2022 Archived Content

Immuno-Oncology Therapeutic Development

Predictive Biomarkers, Preclinical and Translational Studies

23 - 24 May 2022 ALL TIMES BST

The strategies for development of immune-oncology treatments exhibit some important differences compared to other cancer therapy. Essentially all of these products are a form of precision medicine, making the role of predictive biomarkers for patient selection, assessment of response or risk of toxicity, critical for further evaluating success. Preclinical studies also face important challenges, including the choice of in vitro and animal models that more accurately predict efficacy and toxicity. Better preclinical and translational studies are even more important when it is combination therapies that are being developed.

Monday, 23 May

07:30 Registration & Morning Coffee (Quayside Foyer)

ROOM LOCATION: Quayside 2

PREDICTIVE BIOMARKERS

08:30

Chairperson's 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
08:40

Exosomes, Proteins, and Metabolites 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

Circulating exosomes have emerged as means for a multitude of cancer applications including early detection, tumor molecular classification, prediction of therapeutic response, and monitoring of tumor progression and regression. The informative value of circulating exosomes across several types of solid tumors will be presented that are indicative of the value of circulating exosomes as a liquid biopsy approach.

09:10

Liquid Biopsy: From Discovery to Clinical Implementation

Simon Joosse, PhD, Associate Professor, Department of Tumor Biology, University Medical Center Hamberg-Eppendorf

Circulating tumor cells (CTCs) and DNA (ctDNA) in blood have received enormous attention as new "liquid biopsy" biomarkers. Broad clinical usage will depend on standardization of both pre-analytical and analytical procedures as performed in the European Liquid Biopsy Society (ELBS) consortium (www.elbs.eu). Clinical applications of liquid biopsy analyses include early cancer detection, improved cancer staging, early detection of relapse, real-time monitoring of therapeutic efficacy, and detection of therapeutic targets and resistance mechanisms. 

09:40 Speed Networking
10:10 Networking Coffee Break (Quayside Foyer)
10:30

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

Over the past 10 years, circulating tumor cells (CTCs), circulating tumor DNA (ctDNA) and exosomes have received enormous attention as new biomarkers and subjects of translational research. Although these biomarkers are already used in numerous clinical trials, their clinical utility is still under investigation with promising first results. The development of cancer immunotherapy led to new challenges for the development of relevant biomarkers. Additionally, as the treatment needs to be tracked over time, the development of liquid biopsy-based robust biomarkers is urgently needed. Current challenges of liquid biopsy research, from discovery to clinical application in Immuno-Oncology will be highlighted.

11:00

Quantitative MHC-I Peptide Measurement to Support the Development of Cancer Immunotherapeutic Approaches Using Targeted Mass Spectrometry

Axel Ducret, PhD, Senior Principal Scientist, Pharmaceutical Sciences, Roche Innovation Center Basel, Switzerland

Demonstrating antigen availability on the surface of tumor cells is crucial for the success of cancer immunotherapeutic approaches. We detail in this presentation the quantitative mass spectrometric strategy that we developed to measure generically MHC-I peptides of interest after initial characterization. We present two representative examples for which we demonstrated and quantified MHC-I peptide receptor complexes in relation to in vitro treatment effect.

11:30

High-Dimensional, Longitudinal Immunophenotyping of PBMCS by CyTOF from Resectable Head and Neck Squamous Cell Carcinoma Patients Treated with Durvalumab (anti-PD-L1) with and without Tremelimumab (anti-CTLA-4)

Brian Lee, Graduate Student, Seoul National University, College of Medicine

Combination immune checkpoint inhibitor therapy has shown promising results in treating various cancer types. However, recent studies in HNSCC patients have shown no significant differences in overall survival between durvalumab (aPD-L1) or durvalumab + tremelimumab (aCTLA-4) therapy compared to standard of care. To understand immune landscape dynamics of these patients, we designed a CyTOF panel to broadly identify all major immune cell types and deeply profile regulatory T cells.

12:00 Enjoy Lunch on Your Own
12:40 Networking Refreshment Break (Quayside Foyer)

PRECLINICAL TOXICITY STUDIES

13:20

Chairperson's Remarks

Richard Stebbings, PhD, Principal Scientist, Toxicology, AstraZeneca
13:25

Preclinical Strategies for Modelling and Predicting Toxicity for Immuno-Oncology Treatments

Richard Stebbings, PhD, Principal Scientist, Toxicology, AstraZeneca

Preclinical safety testing of novel immune-oncology therapeutics require assessment of pharmacology driven cytokine release syndrome and on-target off-tumour risk where standard toxicity models are often not applicable. To address these challenges we are applying tumour bearing humanised mouse models, primary cell screens and membrane protein arrays to safety assessment. The benefits and challenges of these approaches will be discussed.

13:55

Understanding Cellular and Molecular Players in Cytokine Release Syndrome (CRS)

Gabrielle Leclercq, PhD, Postdoctoral Scientist, Pharma Research & Early Development, Roche

CRS is one of the major safety liabilities associated with treatment with T cell engaging therapies in the clinic, including CAR T cells and T cell engagers. A series of in vitro and in vivo preclinical studies were conducted to gain a better understanding of the biological mechanisms inducing CRS, providing new avenues for the mitigation of the same. 

14:25

Regulatory T Cells as Determinants of Adaptive and Innate Function in Brain Tumours

Felipe Galvez-Cancino, PhD, UCL Cancer Institute, Department of Hematology, University College London

The role of Tregs in glioblastoma remains controversial and their depletion has the potential to open the door to new treatment strategies for this devastating disease. By combining the analysis of human datasets, the use of a novel Treg targeting antibody and a newly developed mouse model of glioblastoma we have found that Treg depletion leads to deep changes in the adaptive and innate compartments allowing for the rational design of novel combination therapies targeting the glioblastoma microenvironment.

14:55 Refreshment Break with Exhibit and Poster Viewing (Quayside Foyer)

PRECLINICAL STUDIES WITH NOVEL THERAPEUTICS

15:35

MB097: A Potent Therapeutic Consortium of Bacteria Clinically-Defined by Precision Microbiome Profiling of Melanoma Patients

Matthew Robinson, PhD, Vice President, Translational Biology, Microbiotica Ltd.

Microbiotica’s precision microbiome profiling and the MELRESIST clinical study has allowed us to identify a consortium of bacteria, MB097, strongly linked to immune checkpoint inhibitor response across multiple melanoma cohorts. MB097 synergises with anti-PD1 in vivo to inhibit tumour growth. The bacteria also demonstrate multiple interactions with primary human immune cells in vitro, leading to tumour cell killing. MB097 is being scaled up for clinical trials as a novel co-therapy with anti-PD1 in melanoma.

16:05

Preclinical Development of CD47 Bispecific Antibodies for Haematological and Solid Tumours

Nicolas Fischer, PhD, CEO, Light Chain Bioscience

We have developed several bispecific antibody programs targeting the innate immune checkpoint CD47 for different indications in oncology. Two clinical-stage programs aim at selectively blocking CD47 at the surface of malignant cells. In addition, we are also developing two CD47/PD-L1 bispecific antibodies with differentiating mechanisms of action, affording either independent blockade of the immune checkpoints or targeted CD47 inhibition to PD-L1+ cells. The different rationales, key characteristics, and development status of these four CD47 targeting programs will be discussed.  

16:35

CodaLytic, a Novel Codon-Modified Virus for Immunotherapy of Solid Tumors

Johanna Kaufmann, PhD, Executive Vice President, Oncology, Codagenix

In contrast to many oncolytic viruses that are attenuated by either gene deletion or mutation and the armed with immunomodulatory transgenes to promote anti-tumor immune responses, CodaLytic is a novel virotherapeutic that is synthetically engineered to use alternative codons. We will describe its design, efficacy, holistic modulation of the tumor microenvironment, and ability to stimulate potent anti-tumor immune responses in preclinical models. This will support the utility of the virus as a monotherapy and in combination with checkpoint inhibition.

17:05 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 on the conference website for a complete listing of topics and descriptions.

IN-PERSON ONLY: Means of Overcoming Immunosuppressive Mechanisms in the Tumour Microenvironment

Mark S. Cragg, PhD, Professor of Experimental Cancer Biology, School of Cancer Sciences, Faculty of Medicine, University of Southampton
Sophia N. Karagiannis, PhD, Professor, Translational Cancer Immunology & Immunotherapy, Kings College London
  • Different types of suppressive mechanisms to be overcome
  • Target antigen heterogeneity
  • Selection of target antigens for antibody immunotherapy of solid tumours
  • Importance of affinity to the target antigen and to Fc receptors for tissue penetration and efficacy
  • Importance of isotype selection 
  • Are multi-specifics the answer?​

IN-PERSON ONLY: Understanding and Reducing Toxicity

Richard Stebbings, PhD, Principal Scientist, Toxicology, AstraZeneca
Gabrielle Leclercq, PhD, Postdoctoral Scientist, Pharma Research & Early Development, Roche

IN-PERSON ONLY: Potential of Immunocytokines for Cancer Immunotherapy

Dario Neri, PhD, CEO and CSO, Philogen
  • Interrogating the cytokine environment and the immunosuppresive cell populations infiltrating tumours
  • The biology and the problems associated with cytokines
  • Challenges with toxicity
  • Temporal and spatial considerations
  • To what extent can cytokine-based therapeutics improve the therapeutic index?
  • Considerations on mechanisms of action
  • Potential for combination therapy​

IN-PERSON ONLY: Selecting and Evaluating Combination Therapies

Robert Arathoon, PhD, Founder, President & CEO, Kenjockety Biotechnology, Inc.

Rationale for combination therapies

Combinations with immune checkpoint inhibitors

Challenges for preclinical evaluation of combination therapies

Addressing dosing, safety, and efficacy in translational studies

17:50 Reception in the Exhibit Hall with Poster Viewing (Quayside Foyer)
18:50 Close of Day

Tuesday, 24 May

08:00 Registration and Morning Coffee (Quayside Foyer)

ROOM LOCATION: Quayside 2

TRANSLATIONAL STUDIES

08:30

Chairperson's Remarks

Samuel Zhang, PhD, Chief Business Officer, NeoImmuneTech, Inc.
08:35

FAP-CD40: A Novel Tumor-Stroma Targeted CD40 Agonist, Abrogates Systemic Toxicity, and Enables Administration of High Doses to Induce Effective Anti-Tumor Immunity

Christine Trumpfheller, PhD, Senior Scientist, Roche Innovation Center, Zurich

Activation of CD40+ antigen-presenting cells (APCs) aims at generating anti-tumor T cell responses. So far, dose limitations due to on-target systemic toxicity restrict the amount of systemically administered CD40 agonists that can be delivered to the tumor to activate APCs locally. We developed a fibroblast-activation-protein-a (FAP)-CD40 antibody, which activates CD40 solely in presence of the tumour stromal antigen FAP. FAP-CD40 enables safe administration of higher doses, essential for inducing antitumor immunity.

09:05

Promising Early Clinical Efficacy in Immune Cold Tumors with NT-17 in Combination with Pembrolizumab

Samuel Zhang, PhD, Chief Business Officer, NeoImmuneTech, Inc.

NT-I7, a long-acting IL-7, has shown promising early anti-tumor activity in two immune-cold tumor types. In MSS CRC, where pembro had 0% ORR, 3 PR/iPR were observed in 17 evaluable pts treated with NT-I7+pembro, for an iORR of 18%. Furthermore response was also observed in pancreatic cancer, where pembro also had 0% ORR, despite short followup. Responses, once occurred, are durable and continue to deepen over time.

Mourad Ferhat, EMEA Product Manager, Isoplexis

The successes with immune checkpoints and CAR T cells in treating multiple cancer types have established immunotherapy as a powerful curative option for patients. Unfortunately, many patients do not derive benefit or long-term responses, highlighting a pressing need to perform complete investigation of the underlying mechanisms. We will show how high-plex single-cell functional proteomics can help to deliver new insights into biomarker discovery and guide the rational design of next-generation immunotherapies.

10:05

Potent, Tumor-Selective, Bispecific, Antibody Therapeutics Simultaneously Targeting IO and Drug Resistance Antigens in Cancer

Robert Arathoon, PhD, Founder, President & CEO, Kenjockety Biotechnology, Inc.

We have developed BsAbs engineered to preferentially target tumor rather than normal tissue and that demonstrate potent activity in xenograft models of hematologic and solid tumors, both as monotherapy and in combination with other agents. The tumor-selective activity is mediated by simultaneously masking an IO target while antagonizing an efflux transporter, thereby inducing an enhanced immune response and counteracting drug resistance. The potent activity of this dual targeting on tumors and the lack of binding to normal tissue provides for enhanced efficacy with the additional benefit of reduced toxicity.

10:35 Coffee Break with Exhibit and Poster Viewing (Quayside Foyer)

PLENARY ROOM LOCATION: Quayside 1 & 2

PLENARY SESSION

11:15

Chairperson's Remarks

Robert W. Wilkinson, PhD, Senior Director, Oncology R&D, AstraZeneca
11:20

Antibody-Cytokine Fusions as Bispecific Immunomodulatory Proteins

Dario Neri, PhD, CEO and CSO, Philogen

Many solid tumors and hematological malignancies are characterized by an insufficient density and activation of tumor-specific leukocytes (e.g., CD8+ T cells, NK cells) at the site of disease. The antibody-based targeted delivery of suitable cytokine payloads (e.g., IL2, IL12, TNF) to the tumor may activate lymphocytes at the site of disease, mediates a potent anti-cancer activity in preclinical models of cancer, and is being investigated in clinical trials.

11:55

Targeting Regulatory T Cells within Brain Tumours: From Mechanisms to Therapies

Sergio A. Quezada, PhD, Professor, Cancer Immunology & Immunotherapy, University College London

T cells have a critical role in the maintenance of immune homeostasis in mice and man. However, despite their role in maintaining host integrity, their function is known to be hijacked in the context of cancer. In this talk, I will discuss the role of regulatory T cells in the context of the brain tumour microenvironment, as well as old and new strategies to target this compartment, including our previous work on anti-CTLA-4 and the recent development of a new CD25-targeting Treg depleting antibody for mice and man.

12:30 Enjoy Lunch on Your Own
13:10 Coffee Break with Exhibit and Poster Viewing (Quayside Foyer)
13:45

Better Targets, Better Targeting: Iterative CAR T Studies in an Academic Programme

Claire Roddie, PhD, Professor, Haemato-Oncology, UCL Cancer Institute

Here we will discuss current paradigms, recent learnings and future directions for the academic UCL CAR T cell programme, encompassing our plans for blood cancers and our increasing footprint in solid tumours.

14:20

Precision Medicine-Enabled Development of PD-1 Antibody Mono- and Combination Therapy

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

Precision medicine enabled screening studies identified unambiguous effect of PD-1 monoclonal antibody (mAB) activity in more than 30 major cancer types. Precision medicine tools enabled accelerated development and approvals of monotherapy applications. Precision medicine tools led to histology agnostic monotherapy approvals. These same tools provided powerful insights to mechanisms of treatment resistance and informed the selection of combination therapies. PD-1 mAB in combination with a variety of agents are having a major impact in cancer treatment.

14:55 Refreshment Break with Exhibit and Poster Viewing (Quayside Foyer)
14:55 Close of Immuno-Oncology Therapeutic Development Conference
14:25

Regulatory T Cells as Determinants of Adaptive and Innate Function in Brain Tumours

Felipe Galvez-Cancino, PhD, UCL Cancer Institute, Department of Hematology, University College London

The role of Tregs in glioblastoma remains controversial and their depletion has the potential to open the door to new treatment strategies for this devastating disease. By combining the analysis of human datasets, the use of a novel Treg targeting antibody and a newly developed mouse model of glioblastoma we have found that Treg depletion leads to deep changes in the adaptive and innate compartments allowing for the rational design of novel combination therapies targeting the glioblastoma microenvironment.






Register Now

Modulating the Tumour Microenvironment