51
|
Molloy ME, Austin RJ, Lemon BD, Aaron WH, Ganti V, Jones A, Jones SD, Strobel KL, Patnaik P, Sexton K, Tatalick L, Yu TZ, Baeuerle PA, Law CL, Wesche H. Preclinical Characterization of HPN536, a Trispecific, T-Cell-Activating Protein Construct for the Treatment of Mesothelin-Expressing Solid Tumors. Clin Cancer Res 2020; 27:1452-1462. [PMID: 33262134 DOI: 10.1158/1078-0432.ccr-20-3392] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/12/2020] [Accepted: 11/25/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Mesothelin (MSLN) is a glycophosphatidylinositol-linked tumor antigen overexpressed in a variety of malignancies, including ovarian, pancreatic, lung, and triple-negative breast cancer. Early signs of clinical efficacy with MSLN-targeting agents have validated MSLN as a promising target for therapeutic intervention, but therapies with improved efficacy are still needed to address the significant unmet medical need posed by MSLN-expressing cancers. EXPERIMENTAL DESIGN We designed HPN536, a 53-kDa, trispecific, T-cell-activating protein-based construct, which binds to MSLN-expressing tumor cells, CD3ε on T cells, and to serum albumin. Experiments were conducted to assess the potency, activity, and half-life of HPN536 in in vitro assays, rodent models, and in nonhuman primates (NHP). RESULTS HPN536 binds to MSLN-expressing tumor cells and to CD3ε on T cells, leading to T-cell activation and potent redirected target cell lysis. A third domain of HPN536 binds to serum albumin for extension of plasma half-life. In cynomolgus monkeys, HPN536 at doses ranging from 0.1 to 10 mg/kg demonstrated MSLN-dependent pharmacologic activity, was well tolerated, and showed pharmacokinetics in support of weekly dosing in humans. CONCLUSIONS HPN536 is potent, is well tolerated, and exhibits extended half-life in NHPs. It is currently in phase I clinical testing in patients with MSLN-expressing malignancies (NCT03872206).
Collapse
Affiliation(s)
| | | | - Bryan D Lemon
- Harpoon Therapeutics, South San Francisco, California
| | - Wade H Aaron
- Harpoon Therapeutics, South San Francisco, California
| | | | - Adrie Jones
- Harpoon Therapeutics, South San Francisco, California
| | - Susan D Jones
- Harpoon Therapeutics, South San Francisco, California
| | | | | | | | | | - Timothy Z Yu
- Harpoon Therapeutics, South San Francisco, California
| | - Patrick A Baeuerle
- Harpoon Therapeutics, South San Francisco, California.,MPM Capital, Cambridge, Massachusetts.,Institute for Immunology, Ludwig-Maximilians University Munich, Planegg- Martinsried, Munich, Germany
| | - Che-Leung Law
- Harpoon Therapeutics, South San Francisco, California
| | - Holger Wesche
- Harpoon Therapeutics, South San Francisco, California
| |
Collapse
|
52
|
Song L, Xue J, Zhang J, Li S, Liu D, Zhou T. Mechanistic prediction of first-in-human dose for bispecific CD3/EpCAM T-cell engager antibody M701, using an integrated PK/PD modeling method. Eur J Pharm Sci 2020; 158:105584. [PMID: 33039565 DOI: 10.1016/j.ejps.2020.105584] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/08/2020] [Accepted: 10/01/2020] [Indexed: 12/18/2022]
Abstract
AIM M701 is a bispecific CD3/EpCAM T-cell engager antibody to treat malignant ascites. This study aimed to predict in vivo exposure-cytotoxicity relationship and human pharmacokinetics (PK) characteristics of M701, as well as to design optimal starting dose and effective dose for M701 first-in-human (FIH) study. METHOD Mechanistic in vitro PK/PD model was firstly developed based on in vitro data of M701's cytotoxicity and binding affinities with targeting receptors. The cell killing effect of M701 in vitro was driven by tri-molecular synapse, which formed by binding drug to both CD3 receptor on T cells and EpCAM receptor on tumor cells. Human exposure-response (E-R) curve in ascites was estimated using the same model structure with clinical systemic model parameters. Human PK was predicted by allometrically scaling monkey PK data, which was characterized using a two compartment model. Human PK model was integrated into in vivo synapse-based cell killing model to provide human PK/PD characteristics. Integrated human PK/PD model was applied in FIH dose design. Clinical starting dose and effective dose were suggested as the simulated drug concentration in human ascites that achieved the estimated in vivo minimally anticipated biological effect level (MABEL) and pharmacologically active level. Other approaches including PK-driven and receptor occupancy calculation were also employed in this study to verify the starting dose prediction. RESULTS In vitro M701 cytotoxicity curves under 24, 48, 72 h incubations were well captured by mechanistic synapse-based cell killing model. Human E-R curve in ascites was obtained based on in vitro model structure and clinical systematic parameters. We defined 10~20% and 80% of maximum cytotoxicity effect as in vivo MABEL and pharmacologically active level. Human E-R curve indicated in vivo EC10, EC20 and EC80 were 0.56, 1.26 and 31.6 ng/mL. For human PK model, clearance (CL, CLd), distribution volumes (Vc, Vp) and absorption rate were allometrically scaled using exponent of 0.9, 1 and -0.25. Predicted clearance and volume were 0.53- and 1.19-fold of observed data. Simulated average ascites M701 concentrations (calculated as Cave_ ascites = AUCτ/τ) were 0.81 and 32.5 ng/mL under dose of 5 and 200 μg within 2-hour i.p. infusion. By integrating human E-R curve and the simulated PK profile in ascites, we suggested 5 and 200 μg within 2-hour i.p. infusion as MABEL dose and pharmacologically active dose (PAD) for M701 FIH study. PK-driven approach predicted a starting dose of 5 μg, which was comparable to that predicted via PK/PD-driven approach. CONCLUSIONS This study predicted human ascites PK and E-R curve by integrating human PK model into in vivo synapse-based cell killing model. Optimal clinical MABEL dose and PAD of bispecific T cell engager antibody M701 were suggested based on current integrated PK/PD approach.
Collapse
Affiliation(s)
- Ling Song
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Junsheng Xue
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jing Zhang
- Wuhan YZY Biopharma Co., Ltd., Wuhan, HuBei, 430075, China
| | - Si Li
- Wuhan YZY Biopharma Co., Ltd., Wuhan, HuBei, 430075, China
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China.
| | - Tianyan Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
53
|
Breous-Nystrom E, Kronenberg S, Marrer-Berger E, Roth A, Lave T, Singer T. Transforming preclinical assessment to meet clinical relevance with advanced models. CURRENT OPINION IN TOXICOLOGY 2020. [DOI: 10.1016/j.cotox.2020.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
54
|
Morcos PN, Li J, Hosseini I, Li CC. Quantitative Clinical Pharmacology of T-Cell Engaging Bispecifics: Current Perspectives and Opportunities. Clin Transl Sci 2020; 14:75-85. [PMID: 32882099 PMCID: PMC7877841 DOI: 10.1111/cts.12877] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/11/2020] [Indexed: 12/19/2022] Open
Abstract
T-cell directing/engaging bispecifics (TDBs) enable a powerful mode of action by activating T-cells through the creation of artificial immune synapses. Their pharmacological response involves the dynamic inter-relationships among T-cells, tumor cells, and TDBs. This results in complex and challenging issues in understanding pharmacokinetics, tissue distribution, target engagement, and exposure-response relationship. Dosing strategy plays a crucial role in determining the therapeutic window of TDBs because of the desire to maximize therapeutic efficacy in the context of known mechanism-related adverse events, such as cytokine release syndrome and neurological adverse events. Such adverse events are commonly reported as the most prominent events during the initial treatment cycles and dissipate over time. Therefore, the kinetic characterization of the inter-relationships between exposure/target engagement and safety/efficacy outcomes is crucial in designing the optimal dosing regimen to maximize the benefit/risk of TDB agents. In this review, we discuss the key clinical pharmacological considerations in drug discovery and development for TDBs and provide a summary of TDBs currently in clinical development. We also propose forward-looking perspectives and opportunities to derive insights through quantitative clinical pharmacology approaches.
Collapse
Affiliation(s)
- Peter N Morcos
- Pharmaceutical Sciences
- Pharma Research and Early Development (pRED), Roche Innovation Center, New York, New York, USA
| | - Junyi Li
- Department of Clinical Pharmacology, Genentech, Roche, South San Francisco, California, USA
| | - Iraj Hosseini
- Preclinical and Translational Pharmacokinetics, Genentech, Roche, South San Francisco, California, USA
| | - Chi-Chung Li
- Department of Clinical Pharmacology, Genentech, Roche, South San Francisco, California, USA
| |
Collapse
|
55
|
Steiner G, Marban-Doran C, Langer J, Pimenova T, Duran-Pacheco G, Sauter D, Langenkamp A, Solier C, Singer T, Bray-French K, Ducret A. Enabling Routine MHC-II-Associated Peptide Proteomics for Risk Assessment of Drug-Induced Immunogenicity. J Proteome Res 2020; 19:3792-3806. [PMID: 32786679 DOI: 10.1021/acs.jproteome.0c00309] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Major histocompatibility complex-II (MHC-II)-Associated Peptide Proteomics (MAPPs) is a mass spectrometry-based approach to identify and relatively quantitate naturally processed and presented MHC-II-associated peptides that can potentially activate T cells and contribute to the immunogenicity of a drug. Acceptance of the MAPPs technology as an appropriate preclinical (and potentially clinical) immunogenicity risk assessment tool depends not only on its technical stability and robustness but also on the ability to compare results across experiments and donors. To this end, we developed a specialized MAPPs data processing pipeline, dataMAPPs, which presents complex mass spectrometric data sets in the form of heat maps (heatMAPPs), enabling rapid and convenient comparison between conditions and donors. A customized normalization procedure based on identified endogenous peptides standardizes signal intensities within and between donors and enables cross-experimental comparison. We evaluated the technical reproducibility of the MAPPs platform using tool compounds with respect to the most prominent experimental factors and found that the systematic biological differences across donors by far outweighed any technical source of variation. We illustrate the capability of the MAPPs platform to generate data that may be used for preclinical risk assessment of drug-induced immunogenicity and discuss its applicability in the clinics.
Collapse
Affiliation(s)
- Guido Steiner
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Innovation Center Basel, Basel 4070, Switzerland
| | - Céline Marban-Doran
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Innovation Center Basel, Basel 4070, Switzerland
| | - Jessica Langer
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Innovation Center Basel, Basel 4070, Switzerland
| | - Tatiana Pimenova
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Innovation Center Basel, Basel 4070, Switzerland
| | - Gonzalo Duran-Pacheco
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Innovation Center Basel, Basel 4070, Switzerland
| | - Denise Sauter
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Innovation Center Basel, Basel 4070, Switzerland
| | - Anja Langenkamp
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Innovation Center Basel, Basel 4070, Switzerland
| | - Corinne Solier
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Innovation Center Basel, Basel 4070, Switzerland
| | - Thomas Singer
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Innovation Center Basel, Basel 4070, Switzerland
| | - Katharine Bray-French
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Innovation Center Basel, Basel 4070, Switzerland
| | - Axel Ducret
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Innovation Center Basel, Basel 4070, Switzerland
| |
Collapse
|
56
|
Leach MW, Clarke DO, Dudal S, Han C, Li C, Yang Z, Brennan FR, Bailey WJ, Chen Y, Deslandes A, Loberg LI, Mayawala K, Rogge MC, Todd M, Chemuturi NV. Strategies and Recommendations for Using a Data-Driven and Risk-Based Approach in the Selection of First-in-Human Starting Dose: An International Consortium for Innovation and Quality in Pharmaceutical Development (IQ) Assessment. Clin Pharmacol Ther 2020; 109:1395-1415. [PMID: 32757299 DOI: 10.1002/cpt.2009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/02/2020] [Indexed: 01/27/2023]
Abstract
Various approaches to first-in-human (FIH) starting dose selection for new molecular entities (NMEs) are designed to minimize risk to trial subjects. One approach uses the minimum anticipated biological effect level (MABEL), which is a conservative method intended to maximize subject safety and designed primarily for NMEs having high perceived safety risks. However, there is concern that the MABEL approach is being inappropriately used for lower risk molecules with negative impacts on drug development and time to patient access. In addition, ambiguity exists in how MABEL is defined and the methods used to determine it. The International Consortium for Innovation and Quality in Pharmaceutical Development convened a working group to understand current use of MABEL and its impact on FIH starting dose selection, and to make recommendations for FIH dose selection going forward. An industry-wide survey suggested the achieved or estimated maximum tolerated dose, efficacious dose, or recommended phase II dose was > 100-fold higher than the MABEL-based starting dose for approximately one third of NMEs, including trials in patients. A decision tree and key risk factor table were developed to provide a consistent, data driven-based, and risk-based approach for selecting FIH starting doses.
Collapse
Affiliation(s)
- Michael W Leach
- Drug Safety Research and Development, Pfizer, Inc., Cambridge, Massachusetts, USA
| | - David O Clarke
- Nonclinical Safety Assessment, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, USA
| | - Sherri Dudal
- DMPK Project Leads and Translational M&S, Pharmaceutical Sciences, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Chao Han
- Biologics Development Sciences, Janssen Research and Development, LLC, Spring House, Pennsylvania, USA
| | - Chunze Li
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Zheng Yang
- Metabolism and Pharmacokinetics, Bristol-Myers Squibb Co., Princeton, New Jersey, USA
| | | | - Wendy J Bailey
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Yingxue Chen
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Boston, Massachusetts, USA
| | - Antoine Deslandes
- Translational Medicine & Early Development, Sanofi R&D, Centre de Recherche Vitry-sur-Seine 13, Vitry-sur-Seine Cedex, France
| | - Lise I Loberg
- Preclinical Safety, AbbVie, North Chicago, Illinois, USA
| | - Kapil Mayawala
- Quantitative Pharmacology and Pharmacometrics, PPDM, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Mark C Rogge
- Quantitative and Translational Science, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Marque Todd
- Drug Safety Research and Development, Pfizer, Inc., San Diego, California, USA
| | - Nagendra V Chemuturi
- Pharmacokinetic Sciences, Novartis Institute of BioMedical Research, Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
57
|
Betts A, van der Graaf PH. Mechanistic Quantitative Pharmacology Strategies for the Early Clinical Development of Bispecific Antibodies in Oncology. Clin Pharmacol Ther 2020; 108:528-541. [PMID: 32579234 PMCID: PMC7484986 DOI: 10.1002/cpt.1961] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023]
Abstract
Bispecific antibodies (bsAbs) have become an integral component of the therapeutic research strategy to treat cancer. In addition to clinically validated immune cell re‐targeting, bsAbs are being designed for tumor targeting and as dual immune modulators. Explorative preclinical and emerging clinical data indicate potential for enhanced efficacy and reduced systemic toxicity. However, bsAbs are a complex modality with challenges to overcome in early clinical trials, including selection of relevant starting doses using a minimal anticipated biological effect level approach, and predicting efficacious dose despite nonintuitive dose response relationships. Multiple factors can contribute to variability in the clinic, including differences in functional affinity due to avidity, receptor expression, effector to target cell ratio, and presence of soluble target. Mechanistic modeling approaches are a powerful integrative tool to understand the complexities and aid in clinical translation, trial design, and prediction of regimens and strategies to reduce dose limiting toxicities of bsAbs. In this tutorial, the use of mechanistic modeling to impact decision making for bsAbs is presented and illustrated using case study examples.
Collapse
Affiliation(s)
- Alison Betts
- Applied Biomath, Concord, Massachusetts, USA.,Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Piet H van der Graaf
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands.,Certara, Canterbury, UK
| |
Collapse
|
58
|
Deshaies RJ. Multispecific drugs herald a new era of biopharmaceutical innovation. Nature 2020; 580:329-338. [DOI: 10.1038/s41586-020-2168-1] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
|
59
|
Staflin K, Zuch de Zafra CL, Schutt LK, Clark V, Zhong F, Hristopoulos M, Clark R, Li J, Mathieu M, Chen X, Johnston J, Low J, Ybarra R, Slaga D, Yang J, Ovacik M, Dybdal NO, Totpal K, Junttila MR, Ellerman D, Lee G, Dennis MS, Prell R, Junttila TT. Target arm affinities determine preclinical efficacy and safety of anti-HER2/CD3 bispecific antibody. JCI Insight 2020; 5:133757. [PMID: 32271166 DOI: 10.1172/jci.insight.133757] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 03/11/2020] [Indexed: 11/17/2022] Open
Abstract
Systemic cytokine release and on-target/off-tumor toxicity to normal tissues are the main adverse effects limiting the clinical utility of T cell-redirecting therapies. This study was designed to determine how binding affinity for CD3 and tumor target HER2 impact the efficacy and nonclinical safety of anti-HER2/CD3 T cell-dependent antibodies (TDBs). Affinity was found to be a major determinant for the overall tolerability. Higher affinity for CD3 associated with rapidly elevated peripheral cytokine concentrations, weight loss in mice, and poor tolerability in cynomolgus monkeys. A TDB with lower CD3 affinity was better tolerated in cynomolgus monkeys compared with a higher CD3-affinity TDB. In contrast to tolerability, T cell binding affinity had only limited impact on in vitro and in vivo antitumor activity. High affinity for HER2 was critical for the tumor-killing activity of anti-HER2/CD3 TDBs, but higher HER2 affinity also associated with a more severe toxicity profile, including cytokine release and damage to HER2-expressing tissues. The tolerability of the anti-HER2/CD3 was improved by implementing a dose-fractionation strategy. Fine-tuning the affinities for both the tumor target and CD3 is likely a valuable strategy for achieving maximal therapeutic index of CD3 bispecific antibodies.
Collapse
|
60
|
Abstract
A bispecific antibody (bsAb) can simultaneously bind two different epitopes or antigens, allowing for multiple mechanistic functions with synergistic effects. BsAbs have attracted significant scientific attentions and efforts towards their development as drugs for cancers. There are 21 bsAbs currently undergoing clinical trials in China. Here, we review their platform technologies, expression and production, and biological activities and bioassay of these bsAbs, and summarize their structural formats and mechanisms of actions. T-cell redirection and checkpoint inhibition are two main mechanisms of the bsAbs that we discuss in detail. Furthermore, we provide our perspective on the future of bsAb development in China, including CD3-bsAbs for solid tumors and related cytokine release syndromes, expression and chemistry, manufacturing and controls, clinical development, and immunogenicity.
Collapse
Affiliation(s)
- Jing Zhang
- Wuhan YZY Biopharma Co., Ltd, Biolake City C2-1, No. 666 Gaoxin Road, Wuhan, Hubei 430075, China
| | - Jizu Yi
- Wuhan YZY Biopharma Co., Ltd, Biolake City C2-1, No. 666 Gaoxin Road, Wuhan, Hubei 430075, China
| | - Pengfei Zhou
- Wuhan YZY Biopharma Co., Ltd, Biolake City C2-1, No. 666 Gaoxin Road, Wuhan, Hubei 430075, China
| |
Collapse
|
61
|
Kamperschroer C, Shenton J, Lebrec H, Leighton JK, Moore PA, Thomas O. Summary of a workshop on preclinical and translational safety assessment of CD3 bispecifics. J Immunotoxicol 2020; 17:67-85. [DOI: 10.1080/1547691x.2020.1729902] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
| | | | - Hervé Lebrec
- Translational Safety, Amgen Research, South San Francisco, CA, USA
| | | | | | - Oliver Thomas
- Translational Safety, Amgen Research, Munich, Germany
| |
Collapse
|
62
|
Jiang X, Chen X, Jaiprasart P, Carpenter TJ, Zhou R, Wang W. Development of a minimal physiologically-based pharmacokinetic/pharmacodynamic model to characterize target cell depletion and cytokine release for T cell-redirecting bispecific agents in humans. Eur J Pharm Sci 2020; 146:105260. [PMID: 32058058 DOI: 10.1016/j.ejps.2020.105260] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/23/2020] [Accepted: 02/07/2020] [Indexed: 12/22/2022]
Abstract
T cell-redirecting bispecific antibodies (bsAbs) are highly potent tumor-killing molecules. Following bsAb mediated engagement with target cells, T cells get activated and kill target cells while inducing cytokine release, which at higher levels may lead to life-threatening cytokine release syndrome (CRS). Clinical evidence suggests that CRS can be mitigated by implementing a stepwise dosing strategy. Here, we developed a mechanism-based minimal physiologically-based pharmacokinetic/pharmacodynamic (mPBPK/PD) model using reported preclinical and clinical data from blinatumomab. The mPBPK/PD model reasonably captured blinatumomab PK and B cell depletion profiles in blood and in various tissue sites of action (i.e., red marrow perivascular niche, spleen, and lymph nodes) in patients with non-Hodgkin's lymphoma (NHL) and acute lymphoblastic leukemia (ALL). Using interleukin 6 (IL-6) as an example, our model quantitatively characterized the mitigation of cytokine release by a blinatumomab 5-15-60 µg/m2/day stepwise dosing regimen comparing to a 60 µg/m2/day flat dose in NHL patients. Furthermore, by only modifying the system parameters specific for ALL patients, the mPBPK/PD model successfully predicted the mitigation of IL-6 release by a blinatumomab 5-15 µg/m2/day stepwise dosing regimen comparing to a 15 µg/m2/day flat dose. Our work provided a case example to show how mPBPK/PD model can be used to support the discovery and clinical development of T cell-redirecting bsAbs.
Collapse
Affiliation(s)
- Xiling Jiang
- Janssen Research & Development Inc, Spring House, PA, USA
| | - Xi Chen
- Janssen Research & Development Inc, Spring House, PA, USA
| | | | | | - Rebecca Zhou
- Biology Department, Swarthmore College, Swarthmore, PA, USA
| | - Weirong Wang
- Janssen Research & Development Inc, Spring House, PA, USA.
| |
Collapse
|
63
|
Wu L, Seung E, Xu L, Rao E, Lord DM, Wei RR, Cortez-Retamozo V, Ospina B, Posternak V, Ulinski G, Piepenhagen P, Francesconi E, El-Murr N, Beil C, Kirby P, Li A, Fretland J, Vicente R, Deng G, Dabdoubi T, Cameron B, Bertrand T, Ferrari P, Pouzieux S, Lemoine C, Prades C, Park A, Qiu H, Song Z, Zhang B, Sun F, Chiron M, Rao S, Radošević K, Yang ZY, Nabel GJ. Trispecific antibodies enhance the therapeutic efficacy of tumor-directed T cells through T cell receptor co-stimulation. ACTA ACUST UNITED AC 2019; 1:86-98. [DOI: 10.1038/s43018-019-0004-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/23/2019] [Indexed: 12/26/2022]
|
64
|
Iwata Y, Sasaki M, Harada A, Taketo J, Hara T, Akai S, Ishiguro T, Narita A, Kaneko A, Mishima M. Daily ascending dosing in cynomolgus monkeys to mitigate cytokine release syndrome induced by ERY22, surrogate for T-cell redirecting bispecific antibody ERY974 for cancer immunotherapy. Toxicol Appl Pharmacol 2019; 379:114657. [DOI: 10.1016/j.taap.2019.114657] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 07/09/2019] [Indexed: 01/24/2023]
|
65
|
Strohl WR, Naso M. Bispecific T-Cell Redirection versus Chimeric Antigen Receptor (CAR)-T Cells as Approaches to Kill Cancer Cells. Antibodies (Basel) 2019; 8:E41. [PMID: 31544847 PMCID: PMC6784091 DOI: 10.3390/antib8030041] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/23/2019] [Accepted: 06/24/2019] [Indexed: 12/16/2022] Open
Abstract
The concepts for T-cell redirecting bispecific antibodies (TRBAs) and chimeric antigen receptor (CAR)-T cells are both at least 30 years old but both platforms are just now coming into age. Two TRBAs and two CAR-T cell products have been approved by major regulatory agencies within the last ten years for the treatment of hematological cancers and an additional 53 TRBAs and 246 CAR cell constructs are in clinical trials today. Two major groups of TRBAs include small, short-half-life bispecific antibodies that include bispecific T-cell engagers (BiTE®s) which require continuous dosing and larger, mostly IgG-like bispecific antibodies with extended pharmacokinetics that can be dosed infrequently. Most CAR-T cells today are autologous, although significant strides are being made to develop off-the-shelf, allogeneic CAR-based products. CAR-Ts form a cytolytic synapse with target cells that is very different from the classical immune synapse both physically and mechanistically, whereas the TRBA-induced synapse is similar to the classic immune synapse. Both TRBAs and CAR-T cells are highly efficacious in clinical trials but both also present safety concerns, particularly with cytokine release syndrome and neurotoxicity. New formats and dosing paradigms for TRBAs and CAR-T cells are being developed in efforts to maximize efficacy and minimize toxicity, as well as to optimize use with both solid and hematologic tumors, both of which present significant challenges such as target heterogeneity and the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- William R Strohl
- BiStro Biotech Consulting, LLC, 1086 Tullo Farm Rd., Bridgewater, NJ 08807, USA.
| | - Michael Naso
- Century Therapeutics, 3675 Market St., Philadelphia, PA 19104, USA
| |
Collapse
|
66
|
Betts A, Haddish-Berhane N, Shah DK, van der Graaf PH, Barletta F, King L, Clark T, Kamperschroer C, Root A, Hooper A, Chen X. A Translational Quantitative Systems Pharmacology Model for CD3 Bispecific Molecules: Application to Quantify T Cell-Mediated Tumor Cell Killing by P-Cadherin LP DART ®. AAPS J 2019; 21:66. [PMID: 31119428 PMCID: PMC6531394 DOI: 10.1208/s12248-019-0332-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/08/2019] [Indexed: 01/12/2023] Open
Abstract
CD3 bispecific antibody constructs recruit cytolytic T cells to kill tumor cells, offering a potent approach to treat cancer. T cell activation is driven by the formation of a trimolecular complex (trimer) between drugs, T cells, and tumor cells, mimicking an immune synapse. A translational quantitative systems pharmacology (QSP) model is proposed for CD3 bispecific molecules capable of predicting trimer concentration and linking it to tumor cell killing. The model was used to quantify the pharmacokinetic (PK)/pharmacodynamic (PD) relationship of a CD3 bispecific targeting P-cadherin (PF-06671008). It describes the disposition of PF-06671008 in the central compartment and tumor in mouse xenograft models, including binding to target and T cells in the tumor to form the trimer. The model incorporates T cell distribution to the tumor, proliferation, and contraction. PK/PD parameters were estimated for PF-06671008 and a tumor stasis concentration (TSC) was calculated as an estimate of minimum efficacious trimer concentration. TSC values ranged from 0.0092 to 0.064 pM across mouse tumor models. The model was translated to the clinic and used to predict the disposition of PF-06671008 in patients, including the impact of binding to soluble P-cadherin. The predicted terminal half-life of PF-06671008 in the clinic was approximately 1 day, and P-cadherin expression and number of T cells in the tumor were shown to be sensitive parameters impacting clinical efficacy. A translational QSP model is presented for CD3 bispecific molecules, which integrates in silico, in vitro and in vivo data in a mechanistic framework, to quantify and predict efficacy across species.
Collapse
Affiliation(s)
- Alison Betts
- Department of Biomedicine Design, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts, 02139, USA.
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, 2300 RA, Leiden, The Netherlands.
| | | | - Dhaval K Shah
- Department of Pharmaceutical Sciences, 455 Kapoor Hall, University at Buffalo, The State University of New York, Buffalo, New York, 14214-8033, USA
| | - Piet H van der Graaf
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, 2300 RA, Leiden, The Netherlands
| | - Frank Barletta
- Oncology Research Unit, Pfizer Inc., 401 N Middletown Rd., Pearl River, New York, 10965, USA
| | - Lindsay King
- Department of Biomedicine Design, Pfizer Inc., 1 Burtt Road, Andover, Massachusetts, USA
| | - Tracey Clark
- Established Med Business, Pfizer Inc., Eastern Point Rd, Groton, Connecticut, 06340, USA
| | - Cris Kamperschroer
- Department of Immunotoxicology, Pfizer Inc., 558 Eastern Point Road, Groton, Connecticut, 06340, USA
| | - Adam Root
- Department of Biomedicine Design, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts, 02139, USA
| | - Andrea Hooper
- Oncology Research Unit, Pfizer Inc., 401 N Middletown Rd., Pearl River, New York, 10965, USA
| | - Xiaoying Chen
- Department of Clinical Pharmacology, Pfizer Inc., 10555 Science Center Dr., San Diego, California, 92121, USA
| |
Collapse
|
67
|
Trinklein ND, Pham D, Schellenberger U, Buelow B, Boudreau A, Choudhry P, Clarke SC, Dang K, Harris KE, Iyer S, Jorgensen B, Pratap PP, Rangaswamy US, Ugamraj HS, Vafa O, Wiita AP, van Schooten W, Buelow R, Force Aldred S. Efficient tumor killing and minimal cytokine release with novel T-cell agonist bispecific antibodies. MAbs 2019; 11:639-652. [PMID: 30698484 PMCID: PMC6601548 DOI: 10.1080/19420862.2019.1574521] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
T-cell-recruiting bispecific antibodies (T-BsAbs) have shown potent tumor killing activity in humans, but cytokine release-related toxicities have affected their clinical utility. The use of novel anti-CD3 binding domains with more favorable properties could aid in the creation of T-BsAbs with improved therapeutic windows. Using a sequence-based discovery platform, we identified new anti-CD3 antibodies from humanized rats that bind to multiple epitopes and elicit varying levels of T-cell activation. In T-BsAb format, 12 different anti-CD3 arms induce equivalent levels of tumor cell lysis by primary T-cells, but potency varies by a thousand-fold. Our lead CD3-targeting arm stimulates very low levels of cytokine release, but drives robust tumor antigen-specific killing in vitro and in a mouse xenograft model. This new CD3-targeting antibody underpins a next-generation T-BsAb platform in which potent cytotoxicity is uncoupled from high levels of cytokine release, which may lead to a wider therapeutic window in the clinic.
Collapse
Affiliation(s)
| | - Duy Pham
- a Teneobio, Inc ., Menlo Park , CA , USA
| | | | - Ben Buelow
- a Teneobio, Inc ., Menlo Park , CA , USA
| | | | - Priya Choudhry
- b Department of Laboratory Medicine , University of California , San Francisco , CA , USA
| | | | - Kevin Dang
- a Teneobio, Inc ., Menlo Park , CA , USA
| | | | | | | | | | | | | | - Omid Vafa
- a Teneobio, Inc ., Menlo Park , CA , USA
| | - Arun P Wiita
- b Department of Laboratory Medicine , University of California , San Francisco , CA , USA
| | | | | | | |
Collapse
|