1
|
El-Khoueiry A, Saavedra O, Thomas J, Livings C, Garralda E, Hintzen G, Kohlhas L, Vanosmael D, Koch J, Rajkovic E, Ravenstijn P, Nuciforo P, Fehniger TA, Foster M, Berrien-Elliott MM, Wingert S, Stäble S, Morales-Espinosa D, Rivas D, Emig M, Lopez J. First-in-Human Phase I Study of a CD16A Bispecific Innate Cell Engager, AFM24, Targeting EGFR-Expressing Solid Tumors. Clin Cancer Res 2025; 31:1257-1267. [PMID: 39846810 PMCID: PMC11964176 DOI: 10.1158/1078-0432.ccr-24-1991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/02/2024] [Accepted: 01/21/2025] [Indexed: 01/24/2025]
Abstract
PURPOSE Innate immune cell-based therapies have shown promising antitumor activity against solid and hematologic malignancies. AFM24, a bispecific innate cell engager, binds CD16A on NK cells/macrophages and EGFR on tumor cells, redirecting antitumor activity toward tumors. The safety and tolerability of AFM24 were evaluated in this phase I/IIa dose-escalation/dose-expansion study in patients with recurrent or persistent, advanced solid tumors known to express EGFR. PATIENTS AND METHODS The main objective in phase I was to determine the MTD and/or recommended phase II dose. The primary endpoint was the incidence of dose-limiting toxicities during the observation period. Secondary endpoints included the incidence of treatment-emergent adverse events and pharmacokinetics. RESULTS In the dose-escalation phase, 35 patients received AFM24 weekly across seven dose cohorts (14-720 mg). One patient experienced a dose-limiting toxicity of grade 3 infusion-related reaction. Infusion-related reactions were mainly reported after the first infusion; these were manageable with premedication and a gradual increase in infusion rate. Pharmacokinetics was dose-proportional, and CD16A receptor occupancy on NK cells approached saturation between 320 and 480 mg. Paired tumor biopsies demonstrated the activation of innate and adaptive immune responses within the tumor. The best objective response was stable disease in 10/35 patients; four patients had stable disease for 4.3 to 7.1 months. CONCLUSIONS AFM24 was well tolerated, with 480 mg established as the recommended phase II dose. AFM24 could be a novel therapy for patients with EGFR-expressing solid tumors, with suitable tolerability and appropriate pharmacokinetic properties for further development in combination with other immuno-oncology therapeutics.
Collapse
Affiliation(s)
- Anthony El-Khoueiry
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Omar Saavedra
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Barcelona, Spain
- Departamento de Medicina, Universidad Autónoma de Barcelona (UAB), Barcelona, Spain
| | - Jacob Thomas
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Claire Livings
- Drug Development Unit, Royal Marsden NHS Foundation Trust and the Institute of Cancer Research, Sutton, United Kingdom
| | - Elena Garralda
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Barcelona, Spain
| | | | | | | | | | | | | | - Paolo Nuciforo
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Barcelona, Spain
| | - Todd A. Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Mark Foster
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Melissa M. Berrien-Elliott
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | | | | | | | | | | | - Juanita Lopez
- Drug Development Unit, Royal Marsden NHS Foundation Trust and the Institute of Cancer Research, Sutton, United Kingdom
| |
Collapse
|
2
|
Sánchez-Gaona N, Perea D, Curran A, Burgos J, Navarro J, Suanzes P, Falcó V, Martín-Gayo E, Genescà M, Carrillo J, Buzón MJ. NK cell depletion in bispecific antibody therapy is associated with lack of HIV control after ART interruption. Commun Biol 2025; 8:236. [PMID: 39953264 PMCID: PMC11829058 DOI: 10.1038/s42003-025-07651-6] [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: 09/04/2024] [Accepted: 01/31/2025] [Indexed: 02/17/2025] Open
Abstract
HIV infection remains incurable as the virus persists within a latent reservoir of CD4+T cells. Novel approaches to enhance immune responses against HIV are essential for effective control and potential cure of the infection. In this study, we designed a novel tetravalent bispecific antibody (Bi-Ab32/16) to simultaneously target the gp120 viral protein on infected cells, and the CD16a receptor on NK cells. In vitro, Bi-Ab32/16 triggered a potent, specific, and polyfunctional NK-dependent response against HIV-infected cells. Moreover, addition of the Bi-Ab32/16 significantly reduced the latent HIV reservoir after viral reactivation and mediated the clearance of cells harboring intact proviruses in samples from people with HIV (PWH). However, the in vivo preclinical evaluation of Bi-Ab32/16 in humanized mice expressing IL-15 (NSG-Hu-IL-15) revealed a significant decline of NK cells associated with poor virological control after ART interruption. Our study underscores the need to carefully evaluating strategies for sustained NK cell stimulation during ART withdrawal.
Collapse
Affiliation(s)
- N Sánchez-Gaona
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - D Perea
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - A Curran
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - J Burgos
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - J Navarro
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - P Suanzes
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - V Falcó
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - E Martín-Gayo
- Universidad Autónoma de Madrid, Immunology Unit, Hospital Universitario de la Princesa, Madrid, Spain
- CIBERINFEC. ISCIII, Madrid, Spain
| | - M Genescà
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - J Carrillo
- CIBERINFEC. ISCIII, Madrid, Spain
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - M J Buzón
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
3
|
In H, Park M, Lee H, Han KH. Immune Cell Engagers: Advancing Precision Immunotherapy for Cancer Treatment. Antibodies (Basel) 2025; 14:16. [PMID: 39982231 PMCID: PMC11843982 DOI: 10.3390/antib14010016] [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: 01/17/2025] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
Immune cell engagers (ICEs) are an emerging class of immunotherapies designed to harness the immune system's anti-tumor potential through precise targeting and activation of immune effector cells. By engaging T cells, natural killer (NK) cells, and phagocytes, ICEs overcome challenges such as immune evasion and MHC downregulation, addressing critical barriers in cancer treatment. T-cell engagers (TCEs), led by bispecific T-cell engagers (BiTEs), dominate the field, with innovations such as half-life-extended BiTEs, trispecific antibodies, and checkpoint inhibitory T-cell engagers driving their application in hematologic and solid malignancies. NK cell engagers (NKCEs) and phagocyte cell engagers (PCEs) are rapidly progressing, drawing on NK cells' innate cytotoxicity and macrophages' phagocytic abilities to target tumors, particularly in immunosuppressive microenvironments. Since the FDA approval of Blinatumomab in 2014, ICEs have transformed the oncology landscape, with nine FDA-approved products and numerous candidates in clinical trials. Despite challenges such as toxicity, resistance, and limited efficacy in solid tumors, ongoing research into advanced platforms and combination therapies highlights the growing potential of ICEs to provide personalized, scalable, and effective cancer treatments. This review investigates the mechanisms, platforms, research trends, and clinical progress of ICEs, emphasizing their pivotal role in advancing precision immunotherapy and their promise as a cornerstone of next-generation cancer therapies.
Collapse
Affiliation(s)
| | | | | | - Kyung Ho Han
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea
| |
Collapse
|
4
|
Nikkhoi SK, Li G, Hatefi A. Natural killer cell engagers for cancer immunotherapy. Front Oncol 2025; 14:1483884. [PMID: 39911822 PMCID: PMC11794116 DOI: 10.3389/fonc.2024.1483884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
This review article explores the rapidly evolving field of bi-, tri-, and multi-specific NK cell engagers (NKCEs), highlighting their potential as a cutting-edge approach in cancer immunotherapy. NKCEs offer a significant advancement over conventional monoclonal antibodies (mAbs) by enhancing Antibody-Dependent Cellular Cytotoxicity (ADCC). They achieve this by stably and selectively binding to both NK cell activating receptors and tumor-associated antigens (TAAs). Unlike traditional mAbs, which depend on the relatively transient interaction between their Fc region and CD16a, NKCEs establish more robust connections with a range of activating receptors (e.g., CD16a, NKG2D, NKp30, NKp46, NKG2C) and inhibitory receptors (e.g., Siglec-7) on NK cells, thereby increasing cancer cell killing efficacy and specificity. This review article critically examines the strategies for engineering bi-, tri-, and multi-specific NKCEs for cancer immunotherapy, providing an in-depth analysis of the latest advancements in NKCE platform technologies currently under development by pharmaceutical and biotech companies and discussing the preclinical and clinical progress of these products. While NKCEs show great promise, the review underscores the need for continued research to optimize their therapeutic efficacy and to overcome obstacles related to NK cell functionality in cancer patients. Ultimately, this article presents an overview of the current landscape and future prospects of NKCE-based cancer immunotherapy, emphasizing its potential to revolutionize cancer treatment.
Collapse
Affiliation(s)
| | - Geng Li
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ, United States
| | - Arash Hatefi
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ, United States
- Cancer Pharmacology Program, Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
5
|
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
|
6
|
Stenger TD, Miller JS. Therapeutic approaches to enhance natural killer cell cytotoxicity. Front Immunol 2024; 15:1356666. [PMID: 38545115 PMCID: PMC10966407 DOI: 10.3389/fimmu.2024.1356666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/05/2024] [Indexed: 04/14/2024] Open
Abstract
Enhancing the cytotoxicity of natural killer (NK) cells has emerged as a promising strategy in cancer immunotherapy, due to their pivotal role in immune surveillance and tumor clearance. This literature review provides a comprehensive overview of therapeutic approaches designed to augment NK cell cytotoxicity. We analyze a wide range of strategies, including cytokine-based treatment, monoclonal antibodies, and NK cell engagers, and discuss criteria that must be considered when selecting an NK cell product to combine with these strategies. Furthermore, we discuss the challenges and limitations associated with each therapeutic strategy, as well as the potential for combination therapies to maximize NK cell cytotoxicity while minimizing adverse effects. By exploring the wealth of research on this topic, this literature review aims to provide a comprehensive resource for researchers and clinicians seeking to develop and implement novel therapeutic strategies that harness the full potential of NK cells in the fight against cancer. Enhancing NK cell cytotoxicity holds great promise in the evolving landscape of immunotherapy, and this review serves as a roadmap for understanding the current state of the field and the future directions in NK cell-based therapies.
Collapse
Affiliation(s)
- Terran D. Stenger
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | | |
Collapse
|
7
|
Ulitzka M, Harwardt J, Lipinski B, Tran H, Hock B, Kolmar H. Potent Apoptosis Induction by a Novel Trispecific B7-H3xCD16xTIGIT 2+1 Common Light Chain Natural Killer Cell Engager. Molecules 2024; 29:1140. [PMID: 38474651 DOI: 10.3390/molecules29051140] [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: 11/30/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Valued for their ability to rapidly kill multiple tumor cells in succession as well as their favorable safety profile, NK cells are of increasing interest in the field of immunotherapy. As their cytotoxic activity is controlled by a complex network of activating and inhibiting receptors, they offer a wide range of possible antigens to modulate their function by antibodies. In this work, we utilized our established common light chain (cLC)-based yeast surface display (YSD) screening procedure to isolate novel B7-H3 and TIGIT binding monoclonal antibodies. The chicken-derived antibodies showed single- to low-double-digit nanomolar affinities and were combined with a previously published CD16-binding Fab in a 2+1 format to generate a potent NK engaging molecule. In a straightforward, easily adjustable apoptosis assay, the construct B7-H3xCD16xTIGIT showed potent apoptosis induction in cancer cells. These results showcase the potential of the TIGIT NK checkpoint in combination with activating receptors to achieve increased cytotoxic activity.
Collapse
Affiliation(s)
- Michael Ulitzka
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287 Darmstadt, Germany
| | - Julia Harwardt
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287 Darmstadt, Germany
| | - Britta Lipinski
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287 Darmstadt, Germany
| | - Hue Tran
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287 Darmstadt, Germany
| | - Björn Hock
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287 Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287 Darmstadt, Germany
- Centre of Synthetic Biology, Technical University of Darmstadt, 64283 Darmstadt, Germany
| |
Collapse
|
8
|
Boje AS, Pekar L, Koep K, Lipinski B, Rabinovich B, Evers A, Gehlert CL, Krohn S, Xiao Y, Krah S, Zaynagetdinov R, Toleikis L, Poetzsch S, Peipp M, Zielonka S, Klausz K. Impact of antibody architecture and paratope valency on effector functions of bispecific NKp30 x EGFR natural killer cell engagers. MAbs 2024; 16:2315640. [PMID: 38372053 PMCID: PMC10877975 DOI: 10.1080/19420862.2024.2315640] [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: 06/09/2023] [Accepted: 02/02/2024] [Indexed: 02/20/2024] Open
Abstract
Natural killer (NK) cells emerged as a promising effector population that can be harnessed for anti-tumor therapy. In this work, we constructed NK cell engagers (NKCEs) based on NKp30-targeting single domain antibodies (sdAbs) that redirect the cytotoxic potential of NK cells toward epidermal growth factor receptor (EGFR)-expressing tumor cells. We investigated the impact of crucial parameters such as sdAb location, binding valencies, the targeted epitope on NKp30, and the overall antibody architecture on the redirection capacity. Our study exploited two NKp30-specific sdAbs, one of which binds a similar epitope on NKp30 as its natural ligand B7-H6, while the other sdAb addresses a non-competing epitope. For EGFR-positive tumor targeting, humanized antigen-binding domains of therapeutic antibody cetuximab were used. We demonstrate that NKCEs bivalently targeting EGFR and bivalently engaging NKp30 are superior to monovalent NKCEs in promoting NK cell-mediated tumor cell lysis and that the architecture of the NKCE can substantially influence killing capacities depending on the NKp30-targeting sdAb utilized. While having a pronounced impact on NK cell killing efficacy, the capabilities of triggering antibody-dependent cellular phagocytosis or complement-dependent cytotoxicity were not significantly affected comparing the bivalent IgG-like NKCEs with cetuximab. However, the fusion of sdAbs can have a slight impact on the NK cell release of immunomodulatory cytokines, as well as on the pharmacokinetic profile of the NKCE due to unfavorable spatial orientation within the molecule architecture. Ultimately, our findings reveal novel insights for the engineering of potent NKCEs triggering the NKp30 axis.
Collapse
Affiliation(s)
- Ammelie Svea Boje
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| | - Lukas Pekar
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Katharina Koep
- Drug Metabolism and Pharmacokinetics, Merck Healthcare KGaA, Darmstadt, Germany
| | - Britta Lipinski
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Brian Rabinovich
- Department of Oncology and Immuno-Oncology, EMD Serono Research & Development Institute Inc, 45A Middlesex Turnpike, Billerica, MA, USA
| | - Andreas Evers
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Carina Lynn Gehlert
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| | - Steffen Krohn
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| | - Yanping Xiao
- Department of Oncology and Immuno-Oncology, EMD Serono Research & Development Institute Inc, 45A Middlesex Turnpike, Billerica, MA, USA
| | - Simon Krah
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Rinat Zaynagetdinov
- Department of Oncology and Immuno-Oncology, EMD Serono Research & Development Institute Inc, 45A Middlesex Turnpike, Billerica, MA, USA
| | - Lars Toleikis
- Early Protein Supply & Characterization, Merck Healthcare KGaA, Darmstadt, Germany
| | - Sven Poetzsch
- Strategic Innovation, Merck Healthcare KGaA, Darmstadt, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| | - Stefan Zielonka
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| |
Collapse
|
9
|
Heisler J, Kovner D, Izadi S, Zarzar J, Carter PJ. Modulation of the high concentration viscosity of IgG 1 antibodies using clinically validated Fc mutations. MAbs 2024; 16:2379560. [PMID: 39028186 PMCID: PMC11262234 DOI: 10.1080/19420862.2024.2379560] [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: 04/04/2024] [Accepted: 07/09/2024] [Indexed: 07/20/2024] Open
Abstract
The self-association of therapeutic antibodies can result in elevated viscosity and create problems in manufacturing and formulation, as well as limit delivery by subcutaneous injection. The high concentration viscosity of some antibodies has been reduced by variable domain mutations or by the addition of formulation excipients. In contrast, the impact of Fc mutations on antibody viscosity has been minimally explored. Here, we studied the effect of a panel of common and clinically validated Fc mutations on the viscosity of two closely related humanized IgG1, κ antibodies, omalizumab (anti-IgE) and trastuzumab (anti-HER2). Data presented here suggest that both Fab-Fab and Fab-Fc interactions contribute to the high viscosity of omalizumab, in a four-contact model of self-association. Most strikingly, the high viscosity of omalizumab (176 cP) was reduced 10.7- and 2.2-fold by Fc modifications for half-life extension (M252Y:S254T:T256E) and aglycosylation (N297G), respectively. Related single mutations (S254T and T256E) each reduced the viscosity of omalizumab by ~6-fold. An alternative half-life extension Fc mutant (M428L:N434S) had the opposite effect in increasing the viscosity of omalizumab by 1.5-fold. The low viscosity of trastuzumab (8.6 cP) was unchanged or increased by ≤ 2-fold by the different Fc variants. Molecular dynamics simulations provided mechanistic insight into the impact of Fc mutations in modulating electrostatic and hydrophobic surface properties as well as conformational stability of the Fc. This study demonstrates that high viscosity of some IgG1 antibodies can be mitigated by Fc mutations, and thereby offers an additional tool to help design future antibody therapeutics potentially suitable for subcutaneous delivery.
Collapse
Affiliation(s)
- Joel Heisler
- Department of Antibody Engineering, Genentech, Inc, South San Francisco, CA, USA
| | - Daniel Kovner
- Department of Pharmaceutical Development, Genentech, Inc, South San Francisco, CA, USA
| | - Saeed Izadi
- Department of Pharmaceutical Development, Genentech, Inc, South San Francisco, CA, USA
| | - Jonathan Zarzar
- Department of Pharmaceutical Development, Genentech, Inc, South San Francisco, CA, USA
| | - Paul J. Carter
- Department of Antibody Engineering, Genentech, Inc, South San Francisco, CA, USA
| |
Collapse
|
10
|
Liao B, Tumanut C, Li L, Corper A, Challa D, Chang A, Begum H, Farokhi E, Woods C, Fan X. Identification of novel anti-CD16a antibody clones for the development of effective natural killer cell engagers. MAbs 2024; 16:2381261. [PMID: 39048914 PMCID: PMC11271076 DOI: 10.1080/19420862.2024.2381261] [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: 01/11/2024] [Revised: 07/03/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024] Open
Abstract
Natural killer (NK) cells are key players in human innate immunity. Cell engager antibody formats that recruit and activate NK cells more effectively have emerged as a promising immunotherapy approach to target cancer cells through more effective antibody-dependent cell-mediated cytotoxicity (ADCC). Monoclonal antibody drugs with ADCC activity have shown clinical benefit and improved outcomes for patients with certain types of cancer. CD16a, a Fc gamma III receptor, is the major component that is responsible for the ADCC activity of NK cells. Screening AvantGen's yeast displayed human antibody libraries led to the isolation of 2 antibody clones, #1A2 and #2-2A2, that selectively recognize both isoforms (F and V) of CD16a on primary NK cells with high affinity, yet minimally (#1A2) or do not (#2-2A2) cross-react with both allelotypes of CD16b (NA1 and NA2) expressed by neutrophils. Epitope mapping studies revealed that they bind to an epitope dependent on residue Y158 of CD16a, since mutation of Y158 to the corresponding CD16b residue H158 completely abolishes binding to CD16a. When formatted as bispecific antibodies targeting CD16a and a tumor-associated antigen (TAA, e.g. CD19), they exhibit specific binding to NK cells and induce potent NK cell activation upon encountering tumor cells, resulting in effective tumor cell killing. Notably, these bispecific antibody engagers stimulate NK cell cytokine release during co-culture with target cells, resulting in target cell cytotoxicity. These anti-CD16a antibody clones are promising candidates for combination with any TAA of interest, offering the potential for novel NK cell engager-based cancer therapeutics that are minimally affected by the high concentrations of human IgG in the circulation.
Collapse
Affiliation(s)
| | | | - Lin Li
- AvantGen, Inc, San Diego, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Giang KA, Boxaspen T, Diao Y, Nilvebrant J, Kosugi-Kanaya M, Kanaya M, Krokeide SZ, Lehmann F, Svensson Gelius S, Malmberg KJ, Nygren PÅ. Affibody-based hBCMA x CD16 dual engagers for NK cell-mediated killing of multiple myeloma cells. N Biotechnol 2023; 77:139-148. [PMID: 37673373 DOI: 10.1016/j.nbt.2023.09.002] [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: 07/04/2023] [Revised: 08/16/2023] [Accepted: 09/03/2023] [Indexed: 09/08/2023]
Abstract
We describe the development and characterization of the (to date) smallest Natural Killer (NK) cell re-directing human B Cell Maturation Antigen (hBCMA) x CD16 dual engagers for potential treatment of multiple myeloma, based on combinations of small 58 amino acid, non-immunoglobulin, affibody affinity proteins. Affibody molecules to human CD16a were selected from a combinatorial library by phage display resulting in the identification of three unique binders with affinities (KD) for CD16a in the range of 100 nM-3 µM. The affibody exhibiting the highest affinity demonstrated insensitivity towards the CD16a allotype (158F/V) and did not interfere with IgG (Fc) binding to CD16a. For the construction of hBCMA x CD16 dual engagers, different CD16a binding arms, including bi-paratopic affibody combinations, were genetically fused to a high-affinity hBCMA-specific affibody. Such 15-23 kDa dual engager constructs showed simultaneous hBCMA and CD16a binding ability and could efficiently activate resting primary NK cells and trigger specific lysis of a panel of hBCMA-positive multiple myeloma cell lines. Hence, we report a novel class of uniquely small NK cell engagers with specific binding properties and potent functional profiles.
Collapse
Affiliation(s)
- Kim Anh Giang
- Department of Protein Science, Div. Protein Engineering, AlbaNova University Center, KTH Royal Institute of Technology, S-114 21 Stockholm, Sweden
| | - Thorstein Boxaspen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway
| | - Yumei Diao
- Oncopeptides AB, S-171 48 Stockholm, Sweden
| | - Johan Nilvebrant
- Department of Protein Science, Div. Protein Engineering, AlbaNova University Center, KTH Royal Institute of Technology, S-114 21 Stockholm, Sweden
| | - Mizuha Kosugi-Kanaya
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway
| | - Minoru Kanaya
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway
| | - Silje Zandstra Krokeide
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway
| | | | | | - Karl-Johan Malmberg
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway.
| | - Per-Åke Nygren
- Department of Protein Science, Div. Protein Engineering, AlbaNova University Center, KTH Royal Institute of Technology, S-114 21 Stockholm, Sweden; Science For Life Laboratory, S-171 65 Solna, Sweden.
| |
Collapse
|
12
|
Huan T, Guan B, Li H, Tu X, Zhang C, Tang B. Principles and current clinical landscape of NK cell engaging bispecific antibody against cancer. Hum Vaccin Immunother 2023; 19:2256904. [PMID: 37772505 PMCID: PMC10543353 DOI: 10.1080/21645515.2023.2256904] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/05/2023] [Indexed: 09/30/2023] Open
Abstract
Monoclonal antibody-based targeted therapies have greatly improved treatment options for patients by binding to the innate immune system. However, the long-term efficacy of such antibodies is limited by mechanisms of drug resistance. Over the last 50 years, with advances in protein engineering technology, more and more bispecific antibody (bsAb) platforms have been engineered to meet diverse clinical needs. Bispecific NK cell engagers (BiKEs) or tri-specific NK cell engagers (TriKEs) allow for direct targeting of immune cells to tumors, and therefore resistance and serious adverse effects are greatly reduced. Many preclinical and clinical trials are currently underway, depicting the promise of antibody-based natural killer cell engager therapeutics. In this review, we compile worldwide efforts to explore the involvement of NK cells in bispecific antibodies. With a particular emphasis on lessons learned, we focus on preclinical and clinical studies in malignancies and discuss the reasons for the limited success of NK-cell engagers against solid tumors, offering plausible new ideas for curing some advanced cancers shortly.
Collapse
Affiliation(s)
- Tian Huan
- Department of General Surgery, Jinhu County People’s Hospital, Huaian, Jiangsu, China
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bugao Guan
- Department of General Surgery, Jinhu County People’s Hospital, Huaian, Jiangsu, China
| | - Hongbo Li
- Department of General Surgery, Jinhu County People’s Hospital, Huaian, Jiangsu, China
| | - Xiu Tu
- Department of General Surgery, Jinhu County People’s Hospital, Huaian, Jiangsu, China
| | - Chi Zhang
- Department of General Surgery, Jinhu County People’s Hospital, Huaian, Jiangsu, China
| | - Bin Tang
- Department of General Surgery, Jinhu County People’s Hospital, Huaian, Jiangsu, China
- Department of Central Laboratory, Jinhu County People’s Hospital, Huaian, Jiangsu, China
| |
Collapse
|
13
|
Tapia-Galisteo A, Álvarez-Vallina L, Sanz L. Bi- and trispecific immune cell engagers for immunotherapy of hematological malignancies. J Hematol Oncol 2023; 16:83. [PMID: 37501154 PMCID: PMC10373336 DOI: 10.1186/s13045-023-01482-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
Immune cell engagers are engineered antibodies with at least one arm binding a tumor-associated antigen and at least another one directed against an activating receptor in immune effector cells: CD3 for recruitment of T cells and CD16a for NK cells. The first T cell engager (the anti-CD19 blinatumomab) was approved by the FDA in 2014, but no other one hit the market until 2022. Now the field is gaining momentum, with three approvals in 2022 and 2023 (as of May): the anti-CD20 × anti-CD3 mosunetuzumab and epcoritamab and the anti-B cell maturation antigen (BCMA) × anti-CD3 teclistamab, and another three molecules in regulatory review. T cell engagers will likely revolutionize the treatment of hematological malignancies in the short term, as they are considerably more potent than conventional monoclonal antibodies recognizing the same tumor antigens. The field is thriving, with a plethora of different formats and targets, and around 100 bispecific T cell engagers more are already in clinical trials. Bispecific NK cell engagers are also in early-stage clinical studies and may offer similar efficacy with milder side effects. Trispecific antibodies (engaging either T cell or NK cell receptors) raise the game even further with a third binding moiety, which allows either the targeting of an additional tumor-associated antigen to increase specificity and avoid immune escape or the targeting of additional costimulatory receptors on the immune cell to improve its effector functions. Altogether, these engineered molecules may change the paradigm of treatment for relapsed or refractory hematological malignancies.
Collapse
Affiliation(s)
- Antonio Tapia-Galisteo
- Immuno-Oncology and Immunotherapy Group, Biomedical Research Institute Hospital Universitario, 12 de Octubre, Madrid, Spain
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario, 12 de Octubre, Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Luis Álvarez-Vallina
- Immuno-Oncology and Immunotherapy Group, Biomedical Research Institute Hospital Universitario, 12 de Octubre, Madrid, Spain.
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario, 12 de Octubre, Madrid, Spain.
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain.
| | - Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain.
| |
Collapse
|
14
|
Seo H, Verma A, Kinzel M, Huang Q, Mahoney DJ, Jacquelot N. Targeting Potential of Innate Lymphoid Cells in Melanoma and Other Cancers. Pharmaceutics 2023; 15:2001. [PMID: 37514187 PMCID: PMC10384206 DOI: 10.3390/pharmaceutics15072001] [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: 06/13/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Reinvigorating the killing function of tumor-infiltrating immune cells through the targeting of regulatory molecules expressed on lymphocytes has markedly improved the prognosis of cancer patients, particularly in melanoma. While initially thought to solely strengthen adaptive T lymphocyte anti-tumor activity, recent investigations suggest that other immune cell subsets, particularly tissue-resident innate lymphoid cells (ILCs), may benefit from immunotherapy treatment. Here, we describe the recent findings showing immune checkpoint expression on tissue-resident and tumor-infiltrating ILCs and how their effector function is modulated by checkpoint blockade-based therapies in cancer. We discuss the therapeutic potential of ILCs beyond the classical PD-1 and CTLA-4 regulatory molecules, exploring other possibilities to manipulate ILC effector function to further impede tumor growth and quench disease progression.
Collapse
Affiliation(s)
- Hobin Seo
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| | - Amisha Verma
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Megan Kinzel
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| | - Qiutong Huang
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, QLD 4102, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Douglas J Mahoney
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| | - Nicolas Jacquelot
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
15
|
Shin E, Bak SH, Park T, Kim JW, Yoon SR, Jung H, Noh JY. Understanding NK cell biology for harnessing NK cell therapies: targeting cancer and beyond. Front Immunol 2023; 14:1192907. [PMID: 37539051 PMCID: PMC10395517 DOI: 10.3389/fimmu.2023.1192907] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/30/2023] [Indexed: 08/05/2023] Open
Abstract
Gene-engineered immune cell therapies have partially transformed cancer treatment, as exemplified by the use of chimeric antigen receptor (CAR)-T cells in certain hematologic malignancies. However, there are several limitations that need to be addressed to target more cancer types. Natural killer (NK) cells are a type of innate immune cells that represent a unique biology in cancer immune surveillance. In particular, NK cells obtained from heathy donors can serve as a source for genetically engineered immune cell therapies. Therefore, NK-based therapies, including NK cells, CAR-NK cells, and antibodies that induce antibody-dependent cellular cytotoxicity of NK cells, have emerged. With recent advances in genetic engineering and cell biology techniques, NK cell-based therapies have become promising approaches for a wide range of cancers, viral infections, and senescence. This review provides a brief overview of NK cell characteristics and summarizes diseases that could benefit from NK-based therapies. In addition, we discuss recent preclinical and clinical investigations on the use of adoptive NK cell transfer and agents that can modulate NK cell activity.
Collapse
Affiliation(s)
- Eunju Shin
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Seong Ho Bak
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Taeho Park
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Jin Woo Kim
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Suk-Ran Yoon
- Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Haiyoung Jung
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Ji-Yoon Noh
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| |
Collapse
|
16
|
Russler-Germain DA, Ghobadi A. T-cell redirecting therapies for B-cell non-Hodgkin lymphoma: recent progress and future directions. Front Oncol 2023; 13:1168622. [PMID: 37465110 PMCID: PMC10351267 DOI: 10.3389/fonc.2023.1168622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/21/2023] [Indexed: 07/20/2023] Open
Abstract
Several key advances in the treatment of B-cell non-Hodgkin lymphoma (B-NHL) over the past two decades have strategically exploited B-cell lineage markers suitable for targeting by immunotherapies. First, the addition of the anti-CD20 monoclonal antibody (mAb) rituximab to a range of standard therapies conferred remarkable outcomes improvements in diverse settings, perhaps most prominently an overall survival advantage in newly diagnosed diffuse large B-cell lymphoma (DLBCL). Subsequently, multiple chimeric antigen receptor (CAR) T-cell therapies targeting CD19 have revolutionized the treatment of relapsed/refractory (rel/ref) DLBCL and are active in other B-NHL subtypes as well. Most recently, the longstanding aspiration to exploit patients' endogenous T-cells to combat lymphoma has been achieved via T-cell redirecting therapies such as bispecific antibodies (BsAbs) that incorporate dual targeting of a T-cell antigen such as CD3 plus a B-cell antigen such as CD19 or CD20 expressed by the tumor. These novel agents have demonstrated impressive activity as monotherapies in patients with heavily pre-treated, rel/ref B-NHL of a variety of subtypes. Now, myriad clinical trials are exploring combinations of T-cell redirectors with targeted therapies, antibody-drug conjugates, conventional chemotherapy, and even new immunotherapies. Here, we highlight key landmarks in the development of T-cell redirecting therapies for the treatment of B-NHL, emerging evidence and lessons from recent clinical trials, and exciting new directions in this arena.
Collapse
Affiliation(s)
- David A. Russler-Germain
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, United States
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Armin Ghobadi
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, United States
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
17
|
Braun T, Schrader A. Education and Empowering Special Forces to Eradicate Secret Defectors: Immune System-Based Treatment Approaches for Mature T- and NK-Cell Malignancies. Cancers (Basel) 2023; 15:cancers15092532. [PMID: 37173999 PMCID: PMC10177197 DOI: 10.3390/cancers15092532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Mature T- and NK-cell leukemia/lymphoma (MTCL/L) constitute a heterogeneous group of, currently, 30 distinct neoplastic entities that are overall rare, and all present with a challenging molecular markup. Thus, so far, the use of first-line cancer treatment modalities, including chemotherapies, achieve only limited clinical responses associated with discouraging prognoses. Recently, cancer immunotherapy has evolved rapidly, allowing us to help patients with, e.g., solid tumors and also relapsed/refractory B-cell malignancies to achieve durable clinical responses. In this review, we systematically unveiled the distinct immunotherapeutic approaches available, emphasizing the special impediments faced when trying to employ immune system defense mechanisms to target 'one of their own-gone mad'. We summarized the preclinical and clinical efforts made to employ the various platforms of cancer immunotherapies including antibody-drug conjugates, monoclonal as well as bispecific antibodies, immune-checkpoint blockades, and CAR T cell therapies. We emphasized the challenges to, but also the goals of, what needs to be done to achieve similar successes as seen for B-cell entities.
Collapse
Affiliation(s)
- Till Braun
- Department I of Internal Medicine, Center for Integrated Oncology, Aachen-Bonn-Cologne-Duesseldorf, Excellence Cluster for Cellular Stress Response and Aging-Associated Diseases, Center for Molecular Medicine Cologne, University of Cologne, 50937 Cologne, Germany
| | - Alexandra Schrader
- Department I of Internal Medicine, Center for Integrated Oncology, Aachen-Bonn-Cologne-Duesseldorf, Excellence Cluster for Cellular Stress Response and Aging-Associated Diseases, Center for Molecular Medicine Cologne, University of Cologne, 50937 Cologne, Germany
- Lymphoma Immuno Biology Team, Equipe Labellisée LIGUE 2023, Centre International de Recherche en Infectiologie, INSERM U1111-CNRS UMR5308, Faculté de Médecine Lyon-Sud, Hospices Civils de Lyon, Université Claude Bernard Lyon I-ENS de Lyon, 69921 Lyon, France
| |
Collapse
|
18
|
Yu Y. The Function of NK Cells in Tumor Metastasis and NK Cell-Based Immunotherapy. Cancers (Basel) 2023; 15:cancers15082323. [PMID: 37190251 DOI: 10.3390/cancers15082323] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/09/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Metastatic tumors cause the most deaths in cancer patients. Treating metastasis remains the primary goal of current cancer research. Although the immune system prevents and kills the tumor cells, the function of the immune system in metastatic cancer has been unappreciated for decades because tumors are able to develop complex signaling pathways to suppress immune responses, leading them to escape detection and elimination. Studies showed NK cell-based therapies have many advantages and promise for fighting metastatic cancers. We here review the function of the immune system in tumor progression, specifically focusing on the ability of NK cells in antimetastasis, how metastatic tumors escape the NK cell attack, as well as the recent development of effective antimetastatic immunotherapies.
Collapse
Affiliation(s)
- Yanlin Yu
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
19
|
Harwardt J, Carrara SC, Bogen JP, Schoenfeld K, Grzeschik J, Hock B, Kolmar H. Generation of a symmetrical trispecific NK cell engager based on a two-in-one antibody. Front Immunol 2023; 14:1170042. [PMID: 37081888 PMCID: PMC10110854 DOI: 10.3389/fimmu.2023.1170042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
To construct a trispecific IgG-like antibody at least three different binding moieties need to be combined, which results in a complex architecture and challenging production of these molecules. Here we report for the first time the construction of trispecific natural killer cell engagers based on a previously reported two-in-one antibody combined with a novel anti-CD16a common light chain module identified by yeast surface display (YSD) screening of chicken-derived immune libraries. The resulting antibodies simultaneously target epidermal growth factor receptor (EGFR), programmed death-ligand 1 (PD-L1) and CD16a with two Fab fragments, resulting in specific cellular binding properties on EGFR/PD-L1 double positive tumor cells and a potent ADCC effect. This study paves the way for further development of multispecific therapeutic antibodies derived from avian immunization with desired target combinations, valencies, molecular symmetries and architectures.
Collapse
Affiliation(s)
- Julia Harwardt
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Stefania C. Carrara
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Biologics Technology and Development, Ferring Darmstadt Laboratory, Darmstadt, Germany
| | - Jan P. Bogen
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Biologics Technology and Development, Ferring Darmstadt Laboratory, Darmstadt, Germany
| | - Katrin Schoenfeld
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Julius Grzeschik
- Biologics Technology and Development, Ferring Biologics Innovation Centre, Epalinges, Switzerland
| | - Björn Hock
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany
- *Correspondence: Harald Kolmar,
| |
Collapse
|
20
|
Nikkhoi SK, Li G, Eleya S, Yang G, Vandavasi VG, Hatefi A. Bispecific killer cell engager with high affinity and specificity toward CD16a on NK cells for cancer immunotherapy. Front Immunol 2023; 13:1039969. [PMID: 36685519 PMCID: PMC9852913 DOI: 10.3389/fimmu.2022.1039969] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/30/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction The Fc region of monoclonal antibodies (mAbs) interacts with the CD16a receptor on natural killer (NK) cells with "low affinity" and "low selectivity". This low affinity/selectivity interaction results in not only suboptimal anticancer activity but also induction of adverse effects. CD16a on NK cells binds to the antibody-coated cells, leading to antibody-dependent cell-mediated cytotoxicity (ADCC). Recent clinical data have shown that the increased binding affinity between mAb Fc region and CD16a receptor is responsible for significantly improved therapeutic outcomes. Therefore, the objective of this study was to develop a bispecific killer cell engager (BiKE) with high affinity and specificity/selectivity toward CD16a receptor for NK cell-based cancer immunotherapy. Methods To engineer BiKE, a llama was immunized, then high binding anti-CD16a and anti-HER2 VHH clones were isolated using phage display. ELISA, flow cytometry, and biolayer interferometry (BLI) data showed that the isolated anti-CD16a VHH has high affinity (sub-nanomolar) toward CD16a antigen without cross-reactivity with CD16b-NA1 on neutrophils or CD32b on B cells. Similarly, the data showed that the isolated anti-HER2 VHH has high affinity/specificity toward HER2 antigen. Using a semi-flexible linker, anti-HER2 VHH was recombinantly fused with anti-CD16a VHH to create BiKE:HER2/CD16a. Then, the ability of BiKE:HER2/CD16a to activate NK cells to release cytokines and kill HER2+ cancer cells was measured. As effector cells, both high-affinity haNK92 (CD16+, V176) and low-affinity laNK92 (CD16+, F176) cells were used. Results and discussion The data showed that the engineered BiKE:HER2/CD16a activates haNK92 and laNK92 cells to release cytokines much greater than best-in-class mAbs in the clinic. The cytotoxicity data also showed that the developed BiKE induces higher ADCC to both ovarian and breast cancer cells in comparison to Trazimera™ (trastuzumab). According to the BLI data, BiKE:HER2/CD16 recognizes a different epitope on CD16a antigen than IgG-based mAbs; thus, it provides the opportunity for not only monotherapy but also combination therapy with other antibody drugs such as checkpoint inhibitors and antibody-drug conjugates. Taken together, the data demonstrate the creation of a novel BiKE with high affinity and specificity toward CD16a on NK cells with the potential to elicit a superior therapeutic response in patients with HER2+ cancer than existing anti-HER2 mAbs.
Collapse
Affiliation(s)
| | - Geng Li
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ, United States
| | - Suha Eleya
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ, United States
| | - Ge Yang
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ, United States
| | - Venu Gopal Vandavasi
- Department of Chemistry, Biophysics Core Facility, Princeton University, Princeton, NJ, United States
| | - Arash Hatefi
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ, United States
- Cancer Pharmacology Program, Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, United States
| |
Collapse
|
21
|
Topp M, Dlugosz-Danecka M, Skotnicki AB, Salogub G, Viardot A, Klein AK, Hess G, Michel CS, Grosicki S, Gural A, Schwarz SE, Pietzko K, Gärtner U, Strassz A, Alland L, Mayer J. Safety of AFM11 in the treatment of patients with B-cell malignancies: findings from two phase 1 studies. Trials 2023; 24:4. [PMID: 36597128 PMCID: PMC9808944 DOI: 10.1186/s13063-022-06982-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/05/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The prognosis for patients with relapsed and/or refractory (R/R) non-Hodgkin's lymphoma (NHL) or acute lymphoblastic leukaemia (ALL) remains poor, with existing treatments having significant side effects. Developed for the treatment of these cancers, AFM11 is a tetravalent, bispecific humanised recombinant antibody construct (TandAb®) designed to bind to human CD19 and CD3 and lead to the activation of T cells inducing apoptosis and killing of malignant B cells. METHODS Two open-label, multicentre, dose-escalation phase 1 studies evaluated the safety, pharmacokinetics and activity of AFM11 in patients with R/R CD19-positive B cell NHL (AFM11-101) and in patients with CD19 + B-precursor Philadelphia-chromosome negative ALL (AFM11-102). Adverse events (AEs) were assessed and recorded; imaging (NHL) or bone marrow assessment (ALL) were used to evaluate response. Additional pharmacodynamic assays undertaken included cytokine release analysis and B-cell and T-cell depletion. RESULTS In AFM11-101, 16 patients with R/R NHL received AFM11 in five different dose cohorts. Of which, 14 experienced drug-related treatment-emergent AEs (TEAEs) [including five serious AEs (SAEs)], five patients experienced dose-limiting toxicity (DLT) and ten patients discontinued the study. The high number of neurological events led to a decrease in infusion frequency during the study. No objective response to treatment was observed. In AFM11-102, 17 patients with R/R ALL received AFM11 in six different dose cohorts. Thirteen patients experienced drug-related TEAEs (including four SAEs), DLTs occurred in two patients and five patients discontinued the study. An objective response was recorded in three patients. The maximum tolerated dose could not be determined in either study due to early termination. CONCLUSIONS AFM11 treatment was associated with frequent neurological adverse reactions that were severe in some patients. In ALL, some signs of activity, albeit short-lived, were observed whereas no activity was observed in patients with NHL; therefore, further clinical development was terminated. TRIAL REGISTRATION NCT02106091 . Safety Study to Assess AFM11 in Patients With Relapsed and/or Refractory CD19 Positive B-cell NHL. Registered April 2014. NCT02848911 . Safety Study to Assess AFM11 in Patients With Relapsed or Refractory Adult B-precursor ALL. Registered July 2016.
Collapse
Affiliation(s)
- Max Topp
- grid.411760.50000 0001 1378 7891Universitätsklinikum Würzburg, Würzburg, Germany
| | - Monika Dlugosz-Danecka
- grid.418165.f0000 0004 0540 2543Maria Sklodowska-Curie National Research Institute of Oncology, Kraków, Poland
| | - Aleksander B. Skotnicki
- grid.5522.00000 0001 2162 9631Department of Haematology, Jagiellonian University, Kraków, Poland
| | - Galina Salogub
- Almazova National Medical Research Center, St Petersburg, Russia
| | - Andreas Viardot
- grid.410712.10000 0004 0473 882XKlinik Für Innere Medizin III, Universitätsklinikum Ulm, Ulm, Germany
| | - Andreas K. Klein
- grid.67033.310000 0000 8934 4045Tufts Medical Center, Boston, MA USA
| | - Georg Hess
- grid.410607.4Universitätsmedizin Mainz, Mainz, Germany
| | | | - Sebastian Grosicki
- grid.411728.90000 0001 2198 0923Department of Hematology and Cancer Prevention, Faculty of Health Sciences, Medical University of Silesia, Katowice, Poland
| | - Alex Gural
- grid.17788.310000 0001 2221 2926Hadassah Medical Center, Jerusalem, Israel
| | | | - Kerstin Pietzko
- grid.432627.60000 0004 0631 9610Affimed GmbH, Heidelberg, Germany
| | - Ulrike Gärtner
- grid.432627.60000 0004 0631 9610Affimed GmbH, Heidelberg, Germany
| | - András Strassz
- grid.432627.60000 0004 0631 9610Affimed GmbH, Heidelberg, Germany
| | - Leila Alland
- grid.432627.60000 0004 0631 9610Affimed GmbH, Heidelberg, Germany
| | - Jiri Mayer
- University Hospital Brno, and Masaryk University, Brno, Czech Republic
| |
Collapse
|
22
|
Müller T, Tasser C, Tesar M, Fucek I, Schniegler-Mattox U, Koch J, Ellwanger K. Selection of bispecific antibodies with optimal developability using FcRn‑Ph‑HPLC as an optimized FcRn affinity chromatography method. MAbs 2023; 15:2245519. [PMID: 37599441 PMCID: PMC10443974 DOI: 10.1080/19420862.2023.2245519] [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: 05/04/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/22/2023] Open
Abstract
A challenge when developing therapeutic antibodies is the identification of candidates with favorable pharmacokinetics (PK) early in development. A key determinant of immunoglobulin (IgG) serum half‑life in vivo is the efficiency of pH-dependent binding to the neonatal Fc receptor (FcRn). Numerous studies have proposed techniques to assess FcRn binding of IgG-based therapeutics in vitro, enabling prediction of serum half-life prior to clinical assessment. FcRn high-performance liquid chromatography (HPLC) assays FcRn binding of therapeutic IgGs across a pH gradient, allowing the correlation of IgG column retention time to the half‑life of a therapeutic IgG in vivo. However, as FcRn retention time cannot be directly compared to an in vivo parameter, modifications to FcRn-HPLC are required to enable interpretation of the data within a physiological context, to provide more accurate estimations of serum half-life. This study presents an important modification to this method, FcRn-pH-HPLC, which reproducibly measures FcRn dissociation pH, allowing correlation with previously established half-lives of therapeutic antibodies. Furthermore, the influence of incorporating various antibody modifications, binding modules, and their orientations within IgGs and bispecifics on FcRn dissociation pH was evaluated using antibodies from the redirected optimized cell killing (ROCK®) platform. Target and effector antigen-binding domain sequences, their presentation format and orientation within a bispecific antibody alter FcRn retention; tested Fc domain modifications and incorporating stabilizing disulfide bonds had minimal effect. This study may inform the generation of mono-, bi- and multi-specific antibodies with tailored half-lives based on FcRn binding properties in vitro, to differentiate antibody-based therapeutic candidates with optimal developability.
Collapse
Affiliation(s)
- Thomas Müller
- Discovery Research and Translational Immunology, Affimed GmbH, Heidelberg, Germany
| | - Carolin Tasser
- Discovery Research and Translational Immunology, Affimed GmbH, Heidelberg, Germany
| | - Michael Tesar
- Discovery Research and Translational Immunology, Affimed GmbH, Heidelberg, Germany
| | - Ivica Fucek
- Discovery Research and Translational Immunology, Affimed GmbH, Heidelberg, Germany
| | | | - Joachim Koch
- Discovery Research and Translational Immunology, Affimed GmbH, Heidelberg, Germany
| | - Kristina Ellwanger
- Discovery Research and Translational Immunology, Affimed GmbH, Heidelberg, Germany
| |
Collapse
|
23
|
Zielonka S, Krah S, Arras P, Lipinski B, Zimmermann J, Boje AS, Klausz K, Peipp M, Pekar L. Affinity Maturation of the Natural Ligand (B7-H6) for Natural Cytotoxicity Receptor NKp30 by Yeast Surface Display. Methods Mol Biol 2023; 2681:231-248. [PMID: 37405651 DOI: 10.1007/978-1-0716-3279-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
In recent years, the development of bispecific antibodies (bsAbs) has experienced tremendous progress for disease treatment, and consequently, a plethora of bsAbs is currently scrutinized in clinical trials. Besides antibody scaffolds, multifunctional molecules referred to as immunoligands have been developed. These molecules typically harbor a natural ligand entity for the engagement of a specific receptor, while binding to the additional antigen is facilitated by an antibody-derived paratope. Immunoligands can be exploited to conditionally activate immune cells, e.g., natural killer (NK) cells, in the presence of tumor cells, ultimately causing target-dependent tumor cell lysis. However, many ligands naturally show only moderate affinities toward their cognate receptor, potentially hampering killing capacities of immunoligands. Herein, we provide protocols for yeast surface display-based affinity maturation of B7-H6, the natural ligand of NK cell-activating receptor NKp30.
Collapse
Affiliation(s)
- Stefan Zielonka
- Protein Engineering and Antibody Technologies (PEAT), Merck Healthcare KGaA, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Simon Krah
- Protein Engineering and Antibody Technologies (PEAT), Merck Healthcare KGaA, Darmstadt, Germany
| | - Paul Arras
- Protein Engineering and Antibody Technologies (PEAT), Merck Healthcare KGaA, Darmstadt, Germany
| | - Britta Lipinski
- Protein Engineering and Antibody Technologies (PEAT), Merck Healthcare KGaA, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Jasmin Zimmermann
- Protein Engineering and Antibody Technologies (PEAT), Merck Healthcare KGaA, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Ammelie Svea Boje
- Stem Cell Transplantation and Immunotherapy, Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Katja Klausz
- Stem Cell Transplantation and Immunotherapy, Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Matthias Peipp
- Stem Cell Transplantation and Immunotherapy, Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Lukas Pekar
- Protein Engineering and Antibody Technologies (PEAT), Merck Healthcare KGaA, Darmstadt, Germany.
| |
Collapse
|
24
|
Kazandjian D, Kowalski A, Landgren O. T cell redirecting bispecific antibodies for multiple myeloma: emerging therapeutic strategies in a changing treatment landscape. Leuk Lymphoma 2022; 63:3032-3043. [PMID: 36059239 PMCID: PMC10113039 DOI: 10.1080/10428194.2022.2113532] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/20/2022] [Accepted: 08/02/2022] [Indexed: 01/11/2023]
Abstract
In recent years, the treatment landscape of multiple myeloma has continued to evolve with the introduction of novel immunotherapies. This progress has translated to improved overall survival for patients, but an unmet need remains in the heavily pretreated and high-risk subsets of patients. Emerging immunotherapies in the form of CAR-T cell therapies have been approved for multiple myeloma. However, CAR-T cell therapy has logistical limitations and there is a need for immunotherapies that are readily available, safe, and effective in RRMM. Currently, pending approval, there are many "off the shelf" bispecific antibodies being developed that target BCMA, GPRC5D, FcRH5 and other cell surface proteins. Preliminary efficacy data has suggested that these bispecific antibody therapies have similar response rates (∼50-80%) in heavily pretreated patients. Similarly, to CAR-T cell therapy, cytokine release syndrome and immune effector cell associated neurotoxicity syndrome are adverse events of key interest and incidence range from ∼40 to 90% and 3 to 20%, respectively. In this review, we highlight the various bispecific immunotherapies under development in the treatment of multiple myeloma with a focus on the data from clinical phase I and II studies.
Collapse
Affiliation(s)
- Dickran Kazandjian
- Myeloma Program, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami
| | - Andrew Kowalski
- Myeloma Program, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami
| | - Ola Landgren
- Myeloma Program, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami
| |
Collapse
|
25
|
Klausz K, Pekar L, Boje AS, Gehlert CL, Krohn S, Gupta T, Xiao Y, Krah S, Zaynagetdinov R, Lipinski B, Toleikis L, Poetzsch S, Rabinovich B, Peipp M, Zielonka S. Multifunctional NK Cell–Engaging Antibodies Targeting EGFR and NKp30 Elicit Efficient Tumor Cell Killing and Proinflammatory Cytokine Release. THE JOURNAL OF IMMUNOLOGY 2022; 209:1724-1735. [DOI: 10.4049/jimmunol.2100970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 08/23/2022] [Indexed: 01/04/2023]
Abstract
Abstract
In this work, we have generated novel Fc-comprising NK cell engagers (NKCEs) that bridge human NKp30 on NK cells to human epidermal growth factor receptor (EGFR) on tumor cells. Camelid-derived VHH single-domain Abs specific for human NKp30 and a humanized Fab derived from the EGFR-specific therapeutic Ab cetuximab were used as binding arms. By combining camelid immunization with yeast surface display, we were able to isolate a diverse panel of NKp30-specific VHHs against different epitopes on NKp30. Intriguingly, NKCEs built with VHHs that compete for binding to NKp30 with B7-H6, the natural ligand of NKp30, were significantly more potent in eliciting tumor cell lysis of EGFR-positive tumor cells than NKCEs harboring VHHs that target different epitopes on NKp30 from B7-H6. We demonstrate that the NKCEs can be further improved with respect to killing capabilities by concomitant engagement of FcγRIIIa and that soluble B7-H6 does not impede cytolytic capacities of all scrutinized NKCEs at significantly higher B7-H6 concentrations than observed in cancer patients. Moreover, we show that physiological processes requiring interactions between membrane-bound B7-H6 and NKp30 on NK cells are unaffected by noncompeting NKCEs still eliciting tumor cell killing at low picomolar concentrations. Ultimately, the NKCEs generated in this study were significantly more potent in eliciting NK cell–mediated tumor cell lysis than cetuximab and elicited a robust release of proinflammatory cytokines, both features which might be beneficial for antitumor therapy.
Collapse
Affiliation(s)
- Katja Klausz
- *Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian Albrechts University Kiel, Kiel, Germany
| | - Lukas Pekar
- †Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Ammelie Svea Boje
- *Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian Albrechts University Kiel, Kiel, Germany
| | - Carina Lynn Gehlert
- *Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian Albrechts University Kiel, Kiel, Germany
| | - Steffen Krohn
- *Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian Albrechts University Kiel, Kiel, Germany
| | - Tushar Gupta
- ‡Protein Engineering and Antibody Technologies, EMD Serono Research & Development Institute, Inc., Billerica, MA
| | - Yanping Xiao
- §Department of Oncology and Immuno-oncology, EMD Serono Research & Development Institute, Inc., Billerica, MA
| | - Simon Krah
- †Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Rinat Zaynagetdinov
- §Department of Oncology and Immuno-oncology, EMD Serono Research & Development Institute, Inc., Billerica, MA
| | - Britta Lipinski
- †Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
- ¶Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany; and
| | - Lars Toleikis
- †Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Sven Poetzsch
- ‖Strategic Innovation, Merck Healthcare KGaA, Darmstadt, Germany
| | - Brian Rabinovich
- §Department of Oncology and Immuno-oncology, EMD Serono Research & Development Institute, Inc., Billerica, MA
| | - Matthias Peipp
- *Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian Albrechts University Kiel, Kiel, Germany
| | - Stefan Zielonka
- †Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
- ¶Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany; and
| |
Collapse
|
26
|
Pinto S, Pahl J, Schottelius A, Carter PJ, Koch J. Reimagining antibody-dependent cellular cytotoxicity in cancer: the potential of natural killer cell engagers. Trends Immunol 2022; 43:932-946. [PMID: 36306739 DOI: 10.1016/j.it.2022.09.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 01/12/2023]
Abstract
Bi-, tri- and multispecific antibodies have enabled the development of targeted cancer immunotherapies redirecting immune effector cells to eliminate malignantly transformed cells. These antibodies allow for simultaneous binding of surface antigens on malignant cells and activating receptors on innate immune cells, such as natural killer (NK) cells, macrophages, and neutrophils. Significant progress with such antibodies has been achieved, particularly in hematological malignancies. Nevertheless, several major challenges remain, including increasing their immunotherapeutic efficacy in a greater proportion of patients, particularly in those harboring solid tumors, and overcoming dose-limiting toxicities and immunogenicity. Here, we discuss novel antibody-engineering developments designed to maximize the potential of NK cells by NK cell engagers mediating antibody-dependent cellular cytotoxicity (ADCC), thereby expanding the armamentarium for cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Paul J Carter
- Genentech, Department of Antibody Engineering, San Francisco, CA, USA
| | | |
Collapse
|
27
|
Harnessing natural killer cells for cancer immunotherapy: dispatching the first responders. Nat Rev Drug Discov 2022; 21:559-577. [PMID: 35314852 PMCID: PMC10019065 DOI: 10.1038/s41573-022-00413-7] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2022] [Indexed: 02/07/2023]
Abstract
Natural killer (NK) cells have crucial roles in the innate immunosurveillance of cancer and viral infections. They are 'first responders' that can spontaneously recognize abnormal cells in the body, rapidly eliminate them through focused cytotoxicity mechanisms and potently produce pro-inflammatory cytokines and chemokines that recruit and activate other immune cells to initiate an adaptive response. From the initial discovery of the diverse cell surface receptors on NK cells to the characterization of regulatory events that control their function, our understanding of the basic biology of NK cells has improved dramatically in the past three decades. This advanced knowledge has revealed increased mechanistic complexity, which has opened the doors to the development of a plethora of exciting new therapeutics that can effectively manipulate and target NK cell functional responses, particularly in cancer patients. Here, we summarize the basic mechanisms that regulate NK cell biology, review a wide variety of drugs, cytokines and antibodies currently being developed and used to stimulate NK cell responses, and outline evolving NK cell adoptive transfer approaches to treat cancer.
Collapse
|
28
|
Peipp M, Klausz K, Boje AS, Zeller T, Zielonka S, Kellner C. Immunotherapeutic targeting of activating natural killer cell receptors and their ligands in cancer. Clin Exp Immunol 2022; 209:22-32. [PMID: 35325068 PMCID: PMC9307233 DOI: 10.1093/cei/uxac028] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/15/2022] [Accepted: 03/22/2022] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells exert an important role in cancer immune surveillance. Recognition of malignant cells and controlled activation of effector functions are facilitated by the expression of activating and inhibitory receptors, which is a complex interplay that allows NK cells to discriminate malignant cells from healthy tissues. Due to their unique profile of effector functions, the recruitment of NK cells is attractive in cancer treatment and a key function of NK cells in antibody therapy is widely appreciated. In recent years, besides the low-affinity fragment crystallizable receptor for immunoglobulin G (FcγRIIIA), the activating natural killer receptors p30 (NKp30) and p46 (NKp46), as well as natural killer group 2 member D (NKG2D), have gained increasing attention as potential targets for bispecific antibody-derivatives to redirect NK cell cytotoxicity against tumors. Beyond modulation of the receptor activity on NK cells, therapeutic targeting of the respective ligands represents an attractive approach. Here, novel therapeutic approaches to unleash NK cells by engagement of activating NK-cell receptors and alternative strategies targeting their tumor-expressed ligands in cancer therapy are summarized.
Collapse
Affiliation(s)
- Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ammelie Svea Boje
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Tobias Zeller
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Stefan Zielonka
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Christian Kellner
- Correspondence: Christian Kellner, Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
29
|
Designing antibodies as therapeutics. Cell 2022; 185:2789-2805. [PMID: 35868279 DOI: 10.1016/j.cell.2022.05.029] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [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
|
30
|
Ordóñez-Reyes C, Garcia-Robledo JE, Chamorro DF, Mosquera A, Sussmann L, Ruiz-Patiño A, Arrieta O, Zatarain-Barrón L, Rojas L, Russo A, de Miguel-Perez D, Rolfo C, Cardona AF. Bispecific Antibodies in Cancer Immunotherapy: A Novel Response to an Old Question. Pharmaceutics 2022; 14:pharmaceutics14061243. [PMID: 35745815 PMCID: PMC9229626 DOI: 10.3390/pharmaceutics14061243] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 01/14/2023] Open
Abstract
Immunotherapy has redefined the treatment of cancer patients and it is constantly generating new advances and approaches. Among the multiple options of immunotherapy, bispecific antibodies (bsAbs) represent a novel thoughtful approach. These drugs integrate the action of the immune system in a strategy to redirect the activation of innate and adaptive immunity toward specific antigens and specific tumor locations. Here we discussed some basic aspects of the design and function of bsAbs, their main challenges and the state-of-the-art of these molecules in the treatment of hematological and solid malignancies and future perspectives.
Collapse
Affiliation(s)
- Camila Ordóñez-Reyes
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
| | - Juan Esteban Garcia-Robledo
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ 85054, USA
| | - Diego F. Chamorro
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
| | - Andrés Mosquera
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
| | - Liliana Sussmann
- Department of Neurology, Fundación Universitaria de Ciencias de la Salud, Bogotá 111221, Colombia;
| | - Alejandro Ruiz-Patiño
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
| | - Oscar Arrieta
- Thoracic Oncology Unit and Personalized Oncology Laboratory, National Cancer Institute (INCan), Mexico City 14080, Mexico; (O.A.); (L.Z.-B.)
| | - Lucia Zatarain-Barrón
- Thoracic Oncology Unit and Personalized Oncology Laboratory, National Cancer Institute (INCan), Mexico City 14080, Mexico; (O.A.); (L.Z.-B.)
| | - Leonardo Rojas
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
| | | | - Diego de Miguel-Perez
- Center for Thoracic Oncology, Tisch Cancer Institute and Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (D.d.M.-P.); (C.R.)
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute and Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (D.d.M.-P.); (C.R.)
| | - Andrés F. Cardona
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
- Direction of Research, Science and Education, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC), Bogotá 110131, Colombia
- Correspondence: ; Tel.: +57-(1)-6190052; Fax: +57-(1)-6190053
| |
Collapse
|
31
|
Vaněk O, Kalousková B, Abreu C, Nejadebrahim S, Skořepa O. Natural killer cell-based strategies for immunotherapy of cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 129:91-133. [PMID: 35305726 DOI: 10.1016/bs.apcsb.2022.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Natural killer (NK) cells are a family of lymphocytes with a natural ability to kill infected, harmed, or malignantly transformed cells. As these cells are part of the innate immunity, the cytotoxic mechanisms are activated upon recognizing specific patterns without prior antigen sensitization. This recognition is crucial for NK cell function in the maintenance of homeostasis and immunosurveillance. NK cells not only act directly toward malignant cells but also participate in the complex immune response by producing cytokines or cross-talk with other immune cells. Cancer may be seen as a break of all immune defenses when malignant cells escape the immunity and invade surrounding tissues creating a microenvironment supporting tumor progression. This process may be reverted by intervening immune response with immunotherapy, which may restore immune recognition. NK cells are important effector cells for immunotherapy. They may be used for adoptive cell transfer, genetically modified with chimeric antigen receptors, or triggered with appropriate antibodies and other antibody-fragment-based recombinant therapeutic proteins tailored specifically for NK cell engagement. NK cell receptors, responsible for target recognition and activation of cytotoxic response, could also be targeted in immunotherapy, for example, by various bi-, tri-, or multi-specific fusion proteins designed to bridge the gap between tumor markers present on target cells and activation receptors expressed on NK cells. However, this kind of immunoactive therapeutics may be developed only with a deep functional and structural knowledge of NK cell receptor: ligand interactions. This review describes the recent developments in the fascinating protein-engineering field of NK cell immunotherapeutics.
Collapse
Affiliation(s)
- Ondřej Vaněk
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic.
| | - Barbora Kalousková
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Celeste Abreu
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Shiva Nejadebrahim
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Ondřej Skořepa
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
32
|
Chen RP, Shinoda K, Rampuria P, Jin F, Bartholomew T, Zhao C, Yang F, Chaparro-Riggers J. Bispecific antibodies for immune cell retargeting against cancer. Expert Opin Biol Ther 2022; 22:965-982. [PMID: 35485219 DOI: 10.1080/14712598.2022.2072209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Following the approval of the T-cell engaging bispecific antibody blinatumomab, immune cell retargeting with bispecific or multispecific antibodies has emerged as a promising cancer immunotherapy strategy, offering alternative mechanisms compared to immune checkpoint blockade. As we gain more understanding of the complex tumor microenvironment, rules and design principles have started to take shape on how to best harness the immune system to achieve optimal anti-tumor activities. AREAS COVERED In the present review, we aim to summarize the most recent advances and challenges in using bispecific antibodies for immune cell retargeting and to provide insights into various aspects of antibody engineering. Discussed herein are studies that highlight the importance of considering antibody engineering parameters, such as binding epitope, affinity, valency, and geometry to maximize the potency and mitigate the toxicity of T cell engagers. Beyond T cell engaging bispecifics, other bispecifics designed to recruit the innate immune system are also covered. EXPERT OPINION Diverse and innovative molecular designs of bispecific/multispecific antibodies have the potential to enhance the efficacy and safety of immune cell retargeting for the treatment of cancer. Whether or not clinical data support these different hypotheses, especially in solid tumor settings, remains to be seen.
Collapse
Affiliation(s)
- Rebecca P Chen
- Pfizer BioMedicine Design, Pfizer Inc, San Diego, CA, USA
| | - Kenta Shinoda
- Pfizer BioMedicine Design, Pfizer Inc, Cambridge, MA, USA
| | | | - Fang Jin
- Pfizer BioMedicine Design, Pfizer Inc, Cambridge, MA, USA
| | | | - Chunxia Zhao
- Pfizer BioMedicine Design, Pfizer Inc, Cambridge, MA, USA
| | - Fan Yang
- Pfizer BioMedicine Design, Pfizer Inc, San Diego, CA, USA
| | | |
Collapse
|
33
|
Tarannum M, Romee R, Shapiro RM. Innovative Strategies to Improve the Clinical Application of NK Cell-Based Immunotherapy. Front Immunol 2022; 13:859177. [PMID: 35401529 PMCID: PMC8990319 DOI: 10.3389/fimmu.2022.859177] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/28/2022] [Indexed: 12/31/2022] Open
Abstract
Natural killer cells constitute a part of the innate immune system that mediates an effective immune response towards virus-infected and malignant cells. In recent years, research has focused on exploring and advancing NK cells as an active immunotherapy platform. Despite major advances, there are several key challenges that need to be addressed for the effective translation of NK cell research to clinical applications. This review highlights some of these challenges and the innovative strategies being developed to overcome them, including in vitro expansion, in vivo persistence, infiltration to the tumor site, and prevention of exhaustion.
Collapse
Affiliation(s)
- Mubin Tarannum
- Division of Stem Cell Transplant and Cellular Therapy, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Rizwan Romee
- Division of Stem Cell Transplant and Cellular Therapy, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Roman M Shapiro
- Division of Stem Cell Transplant and Cellular Therapy, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
34
|
Nahi H, Chrobok M, Meinke S, Gran C, Marquardt N, Afram G, Sutlu T, Gilljam M, Stellan B, Wagner AK, Blomberg P, Holmqvist PH, Walther-Jallow L, Mellström K, Liwing J, Gustafsson C, Månsson R, Klimkowska M, Gahrton G, Lund J, Ljungman P, Ljunggren HG, Alici E. Autologous NK cells as consolidation therapy following stem cell transplantation in multiple myeloma. Cell Rep Med 2022; 3:100508. [PMID: 35243416 PMCID: PMC8861830 DOI: 10.1016/j.xcrm.2022.100508] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 11/03/2021] [Accepted: 01/05/2022] [Indexed: 02/07/2023]
Abstract
Few approaches have been made toward exploring autologous NK cells in settings of cancer immunotherapy. Here, we demonstrate the feasibility of infusing multiple doses of ex vivo activated and expanded autologous NK cells in patients with multiple myeloma (MM) post-autologous stem cell transplantation. Infused NK cells were detected in circulation up to 4 weeks after the last infusion. Elevations in plasma granzyme B levels were observed following each consecutive NK cell infusion. Moreover, increased granzyme B levels were detected in bone marrow 4 weeks after the last infusion. All measurable patients had objective, detectable responses after NK cell infusions in terms of reduction in M-component and/or minimal residual disease. The present study demonstrates that autologous NK cell-based immunotherapy is feasible in a setting of MM consolidation therapy. It opens up the possibility for usage of autologous NK cells in clinical settings where patients are not readily eligible for allogeneic NK cell-based immunotherapies. Infusing activated and expanded autologous NK cells in patients with MM is possible Infused NK cells are detected in circulation for up to 4 weeks Elevated granzyme B levels are observed following each consecutive NK cell infusion Objective, detectable responses after NK cell infusions are seen in patients
Collapse
Affiliation(s)
- Hareth Nahi
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden.,Department of Hematology, Karolinska University Hospital, SE-14186 Huddinge, Sweden
| | - Michael Chrobok
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden
| | - Stephan Meinke
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden
| | - Charlotte Gran
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden.,Department of Clinical Chemistry, Karolinska University Laboratory, SE-14183 Huddinge, Sweden
| | - Nicole Marquardt
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden
| | - Gabriel Afram
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden.,Department of Hematology, Karolinska University Hospital, SE-14186 Huddinge, Sweden
| | - Tolga Sutlu
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden
| | - Mari Gilljam
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden
| | - Birgitta Stellan
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden
| | - Arnika K Wagner
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden
| | - Pontus Blomberg
- Vecura, Department of Laboratory Medicine, Karolinska Institutet, SE-14186 Stockholm, Sweden.,Vecura, Karolinska Cell Therapy Center, Karolinska University Hospital, SE-14186 Stockholm, Sweden
| | - Per-Henrik Holmqvist
- Vecura, Karolinska Cell Therapy Center, Karolinska University Hospital, SE-14186 Stockholm, Sweden
| | - Lilian Walther-Jallow
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, SE-14183 Huddinge, Sweden
| | - Karin Mellström
- XNK Therapeutics AB, Hälsovägen 7, Novum, SE-14157 Huddinge, Sweden
| | - Johan Liwing
- XNK Therapeutics AB, Hälsovägen 7, Novum, SE-14157 Huddinge, Sweden
| | - Charlotte Gustafsson
- Center for Hematology and Regenerative Medicine, Division of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institutet, SE-14183 Huddinge, Sweden
| | - Robert Månsson
- Center for Hematology and Regenerative Medicine, Division of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institutet, SE-14183 Huddinge, Sweden
| | - Monika Klimkowska
- Pathology Unit, Department of Laboratory Medicine, Karolinska Institutet, SE-14183 Huddinge, Sweden.,Department of Clinical Pathology and Cytology, Karolinska University Hospital, SE-14186 Huddinge, Sweden
| | - Gösta Gahrton
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden
| | - Johan Lund
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden.,Department of Hematology, Karolinska University Hospital, SE-14186 Huddinge, Sweden
| | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital Huddinge, SE-14186 Huddinge, Sweden.,Division of Hematology, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden
| | - Hans-Gustaf Ljunggren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden
| | - Evren Alici
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, SE-14183 Huddinge, Sweden
| |
Collapse
|
35
|
Reusch U, Ellwanger K, Fucek I, Müller T, Schniegler-Mattox U, Koch J, Tesar M. Cryopreservation of Natural Killer Cells Pre-Complexed with Innate Cell Engagers. Antibodies (Basel) 2022; 11:antib11010012. [PMID: 35225870 PMCID: PMC8883965 DOI: 10.3390/antib11010012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/26/2022] [Accepted: 02/03/2022] [Indexed: 12/21/2022] Open
Abstract
Innate cell engager (ICE®) constructs are bispecific tetravalent antibodies targeting specific tumor antigens and simultaneously engaging natural killer (NK) cell and macrophage receptors for the destruction of tumor cells. Pre-complexing of ICE® constructs with adoptive NK cells is a novel approach to enhance NK cell activity. The suitability of such complexes for cryopreservation, whilst retaining the biological activity and specificity, may enable the development of off-the-shelf NK cell products. This study investigates the binding affinity of ICE® constructs targeting EpCAM and NK cell receptors CD16A, NKG2D, or NKp46 to the corresponding antigens, the ICE® antitumor activity, and feasibility of cryopreservation. Cell surface retention assays using primary NK cells confirmed a substantially slower ICE® construct dissociation kinetics compared with control molecules, suggesting the formation of durable complexes independently of the CD16A polymorphism. The high-affinity NK cell and EpCAM/CD16A ICE® complexes were superior to those engaging NKG2D or NKp46 receptors when tested for the NK-cell-mediated elimination of EpCAM-expressing tumor cells. Moreover, the potency and efficacy of these complexes were unaffected after a single freeze–thaw cycle. CD16A-selective ICE® drug candidates complexed with NK cells hold promise as novel cryopreserved off-the-shelf NK cell products with chimeric antigen receptor-like NK cell properties, capable of effective depletion of tumor cells.
Collapse
|
36
|
Abstract
Hodgkin lymphoma is a B-cell malignancy with approximately 85-95% complete remission rate following frontline therapy; however, relapsed/refractory disease occurs in roughly 10-30% of patients after treatment. Salvage therapy conventionally relies upon cytotoxic chemotherapy followed by high-dose chemotherapy and autologous stem cell transplantation. A considerable number of patients experience relapse after transplantation, and further salvage management has included the use of allogeneic transplantation and radiotherapy. In the past decade, novel therapies including, brentuximab vedotin, PD-1 inhibitors, and the incorporation of PET-imaging into management have changed the paradigm of relapsed/refractory disease care. Novel therapies have been investigated in both single and combination regimens with other novel therapies and traditional chemotherapies. There is promising early work into the utility of CD30.CAR-T cell therapy, AFM13, camidanlumab tesirine, novel PD-1 inhibitors, and JAK1/JAK2 inhibition in management. Herein, we will review current salvage therapies in Hodgkin lymphoma and future directions in relapsed/refractory disease management.
Collapse
Affiliation(s)
- Karan Chohan
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
37
|
Kakiuchi-Kiyota S, Ross T, Wallweber HA, Kiefer JR, Schutten MM, Adedeji AO, Cai H, Hendricks R, Cohen S, Myneni S, Liu L, Fullerton A, Corr N, Yu L, de Almeida Nagata D, Zhong S, Leong SR, Li J, Nakamura R, Sumiyoshi T, Li J, Ovacik AM, Zheng B, Dillon M, Spiess C, Wingert S, Rajkovic E, Ellwanger K, Reusch U, Polson AG. A BCMA/CD16A bispecific innate cell engager for the treatment of multiple myeloma. Leukemia 2022; 36:1006-1014. [PMID: 35001074 DOI: 10.1038/s41375-021-01478-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/05/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022]
Abstract
Despite the recent progress, multiple myeloma (MM) is still essentially incurable and there is a need for additional effective treatments with good tolerability. RO7297089 is a novel bispecific BCMA/CD16A-directed innate cell engager (ICE®) designed to induce BCMA+ MM cell lysis through high affinity binding of CD16A and retargeting of NK cell cytotoxicity and macrophage phagocytosis. Unlike conventional antibodies approved in MM, RO7297089 selectively targets CD16A with no binding of other Fcγ receptors, including CD16B on neutrophils, and irrespective of 158V/F polymorphism, and its activity is less affected by competing IgG suggesting activity in the presence of M-protein. Structural analysis revealed this is due to selective interaction with a single residue (Y140) uniquely present in CD16A opposite the Fc binding site. RO7297089 induced tumor cell killing more potently than conventional antibodies (wild-type and Fc-enhanced) and induced lysis of BCMA+ cells at very low effector-to-target ratios. Preclinical toxicology data suggested a favorable safety profile as in vitro cytokine release was minimal and no RO7297089-related mortalities or adverse events were observed in cynomolgus monkeys. These data suggest good tolerability and the potential of RO7297089 to be a novel effective treatment of MM patients.
Collapse
Affiliation(s)
| | | | | | - James R Kiefer
- Genentech Research and Early Development, San Francisco, CA, USA
| | | | | | - Hao Cai
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Robert Hendricks
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Sivan Cohen
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Srividya Myneni
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Luna Liu
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Aaron Fullerton
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Nicholas Corr
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Lanlan Yu
- Genentech Research and Early Development, San Francisco, CA, USA
| | | | - Shelly Zhong
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Steven R Leong
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Ji Li
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Rin Nakamura
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Teiko Sumiyoshi
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Jinze Li
- Genentech Research and Early Development, San Francisco, CA, USA
| | | | - Bing Zheng
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Mike Dillon
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Christoph Spiess
- Genentech Research and Early Development, San Francisco, CA, USA
| | | | | | | | | | - Andrew G Polson
- Genentech Research and Early Development, San Francisco, CA, USA.
| |
Collapse
|
38
|
Yanakieva D, Pekar L, Evers A, Fleischer M, Keller S, Mueller-Pompalla D, Toleikis L, Kolmar H, Zielonka S, Krah S. Beyond bispecificity: Controlled Fab arm exchange for the generation of antibodies with multiple specificities. MAbs 2022; 14:2018960. [PMID: 35014603 PMCID: PMC8757479 DOI: 10.1080/19420862.2021.2018960] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/13/2021] [Indexed: 01/07/2023] Open
Abstract
Controlled Fab arm exchange (cFAE) has proven to be a generic and versatile technology for the efficient generation of IgG-like bispecific antibodies (DuoBodies or DBs), with several in clinical development and one product, amivantamab, approved by the Food and Drug Administration. In this study, we expand the cFAE-toolbox by incorporating VHH-modules at the C-termini of DB-IgGs, termed DB-VHHs. This approach enables the combinatorial generation of tri- and tetraspecific molecules with flexible valencies in a straightforward fashion. Using cFAE, a variety of multispecific molecules was produced and assessed for manufacturability and physicochemical characteristics. In addition, we were able to generate DB-VHHs that efficiently triggered natural killer cell mediated lysis of tumor cells, demonstrating the utility of this format for potential therapeutic applications.
Collapse
Affiliation(s)
- Desislava Yanakieva
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
- Protein Engineering and Antibody Technologies, Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Lukas Pekar
- Protein Engineering and Antibody Technologies, Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Andreas Evers
- Protein Engineering and Antibody Technologies, Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Markus Fleischer
- Protein and Cell Sciences, Merck Healthcare KGaA, Darmstadt, Germany
| | - Stephan Keller
- Protein and Cell Sciences, Merck Healthcare KGaA, Darmstadt, Germany
| | | | - Lars Toleikis
- Protein Engineering and Antibody Technologies, Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Stefan Zielonka
- Protein Engineering and Antibody Technologies, Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Simon Krah
- Protein Engineering and Antibody Technologies, Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| |
Collapse
|
39
|
Bi-specific and Tri-specific NK Cell Engagers: The New Avenue of Targeted NK Cell Immunotherapy. Mol Diagn Ther 2021; 25:577-592. [PMID: 34327614 DOI: 10.1007/s40291-021-00550-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2021] [Indexed: 02/01/2023]
Abstract
Natural killer (NK) cell-mediated cancer immunotherapy has grown significantly over the past two decades. More recently, multi-specific engagers have been developed as cancer therapeutics to effectively arm endogenous NK cells to more potently induce specific cytolytic responses against tumor targets. This review explores the bi- and tri-specific NK/tumor engagers that are emerging as a new generation of immunotherapeutics. These molecules vary in configuration, but they typically have small molecular weights and domains that engage specific tumor antigens and NK cell-activating receptors such as CD16, NKp30, NKp46, and NKG2D. They have demonstrated compelling potential in boosting NK cell cytotoxicity against specific tumor targets. This highly adaptable off-the-shelf platform, which in some formats also integrates cytokines, is poised to revolutionize targeted NK cell immunotherapy, either as a monotherapy or in combination with other effective anti-cancer therapies.
Collapse
|
40
|
Wingert S, Reusch U, Knackmuss S, Kluge M, Damrat M, Pahl J, Schniegler-Mattox U, Mueller T, Fucek I, Ellwanger K, Tesar M, Haneke T, Koch J, Treder M, Fischer W, Rajkovic E. Preclinical evaluation of AFM24, a novel CD16A-specific innate immune cell engager targeting EGFR-positive tumors. MAbs 2021; 13:1950264. [PMID: 34325617 PMCID: PMC8331026 DOI: 10.1080/19420862.2021.1950264] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epidermal growth factor receptor (EGFR)-targeted cancer therapy such as anti-EGFR monoclonal antibodies and tyrosine kinase inhibitors have demonstrated clinical efficacy. However, there remains a medical need addressing limitations of these therapies, which include a narrow therapeutic window mainly due to skin and organ toxicity, and primary and secondary resistance mechanisms of the EGFR-signaling cascade (e.g., RAS-mutated colorectal cancer). Using the redirected optimized cell killing (ROCK®) antibody platform, we have developed AFM24, a novel bispecific, IgG1-scFv fusion antibody targeting CD16A on innate immune cells, and EGFR on tumor cells. We herein demonstrate binding of AFM24 to CD16A on natural killer (NK) cells and macrophages with KD values in the low nanomolar range and to various EGFR-expressing tumor cells. AFM24 was highly potent and effective for antibody-dependent cell-mediated cytotoxicity via NK cells, and also mediated antibody-dependent cellular phagocytosis via macrophages in vitro. Importantly, AFM24 was effective toward a variety of EGFR-expressing tumor cells, regardless of EGFR expression level and KRAS/BRAF mutational status. In vivo, AFM24 was well tolerated up to the highest dose (75 mg/kg) when administered to cynomolgus monkeys once weekly for 28 days. Notably, skin and other toxicities were not observed. A transient elevation of interleukin-6 levels was detected at all dose levels, 2-4 hours post-dose, which returned to baseline levels after 24 hours. These results emphasize the promise of bispecific innate cell engagers as an alternative cancer therapy and demonstrate the potential for AFM24 to effectively target tumors expressing varying levels of EGFR, regardless of their mutational status.Abbreviations: ADA: antidrug antibody; ADCC: antibody-dependent cell-mediated cytotoxicity; ADCP: antibody-dependent cellular phagocytosis; AUC: area under the curve; CAR: chimeric-antigen receptor; CD: Cluster of differentiation; CRC :colorectal cancer; ECD: extracellular domain; EGF: epidermal growth factorEGFR epidermal growth factor receptor; ELISA: enzyme-linked immunosorbent assay; FACS: fluorescence-activated cell sorting; Fc: fragment, crystallizableFv variable fragment; HNSCC: head and neck squamous carcinomaIL interleukinm; Ab monoclonal antibody; MOA: mechanism of action; NK :natural killer; NSCLC: non-small cell lung cancer; PBMC: peripheral blood mononuclear cell; PBS: phosphate-buffered saline; PD: pharmacodynamic; ROCK: redirected optimized cell killing; RSV: respiratory syncytial virus; SABC: specific antibody binding capacity; SD: standard deviation; TAM: tumor-associated macrophage; TKI: tyrosine kinase inhibitor; WT: wildtype.
Collapse
Affiliation(s)
| | - Uwe Reusch
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | | | - Michael Kluge
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | - Michael Damrat
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | - Jens Pahl
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | | | - Thomas Mueller
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | - Ivica Fucek
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | | | - Michael Tesar
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | - Torsten Haneke
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | - Joachim Koch
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | - Martin Treder
- Formerly Affimed GmbH, Heidelberg, Germany. Now: Arjuna Therapeutics, Santiago De Compostela, Spain
| | | | - Erich Rajkovic
- Research & Development, Affimed GmbH, Heidelberg, Germany
| |
Collapse
|
41
|
A functional mammalian display screen identifies rare antibodies that stimulate NK cell-mediated cytotoxicity. Proc Natl Acad Sci U S A 2021; 118:2104099118. [PMID: 34330834 DOI: 10.1073/pnas.2104099118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Therapies that boost the antitumor immune response have shown a great deal of success. Although most of these therapies have focused on enhancing T cell functions, there is a growing interest in developing therapies that can target other immune cell subsets. Like T cells, natural killer (NK) cells are cytotoxic effector cells that play a key role in the antitumor response. To advance the development of NK-based therapies, we developed a functional screen to rapidly identify antibodies that can activate NK cells. We displayed antibodies on a mammalian target cell line and probed their ability to stimulate NK cell-mediated cytotoxicity. From this screen, we identified five antibodies that bound with high affinity to NK cells and stimulated NK cell-mediated cytotoxicity and interferon-γ (IFN-γ) secretion. We demonstrate that these antibodies can be further developed into bispecific antibodies to redirect NK cell-mediated cytotoxicity toward CD20+ B cell lymphoma cells and HER2+ breast cancer cells. While antibodies to two of the receptors, CD16 and NCR1, have previously been targeted as bispecific antibodies to redirect NK cell-mediated cytotoxicity, we demonstrate that bispecific antibodies targeting NCR3 can also potently activate NK cells. These results show that this screen can be used to directly identify antibodies that can enhance antitumor immune responses.
Collapse
|
42
|
Capuano C, Pighi C, Battella S, De Federicis D, Galandrini R, Palmieri G. Harnessing CD16-Mediated NK Cell Functions to Enhance Therapeutic Efficacy of Tumor-Targeting mAbs. Cancers (Basel) 2021; 13:cancers13102500. [PMID: 34065399 PMCID: PMC8161310 DOI: 10.3390/cancers13102500] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/18/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Natural Killer (NK) cells play a major role in cancer immunotherapy based on tumor-targeting mAbs. NK cell-mediated tumor cell killing and cytokine secretion are powerfully stimulated upon interaction with IgG-opsonized tumor cells, through the aggregation of FcγRIIIA/CD16 IgG receptor. Advances in basic and translational NK cell biology have led to the development of strategies that, by improving mAb-dependent antitumor responses, may overcome the current limitations of antibody therapy attributable to tolerance, immunosuppressive microenvironment, and genotypic factors. This review provides an overview of the immunotherapeutic strategies being pursued to improve the efficacy of mAb-induced NK antitumor activity. The exploitation of antibody combinations, antibody-based molecules, used alone or combined with adoptive NK cell therapy, will be uncovered. Within the landscape of NK cell heterogeneity, we stress the role of memory NK cells as promising effectors in the next generation of immunotherapy with the aim to obtain long-lasting tumor control. Abstract Natural killer (NK) cells hold a pivotal role in tumor-targeting monoclonal antibody (mAb)-based activity due to the expression of CD16, the low-affinity receptor for IgG. Indeed, beyond exerting cytotoxic function, activated NK cells also produce an array of cytokines and chemokines, through which they interface with and potentiate adaptive immune responses. Thus, CD16-activated NK cells can concur to mAb-dependent “vaccinal effect”, i.e., the development of antigen-specific responses, which may be highly relevant in maintaining long-term protection of treated patients. On this basis, the review will focus on strategies aimed at potentiating NK cell-mediated antitumor functions in tumor-targeting mAb-based regimens, represented by (a) mAb manipulation strategies, aimed at augmenting recruitment and efficacy of NK cells, such as Fc-engineering, and the design of bi- or trispecific NK cell engagers and (b) the possible exploitation of memory NK cells, whose distinctive characteristics (enhanced responsiveness to CD16 engagement, longevity, and intrinsic resistance to the immunosuppressive microenvironment) may maximize therapeutic mAb antitumor efficacy.
Collapse
Affiliation(s)
- Cristina Capuano
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.C.); (C.P.); (S.B.); (D.D.F.)
| | - Chiara Pighi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.C.); (C.P.); (S.B.); (D.D.F.)
| | - Simone Battella
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.C.); (C.P.); (S.B.); (D.D.F.)
- ReiThera Srl, 00128 Rome, Italy
| | - Davide De Federicis
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.C.); (C.P.); (S.B.); (D.D.F.)
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Ricciarda Galandrini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.C.); (C.P.); (S.B.); (D.D.F.)
- Correspondence: (R.G.); (G.P.)
| | - Gabriella Palmieri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.C.); (C.P.); (S.B.); (D.D.F.)
- Correspondence: (R.G.); (G.P.)
| |
Collapse
|
43
|
van Faassen H, Jo DH, Ryan S, Lowden MJ, Raphael S, MacKenzie CR, Lee SH, Hussack G, Henry KA. Incorporation of a Novel CD16-Specific Single-Domain Antibody into Multispecific Natural Killer Cell Engagers With Potent ADCC. Mol Pharm 2021; 18:2375-2384. [PMID: 33999642 DOI: 10.1021/acs.molpharmaceut.1c00208] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Multispecific antibodies that bridge immune effector and tumor cells have shown promising preclinical and clinical efficacies. Here, we isolated and characterized novel llama single-domain antibodies (sdAbs) against CD16. One sdAb, NRC-sdAb048, bound recombinant human and cynomolgus monkey CD16 ectodomains with equivalent affinity (KD: 1 nM) but did not recognize murine CD16. Binding was similar for human CD16a expressed on NK cells and CD16b (NA2) expressed on neutrophils but dramatically weaker (KD: ∼6 μM) for the CD16b (NA1) allotype. The sdAb stained primary human peripheral blood NK cells. Irrespective of fusion orientation and linker length, bispecific sdAb-sdAb and sdAb-scFv dimers (anti-CD16/EGFR, anti-CD16/HER2, and anti-CD16/CD19) retained full binding affinity for each target, coengaged both antigens simultaneously, elicited ADCC against target antigen-expressing tumor cells in a reporter bioassay, and triggered target-specific activation and degranulation of primary NK cells as measured via interferon-γ and CD107a expression. These molecules may have applications in cancer immunotherapy.
Collapse
Affiliation(s)
- Henk van Faassen
- Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada
| | - Dong-Hyeon Jo
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Shannon Ryan
- Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada
| | - Michael J Lowden
- Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada
| | - Shalini Raphael
- Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada
| | - C Roger MacKenzie
- Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Greg Hussack
- Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada
| | - Kevin A Henry
- Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada.,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
44
|
A phase 1b study of AFM13 in combination with pembrolizumab in patients with relapsed or refractory Hodgkin lymphoma. Blood 2021; 136:2401-2409. [PMID: 32730586 DOI: 10.1182/blood.2019004701] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 07/01/2020] [Indexed: 12/26/2022] Open
Abstract
In relapsed/refractory Hodgkin lymphoma (R/R HL), immunotherapies such as the anti-programmed death-1 inhibitor pembrolizumab have demonstrated efficacy as monotherapy and are playing an increasingly prominent role in treatment. The CD30/CD16A-bispecific antibody AFM13 is an innate immune cell engager, a first-in-class, tetravalent antibody, designed to create a bridge between CD30 on HL cells and the CD16A receptor on natural killer cells and macrophages, to induce tumor cell killing. Early studies of AFM13 have demonstrated signs of efficacy as monotherapy for patients with R/R HL and the combination of AFM13 with pembrolizumab represents a rational new treatment modality. Here, we describe a phase 1b, dose-escalation study to assess the safety and preliminary efficacy of AFM13 in combination with pembrolizumab in patients with R/R HL. The primary objective was estimating the maximum tolerated dose; the secondary objectives were to assess safety, tolerability, antitumor efficacy, pharmacokinetics, and pharmacodynamics. In this heavily pretreated patient population, treatment with the combination of AFM13 and pembrolizumab was generally well tolerated, with similar safety profiles compared to the known profiles of each agent alone. The combination of AFM13 with pembrolizumab demonstrated an objective response rate of 88% at the highest treatment dose, with an 83% overall response rate for the overall population. Pharmacokinetic assessment of AFM13 in the combination setting revealed a half-life of up to 20.6 hours. This proof-of-concept study holds promise as a novel immunotherapy combination worthy of further investigation. This phase 1b study was registered at www.clinicaltrials.gov as NCT02665650.
Collapse
|
45
|
Lamb MG, Rangarajan HG, Tullius BP, Lee DA. Natural killer cell therapy for hematologic malignancies: successes, challenges, and the future. Stem Cell Res Ther 2021; 12:211. [PMID: 33766099 PMCID: PMC7992329 DOI: 10.1186/s13287-021-02277-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/10/2021] [Indexed: 12/20/2022] Open
Abstract
The adoptive transfer of natural killer (NK) cells is an emerging therapy in the field of immuno-oncology. In the last 3 decades, NK cells have been utilized to harness the anti-tumor immune response in a wide range of malignancies, most notably with early evidence of efficacy in hematologic malignancies. NK cells are dysfunctional in patients with hematologic malignancies, and their number and function are further impaired by chemotherapy, radiation, and immunosuppressants used in initial therapy and hematopoietic stem cell transplantation. Restoring this innate immune deficit may lead to improved therapeutic outcomes. NK cell adoptive transfer has proven to be a safe in these settings, even in the setting of HLA mismatch, and a deeper understanding of NK cell biology and optimized expansion techniques have improved scalability and therapeutic efficacy. Here, we review the use of NK cell therapy in hematologic malignancies and discuss strategies to further improve the efficacy of NK cells against these diseases.
Collapse
Affiliation(s)
- Margaret G Lamb
- Division of Hematology, Oncology, and Bone Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Suite 5A.1, Columbus, OH, 43205-2664, USA. .,Department of Pediatrics, The Ohio State University School of Medicine, Columbus, OH, USA.
| | - Hemalatha G Rangarajan
- Division of Hematology, Oncology, and Bone Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Suite 5A.1, Columbus, OH, 43205-2664, USA.,Department of Pediatrics, The Ohio State University School of Medicine, Columbus, OH, USA
| | - Brian P Tullius
- Division of Hematology, Oncology, and Bone Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Suite 5A.1, Columbus, OH, 43205-2664, USA.,Department of Pediatrics, The Ohio State University School of Medicine, Columbus, OH, USA
| | - Dean A Lee
- Division of Hematology, Oncology, and Bone Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Suite 5A.1, Columbus, OH, 43205-2664, USA.,Department of Pediatrics, The Ohio State University School of Medicine, Columbus, OH, USA
| |
Collapse
|
46
|
Pekar L, Klausz K, Busch M, Valldorf B, Kolmar H, Wesch D, Oberg HH, Krohn S, Boje AS, Gehlert CL, Toleikis L, Krah S, Gupta T, Rabinovich B, Zielonka S, Peipp M. Affinity Maturation of B7-H6 Translates into Enhanced NK Cell-Mediated Tumor Cell Lysis and Improved Proinflammatory Cytokine Release of Bispecific Immunoligands via NKp30 Engagement. THE JOURNAL OF IMMUNOLOGY 2020; 206:225-236. [PMID: 33268483 DOI: 10.4049/jimmunol.2001004] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023]
Abstract
Activating NK cell receptors represent promising target structures to elicit potent antitumor immune responses. In this study, novel immunoligands were generated that bridge the activating NK cell receptor NKp30 on NK cells with epidermal growth factor receptor (EGFR) on tumor cells in a bispecific IgG-like format based on affinity-optimized versions of B7-H6 and the Fab arm derived from cetuximab. To enhance NKp30 binding, the solitary N-terminal IgV domain of B7-H6 (ΔB7-H6) was affinity matured by an evolutionary library approach combined with yeast surface display. Biochemical and functional characterization of 36 of these novel ΔB7-H6-derived NK cell engagers revealed an up to 45-fold-enhanced affinity for NKp30 and significantly improved NK cell-mediated, EGFR-dependent killing of tumor cells compared with the NK cell engager based on the wild-type ΔB7-H6 domain. In this regard, potencies (EC50 killing) of the best immunoligands were substantially improved by up to 87-fold. Moreover, release of IFN-γ and TNF-α was significantly increased. Importantly, equipment of the ΔB7-H6-based NK cell engagers with a human IgG1 Fc part competent in Fc receptor binding resulted in an almost 10-fold superior killing of EGFR-overexpressing tumor cells compared with molecules either triggering FcγRIIIa or NKp30. Additionally, INF-γ and TNF-α release was increased compared with molecules solely triggering FcγRIIIa, including the clinically approved Ab cetuximab. Thus, incorporating affinity-matured ligands for NK cell-activating receptors might represent an effective strategy for the generation of potent novel therapeutic agents with unique effector functions in cancer immunotherapy.
Collapse
Affiliation(s)
- Lukas Pekar
- Protein Engineering and Antibody Technologies, Merck KGaA, D-64293 Darmstadt, Germany.,Discovery Pharmacology, Merck KGaA, D-64293 Darmstadt, Germany
| | - Katja Klausz
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, University Hospital Schleswig-Holstein and Christian-Albrechts-University of Kiel, D-24105 Kiel, Germany
| | - Michael Busch
- Discovery Pharmacology, Merck KGaA, D-64293 Darmstadt, Germany
| | - Bernhard Valldorf
- Chemical and Pharmaceutical Development, Merck KGaA, D-64293 Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, D-64287 Darmstadt, Germany
| | - Daniela Wesch
- Institute of Immunology, University Hospital Schleswig-Holstein and Christian-Albrechts-University of Kiel, D-24105 Kiel, Germany; and
| | - Hans-Heinrich Oberg
- Institute of Immunology, University Hospital Schleswig-Holstein and Christian-Albrechts-University of Kiel, D-24105 Kiel, Germany; and
| | - Steffen Krohn
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, University Hospital Schleswig-Holstein and Christian-Albrechts-University of Kiel, D-24105 Kiel, Germany
| | - Ammelie Svea Boje
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, University Hospital Schleswig-Holstein and Christian-Albrechts-University of Kiel, D-24105 Kiel, Germany
| | - Carina Lynn Gehlert
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, University Hospital Schleswig-Holstein and Christian-Albrechts-University of Kiel, D-24105 Kiel, Germany
| | - Lars Toleikis
- Protein Engineering and Antibody Technologies, Merck KGaA, D-64293 Darmstadt, Germany
| | - Simon Krah
- Protein Engineering and Antibody Technologies, Merck KGaA, D-64293 Darmstadt, Germany
| | - Tushar Gupta
- Department of Immuno-oncology, EMD Serono Research & Development Institute Inc., Billerica, MA 01821
| | - Brian Rabinovich
- Department of Immuno-oncology, EMD Serono Research & Development Institute Inc., Billerica, MA 01821
| | - Stefan Zielonka
- Protein Engineering and Antibody Technologies, Merck KGaA, D-64293 Darmstadt, Germany;
| | - Matthias Peipp
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, University Hospital Schleswig-Holstein and Christian-Albrechts-University of Kiel, D-24105 Kiel, Germany;
| |
Collapse
|
47
|
Kuwahara A, Nagai K, Nakanishi T, Kumagai I, Asano R. Functional Domain Order of an Anti-EGFR × Anti-CD16 Bispecific Diabody Involving NK Cell Activation. Int J Mol Sci 2020; 21:ijms21238914. [PMID: 33255436 PMCID: PMC7727810 DOI: 10.3390/ijms21238914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/19/2020] [Accepted: 11/22/2020] [Indexed: 12/12/2022] Open
Abstract
Bispecific antibodies (bsAbs) have emerged as promising therapeutics. A bispecific diabody (bsDb) is a small bsAb consisting of two distinct chimeric single-chain components, with two possible arrangements of the domains. We previously reported the effect of domain order on the function of a humanized bsDb targeting the epidermal growth factor receptor (EGFR) on cancer cells, and CD3 on T cells. Notably, the co-localization of a T-cell receptor (TCR) with CD3 is bulky, potentially affecting the cross-linking ability of bsDbs, due to steric hindrance. Here, we constructed and evaluated humanized bsDbs, with different domain orders, targeting EGFR and CD16 on natural killer (NK) cells (hEx16-Dbs). We predicted minimal effects due to steric hindrance, as CD16 lacks accessory molecules. Interestingly, one domain arrangement displayed superior cytotoxicity in growth inhibition assays, despite similar cross-linking abilities for both domain orders tested. In hEx16-Dbs specifically, domain order might affect the agonistic activity of the anti-CD16 portion, which was supported by a cytokine production test, and likely contributed to the superiority of one of the hEx16-Dbs. Our results indicate that both the target antigen and mode of action of an antibody must be considered in the construction of highly functional bsAbs.
Collapse
Affiliation(s)
- Atsushi Kuwahara
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan; (A.K.); (I.K.)
| | - Keisuke Nagai
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai 980-8579, Japan;
| | - Takeshi Nakanishi
- Department of Applied Chemistry and Bioengineering, Graduate School of Engineering, Osaka City University, Osaka 558-8585, Japan;
| | - Izumi Kumagai
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan; (A.K.); (I.K.)
| | - Ryutaro Asano
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan; (A.K.); (I.K.)
- Correspondence: ; Tel.: +81-42-388-7512
| |
Collapse
|
48
|
Felices M, Lenvik TR, Kodal B, Lenvik AJ, Hinderlie P, Bendzick LE, Schirm DK, Kaminski MF, McElmurry RT, Geller MA, Eckfeldt CE, Vallera DA, Miller JS. Potent Cytolytic Activity and Specific IL15 Delivery in a Second-Generation Trispecific Killer Engager. Cancer Immunol Res 2020; 8:1139-1149. [PMID: 32661096 DOI: 10.1158/2326-6066.cir-19-0837] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/10/2020] [Accepted: 06/25/2020] [Indexed: 12/22/2022]
Abstract
Natural killer (NK) cells are potent immune modulators that can quickly lyse tumor cells and elicit inflammatory responses. These characteristics make them ideal candidates for immunotherapy. However, unlike T cells, NK cells do not possess clonotypic receptors capable of specific antigen recognition and cannot expand via activating receptor signals alone. To enable NK cells with these capabilities, we created and have previously described a tri-specific killer engager (TriKE) platform capable of inducing antigen specificity and cytokine-mediated NK-cell expansion. TriKE molecules have three arms: (i) a single-chain variable fragment (scFv) against the activating receptor CD16 on NK cells to trigger NK-cell activation, (ii) an scFv against a tumor-associated antigen (CD33 here) to induce specific tumor target recognition, and (iii) an IL15 moiety to trigger NK-cell expansion and priming. Here, we demonstrate that by modifying the anti-CD16 scFv with a humanized single-domain antibody against CD16, we improved TriKE functionality. A CD33-targeting second-generation TriKE induced stronger and more specific NK-cell proliferation without T-cell stimulation, enhanced in vitro NK-cell activation and killing of CD33-expressing targets, and improved tumor control in preclinical mouse models. Given these improved functional characteristics, we propose rapid translation of second-generation TriKEs into the clinic.
Collapse
Affiliation(s)
- Martin Felices
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Todd R Lenvik
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Behiye Kodal
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Alexander J Lenvik
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Peter Hinderlie
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Laura E Bendzick
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, Minnesota
| | - Dawn K Schirm
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Michael F Kaminski
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Ron T McElmurry
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Melissa A Geller
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, Minnesota
| | - Craig E Eckfeldt
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Daniel A Vallera
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota
| | - Jeffrey S Miller
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
49
|
Zhu Y, Shen R, Hao R, Wang S, Ho M. Highlights of Antibody Engineering and Therapeutics 2019 in San Diego, USA: Bispecific Antibody Design and Clinical Applications. Antib Ther 2020; 3:146-154. [PMID: 32671329 PMCID: PMC7363406 DOI: 10.1093/abt/tbaa012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Although there are only two bispecific antibody (bsAb) drugs in the market, around 100 bsAb drug candidates are in clinical development. BsAbs have gained fast growing investment and attractions from the biopharmaceutical industry and academia in recent years. Antibody Engineering & Therapeutics 2019 (AET 2019) was held in San Diego, USA, from December 9th to 13th, 2019. This year's AET certainly reflected the trend. In this report, we selected eleven presentations from AET 2019 to highlight bsAbs' design and their potentials in cancer therapy. These presentations have discussed emerging strategies to improve bispecific antibody drugs in efficacy, safety, and production. As compared to CAR-Ts, some T cell-redirecting bsAbs may potentially achieve comparable efficacies with less side effects and toxicities, as evidenced with both preclinical and clinical data reviewed at the conference. Several approaches to reduce T cell engagers' toxicities including conditionally active bsAbs and IgM-based bsAbs were also presented and discussed at the conference. For the first time, The Antibody Society and the Chinese Antibody Society jointly held a special session at the AET.
Collapse
Affiliation(s)
- Yong Zhu
- Chinese Antibody Society, Cambridge, MA 02139, USA,To whom correspondence should be addressed. Yong Zhu or Mitchell Ho. or
| | - Run Shen
- Sorrento Therapeutics, San Diego, CA 92121, USA
| | - Rui Hao
- Pfizer, San Diego, CA 92121, USA
| | - Shouye Wang
- Chinese Antibody Society, Cambridge, MA 02139, USA
| | - Mitchell Ho
- NCI Antibody Engineering Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA,To whom correspondence should be addressed. Yong Zhu or Mitchell Ho. or
| |
Collapse
|
50
|
Maryamchik E, Gallagher KME, Preffer FI, Kadauke S, Maus MV. New directions in chimeric antigen receptor T cell [CAR-T] therapy and related flow cytometry. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 98:299-327. [PMID: 32352629 DOI: 10.1002/cyto.b.21880] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/01/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Chimeric antigen receptor (CAR) T cells provide a promising approach to the treatment of hematologic malignancies and solid tumors. Flow cytometry is a powerful analytical modality, which plays an expanding role in all stages of CAR T therapy, from lymphocyte collection, to CAR T cell manufacturing, to in vivo monitoring of the infused cells and evaluation of their function in the tumor environment. Therefore, a thorough understanding of the new directions is important for designing and implementing CAR T-related flow cytometry assays in the clinical and investigational settings. However, the speed of new discoveries and the multitude of clinical and preclinical trials make it challenging to keep up to date in this complex field. In this review, we summarize the current state of CAR T therapy, highlight the areas of emergent research, discuss applications of flow cytometry in modern cell therapy, and touch upon several considerations particular to CAR detection and assessing the effectiveness of CAR T therapy.
Collapse
Affiliation(s)
- Elena Maryamchik
- Department of Pathology and Laboratory Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Frederic I Preffer
- Clinical Cytometry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stephan Kadauke
- Department of Pathology and Laboratory Medicine, Cell and Gene Therapy Laboratory, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Marcela V Maus
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Cellular Immunotherapy Program, Department of Medicine, Boston, Massachusetts, USA
| |
Collapse
|