Genomics in Precision Oncology Xchange
West Coast
March 30, 2022
Welcome to hubXchange’s West Coast Hybrid Genomics in Precision Oncology 2022, bringing together executives from pharma and biotech to address and find solutions to the key issues faced in genomics-led oncology precision medicine strategies.
Discussion topics will cover Cancer Genomics, Translational Bioinformatics, Data Quality & Access and Genomics Informed Clinical Decisions.
Take advantage of this unique highly interactive meeting format designed for maximum engagement, collaboration and networking with your peers.
Please note this is a HYBRID meeting. Participants can join in-person and virtually. All COVID safety protocols will be adhered to.
Venue Details – DoubleTree by Hilton San Francisco Airport Hotel, 835 Airport Boulevard, Burlingame CA 94010-9949
Cancer Genomics
Opening Address & Keynote: Connecting Biomarker Development with Disease Detection and Monitoring
- Better characterizing disease biology in order to more fully understand tumor heterogeneity
- Enabling more sophisticated diagnostic tests to address therapy-induced tumor evolution
- Improving outcomes with ongoing disease molecular assessment and treatment management based upon molecular endpoints
President & CEO, Stilla Technologies
Philippe is an executive life sciences leader with over 25 years’ experience gained through leadership roles at global organizations serving drug discovery and development organizations and with his strong biomarker landscape knowledge has a proven track record of influencing both translational and clinical programs.
Connecting cancer genomics with environmental stress adaptation: Considerations for drug discovery
- Tumor targeted therapies in genetically defined patient populations have revolutionized cancer treatment paradigms. However, innate or acquired therapy resistance cannot be completely explained by genetic hypotheses.
- It is increasingly recognized that environmental pressures promote tumor dependencies that may or may not be genetically driven.
- This discussion will focus on our emerging understanding of tumor genetic lesions and their cross-talk with environmental stressors.
- Can these learnings inform on the next generation of cancer drugs or on novel therapeutic combinations?
Senior Director, Head of Translational Sciences, Pioneering Medicines
Charles is a Senior Director and Head of Translational Sciences at Pioneering Medicines. Prior to this, he was Senior Principal Scientist at Bristol-Myers Squibb, leading drug discovery programs targeting the interface between tumor-intrinsic biology and the tumor microenvironment. Charles originally trained as an immunologist, completing his PhD in immune cell biology at the MRC National Institute for Medical Research (London, UK) and postdoctoral training in inflammation at Emory University. He previously led a small molecule immuno-oncology group at AstraZeneca (Cambridge, UK), where his team discovered a clinical candidate antisense oligonucleotide targeting FOXP3. Charles also has a longstanding interest to understand the immunomodulatory functions of the PI3K/MTOR signaling pathway in cancer and inflammation.
Beyond genomics for oncology biomarkers: opportunities and challenges of multi-omics approaches
- How multiple mutations lead to the same phenotypes and how can we account for them?
- Can gene expression uncover cellular states related to drug sensitivity?
- What are the challenges to get non-genomic based tests in the clinic?
- Can other omics such as methylation and histone marks be used for translational research?
Senior Bioinformatics Scientist, Genentech
As a computational biologist in oncology research at Genentech, Marc Hafner works towards characterizing new therapeutic targets, supporting the development of new drugs, and identifying the patient populations that would most benefit from these drugs. For his personal research, Marc is leveraging pre-clinical and clinical data including real-world data to understand drug mechanisms of resistance. Marc is also leading the implementation of a drug response software suite that leverage the GR method he developed during his postdoc.
3:45 – 4:45pm
New genomics technologies for cancer target identification and characterization – opportunities and challenges
- Which new technologies are on the horizon for target identification and characterization?
- Are novel technologies in the spatial transcriptomics (Stereo-seq, Slide-seq) and CRISPR single-cell sequencing (TAP-seq) field advanced enough to provide a benefit for target identification and characterization?
- How do the cancer genomics field and target identification benefit from spatial transcriptomics approaches (or other novel technologies)?
- Evaluating gene perturbation effects on single-cell level – challenges and benefits
Senior Scientist, Bioinformatics, Merck
Milena Hornburg is a Senior Scientist in the Genome and Biomarker Science Department at Merck. As part of the oncology computational biology group, her work focuses on novel target identification and characterization working with large NGS datasets spanning from traditional RNA-seq to novel spatial transcriptomics and single-cell CRISPR-seq technology data. Prior to Merck, Milena completed her postdoc at Genentech in the Bioinformatics and Computational Biology department where she collaborated with Oncology Biomarker Development on the investigation of the tumor microenvironment and its’ association with tumor immune infiltration (Hornburg et al. Cancer Cell 2021, Desbois et al. Nature Medicine 2020).
Translational Bioinformatics
Opening Address & Keynote: Connecting Biomarker Development with Disease Detection and Monitoring
- Better characterizing disease biology in order to more fully understand tumor heterogeneity
- Enabling more sophisticated diagnostic tests to address therapy-induced tumor evolution
- Improving outcomes with ongoing disease molecular assessment and treatment management based upon molecular endpoints
President & CEO, Stilla Technologies
Philippe is an executive life sciences leader with over 25 years’ experience gained through leadership roles at global organizations serving drug discovery and development organizations and with his strong biomarker landscape knowledge has a proven track record of influencing both translational and clinical programs.
Discover tumor evolution, understand resistance, inform therapy
- What are the major mechanisms of resistance to targeted therapies in leukemia/ lymphoma and in the solid tumor setting (target escape, mutation, hostile immune microenvironment etc)?
- What new therapies are on the horizon to overcome these challenges?
- How can tumor biopsies be better leveraged to develop robust predictive biomarkers and how can these strategies be incorporated into clinical trial design to accelerate development of novel oncology drug products?
- If there is strong evidence of a predictive tumor biomarker identified during non-clinical evaluation, should better attempts be made in FIH trials to enrich for these patients?
- What are the best technologies currently available to monitor tumor evolution and enable rapid identification of emerging resistance mechanisms? How can this information be used to inform patients around subsequent therapy and/or enrollment onto other clinical trials?
Senior Vice President, Head of Translational Medicine, CERo Therapeutics
John Rossi leads the Translational Medicine team at CERo. Prior to joining, he served as Senior Director of Translational Medicine and Head of Cell Therapy Clinical Pharmacology at Kite, a Gilead Company. While at Kite, he led translational activities supporting the clinical development and global approvals of YESCARTA® and TECARTUS® CAR T cell therapies. At Kite, his team also supported work focusing on the mechanistic understanding of engineered cell therapy products under strategic collaborations with the NCI and numerous leading academic institutions. Prior to Kite, he spent 13 years in oncology drug development at Amgen working with teams to identify novel biomarkers for hematologic and solid tumor drug development programs.
- Multimodal data is a powerful asset in healthcare
- Complementary information across different modalities can lead to more accurate disease diagnoses and outcomes predictions
- Emerging RWD capabilities are uncovering valuable new insights to support drug discovery efforts, clinical development, and commercialization
Senior Director, Data Science, Tempus
Kyle Beauchamp is Senior Director of Data Science at Tempus, where his team uses interdisciplinary methods of data science, machine learning, and computational biology to develop AI-powered clinical tests and RWD-powered advances in oncology research. Before coming to Tempus, he worked at Counsyl/Myriad after receiving his PhD from Stanford and completing post-doctoral work at Memorial Sloan Kettering Cancer Center.
Networking Lunch
Advancements in single-cell multi-omics approaches towards biomarker discovery
- What are the key challenges in preclinical and translational bioinformatics.
- What are the advancements in technologies that can help us interrogate tumor heterogeneity at a single-cell level.
- How can we characterize gene edits using NGS and single-cell technologies to create safer cell therapies.
- What are the challenges in mining these heterogeneous data types.
Associate Director, Bioinformatics, Nkarta Therapeutics
Sombeet is the head of computational biology at Nkarta. Nkarta is a clinical-stage biotechnology company advancing the development of allogeneic natural killer (NK) cell therapies for cancer. Sombeet has extensive experience leading multiple informatics software and computational biology products to market. He is a strategic leader, an inventor, and developer of many industry-first tools and analysis methods. Previously he was a Bioinformatics lead at Mission Bio and ThermoFisher Scientific.
3:45 – 4:45pm
Multi-omics-led oncology biomarkers discovery
- How can we improve our rate of success in translating biomarkers discovered from in-vitro or in-vivo datasets to the clinic?
- Can omics data aggregated across many indications be effectively used for biomarker discovery?
- Can systems biology knowledgebases be effectively integrated into machine learning models for drug response prediction?
- What challenges exist in identifying targets/biomarkers linked to positive selection in functional genomics datasets?
- What opportunities exist to improve genomic annotation of cellular models?
Associate Director, Bioinformatics, IDEAYA Biosciences
Stephen Federowicz received his B.S and PhD in Bioinformatics at the University of California Santa Cruz and University of California San Diego respectively. His research interests have focused on systems biology, functional genomic screens, NGS technologies, and machine learning. He is currently the head of Bioinformatics at IDEAYA Biosciences where he oversees a CRISPR screening focused target ID platform, omics driven MOA studies, and machine learning efforts to identify novel biomarkers and optimal patient selection strategies. Prior to IDEAYA Stephen held roles with increasing levels of responsibility at Intrexon, Roche, and Inscripta.
Data Quality & Access
Opening Address & Keynote: Connecting Biomarker Development with Disease Detection and Monitoring
- Better characterizing disease biology in order to more fully understand tumor heterogeneity
- Enabling more sophisticated diagnostic tests to address therapy-induced tumor evolution
- Improving outcomes with ongoing disease molecular assessment and treatment management based upon molecular endpoints
President & CEO, Stilla Technologies
Philippe is an executive life sciences leader with over 25 years’ experience gained through leadership roles at global organizations serving drug discovery and development organizations and with his strong biomarker landscape knowledge has a proven track record of influencing both translational and clinical programs.
9:05 – 10:05am
- How can we break down data silos within our organizations and make data more findable and accessible?
- Besides partnerships and consortia arrangements, are there other ways to promote inter-organizational data sharing?
- What infrastructure and methodologies can we employ to use, integrate, and/or interpret heterogeneous data types and datasets to uncover biological insights and patient stratification?
- What can be done to facilitate interoperability of databases even within the same organization or dept?
- To better enable precision oncology, should we and can we democratize the use of big data, instead of leaving it to primarily people with data science expertise?
Senior Scientist, Bioinformatics, Genentech
Jieming Chen is an experienced computational biologist, with a proven track record in bioinformatics, population genetics, genomics and protein science. He has expertise in leveraging machine learning techniques, statistical modeling and big data analytics in molecular data analyses – DNA sequencing (microarray, whole exome and whole genome), RNA-seq, ChIP-seq and 3D protein structure analyses. Jieming is experienced working with clinical data, especially clinical trials, and electronic health records. Currently he is focused on cancer immunology, including cancer immunotherapy, neoantigen prediction, and the development of personalized cancer vaccines.
Exploring how NGS is moving the field towards comprehensive characterization
- Advances in NGS technologies to revolutionize personalized cancer vaccines, precision medicine and biomarker discovery.
- Existing challenges that must be addressed for both tissue profiling and liquid biopsy approaches.
- Various prevailing issues and strategies facing the field
Manager, Field Application Scientist, Personalis
Kedar Hastak is a Manager, Field Applications Scientist with more than ten years of basic and translational experience in cancer therapeutics. At Personalis, Kedar provides scientific technical support and customer education on the NeXT Cancer Product Portfolio. Prior to joining Personalis, Kedar was an Instructor/ Senior Scientist at Stanford University’s Division of Oncology, where he led translational research studies to evaluate the efficacy of PARP inhibitors in treating breast and other solid tumor cancer models. Kedar completed his postdoctoral studies at the Cleveland Clinic Foundation and Stanford University, and received his PhD in field of Cancer Biology from Case Western Reserve University.
Overcoming data reproducibility challenges: Definitions, deliverables, processes, and culture
- How do we define reproducibility?
- What are tangible assets we associate with reproducibility?
- How can we initiate, nurture, and maintain best practices in reproducibility?
- Can we create processes and culture that makes reproducibility an integral part of our work?
Senior Computational Immunologist, Pfizer
As a computational immunologist at Pfizer, Robert Amezquita leverages recent advances in single-cell genomics in tandem with large databases of real-world evidence data and clinical compendiums to promote the advancement of promising assets through Pfizer’s preclinical and clinical development pipeline. Applying systems engineering approaches in building out operationalized machine learning platforms (MLOps), Robert enhance his team’s capabilities by building a framework for discoverability, accessibility, reproducibility, and traceability (DART) to drive timely and high-value insights.
Past work spans novel ML based microsatellite instability caller from liquid biopsies at a cancer diagnostics startup, postdoctoral fellowship research of CAR-T cell activity and development at the Immunotherapy Integrated Research Center at Fred Hutch, and the study of CD8 T cell differentiation throughout Robert’s PhD in Yale’s Immunobiology program.
Genomics Informed Clinical Decisions
Opening Address & Keynote: Connecting Biomarker Development with Disease Detection and Monitoring
- Better characterizing disease biology in order to more fully understand tumor heterogeneity
- Enabling more sophisticated diagnostic tests to address therapy-induced tumor evolution
- Improving outcomes with ongoing disease molecular assessment and treatment management based upon molecular endpoints
President & CEO, Stilla Technologies
Philippe is an executive life sciences leader with over 25 years’ experience gained through leadership roles at global organizations serving drug discovery and development organizations and with his strong biomarker landscape knowledge has a proven track record of influencing both translational and clinical programs.
Effective patient stratification based on molecular and clinical data
- Are genomics approaches best used as research tools for identifying responsive pathological populations, or should they be developed into companion diagnostics?
- Do currently-available genomics-based tests offer enough decision-changing information at reasonable price points to justify their wider incorporation in clinical practice?
- Which innovations in genomics do you believe will contribute most to patient stratification in the future?
- What strategies can we employ to move more genomics-scale biomarkers from R&D to clinical applications?
Director, Bioinformatics, Zai Labs (US)
Christopher Szeto leads the Bioinformatics team at Zai Lab (US) guiding precision medicine strategies for multiple clinical-stage oncology products. Prior to his current translational medicine role, Christopher worked at NantOmics/ImmunityBio on molecular diagnostics products in oncology. While there he lead teams of Bioinformaticians and machine-learning experts to derive predictive biomarkers from multi-omics datasets and pathology images. Christopher is a Fulbright scholar from New Zealand who moved to the US in 2006 to study towards a Ph.D. in the Haussler Lab at UCSC, where he was part of data coordination for the TCGA project and developing predictive algorithms in cancer datasets.
Better informing treatment options
- How to enable oncologists to get faster decisions using molecular testing
- How do we better support the practical clinical development of biomarkers
- Identifying and measuring personalized mutations in the patient monitoring setting
President & CEO, Stilla Technologies
Philippe is an executive life sciences leader with over 25 years’ experience gained through leadership roles at global organizations serving drug discovery and development organizations and with his strong biomarker landscape knowledge has a proven track record of influencing both translational and clinical programs.
Networking Lunch
3:45 – 4:45pm
- Thorough understanding of key mechanisms that drive response – association vs causal/driver
- Identification of biomarkers that are reflective of an immunological response and potency
- Distilling information from new technologies like spatial omics, TCR repertoire and scRNAseq for predicting response and further development into effective biomarker strategies
Director, Late-Stage Research, InstilBio
Akshata Udyavar is heading the Late Stage Research team at InstilBio, a clinical stage biotech focused on developing TIL therapies for solid tumors. At Instil, she leads research and in vivo pharmacology teams focused on driving early discovery candidates into the clinic for unmodified and engineered TIL therapies. Prior to Instil, she headed the department of Bioinformatics at Arcus Biosciences, where her team built the research bioinformatics and translational infrastructure and supported research and clinical development of Domvanalimab, Quemliclustat and Etrumadenant. Prior to Arcus, she supported oncology biomarker development teams for Xeloda, Avastin and Atezolizumab in solid tumors at Genentech. She has experience in various facets of immunoncology biotech for 8 years with over 16 years in oncology with prior research experience at St. Jude Children’s hospital and Vanderbilt University.