1
|
Chanchiri I, Christensen EB, Abildgaard N, Barington T, Lund T, Krejcik J. Role of NK Cells in Progression and Treatment of Multiple Myeloma. FRONT BIOSCI-LANDMRK 2025; 30:26205. [PMID: 40302319 DOI: 10.31083/fbl26205] [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: 08/20/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 05/02/2025]
Abstract
Multiple myeloma (MM) is a haematological malignancy originating from terminally differentiated B cells, resulting in significant morbidity and mortality. Currently, MM is regarded as an incurable disease, often exhibiting a relapse-remitting pattern that necessitates multiple lines of therapy. It is now well-established that ineffective immunosurveillance plays a critical role in the progression of MM. Consequently, strategies that redirect immune effector cells against MM have emerged as effective treatment modalities, particularly in cases where standard care therapies fail. T cell-based immunotherapy has gained considerable attention in ongoing clinical trials; however, natural killer (NK) cells, known for their ability to execute cytotoxicity against infected and malignant cells with precision, may offer complementary therapeutic advantages over T cells and possess untapped therapeutic potential. This review seeks to introduce readers to the significance of NK cell-mediated immunosurveillance in the context of MM, explore the potential benefits of redirecting NK cells against MM, and illustrate how current treatment strategies are often reliant on the functionality of NK cells. Most importantly, new promising mechanisms of harnessing NK cell-based immunity against MM are reviewed and put into a clinical perspective to highlight their implications for patient treatment and outcomes.
Collapse
Affiliation(s)
- Iman Chanchiri
- Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark
| | - Emil Birch Christensen
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, 5230 Odense, Denmark
- Department of Clinical Immunology, Odense University Hospital, 5000 Odense, Denmark
| | - Niels Abildgaard
- Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, 5230 Odense, Denmark
| | - Torben Barington
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, 5230 Odense, Denmark
- Department of Clinical Immunology, Odense University Hospital, 5000 Odense, Denmark
| | - Thomas Lund
- Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, 5230 Odense, Denmark
- Centre for Innovative Medical Technology (CIMT), Odense University Hospital, 5000 Odense, Denmark
| | - Jakub Krejcik
- Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark
| |
Collapse
|
2
|
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
|
3
|
Pu J, Liu T, Sharma A, Jiang L, Wei F, Ren X, Schmidt-Wolf IGH, Hou J. Advances in adoptive cellular immunotherapy and therapeutic breakthroughs in multiple myeloma. Exp Hematol Oncol 2024; 13:105. [PMID: 39468695 PMCID: PMC11514856 DOI: 10.1186/s40164-024-00576-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024] Open
Abstract
The basic idea of modulating the immune system to better recognize and fight tumor cells has led to the successful introduction of adoptive cellular immunotherapy (ACT). ACT-based treatment regimens, in which the patient's own immune cells are isolated and subsequently expanded (ex vivo) and reinfused, have also contributed significantly to the development of a personalized treatment strategy. Complementing this, the unprecedented advances in ACTs as chimeric antigen receptor (CAR)-T cell therapies and their derivatives such as CAR-NK, CAR-macrophages, CAR-γδT and CAR-NKT have further maximized the therapeutic outcomes. Herein, we provide a comprehensive overview of the development of ACTs in multiple myeloma (MM) and outline how they have evolved from an experimental form to a mainstay of standard clinical settings. Besides, we provide insights into cytokine-induced killer cell (CIK) therapy, an alternative form of ACT that (as CIK or CAR-CIK) has enormous potential in the clinical spectrum of MM. We also summarize the results of the major preclinical and clinical studies of adoptive cell therapy in MM and address the current challenges (such as cytokine release syndrome (CRS) and neurotoxicity) that limit its complete success in the cancer landscape.
Collapse
Affiliation(s)
- Jingjing Pu
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ting Liu
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, NRW, Germany
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
| | - Liping Jiang
- Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Feng Wei
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300070, China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300070, China.
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany.
| | - Jian Hou
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
4
|
Kremer PG, Lampros EA, Blocker AM, Barb AW. One N-glycan regulates natural killer cell antibody-dependent cell-mediated cytotoxicity and modulates Fc γ receptor IIIa/CD16a structure. eLife 2024; 13:RP100083. [PMID: 39453384 PMCID: PMC11509673 DOI: 10.7554/elife.100083] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024] Open
Abstract
Both endogenous antibodies and a subset of antibody therapeutics engage Fc gamma receptor (FcγR)IIIa/CD16a to stimulate a protective immune response. Increasing the FcγRIIIa/IgG1 interaction improves the immune response and thus represents a strategy to improve therapeutic efficacy. FcγRIIIa is a heavily glycosylated receptor and glycan composition affects antibody-binding affinity. Though our laboratory previously demonstrated that natural killer (NK) cell N-glycan composition affected the potency of one key protective mechanism, antibody-dependent cell-mediated cytotoxicity (ADCC), it was unclear if this effect was due to FcγRIIIa glycosylation. Furthermore, the structural mechanism linking glycan composition to affinity and cellular activation remained undescribed. To define the role of individual amino acid and N-glycan residues, we measured affinity using multiple FcγRIIIa glycoforms. We observed stepwise affinity increases with each glycan truncation step, with the most severely truncated glycoform displaying the highest affinity. Removing the N162 glycan demonstrated its predominant role in regulating antibody-binding affinity, in contrast to four other FcγRIIIa N-glycans. We next evaluated the impact of the N162 glycan on NK cell ADCC. NK cells expressing the FcγRIIIa V158 allotype exhibited increased ADCC following kifunensine treatment to limit N-glycan processing. Notably, an increase was not observed with cells expressing the FcγRIIIa V158 S164A variant that lacks N162 glycosylation, indicating that the N162 glycan is required for increased NK cell ADCC. To gain structural insight into the mechanisms of N162 regulation, we applied a novel protein isotope labeling approach in combination with solution NMR spectroscopy. FG loop residues proximal to the N162 glycosylation site showed large chemical shift perturbations following glycan truncation. These data support a model for the regulation of FcγRIIIa affinity and NK cell ADCC whereby composition of the N162 glycan stabilizes the FG loop and thus the antibody-binding site.
Collapse
Affiliation(s)
- Paul G Kremer
- Department of Biochemistry and Molecular Biology, University of GeorgiaAthensUnited States
| | - Elizabeth A Lampros
- Department of Biochemistry and Molecular Biology, University of GeorgiaAthensUnited States
| | - Allison M Blocker
- Department of Biochemistry and Molecular Biology, University of GeorgiaAthensUnited States
| | - Adam W Barb
- Department of Biochemistry and Molecular Biology, University of GeorgiaAthensUnited States
- Complex Carbohydrate Research Center, University of GeorgiaAthensUnited States
- Department of Chemistry, University of GeorgiaAthensUnited States
| |
Collapse
|
5
|
Kremer PG, Lampros EA, Blocker AM, Barb AW. One N-glycan regulates natural killer cell antibody-dependent cell-mediated cytotoxicity and modulates Fc γ receptor IIIa / CD16a structure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599285. [PMID: 38948809 PMCID: PMC11212880 DOI: 10.1101/2024.06.17.599285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Both endogenous antibodies and a subset of antibody therapeutics engage Fc gamma receptor (FcγR)IIIa / CD16a to stimulate a protective immune response. Increasing the FcγRIIIa/IgG1 interaction improves the immune response and thus represents a strategy to improve therapeutic efficacy. FcγRIIIa is a heavily glycosylated receptor and glycan composition affects antibody-binding affinity. Though our laboratory previously demonstrated that natural killer (NK) cell N-glycan composition affected the potency of one key protective mechanism, antibody-dependent cell-mediated cytotoxicity (ADCC), it was unclear if this effect was due to FcγRIIIa glycosylation. Furthermore, the structural mechanism linking glycan composition to affinity and cellular activation remained undescribed. To define the role of individual amino acid and N-glycan residues we measured affinity using multiple FcγRIIIa glycoforms. We observed stepwise affinity increases with each glycan truncation step with the most severely truncated glycoform displaying the highest affinity. Removing the N162 glycan demonstrated its predominant role in regulating antibody-binding affinity, in contrast to four other FcγRIIIa N-glycans. We next evaluated the impact of the N162 glycan on NK cell ADCC. NK cells expressing the FcγRIIIa V158 allotype exhibited increased ADCC following kifunensine treatment to limit N-glycan processing. Notably, an increase was not observed with cells expressing the FcγRIIIa V158 S164A variant that lacks N162 glycosylation, indicating the N162 glycan is required for increased NK cell ADCC. To gain structural insight into the mechanisms of N162 regulation, we applied a novel protein isotope labeling approach in combination with solution NMR spectroscopy. FG loop residues proximal to the N162 glycosylation site showed large chemical shift perturbations following glycan truncation. These data support a model for the regulation of FcγRIIIa affinity and NK cell ADCC whereby composition of the N162 glycan stabilizes the FG loop and thus the antibody-binding site.
Collapse
Affiliation(s)
- Paul G. Kremer
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA
| | - Elizabeth A. Lampros
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA
| | - Allison M. Blocker
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA
| | - Adam W. Barb
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
- Department of Chemistry, University of Georgia, Athens, GA
| |
Collapse
|
6
|
Braun A, Gouni S, Pulles A, Strati P, Minnema MC, Budde LE. Bispecific Antibody Use in Patients With Lymphoma and Multiple Myeloma. Am Soc Clin Oncol Educ Book 2024; 44:e433516. [PMID: 38935881 DOI: 10.1200/edbk_433516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
This article endeavors to navigate the clinical journey of bispecific antibodies (BsAbs), from elucidating common toxicities and management strategies to examining novel agents and broadening access in community health care. These drugs, commonly through T-cell activation, result in shared adverse events such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. Variations in target antigens and designs, however, might introduce unique toxicities for different BsAbs, warranting specific management approaches. Recent US Food and Drug Administration approvals of BsAbs targeting CD3+ T cells linked to CD20 for non-Hodgkin lymphoma and to B-cell maturation antigen or GPRC5D for multiple myeloma have transformed the treatment landscape for hematologic malignancies. Emerging new agents promise further enhancement and safety, exploring novel antigen targets, innovative structures such as trispecific antibodies, and the engagement of diverse immune cells. Simultaneously, the expansion of BsAbs into community practices is underway, demanding a multifaceted strategy that encompasses educational initiatives, operational adaptations, and collaborative frameworks. This ensures comprehensive treatment access, allowing every patient, irrespective of geographical or socioeconomic status, to benefit from these advancements in cancer therapy.
Collapse
Affiliation(s)
- Adam Braun
- City of Hope National Medical Center, Duarte, CA
| | | | - Astrid Pulles
- Department of Rheumatology & Clinical Immunology, Utrecht University, Utrecht, the Netherlands
| | - Paolo Strati
- MD Anderson Comprehensive Cancer Center, Houston, TX
| | - Monique C Minnema
- Department of Hematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | |
Collapse
|
7
|
Parrondo RD, Ailawadhi S, Cerchione C. Bispecific antibodies for the treatment of relapsed/refractory multiple myeloma: updates and future perspectives. Front Oncol 2024; 14:1394048. [PMID: 38660139 PMCID: PMC11039948 DOI: 10.3389/fonc.2024.1394048] [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: 02/29/2024] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Patients with relapsed/refractory multiple myeloma (RRMM) that are refractory to the five most active anti-MM drugs, so-called penta-refractory MM, have historically had dismal outcomes with subsequent therapies. Progressive immune dysfunction, particularly of the T-cell repertoire, is implicated in the development of disease progression and refractory disease. However, the advent of novel immunotherapies such as bispecific antibodies are rapidly changing the treatment landscape and improving the survival outcomes of patients with RRMM. Bispecific antibodies are antibodies that are engineered to simultaneously engage cytotoxic immune effector cells (T cells or NK cells) and malignant plasma cells via binding to immune effector cell antigens and extracellular plasma cell antigens leading to immune effector cell activation and malignant plasma cell destruction. Currently, bispecific antibodies that bind CD3 on T cells and plasma cell epitopes such as B-cell maturation antigen (BCMA), G-protein coupled receptor family C group 5 member D (GPRC5d), and Fc receptor homologue 5 (FcRH5) are the most advanced in clinical development and are showing unprecedented response rates in patients with RRMM, including patients with penta-refractory disease. In this review article, we explore the available clinical data of bispecific antibodies in RRMM and summarize the efficacy, safety, toxicity, clinical outcomes, mechanisms of resistance, and future directions of these therapies in patients with RRMM.
Collapse
Affiliation(s)
- Ricardo D. Parrondo
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, and Cellular Therapies, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, United States
| | - Sikander Ailawadhi
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, and Cellular Therapies, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, United States
| | - Claudio Cerchione
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
8
|
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: 126] [Impact Index Per Article: 126.0] [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
|
9
|
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
|
10
|
Besla R, Penuel E, Del Rosario G, Cosino E, Myrta S, Dillon M, Lazar GA, Nickles D, Spiess C, Yu SF, Polson AG. T cell-Dependent Bispecific Therapy Enhances Innate Immune Activation and Antibody-Mediated Killing. Cancer Immunol Res 2024; 12:60-71. [PMID: 37902604 DOI: 10.1158/2326-6066.cir-23-0072] [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: 01/27/2023] [Revised: 05/15/2023] [Accepted: 10/26/2023] [Indexed: 10/31/2023]
Abstract
T cell-retargeting therapies have transformed the therapeutic landscape for hematologic diseases. T cell-dependent bispecific antibodies (TDB) function as conditional agonists that induce a polyclonal T-cell response, resulting in target cell destruction and cytokine release. The relationship between this response and its effects on surrounding innate immune populations has not been fully explored. Here we show that treatment with mosunetuzumab in patients results in natural killer (NK) cell activation in the peripheral blood. We modeled this phenomenon in vitro and found that TDB-mediated killing activated NK cells, increasing NK function and antibody-dependent cellular cytotoxicity (ADCC), and enhanced the capability of macrophages to perform antibody-dependent cellular phagocytosis (ADCP). This enhancement was triggered by cytokines released through TDB treatment, with IL2 and IFNγ being major drivers for increased ADCC and ADCP, respectively. Surprisingly, cytolytic ability could be further augmented through neutralization of IL10 for NK cells and TNFα for macrophages. Finally, we showed that TDB treatment enhanced the efficacy of Fc-driven killing to an orthogonal solid tumor target in vivo. These results provide rationale for novel antibody therapy combinations that take advantage of both adaptive and innate immune responses.
Collapse
Affiliation(s)
- Rickvinder Besla
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Elicia Penuel
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Geoff Del Rosario
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Ely Cosino
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | | | - Mike Dillon
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Greg A Lazar
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Dorothee Nickles
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Christoph Spiess
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Shang-Fan Yu
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Andrew G Polson
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| |
Collapse
|
11
|
Guo X, Wu Y, Xue Y, Xie N, Shen G. Revolutionizing cancer immunotherapy: unleashing the potential of bispecific antibodies for targeted treatment. Front Immunol 2023; 14:1291836. [PMID: 38106416 PMCID: PMC10722299 DOI: 10.3389/fimmu.2023.1291836] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/08/2023] [Indexed: 12/19/2023] Open
Abstract
Recent progressions in immunotherapy have transformed cancer treatment, providing a promising strategy that activates the immune system of the patient to find and eliminate cancerous cells. Bispecific antibodies, which engage two separate antigens or one antigen with two distinct epitopes, are of tremendous concern in immunotherapy. The bi-targeting idea enabled by bispecific antibodies (BsAbs) is especially attractive from a medical standpoint since most diseases are complex, involving several receptors, ligands, and signaling pathways. Several research look into the processes in which BsAbs identify different cancer targets such angiogenesis, reproduction, metastasis, and immune regulation. By rerouting cells or altering other pathways, the bispecific proteins perform effector activities in addition to those of natural antibodies. This opens up a wide range of clinical applications and helps patients with resistant tumors respond better to medication. Yet, further study is necessary to identify the best conditions where to use these medications for treating tumor, their appropriate combination partners, and methods to reduce toxicity. In this review, we provide insights into the BsAb format classification based on their composition and symmetry, as well as the delivery mode, focus on the action mechanism of the molecule, and discuss the challenges and future perspectives in BsAb development.
Collapse
Affiliation(s)
- Xiaohan Guo
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yi Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Ying Xue
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Guobo Shen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
12
|
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
|
13
|
Liu Q, Song Q, Luo C, Wei J, Xu Y, Zhao L, Wang Y. A novel bispecific antibody as an immunotherapeutic agent in hepatocellular carcinoma. Mol Immunol 2023; 162:125-132. [PMID: 37677989 DOI: 10.1016/j.molimm.2023.08.007] [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: 03/27/2023] [Revised: 08/04/2023] [Accepted: 08/20/2023] [Indexed: 09/09/2023]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most common and highly fatal malignancies in humans worldwide with increasing prevalence and limited therapeutic options. For many decades, many researchers have attempted to find effective curative methods for HCC and great strides have been made. GPC3 is overexpressed in HCC, but not in normal liver, making it a rational immunotherapeutic target for HCC. GC33, a humanized mAb directed against GPC3, is a safe and well-tolerated therapy choice for patients with HCC, which tested in a phase I trial in advanced HCC patients. Phase II trials of GC33 to evaluate its efficacy and safety in advanced or metastatic HCC, showed no significant differences in overall survival and progression-free survival compared with the placebo. Retrospective analysis indicates that high drug exposure and high CD16 expression may contribute to the clinical efficacy of GC33. Chugai Pharmaceutical has restarted its Phase I trial of GC33, continuing to explore its clinical value targeting GPC3 in solid tumors. To enhance the antitumor potency of GC33, we designed a GPC3/CD16A bispecific antibody (QDEB). In this study, we obtained QDEB at high purity and assessed its effectiveness in the therapy of HCC compared with GC33. In vitro cytotoxicity assays and in vivo experiments demonstrated that QDEB could enhance anti-tumor efficacy compared with GC33. CD16A activation and increased cytokines release were associated with higher anti-tumor activity. In conclusion, this bispecific antibody may possibly help develop new therapeutic strategies for HCC and develop new treatment options in the future.
Collapse
Affiliation(s)
- Qingxia Liu
- Department of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China; Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China
| | - Qifeng Song
- Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China
| | - Cheng Luo
- Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China
| | - Jian Wei
- Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China
| | - Yao Xu
- Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China
| | - Liwen Zhao
- Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China
| | - Yong Wang
- Department of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China; Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China.
| |
Collapse
|
14
|
Zhang M, Lam KP, Xu S. Natural Killer Cell Engagers (NKCEs): a new frontier in cancer immunotherapy. Front Immunol 2023; 14:1207276. [PMID: 37638058 PMCID: PMC10450036 DOI: 10.3389/fimmu.2023.1207276] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/03/2023] [Indexed: 08/29/2023] Open
Abstract
Natural Killer (NK) cells are a type of innate lymphoid cells that play a crucial role in immunity by killing virally infected or tumor cells and secreting cytokines and chemokines. NK cell-mediated immunotherapy has emerged as a promising approach for cancer treatment due to its safety and effectiveness. NK cell engagers (NKCEs), such as BiKE (bispecific killer cell engager) or TriKE (trispecific killer cell engager), are a novel class of antibody-based therapeutics that exhibit several advantages over other cancer immunotherapies harnessing NK cells. By bridging NK and tumor cells, NKCEs activate NK cells and lead to tumor cell lysis. A growing number of NKCEs are currently undergoing development, with some already in clinical trials. However, there is a need for more comprehensive studies to determine how the molecular design of NKCEs affects their functionality and manufacturability, which are crucial for their development as off-the-shelf drugs for cancer treatment. In this review, we summarize current knowledge on NKCE development and discuss critical factors required for the production of effective NKCEs.
Collapse
Affiliation(s)
- Minchuan Zhang
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Kong-Peng Lam
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore, Singapore
| | - Shengli Xu
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
15
|
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
|
16
|
Cencini E, Sicuranza A, Ciofini S, Fabbri A, Bocchia M, Gozzetti A. Tumor-Associated Macrophages in Multiple Myeloma: Key Role in Disease Biology and Potential Therapeutic Implications. Curr Oncol 2023; 30:6111-6133. [PMID: 37504315 PMCID: PMC10378698 DOI: 10.3390/curroncol30070455] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/14/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023] Open
Abstract
Multiple myeloma (MM) is characterized by multiple relapse and, despite the introduction of novel therapies, the disease becomes ultimately drug-resistant. The tumor microenvironment (TME) within the bone marrow niche includes dendritic cells, T-cytotoxic, T-helper, reactive B-lymphoid cells and macrophages, with a complex cross-talk between these cells and the MM tumor cells. Tumor-associated macrophages (TAM) have an important role in the MM pathogenesis, since they could promote plasma cells proliferation and angiogenesis, further supporting MM immune evasion and progression. TAM are polarized towards M1 (classically activated, antitumor activity) and M2 (alternatively activated, pro-tumor activity) subtypes. Many studies demonstrated a correlation between TAM, disease progression, drug-resistance and reduced survival in lymphoproliferative neoplasms, including MM. MM plasma cells in vitro could favor an M2 TAM polarization. Moreover, a possible correlation between the pro-tumor effect of M2 TAM and a reduced sensitivity to proteasome inhibitors and immunomodulatory drugs was hypothesized. Several clinical studies confirmed CD68/CD163 double-positive M2 TAM were associated with increased microvessel density, chemoresistance and reduced survival, independently of the MM stage. This review provided an overview of the biology and clinical relevance of TAM in MM, as well as a comprehensive evaluation of a potential TAM-targeted immunotherapy.
Collapse
Affiliation(s)
- Emanuele Cencini
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Anna Sicuranza
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Sara Ciofini
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Alberto Fabbri
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Monica Bocchia
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Alessandro Gozzetti
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| |
Collapse
|
17
|
Xiao X, Cheng Y, Zheng X, Fang Y, Zhang Y, Sun R, Tian Z, Sun H. Bispecific NK-cell engager targeting BCMA elicits stronger antitumor effects and produces less proinflammatory cytokines than T-cell engager. Front Immunol 2023; 14:1113303. [PMID: 37114050 PMCID: PMC10126364 DOI: 10.3389/fimmu.2023.1113303] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/27/2023] [Indexed: 04/29/2023] Open
Abstract
Bispecific antibodies have attracted more attention in recent years for the treatment of tumors, in which most of them target CD3, which mediates the killing of tumor cells by T cells. However, T-cell engager may cause serious side effects, including neurotoxicity and cytokine release syndrome. More safe treatments are still needed to address unmet medical needs, and NK cell-based immunotherapy is a safer and more effective way to treat tumors. Our study developed two IgG-like bispecific antibodies with the same configuration: BT1 (BCMA×CD3) attracted T cells and tumor cells, while BK1 (BCMA×CD16) attracted NK cells and tumor cells. Our study showed that BK1 mediated NK cell activation and upregulated the expression of CD69, CD107a, IFN-γ and TNF. In addition, BK1 elicited a stronger antitumor effect than BT1 both in vitro and in vivo. Combinatorial treatment (BK1+BT1) showed a stronger antitumor effect than either treatment alone, as indicated by in vitro experiments and in vivo murine models. More importantly, BK1 induced fewer proinflammatory cytokines than BT1 both in vitro and in vivo. Surprisingly, BK1 reduced cytokine production in the combinatorial treatment, suggesting the indispensable role of NK cells in the control of cytokine secretion by T cells. In conclusion, our study compared NK-cell engagers and T-cell engagers targeting BCMA. The results indicated that NK-cell engagers were more effective with less proinflammatory cytokine production. Furthermore, the use of NK-cell engagers in combinatorial treatment helped to reduce cytokine secretion by T cells, suggesting a bright future for NK-cell engagers in clinical settings.
Collapse
Affiliation(s)
- Xinghui Xiao
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Ying Cheng
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xiaodong Zheng
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Yuhang Fang
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Yu Zhang
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Rui Sun
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Zhigang Tian
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
- Hefei TG ImmunoPharma Corporation Limited, Hefei, China
| | - Haoyu Sun
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| |
Collapse
|
18
|
Zhou Y, Cheng L, Liu L, Li X. NK cells are never alone: crosstalk and communication in tumour microenvironments. Mol Cancer 2023; 22:34. [PMID: 36797782 PMCID: PMC9933398 DOI: 10.1186/s12943-023-01737-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Immune escape is a hallmark of cancer. The dynamic and heterogeneous tumour microenvironment (TME) causes insufficient infiltration and poor efficacy of natural killer (NK) cell-based immunotherapy, which becomes a key factor triggering tumour progression. Understanding the crosstalk between NK cells and the TME provides new insights for optimising NK cell-based immunotherapy. Here, we present new advances in direct or indirect crosstalk between NK cells and 9 specialised TMEs, including immune, metabolic, innervated niche, mechanical, and microbial microenvironments, summarise TME-mediated mechanisms of NK cell function inhibition, and highlight potential targeted therapies for NK-TME crosstalk. Importantly, we discuss novel strategies to overcome the inhibitory TME and provide an attractive outlook for the future.
Collapse
Affiliation(s)
- Yongqiang Zhou
- grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000 China ,grid.412643.60000 0004 1757 2902Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China ,Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China
| | - Lu Cheng
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China
| | - Lu Liu
- grid.412643.60000 0004 1757 2902Department of Pediatrics, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xun Li
- The First School of Clinical Medicine, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China. .,Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China. .,Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China.
| |
Collapse
|
19
|
Plesner T, Harrison SJ, Quach H, Lee C, Bryant A, Vangsted A, Estell J, Delforge M, Offner F, Twomey P, Choeurng V, Li J, Hendricks R, Ruppert SM, Sumiyoshi T, Miller K, Cho E, Schjesvold F. Phase I Study of Safety and Pharmacokinetics of RO7297089, an Anti-BCMA/CD16a Bispecific Antibody, in Patients with Relapsed, Refractory Multiple Myeloma. Clin Hematol Int 2023; 5:43-51. [PMID: 36656461 PMCID: PMC10063703 DOI: 10.1007/s44228-022-00023-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/26/2022] [Indexed: 01/20/2023] Open
Abstract
INTRODUCTION This phase 1 trial assessed the safety, pharmacokinetics, and preliminary antitumor activity of RO7297089, an anti-BCMA/CD16a bispecific antibody. METHODS RO7297089 was administered weekly by intravenous infusion to patients with relapsed/refractory multiple myeloma. The starting dose was 60 mg in this dose-escalation study utilizing a modified continual reassessment method with overdose control model. RESULTS Overall, 27 patients were treated at doses between 60 and 1850 mg. The maximally administered dose was 1850 mg due to excipients in the formulation that did not allow for higher doses to be used. The maximum tolerated dose was not reached. The most common adverse events irrespective of grade and relationship to the drug were anemia, infusion-related reaction, and thrombocytopenia. Most common treatment-related grade ≥ 3 toxicities were ALT/AST increase and reduced lymphocyte count. Pharmacokinetic studies suggested non-linear pharmacokinetics and target-mediated drug disposition, with a trend of approaching linear pharmacokinetics at doses of 1080 mg and higher. Partial response was observed in two patients (7%), minimal response in two patients (7%), and stable disease in 14 patients (52%). CONCLUSIONS RO7297089 was well tolerated at doses up to 1850 mg, and the efficacy data supported activity of RO7297089 in multiple myeloma. Combination with other agents may further enhance its potential as an innate immune cell engager in multiple myeloma. TRIAL REGISTRATION ClinicalTrials.gov: NCT04434469; Registered June 16, 2020; https://www. CLINICALTRIALS gov/ct2/show/NCT04434469 .
Collapse
Affiliation(s)
- Torben Plesner
- Vejle Hospital, University of Southern Denmark, Vejle, Denmark.
| | - Simon J Harrison
- Peter MacCallum Cancer Center, Royal Melbourne Hospital, Melbourne and Sir Peter MacCallum Dept of Oncology University of Melbourne, Parkville, Australia
| | - Hang Quach
- St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, Australia
| | - Cindy Lee
- Royal Adelaide Hospital, Adelaide, Australia
| | | | | | - Jane Estell
- Concord Repatriation General Hospital, Concord, Australia
| | | | | | | | - Voleak Choeurng
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital, Oslo, Norway
| | - Junyi Li
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | | | | | - Eunpi Cho
- Genentech, Inc., South San Francisco, CA, USA
| | - Fredrik Schjesvold
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital, Oslo, Norway.,KG Jebsen Center for B Cell Malignancies, University of Oslo, Oslo, Norway
| |
Collapse
|
20
|
Whalen KA, Rakhra K, Mehta NK, Steinle A, Michaelson JS, Baeuerle PA. Engaging natural killer cells for cancer therapy via NKG2D, CD16A and other receptors. MAbs 2023; 15:2208697. [PMID: 37165468 PMCID: PMC10173799 DOI: 10.1080/19420862.2023.2208697] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/26/2023] [Indexed: 05/12/2023] Open
Abstract
The field of immuno-oncology has revolutionized cancer patient care and improved survival and quality of life for patients. Much of the focus in the field has been on exploiting the power of the adaptive immune response through therapeutic targeting of T cells. While these approaches have markedly advanced the field, some challenges remain, and the clinical benefit of T cell therapies does not extend to all patients or tumor indications. Alternative strategies, such as engaging the innate immune system, have become an intense area of focus in the field. In particular, the engagement of natural killer (NK) cells as potent effectors of the innate immune response has emerged as a promising modality in immunotherapy. Here, we review therapeutic approaches for selective engagement of NK cells for cancer therapy, with a particular focus on targeting the key activating receptors NK Group 2D (NKG2D) and cluster of differentiation 16A (CD16A).
Collapse
Affiliation(s)
- Kerry A. Whalen
- Preclinical and Early Development, Cullinan Oncology, Inc, Cambridge, MA, USA
| | - Kavya Rakhra
- Preclinical and Early Development, Cullinan Oncology, Inc, Cambridge, MA, USA
| | - Naveen K. Mehta
- Preclinical and Early Development, Cullinan Oncology, Inc, Cambridge, MA, USA
| | - Alexander Steinle
- Institute for Molecular Medicine, Goethe-University Frankfurt, Frankfurt am Main, Germany
- Preclinical and Early Development, Frankfurt Cancer Institute, Frankfurt am Main, Germany
| | | | - Patrick A. Baeuerle
- Preclinical and Early Development, Cullinan Oncology, Inc, Cambridge, MA, USA
- Institute for Immunology, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
21
|
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: 35] [Impact Index Per Article: 11.7] [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
|
22
|
Sunder-Plassmann V, Aksoy O, Lind J, Pecherstorfer M, Vallet S, Podar K. On the continuous (R)evolution of antibody-based and CAR T cell therapies in multiple myeloma: An early 2022 glance into the future. Expert Opin Pharmacother 2022; 23:1425-1444. [PMID: 35829636 DOI: 10.1080/14656566.2022.2101362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The pace at which the identification of novel therapeutic targets has led to the approval of Multiple Myeloma (MM) agents during the last two decades is nothing more than spectacular. Nevertheless, MM remains an incurable disease. Therefore, there is an urgent need for additional, innovative therapeutics. Immune dysfunction and the tumor-permissive immune bone marrow microenvironment represent hallmarks of MM pathophysiology. Naked monoclonal antibodies directed against SLAMF7 and CD38 already constitute backbones of today's MM therapy. Novel immunotherapeutic modalities including antibody-drug-conjugates (ADC), bispecific antibodies (BsAb) and chimeric-antigen-receptor T cells are on the way to once more revolutionize future MM therapy. AREAS COVERED The present review article summarizes the most recent results on MM immunotherapies presented at the 2021 Annual Meeting of the American Society of Hematology; and throws a glance on ongoing preclinical and clinical efforts aiming at further increasing their efficacy, while reducing their toxicity. EXPERT OPINION With the approvals of the first-in-class BCMA-targeting ADC (belantamab mafodotin) and two BCMA-targeting CAR T cell products (Ide-cel, Cilta-cel); and the approval of the first-in-class BCMAxCD3 BsAb immediately pending, the era of modern next-generation immunotherapies in MM is continuously evolving. Long-term disease-free survival and potential cure of MM are finally within reach.
Collapse
Affiliation(s)
- Vincent Sunder-Plassmann
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences. Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria
| | - Osman Aksoy
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences. Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria
| | - Judith Lind
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences. Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria
| | - Martin Pecherstorfer
- Department of Internal Medicine 2, University Hospital Krems, Mitterweg 10, 3500 Krems an der Donau, Austria
| | - Sonia Vallet
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences. Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria.,Department of Internal Medicine 2, University Hospital Krems, Mitterweg 10, 3500 Krems an der Donau, Austria
| | - Klaus Podar
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences. Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria.,Department of Internal Medicine 2, University Hospital Krems, Mitterweg 10, 3500 Krems an der Donau, Austria
| |
Collapse
|
23
|
Demel I, Koristek Z, Motais B, Hajek R, Jelinek T. Natural killer cells: Innate immune system as a part of adaptive immunotherapy in hematological malignancies. Am J Hematol 2022; 97:802-817. [PMID: 35285978 DOI: 10.1002/ajh.26529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/06/2022] [Accepted: 03/03/2022] [Indexed: 11/06/2022]
Abstract
Natural killer (NK) cells are part of a phylogenetically old defense system, which is characterized by its strong cytolytic function against physiologically stressed cells such as tumor cells and virus-infected cells. Their use in the treatment of hematological malignancies may be more advantageous in several ways when compared with the already established T lymphocyte-based immunotherapy. Given the different mechanisms of action, allogeneic NK cell products can be produced in a non-personal based manner without the risk of the formidable graft-versus-host disease. Advanced manufacturing processes are capable of producing NK cells relatively easily in large and clinically sufficient numbers, useable without subsequent manipulations or after genetic modifications, which can solve the lack of specificity and improve clinical efficacy of NK cell products. This review summarizes the basic characteristics of NK cells and provides a quick overview of their sources. Results of clinical trials in hematological malignancies are presented, and strategies on how to improve the clinical outcome of NK cell therapy are discussed.
Collapse
Affiliation(s)
- Ivo Demel
- Department of Hematooncology University Hospital Ostrava Ostrava Czech Republic
| | - Zdenek Koristek
- Department of Hematooncology University Hospital Ostrava Ostrava Czech Republic
- Faculty of Medicine University of Ostrava Ostrava Czech Republic
| | - Benjamin Motais
- Faculty of Medicine University of Ostrava Ostrava Czech Republic
- Faculty of Science University of Ostrava Ostrava Czech Republic
| | - Roman Hajek
- Department of Hematooncology University Hospital Ostrava Ostrava Czech Republic
- Faculty of Medicine University of Ostrava Ostrava Czech Republic
| | - Tomas Jelinek
- Department of Hematooncology University Hospital Ostrava Ostrava Czech Republic
- Faculty of Medicine University of Ostrava Ostrava Czech Republic
| |
Collapse
|
24
|
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
|