1
|
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
|
2
|
Liu L, Yao W, Wang M, Wang B, Kong F, Fan Z, Fan G. A systematic review of cardiovascular toxicities induced by cancer immune therapies: Underlying mechanisms, clinical manifestations and therapeutic approaches. Semin Cancer Biol 2024; 106-107:179-191. [PMID: 39442678 DOI: 10.1016/j.semcancer.2024.10.004] [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: 04/29/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Immunotherapy has revolutionized the management of various types of cancers, even those previously deemed untreatable. Nonetheless, these medications have been associated with inflammation and damage across various organs. These challenges are exemplified by the adverse cardiovascular impacts of cancer immunotherapy, which need comprehensive understanding, clarification, and management integrated into the overall care of cancer patients. Numerous anticancer immunotherapies have been linked to the prevalence and severity of cardiovascular toxicity. These challenges emphasize the importance of conducting fundamental and applied research to elucidate disease causes, discover prognostic indicators, enhance diagnostic methods, and create successful therapies. Despite the acknowledged importance of T cells, there remains a knowledge gap regarding the inciting antigens, the reasons for their recognition, and the mechanisms of how they contribute to cardiac cell injury. In this review, we summarize the molecular mechanism, epidemiology, diagnosis, pathophysiology and corresponding treatment of cardiovascular toxicity induced by immunotherapy, including immune checkpoint inhibitors (ICIs), adoptive cell therapies (ACT), and bi-specific T-cell engagers (BiTEs) among others. By elucidating these aspects, we aim to provide a better understanding of immunotherapies in cancer treatment and offer guidance for their clinical application.
Collapse
Affiliation(s)
- Li Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Wentao Yao
- Department of Urology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Mi Wang
- Department of Cardiology Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Baohui Wang
- Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fanming Kong
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Zhongguo Fan
- Department of Cardiology Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
3
|
Lai M, Pichardo-Almarza C, Verma M, Shahinuzzaman M, Zhu X, Kimko H. T-cell engagers: model interrogation as a tool to quantify the interplay of relative affinity and target expression on trimer formation. Front Pharmacol 2024; 15:1470595. [PMID: 39439898 PMCID: PMC11493665 DOI: 10.3389/fphar.2024.1470595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/06/2024] [Indexed: 10/25/2024] Open
Abstract
T-cell engagers (TCEs) represent a promising therapeutic strategy for various cancers and autoimmune disorders. These bispecific antibodies act as bridges, connecting T-cell receptors (TCRs) to target cells (either malignant or autoreactive) via interactions with specific tumour-associated antigens (TAAs) or autoantigens to form trimeric synapses, or trimers, that co-localise T-cells with target cells and stimulate their cytotoxic function. Bispecific TCEs are expected to exhibit a bell-shaped dose-response curve, with a defined optimal TCE exposure for maximizing trimer formation. The shape of the dose-response is determined by a non-trivial interplay of binding affinities, exposure and antigens expression levels. Furthermore, excessively low binding to the TCR may reduce efficacy, but mitigate risk of over-stimulating cytokine secretion or induce effector cell exhaustion. These inevitable trade-off highlights the importance of quantitatively understanding the relationship between TCE concentration, target expression, binding affinities, and trimer formation. We utilized a mechanistic target engagement model to show that, if the TCE design parameters are close to the recommended ranges found in the literature, relative affinities for TCR, TAA and target expression levels have qualitatively different, but predictable, effects on the resulting dose-response curve: higher expression levels shift the curve upwards, higher antigen affinity shifts the curve to the left, and higher TCR affinity shifts the curve upwards and to the left.
Collapse
Affiliation(s)
- Massimo Lai
- Clinical Pharmacology and Quantitative Pharmacology, R&D Biopharmaceuticals, AstraZeneca plc, Cambridge, United Kingdom
| | - Cesar Pichardo-Almarza
- Clinical Pharmacology and Quantitative Pharmacology, R&D Biopharmaceuticals, AstraZeneca plc, Cambridge, United Kingdom
| | - Meghna Verma
- Clinical Pharmacology and Quantitative Pharmacology, R&D Biopharmaceuticals, AstraZeneca Plc, Gaithersburg, MD, United States
| | - Md Shahinuzzaman
- Clinical Pharmacology and Quantitative Pharmacology, R&D Biopharmaceuticals, AstraZeneca Plc, Gaithersburg, MD, United States
| | - Xu Zhu
- Clinical Pharmacology and Quantitative Pharmacology, R&D Biopharmaceuticals, AstraZeneca Plc, Waltham, MA, United States
| | - Holly Kimko
- Clinical Pharmacology and Quantitative Pharmacology, R&D Biopharmaceuticals, AstraZeneca Plc, Gaithersburg, MD, United States
| |
Collapse
|
4
|
Yao Z, Zeng Y, Liu C, Jin H, Wang H, Zhang Y, Ding C, Chen G, Wu D. Focusing on CD8 + T-cell phenotypes: improving solid tumor therapy. J Exp Clin Cancer Res 2024; 43:266. [PMID: 39342365 PMCID: PMC11437975 DOI: 10.1186/s13046-024-03195-5] [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: 07/31/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Vigorous CD8+ T cells play a crucial role in recognizing tumor cells and combating solid tumors. How T cells efficiently recognize and target tumor antigens, and how they maintain the activity in the "rejection" of solid tumor microenvironment, are major concerns. Recent advances in understanding of the immunological trajectory and lifespan of CD8+ T cells have provided guidance for the design of more optimal anti-tumor immunotherapy regimens. Here, we review the newly discovered methods to enhance the function of CD8+ T cells against solid tumors, focusing on optimizing T cell receptor (TCR) expression, improving antigen recognition by engineered T cells, enhancing signal transduction of the TCR-CD3 complex, inducing the homing of polyclonal functional T cells to tumors, reversing T cell exhaustion under chronic antigen stimulation, and reprogramming the energy and metabolic pathways of T cells. We also discuss how to participate in the epigenetic changes of CD8+ T cells to regulate two key indicators of anti-tumor responses, namely effectiveness and persistence.
Collapse
Affiliation(s)
- Zhouchi Yao
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yayun Zeng
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Cheng Liu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Huimin Jin
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hong Wang
- Department of Scientific Research, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Yue Zhang
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Chengming Ding
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Guodong Chen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Daichao Wu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
5
|
Tur C, Eckstein M, Velden J, Rauber S, Bergmann C, Auth J, Bucci L, Corte G, Hagen M, Wirsching A, Grieshaber-Bouyer R, Reis P, Kittan N, Wacker J, Rius Rigau A, Ramming A, D'Agostino MA, Hartmann A, Müller F, Mackensen A, Bozec A, Schett G, Raimondo MG. CD19-CAR T-cell therapy induces deep tissue depletion of B cells. Ann Rheum Dis 2024:ard-2024-226142. [PMID: 39153835 DOI: 10.1136/ard-2024-226142] [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: 05/21/2024] [Accepted: 08/01/2024] [Indexed: 08/19/2024]
Abstract
OBJECTIVES CD19-targeting chimeric antigen receptor (CAR) T-cell therapy can induce long-term drug-free remission in patients with autoimmune diseases (AIDs). The efficacy of CD19-CAR T-cell therapy is presumably based on deep tissue depletion of B cells; however, such effect has not been proven in humans in vivo. METHODS Sequential ultrasound-guided inguinal lymph node biopsies were performed at baseline and after CD19-CAR T-cell therapy in patients with AIDs. Results were compared with lymph node biopsies from rituximab (RTX)-treated AID patients with absence of peripheral B cells. Conventional and immunohistochemistry staining were performed on lymph node tissue to assess architecture as well the number of B cells, follicular dendritic cells (FDCs), plasma cells, T cells and macrophages. RESULTS Sequential lymph node biopsies were analysed from five patients with AID before and after CD19-CAR T-cell therapy and from five patients with AID after RTX treatment. In addition, non-lymphoid organ biopsies (colon, kidney and gallbladder) from three additional patients with AID after CD19-CAR T-cell therapy were analysed. CD19+ and CD20+ B cells were completely depleted in the lymph nodes after CD19-CAR T-cell therapy, but not after RTX treatment. Plasma cells, T cells and macrophages in the lymph nodes remained unchanged. Follicular structures were disrupted and FDCs were depleted in the lymph nodes after CD19-CAR T-cell therapy, but not after RTX. Non-lymphoid organs were completely depleted of B cells. DISCUSSION This study demonstrates complete B-cell depletion in secondary lymphoid tissues of patients with AIDs following CD19-CAR T-cell therapy combined with standard lymphodepleting therapy.
Collapse
Affiliation(s)
- Carlo Tur
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
- Division of Rheumatology-Fondazione Policlinico Universitario A. Gemelli, IRCCS-Università Cattolica del Sacro Cuore, Roma, Lazio, Italy
| | - Markus Eckstein
- Institute of Pathology and Comprehensive Cancer Center EMN, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Joachim Velden
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Simon Rauber
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Christina Bergmann
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Janina Auth
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Laura Bucci
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Giulia Corte
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Melanie Hagen
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Andreas Wirsching
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Ricardo Grieshaber-Bouyer
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Petra Reis
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Nicolai Kittan
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Jochen Wacker
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Aleix Rius Rigau
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Andreas Ramming
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Maria-Antonietta D'Agostino
- Division of Rheumatology-Fondazione Policlinico Universitario A. Gemelli, IRCCS-Università Cattolica del Sacro Cuore, Roma, Lazio, Italy
| | - Arndt Hartmann
- Institute of Pathology and Comprehensive Cancer Center EMN, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
| | - Fabian Müller
- Department of Medicine 5-Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Uniklinikum Erlangen, Erlangen, Bayern, Germany
| | - Andreas Mackensen
- Department of Medicine 5-Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Uniklinikum Erlangen, Erlangen, Bayern, Germany
| | - Aline Bozec
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| | - Georg Schett
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
- Division of Rheumatology-Fondazione Policlinico Universitario A. Gemelli, IRCCS-Università Cattolica del Sacro Cuore, Roma, Lazio, Italy
| | - Maria Gabriella Raimondo
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, Erlangen, Germany
| |
Collapse
|
6
|
Xie Z, Protzer U. Activating adaptive immunity by bispecific, T-cell engager antibodies bridging infected and immune-effector cells is a promising novel therapy for chronic hepatitis B. Antiviral Res 2024; 229:105972. [PMID: 39084340 DOI: 10.1016/j.antiviral.2024.105972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/14/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Bispecific antibodies (bsAbs) are engineered immunoglobulins that combine two different antigen-binding sites in one molecule. BsAbs can be divided into two molecular formats: IgG-like and non-IgG-like antibodies. Structural elements of each format have implications for engaging the immune system. T cell engager antibodies (TCEs) are bsAbs designed to engage T cells with target cells. TCEs can be applied not only in cancer but also in infectious disease therapy to activate T-cell responses. In this review, we focus on current literature on the design and use of bsAbs as an innovative strategy to enhance adaptive antiviral immune responses. We summarized the novel T cell-related immunotherapies with a focus on TCEs, that are developed for the treatment of chronic hepatitis B. Chronic infection with the hepatitis B virus (HBV) had a death toll of 1.1 million humans in 2022, mainly due to liver cirrhosis and hepatocellular carcinoma developing in the more than 250 million humans chronically infected. A curative treatment approach for chronic hepatitis B is lacking. Combining antiviral therapy with immune therapies activating T-cell responses is regarded as the most promising therapeutic approach to curing HBV and preventing the sequelae of chronic infection. Attracting functionally intact T cells that are not HBV-specific and, therefore, have not yet been exposed to regulatory mechanisms and activating those at the target site in the liver is a very interesting therapeutic approach that could be achieved by TCEs. Thus, TCEs redirecting T cells toward HBV-positive cells represent a promising strategy for treating chronic hepatitis B and HBV-associated hepatocellular carcinoma.
Collapse
Affiliation(s)
- Zhe Xie
- Institute of Virology, School of Medicine and Health, Technical University of Munich / Helmholtz Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, School of Medicine and Health, Technical University of Munich / Helmholtz Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Sites, Germany.
| |
Collapse
|
7
|
Garaulet G, Báez BB, Medrano G, Rivas-Sánchez M, Sánchez-Alonso D, Martinez-Torrecuadrada JL, Mulero F. Radioimmunotheragnosis in Cancer Research. Cancers (Basel) 2024; 16:2896. [PMID: 39199666 PMCID: PMC11352548 DOI: 10.3390/cancers16162896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
The combination of immunoPET-where an antibody (Ab) is labeled with an isotope for PET imaging-and radioimmunotherapy (RIT), using the same antibody with a therapeutic isotope, offers significant advantages in cancer management. ImmunoPET allows non-invasive imaging of antigen expression, which aids in patient selection for subsequent radioimmunotherapy. It also facilitates the assessment of tumor response to therapy, allowing for treatment adjustments if necessary. In addition, immunoPET provides critical pharmacokinetic data, including antibody biodistribution and clearance rates, which are essential for dosimetry calculations and treatment protocol optimization. There are still challenges to overcome. Identifying appropriate target antigens that are selectively expressed on cancer cells while minimally expressed on normal tissues remains a major hurdle to reduce off-target toxicity. In addition, it is critical to optimize the pharmacokinetics of radiolabeled antibodies to maximize tumor uptake and minimize normal tissue uptake, particularly in vital organs such as the liver and kidney. This approach offers the potential for targeted and personalized cancer therapy with reduced systemic toxicity by exploiting the specificity of monoclonal antibodies and the cytotoxic effects of radiation. However, further research is needed to address remaining challenges and to optimize these technologies for clinical use.
Collapse
Affiliation(s)
- Guillermo Garaulet
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - Bárbara Beatriz Báez
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - Guillermo Medrano
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - María Rivas-Sánchez
- Protein Production Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (M.R.-S.); (D.S.-A.)
| | - David Sánchez-Alonso
- Protein Production Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (M.R.-S.); (D.S.-A.)
| | | | - Francisca Mulero
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| |
Collapse
|
8
|
Liao CY, Engelberts P, Ioan-Facsinay A, Klip JE, Schmidt T, Ruijtenbeek R, Danen EHJ. CD3-engaging bispecific antibodies trigger a paracrine regulated wave of T-cell recruitment for effective tumor killing. Commun Biol 2024; 7:983. [PMID: 39138287 PMCID: PMC11322607 DOI: 10.1038/s42003-024-06682-9] [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: 04/26/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
The mechanism of action of bispecific antibodies (bsAbs) directing T-cell immunity to solid tumors is incompletely understood. Here, we screened a series of CD3xHER2 bsAbs using extracellular matrix (ECM) embedded breast cancer tumoroid arrays exposed to healthy donor-derived T-cells. An initial phase of random T-cell movement throughout the ECM (day 1-2), was followed by a bsAb-dependent phase of active T-cell recruitment to tumoroids (day 2-4), and tumoroid killing (day 4-6). Low affinity HER2 or CD3 arms were compensated for by increasing bsAb concentrations. Instead, a bsAb binding a membrane proximal HER2 epitope supported tumor killing whereas a bsAb binding a membrane distal epitope did not, despite similar affinities and intra-tumoroid localization of the bsAbs, and efficacy in 2D co-cultures. Initial T-cell-tumor contact through effective bsAbs triggered a wave of subsequent T-cell recruitment. This critical surge of T-cell recruitment was explained by paracrine signaling and preceded a full-scale T-cell tumor attack.
Collapse
Affiliation(s)
- Chen-Yi Liao
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | | | | | - Janna Eleonora Klip
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Thomas Schmidt
- Leiden Institute of Physics, Leiden University, Leiden, the Netherlands
| | | | - Erik H J Danen
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
9
|
Hill TF, Narvekar P, Asher GD, Edelstein JN, Camp ND, Grimm A, Thomas KR, Leiken MD, Molloy KM, Cook PJ, Arlauckas SP, Morgan RA, Tasian SK, Rawlings DJ, James RG. Human plasma cells engineered to secrete bispecifics drive effective in vivo leukemia killing. Mol Ther 2024; 32:2676-2691. [PMID: 38959896 PMCID: PMC11405176 DOI: 10.1016/j.ymthe.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/09/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024] Open
Abstract
Bispecific antibodies are an important tool for the management and treatment of acute leukemias. As a next step toward clinical translation of engineered plasma cells, we describe approaches for secretion of bispecific antibodies by human plasma cells. We show that human plasma cells expressing either fragment crystallizable domain-deficient anti-CD19 × anti-CD3 (blinatumomab) or anti-CD33 × anti-CD3 bispecific antibodies mediate T cell activation and direct T cell killing of B acute lymphoblastic leukemia or acute myeloid leukemia cell lines in vitro. We demonstrate that knockout of the self-expressed antigen, CD19, boosts anti-CD19-bispecific secretion by plasma cells and prevents self-targeting. Plasma cells secreting anti-CD19-bispecific antibodies elicited in vivo control of acute lymphoblastic leukemia patient-derived xenografts in immunodeficient mice co-engrafted with autologous T cells. In these studies, we found that leukemic control elicited by engineered plasma cells was similar to CD19-targeted chimeric antigen receptor-expressing T cells. Finally, the steady-state concentration of anti-CD19 bispecifics in serum 1 month after cell delivery and tumor eradication was comparable with that observed in patients treated with a steady-state infusion of blinatumomab. These findings support further development of ePCs for use as a durable delivery system for the treatment of acute leukemias, and potentially other cancers.
Collapse
Affiliation(s)
- Tyler F Hill
- University of Washington, Medical Scientist Training Program, Seattle, WA, USA; Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Parnal Narvekar
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Gregory D Asher
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | | | - Nathan D Camp
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Annaiz Grimm
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Kerri R Thomas
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | | | | | - Peter J Cook
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | | | | | - Sarah K Tasian
- Children's Hospital of Philadelphia, Division of Oncology and Center for Childhood Cancer Research, Philadelphia, PA, USA; Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David J Rawlings
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA; University of Washington, Departments of Pediatrics and Immunology, Seattle, WA, USA
| | - Richard G James
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA; University of Washington, Departments of Pediatrics and Pharmacology, Seattle, WA, USA.
| |
Collapse
|
10
|
Coënon L, Rigal E, Courot H, Multrier C, Zemiti S, Lambour J, Pugnière M, de Toledo M, Bossis G, Cartron G, Robert B, Martineau P, Fauvel B, Presumey J, Villalba M. Generation of non-genetically modified, CAR-like, NK cells. J Immunother Cancer 2024; 12:e009070. [PMID: 39029925 PMCID: PMC11261687 DOI: 10.1136/jitc-2024-009070] [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: 06/27/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Natural killer (NK) cell therapy is considered an attractive and safe strategy for anticancer therapy. Nevertheless, when autologous or allogenic NK cells are used alone, the clinical benefit has been disappointing. This is partially due to the lack of target specificity. Recently, CD19-specific chimeric antigen receptor (CAR)-NK cells have proven to be safe and potent in patients with B-cell tumors. However, the generation of CAR-NK cells is a complicated manufacturing process. We aim at developing a targeted NK cell therapy without the need for cellular genetic modifications. We took advantage of the natural expression of the IgG Fc receptor CD16a (FcγRIIIa) to induce strong antigen-specific effector functions through antibody-dependent cell-mediated cytotoxicity (ADCC). We have generated the new technology "Pin", which enables the arming of modified monoclonal antibodies (mAbs) onto the CD16a of ex vivo expanded NK (eNK) cells. Methods Ex vivo eNK were prepared from umbilical cord blood cells and expanded using interleukin (IL)-2/IL-15 and Epstein-Barr virus (EBV)-transformed B-lymphoblastoid feeder cells. mAbs were engineered with four substitutions called Pin mutations to increase their affinity to CD16a. eNK were incubated with anti-CD20 or anti-CD19 Pin-mAbs to generate "armed" eNK and were used to assess effector functions in vitro on cancer cell lines, lymphoma patient cells and in vivo. RESULTS CD16a/Pin-mAb interaction is stable for several days and Pin-mAb eNK inherit the mAb specificity and exclusively induce ADCC against targets expressing the cognate antigen. Hence, Pin-mAbs confer long-term selectivity to eNK, which allows specific elimination of the target cells in several in vivo mouse models. Finally, we showed that it is possible to arm eNK with at least two Pin-mAbs simultaneously, to increase efficacy against heterogenous cancer cell populations. CONCLUSIONS The Pin technology provides an off-the-shelf NK cell therapy platform to generate CAR-like NK cells, without genetic modifications, that easily target multiple tumor antigens.
Collapse
MESH Headings
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Humans
- Animals
- Mice
- Receptors, IgG/metabolism
- Receptors, IgG/immunology
- Immunotherapy, Adoptive/methods
- Cell Line, Tumor
- Antigens, CD19/immunology
- Antibody-Dependent Cell Cytotoxicity
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Xenograft Model Antitumor Assays
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/pharmacology
Collapse
Affiliation(s)
- Loïs Coënon
- IRMB, INSERM U1183, University of Montpellier, CHU Montpellier, Montpellier, France
| | | | | | - Caroline Multrier
- IRMB, INSERM U1183, University of Montpellier, CHU Montpellier, Montpellier, France
| | - Sara Zemiti
- IRMB, INSERM U1183, University of Montpellier, CHU Montpellier, Montpellier, France
| | - Jennifer Lambour
- IRMB, INSERM U1183, University of Montpellier, CHU Montpellier, Montpellier, France
| | - Martine Pugnière
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | | | - Guillaume Cartron
- Department of Clinical Hematology, CHU Montpellier, Montpellier, France
| | - Bruno Robert
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Pierre Martineau
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | | | - Martin Villalba
- IRMB, INSERM U1183, University of Montpellier, CHU Montpellier, Montpellier, France
| |
Collapse
|
11
|
Li R, Shi X, Zhang J, Liu B, Shen J, Liu H, Zhou J. CaCO 3-Encapsulated polydopamine with an adsorbed TLR7 agonist for improved tumor photothermal immunotherapy. Heliyon 2024; 10:e33837. [PMID: 39050425 PMCID: PMC11268191 DOI: 10.1016/j.heliyon.2024.e33837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
Because of the tumor's recurrence and significant metastasis, the standard single-therapy paradigm has failed to meet clinical requirements. Recently, researchers have focused their emphasis on phototherapy and immunogenic cell death (ICD) techniques. In response to the current problems of immunotherapy, a multifunctional drug delivery nanosystem (PDA-IMQ@CaCO3-blinatumomab, PICB) was constructed by using high physiological compatibility of polydopamine (PDA) and calcium carbonate (CaCO3). Toll-like receptor 7 (TLR7) agonist imiquimod (IMQ) and bispecific antibody (BsAb) blinatumomab were loaded onto PDA-CaCO3 nanoparticles (NPs). The findings revealed that the system exhibited the advantages of good dispersion, high stability, excellent physiological compatibility, low toxicity, and high drug loading rate. Compared to the control group, it resulted in a 2.4-fold decrease in FOXP3+ regulatory T-cells within the tumor and a 5.0-fold increase in CD4+ effector T-cells, and promoted the production of damage-related molecular patterns to reinvigorate the ICD effect. PICB had a strong inhibitory effect on tumor growth in 4T1 tumor-bearing mice, and has no toxicity to other organs. Therefore, the multifunctional drug delivery nanosystem constructed in this study could effectively exert the properties of various components in vivo, fully demonstrate the synergistic effect between immunotherapy and photothermal therapy, thus significantly improving the tumor therapeutic efficacy, and has a promising clinical application.
Collapse
Affiliation(s)
- RuYan Li
- College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - XianDong Shi
- College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - JingYi Zhang
- College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - BaoQing Liu
- College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Jian Shen
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Biofunctional Materials, Nanjing Normal University, Nanjing, 210023, China
| | - HaiLong Liu
- College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - JiaHong Zhou
- College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| |
Collapse
|
12
|
Wen X, Xu G. Nephrotoxicity in Bispecific Antibodies Recipients: Focus on T-Cell-Engaging Bispecific Antibodies. Onco Targets Ther 2024; 17:545-556. [PMID: 39006885 PMCID: PMC11245674 DOI: 10.2147/ott.s465679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 05/31/2024] [Indexed: 07/16/2024] Open
Abstract
Recently, bispecific antibodies (BsAbs) are evolving the landscape of cancer treatment and have significantly improved the outcomes of relapsed or refractory cancer patients. As increasing BsAbs entered clinical practice, specific toxicities have emerged, and renal side-effects have been described. However, there are a lack of studies analyzing the nephrotoxicity in the anti-cancer BsAbs recipients systematically. In this review, we demonstrate the etiologies, mechanisms, other risk factors and treatment options of kidney injury in the BsAbs recipients to provide a more comprehensive insight into the nephrotoxicity post-BsAbs therapy. Significantly, due to the limited clinical trial data on each subject, we mainly conclude the related etiologies, mechanisms, and risk factors of nephrotoxicity that occur in T-cell-engaging BsAbs recipients. Nephrotoxicity associated with non-T-cell BsAbs may be associated with adverse nephrotoxicity of related monoclonal antibodies to two specific antigens. The aim of this paper is to provide nephrologists and oncologists with theoretical knowledge to provide better medical management for recipients who receive BsAbs, especially T-cell-engaging BsAbs treatment.
Collapse
Affiliation(s)
- Xiaoli Wen
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, People's Republic of China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, People's Republic of China
| |
Collapse
|
13
|
Rosenbaum D, Saftig P. New insights into the function and pathophysiology of the ectodomain sheddase A Disintegrin And Metalloproteinase 10 (ADAM10). FEBS J 2024; 291:2733-2766. [PMID: 37218105 DOI: 10.1111/febs.16870] [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: 03/28/2023] [Revised: 05/11/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023]
Abstract
The 'A Disintegrin And Metalloproteinase 10' (ADAM10) has gained considerable attention due to its discovery as an 'α-secretase' involved in the nonamyloidogenic processing of the amyloid precursor protein, thereby possibly preventing the excessive generation of the amyloid beta peptide, which is associated with the pathogenesis of Alzheimer's disease. ADAM10 was found to exert many additional functions, cleaving about 100 different membrane proteins. ADAM10 is involved in many pathophysiological conditions, ranging from cancer and autoimmune disorders to neurodegeneration and inflammation. ADAM10 cleaves its substrates close to the plasma membrane, a process referred to as ectodomain shedding. This is a central step in the modulation of the functions of cell adhesion proteins and cell surface receptors. ADAM10 activity is controlled by transcriptional and post-translational events. The interaction of ADAM10 with tetraspanins and the way they functionally and structurally depend on each other is another topic of interest. In this review, we will summarize findings on how ADAM10 is regulated and what is known about the biology of the protease. We will focus on novel aspects of the molecular biology and pathophysiology of ADAM10 that were previously poorly covered, such as the role of ADAM10 on extracellular vesicles, its contribution to virus entry, and its involvement in cardiac disease, cancer, inflammation, and immune regulation. ADAM10 has emerged as a regulator controlling cell surface proteins during development and in adult life. Its involvement in disease states suggests that ADAM10 may be exploited as a therapeutic target to treat conditions associated with a dysfunctional proteolytic activity.
Collapse
Affiliation(s)
- David Rosenbaum
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Germany
| | - Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Germany
| |
Collapse
|
14
|
Ochi T, Konishi T, Takenaka K. Bispecific antibodies for multiple myeloma: past, present and future. Int J Hematol 2024; 120:23-33. [PMID: 38613724 DOI: 10.1007/s12185-024-03766-4] [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: 01/30/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/15/2024]
Abstract
Despite the development of various therapeutic agents, multiple myeloma remains incurable. Recently, T-cell redirected immunotherapy has become a promising strategy for the treatment of refractory myeloma. Clinical trials using chimeric antigen receptor (CAR)-T cells and bispecific antibodies have demonstrated successful anti-myeloma responses in triple-class-refractory patients. However, unique and unwanted immune effects associated with on-target/off-target reactivity of activated immune cells need to be considered and properly managed. This review summarizes recent advances in bispecific antibodies for the treatment of refractory myeloma. It outlines the history of their development, along with a discussion of their mechanisms of action and their current and potential future role in myeloma therapy. As more evidence emerges to inform the timing of CAR-T-cell therapy, the results of clinical trials and off-the-shelf nature of bispecifics also suggest the timing of their treatment. These findings will promote further development and application of bispecifics for refractory myeloma in combination with other appropriate agents.
Collapse
Affiliation(s)
- Toshiki Ochi
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, 791-0295, Japan.
- Division of Immune Regulation, Proteo-Science Center, Ehime University, Toon, Ehime, 791-0295, Japan.
| | - Tatsuya Konishi
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, 791-0295, Japan
| | - Katsuto Takenaka
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, 791-0295, Japan
| |
Collapse
|
15
|
Gao T, Irie A, Kouwaki T, Oshiumi H. Development of a single-chain variable antibody fragment against a conserved region of the SARS-CoV-2 spike protein. Sci Rep 2024; 14:14419. [PMID: 38909102 PMCID: PMC11193732 DOI: 10.1038/s41598-024-64103-7] [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: 11/24/2023] [Accepted: 06/05/2024] [Indexed: 06/24/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has prolonged the duration of the pandemic because of the continuous emergence of new variant strains. The emergence of these mutant strains makes it difficult to detect the virus with the existing antibodies; thus, the development of novel antibodies that can target both the variants as well as the original strain is necessary. In this study, we generated a high-affinity monoclonal antibody (5G2) against the highly conserved region of the SARS-CoV-2 spike protein to detect the protein variants. Moreover, we generated its single-chain variable antibody fragment (sc5G2). The sc5G2 expressed in mammalian and bacterial cells detected the spike protein of the original SARS-CoV-2 and variant strains. The resulting sc5G2 will be a useful tool to detect the original SARS-CoV-2 and variant strains.
Collapse
Affiliation(s)
- Tingyu Gao
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Atsushi Irie
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan.
| | - Takahisa Kouwaki
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan.
| |
Collapse
|
16
|
Subklewe M, Magno G, Gebhardt C, Bücklein V, Szelinski F, Arévalo HJR, Hänel G, Dörner T, Zugmaier G, von Bergwelt-Baildon M, Skapenko A, Schulze-Koops H. Application of blinatumomab, a bispecific anti-CD3/CD19 T-cell engager, in treating severe systemic sclerosis: A case study. Eur J Cancer 2024; 204:114071. [PMID: 38691878 DOI: 10.1016/j.ejca.2024.114071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024]
Abstract
Systemic sclerosis, a severe inflammatory autoimmune disease, shares a common thread with cancer through the underlying mechanism of inflammation. This inflammatory milieu not only drives the immune dysregulation characteristic of autoimmune diseases but also plays a pivotal role in the pathogenesis of cancer. Among the cellular components involved, B cells have emerged as key players in hematologic tumor and autoimmune disease, contributing to immune dysregulation and persistent tissue fibrosis in systemic sclerosis, as well as tumor progression and immune evasion in cancer. Consequently, novel therapeutic strategies targeting B cells hold promise in both conditions. Recent exploration of CD19 CAR T cells in severe systemic sclerosis patients has shown great potential, but also introduced possible risks and drawbacks associated with viral vectors, prolonged CAR T cell persistence, lengthy production timelines, high costs, and the necessity of conditioning patients with organotoxic and fertility-damaging chemotherapy. Given these challenges, alternative CD19-depleting approaches are of high interest for managing severe systemic autoimmune diseases. Here, we present the pioneering use of blinatumomab, a bispecific anti-CD3/anti-CD19 T cell engager in a patient with progressive, severe systemic sclerosis, offering a promising alternative for such challenging cases.
Collapse
Affiliation(s)
- Marion Subklewe
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany; Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany; German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany.
| | - Giulia Magno
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany.
| | - Christina Gebhardt
- Division of Rheumatology and Clinical Immunology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany.
| | - Veit Bücklein
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany; Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany; German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany.
| | - Franziska Szelinski
- Department of Medicine, Rheumatology and Clinical Immunology, Charite University Hospital, Berlin, Germany.
| | | | - Gerulf Hänel
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany.
| | - Thomas Dörner
- Department of Medicine, Rheumatology and Clinical Immunology, Charite University Hospital, Berlin, Germany.
| | | | - Michael von Bergwelt-Baildon
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany; German Cancer Consortium (DKTK), Munich and Berlin sites, and German Cancer Research Center, Heidelberg, Germany.
| | - Alla Skapenko
- Division of Rheumatology and Clinical Immunology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany.
| | - Hendrik Schulze-Koops
- Division of Rheumatology and Clinical Immunology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
17
|
Paul S, Konig MF, Pardoll DM, Bettegowda C, Papadopoulos N, Wright KM, Gabelli SB, Ho M, van Elsas A, Zhou S. Cancer therapy with antibodies. Nat Rev Cancer 2024; 24:399-426. [PMID: 38740967 PMCID: PMC11180426 DOI: 10.1038/s41568-024-00690-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/29/2024] [Indexed: 05/16/2024]
Abstract
The greatest challenge in cancer therapy is to eradicate cancer cells with minimal damage to normal cells. Targeted therapy has been developed to meet that challenge, showing a substantially increased therapeutic index compared with conventional cancer therapies. Antibodies are important members of the family of targeted therapeutic agents because of their extraordinarily high specificity to the target antigens. Therapeutic antibodies use a range of mechanisms that directly or indirectly kill the cancer cells. Early antibodies were developed to directly antagonize targets on cancer cells. This was followed by advancements in linker technologies that allowed the production of antibody-drug conjugates (ADCs) that guide cytotoxic payloads to the cancer cells. Improvement in our understanding of the biology of T cells led to the production of immune checkpoint-inhibiting antibodies that indirectly kill the cancer cells through activation of the T cells. Even more recently, bispecific antibodies were synthetically designed to redirect the T cells of a patient to kill the cancer cells. In this Review, we summarize the different approaches used by therapeutic antibodies to target cancer cells. We discuss their mechanisms of action, the structural basis for target specificity, clinical applications and the ongoing research to improve efficacy and reduce toxicity.
Collapse
Affiliation(s)
- Suman Paul
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Maximilian F Konig
- Division of Rheumatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Katharine M Wright
- Discovery Chemistry, Merck Research Laboratory, Merck and Co, West Point, PA, USA
| | - Sandra B Gabelli
- Discovery Chemistry, Merck Research Laboratory, Merck and Co, West Point, PA, USA.
| | - Mitchell Ho
- Antibody Engineering Program, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | | | - Shibin Zhou
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Bucci L, Hagen M, Rothe T, Raimondo MG, Fagni F, Tur C, Wirsching A, Wacker J, Wilhelm A, Auger JP, Pachowsky M, Eckstein M, Alivernini S, Zoli A, Krönke G, Uderhardt S, Bozec A, D'Agostino MA, Schett G, Grieshaber-Bouyer R. Bispecific T cell engager therapy for refractory rheumatoid arthritis. Nat Med 2024; 30:1593-1601. [PMID: 38671240 DOI: 10.1038/s41591-024-02964-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/01/2024] [Indexed: 04/28/2024]
Abstract
Bispecific T cell engagers (BiTEs) kill B cells by engaging T cells. BiTEs are highly effective in acute lymphoblastic leukemia. Here we treated six patients with multidrug-resistant rheumatoid arthritis (RA) with the CD19xCD3 BiTE blinatumomab under compassionate use. Low doses of blinatumomab led to B cell depletion and concomitant decrease of T cells, documenting their engager function. Treatment was safe, with brief increase in body temperature and acute phase proteins during first infusion but no signs of clinically relevant cytokine-release syndrome. Blinatumomab led to a rapid decline in RA clinical disease activity in all patients, improved synovitis in ultrasound and FAPI-PET-CT and reduced autoantibodies. High-dimensional flow cytometry analysis of B cells documented an immune reset with depletion of activated memory B cells, which were replaced by nonclass-switched IgD-positive naïve B cells. Together, these data suggest the feasibility and potential for BiTEs to treat RA. This approach warrants further exploration on other B-cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Laura Bucci
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Melanie Hagen
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tobias Rothe
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Maria Gabriella Raimondo
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Filippo Fagni
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Carlo Tur
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Rheumatology, Fondazione Policlinico Universitario A Gemelli, IRCSS, Catholic University of Sacred Heart, Rome, Italy
| | - Andreas Wirsching
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jochen Wacker
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Artur Wilhelm
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Rheumatology, Charite, Berlin, Germany
| | - Jean-Philippe Auger
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Milena Pachowsky
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Markus Eckstein
- Department of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefano Alivernini
- Department of Rheumatology, Fondazione Policlinico Universitario A Gemelli, IRCSS, Catholic University of Sacred Heart, Rome, Italy
| | - Angelo Zoli
- Department of Rheumatology, Fondazione Policlinico Universitario A Gemelli, IRCSS, Catholic University of Sacred Heart, Rome, Italy
| | - Gerhard Krönke
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Rheumatology, Charite, Berlin, Germany
| | - Stefan Uderhardt
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Maria-Antonietta D'Agostino
- Department of Rheumatology, Fondazione Policlinico Universitario A Gemelli, IRCSS, Catholic University of Sacred Heart, Rome, Italy
| | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.
- Department of Rheumatology, Fondazione Policlinico Universitario A Gemelli, IRCSS, Catholic University of Sacred Heart, Rome, Italy.
- Karolinska Institutet, Stockholm, Sweden.
| | - Ricardo Grieshaber-Bouyer
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
19
|
Suwanchiwasiri K, Phanthaphol N, Somboonpatarakun C, Yuti P, Sujjitjoon J, Luangwattananun P, Maher J, Yenchitsomanus PT, Junking M. Bispecific T cell engager-armed T cells targeting integrin ανβ6 exhibit enhanced T cell redirection and antitumor activity in cholangiocarcinoma. Biomed Pharmacother 2024; 175:116718. [PMID: 38744221 DOI: 10.1016/j.biopha.2024.116718] [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: 02/12/2024] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
Advanced cholangiocarcinoma (CCA) presents a clinical challenge due to limited treatment options, necessitating exploration of innovative therapeutic approaches. Bispecific T cell engager (BTE)-armed T cell therapy shows promise in hematological and solid malignancies, offering potential advantages in safety over continuous BTE infusion. In this context, we developed a novel BTE, targeting CD3 on T cells and integrin αvβ6, an antigen elevated in various epithelial malignancies, on cancer cells. The novel BTE was generated by fusing an integrin αvβ6-binding peptide (A20) to an anti-CD3 (OKT3) single-chain variable fragment (scFv) through a G4S peptide linker (A20/αCD3 BTE). T cells were then armed with A20/αCD3 BTE (A20/αCD3-armed T cells) and assessed for antitumor activity. Our results highlight the specific binding of A20/αCD3 BTE to CD3 on T cells and integrin αvβ6 on target cells, effectively redirecting T cells towards these targets. After co-culture, A20/αCD3-armed T cells exhibited significantly heightened cytotoxicity against integrin αvβ6-expressing target cells compared to unarmed T cells in both KKU-213A cells and A375.β6 cells. Moreover, in a five-day co-culture, A20/αCD3-armed T cells demonstrated superior cytotoxicity against KKU-213A spheroids compared to unarmed T cells. Importantly, A20/αCD3-armed T cells exhibited an increased proportion of the effector memory T cell (Tem) subset, upregulation of T cell activation markers, enhanced T cell proliferation, and increased cytolytic molecule/cytokine production, when compared to unarmed T cells in an integrin αvβ6-dependent manner. These findings support the potential of A20/αCD3-armed T cells as a novel therapeutic approach for integrin αvβ6-expressing cancers.
Collapse
Affiliation(s)
- Kwanpirom Suwanchiwasiri
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok, Thailand; Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nattaporn Phanthaphol
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; School of Cardiovascular and Medical Health, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK.
| | - Chalermchai Somboonpatarakun
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pornpimon Yuti
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jatuporn Sujjitjoon
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Piriya Luangwattananun
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - John Maher
- King's College London, School of Cancer and Pharmaceutical Sciences, CAR Mechanics Lab, Guy's Cancer Centre, Great Maze Pond, London, United Kingdom
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Mutita Junking
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
20
|
Mitra A, Kumar A, Amdare NP, Pathak R. Current Landscape of Cancer Immunotherapy: Harnessing the Immune Arsenal to Overcome Immune Evasion. BIOLOGY 2024; 13:307. [PMID: 38785789 PMCID: PMC11118874 DOI: 10.3390/biology13050307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024]
Abstract
Cancer immune evasion represents a leading hallmark of cancer, posing a significant obstacle to the development of successful anticancer therapies. However, the landscape of cancer treatment has significantly evolved, transitioning into the era of immunotherapy from conventional methods such as surgical resection, radiotherapy, chemotherapy, and targeted drug therapy. Immunotherapy has emerged as a pivotal component in cancer treatment, harnessing the body's immune system to combat cancer and offering improved prognostic outcomes for numerous patients. The remarkable success of immunotherapy has spurred significant efforts to enhance the clinical efficacy of existing agents and strategies. Several immunotherapeutic approaches have received approval for targeted cancer treatments, while others are currently in preclinical and clinical trials. This review explores recent progress in unraveling the mechanisms of cancer immune evasion and evaluates the clinical effectiveness of diverse immunotherapy strategies, including cancer vaccines, adoptive cell therapy, and antibody-based treatments. It encompasses both established treatments and those currently under investigation, providing a comprehensive overview of efforts to combat cancer through immunological approaches. Additionally, the article emphasizes the current developments, limitations, and challenges in cancer immunotherapy. Furthermore, by integrating analyses of cancer immunotherapy resistance mechanisms and exploring combination strategies and personalized approaches, it offers valuable insights crucial for the development of novel anticancer immunotherapeutic strategies.
Collapse
Affiliation(s)
- Ankita Mitra
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida 201309, Uttar Pradesh, India
| | - Nitin P. Amdare
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
21
|
Frebel K, Albring JC, Wohlgemuth A, Schwöppe C, Hailfinger S, Lenz G, Stelljes M. Comparison of antibody-based immunotherapeutics for malignant hematological disease in an experimental murine model. Blood Adv 2024; 8:1934-1945. [PMID: 38197968 PMCID: PMC11021910 DOI: 10.1182/bloodadvances.2023011647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/11/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
ABSTRACT Antibody-based immunotherapies have revolutionized leukemia and lymphoma treatment, with animal studies being crucial in evaluating effectiveness and side effects. By targeting the evolutionary conserved Slamf7 immune receptor, which is naturally expressed by the murine multiple myeloma cell line MPC-11, we have developed a syngeneic mouse model for direct comparison of 3 immunotherapies: monoclonal antibodies (mAb), bispecific T-cell engagers (BiTE), and chimeric antigen receptor (CAR) T cells (CART), all targeting Slamf7. Slamf7-BiTE is a bispecific single-chain antibody consisting of α-Slamf7 and α-CD3 Fv fragments joined through a Gly-Ser linker, and Slamf7-CART comprises the α-Slamf7 Fv fragment fused to the msCD8α transmembrane and msCD28, 4-1BB, and CD3ζ intracellular signaling domains. Slamf7-BiTE and Slamf7-CART effectively killed MPC-11 cells in vitro, independently of Slamf7-mediated inhibitory signaling by self-ligation. After chimerizing the constant region of the rat-anti-mouse Slamf7 antibody to mouse Fc-immunoglobulin G2a for enhanced effector functions, Slamf7-mAb triggered antigen-specific antibody-dependent cellular cytotoxicity by binding to Fcγ receptor IV. In vivo, all 3 immunotherapies showed antitumor effects against Slamf7-expressing targets. Unlike Slamf7-mAb, Slamf7-BiTE led to considerable side effects in test animals, including weight loss and general malaise, which were also observed to a lesser extent after Slamf7-CART infusion. In allogeneic transplant, Slamf7-BiTE and Slamf7-CART maintained activity compared with the nontransplant setting, whereas Slamf7-mAb displayed enhanced antimyeloma activity. In summary, our model faithfully replicates treatment efficacy and side effects detected after human immunotherapy. It aids in developing and improving immunotherapies and may help devise novel approaches to mitigate undesired effects in steady state and allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Karin Frebel
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| | - Jörn C. Albring
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| | - Anika Wohlgemuth
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| | - Christian Schwöppe
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| | - Stephan Hailfinger
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| | - Matthias Stelljes
- Department of Medicine A, Hematology, Oncology, Hemostaseology and Pneumology, University Hospital Münster, Münster, Germany
| |
Collapse
|
22
|
Cheng L, Chen L, Shi Y, Gu W, Ding W, Zheng X, Liu Y, Jiang J, Zheng Z. Efficacy and safety of bispecific antibodies vs. immune checkpoint blockade combination therapy in cancer: a real-world comparison. Mol Cancer 2024; 23:77. [PMID: 38627681 PMCID: PMC11020943 DOI: 10.1186/s12943-024-01956-6] [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: 11/22/2023] [Accepted: 02/07/2024] [Indexed: 04/19/2024] Open
Abstract
Emerging tumor immunotherapy methods encompass bispecific antibodies (BSABs), immune checkpoint inhibitors (ICIs), and adoptive cell immunotherapy. BSABs belong to the antibody family that can specifically recognize two different antigens or epitopes on the same antigen. These antibodies demonstrate superior clinical efficacy than monoclonal antibodies, indicating their role as a promising tumor immunotherapy option. Immune checkpoints are also important in tumor immunotherapy. Programmed cell death protein-1 (PD-1) is a widely acknowledged immune checkpoint target with effective anti-tumor activity. PD-1 inhibitors have demonstrated notable therapeutic efficacy in treating hematological and solid tumors; however, more than 50% of patients undergoing this treatment exhibit a poor response. However, ICI-based combination therapies (ICI combination therapies) have been demonstrated to synergistically increase anti-tumor effects and immune response rates. In this review, we compare the clinical efficacy and side effects of BSABs and ICI combination therapies in real-world tumor immunotherapy, aiming to provide evidence-based approaches for clinical research and personalized tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Linyan Cheng
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Lujun Chen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China
- Institute for Cell Therapy of Soochow University, Changzhou, China
| | - Yuan Shi
- Laboratory of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Weiying Gu
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Weidong Ding
- Department of Hematology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China.
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China.
- Institute for Cell Therapy of Soochow University, Changzhou, China.
| | - Yan Liu
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China.
| | - Jingting Jiang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China.
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China.
- Institute for Cell Therapy of Soochow University, Changzhou, China.
| | - Zhuojun Zheng
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China.
| |
Collapse
|
23
|
Tapia-Galisteo A, Sánchez-Rodríguez I, Narbona J, Iglesias-Hernández P, Aragón-García S, Jiménez-Reinoso A, Compte M, Khan S, Tsuda T, Chames P, Lacadena J, Álvarez-Vallina L, Sanz L. Combination of T cell-redirecting strategies with a bispecific antibody blocking TGF-β and PD-L1 enhances antitumor responses. Oncoimmunology 2024; 13:2338558. [PMID: 38623463 PMCID: PMC11018002 DOI: 10.1080/2162402x.2024.2338558] [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: 11/07/2023] [Accepted: 03/30/2024] [Indexed: 04/17/2024] Open
Abstract
T cell-based immunotherapies for solid tumors have not achieved the clinical success observed in hematological malignancies, partially due to the immunosuppressive effect promoted by the tumor microenvironment, where PD-L1 and TGF-β play a pivotal role. However, durable responses to immune checkpoint inhibitors remain limited to a minority of patients, while TGF-β inhibitors have not reached the market yet. Here, we describe a bispecific antibody for dual blockade of PD-L1 and TFG-β, termed AxF (scFv)2, under the premise that combination with T cell redirecting strategies would improve clinical benefit. The AxF (scFv)2 antibody was well expressed in mammalian and yeast cells, bound both targets and inhibited dose-dependently the corresponding signaling pathways in luminescence-based cellular reporter systems. Moreover, combined treatment with trispecific T-cell engagers (TriTE) or CAR-T cells significantly boosted T cell activation status and cytotoxic response in breast, lung and colorectal (CRC) cancer models. Importantly, the combination of an EpCAMxCD3×EGFR TriTE with the AxF (scFv)2 delayed CRC tumor growth in vivo and significantly enhanced survival compared to monotherapy with the trispecific antibody. In summary, we demonstrated the feasibility of concomitant blockade of PD-L1 and TGF-β by a single molecule, as well as its therapeutic potential in combination with different T cell redirecting agents to overcome tumor microenvironment-mediated immunosuppression.
Collapse
Affiliation(s)
- Antonio Tapia-Galisteo
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Cancer Immunotherapy Unit (UNICA), Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-oncology and Immunotherapy Group, Biomedical Research Institute Hospital 12 de Octubre, Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Iñigo Sánchez-Rodríguez
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Javier Narbona
- Department of Biochemistry and Molecular Biology, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Patricia Iglesias-Hernández
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Saray Aragón-García
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Anaïs Jiménez-Reinoso
- Cancer Immunotherapy Unit (UNICA), Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-oncology and Immunotherapy Group, Biomedical Research Institute Hospital 12 de Octubre, Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marta Compte
- Department of Antibody Engineering, Leadartis SL, Madrid, Spain
| | - Shaukat Khan
- Nemours Children’s Health Delaware, Wilmington, Delaware, USA
| | - Takeshi Tsuda
- Nemours Children’s Health Delaware, Wilmington, Delaware, USA
| | - Patrick Chames
- Aix Marseille Univ, CNRS, INSERM, Institute Paoli-Calmettes, CRCM, Marseille, France
| | - Javier Lacadena
- Department of Biochemistry and Molecular Biology, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-oncology and Immunotherapy Group, Biomedical Research Institute Hospital 12 de Octubre, Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| |
Collapse
|
24
|
Chen M, Zhang J, Li H, Deng Y, Huang Y, Shen W, Zeng Y, Ci T. Engineered platelet-based immune engager for tumor post-surgery treatment. BIOMATERIALS ADVANCES 2024; 158:213796. [PMID: 38342024 DOI: 10.1016/j.bioadv.2024.213796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/13/2024]
Abstract
Tumor metastasis and recurrence are principal reasons for the high mortality and poor prognosis of cancers. Inefficient engagement between T cell and tumor cell, as well as the universal existence of immune checkpoints, are important factors to the limited immunological surveillance of the immune systems to tumor cells. Herein, an immune engager based on engineered platelets with CD3 antibody modification (P-aCD3) was constructed to facilitate the contact between T cell and tumor cell via providing the anchoring sites of above two cells. Combined with the immune checkpoint blockade strategy, P-aCD3 effectively enhanced T cell mediated cytotoxicity and inhibited tumor recurrence and metastasis in mice melanoma postoperative model and breast cancer model, resulting in significantly prolonged survival of mice.
Collapse
Affiliation(s)
- Mo Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province 210009, China
| | - Jinniu Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Huangjuan Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province 210009, China
| | - Yueyang Deng
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province 210009, China
| | - Yun Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenhao Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yixing Zeng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tianyuan Ci
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
25
|
Jabbour E, Zugmaier G, Agrawal V, Martínez-Sánchez P, Rifón Roca JJ, Cassaday RD, Böll B, Rijneveld A, Abdul-Hay M, Huguet F, Cluzeau T, Díaz MT, Vucinic V, González-Campos J, Rambaldi A, Schwartz S, Berthon C, Hernández-Rivas JM, Gordon PR, Brüggemann M, Hamidi A, Chen Y, Wong HL, Panwar B, Katlinskaya Y, Markovic A, Kantarjian H. Single agent subcutaneous blinatumomab for advanced acute lymphoblastic leukemia. Am J Hematol 2024; 99:586-595. [PMID: 38317420 DOI: 10.1002/ajh.27227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 02/07/2024]
Abstract
Blinatumomab is a BiTE® (bispecific T-cell engager) molecule that redirects CD3+ T-cells to engage and lyse CD19+ target cells. Here we demonstrate that subcutaneous (SC) blinatumomab can provide high efficacy and greater convenience of administration. In the expansion phase of a multi-institutional phase 1b trial (ClinicalTrials.gov, NCT04521231), heavily pretreated adults with relapsed/refractory B-cell acute lymphoblastic leukemia (R/R B-ALL) received SC blinatumomab at two doses: (1) 250 μg once daily (QD) for week 1 and 500 μg three times weekly (TIW) thereafter (250 μg/500 μg) or (2) 500 μg QD for week 1 and 1000 μg TIW thereafter (500 μg/1000 μg). The primary endpoint was complete remission/complete remission with partial hematologic recovery (CR/CRh) within two cycles. At the data cutoff of September 15, 2023, 29 patients were treated: 14 at the 250 μg/500 μg dose and 13 at 500 μg/1000 μg dose. Data from two ineligible patients were excluded. At the end of two cycles, 12 of 14 patients (85.7%) from the 250 μg/500 μg dose achieved CR/CRh of which nine patients (75.0%) were negative for measurable residual disease (MRD; <10-4 leukemic blasts). At the 500 μg/1000 μg dose, 12 of 13 patients (92.3%) achieved CR/CRh; all 12 patients (100.0%) were MRD-negative. No treatment-related grade 4 cytokine release syndrome (CRS) or neurologic events (NEs) were reported. SC injections were well tolerated and all treatment-related grade 3 CRS and NEs responded to standard-of-care management, interruption, or discontinuation. Treatment with SC blinatumomab resulted in high efficacy, with high MRD-negativity rates and acceptable safety profile in heavily pretreated adults with R/R B-ALL.
Collapse
Affiliation(s)
- Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Vaibhav Agrawal
- Department of Hematology and Hematopoietic Cell Transplantation, Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, California, USA
| | - Pilar Martínez-Sánchez
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - José J Rifón Roca
- Hematology and Hemotherapy Department, Clínica Universidad de Navarra, Pamplona, Spain
| | - Ryan D Cassaday
- Division of Hematology and Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Boris Böll
- First Department of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Dusseldorf, University of Cologne, Cologne, Germany
| | - Anita Rijneveld
- Department of Hematology, Erasmus University Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Maher Abdul-Hay
- Perlmutter Cancer Center, New York University Langone Health, New York, New York, USA
| | - Françoise Huguet
- Department of Hematology, Institut Universitaire du Cancer-Oncopole CHU de Toulouse, Toulouse, France
| | | | - Mar Tormo Díaz
- Hematology Department, Hospital Clínico Universitario de Valencia, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - Vladan Vucinic
- Department of Hematology and Cell Therapy, University Hospital Leipzig, Leipzig, Germany
| | | | - Alessandro Rambaldi
- Department of Oncology-Hematology, University of Milan, Milan and Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Stefan Schwartz
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität and Humboldt-Universität zu Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Céline Berthon
- Centre Hospitalier Universitaire de Lille, Lille, France
| | - Jesús María Hernández-Rivas
- IBSAL, IBMCC, CSIC, Cancer Research Center, University of Salamanca, Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
- CIBERONC, Research Group CB16/12/00233, Salamanca, Spain
| | | | - Monika Brüggemann
- Department of Hematology, University of Schleswig-Holstein, Kiel, Germany
| | - Ali Hamidi
- Amgen Inc., Thousand Oaks, California, USA
| | - Yuqi Chen
- Amgen Inc., Thousand Oaks, California, USA
| | - Hansen L Wong
- Clinical Pharmacology, Modeling and Simulation, Amgen Inc., South San Francisco, California, USA
| | | | | | | | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
26
|
Zhou X, Xiao X, Kortuem KM, Einsele H. Bispecific Antibodies in the Treatment of Multiple Myeloma. Hematol Oncol Clin North Am 2024; 38:361-381. [PMID: 38199897 DOI: 10.1016/j.hoc.2023.12.003] [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] [Indexed: 01/12/2024]
Abstract
The treatment of multiple myeloma (MM) is evolving rapidly. In recent years, T-cell-based novel immunotherapies emerged as new treatment strategies for patients with relapsed/refractory MM, including highly effective new options like chimeric antigen receptor (CAR)-modified T cells and bispecific antibodies (bsAbs). Currently, B-cell maturation antigen is the most commonly used target antigen for CAR T-cell and bsAb therapies in MM. Results from different clinical trials have demonstrated promising efficacy and acceptable safety profile of bsAb in RRMM.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Xianghui Xiao
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Klaus Martin Kortuem
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany.
| |
Collapse
|
27
|
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: 0] [Impact Index Per Article: 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
|
28
|
Kastenschmidt JM, Schroers-Martin JG, Sworder BJ, Sureshchandra S, Khodadoust MS, Liu CL, Olsen M, Kurtz DM, Diehn M, Wagar LE, Alizadeh AA. A human lymphoma organoid model for evaluating and targeting the follicular lymphoma tumor immune microenvironment. Cell Stem Cell 2024; 31:410-420.e4. [PMID: 38402619 PMCID: PMC10960522 DOI: 10.1016/j.stem.2024.01.012] [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: 09/01/2023] [Revised: 12/11/2023] [Accepted: 01/30/2024] [Indexed: 02/27/2024]
Abstract
Heterogeneity in the tumor microenvironment (TME) of follicular lymphomas (FLs) can affect clinical outcomes. Current immunotherapeutic strategies, including antibody- and cell-based therapies, variably overcome pro-tumorigenic mechanisms for sustained disease control. Modeling the intact FL TME, with its native, syngeneic tumor-infiltrating leukocytes, is a major challenge. Here, we describe an organoid culture method for cultivating patient-derived lymphoma organoids (PDLOs), which include cells from the native FL TME. We define the robustness of this method by successfully culturing cryopreserved FL specimens from diverse patients and demonstrate the stability of TME cellular composition, tumor somatic mutations, gene expression profiles, and B/T cell receptor dynamics over 3 weeks. PDLOs treated with CD3:CD19 and CD3:CD20 therapeutic bispecific antibodies showed B cell killing and T cell activation. This stable system offers a robust platform for advancing precision medicine efforts in FL through patient-specific modeling, high-throughput screening, TME signature identification, and treatment response evaluation.
Collapse
Affiliation(s)
- Jenna M Kastenschmidt
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92617, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92617, USA; Cancer Research Institute, University of California, Irvine, Irvine, CA 92617, USA
| | | | - Brian J Sworder
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Suhas Sureshchandra
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92617, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92617, USA; Cancer Research Institute, University of California, Irvine, Irvine, CA 92617, USA
| | - Michael S Khodadoust
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA
| | - Chih Long Liu
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mari Olsen
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - David M Kurtz
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Lisa E Wagar
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA 92617, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92617, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92617, USA; Cancer Research Institute, University of California, Irvine, Irvine, CA 92617, USA.
| | - Ash A Alizadeh
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Division of Hematology, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
29
|
Davis JD, Bravo Padros M, Conrado DJ, Ganguly S, Guan X, Hassan HE, Hazra A, Irvin SC, Jayachandran P, Kosloski MP, Lin KJ, Mukherjee K, Paccaly A, Papachristos A, Partridge MA, Prabhu S, Visich J, Welf ES, Xu X, Zhao A, Zhu M. Subcutaneous Administration of Monoclonal Antibodies: Pharmacology, Delivery, Immunogenicity, and Learnings From Applications to Clinical Development. Clin Pharmacol Ther 2024; 115:422-439. [PMID: 38093583 DOI: 10.1002/cpt.3150] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/06/2023] [Indexed: 01/11/2024]
Abstract
Subcutaneous (s.c.) administration of monoclonal antibodies (mAbs) can reduce treatment burden for patients and healthcare systems compared with intravenous (i.v.) infusion through shorter administration times, made possible by convenient, patient-centric devices. A deeper understanding of clinical pharmacology principles related to efficacy and safety of s.c.-administered mAbs over the past decade has streamlined s.c. product development. This review presents learnings from key constituents of the s.c. mAb development pathway, including pharmacology, administration variables, immunogenicity, and delivery devices. Restricted mAb transportation through the hypodermis explains their incomplete absorption at a relatively slow rate (pharmacokinetic (PK)) and may impact mAb-cellular interactions and/or onset and magnitude of physiological responses (pharmacodynamic). Injection volumes, formulation, rate and site of injection, and needle attributes may affect PKs and the occurrence/severity of adverse events like injection-site reactions or pain, with important consequences for treatment adherence. A review of immunogenicity data for numerous compounds reveals that incidence of anti-drug antibodies (ADAs) is generally comparable across i.v. and s.c. routes, and complementary factors including response magnitude (ADA titer), persistence over time, and neutralizing antibody presence are needed to assess clinical impact. Finally, four case studies showcase how s.c. biologics have been clinically developed: (i) by implementation of i.v./s.c. bridging strategies to streamline PD-1/PD-L1 inhibitor development, (ii) through co-development with i.v. presentations for anti-severe acute respiratory syndrome-coronavirus 2 antibodies to support rapid deployment of both formulations, (iii) as the lead route for bispecific T cell engagers (BTCEs) to mitigate BTCE-mediated cytokine release syndrome, and (iv) for pediatric patients in the case of dupilumab.
Collapse
Affiliation(s)
- John D Davis
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | | | | | - Samit Ganguly
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Xiaowen Guan
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Hazem E Hassan
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Anasuya Hazra
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Susan C Irvin
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | | | | | - Kuan-Ju Lin
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | | | - Anne Paccaly
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | | | | | - Saileta Prabhu
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | | | - Erik S Welf
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Xiaoying Xu
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - An Zhao
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Min Zhu
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| |
Collapse
|
30
|
Yang K, Yi T. Tumor cell stemness in gastrointestinal cancer: regulation and targeted therapy. Front Mol Biosci 2024; 10:1297611. [PMID: 38455361 PMCID: PMC10918437 DOI: 10.3389/fmolb.2023.1297611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/14/2023] [Indexed: 03/09/2024] Open
Abstract
The cancer stem cells are a rare group of self-renewable cancer cells capable of the initiation, progression, metastasis and recurrence of tumors, and also a key contributor to the therapeutic resistance. Thus, understanding the molecular mechanism of tumor stemness regulation, especially in the gastrointestinal (GI) cancers, is of great importance for targeting CSC and designing novel therapeutic strategies. This review aims to elucidate current advancements in the understanding of CSC regulation, including CSC biomarkers, signaling pathways, and non-coding RNAs. We will also provide a comprehensive view on how the tumor microenvironment (TME) display an overall tumor-promoting effect, including the recruitment and impact of cancer-associated fibroblasts (CAFs), the establishment of an immunosuppressive milieu, and the induction of angiogenesis and hypoxia. Lastly, this review consolidates mainstream novel therapeutic interventions targeting CSC stemness regulation.
Collapse
Affiliation(s)
- Kangqi Yang
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tuo Yi
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Ghobadi A, Foley NC, Cohen J, Rettig MP, Cashen AF, Gehrs L, Christ S, Street E, Wallace N, Ritchey J, Mehta-Shah N, Westervelt P, Fehniger TA, Kahl B, Bartlett NL, DiPersio JF. Blinatumomab consolidation post-autologous stem cell transplantation in patients with diffuse large B-cell lymphoma. Blood Adv 2024; 8:513-522. [PMID: 37871306 PMCID: PMC10835165 DOI: 10.1182/bloodadvances.2023011130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/18/2023] [Accepted: 10/07/2023] [Indexed: 10/25/2023] Open
Abstract
ABSTRACT Outcomes in patients with relapsed diffuse large B-cell lymphoma (DLBCL) who undergo autologous stem cell transplant (auto-SCT) are poor. Blinatumomab is a CD3/CD19 bispecific T-cell engager that directs cytotoxic T cells to CD19+ cells. Here, we performed a pilot study of blinatumomab consolidation after auto-SCT for 14 patients with DLBCL or transformed follicular lymphoma. All patients underwent standard-of-care auto-SCT with carmustine, etoposide, cytarabine, and melphalan (BEAM) conditioning followed by 1 cycle (4 weeks continuous infusion) of blinatumomab consolidation starting at day 42 after auto-SCT. All 14 patients treated on study completed BEAM auto-SCT and 1 cycle of posttransplant blinatumomab. Five patients developed grade 1 cytokine release syndrome (CRS), with no grade 2 or higher CRS. Immune effector cell-associated neurotoxicity syndrome was not observed. Patients were followed up for 3 years after auto-SCT, with median follow-up of 37 (range, 12-65) months. One-hundred days after auto-SCT (1 month after blinatumomab consolidation), 12 patients (86%) had achieved complete remission. At 1 year after auto-SCT, 7 patients (50%) remained in CR, and 1 patient had died of progressive disease. Patients who relapsed had a lower CD8:CD4 T-cell ratio before starting blinatumomab than patients who remained in remission. This pilot study demonstrates blinatumomab consolidation after auto-SCT is safe and well tolerated. Strategies to increase the CD8:CD4 ratio and use additional cycles of consolidation in a larger randomized trial are needed to confirm the efficacy of consolidation with blinatumomab after auto-SCT. This trial was registered at www.clinicaltrials.gov as #NCT03072771.
Collapse
Affiliation(s)
- Armin Ghobadi
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Nicole C. Foley
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Jared Cohen
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Michael P. Rettig
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Amanda F. Cashen
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Leah Gehrs
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Stephanie Christ
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Emily Street
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Nicholas Wallace
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Julie Ritchey
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Neha Mehta-Shah
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Peter Westervelt
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Todd A. Fehniger
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Brad Kahl
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - Nancy L. Bartlett
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| | - John F. DiPersio
- Washington University School of Medicine, Department of Medicine, Division of Medical Oncology, St. Louis, MO
| |
Collapse
|
32
|
Martin GH, Gonon A, Martin-Jeantet P, Renart-Depontieu F, Biesova Z, Cifuentes A, Mukherjee A, Thisted T, Doerner A, Campbell DO, Bourré L, van der Horst EH, Rezza A, Thiam K. Myeloid and dendritic cells enhance therapeutics-induced cytokine release syndrome features in humanized BRGSF-HIS preclinical model. Front Immunol 2024; 15:1357716. [PMID: 38384461 PMCID: PMC10880010 DOI: 10.3389/fimmu.2024.1357716] [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: 12/18/2023] [Accepted: 01/23/2024] [Indexed: 02/23/2024] Open
Abstract
Objectives Despite their efficacy, some immunotherapies have been shown to induce immune-related adverse events, including the potentially life-threatening cytokine release syndrome (CRS), calling for reliable and translational preclinical models to predict potential safety issues and investigate their rescue. Here, we tested the reliability of humanized BRGSF mice for the assessment of therapeutics-induced CRS features in preclinical settings. Methods BRGSF mice reconstituted with human umbilical cord blood CD34+ cells (BRGSF-CBC) were injected with anti-CD3 antibody (OKT3), anti-CD3/CD19 bispecific T-cell engager Blinatumomab, or VISTA-targeting antibody. Human myeloid and dendritic cells' contribution was investigated in hFlt3L-boosted BRGSF-CBC mice. OKT3 treatment was also tested in human PBMC-reconstituted BRGSF mice (BRGSF-PBMC). Cytokine release, immune cell distribution, and clinical signs were followed. Results OKT3 injection in BRGSF-CBC mice induced hallmark features of CRS, specifically inflammatory cytokines release, modifications of immune cell distribution and activation, body weight loss, and temperature drop. hFlt3L-boosted BRGSF-CBC mice displayed enhanced CRS features, revealing a significant role of myeloid and dendritic cells in this process. Clinical CRS-managing treatment Infliximab efficiently attenuated OKT3-induced toxicity. Comparison of OKT3 treatment's effect on BRGSF-CBC and BRGSF-PBMC mice showed broadened CRS features in BRGSF-CBC mice. CRS-associated features were also observed in hFlt3L-boosted BRGSF-CBC mice upon treatment with other T-cell or myeloid-targeting compounds. Conclusions These data show that BRGSF-CBC mice represent a relevant model for the preclinical assessment of CRS and CRS-managing therapies. They also confirm a significant role of myeloid and dendritic cells in CRS development and exhibit the versatility of this model for therapeutics-induced safety assessment.
Collapse
|
33
|
Wei J, Zheng H, Dai S, Liu M. A bibliometric and knowledge-map analysis of bispecific antibodies in cancer immunotherapy from 2000 to 2023. Heliyon 2024; 10:e23929. [PMID: 38312701 PMCID: PMC10835268 DOI: 10.1016/j.heliyon.2023.e23929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/05/2023] [Accepted: 12/15/2023] [Indexed: 02/06/2024] Open
Abstract
Background Bispecific antibody (BsAb)-based cancer immunotherapy has provided new avenues for the treatment of various malignancies. The approval of Blinatumomab has encouraged further investigation into these treatments, and a series of preclinical and clinical trials have been conducted, together with the publication of numerous articles. Here, the knowledge structure of BsAb-based cancer immunotherapy is summarized using bibliometric analysis to provide in-depth insight into current research trends and foci. Methods The studies included in the bibliometric analysis of BsAbs in cancer immunotherapy were retrieved from the online Web of Science Core Collection (WOSCC) database on April 16th, 2023. Visualization analysis was performed with the help of CtieSpace (version 6.2.2.msi [64-bit]), VOSviewer (version 1.6.19), R (version 4.2.1), and the Bibliometric analysis platform (R-based online data processing tool). Results A total of 1750 papers were identified. Analysis of annual publications and total citations indicated that publications have increased steadily over the past few decades. The USA, followed by Germany, had largest number of publications, making significant contributions to the field. The Memorial Sloan Kettering Cancer Center received the highest number of citations (n = 3769). However, its collaboration and cooperation with different institutions require further strengthening. MAbs and Clinical Cancer Research published the most papers, while Blood and Cancer Research were the most commonly co-cited journals. DM Goldenberg from the USA published the most articles with the highest H-index (34), and the most co-cited author (2137 citations) was PA Baeuerle; both these authors have distinguished achievements in this field. Analysis of co-cited references and keywords showed that the hotspots and research focus on the use of BsAbs for solid tumors have increased rapidly while the application of BsAb immunotherapy in hematologic malignancies has expanded significantly. The hot topics in the field included cytokine release syndrome, the efficacy and safety of BsAbs, resistance mechanisms, and the exploration and optimization of combination therapies. Conclusion Cancer immunotherapies based on BsAbs are a hot topic in research. Current studies focus on the construction and optimization of BsAb structure, as well as their combination with other treatment modalities to improve their efficacy and overcome resistance. Furthermore, it is expected that the ongoing investigation of BsAb-based immunotherapy for solid tumors will bear fruit with significant clinical application prospects in the near future.
Collapse
Affiliation(s)
- Jing Wei
- Department of Medical Oncology/Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Huilan Zheng
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610075, China
| | - Shuang Dai
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ming Liu
- Department of Medical Oncology/Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| |
Collapse
|
34
|
Ha Thi HT, Than VT. Recent applications of RNA therapeutic in clinics. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 203:115-150. [PMID: 38359994 DOI: 10.1016/bs.pmbts.2023.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Ribonucleic acid (RNA) therapy has been extensively researched for several decades and has garnered significant attention in recent years owing to its potential in treating a broad spectrum of diseases. It falls under the domain of gene therapy, leveraging RNA molecules as a therapeutic approach in medicine. RNA can be targeted using small-molecule drugs, or RNA molecules themselves can serve as drugs by interacting with proteins or other RNA molecules. While several RNA drugs have been granted clinical approval, numerous RNA-based therapeutics are presently undergoing clinical investigation or testing for various conditions, including genetic disorders, viral infections, and diverse forms of cancer. These therapies offer several advantages, such as high specificity, enabling precise targeting of disease-related genes or proteins, cost-effectiveness, and a relatively straightforward manufacturing process. Nevertheless, successful translation of RNA therapies into widespread clinical use necessitates addressing challenges related to delivery, stability, and potential off-target effects. This chapter provides a comprehensive overview of the general concepts of various classes of RNA-based therapeutics, the mechanistic basis of their function, as well as recent applications of RNA therapeutic in clinics.
Collapse
Affiliation(s)
- Huyen Trang Ha Thi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea.
| | - Van Thai Than
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam; Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
| |
Collapse
|
35
|
Hoelzer D, Bassan R, Boissel N, Roddie C, Ribera JM, Jerkeman M. ESMO Clinical Practice Guideline interim update on the use of targeted therapy in acute lymphoblastic leukaemia. Ann Oncol 2024; 35:15-28. [PMID: 37832649 DOI: 10.1016/j.annonc.2023.09.3112] [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] [Received: 03/27/2023] [Revised: 09/14/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Affiliation(s)
- D Hoelzer
- ONKOLOGIKUM Frankfurt am Museumsufer, Frankfurt, Germany
| | - R Bassan
- Hematology Unit, Ospedale dell'Angelo e Ospedale SS, Giovanni e Paolo, Mestre-Venezia, Italy
| | - N Boissel
- Hematology Department, Saint-Louis Hospital, APHP, Institut de Recherche Saint-Louis, Université de Paris Cité, Paris, France
| | - C Roddie
- Research Department of Haematology, UCL Cancer Institute, London, UK
| | - J M Ribera
- Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Jose Carreras Research Institute, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - M Jerkeman
- Department of Oncology, Skåne University Hospital and Lund University, Lund, Sweden
| |
Collapse
|
36
|
Surowka M, Klein C. A pivotal decade for bispecific antibodies? MAbs 2024; 16:2321635. [PMID: 38465614 PMCID: PMC10936642 DOI: 10.1080/19420862.2024.2321635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/12/2024] Open
Abstract
Bispecific antibodies (bsAbs) are a class of antibodies that can mediate novel mechanisms of action compared to monospecific monoclonal antibodies (mAbs). Since the discovery of mAbs and their adoption as therapeutic agents in the 1980s and 1990s, the development of bsAbs has held substantial appeal. Nevertheless, only three bsAbs (catumaxomab, blinatumomab, emicizumab) were approved through the end of 2020. However, since then, 11 bsAbs received regulatory agency approvals, of which nine (amivantamab, tebentafusp, mosunetuzumab, cadonilimab, teclistamab, glofitamab, epcoritamab, talquetamab, elranatamab) were approved for the treatment of cancer and two (faricimab, ozoralizumab) in non-oncology indications. Notably, of the 13 currently approved bsAbs, two, emicizumab and faricimab, have achieved blockbuster status, showing the promise of this novel class of therapeutics. In the 2020s, the approval of additional bsAbs can be expected in hematological malignancies, solid tumors and non-oncology indications, establishing bsAbs as essential part of the therapeutic armamentarium.
Collapse
Affiliation(s)
- Marlena Surowka
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Roche Glycart AG, Schlieren, Switzerland
| | - Christian Klein
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Roche Glycart AG, Schlieren, Switzerland
| |
Collapse
|
37
|
Kuwahara A, Nazuka M, Kuroki Y, Ito K, Watanabe S, Kumagai I, Asano R. Functional integration of protein A binding ability to antibody fragments for convenient and tag-free purification. Bioengineered 2023; 14:2259093. [PMID: 37732741 PMCID: PMC10515673 DOI: 10.1080/21655979.2023.2259093] [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: 10/11/2022] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
Although the development of small therapeutic antibodies is important, the affinity tags used for their purification often result in heterogeneous production and immunogenicity. In this study, we integrated Staphylococcus aureus protein A (SpA) binding ability into antibody fragments for convenient and tag-free purification. SpA affinity chromatography is used as a global standard purification method for conventional antibodies owing to its high binding affinity to the Fc region. SpA also has a binding affinity for some variable heavy domains (VH) classified in the VH3 subfamily. Through mutagenesis based on alignment and structural modeling results using the SpA-VH3 cocrystal structure, we integrated the SpA-binding ability into the anti-CD3 single-chain Fv. Furthermore, we applied this mutagenesis approach to more complicated small bispecific antibodies and successfully purified the antibodies using SpA affinity chromatography. The antibodies retained their biological function after purification. Integration of SpA-binding ability into conventional antibody fragments simplifies the purification and monitoring of the production processes and, thus, is an ideal strategy for accelerating the development of small therapeutic antibodies. Furthermore, because of its immunoactivity, the anti-CD3 variable region with SpA-binding ability is an effective building block for developing engineered cancer therapeutic antibodies without the Fc region.
Collapse
Affiliation(s)
- Atsushi Kuwahara
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Misae Nazuka
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Yuri Kuroki
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kohei Ito
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | | | - Izumi Kumagai
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ryutaro Asano
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
38
|
Awuah D, Li L, Williams L, Urak R, Kujawski M, Forman SJ, Shively JE, Wang X. Ex-vivo CS1-OKT3 dual specific bivalent antibody-armed effector T cells mediate cellular immunity against multiple myeloma. Sci Rep 2023; 13:20853. [PMID: 38012196 PMCID: PMC10682018 DOI: 10.1038/s41598-023-47115-7] [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: 07/26/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023] Open
Abstract
Bispecific T cell engaging antibodies (bsAbs) have emerged as novel and powerful therapeutic agents for redirecting T cells towards antigen-specific tumor killing. The cell surface glycoprotein and SLAM family member, CS1, exhibits stable and high-level expression on malignant plasma cells including multiple myeloma, which is indicative of an ideal target for bsAb therapy. Here, we developed a CS1 bsAb (CS1-dbBiTE) using Click chemistry to conjugate intact anti-CS1 antibody (Elotuzumab) and anti-huOKT3 antibody at their respective hinge regions. Using a cellular therapy approach, human T cells were armed ex-vivo with CS1-dbBiTE prior to examining effector activity. Our data indicates that arming T cells with CS1-dbBiTE induced T cell activation and expansion and subsequent cytotoxic activity against CS1-bearing MM tumors, demonstrated by significant CD107a expression as well as inflammatory cytokine secretion. As expected, CS1-dbBiTE armed T cells showed significantly reduced effector activity in the absence of CS1 expression. Similarly, in MM mouse xenograft studies, armed T cells exhibited effective anti-tumor efficacy highlighted by reduced tumor burden in MM.1S tumor-bearing mice compared to controls. On the basis of these findings, the rationale for CS1 targeting by human T cells armed with CS1-dbBiTE presents a potentially effective therapeutic approach for targeting MM.
Collapse
Affiliation(s)
- Dennis Awuah
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, USA
| | - Lin Li
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Lindsay Williams
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Ryan Urak
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, USA
| | - Maciej Kujawski
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Stephen J Forman
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, USA
| | - John E Shively
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Xiuli Wang
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, USA.
- T Cell Therapeutics Research Laboratory, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
39
|
Ramapriyan R, Sun J, Curry A, Richardson LG, Ramesh T, Gaffey MA, Gedeon PC, Gerstner ER, Curry WT, Choi BD. The Role of Antibody-Based Therapies in Neuro-Oncology. Antibodies (Basel) 2023; 12:74. [PMID: 37987252 PMCID: PMC10660525 DOI: 10.3390/antib12040074] [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: 08/30/2023] [Revised: 10/16/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
This review explores the evolving landscape of antibody-based therapies in neuro-oncology, in particular, immune checkpoint inhibitors and immunomodulatory antibodies. We discuss their mechanisms of action, blood-brain barrier (BBB) penetration, and experience in neuro-oncological conditions. Evidence from recent trials indicates that while these therapies can modulate the tumor immune microenvironment, their clinical benefits remain uncertain, largely due to challenges with BBB penetration and tumor-derived immunosuppression. This review also examines emerging targets such as TIGIT and LAG3, the potential of antibodies in modulating the myeloid compartment, and tumor-specific targets for monoclonal antibody therapy. We further delve into advanced strategies such as antibody-drug conjugates and bispecific T cell engagers. Lastly, we explore innovative techniques being investigated to enhance antibody delivery, including CAR T cell therapy. Despite current limitations, these therapies hold significant therapeutic potential for neuro-oncology. Future research should focus on optimizing antibody delivery to the CNS, identifying novel biological targets, and discovering combination therapies to address the hostile tumor microenvironment.
Collapse
Affiliation(s)
- Rishab Ramapriyan
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
- Harvard Medical School, Boston, MA 02115, USA (E.R.G.)
| | - Jing Sun
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
| | - Annabel Curry
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
| | - Leland G. Richardson
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
| | - Tarun Ramesh
- Harvard Medical School, Boston, MA 02115, USA (E.R.G.)
| | - Matthew A. Gaffey
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
| | - Patrick C. Gedeon
- Harvard Medical School, Boston, MA 02115, USA (E.R.G.)
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Elizabeth R. Gerstner
- Harvard Medical School, Boston, MA 02115, USA (E.R.G.)
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - William T. Curry
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
- Harvard Medical School, Boston, MA 02115, USA (E.R.G.)
| | - Bryan D. Choi
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA (A.C.); (L.G.R.); (W.T.C.)
- Harvard Medical School, Boston, MA 02115, USA (E.R.G.)
| |
Collapse
|
40
|
Walseng E, Wang B, Yang C, Patel P, Zhao C, Zhang H, Zhao P, Mazor Y. Conformation-selective rather than avidity-based binding to tumor associated antigen derived peptide-MHC enables targeting of WT1-pMHC low expressing cancer cells by anti-WT1-pMHC/CD3 T cell engagers. Front Immunol 2023; 14:1275304. [PMID: 38022650 PMCID: PMC10667733 DOI: 10.3389/fimmu.2023.1275304] [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: 08/09/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
T cell engagers, a category of T cell-retargeting immunotherapy, are rapidly transforming clinical cancer care. However, the lack of tumor-specific targets poses a significant roadblock for broad adaptation of this therapeutic modality in many indications, often resulting in systemic on-target off-tumor toxicity. Though various tumor-derived intracellular mutations provide a massive pool of potential tumor-specific antigens, targeting them is extremely challenging, partly due to the low copy number of tumor associated antigen (TAA)-derived pMHC on tumor cell surface. Further, the interplay of binding geometry and format valency in relation to the capacity of a T cell engager to efficiently target low density cell-surface pMHC is not well understood. Using the Wilms' tumor 1 (WT1) oncoprotein as a proof-of-principle TAA, combined with an array of IgG-like T cell engager modalities that differ in their anti-TAA valency and binding geometry, we show that the ability to induce an immunological synapse formation, resulting in potent killing of WT1 positive cancer cell lines is primarily dependent on the distinct geometrical conformations between the Fab arms of anti-WT1-HLA-A*02:01 and anti-CD3. The augmented avidity conferred by the binding of two anti-WT1-HLA-A*02:01 Fab arms has only minimal influence on cell killing potency. These findings demonstrate the need for careful examination of key design parameters for the development of next-generation T cell engagers targeting low density TAA-pMHCs on tumor cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yariv Mazor
- Biologics Engineering, Biopharmaceutical R&D, AstraZeneca, Gaithersburg, MD, United States
| |
Collapse
|
41
|
Mancusi A, Zorutti F, Ruggeri L, Bonato S, Tricarico S, Zei T, Iacucci Ostini R, Viglione V, Sembenico R, Sciabolacci S, Cardinali V, Martelli MF, Mecucci C, Carotti A, Martelli MP, Velardi A, Pierini A. Blinatumomab Redirects Donor Lymphocytes against CD19 + Acute Lymphoblastic Leukemia without Relevant Bystander Alloreactivity after Haploidentical Hematopoietic Stem Cell Transplantation. Int J Mol Sci 2023; 24:16105. [PMID: 38003295 PMCID: PMC10671266 DOI: 10.3390/ijms242216105] [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: 08/16/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Blinatumomab alone or with donor leukocyte infusions (DLI) has been used after allogeneic hematopoietic stem cell transplantation (HSCT) as a salvage therapy in relapsing patients with CD19+ hematological malignancies. It was effective in a fraction of them, with low incidence of Graft-versus-Host Disease (GvHD). Immunosuppressive drugs used as GvHD prophylaxis hinder T cell function and reduce the efficacy of the treatment. Because T cell-depleted haploidentical HSCT with donor regulatory and conventional T cells (Treg/Tcon haploidentical HSCT) does not require post-transplant immunosuppression, it is an ideal platform for the concomitant use of blinatumomab and DLI. However, the risk of GvHD is high because the donor is haploidentical. We treated two patients with CD19+ acute lymphoblastic leukemia (ALL) who had relapsed after Treg/Tcon haploidentical HSCT with blinatumomab and DLI. Despite the mismatch for one HLA haplotype, they did not develop GvHD and achieved complete remission with negative minimal residual disease. Consistently, we found that blinatumomab did not enhance T cell alloreactivity in vitro. Eventually, the two patients relapsed again because of their high disease risk. This study suggests that treatment with blinatumomab and DLI can be feasible to treat relapse after haploidentical transplantation, and its pre-emptive use should be considered to improve efficacy.
Collapse
Affiliation(s)
- Antonella Mancusi
- Division of Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy; (A.M.)
| | - Francesco Zorutti
- Division of Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy; (A.M.)
| | - Loredana Ruggeri
- Division of Hematology and Clinical Immunology, Azienda Ospedaliera S. Maria Della Misericordia, 06132 Perugia, Italy
| | - Samanta Bonato
- Division of Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy; (A.M.)
| | - Sara Tricarico
- Division of Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy; (A.M.)
| | - Tiziana Zei
- Division of Hematology and Clinical Immunology, Azienda Ospedaliera S. Maria Della Misericordia, 06132 Perugia, Italy
| | - Roberta Iacucci Ostini
- Division of Hematology and Clinical Immunology, Azienda Ospedaliera S. Maria Della Misericordia, 06132 Perugia, Italy
| | - Valerio Viglione
- Division of Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy; (A.M.)
| | - Rebecca Sembenico
- Division of Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy; (A.M.)
| | - Sofia Sciabolacci
- Division of Hematology and Clinical Immunology, Azienda Ospedaliera S. Maria Della Misericordia, 06132 Perugia, Italy
| | - Valeria Cardinali
- Division of Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy; (A.M.)
| | - Massimo Fabrizio Martelli
- Division of Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy; (A.M.)
| | - Cristina Mecucci
- Division of Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy; (A.M.)
| | - Alessandra Carotti
- Division of Hematology and Clinical Immunology, Azienda Ospedaliera S. Maria Della Misericordia, 06132 Perugia, Italy
| | - Maria Paola Martelli
- Division of Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy; (A.M.)
| | - Andrea Velardi
- Division of Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy; (A.M.)
| | - Antonio Pierini
- Division of Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy; (A.M.)
| |
Collapse
|
42
|
Arendt AM, Heubach F, Maier CP, Giardino S, Jung G, Kowalewski E, Rabsteyn A, Amorelli G, Seitz C, Schlegel P, Handgretinger R, Lang P. Targeting GD2 after allogeneic SCT: effector cell composition defines the optimal use of ch14.18 and the bispecific antibody construct NG-CU (GD2-CD3). Cancer Immunol Immunother 2023; 72:3813-3824. [PMID: 37742286 PMCID: PMC10576705 DOI: 10.1007/s00262-023-03536-x] [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: 05/11/2023] [Accepted: 08/24/2023] [Indexed: 09/26/2023]
Abstract
We investigated whether T cell-recruiting bispecific anti-CD3/GD2 antibody NG-CU might be an alternative to therapeutic anti-GD2 monoclonal antibody (mAb) ch14.18, mediating complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC) through natural killer (NK) cells for immunotherapy in high-risk/relapsed neuroblastoma after autologous/allogeneic stem cell transplantation (auto/alloSCT). Different antibody concentrations and effector-to-target ratios (E:T) were evaluated using xCELLigence RTCA system, peripheral blood mononuclear cells (PBMCs) (healthy donors and patients after alloSCT), and neuroblastoma cell lines (LS/LAN-1). Mean specific lysis of LS cells utilizing PBMCs from healthy donors and ch14.18 (1 µg/ml) was 40/66/75% after 12/24/48 h compared to 66/93/100% in the presence of NG-CU (100 ng/ml). NG-CU showed enhanced cytotoxicity compared to ch14.18, even at lower concentrations and E:T ratios, and completely eradicated LS cells after 72 h. To decipher the influence of effector cell subsets on lysis, different ratios of T and NK cells were tested. At a ratio of 1:1, ch14.18 was more effective than NG-CU. Using patient PBMCs taken at different time points posttransplant, significant lysis with both constructs was detectable depending on percentages and total numbers of T and NK cells; in the early posttransplant phase, NK cells were predominant and ch14.18 was superior, whereas later on, T cells represented the majority of immune cells and NG-CU was more effective. Our study highlights the importance of analyzing effector cell subsets in patients before initiating antibody-based therapy. Consequently, we propose an adjusted administration of both antibody constructs, considering the state of posttransplant immune recovery, to optimize anti-tumor activity.
Collapse
Affiliation(s)
- A-M Arendt
- Department of General Pediatrics, Oncology/Hematology, Children's University Hospital Tuebingen, Hoppe-Seyler-Str. 1, 72076, Tübingen, Germany.
| | - F Heubach
- Department of General Pediatrics, Oncology/Hematology, Children's University Hospital Tuebingen, Hoppe-Seyler-Str. 1, 72076, Tübingen, Germany
| | - C P Maier
- Department of General Pediatrics, Oncology/Hematology, Children's University Hospital Tuebingen, Hoppe-Seyler-Str. 1, 72076, Tübingen, Germany
- Department of Hematology/Oncology, Center for Internal Medicine, University Hospital, Tübingen, Germany
| | - S Giardino
- Hematopoietic Stem Cell Transplantation Unit, Department of Hematology and Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - G Jung
- Interfaculty Institute for Cell Biology, Department of Immunology, Eberhard Karls University Tuebingen, Tübingen, Germany
| | - E Kowalewski
- Department of General Pediatrics, Oncology/Hematology, Children's University Hospital Tuebingen, Hoppe-Seyler-Str. 1, 72076, Tübingen, Germany
| | - A Rabsteyn
- Department of General Pediatrics, Oncology/Hematology, Children's University Hospital Tuebingen, Hoppe-Seyler-Str. 1, 72076, Tübingen, Germany
| | - G Amorelli
- Department of General Pediatrics, Oncology/Hematology, Children's University Hospital Tuebingen, Hoppe-Seyler-Str. 1, 72076, Tübingen, Germany
| | - C Seitz
- Department of General Pediatrics, Oncology/Hematology, Children's University Hospital Tuebingen, Hoppe-Seyler-Str. 1, 72076, Tübingen, Germany
| | - P Schlegel
- Department of General Pediatrics, Oncology/Hematology, Children's University Hospital Tuebingen, Hoppe-Seyler-Str. 1, 72076, Tübingen, Germany
| | - R Handgretinger
- Department of General Pediatrics, Oncology/Hematology, Children's University Hospital Tuebingen, Hoppe-Seyler-Str. 1, 72076, Tübingen, Germany
| | - P Lang
- Department of General Pediatrics, Oncology/Hematology, Children's University Hospital Tuebingen, Hoppe-Seyler-Str. 1, 72076, Tübingen, Germany
| |
Collapse
|
43
|
Lutz S, Klausz K, Albici AM, Ebinger L, Sellmer L, Teipel H, Frenzel A, Langner A, Winterberg D, Krohn S, Hust M, Schirrmann T, Dübel S, Scherließ R, Humpe A, Gramatzki M, Kellner C, Peipp M. Novel NKG2D-directed bispecific antibodies enhance antibody-mediated killing of malignant B cells by NK cells and T cells. Front Immunol 2023; 14:1227572. [PMID: 37965326 PMCID: PMC10641740 DOI: 10.3389/fimmu.2023.1227572] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/05/2023] [Indexed: 11/16/2023] Open
Abstract
The activating receptor natural killer group 2, member D (NKG2D) represents an attractive target for immunotherapy as it exerts a crucial role in cancer immunosurveillance by regulating the activity of cytotoxic lymphocytes. In this study, a panel of novel NKG2D-specific single-chain fragments variable (scFv) were isolated from naïve human antibody gene libraries and fused to the fragment antigen binding (Fab) of rituximab to obtain [CD20×NKG2D] bibodies with the aim to recruit cytotoxic lymphocytes to lymphoma cells. All bispecific antibodies bound both antigens simultaneously. Two bibody constructs, [CD20×NKG2D#3] and [CD20×NKG2D#32], efficiently activated natural killer (NK) cells in co-cultures with CD20+ lymphoma cells. Both bibodies triggered NK cell-mediated lysis of lymphoma cells and especially enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) by CD38 or CD19 specific monoclonal antibodies suggesting a synergistic effect between NKG2D and FcγRIIIA signaling pathways in NK cell activation. The [CD20×NKG2D] bibodies were not effective in redirecting CD8+ T cells as single agents, but enhanced cytotoxicity when combined with a bispecific [CD19×CD3] T cell engager, indicating that NKG2D signaling also supports CD3-mediated T cell activation. In conclusion, engagement of NKG2D with bispecific antibodies is attractive to directly activate cytotoxic lymphocytes or to support their activation by monoclonal antibodies or bispecific T cell engagers. As a perspective, co-targeting of two tumor antigens may allow fine-tuning of antibody cancer therapies. Our proposed combinatorial approach is potentially applicable for many existing immunotherapies but further testing in different preclinical models is necessary to explore the full potential.
Collapse
Affiliation(s)
- Sebastian Lutz
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Anca-Maria Albici
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Lea Ebinger
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Lea Sellmer
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Hannah Teipel
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | | | - Anna Langner
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Dorothee Winterberg
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Steffen Krohn
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Michael Hust
- YUMAB GmbH, Braunschweig, Germany
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | | | - Stefan Dübel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Kiel, Germany
| | - Andreas Humpe
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Martin Gramatzki
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Christian Kellner
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| |
Collapse
|
44
|
Konrad CV, Iversen EF, Gunst JD, Monrad I, Holleufer A, Hartmann R, Østergaard LJ, Søgaard OS, Schleimann MH, Tolstrup M. Redirector of Vaccine-induced Effector Responses (RoVER) for specific killing of cellular targets. EBioMedicine 2023; 96:104785. [PMID: 37672868 PMCID: PMC10485592 DOI: 10.1016/j.ebiom.2023.104785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND In individuals with malignancy or HIV-1 infection, antigen-specific cytotoxic T lymphocytes (CTLs) often display an exhausted phenotype with impaired capacity to eliminate the disease. Existing cell-based immunotherapy strategies are often limited by the requirement for adoptive transfer of CTLs. We have developed an immunotherapy technology in which potent CTL responses are generated in vivo by vaccination and redirected to eliminate target cells using a bispecific Redirector of Vaccine-induced Effector Responses (RoVER). METHODS Following Yellow fever (YF) 17D vaccination of 51 healthy volunteers (NCT04083430), single-epitope YF-specific CTL responses were quantified by tetramer staining and multi-parameter flow cytometry. RoVER-mediated redirection of YF-specific CTLs to kill antigen-expressing Raji-Env cells, autologous CD19+ B cells or CD4+ T cells infected in vitro with a full-length HIV-1-eGFP was assessed in cell killing assays. Moreover, secreted IFN-γ, granzyme B, and TNF-α were analyzed by mesoscale multiplex assays. FINDINGS YF-17D vaccination induced strong epitope-specific CTL responses in the study participants. In cell killing assays, RoVER-mediated redirection of YF-specific CTLs to autologous CD19+ B cells or HIV-1-infected CD4+ cells resulted in 58% and 53% killing at effector to target ratio 1:1, respectively. INTERPRETATION We have developed an immunotherapy technology in which epitope-specific CTLs induced by vaccination can be redirected to kill antigen-expressing target cells by RoVER linking. The RoVER technology is highly specific and can be adapted to recognize various cell surface antigens. Importantly, this technology obviates the need for adoptive transfer of CTLs. FUNDING This work was funded by the Novo Nordisk Foundation (Hallas Møller NNF10OC0054577).
Collapse
Affiliation(s)
- Christina V Konrad
- Department of Clinical Medicine, Aarhus University, Aarhus C, 8000, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark
| | - Emma F Iversen
- Department of Clinical Medicine, Aarhus University, Aarhus C, 8000, Denmark
| | - Jesper D Gunst
- Department of Clinical Medicine, Aarhus University, Aarhus C, 8000, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark
| | - Ida Monrad
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark
| | - Andreas Holleufer
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, 8000, Denmark
| | - Rune Hartmann
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, 8000, Denmark
| | - Lars J Østergaard
- Department of Clinical Medicine, Aarhus University, Aarhus C, 8000, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark
| | - Ole S Søgaard
- Department of Clinical Medicine, Aarhus University, Aarhus C, 8000, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark
| | - Mariane H Schleimann
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark
| | - Martin Tolstrup
- Department of Clinical Medicine, Aarhus University, Aarhus C, 8000, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark.
| |
Collapse
|
45
|
Hill TF, Narvekar P, Asher G, Camp N, Thomas KR, Tasian SK, Rawlings DJ, James RG. Human plasma cells engineered to secrete bispecifics drive effective in vivo leukemia killing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554523. [PMID: 37662410 PMCID: PMC10473709 DOI: 10.1101/2023.08.24.554523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Bispecific antibodies are an important tool for the management and treatment of acute leukemias. Advances in genome-engineering have enabled the generation of human plasma cells that secrete therapeutic proteins and are capable of long-term in vivo engraftment in humanized mouse models. As a next step towards clinical translation of engineered plasma cells (ePCs) towards cancer therapy, here we describe approaches for the expression and secretion of bispecific antibodies by human plasma cells. We show that human ePCs expressing either fragment crystallizable domain deficient anti-CD19 × anti-CD3 (blinatumomab) or anti-CD33 × anti-CD3 bispecific antibodies mediate T cell activation and direct T cell killing of specific primary human cell subsets and B-acute lymphoblastic leukemia or acute myeloid leukemia cell lines in vitro. We demonstrate that knockout of the self-expressed antigen, CD19, boosts anti-CD19 bispecific secretion by ePCs and prevents self-targeting. Further, anti-CD19 bispecific-ePCs elicited tumor eradication in vivo following local delivery in flank-implanted Raji lymphoma cells. Finally, immunodeficient mice engrafted with anti-CD19 bispecific-ePCs and autologous T cells potently prevented in vivo growth of CD19+ acute lymphoblastic leukemia in patient-derived xenografts. Collectively, these findings support further development of ePCs for use as a durable, local delivery system for the treatment of acute leukemias, and potentially other cancers.
Collapse
Affiliation(s)
- Tyler F. Hill
- University of Washington, Medical Scientist Training Program, Seattle WA
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Parnal Narvekar
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Gregory Asher
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Nathan Camp
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Kerri R. Thomas
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Sarah K. Tasian
- Children’s Hospital of Philadelphia, Division of Oncology and Center for Childhood Cancer Research, Philadelphia PA
- Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia PA
| | - David J. Rawlings
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
- University of Washington, Departments of Pediatrics and Immunology, Seattle WA
| | - Richard G. James
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
- University of Washington, Departments of Pediatrics and Pharmacology, Seattle WA
| |
Collapse
|
46
|
Tang L, Huang Z, Mei H, Hu Y. Immunotherapy in hematologic malignancies: achievements, challenges and future prospects. Signal Transduct Target Ther 2023; 8:306. [PMID: 37591844 PMCID: PMC10435569 DOI: 10.1038/s41392-023-01521-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 08/19/2023] Open
Abstract
The immune-cell origin of hematologic malignancies provides a unique avenue for the understanding of both the mechanisms of immune responsiveness and immune escape, which has accelerated the progress of immunotherapy. Several categories of immunotherapies have been developed and are being further evaluated in clinical trials for the treatment of blood cancers, including stem cell transplantation, immune checkpoint inhibitors, antigen-targeted antibodies, antibody-drug conjugates, tumor vaccines, and adoptive cell therapies. These immunotherapies have shown the potential to induce long-term remission in refractory or relapsed patients and have led to a paradigm shift in cancer treatment with great clinical success. Different immunotherapeutic approaches have their advantages but also shortcomings that need to be addressed. To provide clinicians with timely information on these revolutionary therapeutic approaches, the comprehensive review provides historical perspectives on the applications and clinical considerations of the immunotherapy. Here, we first outline the recent advances that have been made in the understanding of the various categories of immunotherapies in the treatment of hematologic malignancies. We further discuss the specific mechanisms of action, summarize the clinical trials and outcomes of immunotherapies in hematologic malignancies, as well as the adverse effects and toxicity management and then provide novel insights into challenges and future directions.
Collapse
Affiliation(s)
- Lu Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China
| | - Zhongpei Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| |
Collapse
|
47
|
Tapia-Galisteo A, Álvarez-Vallina L, Sanz L. Bi- and trispecific immune cell engagers for immunotherapy of hematological malignancies. J Hematol Oncol 2023; 16:83. [PMID: 37501154 PMCID: PMC10373336 DOI: 10.1186/s13045-023-01482-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [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
|
48
|
Liu C, Zhou J, Kudlacek S, Qi T, Dunlap T, Cao Y. Population dynamics of immunological synapse formation induced by bispecific T cell engagers predict clinical pharmacodynamics and treatment resistance. eLife 2023; 12:e83659. [PMID: 37490053 PMCID: PMC10368424 DOI: 10.7554/elife.83659] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 06/01/2023] [Indexed: 07/26/2023] Open
Abstract
Effector T cells need to form immunological synapses (IS) with recognized target cells to elicit cytolytic effects. Facilitating IS formation is the principal pharmacological action of most T cell-based cancer immunotherapies. However, the dynamics of IS formation at the cell population level, the primary driver of the pharmacodynamics of many cancer immunotherapies, remains poorly defined. Using classic immunotherapy CD3/CD19 bispecific T cell engager (BiTE) as our model system, we integrate experimental and theoretical approaches to investigate the population dynamics of IS formation and their relevance to clinical pharmacodynamics and treatment resistance. Our models produce experimentally consistent predictions when defining IS formation as a series of spatiotemporally coordinated events driven by molecular and cellular interactions. The models predict tumor-killing pharmacodynamics in patients and reveal trajectories of tumor evolution across anatomical sites under BiTE immunotherapy. Our models highlight the bone marrow as a potential sanctuary site permitting tumor evolution and antigen escape. The models also suggest that optimal dosing regimens are a function of tumor growth, CD19 expression, and patient T cell abundance, which confer adequate tumor control with reduced disease evolution. This work has implications for developing more effective T cell-based cancer immunotherapies.
Collapse
Affiliation(s)
- Can Liu
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Jiawei Zhou
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Stephan Kudlacek
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Timothy Qi
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Tyler Dunlap
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, United States
| |
Collapse
|
49
|
Abdulhaq H, Hwang A, Mahmood O. Targeted Treatment of Adults with Relapsed or Refractory Diffuse Large B-Cell Lymphoma (DLBCL): Tafasitamab in Context. Onco Targets Ther 2023; 16:617-629. [PMID: 37492075 PMCID: PMC10364833 DOI: 10.2147/ott.s372783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/06/2023] [Indexed: 07/27/2023] Open
Abstract
The outcomes of Relapsed/Refractory (R/R) Diffuse Large B-cell lymphoma have been historically poor. The recent development of several novel therapies including CD19 directed agents has improved the prognosis of this disease significantly. Chimeric antigen receptor (CAR) T-cell therapy has drastically changed the treatment of R/R DLBCL, but it is still associated with significant barriers and limited access. Tafasitamab (an anti-CD19 engineered monoclonal antibody), in addition to lenalidomide, has shown significant efficacy with exceptionally durable responses in patients with R/R DLBCL who are ineligible for autologous stem cell transplantation (ASCT). Tafasitamab-lenalidomide and certain other therapies (ie, antibody-drug conjugates and bispecific antibodies) are important treatment options for patients who are ineligible for CAR-T due to co-morbidities or lack of access, and patients with rapid progression of disease who are unable to wait for manufacturing of CAR-T. This review will thus discuss currently approved and recently studied targeted treatment options for patients with R/R DLBCL with an emphasis on CAR-T alternative options, particularly Tafasitamab-lenalidomide.
Collapse
Affiliation(s)
- Haifaa Abdulhaq
- Division of Hematology/Oncology, University of California San Francisco, Fresno, CA, USA
| | - Andrew Hwang
- Division of Hematology/Oncology, University of California San Francisco, Fresno, CA, USA
| | - Omar Mahmood
- Division of Hematology/Oncology, University of California San Francisco, Fresno, CA, USA
| |
Collapse
|
50
|
van de Donk NWCJ, Zweegman S. T-cell-engaging bispecific antibodies in cancer. Lancet 2023; 402:142-158. [PMID: 37271153 DOI: 10.1016/s0140-6736(23)00521-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/06/2023] [Accepted: 03/02/2023] [Indexed: 06/06/2023]
Abstract
T-cell-engaging bispecific antibodies (BsAbs) simultaneously bind to antigens on tumour cells and CD3 subunits on T cells. This simultaneous binding results in the recruitment of T cells to the tumour, followed by T-cell activation and degranulation, and tumour cell elimination. T-cell-engaging BsAbs have shown substantial activity in several haematological malignancies by targeting CD19 in acute lymphoblastic leukaemia, CD20 in B-cell non-Hodgkin lymphoma, and BCMA and GPRC5D in multiple myeloma. Progress with solid tumours has been slower, in part due to the paucity of therapeutic targets with a tumour-specific expression profile, which is needed to limit on-target off-tumour side-effects. Nevertheless, BsAb-mediated recognition of a peptide fragment of gp100 presented by HLA-A2:01 molecules has shown marked activity in patients with unresectable or metastatic uveal melanoma. Cytokine release syndrome is the most frequent toxicity associated with BsAb treatment and is caused by activated T cells secreting proinflammatory cytokines. Understanding of resistance mechanisms has resulted in the development of new T cell-redirecting formats and novel combination strategies, which are expected to further improve depth and duration of response.
Collapse
Affiliation(s)
- Niels W C J van de Donk
- Amsterdam University Medical Centres, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands.
| | - Sonja Zweegman
- Amsterdam University Medical Centres, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|