?
Associate Director, Biomarker & Biopharma Programs
HalioDx
Katir Patel is a classically trained immunology and immuno-oncology specialist with a professional focus on multi-omic approaches to biomarker strategies and diagnostic development. Experience supporting translational research to late-stage clinical trials involving immunotherapies and cancer diagnostics. Current focus includes biomarker consultation and account management of large-scale biopharma projects, strategic partnerships with technology platforms and KOL networks, and market development activities that drive adoption of biomarker technologies that drive successful drug development.
Katir has led biomarker programs at both large scale and startup companies focused on immuno oncology research for over 6 years.
He holds a Ph.D. in immunology and infectious diseases from the University of Massachusetts, Amherst.
- What is the current “standard” with respect to endpoints in our clinical IO trials, and should it be challenged?
- How can we leverage current immune outputs to better define efficacy potential, and bring novel agents to patients sooner?
- What newer technologies can strengthen our confidence in immune markers of potential clinical efficacy?
- What regulatory burdens can we anticipate in considering these potential novel endpoints?
Vice President, Clinical Development
Inovio Pharmaceuticals
Jeffrey Skolnik directs several of Inovio’s clinical-stage cancer immunotherapy programs, overseeing DNA immunotherapies aimed to treat cancers. He was previously at Tetralogic Pharmaceuticals, as Vice President of Clinical Research and later Chief Medical Officer, overseeing all clinical programs in oncology, infectious diseases and dermatology. He was also a Medical Affairs lead in oncology at GSK and held several positions at AstraZeneca including Medical Science Director for early and late stage compounds. Jeffrey received his pediatric training at Children’s Hospital, Boston and completed his hematology/oncology training at the Children’s Hospital of Philadelphia, where he is as Adjunct Assistant Professor of Pediatrics.
9:10 – 10:10am
- What is really a predictive biomarker? When do you really need to think
about one for your clinical development program? - Genomic profiling is becoming more accessible to cancer patients,
including next-generation sequencing (NGS). Does your program
require a companion diagnostic? - What is the challenge of having a Master Protocol? → Basket vs.
Umbrella Trial Designs - Pembrolizumab received the first agnostic indication in oncology →
MSI-H or dMMR. There are now other tissue-agnostic approvals. If your
program gets accelerated approval, how should you plan the
confirmatory study? - What makes a tissue-agnostic approach so appealing in oncology?
Operationally, how you can achieve best approach?
Vice President, Clinical Development
TG Therapeutics
Alejandro Ricart M.D. is Vice President, Clinical Development at TG Therapeutics. Prior to this role, Alex was Executive Medical Director at Oncology Early Development Strategy & Innovation at Novartis Pharmaceuticals Corporation. Alex was also Senior Director Late Development for Merck Oncology. He played a key role in Pembrolizumab Breakthrough Therapy Designation and in the sBLA for Hodgkin lymphoma. Alex was also a Director of Biotechnology and Oncology at Pfizer in San Diego, California. He participated in the clinical development of several investigational compounds, including Axitinib and Bosutinib. He has published more than 80 journal articles, abstracts, reviews, and book chapters.
1:00 – 1:30pm
Lung cancer is an extreme case for drug development:
a) some targeted therapies have gained accelerated approval based on objective response rate (ORR) in single arm Phase II trials but
b) at the same time the number of unknowns for trial design (and the associated decisions/risk) such as subgroups, biomarkers, drug combination, etc. are substantial.
Model informed Drug Development (MiDD) – especially disease-centric and mechanistic in silico trials – are promising tools to alleviate the complexity burden by exploring how the trial variables influence the outcome and by building mechanistic understanding of this impact.
In this context, we built an in silico lung cancer model using NOVA’s Jinkō® platform based on mechanistic knowledge and data to serve as a virtual control arm that can be used to better inform Phase II design decisions.
In our use case of in silico clinical trials we highlight tumor growth inhibition by tyrosine kinase inhibitor (EGFR-TKIs) in patients harboring a specific EGFR mutation. The predictions by such a mechanistic approach are compared with tumor size measurements (TNM) and time to progression reference data not not used to inform the model. Tumoral heterogeneity, age, gender, initial clinical stage, smoking status and ethnicity can be taken into account and drive variability as covariates of treatment efficacy in the in silico trial through mechanistic hypotheses.
The platform allows for leveraging survival data serving for translation between endpoints and can therefore improve sample size estimations of pivotal/confirmatory trials in the data poor-settings of accelerated approval.
The presentation will lay down the architecture of this model and present its functioning from molecular level to patient population as well as future directions for biomarker exploration.
1:40 – 2:40pm
Overcoming IO trial challenges in the era of a pandemic
- How did COVID-19 impact IO clinical studies and what changes may be adopted in future studies?
- What impact did the pandemic have on clinical development strategies and how have regulators responded?
- Can we leverage the experience gained from the rapid development of COVID-19 therapeutics/vaccine and apply to IO studies?
Executive Director, Immuno-Oncology
Partner Therapeutics
Fiona has over 15 years of oncology drug development experience leading clinical development and translational science teams. At PTx, she is responsible for the clinical development of sargramostim in COVID-19 and oncology indications. Previously, she lead the clinical development of the BET inhibitor program at Constellation Pharmaceuticals, the elacestrant program at Radius Health, the isatuximab and clolar programs at Sanofi and was clinical operations lead on the erismodegib program at Novartis. Fiona obtained her BSc in Immunology from University of Edinburgh and PhD in cancer biology from the University of Newcastle.
Cell Therapies
- What issues have you faced in stratifying patient populations?
- How have you overcome specificity hurdles with ELISAs and other assays?
- What challenges have you encountered when moving toward decentralized testing modalities?
- How are you performing sample collection during the pandemic?
Director, International Business Development,
Rapid Novor
Anthony has been heading the business development at Rapid Novor for nearly 4 years, merging his previous 8 years of experience in the business roles with a strong passion for biomedical research, stemming from his training in biomedical engineering. His current efforts are focused on the growth of the REpAb™and NovorIg™ platforms with pharmaceutical and biotech companies globally. His partnerships are focused on using REpAb in patients or animal models to sequence monoclonal antibodies from polyclonal sera, as well as immune system profiling using NovorIg, the team’s NGS based analysis to monitor the relative quantity of specific antibodies over time. Anthony’s a maker and in his spare time takes advantage of the Toronto HackLab to do biological research and other engineering projects.
- Solid tumor heterogeneity: how to choose the best target?
- The hostile tumor microenvironment doesn’t want your immune cell to come in, unless…
- T cell, NK cell, macrophage… is it a team sport or a sniper attack?
- Combinations with checkpoint inhibitors: a must?
Chief Medical Officer
Carisma Therapeutics
Debora Barton joined Carisma Therapeutics in November 2019. She brings 18 years of oncology experience, both in academia as a practicing physician and in the biotechnology/pharmaceutical industry.
Most recently, she served in key senior executive positions in cellular therapy and radiopharmaceutical biotech companies including Iovance Biotherapeutics and Advanced Accelerator Applications, acquired by Novartis during Debora’s tenure. In her leadership roles, Debora built Clinical Development and Drug Safety teams setting up infrastructure for the conduct of clinical studies and was instrumental in obtaining FDA Breakthrough Designation for a cell therapy product and in the approval of a radiopharmaceutical agent by both FDA and EMA.
Previously, Debora spent 10 years at Celgene and Novartis, always focusing on improving the lives of cancer patients. She holds an MD from Pontificia Universidade Catolica Sao Paulo (PUC-SP) and completed her fellowship in Oncology at Federal University of Sao Paulo (UNIFESP) in Brazil.
8:30-9:00am
9:10 – 10:10am
- Tumor heterogeneity and antigen escape mechanisms of tumor resistance to CAR treatment?
- Multi-targeted CAR therapeutics to address tumor resistance, the challenge and the opportunity?
- The optimal format of Multi-targeted CAR? Tandem, multicistronic, or else?
- The emerging armor molecules to facilitate CAR activation against solid tumor?
- Combination therapy to tackle TME?
Chief Scientific Officer
Elpis Biopharmaceuticals
Kehao Zhao, is the Co-founder and Chief Scientific Officer of Elpis Biopharmaceuticals. He and his company have developed proprietary mRNA Display technologies for antibody discovery and protein engineering. Through those technologies, he is leading a team to develop antibody and cellular immunotherapeutics that target and activate the immune response to eradicate tumors. Kehao was a senior investigator at Novartis Institutes for Biomedical Research leading antibody therapeutics platform and cancer epigenetics research. Previously, Kehao was a scientist at Procter & Gamble Pharmaceuticals. Kehao completed postdoctoral training in Gene Expression & Regulation at the Wistar institute and received his PhD from the Institute of Biophysics at the Chinese Academy of Sciences. Kehao has published over 30 scientific research papers.
1:00 – 1:30pm
As a full-service CRO with a specialized focus on Advanced Therapies including cell and gene therapies, Medpace provides comprehensive solutions to the unique and rapidly evolving clinical, operational and regulatory challenges of cellular, tissue and gene therapy medicines. This presentation will focus on operational considerations for cell and gene therapy studies with a focus on cell tracking.
Experts
- A specialized physician team with clinical experience in cellular, tissue and advanced therapy, further supported by our core group of Medpace sub-specialty physicians
- Global and strategic regulatory leadership from a team with hands-on and in-depth experience in the specific requirements of advanced therapies
- Dedicated clinical operations team with extensive experience in advanced therapy and rare diseases
Experience
- Medpace is a leader in the advanced therapy space with experience in many therapeutic areas
- Our areas of expertise include hematopoietic cell transplantation gene therapy, gene editing, graft versus host disease, cellular therapy, tissue therapy, adoptive immunotherapy, and tissue engineered products
Execution
- Processes, including cell-tracking, have been implemented to streamline trial oversight
- Wholly-owned Imaging Core Lab and Central Laboratories provide cohesive, integrated, and standardized trial management
1:40 – 2:40pm
- Access of IPSCs and characterization of these lines for commercial use
- Challenges in differentiation of IPSC’s to NK and T cell phenotypes
- Do you have to start with IPSC’s to get to CARs?
- Are there other cell types besides NK and T cells that are being explored for cell therapy?
Vice President, Oncology Innovation
Janssen Pharmaceuticals
Marco Gottardis has 25 years of experience as a cancer research investigator in both biotech/large pharma and over 29 years of experience overall in translational cancer research. He is currently VP of Oncology and Innovation Leader at Janssen Pharmaceuticals and is exploring new uses of Machine Learning and AI platforms for use in drug discovery and competitive technology landscape analyses. In earlier roles at Janssen, Marco was Prostate DAS Leader and under his leadership, developed a significant R&D portfolio for Janssen.
Prior to Janssen, he was Executive Director of Oncology and Immuno-Oncology Research and co-leader of the Oncology Discovery Group at BMS and Senior Investigator at LIGAND Pharmaceuticals
Marco obtained his BS from Columbia College, Columbia University, his Ph.D. at the Wisconsin Clinical Cancer Center, University of Wisconsin. He completed his post-doctoral fellowship at Vincent T Lombardi Cancer Center Georgetown University. Marco has co-authored over 100 publications and is co-inventor on multiple patents. He is externally recognized as an industry and scientific leader in prostate cancer research and oncology.
Next-Generation Therapies
- From checkpoint inhibitors towards neoepitope vaccines, does one size fits it all?
- Which functional assays; Types and Stages
- From early characterization towards clinical biomarkers
- Strengths & Limitations?
- Challenges?
Founder & Chief Technology Officer
ImmunXperts
Sofie Pattijn has over 20 years of experience in the field of immunogenicity assessment (vaccines and biotherapeutics) and in vitro assay development with a focus on functional assays for immunogenicity, immune oncology and Cell and Gene Therapy products. She has extensive hands-on lab experience and has managed and coached several In Vitro teams over the last decade. From 2008 till 2013 she was Head of the In Vitro Immunogenicity group at AlgoNomics (Ghent, Belgium) and Lonza Applied Protein Services (Cambridge, UK). Prior to that, she worked at Innogenetics, Belgium for over 15 years.
- How do different Type I IFN stimulators differ in how they elicit an immune response?
- How does dosing regimen and delivery strategy (local vs systemic) of Type IFN stimulators impact activity?
- How will the type and/or strength of stimulus impact combination strategies in the future?
Director, Immuno-Oncology Biology
Takeda Oncology
Adnan Abu-Yousif leads the Biology team within the Cold-to-Hot Pillar of IO-Drug Discovery Unit at Takeda Oncology. He has served as the discovery lead for Takeda’s STING assets, TAK-164 and others and is a member of a number of Joint Steering Committees to advance Takeda’s discovery portfolio. Prior to joining Takeda, he contributed to the discovery and translational support of Merrimack Pharmaceuticals pipeline. Adnan has a Ph.D. in Pharmacology, Toxicology and Experimental Therapeutics from the University of Kansas.
9:10 – 10:10am
Understanding the tumor microenvironment to improve therapeutic strategies and outcomes
- Why does the tumor microenvironment matter?
- When thinking of TME analysis, what are the most appropriate considerations, and then what are the best techniques?
- What are some of the challenges associated with the TME, aow do we best manage the transient nature of cellular phenotypes to gain a better grasp of TME dynamics?
- When thinking of therapeutic delivery in to the TME, what must we contend with to ensure proper targeting?
Senior Scientist, Translational Pathology
Jounce Therapeutics
Edward has extensive oncology research experience in multiple cancers across academia and industry. He worked at the Dana-Farber Cancer Institute and at Brigham and Women’s and Harvard Medical School before moving to PerkinElmer where he gained deeper insights into cancer and the tumor microenvironment. This led to Edward’s nomination to serve as member on the Society for Immunotherapy in Cancer (SITC) Biomarker Task Force, which he still does. He is currently a Principal Scientist in the Translational Pathology group at Jounce Therapeutics, helping to characterize tissue metrics in preclinical models, as well as in clinical samples derived from our clinical trials to advance development of novel cancer immunotherapies.
Edward completed his undergraduate education at Stony Brook University in New York and has a Ph.D. from Boston College.
1:00 – 1:30pm
The evolving research field of immuno-oncology focuses on an individual’s immune system as innovative treatment approaches to combat cancer. The goal of cancer immunotherapies is to initiate a self-sustaining cycle of cancer immunity, overcome tumor evasion mechanisms and promote conditions that favor immune protection. Immune checkpoint molecules have been identified as critical players in the regulation of NK cell- and T cell-mediated immune responses. Besides intact transmembrane proteins, soluble isoforms and variants of checkpoint molecules function as immune adjuvants or decoy receptors and may also influence clinical efficacy of checkpoint modulator drugs. The systematic analysis of these soluble biomarkers should help to shed light on the regulation of checkpoint pathways and facilitate research for future immunotherapeutic treatments. This presentation will provide an understanding of immunoassay technologies that enable the simultaneous detection of multiple soluble immune stimulatory and inhibitory factors, and a more comprehensive picture of cancer immunity in a blood sample.
1:40 – 2:40pm
- What are the most accessible and useful types of AI?
- How to leverage AI without falling down the rabbit hole?
- Where is AI most impactful in digital pathology: image analysis or data analysis?
- AI pitfalls 101: what are the risks of relying too heavily on AI predictive models (risks of subsampling, overfitting, class imbalance, and small sample size)?
- What types of challenges is AI not suitable for?
Associate Principal Scientist
AstraZeneca
Michael Surace is an Associate Principal Scientist at AstraZeneca in Gaithersburg, MD where he designs, develops, validates, and deploys multiplex immunofluorescence assays for clinical and research applications in translational oncology. He was trained in innate immunity and molecular biology as a graduate student at Virginia Tech where he studied cell signaling mechanisms controlling macrophage polarization and in neuroscience and biochemistry in postdocs at Virginia Commonwealth University where he studied microglia and astrocyte immune and inflammatory function in neurodegenerative and autoimmune CNS disorders. He moved into biotech and immune oncology which led him to pursue technologies to more intricately describe spatial and phenotypic features of the TME, generating datasets which require AI to fully leverage for patient benefit. He is an AI-facing bench scientist navigating the fast-growing field of technologies, techniques, commercial products, and services seeking to deliver on the promise of AI.