1
|
Ellerman DA. The Evolving Applications of Bispecific Antibodies: Reaping the Harvest of Early Sowing and Planting New Seeds. BioDrugs 2024:10.1007/s40259-024-00691-0. [PMID: 39673023 DOI: 10.1007/s40259-024-00691-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2024] [Indexed: 12/15/2024]
Abstract
After decades of gradual progress from conceptualization to early clinical trials (1960-2000), the therapeutic potential of bispecific antibodies (bisp Abs) is now being fully realized. Insights gained from both successful and unsuccessful trials are helping to identify which mechanisms of action, antibody formats, and targets prove most effective, and which may benefit from further refinement. While T-cell engagers remain the most commonly used class of bisp Abs, current efforts aim to increase their effectiveness by co-engaging costimulatory molecules, reducing escape mechanisms, and countering immunosuppression. Strategies to minimize cytokine release syndrome (CRS) are also actively under development. In addition, novel antibody formats that are selectively activated within tumors are an exciting area of research, as is the precise targeting of specific T-cell subsets. Beyond T cells, the recruitment of macrophages and dendritic cells is attracting increasing interest, with researchers exploring various macrophage receptors to promote phagocytosis or to carry out specialized functions, such as the immunologically silent clearance of amyloid-beta plaques in the brain. While bisp Abs targeting B cells are relatively limited, they are primarily aimed at inhibiting B-cell activity in autoimmune diseases. Another evolving application involves the forced interaction between proteins. Beyond the successful development of Hemlibra for hemophilia, bispecific antibodies that mimic cytokine activity are being explored. Additionally, the recruitment of cell surface ubiquitin ligases and other enzymes represents a novel and promising therapeutic strategy. In regard to antibody formats, some applications such as the combination of T-cell engagers with costimulatory molecules are driving the development of trispecific antibodies, at least in preclinical settings. However, the increasing structural complexity of multispecific antibodies often leads to more challenging development paths, and the number of multispecific antibodies in clinical trials remains low. The clinical success of certain applications and well-established production methods position this therapeutic class to expand its benefits into other therapeutic areas.
Collapse
Affiliation(s)
- Diego A Ellerman
- Antibody Engineering Department, Genentech Inc, South San Francisco, USA.
| |
Collapse
|
2
|
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
|
3
|
Liu J, Zhu J. Progresses of T-cell-engaging bispecific antibodies in treatment of solid tumors. Int Immunopharmacol 2024; 138:112609. [PMID: 38971103 DOI: 10.1016/j.intimp.2024.112609] [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: 05/27/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/08/2024]
Abstract
T-cell-engaging bispecific antibody (TCB) therapies have emerged as a promising immunotherapeutic approach, effectively redirecting effector T cells to selectively eliminate tumor cells. The therapeutic potential of TCBs has been well recognized, particularly with the approval of multiple TCBs in recent years for the treatment of hematologic malignancies as well as some solid tumors. However, TCBs encounter multiple challenges in treating solid tumors, such as on-target off-tumor toxicity, cytokine release syndrome (CRS), and T cell dysfunction within the immunosuppressive tumor microenvironment, all of which may impact their therapeutic efficacy. In this review, we summarize clinical data on TCBs for solid tumor treatment, highlight the challenges faced, and discuss potential solutions based on emerging strategies from current clinical and preclinical research. These solutions include TCB structural optimization, target selection, and combination strategies. This comprehensive analysis aims to guide the development of TCBs from design to clinical application, addressing the evolving landscape of cancer immunotherapy.
Collapse
Affiliation(s)
- Junjun Liu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianwei Zhu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; Jecho Laboratories, Inc., Frederick, MD 21704, USA.
| |
Collapse
|
4
|
Goebeler ME, Stuhler G, Bargou R. Bispecific and multispecific antibodies in oncology: opportunities and challenges. Nat Rev Clin Oncol 2024; 21:539-560. [PMID: 38822215 DOI: 10.1038/s41571-024-00905-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2024] [Indexed: 06/02/2024]
Abstract
Research into bispecific antibodies, which are designed to simultaneously bind two antigens or epitopes, has advanced enormously over the past two decades. Owing to advances in protein engineering technologies and considerable preclinical research efforts, bispecific antibodies are constantly being developed and optimized to improve their efficacy and to mitigate toxicity. To date, >200 of these agents, the majority of which are bispecific immune cell engagers, are in either preclinical or clinical evaluation. In this Review, we discuss the role of bispecific antibodies in patients with cancer, including history and development, as well as innovative targeting strategies, clinical applications, and adverse events. We also discuss novel alternative bispecific antibody constructs, such as those targeting two antigens expressed by tumour cells or cells located in the tumour microenvironment. Finally, we consider future research directions in this rapidly evolving field, including innovative antibody engineering strategies, which might enable more effective delivery, overcome resistance, and thus optimize clinical outcomes.
Collapse
Affiliation(s)
- Maria-Elisabeth Goebeler
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany.
- National Center for Tumour Diseases, NCT WERA, University Hospital Würzburg, Würzburg, Germany.
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.
| | - Gernot Stuhler
- National Center for Tumour Diseases, NCT WERA, University Hospital Würzburg, Würzburg, Germany
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Ralf Bargou
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
- National Center for Tumour Diseases, NCT WERA, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
5
|
Kamperschroer C, Guffroy M, Shen A, Dokmanovich M, Stubbs M, O'Donnell LM. Nonclinical Investigation of Cytokine Mitigation Strategies for T-cell-Engaging Bispecifics in the Cynomolgus Macaque. J Immunother 2024; 47:160-171. [PMID: 38562119 DOI: 10.1097/cji.0000000000000512] [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: 01/18/2023] [Accepted: 01/29/2024] [Indexed: 04/04/2024]
Abstract
SUMMARY T-cell-directed cancer therapies such as T-cell-engaging bispecifics (TCBs) are commonly associated with cytokine release syndrome and associated clinical signs that can limit their tolerability and therapeutic benefit. Strategies for reducing cytokine release are therefore needed. Here, we report on studies performed in cynomolgus monkeys to test different approaches for mitigating cytokine release with TCBs. A "priming dose" as well as subcutaneous dosing reduced cytokine release compared with intravenous dosing but did not affect the intended T-cell response to the bispecific. As another strategy, cytokines or cytokine responses were blocked with an anti-IL-6 antibody, dexamethasone, or a JAK1/TYK2-selective inhibitor, and the effects on toxicity as well as T-cell responses to a TCB were evaluated. The JAK1/TYK2 inhibitor and dexamethasone prevented CRS-associated clinical signs on the day of TCB administration, but the anti-IL-6 had little effect. All interventions allowed for functional T-cell responses and expected damage to target-bearing tissues, but the JAK1/TYK2 inhibitor prevented the upregulation of activation markers on T cells, suggesting the potential for suppression of T-cell responses. Our results suggest that short-term prophylactic dexamethasone treatment may be an effective option for blocking cytokine responses without affecting desired T-cell responses to TCBs.
Collapse
Affiliation(s)
| | | | - Amy Shen
- Preclinical Safety, Research and Development, Sanofi
| | | | - Makeida Stubbs
- Pfizer Inc., Clinical Development and Operations, Groton, CT
| | | |
Collapse
|
6
|
Lampe H, Tam L, Hansen AR. Bi-specific T-cell engagers (BiTEs) in prostate cancer and strategies to enhance development: hope for a BiTE-r future. Front Pharmacol 2024; 15:1399802. [PMID: 38873417 PMCID: PMC11169794 DOI: 10.3389/fphar.2024.1399802] [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: 03/12/2024] [Accepted: 05/13/2024] [Indexed: 06/15/2024] Open
Abstract
Metastatic castrate resistant prostate cancer (mCRPC) continues to have poor survival rates due to limited treatment options. Bi-specific T cell engagers (BiTEs) are a promising class of novel immunotherapies with demonstrated success in haematological malignancies and melanoma. BiTEs developed for tumour associated antigens in prostate cancer have entered clinical testing. These trials have been hampered by high rates of treatment related adverse events, minimal or transient anti-tumour efficacy and generation of high titres of anti-drug antibodies. This paper aims to analyse the challenges faced by the different BiTE therapy constructs and the mCRPC tumour microenvironment that result in therapeutic resistance and identify possible strategies to overcome these issues.
Collapse
Affiliation(s)
| | | | - Aaron R. Hansen
- Department of Medical Oncology, Division of Cancer Care Services, Princess Alexandra Hospital, Metro South Health Service, Queensland Health, Brisbane, QLD, Australia
| |
Collapse
|
7
|
Klein C, Brinkmann U, Reichert JM, Kontermann RE. The present and future of bispecific antibodies for cancer therapy. Nat Rev Drug Discov 2024; 23:301-319. [PMID: 38448606 DOI: 10.1038/s41573-024-00896-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 03/08/2024]
Abstract
Bispecific antibodies (bsAbs) enable novel mechanisms of action and/or therapeutic applications that cannot be achieved using conventional IgG-based antibodies. Consequently, development of these molecules has garnered substantial interest in the past decade and, as of the end of 2023, 14 bsAbs have been approved: 11 for the treatment of cancer and 3 for non-oncology indications. bsAbs are available in different formats, address different targets and mediate anticancer function via different molecular mechanisms. Here, we provide an overview of recent developments in the field of bsAbs for cancer therapy. We focus on bsAbs that are approved or in clinical development, including bsAb-mediated dual modulators of signalling pathways, tumour-targeted receptor agonists, bsAb-drug conjugates, bispecific T cell, natural killer cell and innate immune cell engagers, and bispecific checkpoint inhibitors and co-stimulators. Finally, we provide an outlook into next-generation bsAbs in earlier stages of development, including trispecifics, bsAb prodrugs, bsAbs that induce degradation of tumour targets and bsAbs acting as cytokine mimetics.
Collapse
Affiliation(s)
- Christian Klein
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland.
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | | | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University Stuttgart, Stuttgart, Germany.
| |
Collapse
|
8
|
McCue AC, Demarest SJ, Froning KJ, Hickey MJ, Antonysamy S, Kuhlman B. Engineering a tumor-selective prodrug T-cell engager bispecific antibody for safer immunotherapy. MAbs 2024; 16:2373325. [PMID: 38962811 PMCID: PMC11225918 DOI: 10.1080/19420862.2024.2373325] [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: 02/09/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024] Open
Abstract
T-cell engaging (TCE) bispecific antibodies are potent drugs that trigger the immune system to eliminate cancer cells, but administration can be accompanied by toxic side effects that limit dosing. TCEs function by binding to cell surface receptors on T cells, frequently CD3, with one arm of the bispecific antibody while the other arm binds to cell surface antigens on cancer cells. On-target, off-tumor toxicity can arise when the target antigen is also present on healthy cells. The toxicity of TCEs may be ameliorated through the use of pro-drug forms of the TCE, which are not fully functional until recruited to the tumor microenvironment. This can be accomplished by masking the anti-CD3 arm of the TCE with an autoinhibitory motif that is released by tumor-enriched proteases. Here, we solve the crystal structure of the antigen-binding fragment of a novel anti-CD3 antibody, E10, in complex with its epitope from CD3 and use this information to engineer a masked form of the antibody that can activate by the tumor-enriched protease matrix metalloproteinase 2 (MMP-2). We demonstrate with binding experiments and in vitro T-cell activation and killing assays that our designed prodrug TCE is capable of tumor-selective T-cell activity that is dependent upon MMP-2. Furthermore, we demonstrate that a similar masking strategy can be used to create a pro-drug form of the frequently used anti-CD3 antibody SP34. This study showcases an approach to developing immune-modulating therapeutics that prioritizes safety and has the potential to advance cancer immunotherapy treatment strategies.
Collapse
Affiliation(s)
- Amelia C. McCue
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA
| | | | - Karen J. Froning
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA, USA
| | - Michael J. Hickey
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA, USA
| | | | - Brian Kuhlman
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
9
|
Surowka M, Klein C. A pivotal decade for bispecific antibodies? MAbs 2024; 16:2321635. [PMID: 38465614 PMCID: PMC10936642 DOI: 10.1080/19420862.2024.2321635] [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: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/12/2024] Open
Abstract
Bispecific antibodies (bsAbs) are a class of antibodies that can mediate novel mechanisms of action compared to monospecific monoclonal antibodies (mAbs). Since the discovery of mAbs and their adoption as therapeutic agents in the 1980s and 1990s, the development of bsAbs has held substantial appeal. Nevertheless, only three bsAbs (catumaxomab, blinatumomab, emicizumab) were approved through the end of 2020. However, since then, 11 bsAbs received regulatory agency approvals, of which nine (amivantamab, tebentafusp, mosunetuzumab, cadonilimab, teclistamab, glofitamab, epcoritamab, talquetamab, elranatamab) were approved for the treatment of cancer and two (faricimab, ozoralizumab) in non-oncology indications. Notably, of the 13 currently approved bsAbs, two, emicizumab and faricimab, have achieved blockbuster status, showing the promise of this novel class of therapeutics. In the 2020s, the approval of additional bsAbs can be expected in hematological malignancies, solid tumors and non-oncology indications, establishing bsAbs as essential part of the therapeutic armamentarium.
Collapse
Affiliation(s)
- Marlena Surowka
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Roche Glycart AG, Schlieren, Switzerland
| | - Christian Klein
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Roche Glycart AG, Schlieren, Switzerland
| |
Collapse
|
10
|
Alsajjan R, Mason WP. Bispecific T-Cell Engagers and Chimeric Antigen Receptor T-Cell Therapies in Glioblastoma: An Update. Curr Oncol 2023; 30:8501-8549. [PMID: 37754534 PMCID: PMC10529026 DOI: 10.3390/curroncol30090619] [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: 05/21/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/28/2023] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor in adults. The prognosis is extremely poor even with standard treatment of maximal safe resection, radiotherapy, and chemotherapy. Recurrence is inevitable within months, and treatment options are very limited. Chimeric antigen receptor T-cell therapy (CART) and bispecific T-cell engagers (TCEs) are two emerging immunotherapies that can redirect T-cells for tumor-specific killing and have shown remarkable success in hematological malignancies and been under extensive study for application in glioblastoma. While there have been multiple clinical trials showing preliminary evidence of safety and efficacy for CART, bispecific TCEs are still in the early stages of clinical testing, with preclinical studies showing very promising results. However, there are multiple shared challenges that need to be addressed in the future, including the route of delivery, antigen escape, the immunosuppressive tumor microenvironment, and toxicity resulting from the limited choice of tumor-specific antigens. Efforts are underway to optimize the design of both these treatments and find the ideal combination therapy to overcome these challenges. In this review, we describe the work that has been performed as well as novel approaches in glioblastoma and in other solid tumors that may be applicable in the future.
Collapse
Affiliation(s)
- Roa Alsajjan
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada
- Division of Neurology, Department of Medicine, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Warren P. Mason
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada
| |
Collapse
|
11
|
Qian L, Lin X, Gao X, Khan RU, Liao JY, Du S, Ge J, Zeng S, Yao SQ. The Dawn of a New Era: Targeting the "Undruggables" with Antibody-Based Therapeutics. Chem Rev 2023. [PMID: 37186942 DOI: 10.1021/acs.chemrev.2c00915] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The high selectivity and affinity of antibodies toward their antigens have made them a highly valuable tool in disease therapy, diagnosis, and basic research. A plethora of chemical and genetic approaches have been devised to make antibodies accessible to more "undruggable" targets and equipped with new functions of illustrating or regulating biological processes more precisely. In this Review, in addition to introducing how naked antibodies and various antibody conjugates (such as antibody-drug conjugates, antibody-oligonucleotide conjugates, antibody-enzyme conjugates, etc.) work in therapeutic applications, special attention has been paid to how chemistry tools have helped to optimize the therapeutic outcome (i.e., with enhanced efficacy and reduced side effects) or facilitate the multifunctionalization of antibodies, with a focus on emerging fields such as targeted protein degradation, real-time live-cell imaging, catalytic labeling or decaging with spatiotemporal control as well as the engagement of antibodies inside cells. With advances in modern chemistry and biotechnology, well-designed antibodies and their derivatives via size miniaturization or multifunctionalization together with efficient delivery systems have emerged, which have gradually improved our understanding of important biological processes and paved the way to pursue novel targets for potential treatments of various diseases.
Collapse
Affiliation(s)
- Linghui Qian
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xuefen Lin
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xue Gao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Rizwan Ullah Khan
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jia-Yu Liao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shubo Du
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Jingyan Ge
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore, 117544
| |
Collapse
|
12
|
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: 26] [Impact Index Per Article: 13.0] [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
|
13
|
Tapia-Galisteo A, Compte M, Álvarez-Vallina L, Sanz L. When three is not a crowd: trispecific antibodies for enhanced cancer immunotherapy. Theranostics 2023; 13:1028-1041. [PMID: 36793863 PMCID: PMC9925307 DOI: 10.7150/thno.81494] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 12/31/2022] [Indexed: 02/04/2023] Open
Abstract
Despite the clinical success of the first bispecific antibody approved by the FDA against B cell malignancies (blinatumomab), many obstacles remain such as dosing, treatment resistance, and modest efficacy in solid tumors. To overcome these limitations, considerable efforts have been dedicated to the development of multispecific antibodies, opening up new avenues to address both the complex biology of cancer and the onset of anti-tumoral immune responses. Simultaneous targeting of two tumor-associated antigens is presumed to enhance cancer cell selectivity and reduce immune escape. Co-engagement of CD3, along with agonists of co-stimulatory molecules or antagonists of co-inhibitory immune checkpoint receptors in a single molecule, may revert T cell exhaustion. Similarly, targeting of two activating receptors in NK cells may improve their cytotoxic potency. And these are only examples of the potential of antibody-based molecular entities engaging three (or more) relevant targets. From the perspective of health care costs, multispecific antibodies are appealing, since a similar (or superior) therapeutic effect could be obtained with a single therapeutic agent as with a combination of different monoclonal antibodies. Despite challenges in production, multispecific antibodies are endowed with unprecedented properties, which may render them more potent biologics for cancer therapy.
Collapse
Affiliation(s)
- Antonio Tapia-Galisteo
- Immuno-oncology and Immunotherapy Group, Biomedical Research Institute Hospital 12 de Octubre, Madrid, Spain.,Cancer Immunotherapy Unit (UNICA), Hospital Universitario 12 de Octubre, Madrid, Spain.,H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marta Compte
- Department of Antibody Engineering, Leadartis S.L., Madrid, Spain
| | - Luis Álvarez-Vallina
- Immuno-oncology and Immunotherapy Group, Biomedical Research Institute Hospital 12 de Octubre, Madrid, Spain.,Cancer Immunotherapy Unit (UNICA), Hospital Universitario 12 de Octubre, Madrid, Spain.,H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| |
Collapse
|
14
|
Vasic V, Buldun C, Ritz M, Dickopf S, Georges GJ, Spick C, Peuker A, Meier T, Mayer K, Brinkmann U. Targeted chain-exchange-mediated reconstitution of a split type-I cytokine for conditional immunotherapy. MAbs 2023; 15:2245111. [PMID: 37608616 PMCID: PMC10448976 DOI: 10.1080/19420862.2023.2245111] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 07/17/2023] [Accepted: 08/02/2023] [Indexed: 08/24/2023] Open
Abstract
Antibody-cytokine fusions targeted against tumor-associated antigens (TAAs) are promising cancer immunotherapy agents, with many such molecules currently undergoing clinical trials. However, due to the limited number of tumor-specific targets, on-target off-tumor effects can lead to systemic toxicity. Additionally, targeted cytokines can be scavenged by cytokine receptors on peripheral cells, decreasing tumor penetration. This study aims at overcoming these issues by engineering a platform for targeted conditionally active type I cytokines. Building on our previously reported PACE (Prodrug-Activating Chain Exchange) platform, we split the type I cytokine interleukin-4 (IL-4) to create two inactive IL-4 prodrugs, and fused these split IL-4 counterparts to the C-termini of antibody-like molecules that undergo proximity-induced chain exchange. In doing so, we developed IL-4 prodrugs that preferentially reconstitute into active IL-4 on target cells. We demonstrate that pre-assembled split IL-4 (without additional inactivation) retains activity and present two different strategies of splitting and inactivating IL-4. Using an IL-4 responsive cell-line, we show that IL-4 prodrugs are targeted to TAAs on target cells and regain activity upon chain exchange, primarily in a cis-activation setting. Furthermore, we demonstrate that split IL-4 complementation is also possible in a trans-activation setting, which opens up the possibility for activation of immune cells in the tumor vicinity. We demonstrate that targeted on-cell prodrug conversion is more efficient than nonspecific activation in-solution. Due to the structural similarity between IL-4 and other type I cytokines relevant in cancer immunotherapy such as IL-2, IL-15, and IL-21, cytokine-PACE may be expanded to develop a variety of targeted conditionally active cytokines for cancer immunotherapy.
Collapse
Affiliation(s)
- Vedran Vasic
- Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Can Buldun
- Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
- Bellevue Asset Management, Küsnacht, Switzerland
| | - Manfred Ritz
- Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
- Department of Chemistry, Technical University of Munich, Garching, Germany
| | - Steffen Dickopf
- Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
- Discovery Biology, Morphosys AG, Planegg, Germany
| | - Guy J. Georges
- Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Christian Spick
- Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Alessa Peuker
- Reagent Research and Design, Roche Diagnostics GmbH, Penzberg, Germany
| | - Thomas Meier
- Reagent Research and Design, Roche Diagnostics GmbH, Penzberg, Germany
| | - Klaus Mayer
- Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
15
|
Liu Y, Nguyen AW, Maynard JA. Engineering antibodies for conditional activity in the solid tumor microenvironment. Curr Opin Biotechnol 2022; 78:102809. [PMID: 36182870 DOI: 10.1016/j.copbio.2022.102809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 12/14/2022]
Abstract
Antibody-based therapeutics enjoy considerable clinical and commercial successes as cancer treatments. However, they can also cause serious toxicities due to recognition of tumor-associated antigens in noncancerous tissues, which can prevent antibody use in certain patient populations and therapeutic modalities. Here, we discuss recent efforts to develop advanced antibody therapeutics with activities restricted to the solid tumor microenvironment. With the intent of decreasing toxicities and expanding therapeutic windows, protein engineering strategies can render ligand binding sensitive to multiple tumor-specific characteristics. These triggers can be intrinsic to solid tumor microenvironments, such as low pH, high extracellular ATP, and the presence of specific proteases. Emerging strategies rely instead on exogenous triggers such as light and ultrasound to provide spatial and temporal control over antibody activation. These multilayered approaches to targeting diseased tissues are expected to usher in a new generation of precision therapeutics.
Collapse
Affiliation(s)
- Yutong Liu
- Department of Chemical Engineering, University of Texas, Austin, TX 78712, USA
| | - Annalee W Nguyen
- Department of Chemical Engineering, University of Texas, Austin, TX 78712, USA.
| | - Jennifer A Maynard
- Department of Chemical Engineering, University of Texas, Austin, TX 78712, USA.
| |
Collapse
|
16
|
Boustany LM, LaPorte SL, Wong L, White C, Vinod V, Shen J, Yu W, Koditek D, Winter MB, Moore SJ, Mei L, Diep L, Huang Y, Liu S, Vasiljeva O, West J, Richardson J, Irving B, Belvin M, Kavanaugh WM. A Probody T Cell-Engaging Bispecific Antibody Targeting EGFR and CD3 Inhibits Colon Cancer Growth with Limited Toxicity. Cancer Res 2022; 82:4288-4298. [PMID: 36112781 PMCID: PMC9664135 DOI: 10.1158/0008-5472.can-21-2483] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 05/09/2022] [Accepted: 09/14/2022] [Indexed: 01/07/2023]
Abstract
T cell-engaging bispecific antibodies (TCB) are highly potent therapeutics that can recruit and activate cytotoxic T cells to stimulate an antitumor immune response. However, the development of TCBs against solid tumors has been limited by significant on-target toxicity to normal tissues. Probody therapeutics have been developed as a novel class of recombinant, protease-activated antibody prodrugs that are "masked" to reduce antigen binding in healthy tissues but can become conditionally unmasked by proteases that are preferentially active in the tumor microenvironment (TME). Here, we describe the preclinical efficacy and safety of CI107, a Probody TCB targeting EGFR and CD3. In vitro, the protease-activated, unmasked CI107 effectively bound EGFR and CD3 expressed on the surface of cells and induced T-cell activation, cytokine release, and cytotoxicity toward tumor cells. In contrast, dually masked CI107 displayed a >500-fold reduction in antigen binding and >15,000-fold reduction in cytotoxic activity. In vivo, CI107 potently induced dose-dependent tumor regression of established colon cancer xenografts in mice engrafted with human peripheral blood mononuclear cells. Furthermore, the MTD of CI107 in cynomolgus monkeys was more than 60-fold higher than that of the unmasked TCB, and much lower levels of toxicity were observed in animals receiving CI107. Therefore, by localizing activity to the TME and thus limiting toxicity to normal tissues, this Probody TCB demonstrates the potential to expand clinical opportunities for TCBs as effective anticancer therapies for solid tumor indications. SIGNIFICANCE A conditionally active EGFR-CD3 T cell-engaging Probody therapeutic expands the safety window of bispecific antibodies while maintaining efficacy in preclinical solid tumor settings.
Collapse
Affiliation(s)
| | | | - Laurie Wong
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Clayton White
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Veena Vinod
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Joel Shen
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Wendy Yu
- CytomX Therapeutics, Inc, South San Francisco, California
| | - David Koditek
- CytomX Therapeutics, Inc, South San Francisco, California
| | | | | | - Li Mei
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Linnea Diep
- CytomX Therapeutics, Inc, South San Francisco, California
| | | | - Shouchun Liu
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Olga Vasiljeva
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Jim West
- CytomX Therapeutics, Inc, South San Francisco, California
| | | | - Bryan Irving
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Marcia Belvin
- CytomX Therapeutics, Inc, South San Francisco, California.,Corresponding Author: Marcia Belvin, CytomX Therapeutics, Inc, South San Francisco, CA 94080. Phone: 650-892-9803, E-mail:
| | | |
Collapse
|
17
|
Wei J, Yang Y, Wang G, Liu M. Current landscape and future directions of bispecific antibodies in cancer immunotherapy. Front Immunol 2022; 13:1035276. [PMID: 36389699 PMCID: PMC9650279 DOI: 10.3389/fimmu.2022.1035276] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/14/2022] [Indexed: 07/31/2023] Open
Abstract
Recent advances in cancer immunotherapy using monoclonal antibodies have dramatically revolutionized the therapeutic strategy against advanced malignancies, inspiring the exploration of various types of therapeutic antibodies. Bispecific antibodies (BsAbs) are recombinant molecules containing two different antigens or epitopes identifying binding domains. Bispecific antibody-based tumor immunotherapy has gained broad potential in preclinical and clinical investigations in a variety of tumor types following regulatory approval of newly developed technologies involving bispecific and multispecific antibodies. Meanwhile, a series of challenges such as antibody immunogenicity, tumor heterogeneity, low response rate, treatment resistance, and systemic adverse effects hinder the application of BsAbs. In this review, we provide insights into the various architecture of BsAbs, focus on BsAbs' alternative different mechanisms of action and clinical progression, and discuss relevant approaches to overcome existing challenges in BsAbs clinical application.
Collapse
Affiliation(s)
- Jing Wei
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yueyao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Gang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Ming Liu
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW T-cell-engaging antibodies or T-cell engagers (TCEs) can connect a patient's cytotoxic T cells with cancer cells, leading to potent redirected lysis. Until very recently, only one TCE was approved, the CD19/CD3-bispecific blinatumomab. Many new TCEs in late-stage clinical development target various hematopoietic lineage markers like CD20, BCMA, or CD123. Although very compelling single-agent activity of TCEs was observed with various blood-borne cancers, therapy of solid tumor indications has thus far been less successful. RECENT FINDINGS The approval in 2022 of the gp100 peptide-major histocompatibility complex (MHC)/CD3 bispecific TCE tebentafusp in uveal melanoma confirms that TCEs can also efficiently work against solid tumors. TCEs targeting peptide-MHC complexes will expand the target space for solid tumor therapy to intracellular targets. Likewise, early clinical trial data from TCEs targeting DLL3 in small cell lunger cancer showed promising antitumor activity. Various technologies for conditional activation of TCEs in the tumor microenvironment (TME) may expand the scope of conventional surface targets that suffer from a narrow therapeutic window. Finally, pharmacological enhancements for TCE therapies by engagement of certain costimulatory receptors and cytokines, or blockade of checkpoints, are showing promise. SUMMARY Targeting peptide-MHC complexes, conditional TCE technologies, and concepts enhancing TCE-activated T cells are paving the way towards overcoming challenges associated with solid tumor therapy.
Collapse
|
19
|
Avanzino BC, Prabhakar K, Dalvi P, Hartstein S, Kehm H, Balasubramani A, Boudreau AA, Buelow B, Chang K, Davison LM, Iyer S, Kalwit V, Lewis Wilson K, Malik-Chaudhry HK, Pierson W, Pineda G, Rangaswamy US, Saiganesh S, Schellenberger U, Ugamraj HS, Yabut RD, Buelow R, Chapman J, Trinklein ND, Harris KE. A T-cell engaging bispecific antibody with a tumor-selective bivalent folate receptor alpha binding arm for the treatment of ovarian cancer. Oncoimmunology 2022; 11:2113697. [PMID: 36016696 PMCID: PMC9397469 DOI: 10.1080/2162402x.2022.2113697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 07/20/2022] [Accepted: 08/11/2022] [Indexed: 11/22/2022] Open
Abstract
The use of T-cell engagers (TCEs) to treat solid tumors is challenging, and several have been limited by narrow therapeutic windows due to substantial on-target, off-tumor toxicities due to the expression of low levels of target antigens on healthy tissues. Here, we describe TNB-928B, a fully human TCE that has a bivalent binding arm for folate receptor alpha (FRα) to selectively target FRα overexpressing tumor cells while avoiding the lysis of cells with low levels of FRα expression. The bivalent design of the FRα binding arm confers tumor selectivity due to low-affinity but high-avidity binding to high FRα antigen density cells. TNB-928B induces preferential effector T-cell activation, proliferation, and selective cytotoxic activity on high FRα expressing cells while sparing low FRα expressing cells. In addition, TNB-928B induces minimal cytokine release compared to a positive control TCE containing OKT3. Moreover, TNB-928B exhibits substantial ex vivo tumor cell lysis using endogenous T-cells and robust tumor clearance in vivo, promoting T-cell infiltration and antitumor activity in mouse models of ovarian cancer. TNB-928B exhibits pharmacokinetics similar to conventional antibodies, which are projected to enable favorable administration in humans. TNB-928B is a novel TCE with enhanced safety and specificity for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Brian C. Avanzino
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| | - Kirthana Prabhakar
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| | - Pranjali Dalvi
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| | - Sharon Hartstein
- Teneobio, Inc, Newark, CA, United States
- Therapeutic Discovery, Amgen Inc., Newark, CA, USA
| | | | - Aarti Balasubramani
- Teneobio, Inc, Newark, CA, United States
- Therapeutic Discovery, Amgen Inc., Newark, CA, USA
| | | | - Ben Buelow
- Teneobio, Inc, Newark, CA, United States
| | | | | | | | - Vidyut Kalwit
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| | - Kristin Lewis Wilson
- Translational Safety & Bioanalytical Sciences, Amgen Inc., South San Francisco, CA, USA
| | | | - Will Pierson
- Division of Gynecologic Oncology, University of California, San Francisco, CA, USA
- University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Geovanni Pineda
- Division of Gynecologic Oncology, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Udaya S. Rangaswamy
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| | - Sowmya Saiganesh
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| | | | - Harshad S. Ugamraj
- Teneobio, Inc, Newark, CA, United States
- Process Development, Amgen Inc., Newark, CA, USA
| | - Rodolfovan D. Yabut
- Translational Safety & Bioanalytical Sciences, Amgen Inc., South San Francisco, CA, USA
| | | | - Jocelyn Chapman
- Division of Gynecologic Oncology, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | | | - Katherine E. Harris
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| |
Collapse
|
20
|
Designing antibodies as therapeutics. Cell 2022; 185:2789-2805. [PMID: 35868279 DOI: 10.1016/j.cell.2022.05.029] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/18/2022] [Accepted: 05/31/2022] [Indexed: 12/25/2022]
Abstract
Antibody therapeutics are a large and rapidly expanding drug class providing major health benefits. We provide a snapshot of current antibody therapeutics including their formats, common targets, therapeutic areas, and routes of administration. Our focus is on selected emerging directions in antibody design where progress may provide a broad benefit. These topics include enhancing antibodies for cancer, antibody delivery to organs such as the brain, gastrointestinal tract, and lungs, plus antibody developability challenges including immunogenicity risk assessment and mitigation and subcutaneous delivery. Machine learning has the potential, albeit as yet largely unrealized, for a transformative future impact on antibody discovery and engineering.
Collapse
|
21
|
Chen TT. Conditionally active T cell engagers for the treatment of solid tumors: rationale and clinical development. Expert Opin Biol Ther 2022; 22:955-963. [PMID: 35857922 DOI: 10.1080/14712598.2022.2098674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION T cell engagers are a class of bispecific molecules that induce highly potent T cell-dependent cytotoxicity by bringing T cell activating receptors into proximity with cancer-associated cell surface antigens. However, because of their high potency, there is a greater risk of on-target/off-tumor toxicity owing to normal tissues having tumor antigen expression even at low levels. To reduce these adverse events, the dysregulated activity of proteases within the tumor microenvironment has recently been explored to create inert prodrugs that become conditionally active engagers after their cleavage by these enzymes. AREAS COVERED T-cell engagers that have been introduced for clinical use, and their respective successes and failures are reviewed. The unique challenges of these bispecific molecules for treating solid tumors and prior technologies used to exploit the proteolytic tumor microenvironment to create better-tolerated prodrugs and how that experience has led to the current series of conditionally active T-cell engagers, are discussed. EXPERT OPINION Methods for modulating the serum half-life of both inert and activated T cell engagers could have important ramifications in how they infiltrate tumors and prevent toxicity. Alternative features of the tumor microenvironment can also be leveraged in the development of conditional T cell engagers.
Collapse
Affiliation(s)
- T Timothy Chen
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Development Center Americas, Inc
| |
Collapse
|
22
|
Li M, Mei S, Yang Y, Shen Y, Chen L. Strategies to mitigate the on- and off-target toxicities of recombinant immunotoxins: an antibody engineering perspective. Antib Ther 2022; 5:164-176. [PMID: 35928456 PMCID: PMC9344849 DOI: 10.1093/abt/tbac014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/14/2021] [Accepted: 06/14/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Targeted cancer therapies using immunotoxins has achieved remarkable efficacies in hematological malignancies. However, the clinical development of immunotoxins is also faced with many challenges like anti-drug antibodies and dose-limiting toxicity issues. Such a poor efficacy/safety ratio is also the major hurdle in the research and development of antibody-drug conjugates. From an antibody engineering perspective, various strategies were summarized/proposed to tackle the notorious on target off tumor toxicity issues, including passive strategy (XTENylation of immunotoxins) and active strategies (modulating the affinity and valency of the targeting moiety of immunotoxins, conditionally activating immunotoxins in the tumor microenvironments and reconstituting split toxin to reduce systemic toxicity etc.). By modulating the functional characteristics of the targeting moiety and the toxic moiety of immunotoxins, selective tumor targeting can be augmented while sparing the healthy cells in normal tissues expressing the same target of interest. If successful, the improved therapeutic index will likely help to address the dose-limiting toxicities commonly observed in the clinical trials of various immunotoxins.
Collapse
Affiliation(s)
- Mengyu Li
- Department of Postgraduate , Jiangxi University of Traditional Chinese Medicine, Nanchang, P.R. China
- Joint Graduate School , Yangtze Delta Drug Advanced Research Institute, Nantong, P.R. China
- Joint Graduate School , Yangtze Delta Pharmaceutical College, Nantong, P.R. China
| | - Sen Mei
- Biotherapeutics , Biocytogen Jiangsu Co. Ltd, Nantong, P.R. China
| | - Yi Yang
- Joint Graduate School , Yangtze Delta Drug Advanced Research Institute, Nantong, P.R. China
- Joint Graduate School , Yangtze Delta Pharmaceutical College, Nantong, P.R. China
- Institute of Innovative Medicine , Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing, P.R. China
| | - Yuelei Shen
- Joint Graduate School , Yangtze Delta Drug Advanced Research Institute, Nantong, P.R. China
- Joint Graduate School , Yangtze Delta Pharmaceutical College, Nantong, P.R. China
- Biotherapeutics , Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing, P.R. China
- Institute of Innovative Medicine , Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing, P.R. China
| | - Lei Chen
- Biotherapeutics , Biocytogen Jiangsu Co. Ltd, Nantong, P.R. China
- Biotherapeutics , Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing, P.R. China
| |
Collapse
|
23
|
Exploiting protease activation for therapy. Drug Discov Today 2022; 27:1743-1754. [PMID: 35314338 PMCID: PMC9132161 DOI: 10.1016/j.drudis.2022.03.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/14/2022] [Accepted: 03/15/2022] [Indexed: 02/08/2023]
Abstract
Proteases have crucial roles in homeostasis and disease; and protease inhibitors and recombinant proteases in enzyme replacement therapy have become key therapeutic applications of protease biology across several indications. This review briefly summarises therapeutic approaches based on protease activation and focuses on how recent insights into the spatial and temporal control of the proteolytic activation of growth factors and interleukins are leading to unique strategies for the discovery of new medicines. In particular, two emerging areas are covered: the first is based on antibody therapies that target the process of proteolytic activation of the pro-form of proteins rather than their mature form; the second covers a potentially new class of biopharmaceuticals using engineered, proteolytically activable and initially inactive pro-forms of antibodies or effector proteins to increase specificity and improve the therapeutic window.
Collapse
|
24
|
Dettling DE, Kwok E, Quach L, Datt A, Degenhardt JD, Panchal A, Seto P, Krakow JL, Wall R, Hillier BJ, Zhu Y, Vinogradova M, DuBridge RB, May C. Regression of EGFR positive established solid tumors in mice with the conditionally active T cell engager TAK-186. J Immunother Cancer 2022; 10:jitc-2021-004336. [PMID: 35728872 PMCID: PMC9214390 DOI: 10.1136/jitc-2021-004336] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2022] [Indexed: 11/16/2022] Open
Abstract
Background Despite clinical success with T cell engagers (TCEs) targeting hematological malignancies, achieving a safe and efficacious dose in patients with solid tumors remains challenging. Due to potency, low levels of target antigen expression on normal tissues may not be tolerated. To overcome this, we engineered a novel conditionally active TCE design called COBRA (Conditional Bispecific Redirected Activation). Administered as prodrugs, COBRAs bind to cell surface antigens on both normal and tumor tissues but are preferentially activated within the tumor microenvironment. Methods A COBRA was engineered to target EGFR, TAK-186. The potency of precleaved TAK-186 relative to a non-cleavable control was assessed in vitro. Mice bearing established solid tumors expressing a range of EGFR levels were administered a single bolus of human T cells, and concurrently treated with TAK-186 and associated controls intravenously. We assessed the plasma and tumor exposure of intact and cleaved TAK-186. Results TAK-186 shows potent redirected T cell killing of antigen expressing tumor cells. In vivo efficacy studies demonstrate regressions of established solid tumors, dependent on intratumoral COBRA cleavage. Pharmacokinetic studies reveal TAK-186 is stable in circulation, but once activated is rapidly cleared due to loss of its albumin-binding half-life extension domain. Conclusions The studies shown support the advancement of TAK-186, and the pursuit of additional COBRA TCEs for the treatment of solid tumors.
Collapse
Affiliation(s)
- Danielle E Dettling
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Eilene Kwok
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Lucy Quach
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Aakash Datt
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Jeremiah D Degenhardt
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Anand Panchal
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Pui Seto
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Jessica L Krakow
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Russell Wall
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Brian J Hillier
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Ying Zhu
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Maia Vinogradova
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Robert B DuBridge
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Chad May
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| |
Collapse
|
25
|
Chai AWY, Yee PS, Cheong SC. Rational Combinations of Targeted Therapy and Immune Checkpoint Inhibitors in Head and Neck Cancers. Front Oncol 2022; 12:837835. [PMID: 35372020 PMCID: PMC8968950 DOI: 10.3389/fonc.2022.837835] [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: 12/17/2021] [Accepted: 02/17/2022] [Indexed: 12/21/2022] Open
Abstract
Immunotherapy, especially the immune checkpoint inhibitors (ICIs) such as the pembrolizumab and nivolumab have contributed to significant improvements in treatment outcomes and survival of head and neck cancer (HNC) patients. Still, only a subset of patients benefits from ICIs and hence the race is on to identify combination therapies that could improve response rates. Increasingly, genetic alterations that occur within cancer cells have been shown to modulate the tumor microenvironment resulting in immune evasion, and these have led to the emergence of trials that rationalize a combination of targeted therapy with immunotherapy. In this review, we aim to provide an overview of the biological rationale and current strategies of combining targeted therapy with the approved ICIs in HNC. We summarize the ongoing combinatorial clinical trials and discuss emerging immunomodulatory targets. We also discuss the challenges and gaps that have yet to be addressed, as well as future perspectives in combining these different drug classes.
Collapse
Affiliation(s)
- Annie Wai Yeeng Chai
- Translational Cancer Biology Research Unit, Cancer Research Malaysia, Subang Jaya, Malaysia
| | - Pei San Yee
- Translational Cancer Biology Research Unit, Cancer Research Malaysia, Subang Jaya, Malaysia
| | - Sok Ching Cheong
- Translational Cancer Biology Research Unit, Cancer Research Malaysia, Subang Jaya, Malaysia
- Department of Oral and Maxillofacial Clinical Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
26
|
Bagheri A, Culp PA, DuBridge RB, Chen THT. CRISPR/Cas9 disruption of EpCAM Exon 2 results in cell-surface expression of a truncated protein targeted by an EpCAM specific T cell engager. Biochem Biophys Rep 2022; 29:101205. [PMID: 35071801 PMCID: PMC8761601 DOI: 10.1016/j.bbrep.2022.101205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/24/2021] [Accepted: 01/06/2022] [Indexed: 11/02/2022] Open
Abstract
CRISPR/Cas9 gene-editing technology allows researchers to study protein function by specifically introducing double-stranded breaks in the gene of interest then analyze its subsequent loss in sensitive biological assays. To help characterize one of a series of highly potent, conditionally active, T cell engaging bispecific molecules called COBRA™, the human EpCAM gene was disrupted in HT29 cells using CRISPR/Cas9 and guide RNA targeting its Exon 2. Although a commercially available antibody indicated loss of cell-surface expression, the EpCAM targeting bispecific COBRA was still able to lyse these cells in a T cell dependent cellular cytotoxicity assay. RT-PCR sequence analysis of these cells showed a major alternative transcript generated after CRISPR/Cas9, with Exon 1 and 3 spliced together in-frame, skipping Exon 2 completely, to express a truncated cell-surface receptor recognized by the EpCAM-COBRA. Researchers who use CRISPR/Cas9 must be cognizant of this potential to express alternative versions of their proteins and use sensitive orthogonal detection methods to ensure complete gene disruption. CRISPR/Cas9 allows researchers to target gene disruption and analyze its loss of expression. After CRISPR/Cas9 targeting the EpCAM gene, no protein was detected after staining with a commercial antibody. An in-house EpCAM T-cell engager was able to lyse these supposedly EpCAM KO cells. cDNA analysis showed an alternative mRNA transcript arose after CRISPR/Cas9, creating a truncated version on the cell's surface. Scientists using CRISPR must be aware of the potential to make alternate forms and try other means to ensure no expression.
Collapse
|
27
|
Dickopf S, Buldun C, Vasic V, Georges G, Hage C, Mayer K, Forster M, Wessels U, Stubenrauch KG, Benz J, Ehler A, Lauer ME, Ringler P, Kobold S, Endres S, Klein C, Brinkmann U. Prodrug-Activating Chain Exchange (PACE) converts targeted prodrug derivatives to functional bi- or multispecific antibodies. Biol Chem 2022; 403:495-508. [PMID: 35073465 PMCID: PMC9125802 DOI: 10.1515/hsz-2021-0401] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/23/2021] [Indexed: 12/26/2022]
Abstract
Driven by the potential to broaden the target space of conventional monospecific antibodies, the field of multi-specific antibody derivatives is growing rapidly. The production and screening of these artificial proteins entails a high combinatorial complexity. Antibody-domain exchange was previously shown to be a versatile strategy to produce bispecific antibodies in a robust and efficient manner. Here, we show that the domain exchange reaction to generate hybrid antibodies also functions under physiological conditions. Accordingly, we modified the exchange partners for use in therapeutic applications, in which two inactive prodrugs convert into a product with additional functionalities. We exemplarily show the feasibility for generating active T cell bispecific antibodies from two inactive prodrugs, which per se do not activate T cells alone. The two complementary prodrugs harbor antigen-targeting Fabs and non-functional anti-CD3 Fvs fused to IgG-CH3 domains engineered to drive chain-exchange reactions between them. Importantly, Prodrug-Activating Chain Exchange (PACE) could be an attractive option to conditionally activate therapeutics at the target site. Several examples are provided that demonstrate the efficacy of PACE as a new principle of cancer immunotherapy in vitro and in a human xenograft model.
Collapse
Affiliation(s)
- Steffen Dickopf
- Large Molecule Research (LMR), Roche Innovation Center Munich , Roche Pharma Research and Early Development (pRED) , Penzberg , Germany
| | - Can Buldun
- Large Molecule Research (LMR), Roche Innovation Center Munich , Roche Pharma Research and Early Development (pRED) , Penzberg , Germany
| | - Vedran Vasic
- Large Molecule Research (LMR), Roche Innovation Center Munich , Roche Pharma Research and Early Development (pRED) , Penzberg , Germany
| | - Guy Georges
- Large Molecule Research (LMR), Roche Innovation Center Munich , Roche Pharma Research and Early Development (pRED) , Penzberg , Germany
| | - Carina Hage
- Discovery Oncology, Roche Innovation Center Munich , Roche Pharma Research and Early Development (pRED) , Penzberg , Germany
| | - Klaus Mayer
- Large Molecule Research (LMR), Roche Innovation Center Munich , Roche Pharma Research and Early Development (pRED) , Penzberg , Germany
| | - Matthias Forster
- Large Molecule Research (LMR), Roche Innovation Center Munich , Roche Pharma Research and Early Development (pRED) , Penzberg , Germany
| | - Uwe Wessels
- Pharmaceutical Sciences (PS), Roche Innovation Center Munich , Roche Pharma Research and Early Development (pRED) , Penzberg , Germany
| | - Kay-Gunnar Stubenrauch
- Pharmaceutical Sciences (PS), Roche Innovation Center Munich , Roche Pharma Research and Early Development (pRED) , Penzberg , Germany
| | - Jörg Benz
- Small Molecule Research, Roche Innovation Center Basel , Roche Pharma Research and Early Development (pRED) , Basel , Switzerland
| | - Andreas Ehler
- Small Molecule Research, Roche Innovation Center Basel , Roche Pharma Research and Early Development (pRED) , Basel , Switzerland
| | - Matthias E. Lauer
- Chemical Biology, Roche Innovation Center Basel , Roche Pharma Research and Early Development (pRED) , Basel , Switzerland
| | - Philippe Ringler
- Center for Cellular Imaging and Nano Analytics , Biozentrum University of Basel , Basel , Switzerland
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV , University Hospital, Ludwig Maximilians University of Munich, German Center for Lung Research (DZL) , Munich , Germany
- German Center for Translational Cancer Research (DKTK) , Partner Site Munich , Munich , Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV , University Hospital, Ludwig Maximilians University of Munich, German Center for Lung Research (DZL) , Munich , Germany
- German Center for Translational Cancer Research (DKTK) , Partner Site Munich , Munich , Germany
| | - Christian Klein
- Discovery Oncology, Roche Innovation Center Zurich , Roche Pharma Research and Early Development (pRED) , Schlieren , Switzerland
| | - Ulrich Brinkmann
- Large Molecule Research (LMR), Roche Innovation Center Munich , Roche Pharma Research and Early Development (pRED) , Penzberg , Germany
| |
Collapse
|
28
|
Krakow J, Hammel M, Zhu Y, Hillier BJ, Paolella B, Desmarais A, Wall R, Chen THT, Pei R, Karunatilake C, DuBridge R, Vinogradova M. Structural arrangement of the VH and VL domains in the COBRA™ T-cell engaging single-chain diabody. Antib Ther 2022; 5:1-10. [PMID: 35005430 PMCID: PMC8719580 DOI: 10.1093/abt/tbab028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND COBRA™ (COnditional Bispecific Redirected Activation) T-cell engagers are designed to target solid tumors as a single polypeptide chain prodrug that becomes activated by proteolysis in the tumor microenvironment. One COBRA molecule comprises seven Ig domains: three single-domain antibodies (sdAbs) recognizing a tumor target or human serum albumin (HSA), and CD3ε-binding variable fragment heavy chain (VH) and variable fragment light chain (VL) and their inactivated counterparts, VHi and VLi. Pairing of VH and VL, and VLi and VHi into single-chain variable fragments (Fv) is prevented by shortened inter-domain linkers. Instead, VH and VL are expected to interact with VLi and VHi, respectively, thus making a diabody whose binding to CD3ε on the T-cells is impaired. METHODS We analyzed the structure of an epidermal growth factor receptor (EGFR) COBRA in solution using negative stain electron microscopy (EM) and small-angle X-ray scattering (SAXS). RESULTS We found that this EGFR COBRA forms stable monomers with a very dynamic interdomain arrangement. At most, only five domains at a time appeared ordered, and only one VH-VL pair was found in the Fv orientation. Nonenzymatic posttranslational modifications suggest that the CDR3 loops in the VL-VHi pair are exposed but are buried in the VH-VLi pair. The MMP9 cleavage rate of the prodrug when bound to recombinant EGFR or HSA is not affected, indicating positioning of the MMP9-cleavable linker away from the EGFR and HSA binding sites. CONCLUSION Here, we propose a model for EGFR COBRA where VH and VLi form an Fv, and VL and VHi do not, possibly interacting with other Ig domains. SAXS and MMP9 cleavage analyses suggest that all COBRA molecules tested have a similar structural architecture.
Collapse
Affiliation(s)
- Jessica Krakow
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Brisbane, CA, USA
| | - Michal Hammel
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Ying Zhu
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Brisbane, CA, USA
| | - Brian J Hillier
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Brisbane, CA, USA
| | - Bryce Paolella
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Brisbane, CA, USA
| | - Austin Desmarais
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Brisbane, CA, USA
| | - Rusty Wall
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Brisbane, CA, USA
| | - Tseng-Hui T Chen
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Brisbane, CA, USA
| | - Rex Pei
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Brisbane, CA, USA
| | - Chulani Karunatilake
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Brisbane, CA, USA
| | - Robert DuBridge
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Brisbane, CA, USA
| | - Maia Vinogradova
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Brisbane, CA, USA
| |
Collapse
|
29
|
Orozco CT, Bersellini M, Irving LM, Howard WW, Hargreaves D, Devine PWA, Siouve E, Browne GJ, Bond NJ, Phillips JJ, Ravn P, Jackson SE. Mechanistic insights into the rational design of masked antibodies. MAbs 2022; 14:2095701. [PMID: 35799328 PMCID: PMC9272835 DOI: 10.1080/19420862.2022.2095701] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Although monoclonal antibodies have greatly improved cancer therapy, they can trigger side effects due to on-target, off-tumor toxicity. Over the past decade, strategies have emerged to successfully mask the antigen-binding site of antibodies, such that they are only activated at the relevant site, for example, after proteolytic cleavage. However, the methods for designing an ideal affinity-based mask and what parameters are important are not yet well understood. Here, we undertook mechanistic studies using three masks with different properties and identified four critical factors: binding site and affinity, as well as association and dissociation rate constants, which also played an important role. HDX-MS was used to identify the location of binding sites on the antibody, which were subsequently validated by obtaining a high-resolution crystal structure for one of the mask-antibody complexes. These findings will inform future designs of optimal affinity-based masks for antibodies and other therapeutic proteins.
Collapse
Affiliation(s)
- Carolina T Orozco
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.,Biologics Engineering, R&D, AstraZeneca, Cambridge, UK.,Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | | | | | - Wesley W Howard
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Paul W A Devine
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | - Elise Siouve
- Biologics Engineering, R&D, AstraZeneca, Cambridge, UK.,Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | | | - Nicholas J Bond
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | | | - Peter Ravn
- Biologics Engineering, R&D, AstraZeneca, Cambridge, UK.,Department of Biotherapeutic Discovery, H. Lundbeck A/S, Copenhagen, Denmark
| | - Sophie E Jackson
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
30
|
Crawford A, Chiu D. Targeting Solid Tumors Using CD3 Bispecific Antibodies. Mol Cancer Ther 2021; 20:1350-1358. [PMID: 34045228 DOI: 10.1158/1535-7163.mct-21-0073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/06/2021] [Accepted: 05/24/2021] [Indexed: 11/16/2022]
Abstract
Immunotherapies to treat cancer have made tremendous progress over the past decade. In particular, T cell-directed therapies have gained considerable attention with CD3 bispecific antibodies and CAR T cells showing potent responses against hematologic tumors. At present, the ability to adapt these therapeutics to treat solid tumors is less established. Herein, we discuss recent advances in T cell-engaging CD3 bispecific antibodies targeting solid tumors, potential mechanisms of resistance, and future prospects. A better understanding of the mechanisms of immune evasion in solid tumors will enable the development of strategies to overcome this resistance and inform choices of therapeutic combinations.
Collapse
Affiliation(s)
| | - Danica Chiu
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| |
Collapse
|
31
|
Lucchi R, Bentanachs J, Oller-Salvia B. The Masking Game: Design of Activatable Antibodies and Mimetics for Selective Therapeutics and Cell Control. ACS CENTRAL SCIENCE 2021; 7:724-738. [PMID: 34079893 PMCID: PMC8161478 DOI: 10.1021/acscentsci.0c01448] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Indexed: 05/04/2023]
Abstract
The high selectivity and affinity of antibody binding have made antibodies all-pervasive tools in therapy, diagnosis, and basic science. A plethora of chemogenetic approaches has been devised to make antibodies responsive to stimuli ranging from light to enzymatic activity, temperature, pH, ions, and effector molecules. Within a single decade, the field of activatable antibodies has yielded marketed therapeutics capable of engaging antigens that could not be targeted with traditional antibodies, as well as new tools to control intracellular protein location and investigate biological processes. Many opportunities remain untapped, waiting for more efficient and generally applicable masking strategies to be developed at the interface between chemistry and biotechnology.
Collapse
Affiliation(s)
- Roberta Lucchi
- Grup d’Enginyeria
de Materials, Institut Químic de
Sarrià (IQS), Universitat Ramon Llull, 08017 Barcelona, Spain
| | - Jordi Bentanachs
- Grup d’Enginyeria
de Materials, Institut Químic de
Sarrià (IQS), Universitat Ramon Llull, 08017 Barcelona, Spain
| | - Benjamí Oller-Salvia
- Grup d’Enginyeria
de Materials, Institut Químic de
Sarrià (IQS), Universitat Ramon Llull, 08017 Barcelona, Spain
| |
Collapse
|
32
|
Singh A, Dees S, Grewal IS. Overcoming the challenges associated with CD3+ T-cell redirection in cancer. Br J Cancer 2021; 124:1037-1048. [PMID: 33469153 PMCID: PMC7960983 DOI: 10.1038/s41416-020-01225-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/24/2020] [Accepted: 11/30/2020] [Indexed: 01/30/2023] Open
Abstract
The development of bispecific antibodies that redirect the cytotoxic activity of CD3+ T cells to tumours is a promising immunotherapeutic strategy for the treatment of haematological malignancies and solid cancers. Since the landmark FDA approval at the end of 2014 of the anti-CD3 × anti-CD19 bispecific antibody blinatumomab (Blincyto®) for the treatment of relapsed/refractory B-cell acute lymphoblastic leukaemia, ~100 clinical trials investigating the safety and efficacy of CD3+ bispecific T-cell redirectors for cancer have been initiated. However, despite early success, numerous challenges pertaining to CD3+ T-cell redirection in the context of cancer exist, including the recruitment of counterproductive CD3+ T-cell subsets, the release of systemic cytokines, the expansion of immune checkpoint molecules, the presence of an immunosuppressive tumour microenvironment, tumour antigen loss/escape, on-target off-tumour toxicity and suboptimal potency. The aim of the present review is to discuss novel approaches to overcome the key challenges associated with CD3+ bispecific T-cell redirection in order to achieve an optimal balance of anti-tumour activity and safety.
Collapse
Affiliation(s)
- Ajit Singh
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sundee Dees
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA, USA
| | - Iqbal S Grewal
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA, USA.
| |
Collapse
|
33
|
Brouillard A, Deshpande N, Kulkarni AA. Engineered Multifunctional Nano- and Biological Materials for Cancer Immunotherapy. Adv Healthc Mater 2021; 10:e2001680. [PMID: 33448159 DOI: 10.1002/adhm.202001680] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/21/2020] [Indexed: 12/19/2022]
Abstract
Cancer immunotherapy is set to emerge as the future of cancer therapy. However, recent immunotherapy trials in different cancers have yielded sub-optimal results, with durable responses seen in only a small fraction of patients. Engineered multifunctional nanomaterials and biological materials are versatile platforms that can elicit strong immune responses and improve anti-cancer efficacy when applied to cancer immunotherapy. While there are traditional systems such as polymer- and lipid-based nanoparticles, there is a wide variety of other materials with inherent and additive properties that can allow for more potent activation of the immune system. By synthesizing and applying multifunctional strategies, it allows for a more extensive and more effective repertoire of tools to use in the wide variety of situations that cancer presents itself. Here, several types of nanoscale and biological material strategies and platforms that provide their inherent benefits for targeting and activating multiple aspects of the immune system are discussed. Overall, this review aims to provide a comprehensive understanding of recent advances in the field of multifunctional cancer immunotherapy and trends that pave the way for more diverse and tactical regression of tumors through soliciting responses by either the adaptive or innate immune system, and even both simultaneously.
Collapse
Affiliation(s)
- Anthony Brouillard
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
| | - Nilesh Deshpande
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
| | - Ashish A. Kulkarni
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
- Center for Bioactive Delivery Institute for Applied Life Sciences University of Massachusetts Amherst MA 01003 USA
| |
Collapse
|
34
|
Middelburg J, Kemper K, Engelberts P, Labrijn AF, Schuurman J, van Hall T. Overcoming Challenges for CD3-Bispecific Antibody Therapy in Solid Tumors. Cancers (Basel) 2021; 13:287. [PMID: 33466732 PMCID: PMC7829968 DOI: 10.3390/cancers13020287] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy of cancer with CD3-bispecific antibodies is an approved therapeutic option for some hematological malignancies and is under clinical investigation for solid cancers. However, the treatment of solid tumors faces more pronounced hurdles, such as increased on-target off-tumor toxicities, sparse T-cell infiltration and impaired T-cell quality due to the presence of an immunosuppressive tumor microenvironment, which affect the safety and limit efficacy of CD3-bispecific antibody therapy. In this review, we provide a brief status update of the CD3-bispecific antibody therapy field and identify intrinsic hurdles in solid cancers. Furthermore, we describe potential combinatorial approaches to overcome these challenges in order to generate selective and more effective responses.
Collapse
Affiliation(s)
- Jim Middelburg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Kristel Kemper
- Genmab, 3584 CT Utrecht, The Netherlands; (K.K.); (P.E.); (A.F.L.); (J.S.)
| | - Patrick Engelberts
- Genmab, 3584 CT Utrecht, The Netherlands; (K.K.); (P.E.); (A.F.L.); (J.S.)
| | - Aran F. Labrijn
- Genmab, 3584 CT Utrecht, The Netherlands; (K.K.); (P.E.); (A.F.L.); (J.S.)
| | - Janine Schuurman
- Genmab, 3584 CT Utrecht, The Netherlands; (K.K.); (P.E.); (A.F.L.); (J.S.)
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| |
Collapse
|