ADC


The complexity of antibody-drug conjugates with its many moving parts makes this field incredibly challenging yet full of potential for innovation. From novel targeting ligands to new conjugation methods, from multiple payloads to changing the drug-antibody ratio, all these are challenging the convention for the design and development of next-generation ADCs. At Antibody-Drug Conjugates I: New Ligands, Payloads and Alternative Formats, we invite scientists to present their new technologies and showcase how they are redefining the rules in the field of ADCs.

Continue your exploration of the antibody-drug conjugates field by attending the accompanying meeting, Antibody-Drug Conjugates II: Advancing toward the Clinic on April 28-29.

WEDNESDAY, APRIL 27

7:00 am Registration and Morning Coffee

8:00 Chairperson’s Remarks

Christopher D. Thanos, Ph.D., Senior Director, Biotherapeutics Discovery, Halozyme Therapeutics, Inc.


8:10 KEYNOTE PRESENTATION:

Determination of Cellular Processing Rates Points to Key Parameters for Antibody-Drug Conjugate Design

K_Dane_WittrupK. Dane Wittrup, Ph.D., C.P. Dubbs Professor, Chemical Engineering & Biological Engineering, Associate Director, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology

Numerous processing steps occur before the active drug component of an ADC can reach its intracellular target. Increased understanding of ADC cellular processing may facilitate more rational design of ADCs. In this work, we present a generalizable method to determine kinetic parameters, which can be used in a basic model for cellular processing of ADCs and can be incorporated into larger scale pharmacokinetic/pharmacodynamic models.

8:40 KEYNOTE PRESENTATION:

Combining Antibody-Drug Conjugates and Immune-Mediated Cancer Therapy: What to Expect?

Hans-Peter GerberHans-Peter Gerber, Ph.D., Vice President, CSO, Bioconjugates Discovery & Development, Oncology Research Unit East, Pfizer Worldwide Research & Development

Toxins targeting DNA like anthracyclines or tubulin poisons like vinblastine can stimulate the innate immune system. When combined immuno-oncology (IO) compounds, both classes of toxins further enhanced the adaptive immune response and improved the anti-tumor responses. Therefore, identification of optimal combination regimens between ADCs employing different classes of toxins and IO compounds holds strong promise to overcome the key limitations of current immune checkpoint inhibitors, by increasing the recruitment and infiltration of CD8+ effector T cells to the tumor.


ALTERNATIVE FORMATS AND NEW TARGETING LIGANDS

9:10 Small Ligand-Targeted Drug Conjugates: An Alternative to ADCs

Phillip_LowPhilip S. Low, Ph.D., Director of the Purdue Center for Drug Discovery, Ralph C. Corley Distinguished Professor, Department of Chemistry, Purdue University

We have developed small molecule ligands for use in targeting attached drugs to pathologic cells, thereby avoiding collateral toxicity to healthy cells. We have also developed low molecular weight targeting ligands to deliver attached drugs selectively to cancers that over-express PSMA, CCK2 receptor, neurokinin 1 receptor, carbonic anhydrase IX , and several other tumor-specific receptors. Finally, ligand-targeted imaging and therapeutic agents for autoimmune, inflammatory, and infectious diseases (e.g. malaria, rheumatoid arthritis, multiple sclerosis, psoriasis, atherosclerosis, osteoarthritis, etc.) will also be described.

9:40 Targeted Protein Therapeutics (TPTs) for the Treatment of Cancer

Greg_AdamsGreg Adams, Ph.D., Chief Development Officer, Viventia Biotech

TPTs are fully biologic constructs containing antibody fragments and protein toxin payloads in a single engineered molecule. The preclinical/clinical development of TPTs employing fully deimmunized payloads for the treatment of systemic disease and non-deimmunized payloads for use in the treatment of loco regional disease will be discussed.

10:10 Coffee Break in the Exhibit Hall with Poster Viewing

10:55 Development of Probody-Drug Conjugates Targeting Highly Expressed Tumor Antigens

Luc_DesnoyersLuc Desnoyers, Ph.D., Director, Oncology, CytomX Therapeutics, Inc.

PDCs are antibody prodrugs that are designed to be activated in tumors while avoiding binding to normal tissues. PDCs can safely enable targeting of antigens with broad, persistent & very high expression in cancer that are also expressed in normal tissues, and therefore cannot be approached with traditional Antibody-Drug Conjugates. Such targets can show >70% prevalence at 3+ expression in many cancer types. Preclinical proof of concept for safety, efficacy & developability of PDCs to high expression targets will be shown.

11:25 Immunomodulation by CO Delivered from Artificial Metalloproteins

Goncalo_BernardesGoncalo J.L. Bernardes, Ph.D., Group Leader, Chemistry, University of Cambridge

A new class of therapeutic metalloproteins allows for the controlled and targeted delivery of carbon monoxide (CO) into tumors. When administered into tumor bearing mice, the CO-releasing metalloproteins result in strong tumor growth retardation. The CO-mediated effect is due to the combined downregulation of important angiogenic factors as well as activation of CD8 cytotoxic T cells. Finally, when used in combination with current standard of care chemotherapeutic drugs, the novel CO immune-modulation treatment results in cancer cures in mice.

11:55 Drug Conjugation and Delivery Enabled by a Tumor-Targeting Peptide-Fc Fusion

Jennifer_ChochranJennifer Cochran, Ph.D., Associate Professor, Bioengineering, Stanford University

We are creating novel peptide-drug conjugates for targeted delivery of chemotherapeutic agents to tumors. As an example, an integrin targeted peptide-Fc fusion, conjugated to an auristatin derivative, was effective as a single agent at inducing regression and prolonged survival in tumor xenograft models. These studies provide proof-of-concept for further development of peptide-drug conjugates as attractive alternatives to ADCs for tumor targeting and drug delivery applications.

12:25pm Selected Poster Presentation:

Recent Advances in Chemistry & Biology of Protein Drug Conjugates (PDC)

Daniel Custar, Ph.D., Scientist, Oncology, Eisai Inc.

Preclinical work around a novel peptide drug conjugate (PDC) will be disclosed. The syntheses of a suitable cytotoxic warhead, along with linker development, and conjugation to a novel peptide will be outlined. Finally the in vitro stability and in vivo efficacy and PK data will be presented.

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

1:55 Session Break


NOVEL APPLICATIONS OF ADCS

2:10 Chairperson’s Remarks

Hans-Peter Gerber, Ph.D., Vice President and CSO, Bioconjugates Discovery & Development, Oncology Research Unit East, Pfizer Worldwide R&D

2:15 Development of a Novel Protease Cleavable Linker for Anti-Staphylococcus Aureus Antibody-Antibiotic Conjugates

Martine_DarwishMartine Darwish, Senior Scientific Researcher, Protein Chemistry, Genentech

Antibody-Antibiotic Conjugates (AACs) employ an antibody specific for cell wall components of Staphylococcus aureus conjugated with a potent antibiotic. We describe the development of a peptide linker that is cleaved by staphopain B, a secreted endopeptidase of S. aureus. The resultant AAC has demonstrated efficacy in in vitro and in vivo models of MRSA infection, providing a novel mechanism by which to target MRSA infections and release payload in a disease specific manner.

2:45 MM302, A Novel Antibody-Drug Conjugated Liposomal Doxorubicin That Specifically Her2-Overexpressing Cancer Cells

Istvan Molnar, MD., Vice President, Clinical Development, Merrimack Pharmaceuticals

Despite improvements in treatment, HER2-positive metastatic breast cancer remains a life-threatening disease. MM-302 is a HER2-targeted PEGylated liposome that encapsulates doxorubicin to facilitate its specific delivery to HER2-overexpressing tumors. Preclinical studies revealed synergistic antitumor activity for MM-302 in combination with trastuzumab. Phase 1 clinical investigation showed an acceptable safety profile and indicated promising clinical activity. MM-302 is currently under investigation in a randomized phase 2 clinical trial (HERMIONE).

3:15 Developing Site-Specifically Modified ADCs Using a Chemoenzymatic Approach

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

Antibody-drug conjugates (ADCs) have become de rigueur for pharmaceutical oncology drug development pipelines. We have developed the SMARTagTM technology platform, which enables precise, programmable, site-selective chemical protein modification. We will highlight progress in developing these SMARTagTM ADCs with a focus on preclinical studies as well as highlight our progress in cell line development and manufacturing of bioconjugates using this chemoenzymatic approach.

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

4:45 Problem-Solving Breakout Discussions

  • The pros and cons of large and small molecular weight ligand-targeted drug conjugates
  • Philip S. Low, Ph.D., Director of the Purdue Center for Drug Discovery, Ralph C. Corley Distinguished Professor, Department of Chemistry, Purdue University

    • The large size of ADCs assures a large AUC but limits tumor penetration. Which property is most important for therapeutic efficacy? Is there an optimal ligand size that yields the best AUC and tumor penetration?
    • Is AUC important when the number of targeted receptors is limited? How does receptor recycling rate impact this answer?
    • Is there a relationship between targeted receptor number and the potency of the targeted cytotoxic drug required for therapeutic efficacy?
    • How can one design a linker in a ligand-targeted drug conjugate that is stable in circulation and cleavable upon endocytosis into the diseased cell?
  • ADCs : Past, Present and Future
  • Peter Hofland, Ph.D., Managing Partner, Publisher and Executive Editor, ADC Review

    • Past: Coining the turn ‘magic bullet; the German physician and scientist Paul Ehrlich proposed, more than a century ago, the concept of selectively delivering a cytotoxic drug to a tumor via a targeting agent. Ehrlich’s concept of targeted therapy was first exemplified when methotrexate (MTX) was linked to an antibody targeting leukemia cells. In 1975, the landmark development of mouse mAbs using hybridoma technology by Kohler and Milstein greatly advanced the field of ADC. The first ‘real’ ADC, gemtuzumab ozogamicin (Mylotarg) was approved in 2000.  However, despite initially encouraging clinical resuls, the drug was withdrawn from the market in 2010 owing to a lack of improvement in overall survival. 
    • Present: Today there are two approved ADC: brentuximab vedotin (Adcetris; Seattle Genetics) and ado-trastuzumab emtansine (Kadcyla; Genentech/Roche). With nearly 50 in clinical trials, and an average clinical development time of ~6 years for ADCs, the expectation is that in the next 3 – 4 years the number of new, approved, ADCs will increase dramatically. But what are some of the hurdles to be expected in bringing novel ADCs to the clinic? ADCs in clinical development are directed against a range of different antigenic targets. However, there is a limited number of cytotoxic drugs, including calicheamicin, auristatins, maytansinoids, duocarmycins and pyrolidobenzodiazepines (PBDs) confirming the difficulties of finding fitting cytotoxic drugs as payloads in ADCs. How do these highly potent agents support an average of drug-to-antibody ratio (DAR) of 2 to 4? Are they not too hydrophobic? Are they linkable? Are they accessible by simple synthetic pathways? Manufacturable? What about a case for moderate toxic payloads? What about the sensitivity of cytotoxic agents to multidrug resistance (MDR) mechanisms?
    • Future: Progress in site-specific conjugation modalities, optimization of linkers with balanced stability and identification of novel, potent cytotoxic agents are expected to pave the way for a better understanding of factors such as ADC efficacy, PK and safety.  What are the complexities? A robust clinical pipeline, evolving clinical data, technological advancements and a better understanding of the biology of cancer and hematological malignancies, may help in the development of novel ADCs. What are the challenges? What are your expectations?

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

    7:00 End of Day

    THURSDAY, APRIL 28

    8:00 am Morning Coffee


    ENGINEERING AND CONJUGATION CHEMISTRY TO IMPROVE PROPERTIES OF ADCs

    8:30 Chairperson’s Remarks

    Greg Adams, Ph.D., Chief Development Officer, Viventia Biotech

    8:35 Engineering a Tumor-Specific, Next-Generation Anti-EGFR ADC Development Candidate

    Chris_ThanosChristopher D. Thanos, Ph.D., Senior Director, Biotherapeutics Discovery, Halozyme Therapeutics, Inc.

    9:05 Selected Poster Presentation I:

    Site-Specific Antibody Drug Conjugates Using a Unique Lysine Residue

    Alex Nanna, Graduate Student, Cancer Biology, Scripps Research Institute

    Current strategies to produce site-specific ADCs mostly rely on mutations or non-selective conjugation chemistry. Here we present a novel strategy to produce site-specific ADCs using a chemically programmable antibody. This approach is mutation-free and drug conjugation proceeds rapidly at neutral pH in only 2 hours. Extensive optimization was required combining synthetic chemistry and antibody engineering to product potent ADCs targeting human epidermal growth factor receptor 2 (HER2), folate receptor 1 (FOLR1) and CD138.

    9:20 Selected Poster Presentation II: 

    Antibody-Drug Conjugates Targeting Sialyl-Tn (STn) Demonstrate in vitro and in vivo Anti-Tumor Efficacy

    Jillian_PrendergastJillian Prendergast, Ph.D., Scientist I, R&D, Siamab Therapeutics

    Targeted therapeutics today must differentiate between normal and malignant tumor cells. At Siamab Therapeutics, we have focused on a target - Sialyl-Tn (STn) - that shows marked differences in expression between normal and cancer tissues; its presence in tumors is also associated with poor prognosis and reduced survival. Clinical approaches to targeting STn have thus far consisted of an STn vaccine (Theratope), which failed in advanced clinical trials due to poor efficacy, but demonstrated strong safety with STn as a target. Early generation commercial mouse antibodies have targeted STn, but have poor specificity, binding additional glycans or proteins. Siamab Therapeutics has addressed these concerns by developing a highly specific, high affinity anti-STn mAb panel. Lead candidates demonstrate single digit nanomolar EC50s in ELISA/flow cytometry assays, target selective cell internalization, and STn specific glycan binding on Siamab's proprietary glycan array. Tumor microarray experiments revealed a desirable staining pattern and frequency across several cancer indications with minimal to no staining of normal tissues. We have formatted these lead anti-STn mAbs as antibody drug conjugates (ADCs) and they demonstrate target specific cell killing in vitro with single digit IC50s. A multiple dose ICR SCID subcutaneous xenograft in vivo cancer model demonstrated statistically significant tumor growth inhibition (%T/C) ranging from 3.0 - 3.6% of these anti-STn ADCs compared to vehicle alone. Humanization was recently completed for these lead mAbs, and target specificity, affinity and cytotoxicity were maintained with additional validation underway. Our data demonstrates that high-affinity, STn-specific mAbs show promise as therapeutics for solid tumors.


    9:35 Delivering the Future of Oncology – Manufacturing Next Generation Bioconjugates & Combination Therapies

    Rohrer_ThomasThomas Rohrer, Senior Manager, Antibody Drug Conjugation Project Evaluation, Lonza

    • A brief history of oncology treatments and bioconjugates
    • Lessons learned and challenges associated with ADCs
    • What does the future look like for ADCs? Novel conjugation technologies, cytotoxic drugs, and different linker chemistries
    • How the experiences from one manufacturing technology may be applied to the future of oncology

     

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

    11:05 A Plug-and-Play Approach to Antibody-Based Therapeutics via a Chemoselective “Dual Click” Strategy

    Vijay_ChudasamaVijay Chudasama, Ph.D., MSc, Lecturer, Chemistry, University College London

    There is a clear demand for the construction of novel antibody-drug conjugate (ADC) platforms that offer greater stability, homogeneity and flexibility. A significant step towards the ideal platform for next generation antibody-based therapeutics is presented. Our technology provides decorated antibody constructs that are highly stable, with complete retention of antibody binding/structure post-modification. It combines site-specific functionalisation with exceptional versatility via the functional re-bridging of interchain disulfide bonds native to antibodies.

    11:35 Improving the Therapeutic Window of an Antibody-Drug Conjugate by a New Stable Conjugation Method

    Jinwong_JungJinwon Jung, Ph.D., Principal Scientist, R&D Center, ABL BIO

    To overcome the limitation of ADC due to off-target toxicity and narrow therapeutic window, we have developed s new conjugation technique that attaches drugs to N-terminal of an antibody through amine bond by reductive alkylation reaction (NTERM). NTERM ADC showed superior in vitro and in vivo stability as well as tolerability than commonly used thiol-conjugate and lysine conjugate. Therefore, NTERM can be a novel conjugation method to improve therapeutic window.

    12:05 pm A Non-Genetic Approach to ADCs with Improved Therapeutic Index with GlycoConnect™ and HydraSpace™ Technology

    Floris_vanDelftFloris van Delft, Ph.D., Co-Founder and CSO, Synaffix

    GlycoConnect™ is a highly efficient technology to obtain, without protein engineering, antibody-drug conjugates (ADCs) in a two-stage process involving (a) enzymatic N-glycan remodeling and (b) copper-free click attachment of payload. Conjugation of highly hydrophobic payloads is accommodated with polar HydraSpace™ technology. Excellent in vivo efficacy and high tolerability is demonstrated, thus paving the way for the next generation of ADCs with an improved therapeutic index.

    12:35 End of Antibody-Drug Conjugates I: New Ligands, Payloads and Alternative Formats

    5:15 Registration for Dinner Short Courses


    Recommended Dinner Short Course*

    SC13: Critical Considerations for the Design and Development of Antibody-Drug Conjugates

    *Separate registration required.


    Register Now

    View By:


    Premier Sponsors

       FairJourneyBiologics Integral-Molecular_NEWLONZAOmniAb  Samsung_Biologics  UnchainedLabs