1
|
Ma S, Yu J, Caligiuri MA. Natural killer cell-based immunotherapy for cancer. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf036. [PMID: 40246292 DOI: 10.1093/jimmun/vkaf036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/18/2025] [Indexed: 04/19/2025]
Abstract
Natural killer (NK) cells are emerging as a promising tool for cancer immunotherapy due to their innate ability to selectively recognize and eliminate cancer cells. Over the past 3 decades, strategies to harness NK cells have included cytokines, small molecules, antibodies, and the adoptive transfer of autologous or allogeneic NK cells, both unmodified and genetically engineered. Despite favorable safety profiles in clinical trials, challenges such as limited in vivo persistence, exhaustion, and the suppressive tumor microenvironment continue to hinder their efficacy and durability. This review categorizes NK cell-based therapies into 3 major approaches: (i) cellular therapies, including unmodified and chimeric antigen receptor-engineered NK cells; (ii) cytokine-based strategies such as interleukin-2 and interleukin-15 derivatives; and (iii) antibody-based therapies, including immune checkpoint inhibitors and NK cell engagers. We highlight these advancements, discuss current limitations, and propose strategies to optimize NK cell-based therapies for improved cancer treatment outcomes.
Collapse
Affiliation(s)
- Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, United States
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, United States
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, United States
| | - Jianhua Yu
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, University of California, Irvine, CA, United States
- Institute for Precision Cancer Therapeutics and Immuno-Oncology, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, United States
- Clemons Family Center for Transformative Cancer Research, University of California, Irvine, Irvine, CA, United States
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, United States
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, United States
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, United States
| |
Collapse
|
2
|
Marr B, Jo D, Jang M, Lee SH. Cytokines in Focus: IL-2 and IL-15 in NK Adoptive Cell Cancer Immunotherapy. Immune Netw 2025; 25:e17. [PMID: 40342841 PMCID: PMC12056295 DOI: 10.4110/in.2025.25.e17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 05/11/2025] Open
Abstract
NK cell adoptive cell therapy (ACT) has emerged as a promising strategy for cancer immunotherapy, offering advantages in scalability, accessibility, efficacy, and safety. Ex vivo activation and expansion protocols, incorporating feeder cells and cytokine cocktails, have enabled the production of highly functional NK cells in clinically relevant quantities. Advances in NK cell engineering, including CRISPR-mediated gene editing and chimeric Ag receptor technologies, have further enhanced cytotoxicity, persistence, and tumor targeting. Cytokine support post-adoptive transfer, particularly with IL-2 and IL-15, remains critical for promoting NK cell survival, proliferation, and anti-tumor activity despite persistent challenges such as regulatory T cell expansion and cytokine-related toxicities. This review explores the evolving roles of IL-2 and IL-15 in NK cell-based ACT, evaluating their potential and limitations, and highlights strategies to optimize these cytokines for effective cancer immunotherapy.
Collapse
Affiliation(s)
- Bryan Marr
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Donghyeon Jo
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mihue Jang
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul 02792, Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Korea
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, Faculty of Medicine and Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
3
|
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
|
4
|
Bisio M, Legato L, Fasano F, Benevolo Savelli C, Boccomini C, Nicolosi M, Santambrogio E, Freilone R, Novo M, Botto B. Bispecific Antibodies for Lymphoid Malignancy Treatment. Cancers (Basel) 2024; 17:94. [PMID: 39796723 PMCID: PMC11719988 DOI: 10.3390/cancers17010094] [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: 10/14/2024] [Revised: 11/30/2024] [Accepted: 12/10/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUD The introduction of highly active immunotherapies has changed the outcome of B-cell non-Hodgkin lymphomas (B-NHLs) in the last two decades. Since then, important progress has been shown using newer and more active immunotherapies, including chimeric antigen receptor T-cell therapy (CAR-T), conjugated monoclonal antibodies, and bispecific antobodies, which currently plays a significant role in the treatment of diffuse large B-cell (DLBCL), follicular (FL), and mantle cell (MCL) lymphoma. PURPOSE In this review, we provide an updated overview of recently completed and ongoing BsAb trials in patients with relapsed/refractory(R/R) B-NHL and Hodgkin's lymphoma, including single-agent results, emerging combinations, safety data, and novel constructs. CONCLUSIONS Bispecific antibodies (BsAbs) are a novel class of "off-the-shelf" T-cell-redirecting drugs capable of targeting various cell-surface antigens. New antigen targets are currently under investigation, such as CD19 × CD3 and CD30 × CD3 or CD30 × CD16, in different settings. BsAbs are among the most promising therapeutic options for lymphoma today since they have demonstrated significant single-agent activity, along with a manageable toxicity profile, in patients with heavily pretreated B-NHL.
Collapse
Affiliation(s)
- Matteo Bisio
- Hematology Division, A.O.U. Città della Salute e della Scienza di Torino, C.so Bramante 88, 10126 Turin, Italy (R.F.); (M.N.)
| | | | | | | | | | | | | | | | | | - Barbara Botto
- Hematology Division, A.O.U. Città della Salute e della Scienza di Torino, C.so Bramante 88, 10126 Turin, Italy (R.F.); (M.N.)
| |
Collapse
|
5
|
Demaria O, Habif G, Vetizou M, Gauthier L, Remark R, Chiossone L, Vagne C, Rebuffet L, Courtois R, Denis C, Le Floch F, Muller M, Girard-Madoux M, Augier S, Lopez J, Carrette B, Maguer A, Vallier JB, Grondin G, Baron W, Galluso J, Yessaad N, Giordano M, Simon L, Chanuc F, Alvarez AB, Perrot I, Bonnafous C, Represa A, Rossi B, Morel A, Morel Y, Paturel C, Vivier E. A tetraspecific engager armed with a non-alpha IL-2 variant harnesses natural killer cells against B cell non-Hodgkin lymphoma. Sci Immunol 2024; 9:eadp3720. [PMID: 39546590 DOI: 10.1126/sciimmunol.adp3720] [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: 03/21/2024] [Revised: 06/07/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Abstract
NK cells offer a promising alternative to T cell therapies in cancer. We evaluated IPH6501, a clinical-stage, tetraspecific NK cell engager (NKCE) armed with a non-alpha IL-2 variant (IL-2v), which targets CD20 and was developed for treating B cell non-Hodgkin lymphoma (B-NHL). CD20-NKCE-IL2v boosts NK cell proliferation and cytotoxicity, showing activity against a range of B-NHL cell lines, including those with low CD20 density. Whereas it presented reduced toxicities compared with those commonly associated with T cell therapies, CD20-NKCE-IL2v showed greater killing efficacy over a T cell engager targeting CD20 in in vitro preclinical models. CD20-NKCE-IL2v also increased the cell surface expression of NK cell-activating receptors, leading to activity against CD20-negative tumor cells. In vivo studies in nonhuman primates and tumor mouse models further validated its efficacy and revealed that CD20-NKCE-IL2v induces peripheral NK cell homing at the tumor site. CD20-NKCE-IL2v emerges as a potential alternative in the treatment landscape of B-NHL.
Collapse
Affiliation(s)
- Olivier Demaria
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Guillaume Habif
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Marie Vetizou
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Laurent Gauthier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Romain Remark
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Laura Chiossone
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Constance Vagne
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Lucas Rebuffet
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Rachel Courtois
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Caroline Denis
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - François Le Floch
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Marianna Muller
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | | | - Séverine Augier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Julie Lopez
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Barbara Carrette
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Aurélie Maguer
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | | | | | - William Baron
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Justine Galluso
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Nadia Yessaad
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Marilyn Giordano
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Léa Simon
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Fabien Chanuc
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | | | - Ivan Perrot
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Cécile Bonnafous
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Agnès Represa
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Benjamin Rossi
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Ariane Morel
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Yannis Morel
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Carine Paturel
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- APHM, Hôpital de la Timone, Marseille-Immunopôle Profiling Platform, Marseille, France
- Paris-Saclay Cancer Cluster, Le Kremlin-Bicêtre, France
- Université Paris-Saclay, Gustave Roussy, INSERM, Prédicteurs moléculaires et nouvelles cibles en oncologie, 94800, Villejuif, France
| |
Collapse
|
6
|
Zhu A, Bai Y, Nan Y, Ju D. Natural killer cell engagers: From bi-specific to tri-specific and tetra-specific engagers for enhanced cancer immunotherapy. Clin Transl Med 2024; 14:e70046. [PMID: 39472273 PMCID: PMC11521791 DOI: 10.1002/ctm2.70046] [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: 05/18/2024] [Revised: 07/25/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Natural killer cell engagers (NKCEs) are a specialised subset of antibodies capable of simultaneously targeting endogenous NK cells and tumour cells, generating precise and effective cytolytic responses against cancer. This review systematically explores NK engagers as a rising star in NK-mediated immunotherapy, specifically focusing on multi-specific engagers. It examines the diverse configuration of NKCEs and how certain biologics could be employed to boost NK activity, including activating receptor engagement and cytokine incorporation. Some challenges and future perspectives of current NKCEs therapy are also discussed, including optimising pharmacokinetics, addressing the immunosuppressive tumour microenvironment and exploring potential combinatorial approaches. By offering an in-depth analysis of the current landscape and future trajectories of multi-specific NKCEs in cancer treatment, this review serves as a valuable resource for understanding this promising field of immunotherapy. HIGHLIGHTS Innovative NKCEs: NK cell engagers (NKCEs) represent a promising new class of immunotherapeutics targeting tumours by activating NK cells. Multi-specific formats: The transition from bi-specific to multi-specific NKCEs enhances their versatility and therapeutic efficacy. MECHANISMS OF ACTION NKCEs have the potential to improve NK cell activation by engaging activating receptors and incorporating cytokines. CLINICAL POTENTIAL Current clinical trials demonstrate the safety and efficacy of various NKCEs across different cancer types. Future research directions: Optimising NKCE designs and exploring combination therapies are essential for overcoming challenges in cancer treatment.
Collapse
Affiliation(s)
- An Zhu
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| | - Yu Bai
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
| | - Yanyang Nan
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| |
Collapse
|
7
|
Bottino C, Picant V, Vivier E, Castriconi R. Natural killer cells and engagers: Powerful weapons against cancer. Immunol Rev 2024; 328:412-421. [PMID: 39180430 PMCID: PMC11659922 DOI: 10.1111/imr.13384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
Abstract
Natural killer (NK) cells are innate immune effectors whose functions rely on receptors binding cytokines, recognizing self-molecules, or detecting danger signals expressed by virus-infected or tumor cells. The potent cytotoxic potential makes NK cells promising candidates for cancer immunotherapy. To enhance their activity strategies include cytokine administration, blocking of immune checkpoints, and designing of antibody-based NK cell engagers (NKCEs). NKCEs represent a cutting-edge approach to cancer therapy: they strengthen the NK-to-target cell interactions and optimize tumor killing, possibly overcoming the immunosuppressive tumor microenvironment. NK cells belong to the innate lymphoid cells (ILCs) and are categorized into different subsets also including cells with a memory-like phenotype: this complexity needs to be explored in the context of cancer immunotherapy, particularly when designing NKCEs. Two strategies to enhance NK cell activity in cancer patients can be adopted: activating patients' own NK cells versus the adoptive transfer of ex vivo activated NK cells. Furthermore, the capability of NKCEs to activate γδ T cells could have a significant synergistic effect in immunotherapy.
Collapse
Affiliation(s)
- Cristina Bottino
- Department of Experimental Medicine (DIMES)University of GenovaGenoaItaly
- Laboratory of Clinical and Experimental ImmunologyIRCCS Istituto Giannina GasliniGenoaItaly
| | - Valentin Picant
- Innate Pharma Research LaboratoriesInnate PharmaMarseilleFrance
| | - Eric Vivier
- Innate Pharma Research LaboratoriesInnate PharmaMarseilleFrance
- Centre National de la Recherche Scientifique, INSERM, Centre d'Immunologie de Marseille‐LuminyAix Marseille UniversitéMarseilleFrance
- Assistance Publique‐Hôpitaux de MarseilleHôpital de la Timone, Marseille ImmunopôleMarseilleFrance
| | - Roberta Castriconi
- Department of Experimental Medicine (DIMES)University of GenovaGenoaItaly
- Laboratory of Clinical and Experimental ImmunologyIRCCS Istituto Giannina GasliniGenoaItaly
| |
Collapse
|
8
|
Yu Y, Lien W, Lin W, Pan Y, Huang S, Mou C, Hu CJ, Mou KY. High-Affinity Superantigen-Based Trifunctional Immune Cell Engager Synergizes NK and T Cell Activation for Tumor Suppression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310204. [PMID: 38937984 PMCID: PMC11434130 DOI: 10.1002/advs.202310204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 06/14/2024] [Indexed: 06/29/2024]
Abstract
The development of immune cell engagers (ICEs) can be limited by logistical and functional restrictions associated with fusion protein designs, thus limiting immune cell recruitment to solid tumors. Herein, a high affinity superantigen-based multivalent ICE is developed for simultaneous activation and recruitment of NK and T cells for tumor treatment. Yeast library-based directed evolution is adopted to identify superantigen variants possessing enhanced binding affinity to immunoreceptors expressed on human T cells and NK cells. High-affinity superantigens exhibiting improved immune-stimulatory activities are then incorporated into a superantigen-based tri-functional yeast-display-enhanced multivalent immune cell engager (STYMIE), which is functionalized with a nanobody, a Neo-2/15 cytokine, and an Fc domain for tumor targeting, immune stimulation, and prolonged circulation, respectively. Intravenous administration of STYMIE enhances NK and T cell recruitment into solid tumors, leading to enhanced inhibition in multiple tumor models. The study offers design principles for multifunctional ICEs.
Collapse
Affiliation(s)
- Yao‐An Yu
- Institute of Biomedical SciencesAcademia SinicaTaipei11529Taiwan
- Doctoral Degree Program of Translational MedicineNational Yang Ming Chiao Tung University and Academia SinicaTaipei112Taiwan
| | - Wan‐Ju Lien
- Institute of Biomedical SciencesAcademia SinicaTaipei11529Taiwan
| | - Wen‐Ching Lin
- Institute of Biomedical SciencesAcademia SinicaTaipei11529Taiwan
| | - Yi‐Chung Pan
- Institute of Biomedical SciencesAcademia SinicaTaipei11529Taiwan
| | - Sin‐Wei Huang
- Institute of Biomedical SciencesAcademia SinicaTaipei11529Taiwan
| | - Chung‐Yuan Mou
- Department of ChemistryNational Taiwan UniversityTaipei10617Taiwan
| | - Che‐Ming Jack Hu
- Institute of Biomedical SciencesAcademia SinicaTaipei11529Taiwan
- Doctoral Degree Program of Translational MedicineNational Yang Ming Chiao Tung University and Academia SinicaTaipei112Taiwan
- Biomedical Translation Research CenterAcademia SinicaTaipei11529Taiwan
| | - Kurt Yun Mou
- Institute of Biomedical SciencesAcademia SinicaTaipei11529Taiwan
| |
Collapse
|
9
|
Imširović V, Wensveen FM, Polić B, Jelenčić V. Maintaining the Balance: Regulation of NK Cell Activity. Cells 2024; 13:1464. [PMID: 39273034 PMCID: PMC11393908 DOI: 10.3390/cells13171464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Natural Killer (NK) cells, integral components of the innate immune system, play a crucial role in the protection against intracellular threats. Their cytotoxic power requires that activation is tightly controlled, and in this, they take a unique position within the immune system. Rather than depending on the engagement of a single activating receptor, their activation involves a delicate balance between inhibitory and activating signals mediated through an array of surface molecules. Only when this cumulative balance surpasses a specific threshold do NK cells initiate their activity. Remarkably, the activation threshold of NK cells remains robust even when cells express vastly different repertoires of inhibitory and activating receptors. These threshold values seem to be influenced by NK cell interactions with their environment during development and after release from the bone marrow. Understanding how NK cells integrate this intricate pattern of stimuli is an ongoing area of research, particularly relevant for cellular therapies seeking to harness the anti-cancer potential of these cells by modifying surface receptor expression. In this review, we will explore some of the current dogmas regarding NK cell activation and discuss recent literature addressing advances in our understanding of this field.
Collapse
Affiliation(s)
| | | | | | - Vedrana Jelenčić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
10
|
Rebuffet L, Melsen JE, Escalière B, Basurto-Lozada D, Bhandoola A, Björkström NK, Bryceson YT, Castriconi R, Cichocki F, Colonna M, Davis DM, Diefenbach A, Ding Y, Haniffa M, Horowitz A, Lanier LL, Malmberg KJ, Miller JS, Moretta L, Narni-Mancinelli E, O'Neill LAJ, Romagnani C, Ryan DG, Sivori S, Sun D, Vagne C, Vivier E. High-dimensional single-cell analysis of human natural killer cell heterogeneity. Nat Immunol 2024; 25:1474-1488. [PMID: 38956378 PMCID: PMC11291291 DOI: 10.1038/s41590-024-01883-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/23/2024] [Indexed: 07/04/2024]
Abstract
Natural killer (NK) cells are innate lymphoid cells (ILCs) contributing to immune responses to microbes and tumors. Historically, their classification hinged on a limited array of surface protein markers. Here, we used single-cell RNA sequencing (scRNA-seq) and cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) to dissect the heterogeneity of NK cells. We identified three prominent NK cell subsets in healthy human blood: NK1, NK2 and NK3, further differentiated into six distinct subgroups. Our findings delineate the molecular characteristics, key transcription factors, biological functions, metabolic traits and cytokine responses of each subgroup. These data also suggest two separate ontogenetic origins for NK cells, leading to divergent transcriptional trajectories. Furthermore, we analyzed the distribution of NK cell subsets in the lung, tonsils and intraepithelial lymphocytes isolated from healthy individuals and in 22 tumor types. This standardized terminology aims at fostering clarity and consistency in future research, thereby improving cross-study comparisons.
Collapse
Grants
- Wellcome Trust
- MR/W031698/1 Medical Research Council
- P01 CA065493 NCI NIH HHS
- P01 CA111412 NCI NIH HHS
- E.V laboratory at CIML and Assistance-Publique des Hôpitaux de Marseille is supported by funding from the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation program (TILC, grant agreement No. 694502 and MInfla-TILC, grand agreement No. 875102), the Agence Nationale de la Recherche including the PIONEER Project (ANR-17-RHUS-0007), MSDAvenir, Innate Pharma and institutional grants awarded to the CIML (INSERM, CNRS and Aix-Marseille University) and Marseille Immunopole.
- D.M.D laboratory is funded by the Medical Research Council (MR/W031698/1) and the Wellcome Trust (110091/Z/15/Z).
- A.D. laboratory is supported by the European Research Council (ERC AdG ILCAdapt, 101055309 to A.D.) and by the German Research Foundation (DFG) (SFB 1444/427826188 and TRR 241/375876048 to A.D., SPP1937/Di764 /9-2 to A.D.). We are grateful to the Benjamin Franklin Flow Cytometry Facility (BFFC) for support in cell sorting. BFFC is supported by DFG Instrument Grants INST 335/597-1 FUGG und INST 335/777-1 FUGG.
- KJM was supported by the Research Council of Norway, Center of Excellence: Precision Immunotherapy Alliance (332727), the US National Cancer Institute (P01 CA111412, P009500901).
- L.M. is funded by Associazione Italiana contro il Cancro (AIRC), 5xmille project n. 21147.
- C.R. laboratory is supported by the ERC Advanced Grant ‘MEM-CLONK’ (101055157) and the Deutsche Forschungsgemeinschaft (DFG) grants SFB TRR241 B02 and RO 3565/7-1.
- D.G.R is supported by funding from the Medical Research Council (MRC) (MC_UU_00028) and Wellcome Trust-Academy of Medical Sciences (WT-AMS) (SBF009\1119).
- S.S. is funded by Ministero dell’Istruzione, dell’Università e della Ricerca: PRIN 2017WC8499_004 and Fondazione AIRC: AIRC 5×1000 project id. 21147.
Collapse
Affiliation(s)
- Lucas Rebuffet
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Janine E Melsen
- Leiden University Medical Center, Willem-Alexander Children's Hospital, Laboratory for Pediatric Immunology, Leiden, the Netherlands
- Leiden University Medical Center, Department of Immunology, Leiden, the Netherlands
| | - Bertrand Escalière
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Daniela Basurto-Lozada
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Avinash Bhandoola
- T Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Yenan T Bryceson
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Division of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
- Sweden Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Roberta Castriconi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- Laboratory of Clinical and Experimental Immunology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Frank Cichocki
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel M Davis
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, South Kensington, London, UK
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Yi Ding
- T Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Dermatology and NIHR Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Amir Horowitz
- Department of Immunology & Immunotherapy, The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lewis L Lanier
- Department of Microbiology and Immunology and the Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, USA
| | - Karl-Johan Malmberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Precision Immunotherapy Alliance, The University of Oslo, Oslo, Norway
- The Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Emilie Narni-Mancinelli
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Chiara Romagnani
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Innate Immunity, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), ein Leibniz Institut, Berlin, Germany
- Berlin University Alliance, Berlin, Germany
| | - Dylan G Ryan
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Dan Sun
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Constance Vagne
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Eric Vivier
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France.
- APHM, Hôpital de la Timone, Marseille-Immunopôle, Marseille, France.
- Paris-Saclay Cancer Cluster, Le Kremlin-Bicêtre, France.
| |
Collapse
|
11
|
Schoenfeld K, Harwardt J, Kolmar H. Better safe than sorry: dual targeting antibodies for cancer immunotherapy. Biol Chem 2024; 405:443-459. [PMID: 38297991 DOI: 10.1515/hsz-2023-0329] [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: 10/17/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
Antibody-based therapies are revolutionizing cancer treatment and experience a steady increase from preclinical and clinical pipelines to market share. While the clinical success of monoclonal antibodies is frequently limited by low response rates, treatment resistance and various other factors, multispecific antibodies open up new prospects by addressing tumor complexity as well as immune response actuation potently improving safety and efficacy. Novel antibody approaches involve simultaneous binding of two antigens on one cell implying increased specificity and reduced tumor escape for dual tumor-associated antigen targeting and enhanced and durable cytotoxic effects for dual immune cell-related antigen targeting. This article reviews antibody and cell-based therapeutics for oncology with intrinsic dual targeting of either tumor cells or immune cells. As revealed in various preclinical studies and clinical trials, dual targeting molecules are promising candidates constituting the next generation of antibody drugs for fighting cancer.
Collapse
Affiliation(s)
- Katrin Schoenfeld
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Strasse 4, D-64287 Darmstadt, Germany
| | - Julia Harwardt
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Strasse 4, D-64287 Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Strasse 4, D-64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|
12
|
Cantoni C, Falco M, Vitale M, Pietra G, Munari E, Pende D, Mingari MC, Sivori S, Moretta L. Human NK cells and cancer. Oncoimmunology 2024; 13:2378520. [PMID: 39022338 PMCID: PMC11253890 DOI: 10.1080/2162402x.2024.2378520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024] Open
Abstract
The long story of NK cells started about 50 y ago with the first demonstration of a natural cytotoxic activity within an undefined subset of circulating leukocytes, has involved an ever-growing number of researchers, fascinated by the apparently easy-to-reach aim of getting a "universal anti-tumor immune tool". In fact, in spite of the impressive progress obtained in the first decades, these cells proved far more complex than expected and, paradoxically, the accumulating findings have continuously moved forward the attainment of a complete control of their function for immunotherapy. The refined studies of these latter years have indicated that NK cells can epigenetically calibrate their functional potential, in response to specific environmental contexts, giving rise to extraordinarily variegated subpopulations, comprehensive of memory-like cells, tissue-resident cells, or cells in various differentiation stages, or distinct functional states. In addition, NK cells can adapt their activity in response to a complex body of signals, spanning from the interaction with either suppressive or stimulating cells (myeloid-derived suppressor cells or dendritic cells, respectively) to the engagement of various receptors (specific for immune checkpoints, cytokines, tumor/viral ligands, or mediating antibody-dependent cell-mediated cytotoxicity). According to this picture, the idea of an easy and generalized exploitation of NK cells is changing, and the way is opening toward new carefully designed, combined and personalized therapeutic strategies, also based on the use of genetically modified NK cells and stimuli capable of strengthening and redirecting their effector functions against cancer.
Collapse
Affiliation(s)
- Claudia Cantoni
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- Laboratory of Clinical and Experimental Immunology, Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Michela Falco
- Laboratory of Clinical and Experimental Immunology, Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Massimo Vitale
- UO Pathology and Experimental Immunology, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Gabriella Pietra
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- UO Pathology and Experimental Immunology, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Enrico Munari
- Pathology Unit, Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Daniela Pende
- UO Pathology and Experimental Immunology, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Maria Cristina Mingari
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- UO Pathology and Experimental Immunology, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| |
Collapse
|
13
|
Rolin C, Zimmer J, Seguin-Devaux C. Bridging the gap with multispecific immune cell engagers in cancer and infectious diseases. Cell Mol Immunol 2024; 21:643-661. [PMID: 38789528 PMCID: PMC11214628 DOI: 10.1038/s41423-024-01176-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
By binding to multiple antigens simultaneously, multispecific antibodies are expected to substantially improve both the activity and long-term efficacy of antibody-based immunotherapy. Immune cell engagers, a subclass of antibody-based constructs, consist of engineered structures designed to bridge immune effector cells to their target, thereby redirecting the immune response toward the tumor cells or infected cells. The increasing number of recent clinical trials evaluating immune cell engagers reflects the important role of these molecules in new therapeutic approaches for cancer and infections. In this review, we discuss how different immune cell types (T and natural killer lymphocytes, as well as myeloid cells) can be bound by immune cell engagers in immunotherapy for cancer and infectious diseases. Furthermore, we explore the preclinical and clinical advancements of these constructs, and we discuss the challenges in translating the current knowledge from cancer to the virology field. Finally, we speculate on the promising future directions that immune cell engagers may take in cancer treatment and antiviral therapy.
Collapse
Affiliation(s)
- Camille Rolin
- Department of Infection and Immunity, Luxembourg Institute of Health, 29 Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg.
- University of Luxembourg, 2 Place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg.
| | - Jacques Zimmer
- Department of Infection and Immunity, Luxembourg Institute of Health, 29 Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Carole Seguin-Devaux
- Department of Infection and Immunity, Luxembourg Institute of Health, 29 Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| |
Collapse
|
14
|
Wu J. Emerging Innate Immune Cells in Cancer Immunotherapy: Promises and Challenges. BioDrugs 2024; 38:499-509. [PMID: 38700835 PMCID: PMC11246812 DOI: 10.1007/s40259-024-00657-2] [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] [Accepted: 03/30/2024] [Indexed: 05/29/2024]
Abstract
Immune checkpoint inhibitor (ICI)-based therapy has made an unprecedented impact on survival benefit for a subset of cancer patients; however, only a subset of cancer patients is benefiting from ICI therapy if all cancer types are considered. With the advanced understanding of interactions of immune effector cell types and tumors, cell-based therapies are emerging as alternatives to patients who could not benefit from ICI therapy. Pioneering work of chimeric antigen receptor T (CAR-T) therapy for hematological malignancies has brought encouragement to a broad range of development for cellular-based cancer immunotherapy, both innate immune cell-based therapies and T-cell-based therapies. Innate immune cells are important cell types due to their rapid response, versatile function, superior safety profiles being demonstrated in early clinical development, and being able to utilize multiple allogeneic cell sources. Efforts on engineering innate immune cells and exploring their therapeutic potential are rapidly emerging. Some of the therapies, such as CD19 CAR natural killer (CAR-NK) cell-based therapy, have demonstrated comparable early efficacy with CD19 CAR-T cells. These studies underscore the significance of developing innate immune cells for cancer therapy. In this review, we focus on the current development of emerging NK cells, γδ T cells, and macrophages. We also present our views on potential challenges and perspectives to overcome these challenges.
Collapse
Affiliation(s)
- Jennifer Wu
- Department of Urology, Feinberg School of Medicine, Robert Lurie Comprehensive Cancer Center, Northwestern University, 303 E. Superior St, Chicago, IL, 60611, USA.
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Robert Lurie Comprehensive Cancer Center, Chicago, IL, USA.
| |
Collapse
|
15
|
Fenis A, Demaria O, Gauthier L, Vivier E, Narni-Mancinelli E. New immune cell engagers for cancer immunotherapy. Nat Rev Immunol 2024; 24:471-486. [PMID: 38273127 DOI: 10.1038/s41577-023-00982-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 01/27/2024]
Abstract
There have been major advances in the immunotherapy of cancer in recent years, including the development of T cell engagers - antibodies engineered to redirect T cells to recognize and kill cancer cells - for the treatment of haematological malignancies. However, the field still faces several challenges to develop agents that are consistently effective in a majority of patients and cancer types, such as optimizing drug dose, overcoming treatment resistance and improving efficacy in solid tumours. A new generation of T cell-targeted molecules was developed to tackle these issues that are potentially more effective and safer. In addition, agents designed to engage the antitumour activities of other immune cells, including natural killer cells and myeloid cells, are showing promise and have the potential to treat a broader range of cancers.
Collapse
Affiliation(s)
- Aurore Fenis
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille Université, Centre National de la Recherche Scientifique, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Olivier Demaria
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Laurent Gauthier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille Université, Centre National de la Recherche Scientifique, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- Assistance Publique-Hôpitaux de Marseille, Hôpital de la Timone, Marseille Immunopôle, Marseille, France
| | - Emilie Narni-Mancinelli
- Aix Marseille Université, Centre National de la Recherche Scientifique, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
| |
Collapse
|
16
|
Huang M, Liu Y, Yan Q, Peng M, Ge J, Mo Y, Wang Y, Wang F, Zeng Z, Li Y, Fan C, Xiong W. NK cells as powerful therapeutic tool in cancer immunotherapy. Cell Oncol (Dordr) 2024; 47:733-757. [PMID: 38170381 DOI: 10.1007/s13402-023-00909-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Natural killer (NK) cells have gained considerable attention and hold great potential for their application in tumor immunotherapy. This is mainly due to their MHC-unrestricted and pan-specific recognition capabilities, as well as their ability to rapidly respond to and eliminate target cells. To artificially generate therapeutic NK cells, various materials can be utilized, such as peripheral blood mononuclear cells (PBMCs), umbilical cord blood (UCB), induced pluripotent stem cells (iPSCs), and NK cell lines. Exploiting the therapeutic potential of NK cells to treat tumors through in vivo and in vitro therapeutic modalities has yielded positive therapeutic results. CONCLUSION This review provides a comprehensive description of NK cell therapeutic approaches for tumors and discusses the current problems associated with these therapeutic approaches and the prospects of NK cell therapy for tumors.
Collapse
Affiliation(s)
- Mao Huang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yixuan Liu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Qijia Yan
- Department of Pathology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Miao Peng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Junshang Ge
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yongzhen Mo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yumin Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Fuyan Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yong Li
- Department of Medicine, Comprehensive Cancer Center, Baylor College of Medicine, Alkek Building, RM N720, Houston, TX, USA
| | - Chunmei Fan
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, 410013, Changsha, Hunan Province, China.
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| |
Collapse
|
17
|
Keener AB. Natural killer cells show their cancer-fighting worth. Nature 2024; 629:S4-S6. [PMID: 38811700 DOI: 10.1038/d41586-024-01427-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
|
18
|
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: 116] [Impact Index Per Article: 116.0] [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
|
19
|
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
|
20
|
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
|
21
|
Cassanello G, Luna de Abia A, Falchi L. Trial watch: bispecific antibodies for the treatment of relapsed or refractory large B-cell lymphoma. Oncoimmunology 2024; 13:2321648. [PMID: 38445082 PMCID: PMC10913711 DOI: 10.1080/2162402x.2024.2321648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/07/2024] Open
Abstract
Immunotherapy has shaped the treatment approach to diffuse large B-cell lymphoma (DLBCL), with rituximab leading to remarkable improvements in outcomes for both relapsed and treatment-naïve patients. Recently, groundbreaking immunotherapies like chimeric antigen receptor T-cells have entered the treatment arena for relapsed/refractory (R/R) DLBCL and gained regulatory approval in several countries. The concept of harnessing a patient's own T-cells to combat cancer has been further explored through the development of bispecific antibodies (BsAbs), a class of engineered antibody products designed to simultaneously target two different antigens. These novel drugs have demonstrated impressive single-agent activity and manageable toxicity in patients with heavily pretreated B-cell non-Hodgkin lymphoma. In this review, we provide an up-to-date overview of recently completed or ongoing BsAbs trials in patients with R/R DLBCL, including single-agent results, emerging combination data, and novel constructs.
Collapse
Affiliation(s)
- Giulio Cassanello
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, USA
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Alejandro Luna de Abia
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, USA
- Hematology Service, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Lorenzo Falchi
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
22
|
Eitler J, Rackwitz W, Wotschel N, Gudipati V, Murali Shankar N, Sidorenkova A, Huppa JB, Ortiz-Montero P, Opitz C, Künzel SR, Michen S, Temme A, Loureiro LR, Feldmann A, Bachmann M, Boissel L, Klingemann H, Wels WS, Tonn T. CAR-mediated targeting of NK cells overcomes tumor immune escape caused by ICAM-1 downregulation. J Immunother Cancer 2024; 12:e008155. [PMID: 38417916 PMCID: PMC10900364 DOI: 10.1136/jitc-2023-008155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2023] [Indexed: 03/01/2024] Open
Abstract
BACKGROUND The antitumor activity of natural killer (NK) cells can be enhanced by specific targeting with therapeutic antibodies that trigger antibody-dependent cell-mediated cytotoxicity (ADCC) or by genetic engineering to express chimeric antigen receptors (CARs). Despite antibody or CAR targeting, some tumors remain resistant towards NK cell attack. While the importance of ICAM-1/LFA-1 interaction for natural cytotoxicity of NK cells is known, its impact on ADCC induced by the ErbB2 (HER2)-specific antibody trastuzumab and ErbB2-CAR-mediated NK cell cytotoxicity against breast cancer cells has not been investigated. METHODS Here we used NK-92 cells expressing high-affinity Fc receptor FcγRIIIa in combination with trastuzumab or ErbB2-CAR engineered NK-92 cells (NK-92/5.28.z) as well as primary human NK cells combined with trastuzumab or modified with the ErbB2-CAR and tested cytotoxicity against cancer cells varying in ICAM-1 expression or alternatively blocked LFA-1 on NK cells. Furthermore, we specifically stimulated Fc receptor, CAR and/or LFA-1 to study their crosstalk at the immunological synapse and their contribution to degranulation and intracellular signaling in antibody-targeted or CAR-targeted NK cells. RESULTS Blockade of LFA-1 or absence of ICAM-1 significantly reduced cell killing and cytokine release during trastuzumab-mediated ADCC against ErbB2-positive breast cancer cells, but not so in CAR-targeted NK cells. Pretreatment with 5-aza-2'-deoxycytidine induced ICAM-1 upregulation and reversed NK cell resistance in ADCC. Trastuzumab alone did not sufficiently activate NK cells and required additional LFA-1 co-stimulation, while activation of the ErbB2-CAR in CAR-NK cells induced efficient degranulation independent of LFA-1. Total internal reflection fluorescence single molecule imaging revealed that CAR-NK cells formed an irregular immunological synapse with tumor cells that excluded ICAM-1, while trastuzumab formed typical peripheral supramolecular activation cluster (pSMAC) structures. Mechanistically, the absence of ICAM-1 did not affect cell-cell adhesion during ADCC, but rather resulted in decreased signaling via Pyk2 and ERK1/2, which was intrinsically provided by CAR-mediated targeting. Furthermore, while stimulation of the inhibitory NK cell checkpoint molecule NKG2A markedly reduced FcγRIIIa/LFA-1-mediated degranulation, retargeting by CAR was only marginally affected. CONCLUSIONS Downregulation of ICAM-1 on breast cancer cells is a critical escape mechanism from trastuzumab-triggered ADCC. In contrast, CAR-NK cells are able to overcome cancer cell resistance caused by ICAM-1 reduction, highlighting the potential of CAR-NK cells in cancer immunotherapy.
Collapse
Affiliation(s)
- Jiri Eitler
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
| | - Wiebke Rackwitz
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Natalie Wotschel
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Venugopal Gudipati
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Nivedha Murali Shankar
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Anastasia Sidorenkova
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Johannes B Huppa
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Paola Ortiz-Montero
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Corinna Opitz
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Stephan R Künzel
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Susanne Michen
- TU Dresden, Medical Faculty and University Hospital Carl Gustav Carus, Department of Neurosurgery, Division of Experimental Neurosurgery and Tumor Immunology, Dresden, Germany
| | - Achim Temme
- TU Dresden, Medical Faculty and University Hospital Carl Gustav Carus, Department of Neurosurgery, Division of Experimental Neurosurgery and Tumor Immunology, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany, National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Liliana Rodrigues Loureiro
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Anja Feldmann
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), Dresden, Germany
| | - Michael Bachmann
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), Dresden, Germany
| | | | | | - Winfried S Wels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Torsten Tonn
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
| |
Collapse
|
23
|
Feng Y, Zhang H, Shao J, Du C, Zhou X, Guo X, Wang Y. Research Progress of Nanomaterials Acting on NK Cells in Tumor Immunotherapy and Imaging. BIOLOGY 2024; 13:153. [PMID: 38534423 DOI: 10.3390/biology13030153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024]
Abstract
The prognosis for cancer patients has declined dramatically in recent years due to the challenges in treating malignant tumors. Tumor immunotherapy, which includes immune target inhibition and chimeric antigen receptor cell treatment, is currently evolving quickly. Among them, natural killer (NK) cells are gradually becoming another preferred cell immunotherapy after T cell immunotherapy due to their unique killing effects in innate and adaptive immunity. NK cell therapy has shown encouraging outcomes in clinical studies; however, there are still some problems, including limited efficacy in solid tumors, inadequate NK cell penetration, and expensive treatment expenses. Noteworthy benefits of nanomaterials include their chemical specificity, biocompatibility, and ease of manufacturing; these make them promising instruments for enhancing NK cell anti-tumor immune responses. Nanomaterials can promote NK cell homing and infiltration, participate in NK cell modification and non-invasive cell tracking and imaging modes, and greatly increase the effectiveness of NK cell immunotherapy. The introduction of NK cell-based immunotherapy research and a more detailed discussion of nanomaterial research in NK cell-based immunotherapy and molecular imaging will be the main topics of this review.
Collapse
Affiliation(s)
- Yachan Feng
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Haojie Zhang
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Jiangtao Shao
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Chao Du
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Xiaolei Zhou
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Xueling Guo
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Yingze Wang
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang 050018, China
| |
Collapse
|
24
|
Vivier E, Rebuffet L, Narni-Mancinelli E, Cornen S, Igarashi RY, Fantin VR. Natural killer cell therapies. Nature 2024; 626:727-736. [PMID: 38383621 DOI: 10.1038/s41586-023-06945-1] [Citation(s) in RCA: 92] [Impact Index Per Article: 92.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/06/2023] [Indexed: 02/23/2024]
Abstract
Natural killer (NK) cells are lymphocytes of the innate immune system. A key feature of NK cells is their ability to recognize a wide range of cells in distress, particularly tumour cells and cells infected with viruses. They combine both direct effector functions against their cellular targets and participate in the generation, shaping and maintenance of a multicellular immune response. As our understanding has deepened, several therapeutic strategies focused on NK cells have been conceived and are currently in various stages of development, from preclinical investigations to clinical trials. Here we explore in detail the complexity of NK cell biology in humans and highlight the role of these cells in cancer immunity. We also analyse the harnessing of NK cell immunity through immune checkpoint inhibitors, NK cell engagers, and infusions of preactivated or genetically modified, autologous or allogeneic NK cell products.
Collapse
Affiliation(s)
- Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France.
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
- APHM, Hôpital de la Timone, Marseille-Immunopôle, Marseille, France.
- Paris-Saclay Cancer Cluster, Le Kremlin-Bicêtre, France.
| | - Lucas Rebuffet
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Emilie Narni-Mancinelli
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Stéphanie Cornen
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | | | | |
Collapse
|
25
|
Zheng W, Ling S, Cao Y, Shao C, Sun X. Combined use of NK cells and radiotherapy in the treatment of solid tumors. Front Immunol 2024; 14:1306534. [PMID: 38264648 PMCID: PMC10803658 DOI: 10.3389/fimmu.2023.1306534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/22/2023] [Indexed: 01/25/2024] Open
Abstract
Natural killer (NK) cells are innate lymphocytes possessing potent tumor surveillance and elimination activity. Increasing attention is being focused on the role of NK cells in integral antitumor strategies (especially immunotherapy). Of note, therapeutic efficacy is considerable dependent on two parameters: the infiltration and cytotoxicity of NK cells in tumor microenvironment (TME), both of which are impaired by several obstacles (e.g., chemokines, hypoxia). Strategies to overcome such barriers are needed. Radiotherapy is a conventional modality employed to cure solid tumors. Recent studies suggest that radiotherapy not only damages tumor cells directly, but also enhances tumor recognition by immune cells through altering molecular expression of tumor or immune cells via the in situ or abscopal effect. Thus, radiotherapy may rebuild a NK cells-favored TME, and thus provide a cost-effective approach to improve the infiltration of NK cells into solid tumors, as well as elevate immune-activity. Moreover, the radioresistance of tumor always hampers the response to radiotherapy. Noteworthy, the puissant cytotoxic activity of NK cells not only kills tumor cells directly, but also increases the response of tumors to radiation via activating several radiosensitization pathways. Herein, we review the mechanisms by which NK cells and radiotherapy mutually promote their killing function against solid malignancies. We also discuss potential strategies harnessing such features in combined anticancer care.
Collapse
Affiliation(s)
- Wang Zheng
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Sunkai Ling
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuandong Cao
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chunlin Shao
- Institution of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
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
|
27
|
Nersesian S, Carter EB, Lee SN, Westhaver LP, Boudreau JE. Killer instincts: natural killer cells as multifactorial cancer immunotherapy. Front Immunol 2023; 14:1269614. [PMID: 38090565 PMCID: PMC10715270 DOI: 10.3389/fimmu.2023.1269614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Natural killer (NK) cells integrate heterogeneous signals for activation and inhibition using germline-encoded receptors. These receptors are stochastically co-expressed, and their concurrent engagement and signaling can adjust the sensitivity of individual cells to putative targets. Against cancers, which mutate and evolve under therapeutic and immunologic pressure, the diversity for recognition provided by NK cells may be key to comprehensive cancer control. NK cells are already being trialled as adoptive cell therapy and targets for immunotherapeutic agents. However, strategies to leverage their naturally occurring diversity and agility have not yet been developed. In this review, we discuss the receptors and signaling pathways through which signals for activation or inhibition are generated in NK cells, focusing on their roles in cancer and potential as targets for immunotherapies. Finally, we consider the impacts of receptor co-expression and the potential to engage multiple pathways of NK cell reactivity to maximize the scope and strength of antitumor activities.
Collapse
Affiliation(s)
- Sarah Nersesian
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Emily B. Carter
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Stacey N. Lee
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | | | - Jeanette E. Boudreau
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
28
|
Momayyezi P, Bilev E, Ljunggren HG, Hammer Q. Viral escape from NK-cell-mediated immunosurveillance: A lesson for cancer immunotherapy? Eur J Immunol 2023; 53:e2350465. [PMID: 37526136 DOI: 10.1002/eji.202350465] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/02/2023]
Abstract
Natural killer (NK) cells are innate lymphocytes that participate in immune responses against virus-infected cells and tumors. As a countermeasure, viruses and tumors employ strategies to evade NK-cell-mediated immunosurveillance. In this review, we examine immune evasion strategies employed by viruses, focusing on examples from human CMV and severe acute respiratory syndrome coronavirus 2. We explore selected viral evasion mechanisms categorized into three classes: (1) providing ligands for the inhibitory receptor NKG2A, (2) downregulating ligands for the activating receptor NKG2D, and (3) inducing the immunosuppressive cytokine transforming growth factor (TGF)-β. For each class, we draw parallels between immune evasion by viruses and tumors, reviewing potential opportunities for overcoming evasion in cancer therapy. We suggest that in-depth investigations of host-pathogen interactions between viruses and NK cells will not only deepen our understanding of viral immune evasion but also shed light on how NK cells counter such evasion attempts. Thus, due to the parallels of immune evasion by viruses and tumors, we propose that insights gained from antiviral NK-cell responses may serve as valuable lessons that can be leveraged for designing future cancer immunotherapies.
Collapse
Affiliation(s)
- Pouria Momayyezi
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Eleni Bilev
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Hans-Gustaf Ljunggren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Quirin Hammer
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| |
Collapse
|
29
|
Yanakieva D, Vollmer L, Evers A, Siegmund V, Arras P, Pekar L, Doerner A, Valldorf B, Kolmar H, Zielonka S, Krah S. Cattle-derived knob paratopes grafted onto peripheral loops of the IgG1 Fc region enable the generation of a novel symmetric bispecific antibody format. Front Immunol 2023; 14:1238313. [PMID: 37942319 PMCID: PMC10628450 DOI: 10.3389/fimmu.2023.1238313] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
In this work we present a novel symmetric bispecific antibody format based on engraftments of cattle-derived knob paratopes onto peripheral loops of the IgG1 Fc region. For this, knob architectures obtained from bovine ultralong CDR-H3 antibodies were inserted into the AB loop or EF loop of the CH3 domain, enabling the introduction of an artificial binding specificity into an IgG molecule. We demonstrate that inserted knob domains largely retain their binding affinities, resulting into bispecific antibody derivatives versatile for effector cell redirection. Essentially, generated bispecifics demonstrated adequate biophysical properties and were not compromised in their Fc mediated functionalities such as FcRn or FcγRIIIa binding.
Collapse
Affiliation(s)
- Desislava Yanakieva
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Lena Vollmer
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Andreas Evers
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Vanessa Siegmund
- Early Protein Supply and Characterization, Merck Healthcare KGaA, Darmstadt, Germany
| | - Paul Arras
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Lukas Pekar
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Achim Doerner
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | | | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Stefan Zielonka
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Simon Krah
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| |
Collapse
|
30
|
Murali Shankar N, Ortiz-Montero P, Kurzyukova A, Rackwitz W, Künzel SR, Wels WS, Tonn T, Knopf F, Eitler J. Preclinical assessment of CAR-NK cell-mediated killing efficacy and pharmacokinetics in a rapid zebrafish xenograft model of metastatic breast cancer. Front Immunol 2023; 14:1254821. [PMID: 37885894 PMCID: PMC10599014 DOI: 10.3389/fimmu.2023.1254821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/12/2023] [Indexed: 10/28/2023] Open
Abstract
Natural killer (NK) cells are attractive effectors for adoptive immunotherapy of cancer. Results from first-in-human studies using chimeric antigen receptor (CAR)-engineered primary NK cells and NK-92 cells are encouraging in terms of efficacy and safety. In order to further improve treatment strategies and to test the efficacy of CAR-NK cells in a personalized manner, preclinical screening assays using patient-derived tumor samples are needed. Zebrafish (Danio rerio) embryos and larvae represent an attractive xenograft model to study growth and dissemination of patient-derived tumor cells because of their superb live cell imaging properties. Injection into the organism's circulation allows investigation of metastasis, cancer cell-to-immune cell-interactions and studies of the tumor cell response to anti-cancer drugs. Here, we established a zebrafish larval xenograft model to test the efficacy of CAR-NK cells against metastatic breast cancer in vivo by injecting metastatic breast cancer cells followed by CAR-NK cell injection into the Duct of Cuvier (DoC). We validated the functionality of the system with two different CAR-NK cell lines specific for PD-L1 and ErbB2 (PD-L1.CAR NK-92 and ErbB2.CAR NK-92 cells) against the PD-L1-expressing MDA-MB-231 and ErbB2-expressing MDA-MB-453 breast cancer cell lines. Injected cancer cells were viable and populated peripheral regions of the larvae, including the caudal hematopoietic tissue (CHT), simulating homing of cancer cells to blood forming sites. CAR-NK cells injected 2.5 hours later migrated to the CHT and rapidly eliminated individual cancer cells throughout the organism. Unmodified NK-92 also demonstrated minor in vivo cytotoxicity. Confocal live-cell imaging demonstrated intravascular migration and real-time interaction of CAR-NK cells with MDA-MB-231 cells, explaining the rapid and effective in vivo cytotoxicity. Thus, our data suggest that zebrafish larvae can be used for rapid and cost-effective in vivo assessment of CAR-NK cell potency and to predict patient response to therapy.
Collapse
Affiliation(s)
- Nivedha Murali Shankar
- Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
- CRTD - Center for Regenerative Therapies TU Dresden, Dresden University of Technology, Dresden, Germany
- Center for Healthy Aging, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Paola Ortiz-Montero
- Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Anastasia Kurzyukova
- CRTD - Center for Regenerative Therapies TU Dresden, Dresden University of Technology, Dresden, Germany
- Center for Healthy Aging, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Wiebke Rackwitz
- Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Stephan R. Künzel
- Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Winfried S. Wels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
| | - Torsten Tonn
- Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
| | - Franziska Knopf
- CRTD - Center for Regenerative Therapies TU Dresden, Dresden University of Technology, Dresden, Germany
- Center for Healthy Aging, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Jiri Eitler
- Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| |
Collapse
|
31
|
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
|
32
|
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: 44] [Impact Index Per Article: 22.0] [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
|
33
|
Bottino C, Vitale C, Dondero A, Castriconi R. B7-H3 in Pediatric Tumors: Far beyond Neuroblastoma. Cancers (Basel) 2023; 15:3279. [PMID: 37444389 DOI: 10.3390/cancers15133279] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
B7-H3 is a 4Ig transmembrane protein that emerged as a tumor-associated antigen in neuroblastoma. It belongs to the B7 family, shows an immunoregulatory role toward NK and T cells, and, therefore, has been included in the growing family of immune checkpoints. Besides neuroblastoma, B7-H3 is expressed by many pediatric cancers including tumors of the central nervous system, sarcomas, and acute myeloid leukemia. In children, particularly those affected by solid tumors, the therapeutic protocols are aggressive and cause important life-threatening side effects. Moreover, despite the improved survival observed in the last decade, a relevant number of patients show therapy resistance and fatal relapses. Immunotherapy represents a new frontier in the cure of cancer patients and the targeting of tumor antigens or immune checkpoints blockade showed exciting results in adults. In this encouraging scenario, researchers and clinicians are exploring the possibility to use immunotherapeutics targeting B7-H3; these include mAbs and chimeric antigen receptor T-cells (CAR-T). These tools are rapidly evolving to improve the efficacy and decrease the unwanted side effects; drug-conjugated mAbs, bi-tri-specific mAbs or CAR-T, and, very recently, NK cell engagers (NKCE), tetra-specific molecules engaging a tumor-associated antigen and NK cells, have been generated. Preclinical data are promising, and clinical trials are ongoing. Hopefully, the B7-H3 targeting will provide important benefits to cancer patients.
Collapse
Affiliation(s)
- Cristina Bottino
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
- Laboratory of Clinical and Experimental Immunology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Chiara Vitale
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| | - Alessandra Dondero
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| | - Roberta Castriconi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| |
Collapse
|
34
|
Parodi M, Astigiano S, Carrega P, Pietra G, Vitale C, Damele L, Grottoli M, Guevara Lopez MDLL, Ferracini R, Bertolini G, Roato I, Vitale M, Orecchia P. Murine models to study human NK cells in human solid tumors. Front Immunol 2023; 14:1209237. [PMID: 37388731 PMCID: PMC10301748 DOI: 10.3389/fimmu.2023.1209237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/02/2023] [Indexed: 07/01/2023] Open
Abstract
Since the first studies, the mouse models have provided crucial support for the most important discoveries on NK cells, on their development, function, and circulation within normal and tumor tissues. Murine tumor models were initially set to study murine NK cells, then, ever more sophisticated human-in-mice models have been developed to investigate the behavior of human NK cells and minimize the interferences from the murine environment. This review presents an overview of the models that have been used along time to study NK cells, focusing on the most popular NOG and NSG models, which work as recipients for the preparation of human-in-mice tumor models, the study of transferred human NK cells, and the evaluation of various enhancers of human NK cell function, including cytokines and chimeric molecules. Finally, an overview of the next generation humanized mice is also provided along with a discussion on how traditional and innovative in-vivo and in-vitro approaches could be integrated to optimize effective pre-clinical studies.
Collapse
Affiliation(s)
- Monica Parodi
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Simonetta Astigiano
- Animal Facility, IRCCS Ospedale Policlinico San Martino Genova, Genova, Italy
| | - Paolo Carrega
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy
| | - Gabriella Pietra
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | - Chiara Vitale
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | - Laura Damele
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Melania Grottoli
- Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | | | - Riccardo Ferracini
- Department of Surgical Sciences, Bone and Dental Bioengineering Laboratory, C.I.R Dental School, University of Turin, Turin, Italy
- Department of Surgical Sciences (DISC), University of Genoa, Genoa, Italy
| | - Giulia Bertolini
- “Epigenomics and Biomarkers of Solid Tumors”, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ilaria Roato
- Department of Surgical Sciences, Bone and Dental Bioengineering Laboratory, C.I.R Dental School, University of Turin, Turin, Italy
| | - Massimo Vitale
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Paola Orecchia
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
35
|
Combes AJ, Samad B, Krummel MF. Defining and using immune archetypes to classify and treat cancer. Nat Rev Cancer 2023:10.1038/s41568-023-00578-2. [PMID: 37277485 DOI: 10.1038/s41568-023-00578-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2023] [Indexed: 06/07/2023]
Abstract
Tumours are surrounded by a host immune system that can suppress or promote tumour growth. The tumour microenvironment (TME) has often been framed as a singular entity, suggesting a single type of immune state that is defective and in need of therapeutic intervention. By contrast, the past few years have highlighted a plurality of immune states that can surround tumours. In this Perspective, we suggest that different TMEs have 'archetypal' qualities across all cancers - characteristic and repeating collections of cells and gene-expression profiles at the level of the bulk tumour. We discuss many studies that together support a view that tumours typically draw from a finite number (around 12) of 'dominant' immune archetypes. In considering the likely evolutionary origin and roles of these archetypes, their associated TMEs can be predicted to have specific vulnerabilities that can be leveraged as targets for cancer treatment with expected and addressable adverse effects for patients.
Collapse
Affiliation(s)
- Alexis J Combes
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA.
- Bakar ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA.
- UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA, USA.
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA.
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - Bushra Samad
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Bakar ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
- UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - Matthew F Krummel
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA.
- Bakar ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA.
- UCSF Immunoprofiler Initiative, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
36
|
Ruf B, Greten TF, Korangy F. Innate lymphoid cells and innate-like T cells in cancer - at the crossroads of innate and adaptive immunity. Nat Rev Cancer 2023; 23:351-371. [PMID: 37081117 DOI: 10.1038/s41568-023-00562-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 04/22/2023]
Abstract
Immunotherapies targeting conventional T cells have revolutionized systemic treatment for many cancers, yet only a subset of patients benefit from these approaches. A better understanding of the complex immune microenvironment of tumours is needed to design the next generation of immunotherapeutics. Innate lymphoid cells (ILCs) and innate-like T cells (ILTCs) are abundant, tissue-resident lymphocytes that have recently been shown to have critical roles in many types of cancers. ILCs and ILTCs rapidly respond to changes in their surrounding environment and act as the first responders to bridge innate and adaptive immunity. This places ILCs and ILTCs as pivotal orchestrators of the final antitumour immune response. In this Review, we outline hallmarks of ILCs and ILTCs and discuss their emerging role in antitumour immunity, as well as the pathophysiological adaptations leading to their pro-tumorigenic function. We explore the pleiotropic, in parts redundant and sometimes opposing, mechanisms that underlie the delicate interplay between the different subsets of ILCs and ILTCs. Finally, we highlight their role in amplifying and complementing conventional T cell functions and summarize immunotherapeutic strategies for targeting ILCs and ILTCs in cancer.
Collapse
Affiliation(s)
- Benjamin Ruf
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Centre for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Centre for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- NCI CCR Liver Cancer Program, National Institutes of Health, Bethesda, MD, USA
| | - Firouzeh Korangy
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Centre for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
37
|
Addressing Natural Killer Cell Dysfunction and Plasticity in Cell-Based Cancer Therapeutics. Cancers (Basel) 2023; 15:cancers15061743. [PMID: 36980629 PMCID: PMC10046032 DOI: 10.3390/cancers15061743] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023] Open
Abstract
Natural killer (NK) cells are cytotoxic group 1 innate lymphoid cells (ILC), known for their role as killers of stressed, cancerous, and virally infected cells. Beyond this cytotoxic function, NK cell subsets can influence broader immune responses through cytokine production and have been linked to central roles in non-immune processes, such as the regulation of vascular remodeling in pregnancy and cancer. Attempts to exploit the anti-tumor functions of NK cells have driven the development of various NK cell-based therapies, which have shown promise in both pre-clinical disease models and early clinical trials. However, certain elements of the tumor microenvironment, such as elevated transforming growth factor (TGF)-β, hypoxia, and indoalemine-2,3-dioxygenase (IDO), are known to suppress NK cell function, potentially limiting the longevity and activity of these approaches. Recent studies have also identified these factors as contributors to NK cell plasticity, defined by the conversion of classical cytotoxic NK cells into poorly cytotoxic, tissue-resident, or ILC1-like phenotypes. This review summarizes the current approaches for NK cell-based cancer therapies and examines the challenges presented by tumor-linked NK cell suppression and plasticity. Ongoing efforts to overcome these challenges are discussed, along with the potential utility of NK cell therapies to applications outside cancer.
Collapse
|
38
|
Lipinski B, Arras P, Pekar L, Klewinghaus D, Boje AS, Krah S, Zimmermann J, Klausz K, Peipp M, Siegmund V, Evers A, Zielonka S. NKp46-specific single domain antibodies enable facile engineering of various potent NK cell engager formats. Protein Sci 2023; 32:e4593. [PMID: 36775946 PMCID: PMC9951198 DOI: 10.1002/pro.4593] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/14/2023]
Abstract
Herein, we describe the generation of potent NK cell engagers (NKCEs) based on single domain antibodies (sdAbs) specific for NKp46 harboring the humanized Fab version of Cetuximab for tumor targeting. After immunization of camelids, a plethora of different VHH domains were retrieved by yeast surface display. Upon reformatting into Fc effector-silenced NKCEs targeting NKp46 and EGFR in a strictly monovalent fashion, the resulting bispecific antibodies elicited potent NK cell-mediated killing of EGFR-overexpressing tumor cells with potencies (EC50 killing) in the picomolar range. This was further augmented via co-engagement of Fcγ receptor IIIa (FcγRIIIa). Importantly, NKp46-specific sdAbs enabled the construction of various NKCE formats with different geometries and valencies which displayed favorable biophysical and biochemical properties without further optimization. By this means, killing capacities were further improved significantly. Hence, NKp46-specific sdAbs are versatile building blocks for the construction of different NKCE formats.
Collapse
Affiliation(s)
- Britta Lipinski
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
- Institute for Organic Chemistry and BiochemistryTechnical University of DarmstadtDarmstadtGermany
| | - Paul Arras
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
| | - Lukas Pekar
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
| | - Daniel Klewinghaus
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
| | - Ammelie Svea Boje
- Division of Antibody‐Based Immunotherapy, Department of Internal Medicine IIUniversity Hospital Schleswig‐Holstein and Christian‐Albrechts‐University KielKielGermany
| | - Simon Krah
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
| | - Jasmin Zimmermann
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
- Institute for Organic Chemistry and BiochemistryTechnical University of DarmstadtDarmstadtGermany
| | - Katja Klausz
- Division of Antibody‐Based Immunotherapy, Department of Internal Medicine IIUniversity Hospital Schleswig‐Holstein and Christian‐Albrechts‐University KielKielGermany
| | - Matthias Peipp
- Division of Antibody‐Based Immunotherapy, Department of Internal Medicine IIUniversity Hospital Schleswig‐Holstein and Christian‐Albrechts‐University KielKielGermany
| | | | - Andreas Evers
- Computational Chemistry and BiologyMerck Healthcare KGaADarmstadtGermany
| | - Stefan Zielonka
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
- Institute for Organic Chemistry and BiochemistryTechnical University of DarmstadtDarmstadtGermany
| |
Collapse
|
39
|
Yao Y, Hu Y, Wang F. Trispecific antibodies for cancer immunotherapy. Immunology 2023. [PMID: 36855956 DOI: 10.1111/imm.13636] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
Despite the clinical success of monoclonal and bispecific antibodies, there are still limitations in the therapeutic effect of malignant tumours, such as low response rate, treatment resistance, and so on, inspiring the exploration of trispecific antibodies (TsAbs). TsAbs further improve the safety and efficacy and has great clinical potential through three targets combination and formats optimization. This article reviews the development history and the target combination features of TsAbs. Although there are still great challenges in the clinical application of TsAbs, it is undeniable that TsAbs may be a breakthrough in the development of antibody drugs.
Collapse
Affiliation(s)
- Yin Yao
- Department of Pharmacy, Fourth People's Hospital of Gui Yang, Guiyang, China
| | - Yiyin Hu
- Department of Nursing, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Fei Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
40
|
Lipinski B, Unmuth L, Arras P, Becker S, Bauer C, Toleikis L, Krah S, Doerner A, Yanakieva D, Boje AS, Klausz K, Peipp M, Siegmund V, Evers A, Kolmar H, Pekar L, Zielonka S. Generation and engineering of potent single domain antibody-based bispecific IL-18 mimetics resistant to IL-18BP decoy receptor inhibition. MAbs 2023; 15:2236265. [PMID: 37469014 PMCID: PMC10361135 DOI: 10.1080/19420862.2023.2236265] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 06/28/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
Here, we generated bispecific antibody (bsAb) derivatives that mimic the function of interleukin (IL)-18 based on single domain antibodies (sdAbs) specific to IL-18 Rα and IL-18 Rβ. For this, camelids were immunized, followed by yeast surface display (YSD)-enabled discovery of VHHs targeting the individual receptor subunits. Upon reformatting into a strictly monovalent (1 + 1) bispecific sdAb architecture, several bsAbs triggered dose-dependent IL-18 R downstream signaling on IL-18 reporter cells, as well as IFN-γ release by peripheral blood mononuclear cells in the presence of low-dose IL-12. However, compared with IL-18, potencies and efficacies were considerably attenuated. By engineering paratope valencies and the spatial orientation of individual paratopes within the overall design architecture, we were able to generate IL-18 mimetics displaying significantly augmented functionalities, resulting in bispecific cytokine mimetics that were more potent than IL-18 in triggering proinflammatory cytokine release. Furthermore, generated IL-18 mimetics were unaffected from inhibition by IL-18 binding protein decoy receptor. Essentially, we demonstrate that this strategy enables the generation of IL-18 mimetics with tailor-made cytokine functionalities.
Collapse
Affiliation(s)
- Britta Lipinski
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Laura Unmuth
- Early Protein Supply and Characterization (EPSC), Merck Healthcare KGaA, Darmstadt, Germany
| | - Paul Arras
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
| | - Stefan Becker
- Early Protein Supply and Characterization (EPSC), Merck Healthcare KGaA, Darmstadt, Germany
| | - Christina Bauer
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
| | - Lars Toleikis
- Early Protein Supply and Characterization (EPSC), Merck Healthcare KGaA, Darmstadt, Germany
| | - Simon Krah
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
| | - Achim Doerner
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
| | - Desislava Yanakieva
- Antibody Discovery and Protein Engineering (ADPE), 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
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian-Albrechts-University Kiel, Kiel, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein and Christian-Albrechts-University Kiel, Kiel, Germany
| | - Vanessa Siegmund
- Early Protein Supply and Characterization (EPSC), Merck Healthcare KGaA, Darmstadt, Germany
| | - Andreas Evers
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Lukas Pekar
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
| | - Stefan Zielonka
- Antibody Discovery and Protein Engineering (ADPE), Merck Healthcare KGaA, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|