1
|
Baskar S, Peng H, Gaglione EM, Carstens EJ, Lindorfer MA, Ahn IE, Herman SEM, Skarzynski M, Chang J, Keyvanfar K, Butera V, Blackburn A, Vire B, Maric I, Stetler-Stevenson M, Yuan CM, Eckhaus MA, Soto S, Farooqui MZH, Taylor RP, Rader C, Wiestner A. Potentiating CD20 monoclonal antibody therapy by targeting complement C3 fragments covalently deposited on lymphoma cells. Blood 2025; 145:1309-1320. [PMID: 39774793 DOI: 10.1182/blood.2024024846] [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: 05/17/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
ABSTRACT Monoclonal antibodies (mAbs) improve survival of patients with mature B-cell malignancies. Fcγ receptor-dependent effector mechanisms kill tumor cells but can promote antigen loss through trogocytosis, contributing to treatment failures. Cell-bound mAbs trigger the complement cascade to deposit C3 activation fragments and lyse cells. Within 24 hours after ofatumumab administration to patients with chronic lymphocytic leukemia (CLL), circulating tumor cells had lost CD20 and were opsonized with C3d, the terminal covalently bound form of complement protein C3. We hypothesized that C3d provides a target to eliminate residual CD20- tumor cells. To test this hypothesis, we generated C8xi, a mouse/human chimeric immunoglobulin G1 (IgG1) that reacts with human but not mouse C3d. C8xi was effective in a patient-derived xenograft model against CD20-, C3d opsonized CLL cells from patients treated with ofatumumab. We also generated rabbit mAbs, 2 of which were chosen because they bound mouse and human C3d with low nanomolar affinity but were minimally cross-reactive with full-length C3. Anti-C3d rabbit/human chimeric IgG1 in combination with ofatumumab or rituximab prolonged survival of xenografted mice that model 3 different types of non-Hodgkin lymphoma (NHL). For example, in a diffuse large B-cell lymphoma model (SU-DHL-6), median survival with single-agent CD20 mAb was 114 days but was not reached for mAb combination treatment (P = .008). In another NHL model (SU-DHL-4), single-agent and combination mAb therapy eradicated lymphoma in most mice. In long-term survivors from both cohorts, there was no evidence of adverse effects. We propose that C3d mAbs combined with complement-fixing CD20 mAbs can overcome antigen-loss escape and increase efficacy of mAb-based therapy.
Collapse
MESH Headings
- Humans
- Animals
- Mice
- Antigens, CD20/immunology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Xenograft Model Antitumor Assays
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/pharmacology
- Complement C3d/immunology
- Complement C3d/antagonists & inhibitors
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Rabbits
- Female
- Rituximab
Collapse
Affiliation(s)
- Sivasubramanian Baskar
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Haiyong Peng
- Department of Immunology and Microbiology, The Herbert Wertheim University of Florida Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL
| | - Erika M Gaglione
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA
| | - Elizabeth J Carstens
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Margaret A Lindorfer
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA
| | - Inhye E Ahn
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Sarah E M Herman
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Martin Skarzynski
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Jing Chang
- Department of Immunology and Microbiology, The Herbert Wertheim University of Florida Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL
| | - Keyvan Keyvanfar
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Vicent Butera
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Amy Blackburn
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Bérengère Vire
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Irina Maric
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Maryalice Stetler-Stevenson
- Flow Cytometry Unit, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Constance M Yuan
- Flow Cytometry Unit, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Michael A Eckhaus
- Division of Veterinary Resources, Office of Research Services, National Institutes of Health, Bethesda, MD
| | - Susan Soto
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Mohammed Z H Farooqui
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA
| | - Christoph Rader
- Department of Immunology and Microbiology, The Herbert Wertheim University of Florida Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL
| | - Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
2
|
Ghasemi N, Holder KA, Ings DP, Grant MD. Enhancement of Human Immunodeficiency Virus-Specific CD8 + T Cell Responses with TIGIT Blockade Involves Trogocytosis. Pathogens 2024; 13:1137. [PMID: 39770396 PMCID: PMC11679564 DOI: 10.3390/pathogens13121137] [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: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Natural killer (NK) and CD8+ T cell function is compromised in human immunodeficiency virus type 1 (HIV-1) infection by increased expression of inhibitory receptors such as TIGIT (T cell immunoreceptor with Ig and ITIM domains). Blocking inhibitory receptors or their ligands with monoclonal antibodies (mAb) has potential to improve antiviral immunity in general and facilitate HIV eradication strategies. We assessed the impact of TIGIT engagement and blockade on cytotoxicity, degranulation, and interferon-gamma (IFN-γ) production by CD8+ T cells from persons living with HIV (PLWH). The effect of TIGIT engagement on non-specific anti-CD3-redirected cytotoxicity was assessed in redirected cytotoxicity assays, and the effect of TIGIT blockade on HIV-specific CD8+ T cell responses was assessed by flow cytometry. In 14/19 cases where peripheral blood mononuclear cells (PBMC) mediated >10% redirected cytotoxicity, TIGIT engagement reduced the level of cytotoxicity to <90% of control values. We selected PLWH with >1000 HIV Gag or Nef-specific IFN-γ spot forming cells per million PBMC to quantify the effects of TIGIT blockade on HIV-specific CD8+ T cell responses by flow cytometry. Cell surface TIGIT expression decreased on CD8+ T cells from 23/40 PLWH following TIGIT blockade and this loss was associated with increased anti-TIGIT mAb fluorescence on monocytes. In total, 6 of these 23 PLWH had enhanced HIV-specific CD8+ T cell degranulation and IFN-γ production with TIGIT blockade, compared to 0/17 with no decrease in cell surface TIGIT expression. Reduced CD8+ T cell TIGIT expression with TIGIT blockade involved trogocytosis by circulating monocytes, suggesting that an effector monocyte population and intact fragment crystallizable (Fc) functions are required for mAb-based TIGIT blockade to effectively enhance HIV-specific CD8+ T cell responses.
Collapse
Affiliation(s)
- Nazanin Ghasemi
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (N.G.); (K.A.H.); (D.P.I.)
| | - Kayla A. Holder
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (N.G.); (K.A.H.); (D.P.I.)
- Department of Biomedical Informatics, University of Colorado School of Medicine, Denver, CO 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Denver, CO 80045, USA
| | - Danielle P. Ings
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (N.G.); (K.A.H.); (D.P.I.)
| | - Michael D. Grant
- Immunology and Infectious Diseases Program, Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (N.G.); (K.A.H.); (D.P.I.)
| |
Collapse
|
3
|
Siconolfi G, Vitali F, Sciarrone MA, Ardito M, Guglielmino V, Romano A, Granata G, Silvestri G, Luigetti M. IgM Flare in Anti-MAG Neuropathy Post Rituximab Treatment: A Clinical Case and a Systematic Review of the Literature. Brain Sci 2024; 14:1294. [PMID: 39766493 PMCID: PMC11674938 DOI: 10.3390/brainsci14121294] [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: 12/03/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Anti-MAG polyneuropathy is a demyelinating peripheral neuropathy associated with IgM monoclonal gammopathies, particularly MGUS (monoclonal gammopathy of undetermined significance) and Waldenström macroglobulinemia. It is characterized by a subacute onset of distal sensory symptoms, with distal motor dysfunction typically appearing only in the later stages of the disease. The condition is caused by the presence of autoantibodies directed against myelin-associated glycoprotein, a structural protein of myelin. This leads to abnormalities in electrophysiological studies, such as markedly delayed distal latencies without conduction blocks or temporal dispersion of potentials. While rituximab (RTX) is the primary treatment, its efficacy is limited, with improvement seen in only 30-50% of patients. Recently, acute worsening of symptoms after RTX treatment has been increasingly reported. METHODS This systematic review compiles case reports and series from inception to June 2024 published on Scopus, PubMed or Cochrane, documenting acute exacerbations after RTX treatment in patients with anti-MAG polyneuropathy. Additionally, we present a case report from our institution that highlights this phenomenon. RESULTS We identified 13 clinical cases of acute deterioration in patients with anti-MAG polyneuropathy. Among these, eight patients (62%) achieved full recovery following additional treatment, while five patients (38%) did not return to their previous level of function. Plasmapheresis led to complete recovery in all four patients who received this intervention. Interestingly, many patients also experienced recovery after discontinuation of rituximab (RTX) treatment without the need for further therapeutic intervention. CONCLUSIONS Acute clinical deterioration following RTX treatment in anti-MAG polyneuropathy is a possible occurrence. However, to date, no studies have assessed the true prevalence of this phenomenon. Further research is warranted to identify potential predictors of worsening following RTX treatment in this patient population.
Collapse
Affiliation(s)
- Giovanni Siconolfi
- Dipartimento Di Neuroscienze, Università Cattolica del Sacro Cuore, Sede Di Roma, 00168 Rome, Italy; (G.S.); (F.V.); (M.A.S.); (M.A.); (V.G.); (A.R.); (G.S.)
| | - Francesca Vitali
- Dipartimento Di Neuroscienze, Università Cattolica del Sacro Cuore, Sede Di Roma, 00168 Rome, Italy; (G.S.); (F.V.); (M.A.S.); (M.A.); (V.G.); (A.R.); (G.S.)
| | - Maria Ausilia Sciarrone
- Dipartimento Di Neuroscienze, Università Cattolica del Sacro Cuore, Sede Di Roma, 00168 Rome, Italy; (G.S.); (F.V.); (M.A.S.); (M.A.); (V.G.); (A.R.); (G.S.)
| | - Michelangelo Ardito
- Dipartimento Di Neuroscienze, Università Cattolica del Sacro Cuore, Sede Di Roma, 00168 Rome, Italy; (G.S.); (F.V.); (M.A.S.); (M.A.); (V.G.); (A.R.); (G.S.)
| | - Valeria Guglielmino
- Dipartimento Di Neuroscienze, Università Cattolica del Sacro Cuore, Sede Di Roma, 00168 Rome, Italy; (G.S.); (F.V.); (M.A.S.); (M.A.); (V.G.); (A.R.); (G.S.)
| | - Angela Romano
- Dipartimento Di Neuroscienze, Università Cattolica del Sacro Cuore, Sede Di Roma, 00168 Rome, Italy; (G.S.); (F.V.); (M.A.S.); (M.A.); (V.G.); (A.R.); (G.S.)
| | - Giuseppe Granata
- Fondazione Policlinico Universitario A. Gemelli IRCCS, UOC Neurofisiopatologia, 00168 Rome, Italy;
| | - Gabriella Silvestri
- Dipartimento Di Neuroscienze, Università Cattolica del Sacro Cuore, Sede Di Roma, 00168 Rome, Italy; (G.S.); (F.V.); (M.A.S.); (M.A.); (V.G.); (A.R.); (G.S.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, UOC Neurologia, 00168 Rome, Italy
| | - Marco Luigetti
- Dipartimento Di Neuroscienze, Università Cattolica del Sacro Cuore, Sede Di Roma, 00168 Rome, Italy; (G.S.); (F.V.); (M.A.S.); (M.A.); (V.G.); (A.R.); (G.S.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, UOC Neurologia, 00168 Rome, Italy
| |
Collapse
|
4
|
Cornell CE, Chorlay A, Krishnamurthy D, Martin NR, Baldauf L, Fletcher DA. Target cell tension regulates macrophage trogocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626490. [PMID: 39677802 PMCID: PMC11642796 DOI: 10.1101/2024.12.02.626490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Macrophages are known to engulf small membrane fragments, or trogocytose, target cells and pathogens, rather than fully phagocytose them. However, little is known about what causes macrophages to choose trogocytosis versus phagocytosis. Here, we report that cortical tension of target cells is a key regulator of macrophage trogocytosis. At low tension, macrophages will preferentially trogocytose antibody-opsonized cells, while at high tension they tend towards phagocytosis. Using model vesicles, we demonstrate that macrophages will rapidly switch from trogocytosis to phagocytosis when membrane tension is increased. Stiffening the cortex of target cells also biases macrophages to phagocytose them, a trend that can be countered by increasing antibody surface density and is captured in a mechanical model of trogocytosis. This work suggests that a distinct molecular pathway for trogocytosis is not required to explain differences in trogocytosis among target cell types and points to a mechanism for target cells to modulate trogocytosis.
Collapse
|
5
|
Jabbarzadeh Kaboli P, Roozitalab G, Farghadani R, Eskandarian Z, Zerrouqi A. c-MET and the immunological landscape of cancer: novel therapeutic strategies for enhanced anti-tumor immunity. Front Immunol 2024; 15:1498391. [PMID: 39664377 PMCID: PMC11632105 DOI: 10.3389/fimmu.2024.1498391] [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: 10/06/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024] Open
Abstract
Cellular mesenchymal-epithelial transition factor (c-MET), also known as hepatocyte growth factor receptor (HGFR), is a crucial receptor tyrosine kinase implicated in various solid tumors, including lung, breast, and liver cancers. The concomitant expression of c-MET and PD-L1 in tumors, such as hepatocellular carcinoma, highlights their prognostic significance and connection to therapeutic resistance. Cancer-associated fibroblasts and mesenchymal stromal cells produce hepatocyte growth factor (HGF), activating c-MET signaling in tumor cells and myeloid-derived suppressor cells (MDSC). This activation leads to metabolic reprogramming and increased activity of enzymes like glutaminase (GLS), indoleamine 2,3-dioxygenase (IDO), and arginase 1 (ARG1), depleting essential amino acids in the tumor microenvironment that are vital for effector immune cell function. This review highlights the interplay between tumor cells and myeloid-derived suppressor cells (MDSCs) that create an immunosuppressive environment while providing targets for c-MET-focused immunotherapy. It emphasizes the clinical implications of c-MET inhibition on the behavior of immune cells such as neutrophils, macrophages, T cells, and NK cells. It explores the potential of c-MET antagonism combined with immunotherapeutic strategies to enhance cancer treatment paradigms. This review also discusses the innovative cancer immunotherapies targeting c-MET, including chimeric antigen receptor (CAR) therapies, monoclonal antibodies, and antibody-drug conjugates, while encouraging the development of a comprehensive strategy that simultaneously tackles immune evasion and enhances anti-tumor efficacy further to improve the clinical prognoses for patients with c-MET-positive malignancies. Despite the challenges and variability in efficacy across different cancer subtypes, continued research into the molecular mechanisms and the development of innovative therapeutic strategies will be crucial.
Collapse
Affiliation(s)
| | - Ghazaal Roozitalab
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Zoya Eskandarian
- Research Institute Children’s Cancer Center, and Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Abdessamad Zerrouqi
- Department of Biochemistry, Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
6
|
Karczmarczyk A, Chojnacki M, Paziewska M, Karp M, Skórka K, Zaleska J, Purkot J, Własiuk P, Giannopoulos K. HLA-G can be transfered via trogocytosis from leukemic cells to T cells in chronic lymphocytic leukemia. Hum Immunol 2024; 85:111178. [PMID: 39541623 DOI: 10.1016/j.humimm.2024.111178] [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: 07/22/2024] [Revised: 10/21/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
In chronic lymphocytic leukemia (CLL) immune escape mechanism allows leukemia cells to proliferate and expand and it might also be responsible for disease progression. Some molecules involved in the regulation of an immune system might represent prognostic value for CLL patients. Among numerous immune escape mechanisms it was shown that the expression of human leukocyte antigen G (HLA-G) might represent one of the agents damaging cellular immune response. In the present study, the expression of the HLA-G molecule and ILT-2 receptor on the surface of leukemic cells, as well as a plasma concentration of soluble HLA-G (sHLA-G) was evaluated. Also, we investigated whether HLA-G could be transferred from leukemic cells to T cells by the mechanism of trogocytosis. We showed higher proportion of leukemic cells expressing HLA-G and increased levels of sHLA-G in CLL patients compared to that of B-cells in healthy volunteers (HVs). Results of our work showed a time-dependent increase in HLA-G expression on CD4+ T cells co-incubated with HLA-G-positive CD19+ cells. Longer coincubation times did significantly increase these proportions (p < 0.001). We have shown that a higher proportion of HLA-G-expressing CD4+ T cells correlated with the clinical stage of the disease according to the Rai classification. Interestingly, we found a higher CD4+HLA-G+ percentage in the group with unmutated immunoglobulin heavy chain variable region (IGHV) genes compared to the group with mutated IGHV gene after 48 h co-culture. In summary, increasing evidence has revealed that, in addition to HLA-G expressed on tumor cells, intercellular transfer of HLA-G among cancer cells and immune cells through trogocytosis plays important roles in mechanism of immune escape, disease progression and poor clinical outcome.
Collapse
MESH Headings
- Humans
- HLA-G Antigens/genetics
- HLA-G Antigens/immunology
- HLA-G Antigens/metabolism
- HLA-G Antigens/blood
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Male
- Middle Aged
- Aged
- Female
- Tumor Escape
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Coculture Techniques
- Antigens, CD19/metabolism
- Antigens, CD19/immunology
- Adult
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Leukocyte Immunoglobulin-like Receptor B1/metabolism
- Aged, 80 and over
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/genetics
- Antigens, CD
Collapse
Affiliation(s)
| | - Michał Chojnacki
- Department of Medical Biology, Institute of Rural Health, Lublin, Poland
| | - Magdalena Paziewska
- Department of Experimental Hematooncology, Medical University of Lublin, Lublin, Poland
| | - Marta Karp
- Department of Experimental Hematooncology, Medical University of Lublin, Lublin, Poland
| | - Katarzyna Skórka
- Department of Experimental Hematooncology, Medical University of Lublin, Lublin, Poland
| | - Joanna Zaleska
- Department of Experimental Hematooncology, Medical University of Lublin, Lublin, Poland
| | - Joanna Purkot
- Department of Experimental Hematooncology, Medical University of Lublin, Lublin, Poland
| | - Paulina Własiuk
- Department of Experimental Hematooncology, Medical University of Lublin, Lublin, Poland
| | - Krzysztof Giannopoulos
- Department of Experimental Hematooncology, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
7
|
Toledo-Stuardo K, Ribeiro CH, González-Herrera F, Matthies DJ, Le Roy MS, Dietz-Vargas C, Latorre Y, Campos I, Guerra Y, Tello S, Vásquez-Sáez V, Novoa P, Fehring N, González M, Rodríguez-Siza J, Vásquez G, Méndez P, Altamirano C, Molina MC. Therapeutic antibodies in oncology: an immunopharmacological overview. Cancer Immunol Immunother 2024; 73:242. [PMID: 39358613 PMCID: PMC11448508 DOI: 10.1007/s00262-024-03814-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/16/2024] [Indexed: 10/04/2024]
Abstract
The biotechnological development of monoclonal antibodies and their immunotherapeutic use in oncology have grown exponentially in the last decade, becoming the first-line therapy for some types of cancer. Their mechanism of action is based on the ability to regulate the immune system or by interacting with targets that are either overexpressed in tumor cells, released into the extracellular milieu or involved in processes that favor tumor growth. In addition, the intrinsic characteristics of each subclass of antibodies provide specific effector functions against the tumor by activating antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, and antibody-dependent cellular phagocytosis, among other mechanisms. The rational design and engineering of monoclonal antibodies have improved their pharmacokinetic and pharmacodynamic features, thus optimizing the therapeutic regimens administered to cancer patients and improving their clinical outcomes. The selection of the immunoglobulin G subclass, modifications to its crystallizable region (Fc), and conjugation of radioactive substances or antineoplastic drugs may all improve the antitumor effects of therapeutic antibodies. This review aims to provide insights into the immunological and pharmacological aspects of therapeutic antibodies used in oncology, with a rational approach at molecular modifications that can be introduced into these biological tools, improving their efficacy in the treatment of cancer.
Collapse
Affiliation(s)
- Karen Toledo-Stuardo
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Carolina H Ribeiro
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Fabiola González-Herrera
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Douglas J Matthies
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - María Soledad Le Roy
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Claudio Dietz-Vargas
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Yesenia Latorre
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Ivo Campos
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Yuneisy Guerra
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Samantha Tello
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Valeria Vásquez-Sáez
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Pedro Novoa
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Nicolás Fehring
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Mauricio González
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Jose Rodríguez-Siza
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Gonzalo Vásquez
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Pamela Méndez
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Claudia Altamirano
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
- Centro Regional de Estudio en Alimentos Saludables, Valparaíso, Chile
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Santiago, Chile
| | - María Carmen Molina
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile.
| |
Collapse
|
8
|
Choudhary R, Gupta V, Khandpur S. Updates on the Management of Autoimmune Bullous Diseases. Indian Dermatol Online J 2024; 15:758-769. [PMID: 39359305 PMCID: PMC11444445 DOI: 10.4103/idoj.idoj_740_23] [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: 09/25/2023] [Revised: 01/09/2024] [Accepted: 02/11/2024] [Indexed: 10/04/2024] Open
Abstract
Background Autoimmune bullous diseases are associated with high morbidity and mortality. Traditionally, systemic corticosteroids and conventional immunosuppressive agents have been the mainstay of treatment, but their broad immunosuppressive effects and long-term complications have prompted the exploration of newer more targeted therapies. Materials and Methods This review explores the evolving landscape of therapeutic options for immunobullous diseases, with a particular focus on pemphigus, bullous pemphigoid (BP), and mucous membrane pemphigoid, by searching PubMed, clinicaltrials.gov, and Cochrane databases for published literature from 2014 to 2023. Results/Discussion We discuss emerging treatments for pemphigus such as B cell modulatory drugs, anti-inflammatory drugs, those inhibiting autoantibody half-life or blister-inducing activity, and stem cell therapy, while offering insights into the level of evidence, potential benefits, and limitations of each approach. The role of biologics and novel therapies like rituximab, omalizumab, and dupilumab in reshaping the management of BP is also discussed. Conclusion The article highlights the need for further research, clinical trials, and comparative studies to determine the most effective and safest treatment options for patients with immunobullous diseases.
Collapse
Affiliation(s)
- Rajat Choudhary
- Department of Dermatology and Venereology, All India Institute of Medical Sciences, New Delhi, India
| | - Vishal Gupta
- Department of Dermatology and Venereology, All India Institute of Medical Sciences, New Delhi, India
| | - Sujay Khandpur
- Department of Dermatology and Venereology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
9
|
Solórzano JL, Menéndez V, Parra E, Solis L, Salazar R, García-Cosío M, Climent F, Fernández S, Díaz E, Francisco-Cruz A, Khoury J, Jiang M, Tamegnon A, Montalbán C, Melero I, Wistuba I, De Andrea C, F. García J. Multiplex spatial analysis reveals increased CD137 expression and m-MDSC neighboring tumor cells in refractory classical Hodgkin Lymphoma. Oncoimmunology 2024; 13:2388304. [PMID: 39135889 PMCID: PMC11318683 DOI: 10.1080/2162402x.2024.2388304] [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: 05/14/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
The Hodgkin and Reed - Sternberg (HRS) cells in classical Hodgkin Lymphoma (cHL) actively modify the immune tumor microenvironment (TME) attracting immunosuppressive cells and expressing inhibitory molecules. A high frequency of myeloid cells in the TME is correlated with an unfavorable prognosis, but more specific and rare cell populations lack precise markers. Myeloid-derived suppressor cells (MDSCs) have been identified in the peripheral blood of cHL patients, where they appear to be correlated with disease aggressiveness. TNFRSF9 (CD137) is a T cell co-stimulator expressed by monocytic and dendritic cells. Its expression has also been described in HRS cells, where it is thought to play a role in reducing antitumor responses. Here, we perform qualitative and quantitative analyses of lymphocytic and MDSC subtypes and determine the CD137 cell distribution in cHL primary tumors using multiplex immunofluorescence and automated multispectral imaging. The results were correlated with patients' clinical features. Cells were stained with specific panels of immune checkpoint markers (PD-1, PD-L1, CD137), tumor-infiltrating T lymphocytes (CD3, PD-1), and monocytic cells/MDSCs (CD68, CD14, CD33, Arg-1, CD11b). This approach allowed us to identify distinct phenotypes and to analyze spatial interactions between immune subpopulations and tumor cells. The results confirm CD137 expression by T, monocytic and HRS cells. In addition, the expression of CD137, T exhausted cells, and monocytic MDSCs (m-MDSCs) in the vicinity of malignant HRS cells were associated with a worse prognosis. Our findings reveal new elements of the TME that mediate immune escape, and confirm CD137 as a candidate target for immunotherapy in cHL.
Collapse
Affiliation(s)
- José L. Solórzano
- Pathology and Molecular Department, MD Anderson Cancer Center, Madrid, Spain
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| | - Victoria Menéndez
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| | - Edwin Parra
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | - Luisa Solis
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | | | | | - Fina Climent
- Pathology Department, Hospital Universitari de Bellvitge, IDIBELL, L’Hospitalet De Llobregat, Barcelona, Spain
| | - Sara Fernández
- Pathology and Molecular Department, MD Anderson Cancer Center, Madrid, Spain
| | - Eva Díaz
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| | | | - Joseph Khoury
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mei Jiang
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | - Auriole Tamegnon
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | - Carlos Montalbán
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| | - Ignacio Melero
- University of Navarra and Instituto de Investigación Sanitaria de Navarra, Pamplona, Navarra, Spain
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | - Carlos De Andrea
- University of Navarra and Instituto de Investigación Sanitaria de Navarra, Pamplona, Navarra, Spain
| | - Juan F. García
- Pathology and Molecular Department, MD Anderson Cancer Center, Madrid, Spain
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| |
Collapse
|
10
|
Jo S, Fischer BR, Cronin NM, Nurmalasari NPD, Loyd YM, Kerkvliet JG, Bailey EM, Anderson RB, Scott BL, Hoppe AD. Antibody surface mobility amplifies FcγR signaling via Arp2/3 during phagocytosis. Biophys J 2024; 123:2312-2327. [PMID: 38321740 PMCID: PMC11331046 DOI: 10.1016/j.bpj.2024.01.036] [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: 06/30/2023] [Revised: 11/07/2023] [Accepted: 01/29/2024] [Indexed: 02/08/2024] Open
Abstract
We report herein that the anti-CD20 therapeutic antibody, rituximab, is rearranged into microclusters within the phagocytic synapse by macrophage Fcγ receptors (FcγR) during antibody-dependent cellular phagocytosis. These microclusters were observed to potently recruit Syk and to undergo rearrangements that were limited by the cytoskeleton of the target cell, with depolymerization of target-cell actin filaments leading to modest increases in phagocytic efficiency. Total internal reflection fluorescence analysis revealed that FcγR total phosphorylation, Syk phosphorylation, and Syk recruitment were enhanced when IgG-FcγR microclustering was enabled on fluid bilayers relative to immobile bilayers in a process that required Arp2/3. We conclude that on fluid surfaces, IgG-FcγR microclustering promotes signaling through Syk that is amplified by Arp2/3-driven actin rearrangements. Thus, the surface mobility of antigens bound by IgG shapes the signaling of FcγR with an unrecognized complexity beyond the zipper and trigger models of phagocytosis.
Collapse
Affiliation(s)
- Seongwan Jo
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota; BioSNTRii, South Dakota State University, Brookings, South Dakota
| | - Brady R Fischer
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota; BioSNTRii, South Dakota State University, Brookings, South Dakota
| | - Nicholas M Cronin
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota; BioSNTRii, South Dakota State University, Brookings, South Dakota
| | - Ni Putu Dewi Nurmalasari
- Department of Nanoscience & Biomedical Engineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; BioSNTRii, South Dakota School of Mines and Technology, Rapid City, South Dakota
| | - Yoseph M Loyd
- Department of Nanoscience & Biomedical Engineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; BioSNTRii, South Dakota School of Mines and Technology, Rapid City, South Dakota
| | - Jason G Kerkvliet
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota; BioSNTRii, South Dakota State University, Brookings, South Dakota
| | - Elizabeth M Bailey
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota; BioSNTRii, South Dakota State University, Brookings, South Dakota
| | - Robert B Anderson
- Department of Nanoscience & Biomedical Engineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; BioSNTRii, South Dakota School of Mines and Technology, Rapid City, South Dakota
| | - Brandon L Scott
- Department of Nanoscience & Biomedical Engineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; BioSNTRii, South Dakota School of Mines and Technology, Rapid City, South Dakota
| | - Adam D Hoppe
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota; BioSNTRii, South Dakota State University, Brookings, South Dakota.
| |
Collapse
|
11
|
Jestrabek H, Kohlhas V, Hallek M, Nguyen PH. Impact of leukemia-associated macrophages on the progression and therapy response of chronic lymphocytic leukemia. Leuk Res 2024; 143:107531. [PMID: 38851084 DOI: 10.1016/j.leukres.2024.107531] [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: 04/17/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
The treatment landscape of chronic lymphocytic leukemia (CLL) has advanced remarkably over the past decade. The advent and approval of the BTK inhibitor ibrutinib and BCL-2 inhibitor venetoclax, as well as monoclonal anti-CD20 antibodies rituximab and obinutuzumab, have resulted in deep remissions and substantially improved survival outcomes for patients. However, CLL remains a complex disease with many patients still experiencing relapse and unsatisfactory treatment responses. CLL cells are highly dependent on their pro-leukemic tumor microenvironment (TME), which comprises different cellular and soluble factors. A large body of evidence suggests that CLL-associated macrophages shaped by leukemic cells play a pivotal role in maintaining CLL cell survival. In this review, we summarize the pro-survival interactions between CLL cells and macrophages, as well as the impact of the current first-line treatment agents, including ibrutinib, venetoclax, and CD20 antibodies on leukemia-associated macrophages.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
- Tumor-Associated Macrophages/immunology
- Tumor-Associated Macrophages/drug effects
- Disease Progression
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Adenine/analogs & derivatives
- Sulfonamides/therapeutic use
- Piperidines/therapeutic use
- Macrophages/pathology
- Macrophages/immunology
Collapse
Affiliation(s)
- Hendrik Jestrabek
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, Cologne 50931, Germany; Mildred Scheel School of Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital of Cologne, Cologne 50931, Germany
| | - Viktoria Kohlhas
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, Cologne 50931, Germany
| | - Michael Hallek
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, Cologne 50931, Germany; Mildred Scheel School of Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital of Cologne, Cologne 50931, Germany
| | - Phuong-Hien Nguyen
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, Cologne 50931, Germany.
| |
Collapse
|
12
|
Taylor RP, Lindorfer MA. Antibody-drug conjugate adverse effects can be understood and addressed based on immune complex clearance mechanisms. Blood 2024; 144:137-144. [PMID: 38643493 DOI: 10.1182/blood.2024024442] [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: 03/01/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/23/2024] Open
Abstract
ABSTRACT Numerous antibody-drug conjugates (ADCs) are being developed for cancer immunotherapy. Although several of these agents have demonstrated considerable clinical efficacy and have won Food and Drug Administration (FDA) approval, in many instances, they have been characterized by adverse side effects (ASEs), which can be quite severe in a fraction of treated patients. The key hypothesis in this perspective is that many of the most serious ASEs associated with the use of ADCs in the treatment of cancer can be most readily explained and understood due to the inappropriate processing of these ADCs via pathways normally followed for immune complex clearance, which include phagocytosis and trogocytosis. We review the key published basic science experiments and clinical observations that support this idea. We propose that it is the interaction of the ADC with Fcγ receptors expressed on off-target cells and tissues that can most readily explain ADC-mediated pathologies, which therefore provides a rationale for the design of protocols to minimize ASEs. We describe measurements that should help identify those patients most likely to experience ASE due to ADC, and we propose readily available treatments as well as therapies under development for other indications that should substantially reduce ASE associated with ADC. Our focus will be on the following FDA-approved ADC for which there are substantial literatures: gemtuzumab ozogamicin and inotuzumab ozogamicin; and trastuzumab emtansine and trastuzumab deruxtecan.
Collapse
Affiliation(s)
- Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA
| | - Margaret A Lindorfer
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA
| |
Collapse
|
13
|
Jabbarzadeh Kaboli P, Chen HF, Babaeizad A, Roustai Geraylow K, Yamaguchi H, Hung MC. Unlocking c-MET: A comprehensive journey into targeted therapies for breast cancer. Cancer Lett 2024; 588:216780. [PMID: 38462033 DOI: 10.1016/j.canlet.2024.216780] [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: 12/20/2023] [Revised: 02/18/2024] [Accepted: 02/29/2024] [Indexed: 03/12/2024]
Abstract
Breast cancer is the most common malignancy among women, posing a formidable health challenge worldwide. In this complex landscape, the c-MET (cellular-mesenchymal epithelial transition factor) receptor tyrosine kinase (RTK), also recognized as the hepatocyte growth factor (HGF) receptor (HGFR), emerges as a prominent protagonist, displaying overexpression in nearly 50% of breast cancer cases. Activation of c-MET by its ligand, HGF, secreted by neighboring mesenchymal cells, contributes to a cascade of tumorigenic processes, including cell proliferation, metastasis, angiogenesis, and immunosuppression. While c-MET inhibitors such as crizotinib, capmatinib, tepotinib and cabozantinib have garnered FDA approval for non-small cell lung cancer (NSCLC), their potential within breast cancer therapy is still undetermined. This comprehensive review embarks on a journey through structural biology, multifaceted functions, and intricate signaling pathways orchestrated by c-MET across cancer types. Furthermore, we highlight the pivotal role of c-MET-targeted therapies in breast cancer, offering a clinical perspective on this promising avenue of intervention. In this pursuit, we strive to unravel the potential of c-MET as a beacon of hope in the fight against breast cancer, unveiling new horizons for therapeutic innovation.
Collapse
Affiliation(s)
- Parham Jabbarzadeh Kaboli
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan
| | - Hsiao-Fan Chen
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan
| | - Ali Babaeizad
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Hirohito Yamaguchi
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan; Department of Biotechnology, Asia University, Taichung, 413, Taiwan.
| |
Collapse
|
14
|
Albanese M, Chen HR, Gapp M, Muenchhoff M, Yang HH, Peterhoff D, Hoffmann K, Xiao Q, Ruhle A, Ambiel I, Schneider S, Mejías-Pérez E, Stern M, Wratil PR, Hofmann K, Amann L, Jocham L, Fuchs T, Ulivi AF, Besson-Girard S, Weidlich S, Schneider J, Spinner CD, Sutter K, Dittmer U, Humpe A, Baumeister P, Wieser A, Rothenfusser S, Bogner J, Roider J, Knolle P, Hengel H, Wagner R, Laketa V, Fackler OT, Keppler OT. Receptor transfer between immune cells by autoantibody-enhanced, CD32-driven trogocytosis is hijacked by HIV-1 to infect resting CD4 T cells. Cell Rep Med 2024; 5:101483. [PMID: 38579727 PMCID: PMC11031382 DOI: 10.1016/j.xcrm.2024.101483] [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: 07/21/2023] [Revised: 12/23/2023] [Accepted: 03/01/2024] [Indexed: 04/07/2024]
Abstract
Immune cell phenotyping frequently detects lineage-unrelated receptors. Here, we report that surface receptors can be transferred from primary macrophages to CD4 T cells and identify the Fcγ receptor CD32 as driver and cargo of this trogocytotic transfer. Filamentous CD32+ nanoprotrusions deposit distinct plasma membrane patches onto target T cells. Transferred receptors confer cell migration and adhesion properties, and macrophage-derived membrane patches render resting CD4 T cells susceptible to infection by serving as hotspots for HIV-1 binding. Antibodies that recognize T cell epitopes enhance CD32-mediated trogocytosis. Such autoreactive anti-HIV-1 envelope antibodies can be found in the blood of HIV-1 patients and, consistently, the percentage of CD32+ CD4 T cells is increased in their blood. This CD32-mediated, antigen-independent cell communication mode transiently expands the receptor repertoire and functionality of immune cells. HIV-1 hijacks this mechanism by triggering the generation of trogocytosis-promoting autoantibodies to gain access to immune cells critical to its persistence.
Collapse
Affiliation(s)
- Manuel Albanese
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany; Department for Clinical Sciences and Community Health (DISCCO), University of Milan, Milan, Italy
| | - Hong-Ru Chen
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany.
| | - Madeleine Gapp
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Maximilian Muenchhoff
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany; German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Hsiu-Hui Yang
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - David Peterhoff
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Katja Hoffmann
- Institute of Virology, University Medical Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Qianhao Xiao
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Adrian Ruhle
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Ina Ambiel
- Department of Infectious Diseases, Heidelberg University, Medical Faculty Heidelberg, Integrative Virology, Center for Integrative Infectious Disease Research (CIID), Heidelberg, Germany; German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Stephanie Schneider
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Ernesto Mejías-Pérez
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Marcel Stern
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Paul R Wratil
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Katharina Hofmann
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Laura Amann
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Linda Jocham
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Thimo Fuchs
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | | | - Simon Besson-Girard
- Institute for Stroke and Dementia Research, University Hospital, LMU München, Munich, Germany
| | - Simon Weidlich
- Technical University of Munich, School of Medicine, University Hospital Rechts der Isar, Department of Internal Medicine II, Munich, Germany
| | - Jochen Schneider
- Technical University of Munich, School of Medicine, University Hospital Rechts der Isar, Department of Internal Medicine II, Munich, Germany
| | - Christoph D Spinner
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany; Technical University of Munich, School of Medicine, University Hospital Rechts der Isar, Department of Internal Medicine II, Munich, Germany
| | - Kathrin Sutter
- University Hospital Essen, University Duisburg-Essen, Institute for Virology and Institute for Translational HIV Research, Essen, Germany
| | - Ulf Dittmer
- University Hospital Essen, University Duisburg-Essen, Institute for Virology and Institute for Translational HIV Research, Essen, Germany
| | - Andreas Humpe
- Department of Transfusion Medicine, Cell Therapeutics, and Hemostaseology, Department of Anesthesiology, University Hospital Munich, Munich, Germany
| | - Philipp Baumeister
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, LMU München, Munich, Germany
| | - Andreas Wieser
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany; Max von Pettenkofer Institute, Medical Microbiology and Hospital Epidemiology, Faculty of Medicine, LMU München, Munich, Germany; Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU München, Munich, Germany
| | - Simon Rothenfusser
- Division of Clinical Pharmacology, University Hospital, LMU München and Unit Clinical Pharmacology (EKliP), Helmholtz Center for Environmental Health, Munich, Germany
| | - Johannes Bogner
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany; Division of Infectious Diseases, University Hospital, Medizinische Klinik und Poliklinik IV, LMU München, Munich, Germany
| | - Julia Roider
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany; Division of Infectious Diseases, University Hospital, Medizinische Klinik und Poliklinik IV, LMU München, Munich, Germany
| | - Percy Knolle
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany; Institute of Molecular Immunology and Experimental Oncology, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Hartmut Hengel
- Institute of Virology, University Medical Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Vibor Laketa
- German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany; Department of Infectious Diseases, Heidelberg University, Medical Faculty Heidelberg, Virology, Center for Integrative Infectious Disease Research (CIID), Heidelberg, Germany
| | - Oliver T Fackler
- Department of Infectious Diseases, Heidelberg University, Medical Faculty Heidelberg, Integrative Virology, Center for Integrative Infectious Disease Research (CIID), Heidelberg, Germany; German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany.
| | - Oliver T Keppler
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany; German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany.
| |
Collapse
|
15
|
Dabkowska A, Domka K, Firczuk M. Advancements in cancer immunotherapies targeting CD20: from pioneering monoclonal antibodies to chimeric antigen receptor-modified T cells. Front Immunol 2024; 15:1363102. [PMID: 38638442 PMCID: PMC11024268 DOI: 10.3389/fimmu.2024.1363102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/25/2024] [Indexed: 04/20/2024] Open
Abstract
CD20 located predominantly on the B cells plays a crucial role in their development, differentiation, and activation, and serves as a key therapeutic target for the treatment of B-cell malignancies. The breakthrough of monoclonal antibodies directed against CD20, notably exemplified by rituximab, revolutionized the prognosis of B-cell malignancies. Rituximab, approved across various hematological malignancies, marked a paradigm shift in cancer treatment. In the current landscape, immunotherapies targeting CD20 continue to evolve rapidly. Beyond traditional mAbs, advancements include antibody-drug conjugates (ADCs), bispecific antibodies (BsAbs), and chimeric antigen receptor-modified (CAR) T cells. ADCs combine the precision of antibodies with the cytotoxic potential of drugs, presenting a promising avenue for enhanced therapeutic efficacy. BsAbs, particularly CD20xCD3 constructs, redirect cytotoxic T cells to eliminate cancer cells, thereby enhancing both precision and potency in their therapeutic action. CAR-T cells stand as a promising strategy for combatting hematological malignancies, representing one of the truly personalized therapeutic interventions. Many new therapies are currently being evaluated in clinical trials. This review serves as a comprehensive summary of CD20-targeted therapies, highlighting the progress and challenges that persist. Despite significant advancements, adverse events associated with these therapies and the development of resistance remain critical issues. Understanding and mitigating these challenges is paramount for the continued success of CD20-targeted immunotherapies.
Collapse
Affiliation(s)
- Agnieszka Dabkowska
- Laboratory of Immunology, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Domka
- Laboratory of Immunology, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Malgorzata Firczuk
- Laboratory of Immunology, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
16
|
Singhal S, Rao AS, Stadanlick J, Bruns K, Sullivan NT, Bermudez A, Honig-Frand A, Krouse R, Arambepola S, Guo E, Moon EK, Georgiou G, Valerius T, Albelda SM, Eruslanov EB. Human Tumor-Associated Macrophages and Neutrophils Regulate Antitumor Antibody Efficacy through Lethal and Sublethal Trogocytosis. Cancer Res 2024; 84:1029-1047. [PMID: 38270915 PMCID: PMC10982649 DOI: 10.1158/0008-5472.can-23-2135] [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: 07/17/2023] [Revised: 11/29/2023] [Accepted: 01/23/2024] [Indexed: 01/26/2024]
Abstract
The clinical benefits of tumor-targeting antibodies (tAb) are modest in solid human tumors. The efficacy of many tAbs is dependent on Fc receptor (FcR)-expressing leukocytes that bind Fc fragments of tAb. Tumor-associated macrophages (TAM) and neutrophils (TAN) represent the majority of FcR+ effectors in solid tumors. A better understanding of the mechanisms by which TAMs and TANs regulate tAb response could help improve the efficacy of cancer treatments. Here, we found that myeloid effectors interacting with tAb-opsonized lung cancer cells used antibody-dependent trogocytosis (ADT) but not antibody-dependent phagocytosis. During this process, myeloid cells "nibbled off" tumor cell fragments containing tAb/targeted antigen (tAg) complexes. ADT was only tumoricidal when the tumor cells expressed high levels of tAg and the effectors were present at high effector-to-tumor ratios. If either of these conditions were not met, which is typical for solid tumors, ADT was sublethal. Sublethal ADT, mainly mediated by CD32hiCD64hi TAM, led to two outcomes: (i) removal of surface tAg/tAb complexes from the tumor that facilitated tumor cell escape from the tumoricidal effects of tAb; and (ii) acquisition of bystander tAgs by TAM with subsequent cross-presentation and stimulation of tumor-specific T-cell responses. CD89hiCD32loCD64lo peripheral blood neutrophils (PBN) and TAN stimulated tumor cell growth in the presence of the IgG1 anti-EGFR Ab cetuximab; however, IgA anti-EGFR Abs triggered the tumoricidal activity of PBN and negated the stimulatory effect of TAN. Overall, this study provides insights into the mechanisms by which myeloid effectors mediate tumor cell killing or resistance during tAb therapy. SIGNIFICANCE The elucidation of the conditions and mechanisms by which human FcR+ myeloid effectors mediate cancer cell resistance and killing during antibody treatment could help develop improved strategies for treating solid tumors.
Collapse
Affiliation(s)
- Sunil Singhal
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Abhishek S. Rao
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jason Stadanlick
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kyle Bruns
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Neil T. Sullivan
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andres Bermudez
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Adam Honig-Frand
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ryan Krouse
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sachinthani Arambepola
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Emily Guo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edmund K. Moon
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas
| | - Thomas Valerius
- Department of Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Steven M. Albelda
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Evgeniy B. Eruslanov
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Cruz-Leal Y, Norris PAA, Gil Gonzalez L, Marjoram D, Wabnitz H, Shan Y, Lazarus AH. Trogocytosis drives red blood cell antigen loss in association with antibody-mediated immune suppression. Blood 2024; 143:807-821. [PMID: 37946269 DOI: 10.1182/blood.2023020860] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/27/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023] Open
Abstract
ABSTRACT Red blood cell (RBC) alloimmunization to paternal antigens during pregnancy can cause hemolytic disease of the fetus and newborn (HDFN). This severe and potentially fatal neonatal disorder can be prevented by the administration of polyclonal anti-D through a mechanism referred to as antibody-mediated immune suppression (AMIS). Although anti-D prophylaxis effectively prevents HDFN, a lack of mechanistic clarity has hampered its replacement with recombinant agents. The major theories behind AMIS induction in the hematologic literature have classically centered around RBC clearance; however, antigen modulation/loss has recently been proposed as a potential mechanism of AMIS. To explore the primary mechanisms of AMIS, we studied the ability of 11 different antibodies to induce AMIS, RBC clearance, antigen loss, and RBC membrane loss in the HOD (hen egg lysozyme-ovalbumin-human Duffy) murine model. Antibodies targeting different portions of the HOD molecule could induce AMIS independent of their ability to clear RBCs; however, all antibodies capable of inducing a strong AMIS effect also caused significant in vivo loss of the HOD antigen in conjunction with RBC membrane loss. In vitro studies of AMIS-inducing antibodies demonstrated simultaneous RBC antigen and membrane loss, which was mediated by macrophages. Confocal live-cell microscopy revealed that AMIS-inducing antibodies triggered RBC membrane transfer to macrophages, consistent with trogocytosis. Furthermore, anti-D itself can induce trogocytosis even at low concentrations, when phagocytosis is minimal or absent. In view of these findings, we propose trogocytosis as a mechanism of AMIS induction.
Collapse
Affiliation(s)
- Yoelys Cruz-Leal
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, ON, Canada
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Peter A A Norris
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, ON, Canada
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institute, Stockholm, Sweden
| | - Lazaro Gil Gonzalez
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Danielle Marjoram
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Hanna Wabnitz
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Yuexin Shan
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Alan H Lazarus
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, ON, Canada
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
18
|
Park S, Kim J, Shin JH. Intercellular Transfer of Immune Regulatory Molecules Via Trogocytosis. Results Probl Cell Differ 2024; 73:131-146. [PMID: 39242377 DOI: 10.1007/978-3-031-62036-2_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] [Indexed: 09/09/2024]
Abstract
Trogocytosis, an active cellular process involving the transfer of plasma membrane and attached cytosol during cell-to-cell contact, has been observed prominently in CD4 T cells interacting with antigen-presenting cells carrying antigen-loaded major histocompatibility complex (MHC) class II molecules. Despite the inherent absence of MHC class II molecules in CD4 T cells, they actively acquire these molecules from encountered antigen-presenting cells, leading to the formation of antigen-loaded MHC class II molecules-dressed CD4 T cells. Subsequently, these dressed CD4 T cells engage in antigen presentation to other CD4 T cells, revealing a dynamic mechanism of immune communication. The transferred membrane proteins through trogocytosis retain their surface localization, thereby altering cellular functions. Concurrently, the donor cells experience a loss of membrane proteins, resulting in functional changes due to the altered membrane properties. This chapter provides a focused exploration into trogocytosis-mediated transfer of immune regulatory molecules and its consequential impact on diverse immune responses.
Collapse
Affiliation(s)
- Soyeon Park
- The interdisciplinary graduate program in integrative biology, Yonsei University, Incheon, South Korea
| | - Jeonghyun Kim
- The interdisciplinary graduate program in integrative biology, Yonsei University, Incheon, South Korea
| | - Jae Hun Shin
- The interdisciplinary graduate program in integrative biology, Yonsei University, Incheon, South Korea.
- Integrative Science and Engineering Division, Underwood International College, Yonsei University, Incheon, South Korea.
| |
Collapse
|
19
|
Shen J, Zhao S, Peng M, Li Y, Zhang L, Li X, Hu Y, Wu M, Xiang S, Wu X, Liu J, Zhang B, Chen Z, Lin D, Liu H, Tang W, Chen J, Sun X, Liao Q, Hide G, Zhou Z, Lun ZR, Wu Z. Macrophage-mediated trogocytosis contributes to destroying human schistosomes in a non-susceptible rodent host, Microtus fortis. Cell Discov 2023; 9:101. [PMID: 37794085 PMCID: PMC10550985 DOI: 10.1038/s41421-023-00603-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 09/13/2023] [Indexed: 10/06/2023] Open
Abstract
Schistosoma parasites, causing schistosomiasis, exhibit typical host specificity in host preference. Many mammals, including humans, are susceptible to infection, while the widely distributed rodent, Microtus fortis, exhibits natural anti-schistosome characteristics. The mechanisms of host susceptibility remain poorly understood. Comparison of schistosome infection in M. fortis with the infection in laboratory mice (highly sensitive to infection) offers a good model system to investigate these mechanisms and to gain an insight into host specificity. In this study, we showed that large numbers of leukocytes attach to the surface of human schistosomes in M. fortis but not in mice. Single-cell RNA-sequencing analyses revealed that macrophages might be involved in the cell adhesion, and we further demonstrated that M. fortis macrophages could be mediated to attach and kill schistosomula with dependence on Complement component 3 (C3) and Complement receptor 3 (CR3). Importantly, we provided direct evidence that M. fortis macrophages could destroy schistosomula by trogocytosis, a previously undescribed mode for killing helminths. This process was regulated by Ca2+/NFAT signaling. These findings not only elucidate a novel anti-schistosome mechanism in M. fortis but also provide a better understanding of host parasite interactions, host specificity and the potential generation of novel strategies for schistosomiasis control.
Collapse
Affiliation(s)
- Jia Shen
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China.
| | - Siyu Zhao
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Mei Peng
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Yanguo Li
- Institute of Drug Discovery Technology, School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Lichao Zhang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Xiaoping Li
- Department of Hepatic Surgery and Liver Transplantation Center, Organ Transplantation Institute, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yunyi Hu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Mingrou Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Suoyu Xiang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Xiaoying Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiahua Liu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Beibei Zhang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Zebin Chen
- Department of Hepatic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Datao Lin
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Huanyao Liu
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenyan Tang
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Chen
- Department of Immunology, Center for Precision Medicine and Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xi Sun
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China
| | - Qi Liao
- Institute of Drug Discovery Technology, School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Geoff Hide
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Salford, UK
| | - Zhijun Zhou
- Department of Laboratory Animals, Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, Hunan, China.
| | - Zhao-Rong Lun
- Biomedical Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Salford, UK.
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Zhongdao Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, China.
| |
Collapse
|
20
|
Roeser A, Lazarus AH, Mahévas M. B cells and antibodies in refractory immune thrombocytopenia. Br J Haematol 2023; 203:43-53. [PMID: 37002711 DOI: 10.1111/bjh.18773] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/11/2023] [Indexed: 04/03/2023]
Abstract
Immune thrombocytopenia (ITP) is an acquired bleeding disorder mediated by pathogenic autoantibodies secreted by plasma cells (PCs) in many patients. In refractory ITP patients, the persistence of splenic and bone marrow autoreactive long-lived PCs (LLPCs) may explain primary failure of rituximab and splenectomy respectively. The reactivation of autoreactive memory B cells generating new autoreactive PCs contributes to relapses after initial response to rituximab. Emerging strategies targeting B cells and PCs aim to prevent the settlement of splenic LLPCs with the combination of anti-BAFF and rituximab, to deplete autoreactive PCs with anti-CD38 antibodies, and to induce deeper B-cell depletion in tissues with novel anti-CD20 monoclonal antibodies and anti-CD19 therapies. Alternative strategies, focused on controlling autoantibody mediated effects, have also been developed, including SYK and BTK inhibitors, complement inhibitors, FcRn blockers and inhibitors of platelet desialylation.
Collapse
Affiliation(s)
- Anaïs Roeser
- Institut Necker Enfants Malades (INEM), INSERM U1151/CNRS UMS 8253, ATIP-Avenir TeamAI2B, Paris, France
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Alan H Lazarus
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Departments of Medicine and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Innovation and Portfolio Management, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Matthieu Mahévas
- Institut Necker Enfants Malades (INEM), INSERM U1151/CNRS UMS 8253, ATIP-Avenir TeamAI2B, Paris, France
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| |
Collapse
|
21
|
Van Wagoner CM, Rivera-Escalera F, Delgadillo NJ, Chu CC, Zent CS, Elliott MR. Antibody-mediated phagocytosis in cancer immunotherapy. Immunol Rev 2023; 319:128-141. [PMID: 37602915 PMCID: PMC10615698 DOI: 10.1111/imr.13265] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023]
Abstract
Unconjugated monoclonal antibodies (mAb) have revolutionized the treatment of many types of cancer. Some of these mAbs promote the clearance of malignant cells via direct cytotoxic effects. More recently, antibody-dependent cellular phagocytosis (ADCP) has been appreciated as a major mechanism of action for a number of widely-used mAbs, including anti-CD20 (rituximab, obinutuzumab), anti-HER2 (trazituzumab), and anti-CD38 (daratumumab). However, as a monotherapy these ADCP-inducing mAbs produce insufficient levels of cytotoxicity in vivo and are not curative. As a result, these mAbs are most effectively used in combination therapies. The efficacy of these mAbs is further hampered by the apparent development of drug resistance by many patients. Here we will explore the role of ADCP in cancer immunotherapy and discuss the key factors that could limit the efficacy of ADCP-inducing mAbs in vivo. Finally, we will discuss current insights and approaches being applied to overcome these limitations.
Collapse
Affiliation(s)
- Carly M. Van Wagoner
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
| | - Fátima Rivera-Escalera
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
| | | | - Charles C. Chu
- Division of Hematology/Oncology, University of Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester, NY, USA
| | - Clive S. Zent
- Division of Hematology/Oncology, University of Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester, NY, USA
| | - Michael R. Elliott
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
22
|
Joo V, Petrovas C, de Leval L, Noto A, Obeid M, Fenwick C, Pantaleo G. A CD64/FcγRI-mediated mechanism hijacks PD-1 from PD-L1/2 interaction and enhances anti-PD-1 functional recovery of exhausted T cells. Front Immunol 2023; 14:1213375. [PMID: 37622123 PMCID: PMC10446174 DOI: 10.3389/fimmu.2023.1213375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAb) targeting the immune checkpoint inhibitor programmed cell death protein 1 (PD-1) have achieved considerable clinical success in anti-cancer therapy through relieving T cell exhaustion. Blockade of PD-1 interaction with its ligands PD-L1 and PD-L2 is an important determinant in promoting the functional recovery of exhausted T cells. Here, we show that anti-PD-1 mAbs act through an alternative mechanism leading to the downregulation of PD-1 surface expression on memory CD4+ and CD8+ T cells. PD-1 receptor downregulation is a distinct process from receptor endocytosis and occurs in a CD14+ monocyte dependent manner with the CD64/Fcγ receptor I acting as the primary factor for this T cell extrinsic process. Importantly, downregulation of surface PD-1 strongly enhances antigen-specific functional recovery of exhausted PD-1+CD8+ T cells. Our study demonstrates a novel mechanism for reducing cell surface levels of PD-1 and limiting the inhibitory targeting by PD-L1/2 and thereby enhancing the efficacy of anti-PD-1 Ab in restoring T cell functionality.
Collapse
Affiliation(s)
- Victor Joo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Constantinos Petrovas
- Institute of Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Laurence de Leval
- Institute of Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Alessandra Noto
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Michel Obeid
- Lausanne Center for Immuno-oncology Toxicities (LCIT), Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Craig Fenwick
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
23
|
Bhandari V, Bril V. FcRN receptor antagonists in the management of myasthenia gravis. Front Neurol 2023; 14:1229112. [PMID: 37602255 PMCID: PMC10439012 DOI: 10.3389/fneur.2023.1229112] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Myasthenia gravis (MG) is an autoimmune disorder characterized by autoantibodies specifically directed against proteins located within the postsynaptic membrane of the neuromuscular junction. These pathogenic autoantibodies can be reduced by therapies such as plasma exchange, IVIG infusions and other immunosuppressive agents. However, there are significant side effects associated with most of these therapies. Since there is a better understanding of the molecular structure and the biological properties of the neonatal Fc receptors (FcRn), it possesses an attractive profile in treating myasthenia gravis. FcRn receptors prevent the catabolism of IgG by impeding their lysosomal degradation and facilitating their extracellular release at physiological pH, consequently extending the IgG half-life. Thus, the catabolism of IgG can be enhanced by blocking the FcRn, leading to outcomes similar to those achieved through plasma exchange with no significant safety concerns. The available studies suggest that FcRn holds promise as a versatile therapeutic intervention, capable of delivering beneficial outcomes in patients with distinct characteristics and varying degrees of MG severity. Efgartigimod is already approved for the treatment of generalized MG, rozanolixizumab is under review by health authorities, and phase 3 trials of nipocalimab and batoclimab are underway. Here, we will review the available data on FcRn therapeutic agents in the management of MG.
Collapse
Affiliation(s)
| | - Vera Bril
- Ellen and Martin Prosserman Centre for Neuromuscular Diseases, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Xiang C, Li Y, Jing S, Han S, He H. Trichomonas gallinae Kills Host Cells Using Trogocytosis. Pathogens 2023; 12:1008. [PMID: 37623968 PMCID: PMC10459183 DOI: 10.3390/pathogens12081008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/18/2023] [Accepted: 07/18/2023] [Indexed: 08/26/2023] Open
Abstract
Trichomonas gallinae (T. gallinae) is an infectious parasite that is prevalent worldwide in poultry and can cause death in both poultry and wild birds. Although studies have shown that T. gallinae damages host cells through direct contact, the mechanism is still unclear. In this study, we found that T. gallinae can kill host cells by ingesting fragments of the host cells, that is, by trogocytosis. Moreover, we found that the PI3K inhibitor wortmannin and the cysteine protease inhibitor E-64D prevented T. gallinae from destroying host cells. To the best of our knowledge, our study has demonstrated for the first time that T. gallinae uses trogocytosis to kill host cells. Understanding this mechanism is crucial for the prevention and control of avian trichomoniasis and will contribute to the development of vaccines and drugs for the prevention and control of avian trichomoniasis.
Collapse
Affiliation(s)
- Chen Xiang
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (C.X.); (S.H.)
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Li
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071000, China; (Y.L.); (S.J.)
| | - Shengfan Jing
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071000, China; (Y.L.); (S.J.)
| | - Shuyi Han
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (C.X.); (S.H.)
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Hongxuan He
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (C.X.); (S.H.)
| |
Collapse
|
25
|
Nguyen TT, Nhu NT, Tran VK, Viet-Nhi NK, Ho XD, Jhan MK, Chen YP, Lin CF. Efficacy and safety of add-on anti-CD20 monoclonal antibody to Bruton tyrosine kinase inhibitor treatment for chronic lymphocytic leukemia: a meta-analysis. Sci Rep 2023; 13:9775. [PMID: 37328530 PMCID: PMC10276018 DOI: 10.1038/s41598-023-36279-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: 01/10/2023] [Accepted: 05/31/2023] [Indexed: 06/18/2023] Open
Abstract
The efficacy of Bruton tyrosine kinase inhibitors (BTKi) remains suboptimal in chronic lymphocytic leukemia (CLL) treatment. A systematic review and meta-analysis were conducted to compare the outcomes of combining anti-CD20 monoclonal antibodies (mAb) with BTKi therapy versus BTKi monotherapy for patients with CLL. We searched for relevant studies in the Pubmed, Medline, Embase, and Cochrane databases until December 2022. We estimated the effective results using a hazard ratio (HR) for survival outcomes and relative risk (RR) for response outcomes and safety. Four randomized controlled trials (including 1056 patients) were found until November 2022 and fulfilled the inclusion criteria. Progression-free survival was significantly improved with the addition of anti-CD20 mAb to BTKi over BTKi (HR 0.70, 95% confidence interval (CI) 0.51-0.97), whereas pooled analysis of overall survival did not favor combination therapy compared to BTKi monotherapy (HR 0.72, 95% CI 0.50-1.04). Combination therapy was related to a statistically better complete response (RR, 2.03; 95% CI 1.01 to 4.06) and an undetectable minimal residual disease rate (RR, 6.43; 95% CI 3.54 to 11.67). The risk of grade ≥ 3 adverse events was comparable between the two groups (RR, 1.08; (95% CI 0.80 to 1.45). Overall, adding anti-CD20 mAb to BTKi revealed superior efficacy than BTKi alone in untreated or previously treated CLL patients without affecting the safety of single-agent BTKi. Conducting further randomized studies to confirm our results and determine the optimal therapy for managing patients with CLL is essential.
Collapse
Affiliation(s)
- Thi Thuy Nguyen
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Department of Oncology, Hue University of Medicine and Pharmacy, Hue University, Hue, Vietnam
| | - Nguyen Thanh Nhu
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Faculty of Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Van Khoi Tran
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Department of Surgery, Hue University of Medicine and Pharmacy, Hue University, Hue, Vietnam
| | - Nguyen-Kieu Viet-Nhi
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Xuan Dung Ho
- Department of Oncology, Hue University of Medicine and Pharmacy, Hue University, Hue, Vietnam
| | - Ming-Kai Jhan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, No. 25, Wuxing St, Xinyi District, Taipei, 110, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Ya-Ping Chen
- Division of Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan
| | - Chiou-Feng Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, No. 25, Wuxing St, Xinyi District, Taipei, 110, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.
- Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei, 110, Taiwan.
| |
Collapse
|
26
|
Advances in antibody-based therapy in oncology. NATURE CANCER 2023; 4:165-180. [PMID: 36806801 DOI: 10.1038/s43018-023-00516-z] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 01/10/2023] [Indexed: 02/22/2023]
Abstract
Monoclonal antibodies are a growing class of targeted cancer therapeutics, characterized by exquisite specificity, long serum half-life, high affinity and immune effector functions. In this review, we outline key advances in the field with a particular focus on recent and emerging classes of engineered antibody therapeutic candidates, discuss molecular structure and mechanisms of action and provide updates on clinical development and practice.
Collapse
|
27
|
Behrens LM, van Egmond M, van den Berg TK. Neutrophils as immune effector cells in antibody therapy in cancer. Immunol Rev 2022; 314:280-301. [PMID: 36331258 DOI: 10.1111/imr.13159] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Tumor-targeting monoclonal antibodies are available for a number of cancer cell types (over)expressing the corresponding tumor antigens. Such antibodies can limit tumor progression by different mechanisms, including direct growth inhibition and immune-mediated mechanisms, in particular complement-dependent cytotoxicity, antibody-dependent cellular phagocytosis, and antibody-dependent cellular cytotoxicity (ADCC). ADCC can be mediated by various types of immune cells, including neutrophils, the most abundant leukocyte in circulation. Neutrophils express a number of Fc receptors, including Fcγ- and Fcα-receptors, and can therefore kill tumor cells opsonized with either IgG or IgA antibodies. In recent years, important insights have been obtained with respect to the mechanism(s) by which neutrophils engage and kill antibody-opsonized cancer cells and these findings are reviewed here. In addition, we consider a number of additional ways in which neutrophils may affect cancer progression, in particular by regulating adaptive anti-cancer immunity.
Collapse
Affiliation(s)
- Leonie M. Behrens
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Vrije Universiteit Amsterdam HV Amsterdam The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology HV Amsterdam The Netherlands
- Amsterdam institute for Infection and Immunity, Cancer Immunology HV Amsterdam The Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Vrije Universiteit Amsterdam HV Amsterdam The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology HV Amsterdam The Netherlands
- Amsterdam institute for Infection and Immunity, Cancer Immunology HV Amsterdam The Netherlands
- Department of Surgery, Amsterdam UMC Vrije Universiteit Amsterdam HV Amsterdam The Netherlands
| | | |
Collapse
|
28
|
Petrini I, Giaccone G. Amivantamab in the Treatment of Metastatic NSCLC: Patient Selection and Special Considerations. Onco Targets Ther 2022; 15:1197-1210. [PMID: 36246734 PMCID: PMC9555392 DOI: 10.2147/ott.s329095] [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: 07/29/2022] [Accepted: 09/22/2022] [Indexed: 11/15/2022] Open
Abstract
Amivantamab is a bispecific antibody that recognizes epidermal growth factor receptor (EGFR) and MET proto-oncogene (MET). In May 2021, the Food and Drug Administration gave an accelerated approval of amivantamab for the treatment of non-small cell lung cancer (NSCLC) patients with EGFR exon 20 insertions (Exon20ins) who progressed after platinum-based chemotherapy. Amivantamab prevents ligand binding to EGFR and MET and the dimerization of the receptors suppressing the downstream signal transduction. Moreover, amivantamab determines antibody dependent cellular cytotoxicity and down regulation of cell surface proteins through internalization of the receptor and trogocytosis. Preliminary results of the Phase I/IB CHRYSALIS trial demonstrated an objective response rate of 40% with a median duration of response of 11.1 months (95% CI 9.6-not reached) in 81 patients treated with amivantamab with pretreated NSCLC with Exon20ins EGFR mutations. In a different cohort of the CHRYSALIS trial, patients with Ex19del and L858R EGFR mutations were enrolled after progression on osimertinib. 121 and 45 patients received amivantamab or a combination with lazertinib, a third-generation tyrosine kinase inhibitor, respectively. The objective response rate was 19% and 36% in patients treated with amivantamab alone or in combination with lazertinib, with a median progression-free survival of 6.9 (95% CI: 3.2-5.3) and 11.1 (95% CI: 3.7-9.5) months, respectively. All 20 patients with Ex19del and L858R EGFR mutations who received amivantamab and lazertinib as their first line treatment achieved an objective response. Amivantamab is currently under evaluation in Phase III clinical trials for the first line treatment of NSCLCs with Exon20ins EGFR mutations in combination with chemotherapy (PAPILLON), for the first line therapy of Ex19del and L858R mutated NSCLCs in combination with lazertinib (MARIPOSA) and in combination with chemotherapy and lazertinib in NSCLCs who progressed on osimertinib (MARIPOSA-2).
Collapse
Affiliation(s)
- Iacopo Petrini
- Medical Oncology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giuseppe Giaccone
- Weill-Cornell Medicine, Meyer Cancer Center, New York, NY, USA,Correspondence: Giuseppe Giaccone, Weill-Cornell Medicine, Meyer Cancer Center, 1300 York Ave. 6th floor, Rm A603C, New York, NY, 10021, USA, Tel +1 646 962-4969, Email
| |
Collapse
|
29
|
Li Y, Basar R, Wang G, Liu E, Moyes JS, Li L, Kerbauy LN, Uprety N, Fathi M, Rezvan A, Banerjee PP, Muniz-Feliciano L, Laskowski TJ, Ensley E, Daher M, Shanley M, Mendt M, Acharya S, Liu B, Biederstädt A, Rafei H, Guo X, Melo Garcia L, Lin P, Ang S, Marin D, Chen K, Bover L, Champlin RE, Varadarajan N, Shpall EJ, Rezvani K. KIR-based inhibitory CARs overcome CAR-NK cell trogocytosis-mediated fratricide and tumor escape. Nat Med 2022; 28:2133-2144. [PMID: 36175679 PMCID: PMC9942695 DOI: 10.1038/s41591-022-02003-x] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 08/09/2022] [Indexed: 01/21/2023]
Abstract
Trogocytosis is an active process that transfers surface material from targeted to effector cells. Using multiple in vivo tumor models and clinical data, we report that chimeric antigen receptor (CAR) activation in natural killer (NK) cells promoted transfer of the CAR cognate antigen from tumor to NK cells, resulting in (1) lower tumor antigen density, thus impairing the ability of CAR-NK cells to engage with their target, and (2) induced self-recognition and continuous CAR-mediated engagement, resulting in fratricide of trogocytic antigen-expressing NK cells (NKTROG+) and NK cell hyporesponsiveness. This phenomenon could be offset by a dual-CAR system incorporating both an activating CAR against the cognate tumor antigen and an NK self-recognizing inhibitory CAR that transferred a 'don't kill me' signal to NK cells upon engagement with their TROG+ siblings. This system prevented trogocytic antigen-mediated fratricide, while sparing activating CAR signaling against the tumor antigen, and resulted in enhanced CAR-NK cell activity.
Collapse
Affiliation(s)
- Ye Li
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rafet Basar
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guohui Wang
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Enli Liu
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Judy S Moyes
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Li
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lucila N Kerbauy
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of Sao Paulo, Sao Paulo, Brazil
- Department of Stem Cell Transplantation and Cellular Therapy, Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Nadima Uprety
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mohsen Fathi
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Ali Rezvan
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Pinaki P Banerjee
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luis Muniz-Feliciano
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tamara J Laskowski
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Emily Ensley
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - May Daher
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mayra Shanley
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mayela Mendt
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sunil Acharya
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bin Liu
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander Biederstädt
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Medicine III: Hematology and Oncology, Technical University of Munich, Munich, Germany
| | - Hind Rafei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xingliang Guo
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luciana Melo Garcia
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Lin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sonny Ang
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Marin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laura Bover
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Navin Varadarajan
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
30
|
Mattei F, Andreone S, Spadaro F, Noto F, Tinari A, Falchi M, Piconese S, Afferni C, Schiavoni G. Trogocytosis in innate immunity to cancer is an intimate relationship with unexpected outcomes. iScience 2022; 25:105110. [PMID: 36185368 PMCID: PMC9515589 DOI: 10.1016/j.isci.2022.105110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/04/2022] [Accepted: 09/07/2022] [Indexed: 11/29/2022] Open
Abstract
Trogocytosis is a cellular process whereby a cell acquires a membrane fragment from a donor cell in a contact-dependent manner allowing for the transfer of surface proteins with functional integrity. It is involved in various biological processes, including cell-cell communication, immune regulation, and response to pathogens and cancer cells, with poorly defined molecular mechanisms. With the exception of eosinophils, trogocytosis has been reported in most immune cells and plays diverse roles in the modulation of anti-tumor immune responses. Here, we report that eosinophils acquire membrane fragments from tumor cells early after contact through the CD11b/CD18 integrin complex. We discuss the impact of trogocytosis in innate immune cells on cancer progression in the context of the evidence that eosinophils can engage in trogocytosis with tumor cells. We also discuss shared and cell-specific mechanisms underlying this process based on in silico modeling and provide a hypothetical molecular model for the stabilization of the immunological synapse operating in granulocytes and possibly other innate immune cells that enables trogocytosis. Trogocytosis in innate immune cells can regulate immune responses to cancer Eosinophils engage in trogocytosis with tumor cells via CD11b/CD18 integrin complex CD11b/CD18 integrin, focal adhesion molecules and actin network enable trogocytosis
Collapse
Affiliation(s)
- Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sara Andreone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Spadaro
- Core Facilities, Microscopy Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Francesco Noto
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Antonella Tinari
- Center for Gender Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Mario Falchi
- National HIV/AIDS Research Center (CNAIDS), Istituto Superiore di Sanità, Rome, Italy
| | - Silvia Piconese
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Italy
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia – Fondazione Cenci Bolognetti, Rome, Italy
| | - Claudia Afferni
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
- Corresponding author
| |
Collapse
|
31
|
Molecular Determinants Underlying the Anti-Cancer Efficacy of CD38 Monoclonal Antibodies in Hematological Malignancies. Biomolecules 2022; 12:biom12091261. [PMID: 36139103 PMCID: PMC9496523 DOI: 10.3390/biom12091261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
CD38 was first discovered as a T-cell antigen and has since been found ubiquitously expressed in various hematopoietic cells, including plasma cells, NK cells, B cells, and granulocytes. More importantly, CD38 expression levels on malignant hematopoietic cells are significantly higher than counterpart healthy cells, thus presenting itself as a promising therapeutic target. In fact, for many aggressive hematological cancers, including CLL, DLBCL, T-ALL, and NKTL, CD38 expression is significantly associated with poorer prognosis and a hyperproliferative or metastatic phenotype. Studies have shown that, beyond being a biomarker, CD38 functionally mediates dysregulated survival, adhesion, and migration signaling pathways, as well as promotes an immunosuppressive microenvironment conducive for tumors to thrive. Thus, targeting CD38 is a rational approach to overcoming these malignancies. However, clinical trials have surprisingly shown that daratumumab monotherapy has not been very effective in these other blood malignancies. Furthermore, extensive use of daratumumab in MM is giving rise to a subset of patients now refractory to daratumumab treatment. Thus, it is important to consider factors modulating the determinants of response to CD38 targeting across different blood malignancies, encompassing both the transcriptional and post-transcriptional levels so that we can diversify the strategy to enhance daratumumab therapeutic efficacy, which can ultimately improve patient outcomes.
Collapse
|
32
|
Lindorfer MA, Taylor RP. FcγR-Mediated Trogocytosis 2.0: Revisiting History Gives Rise to a Unifying Hypothesis. Antibodies (Basel) 2022; 11:antib11030045. [PMID: 35892705 PMCID: PMC9326535 DOI: 10.3390/antib11030045] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 12/25/2022] Open
Abstract
There is increasing interest in the clinical implications and immunology of trogocytosis, a process in which the receptors on acceptor cells remove and internalize cognate ligands from donor cells. We have reported that this phenomenon occurs in cancer immunotherapy, in which cells that express FcγR remove and internalize CD20 and bound mAbs from malignant B cells. This process can be generalized to include other reactions including the immune adherence phenomenon and antibody-induced immunosuppression. We discuss in detail FcγR-mediated trogocytosis and the evidence supporting a proposed predominant role for liver sinusoidal endothelial cells via the action of the inhibitory receptor FcγRIIb2. We describe experiments to test the validity of this hypothesis. The elucidation of the details of FcγR-mediated trogocytosis has the potential to allow for the development of novel therapies that can potentially block or enhance this reaction, depending upon whether the process leads to unfavorable or positive biological effects.
Collapse
|
33
|
Zhang H, Feng L, de Andrade Mello P, Mao C, Near R, Csizmadia E, Chan LLY, Enjyoji K, Gao W, Zhao H, Robson SC. Glycoengineered anti-CD39 promotes anticancer responses by depleting suppressive cells and inhibiting angiogenesis in tumor models. J Clin Invest 2022; 132:e157431. [PMID: 35775486 PMCID: PMC9246388 DOI: 10.1172/jci157431] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/12/2022] [Indexed: 11/17/2022] Open
Abstract
Immunosuppressive cells accumulating in the tumor microenvironment constitute a formidable barrier that interferes with current immunotherapeutic approaches. A unifying feature of these tumor-associated immune and vascular endothelial cells appears to be the elevated expression of ectonucleotidase CD39, which in tandem with ecto-5'-nucleotidase CD73, catalyzes the conversion of extracellular ATP into adenosine. We glycoengineered an afucosylated anti-CD39 IgG2c and tested this reagent in mouse melanoma and colorectal tumor models. We identified major biological effects of this approach on cancer growth, associated with depletion of immunosuppressive cells, mediated through enhanced Fcγ receptor-directed (FcγR-directed), antibody-dependent cellular cytotoxicity (ADCC). Furthermore, regulatory/exhausted T cells lost CD39 expression, as a consequence of antibody-mediated trogocytosis. Most strikingly, tumor-associated macrophages and endothelial cells with high CD39 expression were effectively depleted following antibody treatment, thereby blocking angiogenesis. Tumor site-specific cellular modulation and lack of angiogenesis synergized with chemotherapy and anti-PD-L1 immunotherapy in experimental tumor models. We conclude that depleting suppressive cells and targeting tumor vasculature, through administration of afucosylated anti-CD39 antibody and the activation of ADCC, comprises an improved, purinergic system-modulating strategy for cancer therapy.
Collapse
Affiliation(s)
- Haohai Zhang
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Lili Feng
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Paola de Andrade Mello
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Changchuin Mao
- Antagen Institute for Biomedical Research, Boston, Massachusetts, USA
| | - Richard Near
- Antagen Institute for Biomedical Research, Boston, Massachusetts, USA
| | - Eva Csizmadia
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Leo Li-Ying Chan
- Department of Advanced Technology R&D, Nexcelom from PerkinElmer, Lawrence, Massachusetts, USA
| | - Keiichi Enjyoji
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Wenda Gao
- Antagen Institute for Biomedical Research, Boston, Massachusetts, USA
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Simon C. Robson
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Chen X, Xue L, Ding X, Zhang J, Jiang L, Liu S, Hou H, Jiang B, Cheng L, Zhu Q, Zhang L, Zhou X, Ma J, Liu Q, Li Y, Ren Z, Jiang B, Song X, Song J, Jin W, Wei M, Shen Z, Liu X, Wang L, Li K, Zhang T. An Fc-Competent Anti-Human TIGIT Blocking Antibody Ociperlimab (BGB-A1217) Elicits Strong Immune Responses and Potent Anti-Tumor Efficacy in Pre-Clinical Models. Front Immunol 2022; 13:828319. [PMID: 35273608 PMCID: PMC8902820 DOI: 10.3389/fimmu.2022.828319] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/01/2022] [Indexed: 11/18/2022] Open
Abstract
TIGIT (T-cell immunoglobulin and ITIM domain) has emerged as a promising target in cancer immunotherapy. It is an immune “checkpoint” inhibitor primarily expressed on activated T cells, NK cells and Tregs. Engagement of TIGIT to its ligands PVR and PVR-L2 leads to inhibitory signaling in T cells, promoting functional exhaustion of tumor-infiltrating T lymphocytes. Here, we described the pre-clinical characterization of Ociperlimab (BGB-A1217), a novel humanized IgG1 anti-TIGIT antibody (mAb), and systemically evaluated the contribution of Fc functions in the TIGIT mAb-mediated anti-tumor activities. BGB-A1217 binds to the extracellular domain of human TIGIT with high affinity (KD = 0.135 nM) and specificity, and efficiently blocks the interaction between TIGIT and its ligands PVR or PVR-L2. Cell-based assays show that BGB-A1217 significantly enhances T-cell functions. In addition, BGB-A1217 induces antibody dependent cellular cytotoxicity (ADCC) against Treg cells, activates NK cells and monocytes, and removes TIGIT from T cell surfaces in an Fc-dependent manner, In vivo, BGB-A1217, either alone or in combination with an anti-PD-1 mAb elicits strong immune responses and potent anti-tumor efficacy in pre-clinical models. Moreover, the Fc effector function is critical for the anti-tumor activity of BGB-A1217 in a syngeneic human TIGIT-knock-in mouse model. The observed anti-tumor efficacy is associated with a pharmacodynamic change of TIGIT down-regulation and Treg reduction. These data support the selection of BGB-A1217 with an effector function competent Fc region for clinical development for the treatment of human cancers.
Collapse
Affiliation(s)
- Xin Chen
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Liu Xue
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xiao Ding
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Jing Zhang
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Lei Jiang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Sha Liu
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Hongjia Hou
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Bin Jiang
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Liang Cheng
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Qing Zhu
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Lijie Zhang
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xiaosui Zhou
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Jie Ma
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Qi Liu
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Yucheng Li
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Zhiying Ren
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Beibei Jiang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xiaomin Song
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Jing Song
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Wei Jin
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Min Wei
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Zhirong Shen
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xuesong Liu
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Lai Wang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Kang Li
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Tong Zhang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| |
Collapse
|
35
|
Vo DN, Leventoux N, Campos-Mora M, Gimenez S, Corbeau P, Villalba M. NK Cells Acquire CCR5 and CXCR4 by Trogocytosis in People Living with HIV-1. Vaccines (Basel) 2022; 10:vaccines10050688. [PMID: 35632444 PMCID: PMC9145773 DOI: 10.3390/vaccines10050688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 12/10/2022] Open
Abstract
NK cells play a major role in the antiviral immune response, including against HIV-1. HIV-1 patients have impaired NK cell activity with a decrease in CD56dim NK cells and an increase in the CD56−CD16+ subset, and recently it has been proposed that a population of CD56+NKG2C+KIR+CD57+ cells represents antiviral memory NK cells. Antiretroviral therapy (ART) partly restores the functional activity of this lymphocyte lineage. NK cells when interacting with their targets can gain antigens from them by the process of trogocytosis. Here we show that NK cells can obtain CCR5 and CXCR4, but barely CD4, from T cell lines by trogocytosis in vitro. By UMAP (Uniform Manifold Approximation and Projection), we show that aviremic HIV-1 patients have unique NK cell clusters that include cells expressing CCR5, NKG2C and KIRs, but lack CD57 expression. Viremic patients have a larger proportion of CXCR4+ and CCR5+ NK cells than healthy donors (HD) and this is largely increased in CD107+ cells, suggesting a link between degranulation and trogocytosis. In agreement, UMAP identified a specific NK cell cluster in viremic HIV-1 patients, which contains most of the CD107a+, CCR5+ and CXCR4+ cells. However, this cluster lacks NKG2C expression. Therefore, NK cells can gain CCR5 and CXCR4 by trogocytosis, which depends on degranulation.
Collapse
Affiliation(s)
- Dang-Nghiem Vo
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France; (D.-N.V.); (M.C.-M.)
| | - Nicolas Leventoux
- Institute for Human Genetics, CNRS-Montpellier University, UMR9002, 141 Rue de la Cardonille, CEDEX, 34396 Montpellier, France; (N.L.); (S.G.)
| | - Mauricio Campos-Mora
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France; (D.-N.V.); (M.C.-M.)
| | - Sandrine Gimenez
- Institute for Human Genetics, CNRS-Montpellier University, UMR9002, 141 Rue de la Cardonille, CEDEX, 34396 Montpellier, France; (N.L.); (S.G.)
| | - Pierre Corbeau
- Institute for Human Genetics, CNRS-Montpellier University, UMR9002, 141 Rue de la Cardonille, CEDEX, 34396 Montpellier, France; (N.L.); (S.G.)
- Immunology Department, University Hospital, Place du Pr Debré, CEDEX, 30029 Nîmes, France
- Correspondence: (P.C.); (M.V.)
| | - Martin Villalba
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France; (D.-N.V.); (M.C.-M.)
- IRMB, University Montpellier, INSERM, CNRS, CHU Montpellier, 34295 Montpellier, France
- Institut du Cancer Avignon-Provence Sainte-Catherine, 84000 Avignon, France
- Correspondence: (P.C.); (M.V.)
| |
Collapse
|
36
|
Hasim MS, Marotel M, Hodgins JJ, Vulpis E, Makinson OJ, Asif S, Shih HY, Scheer AK, MacMillan O, Alonso FG, Burke KP, Cook DP, Li R, Petrucci MT, Santoni A, Fallon PG, Sharpe AH, Sciumè G, Veillette A, Zingoni A, Gray DA, McCurdy A, Ardolino M. When killers become thieves: Trogocytosed PD-1 inhibits NK cells in cancer. SCIENCE ADVANCES 2022; 8:eabj3286. [PMID: 35417234 PMCID: PMC9007500 DOI: 10.1126/sciadv.abj3286] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 02/23/2022] [Indexed: 05/12/2023]
Abstract
Trogocytosis modulates immune responses, with still unclear underlying molecular mechanisms. Using leukemia mouse models, we found that lymphocytes perform trogocytosis at high rates with tumor cells. While performing trogocytosis, both Natural Killer (NK) and CD8+ T cells acquire the checkpoint receptor PD-1 from leukemia cells. In vitro and in vivo investigation revealed that PD-1 on the surface of NK cells, rather than being endogenously expressed, was derived entirely from leukemia cells in a SLAM receptor-dependent fashion. PD-1 acquired via trogocytosis actively suppressed NK cell antitumor immunity. PD-1 trogocytosis was corroborated in patients with clonal plasma cell disorders, where NK cells that stained for PD-1 also stained for tumor cell markers. Our results, in addition to shedding light on a previously unappreciated mechanism underlying the presence of PD-1 on NK and cytotoxic T cells, reveal the immunoregulatory effect of membrane transfer occurring when immune cells contact tumor cells.
Collapse
Affiliation(s)
- Mohamed S. Hasim
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- CI3, University of Ottawa, Ottawa, ON, Canada
| | - Marie Marotel
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- CI3, University of Ottawa, Ottawa, ON, Canada
| | - Jonathan J. Hodgins
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- CI3, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Elisabetta Vulpis
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci-Bolognetti, Rome, Italy
| | - Olivia J. Makinson
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- CI3, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Sara Asif
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- CI3, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Han-Yun Shih
- Neuro-Immune Regulome Unit, National Eye Institute, NIH, Bethesda, MD, USA
| | - Amit K. Scheer
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Olivia MacMillan
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- CI3, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Felipe G. Alonso
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Kelly P. Burke
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - David P. Cook
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Rui Li
- Department of Medicine, McGill University, Montréal, QC, Canada
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, QC, Canada
| | - Maria Teresa Petrucci
- Department of Cellular Biotechnology and Hematology, “Sapienza” University of Rome, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci-Bolognetti, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Padraic G. Fallon
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Arlene H. Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | - Giuseppe Sciumè
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci-Bolognetti, Rome, Italy
| | - André Veillette
- Department of Medicine, McGill University, Montréal, QC, Canada
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, QC, Canada
- Department of Medicine, University of Montréal, Montréal, QC, Canada
| | - Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci-Bolognetti, Rome, Italy
| | - Douglas A. Gray
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Arleigh McCurdy
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Division of Hematology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michele Ardolino
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- CI3, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
37
|
Zhao S, Zhang L, Xiang S, Hu Y, Wu Z, Shen J. Gnawing Between Cells and Cells in the Immune System: Friend or Foe? A Review of Trogocytosis. Front Immunol 2022; 13:791006. [PMID: 35185886 PMCID: PMC8850298 DOI: 10.3389/fimmu.2022.791006] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/14/2022] [Indexed: 12/27/2022] Open
Abstract
Trogocytosis occurs when one cell contacts and quickly nibbles another cell and is characterized by contact between living cells and rapid transfer of membrane fragments with functional integrity. Many immune cells are involved in this process, such as T cells, B cells, NK cells, APCs. The transferred membrane molecules including MHC molecules, costimulatory molecules, receptors, antigens, etc. An increasing number of studies have shown that trogocytosis plays an important role in the immune system and the occurrence of relevant diseases. Thus, whether trogocytosis is a friend or foe of the immune system is puzzling, and the precise mechanism underlying it has not yet been fully elucidated. Here, we provide an integrated view of the acquired findings on the connections between trogocytosis and the immune system.
Collapse
Affiliation(s)
- Siyu Zhao
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Lichao Zhang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Suoyu Xiang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Yunyi Hu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Zhongdao Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Jia Shen
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| |
Collapse
|
38
|
Wu TH, Hsieh SC, Li TH, Lu CH, Liao HT, Shen CY, Li KJ, Wu CH, Kuo YM, Tsai CY, Yu CL. Molecular Basis for Paradoxical Activities of Polymorphonuclear Neutrophils in Inflammation/Anti-Inflammation, Bactericide/Autoimmunity, Pro-Cancer/Anticancer, and Antiviral Infection/SARS-CoV-II-Induced Immunothrombotic Dysregulation. Biomedicines 2022; 10:biomedicines10040773. [PMID: 35453523 PMCID: PMC9032061 DOI: 10.3390/biomedicines10040773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 02/06/2023] Open
Abstract
Polymorphonuclear neutrophils (PMNs) are the most abundant white blood cells in the circulation. These cells act as the fast and powerful defenders against environmental pathogenic microbes to protect the body. In addition, these innate inflammatory cells can produce a number of cytokines/chemokines/growth factors for actively participating in the immune network and immune homeostasis. Many novel biological functions including mitogen-induced cell-mediated cytotoxicity (MICC) and antibody-dependent cell-mediated cytotoxicity (ADCC), exocytosis of microvesicles (ectosomes and exosomes), trogocytosis (plasma membrane exchange) and release of neutrophil extracellular traps (NETs) have been successively discovered. Furthermore, recent investigations unveiled that PMNs act as a double-edged sword to exhibit paradoxical activities on pro-inflammation/anti-inflammation, antibacteria/autoimmunity, pro-cancer/anticancer, antiviral infection/COVID-19-induced immunothrombotic dysregulation. The NETs released from PMNs are believed to play a pivotal role in these paradoxical activities, especially in the cytokine storm and immunothrombotic dysregulation in the recent SARS-CoV-2 pandemic. In this review, we would like to discuss in detail the molecular basis for these strange activities of PMNs.
Collapse
Affiliation(s)
- Tsai-Hung Wu
- Division of Nephrology, Taipei Veterans General Hospital, National Yang-Ming Chiao-Tung University, Taipei 11217, Taiwan;
| | - Song-Chou Hsieh
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
| | - Tsu-Hao Li
- Division of Allergy, Immunology and Rheumatology, Shin Kong Wu Ho Shi Hospital, Taipei 11101, Taiwan;
- Institute of Clinical Medicine, National Yang-Ming Chiao-Tung University, Taipei 11217, Taiwan
| | - Cheng-Hsun Lu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
- Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Hsien-Tzung Liao
- Division of Allergy, Immunology and Rheumatology, Taipei Veterans General Hospital, National Yang-Ming Chiao-Tung University, Taipei 11217, Taiwan;
| | - Chieh-Yu Shen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
- Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Ko-Jen Li
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
| | - Cheng-Han Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
- Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Yu-Min Kuo
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
- Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Chang-Youh Tsai
- Division of Allergy, Immunology and Rheumatology, Taipei Veterans General Hospital, National Yang-Ming Chiao-Tung University, Taipei 11217, Taiwan;
- Correspondence: (C.-Y.T.); (C.-L.Y.)
| | - Chia-Li Yu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
- Correspondence: (C.-Y.T.); (C.-L.Y.)
| |
Collapse
|
39
|
In Silico Designed Gain-of-Function Variants of Complement C2 Support Cytocidal Activity of Anticancer Monoclonal Antibodies. Cancers (Basel) 2022; 14:cancers14051270. [PMID: 35267578 PMCID: PMC8909654 DOI: 10.3390/cancers14051270] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/06/2023] Open
Abstract
The molecular target for the classical complement pathway (CP) is defined by surface-bound immunoglobulins. Therefore, numerous anticancer monoclonal antibodies (mAbs) exploit the CP as their effector mechanism. Conversely, the alternative complement pathway (AP) is spontaneously induced on the host and microbial surfaces, but complement inhibitors on host cells prevent its downstream processing. Gain-of-function (GoF) mutations in the AP components that oppose physiological regulation directly predispose carriers to autoimmune/inflammatory diseases. Based on the homology between AP and CP components, we modified the CP component C2 so that it emulates the known pathogenic mutations in the AP component, factor B. By using tumor cell lines and patient-derived leukemic cells along with a set of clinically approved immunotherapeutics, we showed that the supplementation of serum with recombinant GoF C2 variants not only enhances the cytocidal effect of type I anti-CD20 mAbs rituximab and ofatumumab, but also lowers the threshold of mAbs necessary for the efficient lysis of tumor cells and efficiently exploits the leftovers of the drug accumulated in patients' sera after the previous infusion. Moreover, we demonstrate that GoF C2 acts in concert with other therapeutic mAbs, such as type II anti-CD20, anti-CD22, and anti-CD38 specimens, for overcoming cancer cells resistance to complement attack.
Collapse
|
40
|
Sodium stibogluconate and CD47-SIRPα blockade overcome resistance of anti-CD20-opsonized B cells to neutrophil killing. Blood Adv 2021; 6:2156-2166. [PMID: 34942000 PMCID: PMC9006259 DOI: 10.1182/bloodadvances.2021005367] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 11/17/2021] [Indexed: 12/02/2022] Open
Abstract
SSG turns neutrophil trogocytosis of rituximab-opsonized malignant B cells into cell killing. Neutrophil antibody–dependent killing of malignant B cells occurs primarily through FcγRI (CD64).
Anti-CD20 antibodies such as rituximab are broadly used to treat B-cell malignancies. These antibodies can induce various effector functions, including immune cell-mediated antibody-dependent cellular cytotoxicity (ADCC). Neutrophils can induce ADCC toward solid cancer cells by trogoptosis, a cytotoxic mechanism known to be dependent on trogocytosis. However, neutrophils seem to be incapable of killing rituximab-opsonized B-cell lymphoma cells. Nevertheless, neutrophils do trogocytose rituximab-opsonized B-cell lymphoma cells, but this only reduces CD20 surface expression and is thought to render tumor cells therapeutically resistant to further rituximab-dependent destruction. Here, we demonstrate that resistance of B-cell lymphoma cells toward neutrophil killing can be overcome by a combination of CD47-SIRPα checkpoint blockade and sodium stibogluconate (SSG), an anti-leishmaniasis drug and documented inhibitor of the tyrosine phosphatase SHP-1. SSG enhanced neutrophil-mediated ADCC of solid tumor cells but enabled trogoptotic killing of B-cell lymphoma cells by turning trogocytosis from a mechanism that contributes to resistance into a cytotoxic anti-cancer mechanism. Tumor cell killing in the presence of SSG required both antibody opsonization of the target cells and disruption of CD47-SIRPα interactions. These results provide a more detailed understanding of the role of neutrophil trogocytosis in antibody-mediated destruction of B cells and clues on how to further optimize antibody therapy of B-cell malignancies.
Collapse
|
41
|
The Multiple Roles of Trogocytosis in Immunity, the Nervous System, and Development. BIOMED RESEARCH INTERNATIONAL 2021; 2021:1601565. [PMID: 34604381 PMCID: PMC8483919 DOI: 10.1155/2021/1601565] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022]
Abstract
Trogocytosis is a general biological process that involves one cell physically taking small parts of the membrane and other components from another cell. In trogocytosis, one cell seems to take little “bites” from another cell resulting in multiple outcomes from these cell-cell interactions. Trogocytosis was first described in protozoan parasites, which by taking pieces of host cells, kill them and cause tissue damage. Now, it is known that this process is also performed by cells of the immune system with important consequences such as cell communication and activation, elimination of microbial pathogens, and even control of cancer cells. More recently, trogocytosis has also been reported to occur in cells of the central nervous system and in various cells during development. Some of the molecules involved in phagocytosis also participate in trogocytosis. However, the molecular mechanisms that regulate trogocytosis are still a mystery. Elucidating these mechanisms is becoming a research area of much interest. For example, why neutrophils can engage trogocytosis to kill Trichomonas vaginalis parasites, but neutrophils use phagocytosis to eliminate already death parasites? Thus, trogocytosis is a significant process in normal physiology that multiple cells from different organisms use in various scenarios of health and disease. In this review, we present the basic principles known on the process of trogocytosis and discuss the importance in this process to host-pathogen interactions and to normal functions in the immune and nervous systems.
Collapse
|
42
|
Rossignol E, Alter G, Julg B. Antibodies for Human Immunodeficiency Virus-1 Cure Strategies. J Infect Dis 2021; 223:22-31. [PMID: 33586772 DOI: 10.1093/infdis/jiaa165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human immunodeficiency virus (HIV) infection leads to the establishment of a long-lived latent cellular reservoir. One strategy to eliminate quiescent reservoir cells is to reactivate virus replication to induce HIV envelope glycoprotein (Env) expression on the cell surface exposing them to subsequent antibody targeting. Via the interactions between the antibody Fc domain and Fc-γ receptors (FcγRs) that are expressed on innate effector cells, such as natural killer cells, monocytes, and neutrophils, antibodies can mediate the elimination of infected cells. Over the last decade, a multitude of human monoclonal antibodies that are broadly neutralizing across many HIV-1 subtypes have been identified and are currently being explored for HIV eradication strategies. Antibody development also includes novel Fc engineering approaches to increase engagement of effector cells and optimize antireservoir efficacy. In this review, we discuss the usefulness of antibodies for HIV eradication approaches specifically focusing on antibody-mediated strategies to target latently infected cells and options to increase antibody efficacy.
Collapse
Affiliation(s)
- Evan Rossignol
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Boris Julg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA.,Massachusetts General Hospital, Infectious Disease Unit, Boston, Massachusetts, USA
| |
Collapse
|
43
|
Bakhtiari LA, Wells MJ, Gordon VD. High-throughput assays show the timescale for phagocytic success depends on the target toughness. BIOPHYSICS REVIEWS 2021; 2:031402. [PMID: 34632456 PMCID: PMC8485781 DOI: 10.1063/5.0057071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/07/2021] [Indexed: 11/14/2022]
Abstract
Phagocytic immune cells can clear pathogens from the body by engulfing them. Bacterial biofilms are communities of bacteria that are bound together in a matrix that gives biofilms viscoelastic mechanical properties that do not exist for free-swimming bacteria. Since a neutrophil is too small to engulf an entire biofilm, it must be able to detach and engulf a few bacteria at a time if it is to use phagocytosis to clear the infection. We recently found a negative correlation between the target elasticity and phagocytic success. That earlier work used time-consuming, manual analysis of micrographs of neutrophils and fluorescent beads. Here, we introduce and validate flow cytometry as a fast and high-throughput technique that increases the number of neutrophils analyzed per experiment by two orders of magnitude, while also reducing the time required to do so from hours to minutes. We also introduce the use of polyacrylamide gels in our assay for engulfment success. The tunability of polyacrylamide gels expands the mechanical parameter space we can study, and we find that high toughness and yield strain, even with low elasticity, also impact the phagocytic success as well as the timescale thereof. For stiff gels with low-yield strain, and consequent low toughness, phagocytic success is nearly four times greater when neutrophils are incubated with gels for 6 h than after only 1 h of incubation. In contrast, for soft gels with high-yield strain and consequent high toughness, successful engulfment is much less time-sensitive, increasing by less than a factor of two from 1 to 6 h incubation.
Collapse
|
44
|
NK cells eliminate Epstein-Barr virus bound to B cells through a specific antibody-mediated uptake. PLoS Pathog 2021; 17:e1009868. [PMID: 34415956 PMCID: PMC8409624 DOI: 10.1371/journal.ppat.1009868] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 09/01/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
Epstein Barr virus (EBV) causes a highly prevalent and lifelong infection contributing to the development of some malignancies. In addition to the key role played by T cells in controlling this pathogen, NK cells mediate cytotoxicity and IFNγ production in response to EBV-infected B cells in lytic cycle, both directly and through antibody (Ab)-dependent activation. We recently described that EBV-specific Ab-dependent NK cell interaction with viral particles (VP) bound to B cells triggered degranulation and TNFα secretion but not B cell lysis nor IFNγ production. In this report we show that NK cell activation under these conditions reduced B cell transformation by EBV. NK cells eliminated VP from the surface of B cells through a specific and active process which required tyrosine kinase activation, actin polymerization and Ca2+, being independent of proteolysis and perforin. VP were displayed at the NK cell surface before being internalized and partially shuttled to early endosomes and lysosomes. VP transfer was encompassed by a trogocytosis process including the EBV receptor CD21, together with CD19 and CD20. Our study reveals a novel facet of the antibody-dependent NK cell mediated response to this viral infection. Epstein-Barr virus (EBV) is a member of the herpesvirus family which causes a frequent and lifelong infection. The immune system is unable to fully eliminate the virus, which remains dormant in infected B lymphocytes. EBV reactivation leads to the production of new infective particles, spreading to other cells and favoring its transmission. EBV infection goes generally unnoticed in healthy individuals, though it may occasionally cause a disease termed Infectious Mononucleosis, as well as severe disorders in patients with a defective immune response. Remarkably, EBV has oncogenic potential contributing to the development of some tumors, and has been associated to autoimmune diseases. T lymphocytes and Natural Killer (NK) cells play an essential role in the defense against EBV, killing infected cells when the virus reactivates. Antiviral NK cell functions may be also triggered by antibodies (Ab) recognizing infected cells. In this report we provide the first evidence supporting that NK cells in combination with anti-EBV Ab are able to eliminate the virus attached to the surface of B cells, reducing their infection without killing them.
Collapse
|
45
|
McBride HJ, Jassem S, Chow V, Kanakaraj P, Lebrec H, Kuhns S, Ferbas J, Wong M, Thway TM. Non-clinical similarity of biosimilar ABP 798 with rituximab reference product. Biologicals 2021; 72:42-53. [PMID: 34303595 DOI: 10.1016/j.biologicals.2021.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 05/13/2021] [Accepted: 05/20/2021] [Indexed: 01/02/2023] Open
Abstract
ABP 798 is a biosimilar to Rituxan® (rituximab reference product [RP]). Non-clinical assessments relevant to the primary and secondary mechanisms of action (MOA) contribute to the totality of the evidence (TOE) in supporting biosimilarity and are critical in providing scientific evidence for extrapolation of indications. Similarity of ABP 798 with rituximab RP was investigated across a range of biological activities which have potential impact on pharmacokinetics and clinical efficacy with non-clinical assessments relevant to MOA such as CD20 internalization, trogocytosis, binding to primary human natural killer (NK) cells as well as the ability to induce antibody-dependent cellular phagocytosis (ADCP) in peripheral blood mononuclear cells. Additionally, in vitro synergy of ABP 798 or RP with chemotherapeutic agents, in vivo xenograft studies in mice, and toxicological assessments in cynomolgus monkeys (including B cell depletion and toxicokinetics) were also conducted. Results from these non-clinical assessments contribute to the TOE supporting the biosimilarity between ABP 798 and rituximab RP across a range of primary and secondary MOAs and support justification for extrapolation to all indications of use for ABP 798 for which the RP is approved.
Collapse
Affiliation(s)
- Helen J McBride
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| | - Shea Jassem
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| | - Vincent Chow
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| | | | - Herve Lebrec
- Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA.
| | - Scott Kuhns
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| | - John Ferbas
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| | - Min Wong
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| | - Theingi M Thway
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| |
Collapse
|
46
|
Crickx E, Chappert P, Sokal A, Weller S, Azzaoui I, Vandenberghe A, Bonnard G, Rossi G, Fadeev T, Storck S, Fadlallah J, Meignin V, Rivière E, Audia S, Godeau B, Michel M, Weill JC, Reynaud CA, Mahévas M. Rituximab-resistant splenic memory B cells and newly engaged naive B cells fuel relapses in patients with immune thrombocytopenia. Sci Transl Med 2021; 13:13/589/eabc3961. [PMID: 33853929 DOI: 10.1126/scitranslmed.abc3961] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 10/29/2020] [Accepted: 03/22/2021] [Indexed: 01/19/2023]
Abstract
Rituximab (RTX), an antibody targeting CD20, is widely used as a first-line therapeutic strategy in B cell-mediated autoimmune diseases. However, a large proportion of patients either do not respond to the treatment or relapse during B cell reconstitution. Here, we characterize the cellular basis responsible for disease relapse in secondary lymphoid organs in humans, taking advantage of the opportunity offered by therapeutic splenectomy in patients with relapsing immune thrombocytopenia. By analyzing the B and plasma cell immunoglobulin gene repertoire at bulk and antigen-specific single-cell level, we demonstrate that relapses are associated with two responses coexisting in germinal centers and involving preexisting mutated memory B cells that survived RTX treatment and naive B cells generated upon reconstitution of the B cell compartment. To identify distinctive characteristics of the memory B cells that escaped RTX-mediated depletion, we analyzed RTX refractory patients who did not respond to treatment at the time of B cell depletion. We identified, by single-cell RNA sequencing (scRNA-seq) analysis, a population of quiescent splenic memory B cells that present a unique, yet reversible, RTX-shaped phenotype characterized by down-modulation of B cell-specific factors and expression of prosurvival genes. Our results clearly demonstrate that these RTX-resistant autoreactive memory B cells reactivate as RTX is cleared and give rise to plasma cells and further germinal center reactions. Their continued surface expression of CD19 makes them efficient targets for current anti-CD19 therapies. This study thus identifies a pathogenic contributor to autoimmune diseases that can be targeted by available therapeutic agents.
Collapse
Affiliation(s)
- Etienne Crickx
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMS8253, Université Paris Descartes, Sorbonne Paris Cité, 75993 Paris Cedex 14, France.,Service de Médecine Interne, Centre national de référence des cytopénies auto-immunes de l'adulte, Hôpital Henri Mondor, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris Est Créteil, 94000 Créteil, France
| | - Pascal Chappert
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMS8253, Université Paris Descartes, Sorbonne Paris Cité, 75993 Paris Cedex 14, France.,Inovarion, 75005 Paris, France
| | - Aurélien Sokal
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMS8253, Université Paris Descartes, Sorbonne Paris Cité, 75993 Paris Cedex 14, France
| | - Sandra Weller
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMS8253, Université Paris Descartes, Sorbonne Paris Cité, 75993 Paris Cedex 14, France
| | - Imane Azzaoui
- Service de Médecine Interne, Centre national de référence des cytopénies auto-immunes de l'adulte, Hôpital Henri Mondor, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris Est Créteil, 94000 Créteil, France.,INSERM U955, Université Paris Est Créteil (UPEC), 94000 Créteil, France
| | - Alexis Vandenberghe
- Service de Médecine Interne, Centre national de référence des cytopénies auto-immunes de l'adulte, Hôpital Henri Mondor, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris Est Créteil, 94000 Créteil, France.,INSERM U955, Université Paris Est Créteil (UPEC), 94000 Créteil, France
| | - Guillaume Bonnard
- INSERM U955, Université Paris Est Créteil (UPEC), 94000 Créteil, France
| | - Geoffrey Rossi
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMS8253, Université Paris Descartes, Sorbonne Paris Cité, 75993 Paris Cedex 14, France
| | - Tatiana Fadeev
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMS8253, Université Paris Descartes, Sorbonne Paris Cité, 75993 Paris Cedex 14, France
| | - Sébastien Storck
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMS8253, Université Paris Descartes, Sorbonne Paris Cité, 75993 Paris Cedex 14, France
| | - Jehane Fadlallah
- Service d'immunologie clinique, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université Paris Diderot, Sorbonne Paris Cité, 75010 Paris, France
| | - Véronique Meignin
- Service d'anatomopathologie, Hôpital Saint-Louis (AP-HP), 75010 Paris, France
| | - Etienne Rivière
- Service de médecine interne, Hôpital Haut-Lévêque, 33604 Pessac, France
| | - Sylvain Audia
- Service de médecine interne, Hôpital du Bocage, 21000 Dijon, France
| | - Bertrand Godeau
- Service de Médecine Interne, Centre national de référence des cytopénies auto-immunes de l'adulte, Hôpital Henri Mondor, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris Est Créteil, 94000 Créteil, France
| | - Marc Michel
- Service de Médecine Interne, Centre national de référence des cytopénies auto-immunes de l'adulte, Hôpital Henri Mondor, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris Est Créteil, 94000 Créteil, France
| | - Jean-Claude Weill
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMS8253, Université Paris Descartes, Sorbonne Paris Cité, 75993 Paris Cedex 14, France
| | - Claude-Agnès Reynaud
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMS8253, Université Paris Descartes, Sorbonne Paris Cité, 75993 Paris Cedex 14, France
| | - Matthieu Mahévas
- Institut Necker-Enfants Malades, INSERM U1151/CNRS UMS8253, Université Paris Descartes, Sorbonne Paris Cité, 75993 Paris Cedex 14, France. .,Service de Médecine Interne, Centre national de référence des cytopénies auto-immunes de l'adulte, Hôpital Henri Mondor, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris Est Créteil, 94000 Créteil, France.,INSERM U955, Université Paris Est Créteil (UPEC), 94000 Créteil, France
| |
Collapse
|
47
|
Abstract
Immunotherapy marked a milestone in cancer treatment and has shown unprecedented efficacy in a variety of hematological malignancies. Downregulation or loss of target antigens is commonly seen after immunotherapy, which often causes diagnostic dilemma and represents a key mechanism that tumor escapes from immunotherapy. The awareness of phenotypic changes after targeted immunotherapy is important to avoid misdiagnosis. Further understanding of the mechanisms of antigen loss is paramount for the development of therapeutic approaches that can prevent or overcome antigen escape in future immunotherapy.
Collapse
Affiliation(s)
- Ting Zhou
- Flow Cytometry Unit, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Hematopathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hao-Wei Wang
- Flow Cytometry Unit, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Hematopathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
48
|
Capuano C, Pighi C, Battella S, De Federicis D, Galandrini R, Palmieri G. Harnessing CD16-Mediated NK Cell Functions to Enhance Therapeutic Efficacy of Tumor-Targeting mAbs. Cancers (Basel) 2021; 13:cancers13102500. [PMID: 34065399 PMCID: PMC8161310 DOI: 10.3390/cancers13102500] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/18/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Natural Killer (NK) cells play a major role in cancer immunotherapy based on tumor-targeting mAbs. NK cell-mediated tumor cell killing and cytokine secretion are powerfully stimulated upon interaction with IgG-opsonized tumor cells, through the aggregation of FcγRIIIA/CD16 IgG receptor. Advances in basic and translational NK cell biology have led to the development of strategies that, by improving mAb-dependent antitumor responses, may overcome the current limitations of antibody therapy attributable to tolerance, immunosuppressive microenvironment, and genotypic factors. This review provides an overview of the immunotherapeutic strategies being pursued to improve the efficacy of mAb-induced NK antitumor activity. The exploitation of antibody combinations, antibody-based molecules, used alone or combined with adoptive NK cell therapy, will be uncovered. Within the landscape of NK cell heterogeneity, we stress the role of memory NK cells as promising effectors in the next generation of immunotherapy with the aim to obtain long-lasting tumor control. Abstract Natural killer (NK) cells hold a pivotal role in tumor-targeting monoclonal antibody (mAb)-based activity due to the expression of CD16, the low-affinity receptor for IgG. Indeed, beyond exerting cytotoxic function, activated NK cells also produce an array of cytokines and chemokines, through which they interface with and potentiate adaptive immune responses. Thus, CD16-activated NK cells can concur to mAb-dependent “vaccinal effect”, i.e., the development of antigen-specific responses, which may be highly relevant in maintaining long-term protection of treated patients. On this basis, the review will focus on strategies aimed at potentiating NK cell-mediated antitumor functions in tumor-targeting mAb-based regimens, represented by (a) mAb manipulation strategies, aimed at augmenting recruitment and efficacy of NK cells, such as Fc-engineering, and the design of bi- or trispecific NK cell engagers and (b) the possible exploitation of memory NK cells, whose distinctive characteristics (enhanced responsiveness to CD16 engagement, longevity, and intrinsic resistance to the immunosuppressive microenvironment) may maximize therapeutic mAb antitumor efficacy.
Collapse
Affiliation(s)
- Cristina Capuano
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.C.); (C.P.); (S.B.); (D.D.F.)
| | - Chiara Pighi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.C.); (C.P.); (S.B.); (D.D.F.)
| | - Simone Battella
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.C.); (C.P.); (S.B.); (D.D.F.)
- ReiThera Srl, 00128 Rome, Italy
| | - Davide De Federicis
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.C.); (C.P.); (S.B.); (D.D.F.)
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Ricciarda Galandrini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.C.); (C.P.); (S.B.); (D.D.F.)
- Correspondence: (R.G.); (G.P.)
| | - Gabriella Palmieri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.C.); (C.P.); (S.B.); (D.D.F.)
- Correspondence: (R.G.); (G.P.)
| |
Collapse
|
49
|
Miyake K, Karasuyama H. The Role of Trogocytosis in the Modulation of Immune Cell Functions. Cells 2021; 10:cells10051255. [PMID: 34069602 PMCID: PMC8161413 DOI: 10.3390/cells10051255] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 12/16/2022] Open
Abstract
Trogocytosis is an active process, in which one cell extracts the cell fragment from another cell, leading to the transfer of cell surface molecules, together with membrane fragments. Recent reports have revealed that trogocytosis can modulate various biological responses, including adaptive and innate immune responses and homeostatic responses. Trogocytosis is evolutionally conserved from protozoan parasites to eukaryotic cells. In some cases, trogocytosis results in cell death, which is utilized as a mechanism for antibody-dependent cytotoxicity (ADCC). In other cases, trogocytosis-mediated intercellular protein transfer leads to both the acquisition of novel functions in recipient cells and the loss of cellular functions in donor cells. Trogocytosis in immune cells is typically mediated by receptor–ligand interactions, including TCR–MHC interactions and Fcγ receptor-antibody-bound molecule interactions. Additionally, trogocytosis mediates the transfer of MHC molecules to various immune and non-immune cells, which confers antigen-presenting activity on non-professional antigen-presenting cells. In this review, we summarize the recent advances in our understanding of the role of trogocytosis in immune modulation.
Collapse
|
50
|
Nakayama M, Hori A, Toyoura S, Yamaguchi SI. Shaping of T Cell Functions by Trogocytosis. Cells 2021; 10:cells10051155. [PMID: 34068819 PMCID: PMC8151334 DOI: 10.3390/cells10051155] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023] Open
Abstract
Trogocytosis is an active process whereby plasma membrane proteins are transferred from one cell to the other cell in a cell-cell contact-dependent manner. Since the discovery of the intercellular transfer of major histocompatibility complex (MHC) molecules in the 1970s, trogocytosis of MHC molecules between various immune cells has been frequently observed. For instance, antigen-presenting cells (APCs) acquire MHC class I (MHCI) from allografts, tumors, and virally infected cells, and these APCs are subsequently able to prime CD8+ T cells without antigen processing via the preformed antigen-MHCI complexes, in a process called cross-dressing. T cells also acquire MHC molecules from APCs or other target cells via the immunological synapse formed at the cell-cell contact area, and this phenomenon impacts T cell activation. Compared with naïve and effector T cells, T regulatory cells have increased trogocytosis activity in order to remove MHC class II and costimulatory molecules from APCs, resulting in the induction of tolerance. Accumulating evidence suggests that trogocytosis shapes T cell functions in cancer, transplantation, and during microbial infections. In this review, we focus on T cell trogocytosis and the related inflammatory diseases.
Collapse
|