2024 ARCHIVES
Sunday, May 12
Main Conference Registration1:00 pm
Recommended Pre-Conference Short Course2:00 pm
SC5: Targeting Solid Tumors and Understanding the TME
*Separate registration required. See short course page for details.
Tuesday, May 14
Chairperson's Remarks
Mitchell Ho, PhD, Senior Investigator & Deputy Chief, Laboratory of Molecular Biology; Director, Antibody Engineering Program, National Cancer Institute (NCI), National Institutes of Health
Unlocking Novel Therapeutic Targets with Patient-Powered Target Discovery
Jane K. Osbourn, PhD, CSO, Alchemab Therapeutics Ltd.
Identifying genuinely novel targets that have immense therapeutic potential is an increasing challenge across CNS diseases and oncology. Alchemab addresses this challenge by truly disrupting the target discovery process—simply put, we let the patient select the best targets. We have discovered novel disease-relevant targets through our proprietary platform that searches for protective auto-antibodies in patients who are resistant to disease.
Evaluation of Protein/Carbohydrate Combination Epitopes for Targeted Development of Antibodies with Improved Tumor-Specificity
Lisa Kalfhues, PhD, Scientific Project Manager, Glycotope GmbH
Recent advances in antibody engineering have created a portfolio of highly potent therapeutic approaches like antibody-drug conjugates (ADCs), radioimmunoconjugates (RITs), chimeric antigen receptors (CARs), or bispecifics. Due to the increased potency, these drugs are more than ever dependent on very clean targets to prevent side effects. We aim to address this major problem through our approach to develop antibodies with improved tumor specificity and reduced on-target/off-tumor binding, utilizing the aberrant O-glycosylation on tumor cells. A workflow and corresponding database was established to evaluate the O-glycosylation of proteins. This information is subsequently used for targeted discovery of antibodies that bind to protein/carbohydrate combined glycoepitopes (GlycoTargets). A case study will be presented that highlights our workflow to identify suitable GlycoTargets and subsequent development of antibodies.
An Ouyang, Ph.D., Product Manager, Product Development, ACROBiosystems
Developing antibody-drug conjugates faces challenges in selecting suitable antigens for effective targeting while minimizing off-target effects. The lack of well-defined target antigens and tumor heterogeneity adds complexity. As researchers explore innovative strategies like Bispecific antibody-drug conjugates (BsADCs)and tumor microenvironment targeting, using accurate, well-defined target proteins, including B7-H3 and Claudin 18.2, can accelerate ADC antibody screening. Other ADC development solutions are also crucial for successful conjugate crafting.
Refreshment Break in the Exhibit Hall with Poster Viewing4:30 pm
Adding TAPAs to the Menu of Targeting Options
Scott A. Lesley, PhD, CSO, R&D, InduPro Inc.
Most antibody tumor targeting strategies seek to differentiate between disease and normal tissue through the relative abundance of a tumor associated antigens (TAA). Protein dysregulation in cancer can create selective TAAs but such antigens are rare and heterogeneous across patients. While fluid, the cell surface has structure and loose organization of protein neighborhoods. Dysregulation in cancer cells can also result in re-localization of proteins into new neighborhoods. This context of protein neighborhoods provides an additional, underutilized targeting property. We have established a Membrane Interactomics (MInt) platform to identify unique Tumor Associated Proximity Antigens (TAPAs). Using MInt, we have identified several unique proximity-targeting tumor addresses which we have utilized for selective targeting of bispecific ADCs. TAPAs represent and exciting new class of tumor targets.
KnotBodies® - Creating Ion Channel Modulating Antibodies by Fusing Knottins into Antibody Loops
Verity Jackson, PhD, Principal Scientist, Maxion Therapeutics Ltd.
To overcome challenges in antibody discovery against ion channels and GPCRs, Maxion have developed a novel antibody fusion format (KnotBody), by fusing naturally occurring ion channel modulators (knottins) into peripheral CDR loops. This presentation will present our current progress towards developing safe, efficacious and long-acting drugs against previously undruggable targets.
Close of Day6:10 pm
Dinner Short Course Registration6:10 pm
Recommended Dinner Short Course6:30 pm
SC6: Best Practices for Targeting GPCRs, Ion Channels, and Transporters with Monoclonal Antibodies
Wednesday, May 15
Registration and Morning Coffee8:00 am
Daniel A. Vallera, PhD, Lion Scholar and Professor; Director, Section on Molecular Cancer Therapeutics; Professor, Therapeutic Radiology, University of Minnesota Masonic Cancer Center
B7-H7: A Novel Checkpoint Axis and Tumor-Associated Antigen
Tatiana Novobrantseva, PhD, CSO, NextPoint Therapeutics
Immune escape is important for tumor survival. Mechanisms of tumor immune escape can be targeted with therapeutic agents. The B7-H7 immune signaling pathway that NextPoint targets shares similarities with the PD-L1 pathway but is independently expressed and regulated. B7-H7 inhibits activation of T and NK cells via KIR3DL3. Tumors expressing HHLA2 suppress T and NK cells within the tumor. Therefore, blocking B7-H7-KIR3DL3 interaction to enhance the anti-tumor immune response is an attractive strategy for cancer treatment. B7-H7 has also been shown to bind an activating receptor, TMIGD2. To explore the full potential of this axis inhibition, NextPoint Therapeutics is pursuing both B7-H7 and KIR3DL3-specific antibodies, is generating multiple approaches to enable tumor targeting, and is approaching modulation of TMIGD2-mediated activity.
BDC-3042: A Dectin-2 Agonistic Antibody for Tumor-Associated Macrophage-Directed Immunotherapy
Shelley E. Ackerman, PhD, Director, Bolt Biotherapeutics Inc.
This presentation will highlight the preclinical data supporting the development of BDC-3042, a first-in-class Dectin-2 agonistic antibody leading the way in tumor-associated macrophage-directed immunotherapy. This talk sheds light on the potential of harnessing the immune system against cancer through targeted macrophage modulation via a novel target, Dectin-2.
LIGHT (TNFSF14) Co-Stimulation Enhances Myeloid Cell Activation and Anti-Tumor Immunity in the Setting of PD-1 and TIGIT Checkpoint Blockade
George J. Fromm Jr., PhD, CSO, R&D, Shattuck Labs, Inc.
TIGIT-Fc-LIGHT was designed to block all TIGIT-ligand interactions, provide immune co-stimulation to CD8+ T and NK cells via HVEM and myeloid cells by LTBR, and broaden clinical responses into PDL1-low and CPI acquired resistance tumors. TIGIT-Fc-LIGHT differs from antibody blockade of TIGIT, since its co-stimulatory activity (HVEM and LTBR versus DNAM-1) is not directly inhibited by PD1 nor down-regulated on TIL in advanced tumors.
Allison Schulkins, COO, Single Cell Technology, Inc.
Hit generation is critical. Also extremely frustrating when you consider speed vs. quality, lower throughput, limited screening options, having to pare down project scope, developability issues, challenging membrane targets..
There’s a better way: AbTheneum antibody discovery.
We’ve successfully developed assays and delivered data for oncology and beyond for years, combining multiple parallel screening assays in the primary screen to produce multi-layered data with sequences in just 3 weeks.
Coffee Break in the Exhibit Hall with Poster Viewing10:30 am
Targeting Claudin-6 (CLDN6) in Solid Tumors with CAR-T Cells and an Amplifying RNA Vaccine
Sebastian Klobuch, Medical Oncologist, Netherlands Cancer Institute
CAR-T cell therapies in solid tumors are limited by the lack of cancer-specific cell-surface targets. The oncofetal antigen CLDN6 is a promising target that shows high-level aberrant cell membrane expression in many solid tumors. I will discuss CLDN6, the preclinical and clinical anti-tumor activity and safety of CLDN6-targeting CAR-T cells (BNT211-01), and the effect of a CAR-T cell-amplifying RNA vaccine (CARVac) on CAR-T cell expansion and persistence.
Nanobody-Based CAR-T Targeting GPC1 and B7-H3 for Solid Tumors
I will discuss our recent studies on GPC1 and B7-H3 (CD276) as emerging cancer targets, as well as the engineering of CAR T cells for the treatment of pancreatic cancer and neuroblastoma. Additionally, I will describe nanobody technology and single-cell-based transcriptomic analysis of “polyfunctional” CAR T cells to enhance the efficacy of CAR T cell therapy.
Session Break12:10 pm
Luncheon Presentation (Sponsorship Opportunity Available) or Enjoy Lunch on Your Own12:20 pm
Session Break1:20 pm
Developing ImmTAC against PIWIL1, a Promising Novel Colorectal Cancer Target
Christopher Rowley, PhD, Principal Research Scientist, Protein Science Pipeline, Immunocore
ImmTAC bispecifics are designed to elicit a potent T cell response to kill tumor cells. Here we present engineering of IMC-R117C, targeting a peptide derived from PIWIL1, a novel colorectal cancer target, in complex with HLA-A2. We used a proprietary TCR-phage library to identify an optimal TCR with supraphysiological affinity, and an affinity engineering approach that ensures reduced off-target binding; a common challenge with pHLA targeting molecules.
OR641: A Novel Dual Antagonist Antibody that Targets LILRB1 and LILRB2 Inhibitory Receptors and Promotes a Th1-Like Immune Response
Kamal D. Puri, PhD, CSO, OncoResponse, Inc.
The immunosuppression of myeloid cells and lymphocytes within the tumor microenvironment limits efficacy of checkpoint inhibitors. LILRB1 and LILRB2 are inhibitory receptors on immune cells that interact with ligands including HLA Class I and promote an immunosuppressive phenotype of myeloid cells and inhibit T and NK cell cytotoxic activity required for tumor cell death. Dual antagonism of LILRB1 and LILRB2 is a promising strategy to enhance efficacy of checkpoint inhibitors. We have identified OR641 as a dual antagonist antibody that demonstrates superior activity in relieving LILRB1- and LILRB2-mediated immune suppression and enhances both innate and adaptive anti-tumor immunity.
OMO-103: The Molecular Journey of a First-in-Class Direct MYC Inhibitor Mini-Protein
Marie-Eve Beaulieu, PhD, Co-Founder & CSO, Drug Development, Peptomyc SL
A critical cancer driver is MYC, a non-redundant transcription factor essential for cancer cell proliferation and survival. Despite the high interest for a clinical MYC inhibitor, no such drug is currently available, mainly because of the challenges of drugging an intrinsically disordered target localized in the cell’s nuclei. OMO-103, an intravenously delivered cell-penetrating mini-protein, is the first direct MYC inhibitor to have successfully overcome these challenges and completed a clinical trial in human patients showing safety and first signs of clinical activity, supported by molecular target engagement. Here, we present the preclinical development and results from this first-in-human clinical study.
The Monoclonal Antibody NOV2 Targeting Leptin-Dependent NRP-1/OBR Complex, Capable to Enter Cancer Cell Nuclei to Induce DNA Damage and Activate Anti-Tumor Immune Response
Zakia Belaid-Sandal, PhD, CEO & CSO & Co-Founder, THERANOVIR
The anti-NRP-1 antibody “NOV2”, specific to leptin binding domain presents a unique mode of action. It enters cancer cells nuclei to induce their DNA damage and activates thus the anti-tumor response by increasing the number of Tumor Infiltrating CD4+T lymphocytes and CD8+T lymphocytes activation in correlation with metastasis decrease. NOV2 represents a great therapeutic tool for Targeting DNA Damage Response Immuno-Oncology.
Targeting CCR8+ Tregs for Depletion in the Tumor Microenvironment Leads to Robust Anti-Tumor Immunity
Brian Weist, PhD, Senior Research Scientist II, Immunology, Gilead Sciences Inc.
Regulatory T cells are necessary for preventing autoimmune disease but may also inhibit cytolytic T cells in the tumor microenvironment. Reducing tumor-specific Tregs without compromising systemic tolerance has been challenging. CCR8, a chemokine receptor highly expressed on Tregs in solid tumors, serves as an ideal target for ADCC-mediated depletion, leading to robust anti-tumor immunity as a monotherapy as well as in combination with multiple complementary anti-tumor agents.
Ernest Smith, Dr., Chief Scientific Officer / Senior VP Research, Vaccinex Inc.
We have developed a fusion protein technology to enable the direct incorporation of multi-pass membrane proteins such as GPCRs into the membrane of two antigenically distinct poxviruses. The protein of interest is correctly folded and expressed in the cell-derived viral membrane and does not require any detergents or refolding before use. I will describe the use of this technology to discovery antibodies specific for CXCR5 and other multipass membrane proteins.
Q&A and Session Break4:15 pm
Ice Cream Break in the Exhibit Hall with Poster Viewing4:30 pm
SPEED NETWORKING: How Many New Contacts Can You Make?
Mary Ann Brown, Executive Director, Conferences, Cambridge Healthtech Institute
Christina Lingham, Executive Director, Conferences and Fellow, Cambridge Healthtech Institute
Bring yourself and your business cards or e-cards, and be prepared to share and summarize the key elements of your research in a minute. PEGS-Boston will provide a location, timer, and fellow attendees to facilitate the introductions.
STRO-004: A Precisely Designed Tissue Factor-Targeting ADC for Broadened Efficacy & Safety
Garrett G. Gross, PhD, Associate Director, Protein Engineering, Sutro Biopharma Inc.
Sutro's proprietary cell-free protein synthesis and site-specific conjugation platforms enable homogeneous ADCs with best-in-class potential. STRO-004 is a tissue factor-targeting ADC with a stable beta-glucoronidase cleavable linker and TOPO-1 inhibiting Exatecan warhead, precisely positioned by non-natural amino acids with a drug-antibody ratio of 4. STRO-004 demonstrates potent tissue factor-dependent tumor killing in preclinical models and a favorable safety profile in non-human primates, providing a promising clinical candidate for solid-tumor indications.
Conscripting CD180 into Service for Antigen-Specific Immune Responses
Alan Wahl, PhD, CEO, Abacus Bioscience, Inc.
Vaccination is generally ineffective against established disease. To overcome immune tolerance in cancer and chronic infection we’ve focused on stimulating the Antigen Presenting Cell by fusing cancer or viral antigens to antibody derivatives targeting and activating the APC-restricted receptor CD180. This coincident antigen delivery and APC activation via CD180 results in potent yet highly antigen-specific activation of both humoral and cellular response without collateral immune activation. These APC-activating fusion proteins delivering disease antigens clearly demonstrate the potential of this new immunotherapeutic platform.
Cheers to 20 Years Reception in the Exhibit Hall with Poster Viewing6:10 pm
Creating and Fostering a Productive and Effective Mentor-Mentee Relationship
Carter A. Mitchell, PhD, CSO, Purification & Expression, Kemp Proteins, LLC
Deborah Moore-Lai, PhD, Vice President, Protein Development Platform, Abcam
This meet-up is designed for senior scientists that are interested in becoming a mentor for junior scientists: IN-PERSON ONLYOver casual conversation, we will discuss:
Close of Conference7:30 pm
View By:
May 12-13
Display of Biologics
Antibodies for Cancer Therapy
Advances in Immunotherapy
Difficult-to-Express Proteins
ML and Digital Integration in Biotherapeutic Analytics
Biologics for Immunology Indications
May 13-14
Engineering Antibodies
Advancing Multispecific Antibodies
Emerging Targets for Oncology and Beyond
Engineering Cell Therapies
Optimizing Protein Expression
Biophysical Methods
Predicting Immunogenicity with AI/ML Tools
Radiopharmaceutical Therapies
May 15-16
Machine Learning for Protein Engineering
Driving Clinical Success in Antibody-Drug Conjugates
Engineering Bispecific and Multifunctional Antibodies
Next-Generation Immunotherapies
Maximizing Protein Production Workflows
Characterization for Novel Biotherapeutics