Engineering Antibody-Drug Conjugates Banner

 

Making a successful ADC relies not only on the science – the understanding of the underlying mechanism of action of ADCs, but also on the art – the expert combination of the 3 components – antibody, linker and cytotoxic drug – to achieve efficacy and tolerability. By combining the selectivity of targeted treatment with the cytotoxic potency of chemotherapy drugs, ADCs hold the power to revolutionize disease treatment, especially for cancer.
 

However, every novel technology has its challenges. ADCs suffer from low therapeutic window, tumor non-specificity, off-target toxicity, lack of efficacy, to name just a few. At the 8th Annual Engineering Antibody-Drug Conjugates conference, scientists will present their research and explorations on using alternative scaffolds and new payloads to improve half-lives and specificity, optimizing linker-payload chemistry to enhance efficacy, as well as discuss strategies to expand therapeutic window and improve stability of their ADCs.

 

Final Agenda

Recommended Short Course(s)*

SC10: Critical Considerations for the Design and Development of Antibody-Drug Conjugates


*Separate registration required.

WEDNESDAY, MAY 2

7:30 am Registration(Commonwealth Hall) and Morning Coffee (Harbor Level)

ALTERNATIVE SCAFFOLDS AND NEW PAYLOADS
Cityview 2

8:30 Chairperson’s Remarks

Ravi Chari, PhD, Vice President, Chemistry & Biochemistry, ImmunoGen, Inc.

8:40 Abdurin-Drug Conjugates - Small Size and Long Half-Life to Improve Drug Concentration in the Target Tissue

Kurt_GehlsenKurt Gehlsen, PhD, Vice President & CSO, Therapeutics, Research Corporation Technologies, Inc.

Abdurins are a novel antibody-like scaffold that can be engineered to bind to targets of interest and due to an FcRn binding motif, Abdurins have a circulating half-life longer than any other protein scaffold of similar size. Abdurins can be fused with protein toxins, other binding domains and engineered to carry payloads. Abdurin-drug conjugates retained high affinity binding and had in vivo half-life up to 70 hours for certain conjugates.

9:10 Antibody Targeted Amanitin Conjugates (ATACs) - Expanding the ADC Landscape with a New Payload Targeting RNA Polymerase II

Andreas_PahlAndreas Pahl, PhD, CSO, Heidelberg Pharma

Antigen-Targeted Amanitin-Conjugates (ATACs) represent a new class of ADCs using the payload Amanitin. This payload introduces a novel mode of action into oncology therapy, the inhibition of RNA polymerase II. The technology platform includes Amanitin supply, site-specific conjugation, demonstrated safety profile and biomarker. Improvements of the technology and an update of the development of HDP-101 will be presented. HDP-101 is the first ATAC directed against BCMA entering Phase I trials by the end of 2018.

9:40 Preclinical Validation of Site Specifically Conjugated ADCs with Potent Anthracycline Payloads

Roger_BeerliRoger Beerli, PhD, CSO, NBE-Therapeutics AG

We present a novel ADC format based on the site specific conjugation of a derivative of the anthracycline PNU-159682 using the transpeptidase Sortase A. The use of non-cleavable peptide linker provides exquisite stability in vivo, whereas the anthracycline payload endows the ADC with superior potency combined with attractive immune-oncology properties intrinsic to this class of compounds. Homogeneous PNU-ADCs directed against HER2 and ROR1 were generated and shown to have very high anti-tumor efficacy in vivo, both in PDX as well as in syngeneic solid tumor models. In case of HER2, the PNU-ADC exceeded the efficacy of T-DM1 used as a benchmark ADC. In syngeneic breast cancer models, both HER2, as well as ROR1 ADCs resulted in the induction of a long-lasting tumor-selective anti-tumor immunity involving activated CD8 T cells. Importantly, a repeated-dose non-GLP toxicology study in cynomolgus monkeys did not reveal significant toxin-related pathology of PNU-ADCs at any of the evaluated dose-levels, strongly supporting further preclinical and clinical development of this promising new ADC format.

10:10 Coffee Break in the Exhibit Hall with Poster Viewing (Commonwealth Hall)

10:55 Design of a Novel, Glycan Targeting Antimicrobial ADC as a New Therapeutic Strategy for the Treatment of P. aeruginosa Infection

Obadiah_PlanteObadiah J. Plante, PhD, Director, Research, Visterra, Inc.

This presentation will describe Visterra’s approach to the development of a novel antibody-based therapeutic for the treatment of established P. aeruginosa infections. The antibody targets a core region of lipopolysaccharide present at high density on P. aeruginosa and common across serotypes. To provide direct bactericidal activity, we have designed potent anti-microbial peptides that rapidly kill bacteria when directly conjugated to our antibody in the form of an ADC.

11:25 The Development of Antibody Conjugates for Targeted Delivery of siRNA

Chawita_NetirojjanakulChawita Netirojjanakul, PhD, Sr. Scientist, Therapeutic Discovery, Amgen

Advances in small interfering RNA (siRNA) technology result in numerous RNAi-based therapies being pursued in clinical trials. However, several challenges including targeted delivery of siRNA have limited the use of siRNAs as therapeutics. Here, we established a method to prepare and characterize well-defined antibody-siRNA conjugates and demonstrated that using this platform, siRNA can be delivered into specific cells or tissues in vitro and in vivo.

11:55 Attachment Site Cysteine Thiol pKa is a Key Driver for Site-Dependent Stability of Disulfide & Maleimide-Linked THIOMAB(TM) Antibody-Drug Conjugates

Breanna_VollmarBreanna S. Vollmar, PhD, Senior Scientific Researcher, Protein Chemistry, Genentech, Inc.

We set out to understand the underlying mechanisms of site-dependent stability for our THIOMAB™ antibody drug conjugate (TDC) platform utilizing engineered cysteines at specific sites on the antibody. Our observations suggest that cysteine thiol pKa is a significant driver of the circulation stability of TDCs utilizing disulfide or maleimide attachment chemistry and represents a new parameter for the optimization of next generation ADCs utilizing engineered cysteines.

 

12:25 pm Latest Advances Developing Antibody Drug Conjugates (ADCs) and Other Bioconjugates Using SMARTag Technology

David_RabukaDavid Rabuka, Global Head, Research & Development, Chemical Biology, Catalent Biologics, Catalent Pharma Solutions

We have developed the SMARTagTM technology platform, which enables precise, programmable, site-selective chemical protein modification. We will present recent data on our novel protein modification platform and its application to generating novel bioconjugates, including ADCs, utilizing our new conjugation chemistries and linkers. Additionally, we will highlight progress in developing conjugates with a focus on preclinical studies as well as highlight our progress in cell line development and manufacturing of using this chemoenzymatic approach.

12:55 Luncheon Presentation (Sponsorship Opportunity Available) or Enjoy Lunch on your Own

1:55 Session Break

OPTIMIZATION OF LINKER-PAYLOAD CHEMISTRY
Cityview 2

2:10 Chairperson’s Remarks

Kurt Gehlsen, PhD, Vice President & CSO, Therapeutics, Research Corporation Technologies, Inc.

2:15 Chemical Fusion: A General Method for Equimolar Linking of Proteins and Payloads

Alain_WagnerAlain Wagner, PhD, Research Director, Biofunctional Chemistry, Faculty of Pharmacy, University of Strasbourg

We have designed a general conjugation methodology that involves native protein and leads to single DAR conjugates. Our method applies with virtually no limitations to all types of proteins and payloads. It enables preparation of conjugate bearing one, two or more payloads, with precise control for each of them. Noteworthy the simple mono-conjugated DAR 1 opened interesting prospects as a building block to engineer precise protein-oligonucleotide, protein-protein, protein-nanoparticle constructs.

2:45 Enzymatic (Dual) Site-Specific Conjugation of Engineered and Native Antibodies

Philipp_SpycherPhilipp R. Spycher, PhD, PSI Founder Fellow, Center for Radiopharmaceutical Sciences, Paul Scherrer Institut

We will show single and dual site-specific conjugation of engineered antibodies with various functional payloads as well as the controlled modification of antibody-fragments and other proteins using solid-phase immobilized microbial transglutaminase (MTG). Additionally, we will introduce a novel proprietary enzymatic conjugation technology that enables site-specific payload attachment to native antibodies, thus generating well-defined ADCs that have a native IgG antibody structure.

3:15 Poster Highlight I: A Novel Platform for Versatile Payload Conjugating to a Glyco-Engineering tri-Mannosyl Core Antibody

Chun-Chung Lee, PhD, Research Fellow, Institute of Biologics, Development Center for Biotechnology

In this study, we developed a novel site-specific tri-mannosyl core antibody-drug conjugate platform. The novelty of this platform is that we are able to control the antibody conjugating single payload (DAR=2 or 4) or dual payloads (DAR=2A&2B) by one step or stepwise of MGAT-1 and MGAT-2 treatments, respectively. Through this tri-mannosyl core ADC platform, we are able to control the different toxin dosages (DAR=2 or 4) conjugating to the same antibody. As a result, the correlation between conjugating dosages of toxin, safety and efficacy will be methodically evaluated. The platform will also be available for designing the therapeutic antibody conjugated with dual payloads, which will target the same molecule in cancer cells and induce different drug response. The platform is also available for use as companion diagnostics and therapy for cancers when the antibody conjugating both image dye and cancer drugs. 

3:30 Poster Highlight II: Concisely Produced Homogeneous Antibody-Drug Conjugates by a Tryptophan-Selective Protein Bioconjugation

Kounosuke Oisaki, PhD, Lecturer, Graduate School of Pharmaceutical Sciences, University of Tokyo

In 2016, we reported a transition metal-free method for tryptophan (Trp)-selective bioconjugation of proteins. this method exhbits low levels of cross-reactivity and leaves higher-order structures of the protein and various functional groups therein unaffected. Recently, studies aiming at concise production of a homogeneous antibody-drug conjugate (ADC) using the Trp-selective bioconjugation are ongoing in our group. We would like to present recent progress.

3:45 Refreshment Break in the Exhibit Hall with Poster Viewing (Commonwealth Hall)

4:45 Problem-Solving Breakout Discussions (Commonwealth Hall)

Getting Beyond Therapeutic Targets – Delivery Targets Matter Too

Moderator: Jan E. Schnitzer, M.D., Director, Professor, Cellular & Molecular Biology, Proteogenomics Research Institute for Systems Medicine (PRISM)

 

How Do We Improve Drug Concentrations in Targeted Tissues?

Moderator: Kurt Gehlsen, PhD, Vice President & CSO, Therapeutics, Research Corporation Technologies, Inc.

  • ADCs
  • Bispecifics
  • New targeting scaffolds/ligands/peptides
  • How do we assess drug concentration in early trials?

Limitations of First-Generation ADCs in Solid Cancers

David Goldenberg, ScD, MD, Founder and Former CSO, Immunomedics, Inc.

  • Prospects for next-generation ADCs in solid cancers
  • Role of antigen target, drug-antibody ratio, nature of drug, conjugation chemistry, drug resistance.

Improving Therapeutic Index of ADCs

Renu Singh Dhanikula, PhD, Senior Research Investigator, Metabolism and Pharmacokinetics, Bristol-Myers Squibb

  • Off-target toxicity of ADCs
    • In vitro assays to understand non-specific uptake of ADCs
    • Linker chemistry to reduce off-target payload toxicity
  • How we can improve therapeutic index (TI) of ADCs
    • Do site-specific ADCs increase therapeutic index?
    • Does drug/antibody ratio has impact on TI?
  • Combination with Immunoncology/chemotherapy to increase TI

5:45 Networking Reception in the Exhibit Hall with Poster Viewing (Commonwealth Hall)

7:00 End of Day

THURSDAY, MAY 3

8:00 am Registration(Commonwealth Hall) and Morning Coffee (Harbor Level)

8:30 Chairperson’s Remarks

Alain Wagner, PhD, Research Director, Biofunctional Chemistry, Faculty of Pharmacy, University of Strasbourg


8:35 KEYNOTE PRESENTATION: Clinical and Technological Advances in ADCs

Dennis_BenjaminDennis Benjamin, PhD, Senior Vice President, Translational Research, Seattle Genetics

Clinical and preclinical studies of auristatin-based ADCS including brentuximab vedotin will be presented. Recent developments in ADC technology will also be discussed, including engineered antibodies for delivery and novel cytotoxic payloads.

NOVEL TARGETS
Cityview 2

9:05 A Novel Cell Surface Target on Cancer Cells and Development of a Targeted ADC

Ginette_SerreroGinette Serrero, PhD, CEO, A&G Pharmaceutical, Inc.

The key to the ADC approach is the development of an antibody against a desired target preferentially expressed on cancer cells, that results in the internalization of the payload and eventual death of the cell, while limiting the off-target side effects. The presentation will cover antibody development and subsequent conjugation and payload selection for initial proof of concept studies prior to moving the antibody into linker and payload preclinical studies, using a real example of a mAb against a novel CSP target.

Applied Biomath9:35 Computational Exploration of Mechanistic Determinants of ADC Pharmacokinetics Using QSP Modeling Strategies

John Burke, PhD, Co-Founder, President and CEO, Applied BioMath, LLC

The pharmacokinetics of ADC therapeutics typically show a discrepancy between PK of total antibody and of conjugated antibody, carrying one or more payload molecules. This discrepancy is often attributed to deconjugation, however recent evidence suggests that underlying mechanisms may be more complex. This presentation will demonstrate a computational approach to understand the impact of DAR and resulting changes in molecular properties on overall PK and relative payload disposition as observed in preclinical and clinical studies.

10:05 Coffee Break in the Exhibit Hall with Poster Viewing (Commonwealth Hall)

EXPANDING THERAPEUTIC WINDOW AND IMPROVING STABILITY
Cityview 2

11:05 Expanding Therapeutic Windows of Tubulysin Antibody-Drug Conjugates with Stable Hydrophilic Linkers

Robert_ZhaoRobert Yongxin Zhao, PhD, CEO, Hangzhou DAC Biotech Co., Ltd.

To address the liver toxicity issue in our original anti-Her2-tubulysin ADC, we developed a novel stable hydrophilic linker which, while maintaining better efficacy than T-DM1 in xenograft mouse models, proved significantly less toxic than T-DM1 in mice and cynomolgus monkeys. The overall therapeutic window was 4-6 times wider than that of T-DM1. The most percentage of metabolite of tubulysin analog was found in urine of animals demonstrating a clear correlation between its much reduced side effect and the metabolic pathway.

11:35 IMGN632, A Novel ADC with Uniquely Wide Therapeutic Window in Preclinical Models of AML

Yelena_KovtunYelena Kovtun, Associate Director, Pipeline R&D, ImmunoGen, Inc.

IMGN632 is a novel conjugate of anti-CD123 antibody with mono-imine containing Indolinobenzodiazepine payload. Unlike other conjugates (including ADCs with proven clinical success), only IMGN632 demonstrated potent anti-leukemic activity at the concentrations 100-fold lower the levels that impact normal human cells. The strategy to select an optimal antibody, payload, linker and conjugation method to achieve this uniquely wide therapeutic window will be covered in the presentation.

12:05 pm ADC Pumping into Solid Tumors Boosts Drug Potency and Efficacy

Jan_SchnitzerJan E. Schnitzer, MD, Director, Professor, Cellular & Molecular Biology, Proteogenomics Research Institute for Systems Medicine (PRISM)

Current ADC can’t deliver drugs inside solid tumors specifically, rapidly or robustly. Near MTD doses required to drive ADC across endothelial barriers are inadequate to unleash drug potency solely inside tumors. Off-target toxicities minimize therapeutic indices. We circumvent this passive transvascular delivery paradigm by utilizing caveolae pumping and the first antibody to actively penetrate tumors. Immunoconjugates concentrate imaging and therapeutic agents inside resistant and metastatic tumors, enabling precise imaging within 1 hr and boosting therapeutic indices >100-fold (<<<MTD eradication).

12:35 End of Engineering Antibody-Drug Conjugates


Register Now

View By:


Premier Sponsors

   FairJourneyBiologics Integral-Molecular_NEWLONZAOmniAb  Samsung_Biologics  UnchainedLabs