1
|
Lopez JS, Milhem M, Butler MO, Thistlethwaite F, Van Tine BA, D'Angelo SP, Johnson ML, Sato T, Arkenau HT, Edukulla R, Wustner J, Marshall S, Rodon J. Phase 1 study of IMCnyeso, a T cell receptor bispecific ImmTAC targeting NY-ESO-1-expressing malignancies. Cell Rep Med 2025; 6:101994. [PMID: 40054461 DOI: 10.1016/j.xcrm.2025.101994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/07/2024] [Accepted: 02/07/2025] [Indexed: 04/18/2025]
Abstract
IMCnyeso, an immune mobilizing monoclonal T cell receptor against cancer (ImmTAC) bispecific (New York esophageal squamous cell carcinoma [NY-ESO]×CD3) T cell engager, targets an NY-ESO-1/L-antigen family member-1 isoform A (LAGE-1A) peptide presented by histocompatibility leukocyte antigen (HLA)-A∗02:01. In this phase 1 study, 28 HLA-A∗02:01+ patients with advanced NY-ESO-1/LAGE-1A-positive advanced tumors (n = 28) receive IMCnyeso weekly intravenously (dose range: 3-300 μg; 7 dose-escalation cohorts). The primary objective is to identify the maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D); additional objectives include preliminary anti-tumor activity, pharmacokinetics, immunogenicity, and pharmacodynamic changes. The study was terminated before fully enrolling dose escalation, and the MTD was not identified. There are no treatment-related discontinuations or deaths. The most common adverse events are grade 1/2 cytokine release syndrome and associated symptoms. Cytokine induction and transient lymphocyte count decreases are observed at doses 30-300 μg. At these doses, preliminary efficacy includes mixed response (2 patients) and a median overall survival of 12 months. IMCnyeso is well tolerated and, at doses ≥30 μg, induces pharmacodynamic changes consistent with T cell redirection. This study was registered at ClinicalTrials.gov (NCT03515551).
Collapse
Affiliation(s)
- Juanita S Lopez
- Drug Development Unit, Institute of Cancer Research and the Royal Marsden Hospital, London SW7 3RP, UK.
| | - Mohammed Milhem
- Department of Internal Medicine, Division of Hematology/Oncology and BMT, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Marcus O Butler
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto M5G 2M9, ON, Canada; Departments of Medicine and Immunology, University of Toronto, Toronto M5G 2M9, ON, Canada
| | - Fiona Thistlethwaite
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M20 4GJ, UK; Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Brian A Van Tine
- Division of Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Sandra P D'Angelo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Melissa L Johnson
- Lung Cancer Research Program, Sarah Cannon Research Institute at Tennessee Oncology, Nashville, TN 37203, USA
| | - Takami Sato
- Department of Medical Oncology, Sidney Kimmel Comprehensive Cancer Center, Jefferson University, Philadelphia, PA 19107, USA
| | - Hendrik-Tobias Arkenau
- Phase-1 Trials Unit, Sarah Cannon Research Institute, UCL Cancer Institute, University College London, London W1G 6AD, UK
| | | | - Jason Wustner
- Translational Medicine, Immunocore, Conshohocken, PA 19428 USA
| | | | - Jordi Rodon
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston 77030, TX, USA
| |
Collapse
|
2
|
Lefèvre A, Parra-Guillen ZP, Trocóniz IF, Boetsch C, Frances N. Mechanistic PKPD modeling to describe cytokine release associated with CD3 T-cell engager therapies. Front Immunol 2025; 15:1463915. [PMID: 39896804 PMCID: PMC11782561 DOI: 10.3389/fimmu.2024.1463915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 12/19/2024] [Indexed: 02/04/2025] Open
Abstract
Introduction T-cell engagers (TCE), a therapeutic class of cancer immunotherapy (CIT), offer a novel approach to cancer treatment by harnessing and reactivating the patient's immune system to eradicate tumor cells. However, the use of TCE in the clinic can lead to severe side effects, including cytokine release syndrome (CRS). Therefore, innovative dosing strategies need to be implemented to mitigate the risk of developing CRS. Method In the presented work, a mechanistic pharmacokinetics/pharmacodynamics (PKPD) model describing cytokine release following TCE therapy has been developed combining literature knowledge and preclinical data. The model was developed to explore and test hypotheses regarding the mechanisms behind the decrease of cytokine release following two repeated TCE administrations. Results The model is able to successfully reproduce the observed dynamics of cytokine levels associated with the initial and subsequent TCE doses, accounting for different dosing intervals. In addition, the model suggests a mechanism of action that uncouples cytokine release from tumor cell killing. Discussion This model provides an initial mechanistic framework to support the design of experiments and paves the way for the application of mathematical modeling to support clinical dosing regimen selection of any TCE.
Collapse
Affiliation(s)
- Apolline Lefèvre
- Roche Pharma Research and Early Development (pRED), Pharmaceutical Sciences PS, Roche Innovation Center Basel, Basel, Switzerland
- Pharmacometrics & Systems Pharmacology, Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Zinnia P. Parra-Guillen
- Pharmacometrics & Systems Pharmacology, Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Iñaki F. Trocóniz
- Pharmacometrics & Systems Pharmacology, Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Institute of Data Science and Artificial Intelligence (DATAI), University of Navarra, Pamplona, Spain
| | - Christophe Boetsch
- Roche Pharma Research and Early Development (pRED), Pharmaceutical Sciences PS, Roche Innovation Center Basel, Basel, Switzerland
| | - Nicolas Frances
- Roche Pharma Research and Early Development (pRED), Pharmaceutical Sciences PS, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
3
|
Zhou D, Kischel R, Stienen S, Townsley D, Sternjak A, Lutteropp M, Bailis J, Friedrich M, Rattel B, Mehta K, Upreti VV. An Innovative Approach to Optimize First-In-Human Minimal Anticipated Biological Effect Level Based Starting Dose in Oncology Trials for Bispecific T-Cell Engagers: Experience from A Solid Tumor Bispecific T-Cell Engager. Clin Pharmacol Ther 2025; 117:225-231. [PMID: 39285508 DOI: 10.1002/cpt.3431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 08/17/2024] [Indexed: 12/19/2024]
Abstract
Bispecific T-cell engagers (Bi-TCEs) have revolutionized the treatment and management of both hematological and solid tumor indications with opportunities to become best-in-class therapeutics for cancer. However, defining the dose and dosing regimen for the first-in-human (FIH) studies of Bi-TCEs can be challenging, as a high starting dose can expose subjects to serious toxicity while a low starting dose based on traditional minimal anticipated biological effect level (MABEL) approach could lead to lengthy dose escalations that exposes seriously ill patients to sub-therapeutic dosing. Leveraging our in-depth and broad clinical development experience across three generations of Bi-TCEs across both liquid and solid tumor indications, we developed an innovative modified MABEL approach for starting dose selection that integrates knowledge based on the target biology, indication, toxicology, in vitro, in vivo pharmacological evaluations, and translational pharmacokinetic/pharmacodynamic (PK/PD) modeling, together with anticipated safety profile. Compared to the traditional MABEL approach in which high effector to target (E:T) cell ratios are typically used, our innovative approach utilized an optimized E:T cell ratio that better reflects the tumor microenvironment. This modified MABEL approach was successfully applied to FIH dose selection for a half-life extended (HLE) Bi-TCE for gastric cancer. This modified MABEL approach enabled a 10-fold higher starting dose that was deemed safe and well tolerated and saved at least two dose-escalation cohorts before reaching the projected efficacious dose. This approach was successfully accepted by global regulatory agencies and can be applied for Bi-TCEs across both hematological and solid tumor indications for accelerating the clinical development for Bi-TCEs.
Collapse
Affiliation(s)
- Di Zhou
- Clinical Pharmacology Modeling & Simulation, Amgen Inc., South San Francisco, California, USA
| | | | | | - Danielle Townsley
- Oncology Early Development, Amgen Inc., Thousand Oaks, California, USA
| | | | | | - Julie Bailis
- Oncology Research, Amgen Inc., South San Francisco, California, USA
| | | | | | - Khamir Mehta
- Clinical Pharmacology Modeling & Simulation, Amgen Inc., South San Francisco, California, USA
| | - Vijay V Upreti
- Clinical Pharmacology Modeling & Simulation, Amgen Inc., South San Francisco, California, USA
| |
Collapse
|
4
|
van der Graaf PH. First-in-Human Dose Selection for T-Cell Engaging Bispecific Antibodies. Clin Pharmacol Ther 2025; 117:11-13. [PMID: 39690707 DOI: 10.1002/cpt.3487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 12/19/2024]
Affiliation(s)
- Piet H van der Graaf
- Certara, Applied BioSimulation, Canterbury, UK
- Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| |
Collapse
|
5
|
Nagaraja Shastri P, Shah N, Lechmann M, Mody H, Retter MW, Zhu M, Li T, Wang J, Shaik N, Zheng X, Ovacik M, Hua F, Jawa V, Boetsch C, Cao Y, Burke J, Datta K, Gadkar K, Upreti V, Betts A. Industry Perspective on First-in-Human and Clinical Pharmacology Strategies to Support Clinical Development of T-Cell Engaging Bispecific Antibodies for Cancer Therapy. Clin Pharmacol Ther 2025; 117:34-55. [PMID: 39295563 DOI: 10.1002/cpt.3439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/25/2024] [Indexed: 09/21/2024]
Abstract
T-cell-engaging bispecific antibodies (TCEs) that target tumor antigens and T cells have shown great promise in treating cancer, particularly in hematological indications. The clinical development of TCEs often involves a lengthy first-in-human (FIH) trial with many dose-escalation cohorts leading up to an early proof of concept (POC), enabling either a no-go decision or dose selection for further clinical development. Multiple factors related to the target, product, disease, and patient population influence the efficacy and safety of TCEs. The intricate mechanism of action limits the translatability of preclinical models to the clinic, thereby posing challenges to streamline clinical development. In addition, unlike traditional chemotherapy, the top dose and recommended phase II doses (RP2Ds) for TCEs in the clinic are often not guided by the maximum tolerated dose (MTD), but rather based on the integrated dose-response assessment of the benefit/risk profile. These uncertainties pose complex challenges for translational and clinical pharmacologists (PK/PD scientists), as well as clinicians, to design an efficient clinical study that guides development. To that end, experts in the field, under the umbrella of the American Association of Pharmaceutical Scientists, have reviewed learnings from published literature and currently marketed products to share perspectives on the FIH and clinical pharmacology strategies to support early clinical development of TCEs.
Collapse
Affiliation(s)
- Prathap Nagaraja Shastri
- Clinical Pharmacology and Pharmacometrics, Johnson and Johnson Innovative Medicine, Spring House, Pennsylvania, USA
| | - Nirav Shah
- Clinical Pharmacology and Pharmacometrics, Johnson and Johnson Innovative Medicine, Spring House, Pennsylvania, USA
| | - Martin Lechmann
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Penzberg, Germany
| | - Hardik Mody
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - Marc W Retter
- Preclinical PK/PD, Regeneron Pharmaceuticals, Tarrytown, New York, USA
| | - Min Zhu
- Clinical Pharmacology, Regeneron Pharmaceuticals, Tarrytown, New York, USA
| | - Tommy Li
- Clinical Pharmacology, Genmab, Plainsboro, New Jersey, USA
| | - Jun Wang
- Biotherapeutics Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut, USA
| | - Naveed Shaik
- Clinical Pharmacology, Pfizer Oncology Development, San Diego, California, USA
| | - Xirong Zheng
- Clinical Pharmacology, Pharmacometrics and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Meric Ovacik
- Preclinical PK/PD, Genentech, South San Francisco, California, USA
| | - Fei Hua
- Certara Predictive Technology, Certara, Concord, Massachusetts, USA
| | - Vibha Jawa
- Clinical Pharmacology, Pharmacometrics and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Christophe Boetsch
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - John Burke
- Certara Predictive Technology, Certara, Concord, Massachusetts, USA
| | - Kaushik Datta
- Nonclinical Safety, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Kapil Gadkar
- Preclinical PK/PD, Genentech, South San Francisco, California, USA
| | - Vijay Upreti
- Clinical Pharmacology, Modeling & Simulation, Amgen, South San Francisco, California, USA
| | - Alison Betts
- Preclinical & Translational Sciences, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, USA
| |
Collapse
|
6
|
Zhu M, Madia P, Crawford A, Brouwer‐Visser J, Krueger P, Haber L, Peterman M, Uldrick TS, Miller E, Davis JD, Retter MW. Translational findings support regimen selection for first-in-human study of ubamatamab (MUC16 × CD3 bispecific antibody) in patients with recurrent ovarian cancer. Clin Transl Sci 2024; 17:e70082. [PMID: 39652449 PMCID: PMC11627180 DOI: 10.1111/cts.70082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/21/2024] [Accepted: 11/02/2024] [Indexed: 12/12/2024] Open
Abstract
Ubamatamab, a Mucin 16 (MUC16) × cluster of differentiation 3 (CD3) bispecific antibody that promotes T-cell-mediated cytotoxicity of MUC16-expressing cells, is being investigated for the treatment of ovarian cancer. Intravenous administration of ubamatamab, with or without the anti-programmed cell death-1 inhibitor cemiplimab, is being evaluated in a first-in-human study in patients with recurrent ovarian cancer. In vitro cytotoxicity and cytokine data and projected ubamatamab human pharmacokinetic (PK) profiles scaled with monkey PK parameters enabled starting-dose selection in humans. Mouse tumor regression studies identified ubamatamab effective concentrations. Preclinical and clinical PK, cytokine, safety, and efficacy data from dose escalation were integrated to determine expansion regimens. A starting dose of 0.1 mg was selected, which showed acceptable safety in patients. A step-up dosing approach was used to effectively manage cytokine release syndrome. Mouse tumor regression models suggested an ubamatamab efficacious concentration range of 0.4-50 mg/L, consistent with clinical activity observed at ubamatamab trough concentrations ≥5 mg/L. Integrating preclinical and clinical data determined a target trough concentration range of 5-30 mg/L, which supports evaluation of ubamatamab 250 mg with or without cemiplimab and 800 mg monotherapy once every 3 weeks in expansion cohorts. Preclinical data (cytokine release, tumor regression, monkey PK) had translational value in supporting regimen selection in dose escalation and subsequently in dose expansion after integration with patient data from dose escalation.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Mice
- Middle Aged
- Antibodies, Bispecific/pharmacokinetics
- Antibodies, Bispecific/administration & dosage
- Antibodies, Bispecific/adverse effects
- Antibodies, Bispecific/immunology
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- CA-125 Antigen/immunology
- CA-125 Antigen/blood
- CD3 Complex/immunology
- CD3 Complex/antagonists & inhibitors
- Dose-Response Relationship, Drug
- Macaca fascicularis
- Membrane Proteins/immunology
- Membrane Proteins/antagonists & inhibitors
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/immunology
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/immunology
- Translational Research, Biomedical
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Min Zhu
- Regeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | | | | | | | | | - Lauric Haber
- Regeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | | | | | | | | | | |
Collapse
|
7
|
Nolan-Stevaux O, Smith R. Logic-gated and contextual control of immunotherapy for solid tumors: contrasting multi-specific T cell engagers and CAR-T cell therapies. Front Immunol 2024; 15:1490911. [PMID: 39606234 PMCID: PMC11599190 DOI: 10.3389/fimmu.2024.1490911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
CAR-T cell and T cell engager therapies have demonstrated transformational efficacy against hematological malignancies, but achieving efficacy in solid tumors has been more challenging, in large part because of on-target/off-tumor toxicities and sub-optimal T cell anti-tumor cytotoxic functions. Here, we discuss engineering solutions that exploit biological properties of solid tumors to overcome these challenges. Using logic gates as a framework, we categorize the numerous approaches that leverage two inputs instead of one to achieve better cancer selectivity or efficacy in solid tumors with dual-input CAR-Ts or multi-specific TCEs. In addition to the "OR gate" and "AND gate" approaches that leverage dual tumor antigen targeting, we also review "contextual AND gate" technologies whereby continuous cancer-selective inputs such a pH, hypoxia, target density, tumor proteases, and immune-suppressive cytokine gradients can be creatively incorporated in therapy designs. We also introduce the notion of "output directionality" to distinguish dual-input strategies that mechanistically impact cancer cell killing or T cell fitness. Finally, we contrast the feasibility and potential benefits of the various approaches using CAR-T and TCE therapeutics and discuss why the promising "IF/THEN" and "NOT" gate types pertain more specifically to CAR-T therapies, but can also succeed by integrating both technologies.
Collapse
Affiliation(s)
| | - Richard Smith
- Cell Biology Research, Kite Pharma, Foster City, CA, United States
| |
Collapse
|
8
|
Ruuls SR, Parren PWHI. Antibody avidity meets multiple myeloma. NATURE CANCER 2024; 5:1452-1454. [PMID: 39261677 DOI: 10.1038/s43018-024-00805-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Affiliation(s)
| | - Paul W H I Parren
- Gyes, Utrecht, the Netherlands.
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
9
|
Carretero-Iglesia L, Hall OJ, Berret J, Pais D, Estoppey C, Chimen M, Monney T, Loyau J, Dreyfus C, Macoin J, Perez C, Menon V, Gruber I, Laurendon A, Caro LN, Gudi GS, Matsuura T, van der Graaf PH, Blein S, Mbow ML, Croasdale-Wood R, Srivastava A, Dyson MR, Matthes T, Kaya Z, Edwards CM, Edwards JR, Maiga S, Pellat-Deceunynck C, Touzeau C, Moreau P, Konto C, Drake A, Zhukovsky EA, Perro M, Pihlgren M. ISB 2001 trispecific T cell engager shows strong tumor cytotoxicity and overcomes immune escape mechanisms of multiple myeloma cells. NATURE CANCER 2024; 5:1494-1514. [PMID: 39261676 PMCID: PMC11505469 DOI: 10.1038/s43018-024-00821-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 08/07/2024] [Indexed: 09/13/2024]
Abstract
Despite recent advances in immunotherapies targeting single tumor-associated antigens, patients with multiple myeloma eventually relapse. ISB 2001 is a CD3+ T cell engager (TCE) co-targeting BCMA and CD38 designed to improve cytotoxicity against multiple myeloma. Targeting of two tumor-associated antigens by a single TCE resulted in superior cytotoxic potency across a variable range of BCMA and CD38 tumor expression profiles mimicking natural tumor heterogeneity, improved resistance to competing soluble factors and exhibited superior cytotoxic potency on patient-derived samples and in mouse models. Despite the broad expression of CD38 across human tissues, ISB 2001 demonstrated a reduced T cell activation profile in the absence of tumor cells when compared to TCEs targeting CD38 only. To determine an optimal first-in-human dose for the ongoing clinical trial ( NCT05862012 ), we developed an innovative quantitative systems pharmacology model leveraging preclinical data, using a minimum pharmacologically active dose approach, therefore reducing patient exposure to subefficacious doses of therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Vinu Menon
- Ichnos Glenmark Innovation, New York, NY, USA
| | | | | | | | | | - Tomomi Matsuura
- Certara UK Limited, Canterbury Innovation Centre, University Road, Canterbury, United Kingdom
| | - Piet H van der Graaf
- Certara UK Limited, Canterbury Innovation Centre, University Road, Canterbury, United Kingdom
| | | | | | | | | | | | - Thomas Matthes
- Hematology Service, Department of Oncology and Clinical Pathology Service, Department of Diagnostics, University Hospital Geneva, Geneva, Switzerland
| | - Zeynep Kaya
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Institute, University of Oxford, Oxford, United Kingdom
| | - Claire M Edwards
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Institute, University of Oxford, Oxford, United Kingdom
| | - James R Edwards
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Institute, University of Oxford, Oxford, United Kingdom
| | - Sophie Maiga
- Nantes Université, Inserm, CNRS, Université d'Angers, Nantes, France
- SIRIC ILIAD, Angers, Nantes, France
| | | | - Cyrille Touzeau
- Nantes Université, Inserm, CNRS, Université d'Angers, Nantes, France
- SIRIC ILIAD, Angers, Nantes, France
- Service d'Hématologie Clinique, Unité d'Investigation Clinique, CHU, Nantes, France
| | - Philippe Moreau
- Nantes Université, Inserm, CNRS, Université d'Angers, Nantes, France
- SIRIC ILIAD, Angers, Nantes, France
- Service d'Hématologie Clinique, Unité d'Investigation Clinique, CHU, Nantes, France
| | - Cyril Konto
- Ichnos Glenmark Innovation, New York, NY, USA
| | - Adam Drake
- Ichnos Glenmark Innovation, New York, NY, USA
| | | | - Mario Perro
- Ichnos Glenmark Innovation, New York, NY, USA.
| | | |
Collapse
|
10
|
Elmeliegy M, Chen J, Dontabhaktuni A, Gaudy A, Kapitanov GI, Li J, Mim SR, Sharma S, Sun Q, Ait-Oudhia S. Dosing Strategies and Quantitative Clinical Pharmacology for Bispecific T-Cell Engagers Development in Oncology. Clin Pharmacol Ther 2024; 116:637-646. [PMID: 38962850 DOI: 10.1002/cpt.3361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/09/2024] [Indexed: 07/05/2024]
Abstract
Bispecific T-cell Engagers (TCEs) are promising anti-cancer treatments that bind to both the CD3 receptors on T cells and an antigen on the surface of tumor cells, creating an immune synapse, leading to killing of malignant tumor cells. These novel therapies have unique development challenges, with specific safety risks of cytokine release syndrome. These on-target adverse events fortunately can be mitigated and deconvoluted from efficacy via innovative dosing strategies, making clinical pharmacology key in the development of these therapies. This review assesses dose selection and the role of quantitative clinical pharmacology in the development of the first eight approved TCEs. Model informed drug development (MIDD) strategies can be used at every stage to guide TCE development. Mechanistic modeling approaches allow for (1) efficacious yet safe first-in-human dose selection as compared with in vitro minimum anticipated biological effect level (MABEL) approach; (2) rapid escalation and reducing number of patients with subtherapeutic doses through model-based adaptive design; (3) virtual testing of different step-up dosing regimens that may not be feasible to be evaluated in the clinic; and (4) selection and justification of the optimal clinical step-up and full treatment doses. As the knowledge base around TCEs continues to grow, the relevance and utilization of MIDD strategies for supporting the development and dose optimization of these molecules are expected to advance, optimizing the benefit-risk profile for cancer patients.
Collapse
Affiliation(s)
- Mohamed Elmeliegy
- Oncology Research and Development, Pfizer Inc, San Diego, California, USA
| | - Joseph Chen
- Genentech Inc, South San Francisco, California, USA
| | | | | | | | - Junyi Li
- Genentech Inc, South San Francisco, California, USA
| | - Sabiha R Mim
- PharmaPro Consulting Inc, Hillsborough, New Jersey, USA
| | - Sharad Sharma
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Qin Sun
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Sihem Ait-Oudhia
- Quantitative Pharmacology and Pharmacometrics (QP2), Merck & Co., Rahway, New Jersey, USA
| |
Collapse
|
11
|
Matsumoto M, Polli JR, Swaminathan SK, Datta K, Kamperschroer C, Fortin MC, Salian-Mehta S, Dave R, Yang Z, Arora P, Hiura M, Suzuki M, Brennan FR, Sathish J. Beyond MABEL: An Integrative Approach to First in Human Dose Selection of Immunomodulators by the Health and Environmental Sciences Institute (HESI) Immuno-Safety Technical Committee (ITC). Clin Pharmacol Ther 2024; 116:546-562. [PMID: 38847597 DOI: 10.1002/cpt.3316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/13/2024] [Indexed: 08/22/2024]
Abstract
Administration of a new drug candidate in a first-in-human (FIH) clinical trial is a particularly challenging phase in drug development and is especially true for immunomodulators, which are a diverse and complex class of drugs with a broad range of mechanisms of action and associated safety risks. Risk is generally greater for immunostimulators, in which safety concerns are associated with acute toxicity, compared to immunosuppressors, where the risks are related to chronic effects. Current methodologies for FIH dose selection for immunostimulators are focused primarily on identifying the minimum anticipated biological effect level (MABEL), which has often resulted in sub-therapeutic doses, leading to long and costly escalation phases. The Health and Environmental Sciences Institute (HESI) - Immuno-Safety Technical Committee (ITC) organized a project to address this issue through two complementary approaches: (i) an industry survey on FIH dose selection strategies and (ii) detailed case studies for immunomodulators in oncology and non-oncology indications. Key messages from the industry survey responses highlighted a preference toward more dynamic PK/PD approaches as in vitro assays are seemingly not representative of true physiological conditions for immunomodulators. These principles are highlighted in case studies. To address the above themes, we have proposed a revised decision tree, which expands on the guidance by the IQ MABEL Working Group (Leach et al. 2021). This approach facilitates a more refined recommendation of FIH dose selection for immunomodulators, allowing for a nuanced consideration of their mechanisms of action (MOAs) and the associated risk-to-benefit ratio, among other factors.
Collapse
Affiliation(s)
- Mineo Matsumoto
- Review Division, Pharmaceuticals and Medical Devices Agency (PMDA), Chiyoda-ku, Tokyo, Japan
| | - Joseph Ryan Polli
- Pharmacokinetic Sciences, Translational Medicine, Novartis Biomedical Research, Cambridge, Massachusetts, USA
| | | | - Kaushik Datta
- Nonclinical Safety, Bristol-Myers Squibb, New Brunswick, New Jersey, USA
| | | | - Marie C Fortin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, New Jersey, USA
| | - Smita Salian-Mehta
- Nonclinical Safety and Pathobiology, Gilead Sciences, Foster City, California, USA
| | - Rutwij Dave
- Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California, USA
| | - Zheng Yang
- Drug Metabolism and Pharmacokinetics, Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Payal Arora
- Translational Research, Kyowa Kirin Pharmaceuticals, Princeton, New Jersey, USA
| | - Masanori Hiura
- Translational Research, Kyowa Kirin, Sunto-gun, Shizuoka, Japan
| | - Mizuho Suzuki
- Review Division, Pharmaceuticals and Medical Devices Agency (PMDA), Chiyoda-ku, Tokyo, Japan
| | | | - Jean Sathish
- Comparative Medicine and Drug Safety R&D, Pfizer, Pearl River, New York, USA
| |
Collapse
|
12
|
Liao X, Qi T, Zhou J, Liu C, Cao Y. Optimizing Clinical Translation of Bispecific T-cell Engagers through Context Unification with a Quantitative Systems Pharmacology Model. Clin Pharmacol Ther 2024; 116:415-425. [PMID: 38751031 PMCID: PMC11251864 DOI: 10.1002/cpt.3302] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/30/2024] [Indexed: 07/17/2024]
Abstract
Bispecific T-cell engagers (bsTCEs) have emerged as a promising class of cancer immunotherapy. BsTCEs enable physical connections between T cells and tumor cells to enhance T-cell activity against cancer. Despite several marketing approvals, the development of bsTCEs remains challenging, especially at early clinical translational stages. The intricate design of bsTCEs makes their pharmacologic effects and safety profiles highly dependent on patient's immunological and tumor conditions. Such context-dependent pharmacology introduces considerable uncertainty into translational efforts. In this study, we developed a Quantitative Systems Pharmacology (QSP) model, through context unification, that can facilitate the translation of bsTCEs preclinical data into clinical activity. Through characterizing the formation dynamics of immunological synapse (IS) induced by bsTCEs, this model unifies a broad range of contexts related to target affinity, tumor characteristics, and immunological conditions. After rigorous calibration using both experimental and clinical data, the model enables consistent translation of drug potency observed under diverse experimental conditions into predictable exposure-response relationships in patients. Moreover, the model can help identify optimal target-binding affinities and minimum efficacious concentrations across different clinical contexts. This QSP approach holds significant promise for the future development of bsTCEs.
Collapse
Affiliation(s)
- Xiaozhi Liao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA
| | - Timothy Qi
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA
| | - Jiawei Zhou
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA
| | - Can Liu
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
13
|
Lim K, Zhu XS, Zhou D, Ren S, Phipps A. Clinical Pharmacology Strategies for Bispecific Antibody Development: Learnings from FDA-Approved Bispecific Antibodies in Oncology. Clin Pharmacol Ther 2024; 116:315-327. [PMID: 38825990 DOI: 10.1002/cpt.3308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/03/2024] [Indexed: 06/04/2024]
Abstract
Bispecific antibodies, by enabling the targeting of more than one disease-associated antigen or engaging immune effector cells, have both advantages and challenges compared with a combination of two different biological products. As of December 2023, there are 11 U.S. Food and Drug Administration-approved BsAb products on the market. Among these, 9 have been approved for oncology indications, and 8 of these are CD3 T-cell engagers. Clinical pharmacology strategies, including dose-related strategies, are critical for bispecific antibody development. This analysis reviewed clinical studies of all approved bispecific antibodies in oncology and identified dose-related perspectives to support clinical dose optimization and regulatory approvals, particularly in the context of the Food and Drug Administration's Project Optimus: (1) starting doses and dose ranges in first-in-human studies; (2) dose strategies including step-up doses or full doses for recommended phase 2 doses or dose level(s) used for registrational intent; (3) restarting therapy after dose delay; (4) considerations for the introduction of subcutaneous doses; (5) body weight vs. flat dosing strategy; and (6) management of immunogenicity. The learnings arising from this review are intended to inform successful strategies for future bispecific antibody development.
Collapse
Affiliation(s)
- KyoungSoo Lim
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Xu Sue Zhu
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Waltham, Massachusetts, USA
| | - Diansong Zhou
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Waltham, Massachusetts, USA
| | - Song Ren
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Alex Phipps
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
14
|
Yadav R, Schubbert S, Holder PG, Chiang EY, Kiabi N, Bogaert L, Leung I, Rashid R, Avery KN, Bonzon C, Desjarlais JR, Sanjabi S, Sharma A, Lepherd M, Shelton A, Chan P, Liu Y, Joslyn L, Hosseini I, Stefanich EG, Kamath AV, Bernett MJ, Shivva V. Translational PK/PD and the first-in-human dose selection of a PD1/IL15: an engineered recombinant targeted cytokine for cancer immunotherapy. Front Pharmacol 2024; 15:1380000. [PMID: 38887559 PMCID: PMC11181026 DOI: 10.3389/fphar.2024.1380000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/06/2024] [Indexed: 06/20/2024] Open
Abstract
Introduction Interleukin 15 (IL-15) is a potential anticancer agent and numerous engineered IL-15 agonists are currently under clinical investigation. Selective targeting of IL-15 to specific lymphocytes may enhance therapeutic effects while helping to minimize toxicities. Methods We designed and built a heterodimeric targeted cytokine (TaCk) that consists of an anti-programmed cell death 1 receptor antibody (anti-PD-1) and an engineered IL-15. This "PD1/IL15" selectively delivers IL-15 signaling to lymphocytes expressing PD-1. We then investigated the pharmacokinetic (PK) and pharmacodynamic (PD) effects of PD1/IL15 TaCk on immune cell subsets in cynomolgus monkeys after single and repeat intravenous dose administrations. We used these results to determine the first-in-human (FIH) dose and dosing frequency for early clinical trials. Results The PD1/IL15 TaCk exhibited a nonlinear multiphasic PK profile, while the untargeted isotype control TaCk, containing an anti-respiratory syncytial virus antibody (RSV/IL15), showed linear and dose proportional PK. The PD1/IL15 TaCk also displayed a considerably prolonged PK (half-life range ∼1.0-4.1 days) compared to wild-type IL-15 (half-life ∼1.1 h), which led to an enhanced cell expansion PD response. The PD was dose-dependent, durable, and selective for PD-1+ lymphocytes. Notably, the dose- and time-dependent PK was attributed to dynamic TMDD resulting from test article-induced lymphocyte expansion upon repeat administration. The recommended first-in-human (FIH) dose of PD1/IL15 TaCk is 0.003 mg/kg, determined based on a minimum anticipated biological effect level (MABEL) approach utilizing a combination of in vitro and preclinical in vivo data. Conclusion This work provides insight into the complex PK/PD relationship of PD1/IL15 TaCk in monkeys and informs the recommended starting dose and dosing frequency selection to support clinical evaluation of this novel targeted cytokine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Amy Sharma
- Genentech, Inc., South SanFrancisco, CA, United States
| | | | - Amy Shelton
- Genentech, Inc., South SanFrancisco, CA, United States
| | - Pam Chan
- Genentech, Inc., South SanFrancisco, CA, United States
| | - Yanqiu Liu
- Genentech, Inc., South SanFrancisco, CA, United States
| | - Louis Joslyn
- Genentech, Inc., South SanFrancisco, CA, United States
| | - Iraj Hosseini
- Genentech, Inc., South SanFrancisco, CA, United States
| | | | | | | | - Vittal Shivva
- Genentech, Inc., South SanFrancisco, CA, United States
| |
Collapse
|
15
|
Collinge M, Neff-LaFord H, Akella S, Fogal B, Fraser K, Jabbour J, Harper K, Maier CC, Malherbe L, Marshall N, Rao GK, Raman K, Skaggs H, Weber F, Fuller CL. Challenges and gaps in immunosafety evaluation of therapeutics: An IQ DruSafe survey. Regul Toxicol Pharmacol 2024; 150:105630. [PMID: 38642729 DOI: 10.1016/j.yrtph.2024.105630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/15/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Immunotoxicology/immunosafety science is rapidly evolving, with novel modalities and immuno-oncology among the primary drivers of new tools and technologies. The Immunosafety Working Group of IQ/DruSafe sought to better understand some of the key challenges in immunosafety evaluation, gaps in the science, and current limitations in methods and data interpretation. A survey was developed to provide a baseline understanding of the needs and challenges faced in immunosafety assessments, the tools currently being applied across the industry, and the impact of feedback received from regulatory agencies. This survey also focused on current practices and challenges in conducting the T-cell-dependent antibody response (TDAR) and the cytokine release assay (CRA). Respondents indicated that ICH S8 guidance was insufficient for the current needs of the industry portfolio of immunomodulators and novel modalities and should be updated. Other challenges/gaps identified included translation of nonclinical immunosafety assessments to the clinic, and lack of relevant nonclinical species and models in some cases. Key areas of emerging science that will add future value to immunotoxicity assessments include development of additional in vitro and microphysiological system models, as well as application of humanized mouse models. Efforts are ongoing in individual companies and consortia to address some of these gaps and emerging science.
Collapse
Affiliation(s)
| | | | - Surekha Akella
- Abbvie Biotherapeutics Inc., South San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | | - Felix Weber
- F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | | |
Collapse
|
16
|
Saber H, Thompson MD, Leighton JK. Pharmacokinetic models for first-in-human dose selection of immune-activating products in oncology. Regul Toxicol Pharmacol 2024; 149:105616. [PMID: 38561147 DOI: 10.1016/j.yrtph.2024.105616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/06/2024] [Accepted: 03/27/2024] [Indexed: 04/04/2024]
Abstract
Pharmacokinetic (PK) models are increasingly submitted to the FDA to support first-in-human (FIH) dose selection of immune-oncology products. To examine whether a simple PK modeling (SPM) using clearance for scaling was acceptable for dose estimation, FIH(SPM) doses were computed and compared to doses that were safely administered to patients. We concluded that the SPM approach is acceptable in FIH dose estimation, but the variables should be carefully selected for CD3 constructs. For CD3 constructs, use of 60 kg BWh, a clearance exponent of 0.75, and a targeted plasma concentration based on relevant and/or sensitive activity assays was an acceptable approach for FIH dose selection; use of 0.85 as the scaling factor is questionable at this time as it resulted in a FIH dose that was too close to the AHD for one product (7%). Immune activating mAbs were not sensitive to changes in the clearance exponent (0.75-0.85) or body weight (60-70 kg). For PD-1/PD-L1 mAbs, using products' in vitro EC50 in the model resulted in suboptimal FIH doses and clinical data of closely related products informed FIH dose selection. PK models submitted by sponsors were diverse in methods, assumptions, and variables, and the resulting FIH doses were not always optimal.
Collapse
Affiliation(s)
- Haleh Saber
- US Food and Drug Administration, Center for Drug Evaluation and Research, Office of Oncologic Diseases, 10903 New Hampshire Ave, Silver Spring, MD, 20903, United States.
| | - Matthew D Thompson
- US Food and Drug Administration, Center for Drug Evaluation and Research, Office of Oncologic Diseases, 10903 New Hampshire Ave, Silver Spring, MD, 20903, United States
| | - John K Leighton
- US Food and Drug Administration, Center for Drug Evaluation and Research, Office of Oncologic Diseases, 10903 New Hampshire Ave, Silver Spring, MD, 20903, United States
| |
Collapse
|
17
|
Spreafico A, Couselo EM, Irmisch A, Bessa J, Au-Yeung G, Bechter O, Svane IM, Sanmamed MF, Gambardella V, McKean M, Callahan M, Dummer R, Klein C, Umaña P, Justies N, Heil F, Fahrni L, Opolka-Hoffmann E, Waldhauer I, Bleul C, Staack RF, Karanikas V, Fowler S. Phase 1, first-in-human study of TYRP1-TCB (RO7293583), a novel TYRP1-targeting CD3 T-cell engager, in metastatic melanoma: active drug monitoring to assess the impact of immune response on drug exposure. Front Oncol 2024; 14:1346502. [PMID: 38577337 PMCID: PMC10991832 DOI: 10.3389/fonc.2024.1346502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction Although checkpoint inhibitors (CPIs) have improved outcomes for patients with metastatic melanoma, those progressing on CPIs have limited therapeutic options. To address this unmet need and overcome CPI resistance mechanisms, novel immunotherapies, such as T-cell engaging agents, are being developed. The use of these agents has sometimes been limited by the immune response mounted against them in the form of anti-drug antibodies (ADAs), which is challenging to predict preclinically and can lead to neutralization of the drug and loss of efficacy. Methods TYRP1-TCB (RO7293583; RG6232) is a T-cell engaging bispecific (TCB) antibody that targets tyrosinase-related protein 1 (TYRP1), which is expressed in many melanomas, thereby directing T cells to kill TYRP1-expressing tumor cells. Preclinical studies show TYRP1-TCB to have potent anti-tumor activity. This first-in-human (FIH) phase 1 dose-escalation study characterized the safety, tolerability, maximum tolerated dose/optimal biological dose, and pharmacokinetics (PK) of TYRP1-TCB in patients with metastatic melanoma (NCT04551352). Results Twenty participants with cutaneous, uveal, or mucosal TYRP1-positive melanoma received TYRP1-TCB in escalating doses (0.045 to 0.4 mg). All participants experienced ≥1 treatment-related adverse event (TRAE); two participants experienced grade 3 TRAEs. The most common toxicities were grade 1-2 cytokine release syndrome (CRS) and rash. Fractionated dosing mitigated CRS and was associated with lower levels of interleukin-6 and tumor necrosis factor-alpha. Measurement of active drug (dual TYPR1- and CD3-binding) PK rapidly identified loss of active drug exposure in all participants treated with 0.4 mg in a flat dosing schedule for ≥3 cycles. Loss of exposure was associated with development of ADAs towards both the TYRP1 and CD3 domains. A total drug PK assay, measuring free and ADA-bound forms, demonstrated that TYRP1-TCB-ADA immune complexes were present in participant samples, but showed no drug activity in vitro. Discussion This study provides important insights into how the use of active drug PK assays, coupled with mechanistic follow-up, can inform and enable ongoing benefit/risk assessment for individuals participating in FIH dose-escalation trials. Translational studies that lead to a better understanding of the underlying biology of cognate T- and B-cell interactions, ultimately resulting in ADA development to novel biotherapeutics, are needed.
Collapse
Affiliation(s)
- Anna Spreafico
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Eva Muñoz Couselo
- Department of Medical Oncology, Vall d’Hebron University Hospital and Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Anja Irmisch
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Juliana Bessa
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - George Au-Yeung
- Department of Medical Oncology, Peter MacCallum Cancer Center and Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Oliver Bechter
- Department of General Medical Oncology, Universitair Ziekenhuis (UZ), Leuven, Leuven, Belgium
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy and Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Miguel F. Sanmamed
- Department of Medical Oncology, Clínica Universidad de Navarra and Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Valentina Gambardella
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Meredith McKean
- Sarah Cannon Research Institute at Tennessee Oncology, Nashville, TN, United States
| | - Margaret Callahan
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Christian Klein
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Pablo Umaña
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Nicole Justies
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Florian Heil
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Linda Fahrni
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Eugenia Opolka-Hoffmann
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Inja Waldhauer
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Conrad Bleul
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Roland F. Staack
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Vaios Karanikas
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Stephen Fowler
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
18
|
Gevertz JL, Kareva I. Minimally sufficient experimental design using identifiability analysis. NPJ Syst Biol Appl 2024; 10:2. [PMID: 38184643 PMCID: PMC10771435 DOI: 10.1038/s41540-023-00325-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 12/12/2023] [Indexed: 01/08/2024] Open
Abstract
Mathematical models are increasingly being developed and calibrated in tandem with data collection, empowering scientists to intervene in real time based on quantitative model predictions. Well-designed experiments can help augment the predictive power of a mathematical model but the question of when to collect data to maximize its utility for a model is non-trivial. Here we define data as model-informative if it results in a unique parametrization, assessed through the lens of practical identifiability. The framework we propose identifies an optimal experimental design (how much data to collect and when to collect it) that ensures parameter identifiability (permitting confidence in model predictions), while minimizing experimental time and costs. We demonstrate the power of the method by applying it to a modified version of a classic site-of-action pharmacokinetic/pharmacodynamic model that describes distribution of a drug into the tumor microenvironment (TME), where its efficacy is dependent on the level of target occupancy in the TME. In this context, we identify a minimal set of time points when data needs to be collected that robustly ensures practical identifiability of model parameters. The proposed methodology can be applied broadly to any mathematical model, allowing for the identification of a minimally sufficient experimental design that collects the most informative data.
Collapse
Affiliation(s)
- Jana L Gevertz
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, USA.
| | - Irina Kareva
- Quantitative Pharmacology Department, EMD Serono, Merck KGaA, Billerica, MA, USA
| |
Collapse
|
19
|
Guo Y, Quijano Cardé NA, Kang L, Verona R, Banerjee A, Kobos R, Chastain K, Uhlar C, Pillarisetti K, Doyle M, Smit J, Haddish‐Berhane N, Ouellet D. Teclistamab: Mechanism of action, clinical, and translational science. Clin Transl Sci 2024; 17:e13717. [PMID: 38266057 PMCID: PMC10784707 DOI: 10.1111/cts.13717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/31/2023] [Accepted: 12/19/2023] [Indexed: 01/26/2024] Open
Abstract
Multiple myeloma (MM) remains incurable despite improvements in treatment options. B-cell maturation antigen (BCMA) is predominantly expressed in B-lineage cells and represents a promising new target for MM. Teclistamab (TECVAYLITM ) is the first T-cell redirecting bispecific antibody approved for patients with MM. Targeting both CD3 receptor complex on T cells and BCMA on myeloma cells, teclistamab leads to T-cell activation and subsequent lysis of BCMA+ cells. The recommended dose of teclistamab is 1.5 mg/kg subcutaneous weekly after two step-up doses of 0.06 and 0.3 mg/kg, which was selected after review of safety, efficacy, pharmacokinetic, and pharmacodynamic data. Exposure-response analyses of efficacy and safety data were also used to confirm the teclistamab dose. Teclistamab resulted in a high rate of deep and durable responses (63% overall response, 45.5% complete response or better, with 22 months median duration of response) in patients with triple-exposed relapsed/refractory MM. Common adverse reactions included cytokine release syndrome, hematologic abnormalities, and infections. Teclistamab is currently being investigated as monotherapy as well as combination therapy across different MM indications.
Collapse
Affiliation(s)
- Yue Guo
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | | | - Lijuan Kang
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | - Raluca Verona
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | - Arnob Banerjee
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | - Rachel Kobos
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | | | - Clarissa Uhlar
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | | | | | - Jennifer Smit
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | | | | |
Collapse
|
20
|
Frey G, Cugnetti APG, Liu H, Xing C, Wheeler C, Chang HW, Boyle WJ, Short JM. A novel conditional active biologic anti-EpCAM x anti-CD3 bispecific antibody with synergistic tumor selectivity for cancer immunotherapy. MAbs 2024; 16:2322562. [PMID: 38445633 PMCID: PMC10936661 DOI: 10.1080/19420862.2024.2322562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/20/2024] [Indexed: 03/07/2024] Open
Abstract
Epithelial cell adhesion molecule (EpCAM) is a transmembrane glycoprotein that plays several roles in cancer biology. EpCAM is an attractive therapeutic target because of its expression in most solid tumors. However, targeting EpCAM has been challenging because it is also highly expressed in normal epithelial tissues. Initial attempts to develop EpCAM-specific T-cell engagers were unsuccessful due to severe cytokine release effects, as well as serious on-target, off-tumor drug-related toxicities. We developed novel, conditionally active biological (CAB) bispecific antibodies that bind to both EpCAM and CD3 in an acidic tumor microenvironment. In healthy tissues, binding to EpCAM and CD3 is greatly reduced by a novel, dual CAB selection, where each binding domain is independently blocked by the presence of physiological chemicals known as Protein-associated Chemical Switches (PaCS). The CAB anti-EpCAM T-cell engagers displayed the anticipated bispecific binding properties and mediated the potent lysis of EpCAM-positive cancer cell lines through the recruitment of T cells in the tumor microenvironment. Xenograft studies showed that the efficacy of CAB bispecific antibodies is similar to that of a non-CAB anti-EpCAM bispecific antibody, but they have markedly reduced toxicity in non-human primates, indicating an unprecedentedly widened therapeutic index of over 100-fold. These preclinical results indicate that the dual CAB bispecific antibody is potentially both a powerful and safe therapeutic platform and a promising T cell-engaging treatment for patients with EpCAM-expressing tumors.
Collapse
Affiliation(s)
- Gerhard Frey
- Research & Development, BioAtla Inc, San Diego, CA, USA
| | | | - Haizhen Liu
- Research & Development, BioAtla Inc, San Diego, CA, USA
| | - Charles Xing
- Research & Development, BioAtla Inc, San Diego, CA, USA
| | | | | | | | - Jay M. Short
- Research & Development, BioAtla Inc, San Diego, CA, USA
| |
Collapse
|
21
|
Qi T, Liao X, Cao Y. Development of bispecific T cell engagers: harnessing quantitative systems pharmacology. Trends Pharmacol Sci 2023; 44:880-890. [PMID: 37852906 PMCID: PMC10843027 DOI: 10.1016/j.tips.2023.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/20/2023]
Abstract
Bispecific T cell engagers (bsTCEs) have emerged as a promising class of cancer immunotherapy. Several bsTCEs have achieved marketing approval; dozens more are under clinical investigation. However, the clinical development of bsTCEs remains rife with challenges, including nuanced pharmacology, limited translatability of preclinical findings, frequent on-target toxicity, and convoluted dosing regimens. In this opinion article we present a distinct perspective on how quantitative systems pharmacology (QSP) can serve as a powerful tool for overcoming these obstacles. Recent advances in QSP modeling have empowered developers of bsTCEs to gain a deeper understanding of their context-dependent pharmacology, bridge gaps in experimental data, guide first-in-human (FIH) dose selection, design dosing regimens with expanded therapeutic windows, and improve long-term treatment outcomes. We use recent case studies to exemplify the potential of QSP techniques to support future bsTCE development.
Collapse
Affiliation(s)
- Timothy Qi
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xiaozhi Liao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
22
|
Tian Z, Shi C, Yang G, Allen JK, Shi Q, Al-Shami A, Olson JW, Smith MG, Chang Q, Kaur J, You J, Lofton TE, Gonzalez MA, Zhang Q, Zha D, Tasian SK, Jain N, Konopleva MY, Heffernan T, Molldrem JJ. Preclinical development of 1B7/CD3, a novel anti-TSLPR bispecific antibody that targets CRLF2-rearranged Ph-like B-ALL. Leukemia 2023; 37:2006-2016. [PMID: 37634013 PMCID: PMC10539166 DOI: 10.1038/s41375-023-02010-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/03/2023] [Accepted: 08/17/2023] [Indexed: 08/28/2023]
Abstract
Patients harboring CRLF2-rearranged B-lineage acute lymphocytic leukemia (B-ALL) face a 5-year survival rate as low as 20%. While significant gains have been made to position targeted therapies for B-ALL treatment, continued efforts are needed to develop therapeutic options with improved duration of response. Here, first we have demonstrated that patients with CRLF2-rearranged Ph-like ALL harbor elevated thymic stromal lymphopoietin receptor (TSLPR) expression, which is comparable with CD19. Then we present and evaluate the anti-tumor characteristics of 1B7/CD3, a novel CD3-redirecting bispecific antibody (BsAb) that co-targets TSLPR. In vitro, 1B7/CD3 exhibits optimal binding to both human and cynomolgus CD3 and TSLPR. Further, 1B7/CD3 was shown to induce potent T cell activation and tumor lytic activity in both cell lines and primary B-ALL patient samples. Using humanized cell- or patient-derived xenograft models, 1B7/CD3 treatment was shown to trigger dose-dependent tumor remission or growth inhibition across donors as well as induce T cell activation and expansion. Pharmacokinetic studies in murine models revealed 1B7/CD3 to exhibit a prolonged half-life. Finally, toxicology studies using cynomolgus monkeys found that the maximum tolerated dose of 1B7/CD3 was ≤1 mg/kg. Overall, our preclinical data provide the framework for the clinical evaluation of 1B7/CD3 in patients with CRLF2-rearranged B-ALL.
Collapse
Grants
- U01 CA232486 NCI NIH HHS
- U01 CA243072 NCI NIH HHS
- AbbVie, Genentech, F. Hoffman LaRoche, Stemline Therapeutics, Collectis, Calithera, AstraZeneca, Sanofi, Forty Seven, Eli Lilly, Ablynx, Agios, Allogene, Precision Biosciences, Daiichi Sankyo, Rafael Pharmaceutical, Novartis
Collapse
Affiliation(s)
- Ze Tian
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Chunhua Shi
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guojun Yang
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason K Allen
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qing Shi
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amin Al-Shami
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jill Wardell Olson
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melinda G Smith
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qing Chang
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jasbir Kaur
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Junping You
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy E Lofton
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michelle A Gonzalez
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Zhang
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - DongXing Zha
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarah K Tasian
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia; Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Nitin Jain
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marina Y Konopleva
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy Heffernan
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Translational Research to Advance Therapeutics and Innovation in Oncology (TRACTION), The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jeffrey J Molldrem
- ORBIT Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Hematopoietic Biology & Malignancy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
23
|
Niu J, Wang W, Ouellet D. Mechanism-based pharmacokinetic and pharmacodynamic modeling for bispecific antibodies: challenges and opportunities. Expert Rev Clin Pharmacol 2023; 16:977-990. [PMID: 37743720 DOI: 10.1080/17512433.2023.2257136] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/06/2023] [Indexed: 09/26/2023]
Abstract
INTRODUCTION Unlike conventional antibodies, bispecific antibodies (bsAbs) are engineered antibody- or antibody fragment-based molecules that can simultaneously recognize two different epitopes or antigens. Over the past decade, there has been an explosion of bsAbs being developed across therapeutic areas. Development of bsAbs presents unique challenges and mechanism-based pharmacokinetic/pharmacodynamic (PK/PD) modeling has served as a powerful tool to optimize their development and realize their clinical utility. AREAS COVERED In this review, the guiding principles and case examples of how fit-for-purpose, mechanism-based PK/PD models have been applied to answer questions commonly encountered in bsAb development are presented. Such models characterize the key pharmacological elements of bsAbs, and they can be utilized for model-informed drug development. We also include the discussion of challenges, knowledge gaps and future direction for such models. EXPERT OPINION Mechanistic PK/PD modeling is a powerful tool to support the development of bsAbs. These models can be extrapolated to predict treatment outcomes based on mechanisms of action (MoA) and clinical observations to form positive learn-and-confirm cycles during drug development, due to their abilities to differentiate system- and drug-specific parameters. Meanwhile, the models should keep being adapted according to novel drug design and MoA, providing continuous opportunities for model-informed drug development.
Collapse
Affiliation(s)
- Jin Niu
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, Spring House, PA, USA
| | - Weirong Wang
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, Spring House, PA, USA
| | - Daniele Ouellet
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, Spring House, PA, USA
| |
Collapse
|
24
|
Raab MS, Cohen YC, Schjesvold F, Aardalen K, Oka A, Spencer A, Wermke M, Souza AD, Kaufman JL, Cafro AM, Ocio EM, Doki N, Henson K, Trabucco G, Carrion A, Bender FC, Juif PE, Fessehatsion A, Fan L, Stonehouse JP, Blankenship JW, Granda B, De Vita S, Lu H. Preclinical discovery and initial clinical data of WVT078, a BCMA × CD3 bispecific antibody. Leukemia 2023; 37:1349-1360. [PMID: 37024520 DOI: 10.1038/s41375-023-01883-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 04/08/2023]
Abstract
B-cell maturation antigen (BCMA) is an ideal target in multiple myeloma (MM) due to highly specific expression in malignant plasma cells. BCMA-directed therapies including antibody drug conjugates, chimeric antigen receptor-T cells and bispecific antibodies (BsAbs) have shown high response rates in MM. WVT078 is an anti-BCMA× anti-CD3 BsAb that binds to BCMA with subnanomolar-affinity. It was selected based on potent T cell activation and anti-MM activity in preclinical models with favorable tolerability in cynomolgus monkey. In the ongoing first-in-human phase I dose-escalation study (NCT04123418), 33 patients received intravenous WVT078 once weekly at escalated dosing. At the active doses of 48-250 µg/kg tested to date (n = 26), the overall response rate (ORR) was 38.5% (90% CI: 22.6-56.4%) and the complete response rate (CRR, stringent complete response + complete response) was 11.5%, (90% CI: 3.2-27.2%). At the highest dose level tested, the ORR was 75% (3 of 4 patients). 26 (78.8%) patients reported at least one Grade ≥3 AE and 16 of these AEs were suspected to be drug related. 20 patients (60.6%) experienced cytokine release syndrome. WVT078 has an acceptable safety profile and shows preliminary evidence of clinical activity at doses tested to date.
Collapse
Affiliation(s)
- Marc S Raab
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Yael C Cohen
- Department of Hematology, Tel-Aviv Sourasky (Ichilov) Medical Center, Tel Aviv University, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Fredrik Schjesvold
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital, Oslo, Norway
| | | | - Adwait Oka
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Andrew Spencer
- Department of Malignant Haematology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Martin Wermke
- NCT/UCC Early Clinical Trial Unit, Universitätsklinikum Carl Gustav Carus an der Technische Universität, Dresden, Germany
| | - Anita D Souza
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Anna Maria Cafro
- Department of Hematology, Niguarda Hospital, Niguarda, Milan, Italy
| | - Enrique M Ocio
- Hospital Universitario Marqués de Valdecilla (IDIVAL), Universidad de Cantabria, Santander, Spain
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Kristin Henson
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Gina Trabucco
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Ana Carrion
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | | | | | - Liqiong Fan
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | | | - Brian Granda
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Serena De Vita
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
| | - Haihui Lu
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
| |
Collapse
|
25
|
Cattaruzza F, Nazeer A, To M, Hammond M, Koski C, Liu LY, Pete Yeung V, Rennerfeldt DA, Henkensiefken A, Fox M, Lam S, Morrissey KM, Lange Z, Podust VN, Derynck MK, Irving BA, Schellenberger V. Precision-activated T-cell engagers targeting HER2 or EGFR and CD3 mitigate on-target, off-tumor toxicity for immunotherapy in solid tumors. NATURE CANCER 2023; 4:485-501. [PMID: 36997747 PMCID: PMC10132983 DOI: 10.1038/s43018-023-00536-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 03/02/2023] [Indexed: 04/01/2023]
Abstract
To enhance the therapeutic index of T-cell engagers (TCEs), we engineered masked, precision-activated TCEs (XPAT proteins), targeting a tumor antigen (human epidermal growth factor receptor 2 (HER2) or epidermal growth factor receptor (EGFR)) and CD3. Unstructured XTEN polypeptide masks flank the N and C termini of the TCE and are designed to be released by proteases in the tumor microenvironment. In vitro, unmasked HER2-XPAT (uTCE) demonstrates potent cytotoxicity, with XTEN polypeptide masking providing up to 4-log-fold protection. In vivo, HER2-XPAT protein induces protease-dependent antitumor activity and is proteolytically stable in healthy tissues. In non-human primates, HER2-XPAT protein demonstrates a strong safety margin (>400-fold increase in tolerated maximum concentration versus uTCE). HER2-XPAT protein cleavage is low and similar in plasma samples from healthy and diseased humans and non-human primates, supporting translatability of stability to patients. EGFR-XPAT protein confirmed the utility of XPAT technology for tumor targets more widely expressed in healthy tissues.
Collapse
Affiliation(s)
- Fiore Cattaruzza
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Ayesha Nazeer
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Milton To
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Mikhail Hammond
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Caitlin Koski
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Lucas Y Liu
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - V Pete Yeung
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | | | | | - Michael Fox
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Sharon Lam
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Kari M Morrissey
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Zachary Lange
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Vladimir N Podust
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Mika K Derynck
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Bryan A Irving
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | | |
Collapse
|
26
|
Flowers D, Bassen D, Kapitanov GI, Marcantonio D, Burke JM, Apgar JF, Betts A, Hua F. A next generation mathematical model for the in vitro to clinical translation of T-cell engagers. J Pharmacokinet Pharmacodyn 2023; 50:215-227. [PMID: 36790614 DOI: 10.1007/s10928-023-09846-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 02/01/2023] [Indexed: 02/16/2023]
Abstract
T-cell engager (TCE) molecules activate the immune system and direct it to kill tumor cells. The key mechanism of action of TCEs is to crosslink CD3 on T cells and tumor associated antigens (TAAs) on tumor cells. The formation of this trimolecular complex (i.e. trimer) mimics the immune synapse, leading to therapeutic-dependent T-cell activation and killing of tumor cells. Computational models supporting TCE development must predict trimer formation accurately. Here, we present a next-generation two-step binding mathematical model for TCEs to describe trimer formation. Specifically, we propose to model the second binding step with trans-avidity and as a two-dimensional (2D) process where the reactants are modeled as the cell-surface density. Compared to the 3D binding model where the reactants are described in terms of concentration, the 2D model predicts less sensitivity of trimer formation to varying cell densities, which better matches changes in EC50 from in vitro cytotoxicity assay data with varying E:T ratios. In addition, when translating in vitro cytotoxicity data to predict in vivo active clinical dose for blinatumomab, the choice of model leads to a notable difference in dose prediction. The dose predicted by the 2D model aligns better with the approved clinical dose and the prediction is robust under variations in the in vitro to in vivo translation assumptions. In conclusion, the 2D model with trans-avidity to describe trimer formation is an improved approach for TCEs and is likely to produce more accurate predictions to support TCE development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Fei Hua
- Applied BioMath, Concord, MA, USA.
| |
Collapse
|
27
|
Grimaldi C, Ibraghimov A, Kiessling A, Rattel B, Ji C, Fuller CL, Brennan FR, Regenass-Lechner F, Shenton J, Price KD, Piché MS, Steeves MA, Prell R, Dudal S, Kronenberg S, Freebern W, Blanset D. Current nonclinical approaches for immune assessments of immuno-oncology biotherapeutics. Drug Discov Today 2023; 28:103440. [PMID: 36375739 DOI: 10.1016/j.drudis.2022.103440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/30/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Harnessing the immune system to kill tumors has been revolutionary and, as a result, has had an enormous benefit for patients in extending life and resulting in effective cures in some. However, activation of the immune system can come at the cost of undesirable adverse events such as cytokine release syndrome, immune-related adverse events, on-target/off-tumor toxicity, neurotoxicity and tumor lysis syndrome, which are safety risks that can be challenging to assess non-clinically. This article provides a review of the biology and mechanisms that can result in immune-mediated adverse effects and describes industry approaches using in vitro and in vivo models to aid in the nonclinical safety risk assessments for immune-oncology modalities. Challenges and limitations of knowledge and models are also discussed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Sherri Dudal
- Roche Pharmaceutical Research and Early Development, United States
| | - Sven Kronenberg
- Roche Pharmaceutical Research and Early Development, United States
| | | | - Diann Blanset
- Boehringer Ingelheim Pharmaceuticals, Inc., United States.
| |
Collapse
|
28
|
Ball K, Dovedi SJ, Vajjah P, Phipps A. Strategies for clinical dose optimization of T cell-engaging therapies in oncology. MAbs 2023; 15:2181016. [PMID: 36823042 PMCID: PMC9980545 DOI: 10.1080/19420862.2023.2181016] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Innovative approaches in the design of T cell-engaging (TCE) molecules are ushering in a new wave of promising immunotherapies for the treatment of cancer. Their mechanism of action, which generates an in trans interaction to create a synthetic immune synapse, leads to complex and interconnected relationships between the exposure, efficacy, and toxicity of these drugs. Challenges thus arise when designing optimal clinical dose regimens for TCEs with narrow therapeutic windows, with a variety of dosing strategies being evaluated to mitigate key side effects such as cytokine release syndrome, neurotoxicity, and on-target off-tumor toxicities. This review evaluates the current approaches to dose optimization throughout the preclinical and clinical development of TCEs, along with perspectives for improvement of these strategies. Quantitative approaches used to aid the understanding of dose-exposure-response relationships are highlighted, along with opportunities to guide the rational design of next-generation TCE molecules, and optimize their dose regimens in patients.
Collapse
Affiliation(s)
- Kathryn Ball
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Pavan Vajjah
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Alex Phipps
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
29
|
Mayawala K, de Alwis D. Dose Finding in Oncology: What is Impeding Coming of Age? Pharm Res 2022; 39:1817-1822. [PMID: 35474158 PMCID: PMC9314272 DOI: 10.1007/s11095-022-03263-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022]
Abstract
After a drug molecule enters clinical trials, there are primarily three levers to enhance probability of success: patient selection, dose selection and choice of combination agents. Of these, dose selection remains an under-appreciated aspect in oncology drug development despite numerous peer-reviewed publications. Here, we share practical challenges faced by the biopharmaceutical industry that reduce the willingness to invest in dose finding for oncology drugs. First, randomized dose finding admittedly slows down clinical development. To reduce the size of dose finding study, trend in exposure vs. tumor-size analysis can be assessed, instead of a statistical test for non-inferiority between multiple doses. Second, investment in testing a lower dose when benefit-risk at the higher dose is sufficient for regulatory approval (i.e., efficacy at the higher dose is better than standard of care and safety is acceptable) is perceived as low priority. Changing regulatory landscape must be considered to optimize dose in pre-marketing setting as post-marketing changes in dose can be commercially costly. Third, the risk of exposing patients to subtherapeutic exposures with a lower dose should be assessed scientifically instead of assuming a monotonic relationship between dose and efficacy. Only the doses which are expected to be at the plateau of dose/exposure-response curve should be investigated in Phase 1b/2. Overall, changing the perceptions that have been impeding investment in dose finding in oncology requires pragmatic discourse among biopharmaceutical industry, regulatory agencies and academia. These perceptions should also not deter dose finding for recently emerging modalities, including BITEs and CART cell therapies.
Collapse
Affiliation(s)
- Kapil Mayawala
- Oncology Early Development, Clinical Research, Merck and Co., Inc., NJ, Kenilworth, USA.
| | - Dinesh de Alwis
- Oncology Early Development, Clinical Research, Merck and Co., Inc., NJ, Kenilworth, USA
| |
Collapse
|
30
|
Komatsu SI, Kayukawa Y, Miyazaki Y, Kaneko A, Ikegami H, Ishiguro T, Nakamura M, Frings W, Ono N, Sakata K, Fujii T, Kishishita S, Kitazawa T, Endo M, Sano Y. Determination of starting dose of the T cell-redirecting bispecific antibody ERY974 targeting glypican-3 in first-in-human clinical trial. Sci Rep 2022; 12:12312. [PMID: 35853994 PMCID: PMC9296674 DOI: 10.1038/s41598-022-16564-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 07/12/2022] [Indexed: 11/21/2022] Open
Abstract
Currently, ERY974, a humanized IgG4 bispecific T cell-redirecting antibody recognizing glypican-3 and CD3, is in phase I clinical trials. After a first-in-human clinical trial of an anti-CD28 agonist monoclonal antibody resulting in severe life-threatening adverse events, the minimal anticipated biological effect level approach has been considered for determining the first-in-human dose of high-risk drugs. Accordingly, we aimed to determine the first-in-human dose of ERY974 using both the minimal anticipated biological effect level and no observed adverse effect level approaches. In the former, we used the 10% effective concentration value from a cytotoxicity assay using the huH-1 cell line with the highest sensitivity to ERY974 to calculate the first-in-human dose of 4.9 ng/kg, at which maximum drug concentration after 4 h of intravenous ERY974 infusion was equal to the 10% effective concentration value. To determine the no observed adverse effect level, we conducted a single-dose study in cynomolgus monkeys that were intravenously infused with ERY974 (0.1, 1, and 10 μg/kg). The lowest dose of 0.1 μg/kg was determined as the no observed adverse effect level, and the first-in-human dose of 3.2 ng/kg was calculated, considering body surface area and species difference. For the phase I clinical trial, we selected 3.0 ng/kg as a starting dose, which was lower than the first-in-human dose calculated from both the no observed adverse effect level and minimal anticipated biological effect level. Combining these two methods to determine the first-in-human dose of strong immune modulators such as T cell-redirecting antibodies would be a suitable approach from safety and efficacy perspectives.Clinical trial registration: JapicCTI-194805/NCT05022927.
Collapse
Affiliation(s)
- Shun-Ichiro Komatsu
- Discovery Pharmacology Department, Kamakura Research Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Yoko Kayukawa
- Discovery Pharmacology Department, Kamakura Research Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Yoko Miyazaki
- Discovery Pharmacology Department, Kamakura Research Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Akihisa Kaneko
- Discovery Pharmacology Department, Kamakura Research Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Hisashi Ikegami
- Discovery Pharmacology Department, Kamakura Research Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Takahiro Ishiguro
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-1 Nihonbashi-Muromachi 2-Chome, Chuo-ku, Tokyo, 103-8324, Japan
| | - Mikiko Nakamura
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-1 Nihonbashi-Muromachi 2-Chome, Chuo-ku, Tokyo, 103-8324, Japan
| | - Werner Frings
- Discovery Pharmacology Department, Kamakura Research Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Natsuki Ono
- Discovery Pharmacology Department, Kamakura Research Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Kiyoaki Sakata
- Discovery Pharmacology Department, Kamakura Research Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Toshihiko Fujii
- Discovery Pharmacology Department, Kamakura Research Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Shohei Kishishita
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-1 Nihonbashi-Muromachi 2-Chome, Chuo-ku, Tokyo, 103-8324, Japan
| | - Takehisa Kitazawa
- Discovery Pharmacology Department, Kamakura Research Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Mika Endo
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-1 Nihonbashi-Muromachi 2-Chome, Chuo-ku, Tokyo, 103-8324, Japan
| | - Yuji Sano
- Discovery Pharmacology Department, Kamakura Research Division, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan.
| |
Collapse
|
31
|
Carvajal RD, Nathan P, Sacco JJ, Orloff M, Hernandez-Aya LF, Yang J, Luke JJ, Butler MO, Stanhope S, Collins L, McAlpine C, Holland C, Abdullah SE, Sato T. Phase I Study of Safety, Tolerability, and Efficacy of Tebentafusp Using a Step-Up Dosing Regimen and Expansion in Patients With Metastatic Uveal Melanoma. J Clin Oncol 2022; 40:1939-1948. [PMID: 35254876 PMCID: PMC9177239 DOI: 10.1200/jco.21.01805] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/29/2021] [Accepted: 01/26/2022] [Indexed: 12/20/2022] Open
Abstract
PURPOSE This phase I study aimed to define the recommended phase II dose (RP2D) of tebentafusp, a first-in-class T-cell receptor/anti-CD3 bispecific protein, using a three-week step-up dosing regimen, and to assess its safety, pharmacokinetics, pharmacodynamics, and preliminary clinical activity in patients with metastatic uveal melanoma (mUM). METHODS In this open-label, international, phase I/II study, HLA-A*02 or HLA-A*02:01+ patients with mUM received tebentafusp 20 μg once in week 1 and 30 μg once in week 2. Dose escalation (starting at 54 μg) began at week 3 in a standard 3 + 3 design to define RP2D. Expansion-phase patients were treated at the RP2D (20-30-68 μg). Blood and tumor samples were collected for pharmacokinetics/pharmacodynamics assessment, and treatment efficacy was evaluated for all patients with baseline efficacy data as of December 2017. RESULTS Between March 2016 and December 2017, 42 eligible patients who failed a median of two previous treatments were enrolled: 19 in the dose escalation cohort and 23 in an initial dose expansion cohort. Of the dose levels investigated, 68 μg was identified as the RP2D. Most frequent treatment-emergent adverse events regardless of attribution were pyrexia (91%), rash (83%), pruritus (83%), nausea (74%), fatigue (71%), and chills (69%). Toxicity attenuated following the first three doses. The overall response rate was 11.9% (95% CI, 4.0 to 25.6). With a median follow-up of 32.4 months, median overall survival was 25.5 months (range, 0.89-31.1 months) and 1-year overall survival rate was 67%. Treatment was associated with increased tumor T-cell infiltration and transient increases in serum inflammatory mediators. CONCLUSION Using a step-up dosing regimen of tebentafusp allowed a 36% increase in the RP2D compared with weekly fixed dosing, with a manageable side-effect profile and a signal of efficacy in mUM.
Collapse
Affiliation(s)
- Richard D. Carvajal
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
| | - Paul Nathan
- Department of Medical Oncology, Mount Vernon Cancer Centre, London, United Kingdom
| | - Joseph J. Sacco
- Clatterbridge Cancer Centre, Bebington, United Kingdom
- University of Liverpool, Liverpool, United Kingdom
| | - Marlana Orloff
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | | - Jessica Yang
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
| | - Jason J. Luke
- University of Chicago, Chicago, IL
- UPMC/University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA
| | | | | | | | | | | | | | - Takami Sato
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
32
|
Kasichayanula S, Mandlekar S, Shivva V, Patel M, Girish S. Evolution of Preclinical Characterization and Insights into Clinical Pharmacology of Checkpoint Inhibitors Approved for Cancer Immunotherapy. Clin Transl Sci 2022; 15:1818-1837. [PMID: 35588531 PMCID: PMC9372426 DOI: 10.1111/cts.13312] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer immunotherapy has significantly advanced the treatment paradigm in oncology, with approvals of immuno‐oncology agents for over 16 indications, many of them first line. Checkpoint inhibitors (CPIs) are recognized as an essential backbone for a successful anticancer therapy regimen. This review focuses on the US Food and Drug Administration (FDA) regulatory approvals of major CPIs and the evolution of translational advances since their first approval close to a decade ago. In addition, critical preclinical and clinical pharmacology considerations, an overview of the pharmacokinetic and dose/regimen aspects, and a discussion of the future of CPI translational and clinical pharmacology as combination therapy becomes a mainstay of industrial immunotherapy development and in clinical practice are also discussed.
Collapse
Affiliation(s)
| | | | - Vittal Shivva
- Genentech, 1 DNA Way, South San Francisco, 94080, CA
| | - Maulik Patel
- AbbVie Inc., 1000 Gateway Blvd, South San Francisco, 94080, CA
| | - Sandhya Girish
- Gilead Sciences, 310 Lakeside Drive, Foster City, 94404, CA
| |
Collapse
|
33
|
Harding JJ, Garrido-Laguna I, Chen X, Basu C, Dowlati A, Forgie A, Hooper AT, Kamperschroer C, Max SI, Moreau A, Shannon M, Wong GY, Hong DS. A Phase 1 Dose-Escalation Study of PF-06671008, a Bispecific T-Cell-Engaging Therapy Targeting P-Cadherin in Patients With Advanced Solid Tumors. Front Immunol 2022; 13:845417. [PMID: 35493516 PMCID: PMC9047766 DOI: 10.3389/fimmu.2022.845417] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/07/2022] [Indexed: 11/18/2022] Open
Abstract
P-cadherin is a cell-cell adhesion molecule that is overexpressed in several solid tumors. PF-06671008 is a T-cell–redirecting bispecific antibody that engages both P-cadherin on tumors and CD3ϵ on T cells and induces antitumor activity in preclinical models. We conducted a phase 1, open-label, first-in-human, dose-escalation study to characterize the safety and tolerability of PF-06671008, towards determining the recommended phase 2 dose. Adult patients with treatment-refractory solid tumors received PF-06671008 (1.5–400 ng/kg) as a weekly intravenous (IV) infusion on a 21-day/3-week cycle. Parallel cohorts evaluated dosing via subcutaneous injection (SC) or an IV-prime dose. Of the 27 patients enrolled in the study, 24 received PF-06671008 IV in escalating doses, two received SC, and one IV-prime. A dose-limiting toxicity of cytokine release syndrome (CRS) occurred in the 400-ng/kg IV group, prompting evaluation of SC and IV-prime schedules. In all, 25/27 patients who received PF-06671008 reported at least one treatment-related adverse event (TRAE); the most common were CRS (21/27), decreased lymphocyte count (9/27), and hypophosphatemia (8/27). Seven patients permanently discontinued treatment due to adverse events and no treatment-related deaths occurred. Cytokine peak concentrations and CRS grade appeared to positively correlate with Cmax. Although the study was terminated due to limited antitumor activity, it provides important insights into understanding and managing immune-related adverse events resulting from this class of molecules.
Collapse
Affiliation(s)
- James J. Harding
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States
| | | | - Xiaoying Chen
- Early Oncology Development and Clinical Research, Worldwide Research and Development, Pfizer, San Diego, CA, United States
| | - Cynthia Basu
- Early Oncology Development and Clinical Research, Worldwide Research and Development, Pfizer, San Diego, CA, United States
| | - Afshin Dowlati
- University Hospitals Seidman Cancer Center and Case Western Reserve University, Cleveland, OH, United States
| | - Alison Forgie
- Early Clinical Development and Oncology Research, Worldwide Research and Development, Pfizer, San Francisco, CA, United States
| | - Andrea T. Hooper
- Oncology Research and Development, Pfizer, Inc., Pearl River, NY, United States
| | - Cris Kamperschroer
- Drug Safety Research and Development, Worldwide Research and Development, Pfizer, Groton, CT, United States
| | - Steven I. Max
- Early Oncology Development and Clinical Research, Worldwide Research and Development, Pfizer, San Diego, CA, United States
- Janssen Pharmaceutical Companies of Johnson & Johnson, Philadelphia, PA, United States
| | - Allison Moreau
- Early Oncology Development and Clinical Research, Worldwide Research and Development, Pfizer, San Diego, CA, United States
| | - Megan Shannon
- Early Oncology Development and Clinical Research, Worldwide Research and Development, Pfizer, San Diego, CA, United States
| | - Gilbert Y. Wong
- Early Clinical Development and Oncology Research, Worldwide Research and Development, Pfizer, San Francisco, CA, United States
| | - David S. Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: David S. Hong,
| |
Collapse
|
34
|
Dudal S, Bissantz C, Caruso A, David-Pierson P, Driessen W, Koller E, Krippendorff BF, Lechmann M, Olivares-Morales A, Paehler A, Rynn C, Türck D, Van De Vyver A, Wang K, Winther L. Translating pharmacology models effectively to predict therapeutic benefit. Drug Discov Today 2022; 27:1604-1621. [PMID: 35304340 DOI: 10.1016/j.drudis.2022.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/03/2022] [Accepted: 03/11/2022] [Indexed: 12/26/2022]
Abstract
Many in vitro and in vivo models are used in pharmacological research to evaluate the role of targeted proteins in a disease. Understanding the translational relevance and limitation of these models for analyzing the disposition, pharmacokinetic/pharmacodynamic (PK/PD) profile, mechanism, and efficacy of a drug, is essential when selecting the most appropriate model of the disease of interest and predicting clinically efficacious doses of the investigational drug. Here, we review selected animal models used in ophthalmology, infectious diseases, oncology, autoimmune diseases, and neuroscience. Each area has specific challenges around translatability and determination of an efficacious dose: new patient-specific dosing methods could help overcome these limitations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Ken Wang
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | |
Collapse
|
35
|
Iwata Y, Narushima Y, Harada A, Mishima M. Priming treatment with T-cell redirecting bispecific antibody ERY974 reduced cytokine induction without losing cytotoxic activity in vitro by changing the chromatin state in T cells. Toxicol Appl Pharmacol 2022; 441:115986. [PMID: 35304238 DOI: 10.1016/j.taap.2022.115986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/27/2022] [Accepted: 03/10/2022] [Indexed: 11/30/2022]
Abstract
CD3 bispecific constructs are anticipated to become an important form of cancer immunotherapy, but they frequently cause cytokine release syndrome (CRS) that is difficult to manage in clinical contexts. A combination of intra-patient dose escalation and immunosuppressive treatment is widely used to mitigate CRS. Studies suggest that CRS after subsequent doses of CD3 bispecific constructs is less severe than after the priming dose, and that step-up dosing reduces cytokine levels in animals and humans. However, the mechanism underlying the reduced cytokine induction after priming treatment with CD3 bispecific constructs is unclear. To understand human T-cell activation and chromatin states after priming treatment with CD3 bispecific construct targeting CD3ɛ and glypican 3 (ERY974), we examined cytokine levels, cytokine mRNA expression, CD3ɛ expression, CD3-mediated signal transduction, T cell activation markers, cytotoxicity against target cells, and chromatin states in T cells after ERY974 priming treatment or negative control. The second ERY974 treatment decreased cytokines on Day 8, and ERY974 priming treatment changed the chromatin state in T cells. CD3ɛ expression, CD3-mediated signal transduction, T cell activation markers, and cytotoxicity were similar between the priming treatment with ERY974 and negative control. The present study suggests that chromatin state changes in T cells after the priming treatment was a pivotal factor in the mitigation of cytokine release after the second ERY974 treatment.
Collapse
Affiliation(s)
- Yoshika Iwata
- Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan.
| | - Yuta Narushima
- Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Asako Harada
- Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Masayuki Mishima
- Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| |
Collapse
|
36
|
Seung E, Xing Z, Wu L, Rao E, Cortez-Retamozo V, Ospina B, Chen L, Beil C, Song Z, Zhang B, Levit M, Deng G, Hebert A, Kirby P, Li A, Poulton EJ, Vicente R, Garrigou A, Piepenhagen P, Ulinski G, Sanicola-Nadel M, Bangari DS, Qiu H, Pao L, Wiederschain D, Wei R, Yang ZY, Nabel GJ. A trispecific antibody targeting HER2 and T cells inhibits breast cancer growth via CD4 cells. Nature 2022; 603:328-334. [PMID: 35197632 DOI: 10.1038/s41586-022-04439-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 01/18/2022] [Indexed: 12/19/2022]
Abstract
Effective antitumour immunity depends on the orchestration of potent T cell responses against malignancies1. Regression of human cancers has been induced by immune checkpoint inhibitors, T cell engagers or chimeric antigen receptor T cell therapies2-4. Although CD8 T cells function as key effectors of these responses, the role of CD4 T cells beyond their helper function has not been defined. Here we demonstrate that a trispecific antibody to HER2, CD3 and CD28 stimulates regression of breast cancers in a humanized mouse model through a mechanism involving CD4-dependent inhibition of tumour cell cycle progression. Although CD8 T cells directly mediated tumour lysis in vitro, CD4 T cells exerted antiproliferative effects by blocking cancer cell cycle progression at G1/S. Furthermore, when T cell subsets were adoptively transferred into a humanized breast cancer tumour mouse model, CD4 T cells alone inhibited HER2+ breast cancer growth in vivo. RNA microarray analysis revealed that CD4 T cells markedly decreased tumour cell cycle progression and proliferation, and also increased pro-inflammatory signalling pathways. Collectively, the trispecific antibody to HER2 induced T cell-dependent tumour regression through direct antitumour and indirect pro-inflammatory/immune effects driven by CD4 T cells.
Collapse
Affiliation(s)
- Edward Seung
- Sanofi R&D, North America, Cambridge, MA, USA.,ModeX Therapeutics, Natick, MA, USA
| | - Zhen Xing
- Sanofi R&D, North America, Cambridge, MA, USA
| | - Lan Wu
- Sanofi R&D, North America, Cambridge, MA, USA.,ModeX Therapeutics, Natick, MA, USA
| | - Ercole Rao
- Sanofi R&D, Frankfurt, Frankfurt am Main, Germany
| | | | | | - Liqing Chen
- Sanofi R&D, North America, Cambridge, MA, USA
| | | | - Zhili Song
- Sanofi R&D, North America, Cambridge, MA, USA
| | | | | | - Gejing Deng
- Sanofi R&D, North America, Cambridge, MA, USA
| | | | - Patrick Kirby
- Sanofi R&D, North America, Cambridge, MA, USA.,Dragonfly Therapeutics, Waltham, MA, USA
| | - Aiqun Li
- Sanofi R&D, North America, Cambridge, MA, USA
| | | | | | | | | | | | | | | | - Huawei Qiu
- Sanofi R&D, North America, Cambridge, MA, USA
| | - Lily Pao
- Sanofi R&D, North America, Cambridge, MA, USA.
| | - Dmitri Wiederschain
- Sanofi R&D, North America, Cambridge, MA, USA.,Jounce Therapeutics, Cambridge, MA, USA
| | - Ronnie Wei
- Sanofi R&D, North America, Cambridge, MA, USA.,ModeX Therapeutics, Natick, MA, USA
| | - Zhi-Yong Yang
- Sanofi R&D, North America, Cambridge, MA, USA. .,ModeX Therapeutics, Natick, MA, USA.
| | - Gary J Nabel
- Sanofi R&D, North America, Cambridge, MA, USA. .,ModeX Therapeutics, Natick, MA, USA.
| |
Collapse
|
37
|
Frances N, Bacac M, Bray-French K, Christen F, Hinton H, Husar E, Quackenbush E, Schäfer M, Schick E, Vyver AVD, Richter WF. Novel In Vivo and In Vitro Pharmacokinetic/Pharmacodynamic-Based Human Starting Dose Selection for Glofitamab. J Pharm Sci 2021; 111:1208-1218. [PMID: 34953862 DOI: 10.1016/j.xphs.2021.12.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/19/2021] [Accepted: 12/19/2021] [Indexed: 12/30/2022]
Abstract
We present a novel approach for first-in-human (FIH) dose selection of the CD20xCD3 bispecific antibody, glofitamab, based on pharmacokinetic/pharmacodynamic (PKPD) assessment in cynomolgus monkeys to select a high, safe starting dose, with cytokine release (CR) as the PD endpoint. Glofitamab pharmacokinetics were studied in mice and cynomolgus monkeys; PKPD of IL-6, TNF-α and interferon-γ release following glofitamab, with/without obinutuzumab pretreatment (Gpt) was studied in cynomolgus monkeys. Potency differences for CR between cynomolgus monkeys and humans were determined by glofitamab incubation in whole blood of both species. The PKPD model for CR was translated to humans to project a starting dose that did not induce CR exceeding a clinically-predefined threshold. In cynomolgus monkeys, glofitamab showed a species-specific atypical high clearance, with and without B-cell debulking by Gpt. CR was related to glofitamab serum levels and B-cell counts. B-cell reduction by Gpt led to a marked decrease in CR. FIH starting dose (5 µg) was selected based on IL-6 release considering the markedly higher glofitamab in vitro potency in human vs monkey blood. This is a novel PKPD-based approach for selection of FIH starting dose for a CD20xCD3 bispecific antibody in B-cell lymphoma, evidenced in the glofitamab study, NP30179 (NCT03075696).
Collapse
Affiliation(s)
- Nicolas Frances
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Marina Bacac
- Roche Innovation Center Zürich, Roche Pharmaceutical Research and Early Development, Zürich, Switzerland
| | - Katharine Bray-French
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - François Christen
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Heather Hinton
- Roche Innovation Center Zürich, Roche Pharmaceutical Research and Early Development, Zürich, Switzerland
| | - Elisabeth Husar
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Elizabeth Quackenbush
- Roche Innovation Center New York, Roche Pharmaceutical Research and Early Development, New York City, NY
| | - Martin Schäfer
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, Penzberg, Germany
| | - Eginhard Schick
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Arthur Van De Vyver
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Wolfgang F Richter
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland.
| |
Collapse
|
38
|
Van De Vyver A, Eigenmann M, Ovacik M, Pohl C, Herter S, Weinzierl T, Fauti T, Klein C, Lehr T, Bacac M, Walz AC. A Novel Approach for Quantifying the Pharmacological Activity of T-Cell Engagers Utilizing In Vitro Time Course Experiments and Streamlined Data Analysis. AAPS J 2021; 24:7. [PMID: 34862519 PMCID: PMC8817205 DOI: 10.1208/s12248-021-00637-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/11/2021] [Indexed: 11/30/2022] Open
Abstract
CD3-bispecific antibodies are a new class of immunotherapeutic drugs against cancer. The pharmacological activity of CD3-bispecifics is typically assessed through in vitro assays of cancer cell lines co-cultured with human peripheral blood mononuclear cells (PBMCs). Assay results depend on experimental conditions such as incubation time and the effector-to-target cell ratio, which can hinder robust quantification of pharmacological activity. In order to overcome these limitations, we developed a new, holistic approach for quantification of the in vitro dose–response relationship. Our experimental design integrates a time-independent analysis of the dose–response across different time points as an alternative to the static, “snap-shot” analysis based on a single time point commonly used in dose–response assays. We show that the potency values derived from static
in vitro experiments depend on the incubation time, which leads to inconsistent results across multiple assays and compounds. We compared the potency values from the time-independent analysis with a model-based approach. We find comparably accurate potency estimates from the model-based and time-independent analyses and that the time-independent analysis provides a robust quantification of pharmacological activity. This approach may allow for an improved head-to-head comparison of different compounds and test systems and may prove useful for supporting first-in-human dose selection.
Collapse
Affiliation(s)
- Arthur Van De Vyver
- Roche Pharma Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Grenzacherstrasse 124, CH-4070, Basel, Switzerland.,Department of Clinical Pharmacy, Saarland University, Saarbrücken, Germany
| | - Miro Eigenmann
- Roche Pharma Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| | - Meric Ovacik
- Preclinical Translational Pharmacokinetics, South San Francisco, CA, Genentech, USA
| | - Christian Pohl
- Roche Pharma Research and Early Development, Roche Innovation Center Zürich, Wagistrasse 10, 8952, Schlieren, Switzerland
| | - Sylvia Herter
- Roche Pharma Research and Early Development, Roche Innovation Center Zürich, Wagistrasse 10, 8952, Schlieren, Switzerland
| | - Tina Weinzierl
- Roche Pharma Research and Early Development, Roche Innovation Center Zürich, Wagistrasse 10, 8952, Schlieren, Switzerland
| | - Tanja Fauti
- Roche Pharma Research and Early Development, Roche Innovation Center Zürich, Wagistrasse 10, 8952, Schlieren, Switzerland
| | - Christian Klein
- Roche Pharma Research and Early Development, Roche Innovation Center Zürich, Wagistrasse 10, 8952, Schlieren, Switzerland
| | - Thorsten Lehr
- Department of Clinical Pharmacy, Saarland University, Saarbrücken, Germany
| | - Marina Bacac
- Roche Pharma Research and Early Development, Roche Innovation Center Zürich, Wagistrasse 10, 8952, Schlieren, Switzerland
| | - Antje-Christine Walz
- Roche Pharma Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Grenzacherstrasse 124, CH-4070, Basel, Switzerland.
| |
Collapse
|
39
|
Lee SC, Ma JSY, Kim MS, Laborda E, Choi SH, Hampton EN, Yun H, Nunez V, Muldong MT, Wu CN, Ma W, Kulidjian AA, Kane CJ, Klyushnichenko V, Woods AK, Joseph SB, Petrassi M, Wisler J, Li J, Jamieson CAM, Schultz PG, Kim CH, Young TS. A PSMA-targeted bispecific antibody for prostate cancer driven by a small-molecule targeting ligand. SCIENCE ADVANCES 2021; 7:eabi8193. [PMID: 34380625 PMCID: PMC8357232 DOI: 10.1126/sciadv.abi8193] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/23/2021] [Indexed: 06/13/2023]
Abstract
Despite the development of next-generation antiandrogens, metastatic castration-resistant prostate cancer (mCRPC) remains incurable. Here, we describe a unique semisynthetic bispecific antibody that uses site-specific unnatural amino acid conjugation to combine the potency of a T cell-recruiting anti-CD3 antibody with the specificity of an imaging ligand (DUPA) for prostate-specific membrane antigen. This format enabled optimization of structure and function to produce a candidate (CCW702) with specific, potent in vitro cytotoxicity and improved stability compared with a bispecific single-chain variable fragment format. In vivo, CCW702 eliminated C4-2 xenografts with as few as three weekly subcutaneous doses and prevented growth of PCSD1 patient-derived xenograft tumors in mice. In cynomolgus monkeys, CCW702 was well tolerated up to 34.1 mg/kg per dose, with near-complete subcutaneous bioavailability and a PK profile supporting testing of a weekly dosing regimen in patients. CCW702 is being evaluated in a first in-human clinical trial for men with mCRPC who had progressed on prior therapies (NCT04077021).
Collapse
Affiliation(s)
- Sung Chang Lee
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jennifer S Y Ma
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Min Soo Kim
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Eduardo Laborda
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sei-Hyun Choi
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Eric N Hampton
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hwayoung Yun
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vanessa Nunez
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michelle T Muldong
- Department of Urology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christina N Wu
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wenxue Ma
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anna A Kulidjian
- Department of Orthopedic Surgery, Scripps MD Anderson Cancer Center, La Jolla, CA 92093, USA
| | - Christopher J Kane
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Vadim Klyushnichenko
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ashley K Woods
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sean B Joseph
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Mike Petrassi
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - John Wisler
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jing Li
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Christina A M Jamieson
- Department of Urology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Peter G Schultz
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA.
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Chan Hyuk Kim
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Travis S Young
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA.
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
40
|
Kareva I, Zutshi A, Gupta P, Kabilan S. Bispecific antibodies: A guide to model informed drug discovery and development. Heliyon 2021; 7:e07649. [PMID: 34381902 PMCID: PMC8334385 DOI: 10.1016/j.heliyon.2021.e07649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/02/2021] [Accepted: 07/20/2021] [Indexed: 11/27/2022] Open
Abstract
Affinity (KD) optimization of monoclonal antibodies is one of the factors that impacts the stoichiometric binding and the corresponding efficacy of a drug. This impacts the dose and the dosing regimen, making the optimum KD a critical component of drug discovery and development. Its importance is further enhanced for bispecific antibodies, where affinity of the drug needs to be optimized with respect to two targets. Mathematical modeling can have critical impact on lead compound optimization. Here we build on previous work of using mathematical models to facilitate lead compound selection, expanding analysis from two membrane bound targets to soluble targets as well. Our analysis reveals the importance of three factors for lead compound optimization: drug affinity to both targets, target turnover rates, and target distribution throughout the body. We describe a method that leverages this information to help make early stage decisions on whether to optimize affinity, and if so, which arm of the bispecific should be optimized. We apply the proposed approach to a variety of scenarios and illustrate the ability to make improved decisions in each case. We integrate results to develop a bispecific antibody KD optimization guide that can be used to improve resource allocation for lead compound selection, accelerating advancement of better compounds. We conclude with a discussion of possible ways to assess the necessary levels of target engagement for affecting disease as part of an integrative approach for model-informed drug discovery and development.
Collapse
|
41
|
Haber L, Olson K, Kelly MP, Crawford A, DiLillo DJ, Tavaré R, Ullman E, Mao S, Canova L, Sineshchekova O, Finney J, Pawashe A, Patel S, McKay R, Rizvi S, Damko E, Chiu D, Vazzana K, Ram P, Mohrs K, D'Orvilliers A, Xiao J, Makonnen S, Hickey C, Arnold C, Giurleo J, Chen YP, Thwaites C, Dudgeon D, Bray K, Rafique A, Huang T, Delfino F, Hermann A, Kirshner JR, Retter MW, Babb R, MacDonald D, Chen G, Olson WC, Thurston G, Davis S, Lin JC, Smith E. Generation of T-cell-redirecting bispecific antibodies with differentiated profiles of cytokine release and biodistribution by CD3 affinity tuning. Sci Rep 2021; 11:14397. [PMID: 34257348 PMCID: PMC8277787 DOI: 10.1038/s41598-021-93842-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/30/2021] [Indexed: 01/07/2023] Open
Abstract
T-cell-redirecting bispecific antibodies have emerged as a new class of therapeutic agents designed to simultaneously bind to T cells via CD3 and to tumor cells via tumor-cell-specific antigens (TSA), inducing T-cell-mediated killing of tumor cells. The promising preclinical and clinical efficacy of TSAxCD3 antibodies is often accompanied by toxicities such as cytokine release syndrome due to T-cell activation. How the efficacy and toxicity profile of the TSAxCD3 bispecific antibodies depends on the binding affinity to CD3 remains unclear. Here, we evaluate bispecific antibodies that were engineered to have a range of CD3 affinities, while retaining the same binding affinity for the selected tumor antigen. These agents were tested for their ability to kill tumor cells in vitro, and their biodistribution, serum half-life, and anti-tumor activity in vivo. Remarkably, by altering the binding affinity for CD3 alone, we can generate bispecific antibodies that maintain potent killing of TSA + tumor cells but display differential patterns of cytokine release, pharmacokinetics, and biodistribution. Therefore, tuning CD3 affinity is a promising method to improve the therapeutic index of T-cell-engaging bispecific antibodies.
Collapse
Affiliation(s)
- Lauric Haber
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA.
| | - Kara Olson
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Marcus P Kelly
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | | | | | - Richard Tavaré
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Erica Ullman
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Shu Mao
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Lauren Canova
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | | | | | - Arpita Pawashe
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Supriya Patel
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Ryan McKay
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Sahar Rizvi
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | | | | | | | - Priyanka Ram
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Katja Mohrs
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | | | - Jenny Xiao
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | | | - Carlos Hickey
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Cody Arnold
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Jason Giurleo
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Ya Ping Chen
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | | | - Drew Dudgeon
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Kevin Bray
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | | | - Tammy Huang
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Frank Delfino
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Aynur Hermann
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | | | - Marc W Retter
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Robert Babb
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | | | - Gang Chen
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | | | - Gavin Thurston
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Samuel Davis
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - John C Lin
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| | - Eric Smith
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, 10591, USA
| |
Collapse
|
42
|
Poussin M, Sereno A, Wu X, Huang F, Manro J, Cao S, Carpenito C, Glasebrook A, Powell Jr DJ, Demarest SJ. Dichotomous impact of affinity on the function of T cell engaging bispecific antibodies. J Immunother Cancer 2021; 9:e002444. [PMID: 34253637 PMCID: PMC8276301 DOI: 10.1136/jitc-2021-002444] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Bispecific T cell engagers represent the majority of bispecific antibodies (BsAbs) entering the clinic to treat metastatic cancer. The ability to apply these agents safely and efficaciously in the clinic, particularly for solid tumors, has been challenging. Many preclinical studies have evaluated parameters related to the activity of T cell engaging BsAbs, but many questions remain. MAIN BODY This study investigates the impact of affinity of T cell engaging BsAbs with regards to potency, efficacy, and induction of immunomodulatory receptors/ligands using HER-2/CD3 BsAbs as a model system. We show that an IgG BsAb can be as efficacious as a smaller BsAb format both in vitro and in vivo. We uncover a dichotomous relationship between tumor-associated antigen (TAA) affinity and CD3 affinity requirements for cells that express high versus low levels of TAA. HER-2 affinity directly correlated with the CD3 engager lysis potency of HER-2/CD3 BsAbs when HER-2 receptor numbers are high (~200 K/cell), while the CD3 affinity did not impact potency until its binding affinity was extremely low (<600 nM). When HER-2 receptor numbers were lower (~20 K/cell), both HER-2 and CD3 affinity impacted potency. The high affinity anti-HER-2/low CD3 affinity BsAb also demonstrated lower cytokine induction levels in vivo and a dosing paradigm atypical of extremely high potency T cell engaging BsAbs reaching peak efficacy at doses >3 mg/kg. This data confirms that low CD3 affinity provides an opportunity for improved safety and dosing for T cell engaging BsAbs. T cell redirection also led to upregulation of Programmed cell death 1 (PD-1) and 4-1BB, but not CTLA-4 on T cells, and to Programmed death-ligand 1 (PD-L1) upregulation on HER-2HI SKOV3 tumor cells, but not on HER-2LO OVCAR3 tumor cells. Using this information, we combined anti-PD-1 or anti-4-1BB monoclonal antibodies with the HER-2/CD3 BsAb in vivo and demonstrated significantly increased efficacy against HER-2HI SKOV3 tumors via both combinations. CONCLUSIONS Overall, these studies provide an informational dive into the optimization process of CD3 engaging BsAbs for solid tumors indicating that a reduced affinity for CD3 may enable a better therapeutic index with a greater selectivity for the target tumor and a reduced cytokine release syndrome. These studies also provide an additional argument for combining T cell checkpoint inhibition and co-stimulation to achieve optimal efficacy. BACKGROUND
Collapse
Affiliation(s)
- Mathilde Poussin
- Pathology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Arlene Sereno
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
| | - Xiufeng Wu
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
| | - Flora Huang
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
| | - Jason Manro
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
| | - Shanshan Cao
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
| | - Carmine Carpenito
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
- Stelexis, New York, New York, USA
| | - Andrew Glasebrook
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
- Toralgen, San Diego, California, USA
| | - Daniel J Powell Jr
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephen J Demarest
- Eli Lilly and Company Biotechnology Center San Diego, San Diego, California, USA
- Tentarix, San Diego, California, USA
| |
Collapse
|
43
|
Van De Vyver AJ, Marrer-Berger E, Wang K, Lehr T, Walz AC. Cytokine Release Syndrome By T-cell-Redirecting Therapies: Can We Predict and Modulate Patient Risk? Clin Cancer Res 2021; 27:6083-6094. [PMID: 34162679 DOI: 10.1158/1078-0432.ccr-21-0470] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/30/2021] [Accepted: 06/11/2021] [Indexed: 11/16/2022]
Abstract
T-cell-redirecting therapies are promising new therapeutic options in the field of cancer immunotherapy, but the development of these modalities is challenging. A commonly observed adverse event in patients treated with T-cell-redirecting therapies is cytokine release syndrome (CRS). Its clinical manifestation is a burden on patients, and continues to be a big hurdle in the clinical development of this class of therapeutics. We review different T-cell-redirecting therapies, discuss key factors related to cytokine release and potentially leading to CRS, and present clinical mitigation strategies applied for those modalities. We propose to dissect those risk factors into drug-target-disease-related factors and individual patient risk factors. Aiming to optimize the therapeutic intervention of these modalities, we illustrate how the knowledge on drug-target-disease-related factors, such as target expression, binding affinity, and target accessibility, can be leveraged in a model-based framework and highlight with case examples how modeling and simulation is applied to guide drug discovery and development. We draw attention to the current gaps in predicting the individual patient's risk towards a high-grade CRS, which requires further considerations of risk factors related, but not limited to, the patient's demographics, genetics, underlying pathologies, treatment history, and environmental exposures. The drug-target-disease-related factors together with the individual patient's risk factors can be regarded as the patient's propensity for developing CRS in response to therapy. As an outlook, we suggest implementing a risk scoring system combined with mechanistic modeling to enable the prediction of an individual patient's risk of CRS for a given therapeutic intervention.
Collapse
Affiliation(s)
- Arthur J Van De Vyver
- Roche Pharma Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Switzerland. .,Saarland University, Department of Clinical Pharmacy, Saarbrücken, Germany
| | - Estelle Marrer-Berger
- Roche Pharma Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Switzerland
| | - Ken Wang
- Roche Pharma Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Switzerland
| | - Thorsten Lehr
- Saarland University, Department of Clinical Pharmacy, Saarbrücken, Germany
| | - Antje-Christine Walz
- Roche Pharma Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Switzerland
| |
Collapse
|
44
|
Maulana TI, Kromidas E, Wallstabe L, Cipriano M, Alb M, Zaupa C, Hudecek M, Fogal B, Loskill P. Immunocompetent cancer-on-chip models to assess immuno-oncology therapy. Adv Drug Deliv Rev 2021; 173:281-305. [PMID: 33798643 DOI: 10.1016/j.addr.2021.03.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 03/08/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
The advances in cancer immunotherapy come with several obstacles, limiting its widespread use and benefits so far only to a small subset of patients. One of the underlying challenges remains to be the lack of representative nonclinical models that translate to human immunity and are able to predict clinical efficacy and safety outcomes. In recent years, immunocompetent Cancer-on-Chip models emerge as an alternative human-based platform that enables the integration and manipulation of complex tumor microenvironment. In this review, we discuss novel opportunities offered by Cancer-on-Chip models to advance (mechanistic) immuno-oncology research, ranging from design flexibility to multimodal analysis approaches. We then exemplify their (potential) applications for the research and development of adoptive cell therapy, immune checkpoint therapy, cytokine therapy, oncolytic virus, and cancer vaccines.
Collapse
|
45
|
Modeling Pharmacokinetics and Pharmacodynamics of Therapeutic Antibodies: Progress, Challenges, and Future Directions. Pharmaceutics 2021; 13:pharmaceutics13030422. [PMID: 33800976 PMCID: PMC8003994 DOI: 10.3390/pharmaceutics13030422] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/18/2021] [Accepted: 03/18/2021] [Indexed: 12/29/2022] Open
Abstract
With more than 90 approved drugs by 2020, therapeutic antibodies have played a central role in shifting the treatment landscape of many diseases, including autoimmune disorders and cancers. While showing many therapeutic advantages such as long half-life and highly selective actions, therapeutic antibodies still face many outstanding issues associated with their pharmacokinetics (PK) and pharmacodynamics (PD), including high variabilities, low tissue distributions, poorly-defined PK/PD characteristics for novel antibody formats, and high rates of treatment resistance. We have witnessed many successful cases applying PK/PD modeling to answer critical questions in therapeutic antibodies’ development and regulations. These models have yielded substantial insights into antibody PK/PD properties. This review summarized the progress, challenges, and future directions in modeling antibody PK/PD and highlighted the potential of applying mechanistic models addressing the development questions.
Collapse
|
46
|
Schaller TH, Snyder DJ, Spasojevic I, Gedeon PC, Sanchez-Perez L, Sampson JH. First in human dose calculation of a single-chain bispecific antibody targeting glioma using the MABEL approach. J Immunother Cancer 2021; 8:jitc-2019-000213. [PMID: 32273346 PMCID: PMC7254109 DOI: 10.1136/jitc-2019-000213] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2020] [Indexed: 12/13/2022] Open
Abstract
Background First-in-human (FIH) clinical trials require careful selection of a safe yet biologically relevant starting dose. Typically, such starting doses are selected based on toxicity studies in a pharmacologically relevant animal model. However, with the advent of target-specific and highly active immunotherapeutics, both the Food and Drug Administration and the European Medicines Agency have provided guidance that recommend determining a safe starting dose based on a minimum anticipated biological effect level (MABEL) approach. Methods We recently developed a T cell activating bispecific antibody that effectively treats orthotopic patient-derived malignant glioma and syngeneic glioblastoma in mice (hEGFRvIII:CD3 bi-scFv). hEGFRvIII:CD3 bi-scFv is comprized of two single chain antibody fragments (bi-scFvs) that bind mutant epidermal growth factor receptor variant III (EGFRvIII), a mutation frequently seen in malignant glioma, and human CD3ε on T cells, respectively. In order to establish a FIH dose, we used a MABEL approach to select a safe starting dose for hEGFRvIII:CD3 bi-scFv, based on a combination of in vitro data, in vivo animal studies, and theoretical human receptor occupancy modeling. Results Using the most conservative approach to the MABEL assessment, a dose of 57.4 ng hEGFRvIII:CD3 bi-scFv/kg body weight was selected as a safe starting dose for a FIH clinical study. Conclusions The comparison of our MABEL-based starting dose to our in vivo efficacious dose and the theoretical human receptor occupancy strongly supports that our human starting dose of 57.4 ng hEGFRvIII:CD3 bi-scFv/patient kg will be safe.
Collapse
Affiliation(s)
- Teilo H Schaller
- Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, United States.,Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States
| | - David J Snyder
- Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, United States
| | - Ivan Spasojevic
- PK/PD Core Laboratory, Duke Cancer Institute, Durham, North Carolina, United States.,Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States
| | - Patrick C Gedeon
- Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, United States
| | - Luis Sanchez-Perez
- Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, United States
| | - John H Sampson
- Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, United States .,Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, United States.,Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States
| |
Collapse
|
47
|
Considerations on the Calculation of the Human Equivalent Dose from Toxicology Studies for Biologic Anticancer Agents. Clin Pharmacokinet 2021; 60:563-567. [PMID: 33651328 PMCID: PMC8113299 DOI: 10.1007/s40262-021-00987-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2021] [Indexed: 12/23/2022]
|
48
|
Hey A, Baumann A, Kronenberg S, Blaich G, Mohl S, Fagg R, Ulrich P, Rattel B, Richter WF, Kiessling A, Weir L. Nonclinical Development of Biologics: Integrating Safety, Pharmacokinetics, and Pharmacodynamics to Create Smarter and More Flexible Nonclinical Safety Programs Optimizing Animal Use. Int J Toxicol 2021; 40:270-284. [PMID: 33631988 DOI: 10.1177/1091581821994288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Safety assessment of biological drugs has its challenges due to the multiple new different modalities, for example, antibody-drug conjugates, bispecifics, nanobodies, fusion proteins and advanced therapy medicinal products (ATMPs), their different pharmacokinetic and pharmacodynamic properties, and their ability to trigger immunogenicity and toxicity. In the public and in the pharmaceutical industry, there is a strong and general desire to reduce the number of animals used in research and development of drugs and in particular reducing the use of nonhuman primates. Important discussions and activities are ongoing investigating the smarter designs of early research and dose range finding studies, reuse of animals, and replacing animal experiments with in vitro studies. Other important challenges include absence of a relevant species and design of studies and developing genetically modified animals for special investigative toxicology studies. Then, the learnings and challenges from the development of the first ATMPs are available providing valuable insights in the development path for these new potentially transformative treatments. Finally, development of strategies for assessment of immunogenicity and prediction of translation of immunogenicity and associated findings to the clinic. On this, the eighth meeting for the European BioSafe members, these challenges served as the basis for the presentations and discussions during the meeting. This article serves as the workshop report reviewing the presentations and discussions at the meeting.
Collapse
Affiliation(s)
- Adam Hey
- Novartis Pharma, Basel, Switzerland
| | | | - Sven Kronenberg
- 30259Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | | - Silke Mohl
- 30259Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | | | | | | - Wolfgang F Richter
- 30259Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | | | |
Collapse
|
49
|
Wang Y, Pan D, Huang C, Chen B, Li M, Zhou S, Wang L, Wu M, Wang X, Bian Y, Yan J, Liu J, Yang M, Miao L. Dose escalation PET imaging for safety and effective therapy dose optimization of a bispecific antibody. MAbs 2021; 12:1748322. [PMID: 32275842 PMCID: PMC7153848 DOI: 10.1080/19420862.2020.1748322] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Selecting the dose for efficacy and first-in-human studies of bispecific antibodies (BsAbs) is a challenging process. Herein, positron emission tomography (PET) imaging with 89Zr-labeled IBI322, an anti-CD47/PD-L1 BsAb, was used to optimize the safety and effective therapy dose. By labeling with 89Zr, we aimed to assess the pharmacokinetics (PK), safety, and target engagement of IBI322 with dose escalation dynamic PET imaging in humanized transgenic animal models bearing MC38 tumors (knock-in of hCD47 and hPDL1). 89Zr-labeled IBI322 specifically accumulated in tumors with a tumor-to-muscle ratio of 12.37 ± 1.42 at 168 h (0.22 mg/kg) and the biodistribution of normal tissues from PET imaging could be used for preliminary safety prediction. According to the Pearson correlation analysis between the ELISA-quantified serum concentration and heart uptake (%ID/g) (r = 0.980), a modified Patlak model was proposed. The exploratory target-mediated 50% (0.38 mg/kg) and 90% (0.63 mg/kg) inhibitory mass doses were calculated with the current modified Patlak model. The preliminary pharmacodynamics (PD) study with 0.34 mg/kg revealed that the dose prediction was rational. In conclusion, dose escalation PET imaging with 89Zr-labeled antibodies is promising for PK/PD modeling and safety prediction, and helpful for determining rational dosing for preclinical and clinical trials of BsAbs.
Collapse
Affiliation(s)
- Yan Wang
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Donghui Pan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Chenrong Huang
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Bingliang Chen
- Drug Discovery Department, Innovent Biopharmaceutical (Suzhou) Co., Ltd, Suzhou, China
| | - Mingzhu Li
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Shuaixiang Zhou
- Drug Discovery Department, Innovent Biopharmaceutical (Suzhou) Co., Ltd, Suzhou, China
| | - Lizhen Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Min Wu
- Drug Discovery Department, Innovent Biopharmaceutical (Suzhou) Co., Ltd, Suzhou, China
| | - Xinyu Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Yicong Bian
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Junjie Yan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Junjian Liu
- Drug Discovery Department, Innovent Biopharmaceutical (Suzhou) Co., Ltd, Suzhou, China
| | - Min Yang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Liyan Miao
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
50
|
Germovsek E, Cheng M, Giragossian C. Allometric scaling of therapeutic monoclonal antibodies in preclinical and clinical settings. MAbs 2021; 13:1964935. [PMID: 34530672 PMCID: PMC8463036 DOI: 10.1080/19420862.2021.1964935] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/19/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023] Open
Abstract
Constant technological advancement enabled the production of therapeutic monoclonal antibodies (mAbs) and will continue to contribute to their rapid expansion. Compared to small-molecule drugs, mAbs have favorable characteristics, but also more complex pharmacokinetics (PK), e.g., target-mediated nonlinear elimination and recycling by neonatal Fc-receptor. This review briefly discusses mAb biology, similarities and differences in PK processes across species and within human, and provides a detailed overview of allometric scaling approaches for translating mAb PK from preclinical species to human and extrapolating from adults to children. The approaches described here will remain vital in mAb drug development, although more data are needed, for example, from very young patients and mAbs with nonlinear PK, to allow for more confident conclusions and contribute to further growth of this field. Improving mAb PK predictions will facilitate better planning of (pediatric) clinical studies and enable progression toward the ultimate goal of expediting drug development.
Collapse
Affiliation(s)
- Eva Germovsek
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
| | - Ming Cheng
- Development Biologicals, Drug Metabolism And Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, US
| | - Craig Giragossian
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, US
| |
Collapse
|