51
|
Zahavi D, AlDeghaither D, O'Connell A, Weiner LM. Enhancing antibody-dependent cell-mediated cytotoxicity: a strategy for improving antibody-based immunotherapy. Antib Ther 2018; 1:7-12. [PMID: 33928217 PMCID: PMC7990127 DOI: 10.1093/abt/tby002] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 05/25/2018] [Accepted: 06/01/2018] [Indexed: 12/11/2022] Open
Abstract
The targeting of surface antigens expressed on tumor cells by monoclonal antibodies (mAbs) has revolutionized cancer therapeutics. One mechanism of action of antibody-based immunotherapy is the activation of immune effector cells to mediate antibody-dependent cell-mediated cytotoxicity (ADCC). This review will summarize the process of ADCC, its important role in the efficacy of mAb therapy, how to measure it, and finally future strategies for antibody design that can take advantage of it to improve clinical performance.
Collapse
Affiliation(s)
- David Zahavi
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center,3800 Reservoir Rd NW, Washington, DC 20007, USA
| | - Dalal AlDeghaither
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center,3800 Reservoir Rd NW, Washington, DC 20007, USA
| | - Allison O'Connell
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center,3800 Reservoir Rd NW, Washington, DC 20007, USA
| | - Louis M Weiner
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center,3800 Reservoir Rd NW, Washington, DC 20007, USA
| |
Collapse
|
52
|
Wang QM, Tang PMK, Lian GY, Li C, Li J, Huang XR, To KF, Lan HY. Enhanced Cancer Immunotherapy with Smad3-Silenced NK-92 Cells. Cancer Immunol Res 2018; 6:965-977. [PMID: 29915022 DOI: 10.1158/2326-6066.cir-17-0491] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 03/12/2018] [Accepted: 06/12/2018] [Indexed: 12/22/2022]
Abstract
Natural killer (NK) cells, early effectors in anticancer immunity, are paralyzed by TGFβ1, an immunosuppressive cytokine produced by cancer cells. Development and activity of NK cells are largely inhibited in the Smad3-dependent tumor microenvironment. Here, we used genetic engineering to generate a stable SMAD3-silencing human NK cell line, NK-92-S3KD, whose cancer-killing activity and cytokine production were significantly enhanced under TGFβ1-rich condition compared with the parental cell line. Interestingly, we identified that the IFNG gene is a direct E4BP4 target gene. Thus, silencing of SMAD3 allows upregulation of E4BP4 that subsequently promoting interferon-γ (IFNγ) production in the NK-92-S3KD cells. More importantly, NK-92-S3KD immunotherapy increases the production of not only IFNγ, but also granzyme B and perforin in tumors; therefore, inhibiting cancer progression in two xenograft mouse models with human hepatoma (HepG2) and melanoma (A375). Thus, the NK-92-S3KD cell line may be useful for the clinical immunotherapy of cancer. Cancer Immunol Res; 6(8); 965-77. ©2018 AACR.
Collapse
Affiliation(s)
- Qing-Ming Wang
- Li Ka Shing Institute of Health Sciences, Department of Medicine & Therapeutics, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Patrick Ming-Kuen Tang
- Li Ka Shing Institute of Health Sciences, Department of Medicine & Therapeutics, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Guang-Yu Lian
- Li Ka Shing Institute of Health Sciences, Department of Medicine & Therapeutics, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chunjie Li
- Li Ka Shing Institute of Health Sciences, Department of Medicine & Therapeutics, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jinhong Li
- Li Ka Shing Institute of Health Sciences, Department of Medicine & Therapeutics, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiao-Ru Huang
- Li Ka Shing Institute of Health Sciences, Department of Medicine & Therapeutics, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui-Yao Lan
- Li Ka Shing Institute of Health Sciences, Department of Medicine & Therapeutics, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
53
|
Singh K, Marasini B, Chen X, Spearman P. A novel Ebola virus antibody-dependent cell-mediated cytotoxicity (Ebola ADCC) assay. J Immunol Methods 2018; 460:10-16. [PMID: 29894746 DOI: 10.1016/j.jim.2018.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 05/03/2018] [Accepted: 06/06/2018] [Indexed: 12/21/2022]
Abstract
Ebolaviruses are highly virulent pathogens that cause Ebola viral disease (EVD). Data from non-human primate (NHP) models and from human survivors of EVD suggest that anti-Ebola antibodies play an integral role in protection. Antibody-dependent cell-mediated cytotoxicity (ADCC) is a potential mechanism through which anti-Ebola antibodies may mediate protection. We developed a robust Ebola-specific ADCC assay for use in ongoing trials of Ebola vaccines. Stable cell lines for inducible Zaire ebolavirus glycoprotein (EBOV GP) expression were developed to provide a uniform source of target cells in the assay, and were combined with an existing human natural killer (NK) cell line as the effector cell. When applied to commercially available anti-EBOV GP monoclonal antibodies, the assay clearly differentiated antibody with high ADCC activity from those with low or no ADCC activity. Anti-EBOV ADCC activity was also detected in plasma samples from rhesus macaques immunized with a candidate Ebola vaccine. The Ebola ADCC assay reported here will be a useful tool in studying the functionality of anti-EBOV GP antibodies elicited by Ebola vaccines in ongoing and future clinical trials.
Collapse
Affiliation(s)
- Karnail Singh
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Bishal Marasini
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Xuemin Chen
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Paul Spearman
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.
| |
Collapse
|
54
|
Freeman CL, Sehn LH. A tale of two antibodies: obinutuzumabversusrituximab. Br J Haematol 2018; 182:29-45. [DOI: 10.1111/bjh.15232] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Ciara L. Freeman
- Centre for Lymphoid Cancer; British Columbia Cancer and the University of British Columbia; Vancouver BC Canada
| | - Laurie H. Sehn
- Centre for Lymphoid Cancer; British Columbia Cancer and the University of British Columbia; Vancouver BC Canada
| |
Collapse
|
55
|
Schmidt S, Tramsen L, Rais B, Ullrich E, Lehrnbecher T. Natural killer cells as a therapeutic tool for infectious diseases - current status and future perspectives. Oncotarget 2018; 9:20891-20907. [PMID: 29755697 PMCID: PMC5945539 DOI: 10.18632/oncotarget.25058] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/21/2018] [Indexed: 12/12/2022] Open
Abstract
Natural Killer (NK) cells are involved in the host immune response against infections due to viral, bacterial and fungal pathogens, all of which are a significant cause of morbidity and mortality in immunocompromised patients. Since the recovery of the immune system has a major impact on the outcome of an infectious complication, there is major interest in strengthening the host response in immunocompromised patients, either by using cytokines or growth factors or by adoptive cellular therapies transfusing immune cells such as granulocytes or pathogen-specific T-cells. To date, relatively little is known about the potential of adoptively transferring NK cells in immunocompromised patients with infectious complications, although the anti-cancer property of NK cells is already being investigated in the clinical setting. This review will focus on the antimicrobial properties of NK cells and the current standing and future perspectives of generating and using NK cells as immunotherapy in patients with infectious complications, an approach which is promising and might have an important clinical impact in the future.
Collapse
Affiliation(s)
- Stanislaw Schmidt
- Division for Pediatric Hematology and Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Lars Tramsen
- Division for Pediatric Hematology and Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Bushra Rais
- Division of Stem Cell Transplantation and Immunology, Laboratory for Cellular Immunology, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany.,LOEWE Center for Cell and Gene Therapy, Cellular Immunology, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Evelyn Ullrich
- Division of Stem Cell Transplantation and Immunology, Laboratory for Cellular Immunology, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany.,LOEWE Center for Cell and Gene Therapy, Cellular Immunology, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Thomas Lehrnbecher
- Division for Pediatric Hematology and Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany.,LOEWE Center for Cell and Gene Therapy, Cellular Immunology, Johann Wolfgang Goethe University, Frankfurt, Germany
| |
Collapse
|
56
|
Hassenrück F, Knödgen E, Göckeritz E, Midda SH, Vondey V, Neumann L, Herter S, Klein C, Hallek M, Krause G. Sensitive Detection of the Natural Killer Cell-Mediated Cytotoxicity of Anti-CD20 Antibodies and Its Impairment by B-Cell Receptor Pathway Inhibitors. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1023490. [PMID: 29750146 PMCID: PMC5884282 DOI: 10.1155/2018/1023490] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 02/08/2018] [Accepted: 02/11/2018] [Indexed: 11/17/2022]
Abstract
The antibody-dependent cell-mediated cytotoxicity (ADCC) of the anti-CD20 monoclonal antibodies (mAbs) rituximab and obinutuzumab against the cell line Raji and isolated CLL cells and its potential impairment by kinase inhibitors (KI) was determined via lactate dehydrogenase release or calcein retention, respectively, using genetically modified NK92 cells expressing CD16-176V as effector cells. Compared to peripheral blood mononuclear cells, recombinant effector cell lines showed substantial alloreactivity-related cytotoxicity without addition of mAbs but afforded determination of ADCC with reduced interassay variability. The cytotoxicity owing to alloreactivity was less susceptible to interference by KI than the ADCC of anti-CD20 mAbs, which was markedly diminished by ibrutinib, but not by idelalisib. Compared to rituximab, the ADCC of obinutuzumab against primary CLL cells showed approximately 30% higher efficacy and less interference with KI. Irreversible BTK inhibitors at a clinically relevant concentration of 1 μM only weakly impaired the ADCC of anti-CD20 mAbs, with less influence in combinations with obinutuzumab than with rituximab and by acalabrutinib than by ibrutinib or tirabrutinib. In summary, NK cell line-based assays permitted the sensitive detection of ADCC of therapeutic anti-CD20 mAbs against CLL cells and of the interference of KI with this important killing mechanism.
Collapse
MESH Headings
- Adenine/analogs & derivatives
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Antigens, CD20/metabolism
- B-Lymphocytes/drug effects
- Benzamides/pharmacology
- Cell Line, Tumor
- Cytotoxins/pharmacology
- Humans
- Killer Cells, Natural/drug effects
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- Piperidines
- Protein Kinase Inhibitors/pharmacology
- Purines/pharmacology
- Pyrazines/pharmacology
- Pyrazoles/pharmacology
- Pyrimidines/pharmacology
- Quinazolinones/pharmacology
- Receptors, Antigen, B-Cell/metabolism
- Rituximab/pharmacology
Collapse
Affiliation(s)
- Floyd Hassenrück
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Eva Knödgen
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Elisa Göckeritz
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Safi Hasan Midda
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Verena Vondey
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Lars Neumann
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Sylvia Herter
- Roche Innovation Center Zurich, Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Christian Klein
- Roche Innovation Center Zurich, Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Michael Hallek
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| | - Günter Krause
- Department I of Internal Medicine, University of Cologne, Center of Integrated Oncology Cologne Bonn, Kerpener Str. 62, 50931 Köln, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD Cologne), Cologne, Germany
| |
Collapse
|
57
|
Adotevi O, Godet Y, Galaine J, Lakkis Z, Idirene I, Certoux JM, Jary M, Loyon R, Laheurte C, Kim S, Dormoy A, Pouthier F, Barisien C, Fein F, Tiberghien P, Pivot X, Valmary-Degano S, Ferrand C, Morel P, Delabrousse E, Borg C. In situ delivery of allogeneic natural killer cell (NK) combined with Cetuximab in liver metastases of gastrointestinal carcinoma: A phase I clinical trial. Oncoimmunology 2018; 7:e1424673. [PMID: 29721386 DOI: 10.1080/2162402x.2018.1424673] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/16/2017] [Accepted: 01/03/2018] [Indexed: 01/08/2023] Open
Abstract
Despite successful introduction of NK-based cellular therapy in the treatment of myeloid leukemia, the potential use of NK alloreactivity in solid malignancies is still elusive. We performed a phase I clinical trial to assess the safety and efficacy of in situ delivery of allogeneic NK cells combined with cetuximab in liver metastasis of gastrointestinal origin. The conditioning chemotherapy was administrated before the allogeneic NK cells injection via hepatic artery. Three escalating doses were tested (3.106, 8.106 and 12.106 NK cells/kg) following by a high-dose interleukin-2 (IL-2). Cetuximab was administered intravenously every week for 7 weeks. Nine patients with liver metastases of colorectal or pancreatic cancers were included, three per dose level. Hepatic artery injection was successfully performed in all patients with no report of dose-limiting toxicity. Two patients had febrile aplasia requiring a short-term antibiotherapy. Grade 3/4 anemia and thrombopenia were also observed related to the chemotherapy. Objective clinical responses were documented in 3 patients and among them 2 occurred in patients injected with cell products harboring two KIR ligand mismatches and one in a patient with one KIR ligand mismatch. Immune monitoring revealed that most patients presented an increase but transient of IL-15 and IL-7 cytokines levels one week after chemotherapy. Furthermore, a high expansion of FoxP3+regulatory T cells and PD-1+ T cells was observed in all patients, related to IL-2 administration. Our results demonstrated that combining allogeneic NK cells transfer via intra-hepatic artery, cetuximab and a high-dose IL-2 is feasible, well tolerated and may result in clinical responses.
Collapse
Affiliation(s)
- O Adotevi
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, F-25000 Besançon, France
| | - Y Godet
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - J Galaine
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - Z Lakkis
- University Hospital of Besançon, Department of Gastrointestinal and liver surgery, F-25000 Besançon, France
| | - I Idirene
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - J M Certoux
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - M Jary
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, F-25000 Besançon, France
| | - R Loyon
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - C Laheurte
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,Etablissement Français du Sang Bourgogne Franche-Comté, plateforme de BioMonitoring, F-25000 Besançon, France.,INSERM CIC-1431, University Hospital of Besançon, Clinical Investigation Center in Biotherapy, F-25000, Besançon, France
| | - S Kim
- University Hospital of Besançon, Department of Medical Oncology, F-25000 Besançon, France
| | - A Dormoy
- Etablissement Français du Sang Bourgogne Franche-Comté, F-25000 Besançon, France
| | - F Pouthier
- Etablissement Français du Sang Bourgogne Franche-Comté, F-25000 Besançon, France
| | - C Barisien
- Etablissement Français du Sang Bourgogne Franche-Comté, F-25000 Besançon, France
| | - F Fein
- University Hospital of Besançon, Department of Gastroenterology, F-25000 Besançon, France
| | - P Tiberghien
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - X Pivot
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, F-25000 Besançon, France
| | - S Valmary-Degano
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,University Hospital of Besançon, Department of Pathology, F-25000 Besançon, France
| | - C Ferrand
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - P Morel
- Etablissement Français du Sang Bourgogne Franche-Comté, F-25000 Besançon, France
| | - E Delabrousse
- University Hospital of Besançon, Department of Radiology, F-25000, Besançon, France
| | - C Borg
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, F-25000 Besançon, France
| |
Collapse
|
58
|
Salazar N, Carlson JC, Huang K, Zheng Y, Oderup C, Gross J, Jang AD, Burke TM, Lewén S, Scholz A, Huang S, Nease L, Kosek J, Mittelbronn M, Butcher EC, Tu H, Zabel BA. A Chimeric Antibody against ACKR3/CXCR7 in Combination with TMZ Activates Immune Responses and Extends Survival in Mouse GBM Models. Mol Ther 2018; 26:1354-1365. [PMID: 29606504 PMCID: PMC5993942 DOI: 10.1016/j.ymthe.2018.02.030] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 02/21/2018] [Accepted: 02/27/2018] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma (GBM) is the least treatable type of brain tumor, afflicting over 15,000 people per year in the United States. Patients have a median survival of 16 months, and over 95% die within 5 years. The chemokine receptor ACKR3 is selectively expressed on both GBM cells and tumor-associated blood vessels. High tumor expression of ACKR3 correlates with poor prognosis and potential treatment resistance, making it an attractive therapeutic target. We engineered a single chain FV-human FC-immunoglobulin G1 (IgG1) antibody, X7Ab, to target ACKR3 in human and mouse GBM cells. We used hydrodynamic gene transfer to overexpress the antibody, with efficacy in vivo. X7Ab kills GBM tumor cells and ACKR3-expressing vascular endothelial cells by engaging the cytotoxic activity of natural killer (NK) cells and complement and the phagocytic activity of macrophages. Combining X7Ab with TMZ allows the TMZ dosage to be lowered, without compromising therapeutic efficacy. Mice treated with X7Ab and in combination with TMZ showed significant tumor reduction by MRI and longer survival overall. Brain-tumor-infiltrating leukocyte analysis revealed that X7Ab enhances the activation of M1 macrophages to support anti-tumor immune response in vivo. Targeting ACKR3 with immunotherapeutic monoclonal antibodies (mAbs) in combination with standard of care therapies may prove effective in treating GBM.
Collapse
Affiliation(s)
- Nicole Salazar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Jeffrey C Carlson
- Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | | | - Yayue Zheng
- Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Cecilia Oderup
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Julia Gross
- Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Andrew D Jang
- Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Thomas M Burke
- Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Susanna Lewén
- Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Alexander Scholz
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Serina Huang
- Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Leona Nease
- Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Jon Kosek
- Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Michel Mittelbronn
- Institute of Neurology, Edinger Institute, Frankfurt, Germany; Luxembourg Centre of Neuropathology (LCNP), Luxembourg City, Luxembourg; Department of Pathology, Laboratoire National de Santé, Dudelange, Luxembourg; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Eugene C Butcher
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA
| | - Hua Tu
- LakePharma Inc., Belmont, CA, USA
| | - Brian A Zabel
- Palo Alto Veterans Institute for Research (PAVIR), Veterans Affairs Palo Alto Health Care System (VAPAHCS), Palo Alto, CA, USA.
| |
Collapse
|
59
|
Mahaweni NM, Bos GMJ, Mitsiades CS, Tilanus MGJ, Wieten L. Daratumumab augments alloreactive natural killer cell cytotoxicity towards CD38+ multiple myeloma cell lines in a biochemical context mimicking tumour microenvironment conditions. Cancer Immunol Immunother 2018; 67:861-872. [PMID: 29500635 PMCID: PMC5951903 DOI: 10.1007/s00262-018-2140-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 02/22/2018] [Indexed: 12/16/2022]
Abstract
Natural killer (NK) cell-based immunotherapy is a promising novel approach to treat cancer. However, NK cell function has been shown to be potentially diminished by factors common in the tumor microenvironment (TME). In this study, we assessed the synergistic potential of antibody-dependent cell-mediated cytotoxicity (ADCC) and killer immunoglobin-like receptor (KIR)-ligand mismatched NK cells to potentiate NK cell antitumor reactivity in multiple myeloma (MM). Hypoxia, lactate, prostaglandin E2 (PGE2) or combinations were selected to mimic the TME. To investigate this, NK cells from healthy donors were isolated and NK cell ADCC capacity in response to MM cells was assessed in flow cytometry-based cytotoxicity and degranulation (CD107a) assays in the presence of TME factors. Hypoxia, lactate and PGE2 reduced cytotoxicity of NK cells against myeloma target cells. The addition of daratumumab (anti-CD38 antibody) augmented NK-cell cytotoxicity against target cells expressing high CD38, but not against CD38 low or negative target cells also in the presence of TME. Co-staining for inhibitory KIRs and NKG2A demonstrated that daratumumab enhanced degranulation of all NK cell subsets. Nevertheless, KIR-ligand mismatched NK cells were slightly better effector cells than KIR-ligand matched NK cells. In summary, our study shows that combination therapy using strategies to maximize activating NK cell signaling by triggering ADCC in combination with an approach to minimize inhibitory signaling through a selection of KIR-ligand mismatched donors, can help to overcome the NK-suppressive TME. This can serve as a platform to improve the clinical efficacy of NK cells.
Collapse
Affiliation(s)
- Niken M Mahaweni
- Division of Hematology, Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Transplantation Immunology, Tissue Typing Laboratory, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Gerard M J Bos
- Division of Hematology, Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Marcel G J Tilanus
- Department of Transplantation Immunology, Tissue Typing Laboratory, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Lotte Wieten
- Department of Transplantation Immunology, Tissue Typing Laboratory, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
| |
Collapse
|
60
|
Abstract
A group of impressive immunotherapies for cancer treatment, including immune checkpoint-blocking antibodies, gene therapy and immune cell adoptive cellular immunotherapy, have been established, providing new weapons to fight cancer. Natural killer (NK) cells are a component of the first line of defense against tumors and virus infections. Studies have shown dysfunctional NK cells in patients with cancer. Thus, restoring NK cell antitumor functionality could be a promising therapeutic strategy. NK cells that are activated and expanded ex vivo can supplement malfunctional NK cells in tumor patients. Therapeutic antibodies, chimeric antigen receptor (CAR), or bispecific proteins can all retarget NK cells precisely to tumor cells. Therapeutic antibody blockade of the immune checkpoints of NK cells has been suggested to overcome the immunosuppressive signals delivered to NK cells. Oncolytic virotherapy provokes antitumor activity of NK cells by triggering antiviral immune responses. Herein, we review the current immunotherapeutic approaches employed to restore NK cell antitumor functionality for the treatment of cancer.
Collapse
Affiliation(s)
- Yangxi Li
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei 230027, China
| | - Rui Sun
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
61
|
|
62
|
de Vries RD, Nieuwkoop NJ, van der Klis FRM, Koopmans MPG, Krammer F, Rimmelzwaan GF. Primary Human Influenza B Virus Infection Induces Cross-Lineage Hemagglutinin Stalk-Specific Antibodies Mediating Antibody-Dependent Cellular Cytoxicity. J Infect Dis 2017; 217:3-11. [PMID: 29294018 PMCID: PMC5853962 DOI: 10.1093/infdis/jix546] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/04/2017] [Accepted: 10/09/2017] [Indexed: 11/13/2022] Open
Abstract
Influenza A virus (IAV) and influenza B virus (IBV) cause substantial morbidity and mortality during annual epidemics. Two distinct lineages of IBV are distinguished, based on variation in hemagglutinin (HA): B/Victoria/2/87-like (B/Vic) and B/Yamagata/16/88-like (B/Yam). Here, we show that, in humans, primary IBV infection with either lineage induces HA-specific antibody-dependent cellular cytotoxicity (ADCC)-mediating antibodies. IBV infection induced antibodies specific to the HA head and stalk, but only HA stalk-specific antibodies mediated ADCC efficiently and displayed cross-reactivity with IBV of both lineages. This corresponds to recent findings that 2 points of contact between the effector and target cell (ie, HA and sialic acid, respectively, and the fragment crystallizable [Fc] domain and Fcγ receptor IIIα, respectively) are required for efficient ADCC activity and that antibodies specific for the receptor-binding site located in the head domain of HA therefore fail to mediate ADCC. Potentially, ADCC-mediating antibodies directed to the HA stalk of IBV contribute to cross-protective immunity to IBV of both lineages.
Collapse
Affiliation(s)
- Rory D de Vries
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Nella J Nieuwkoop
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Fiona R M van der Klis
- Center for Infectious Disease Control, National Institute of Public Health and the Environment, Bilthoven, the Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Guus F Rimmelzwaan
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
63
|
Sung AP, Tang JJJ, Guglielmo MJ, Redelman D, Smith-Gagen J, Bateman L, Hudig D. An improved method to quantify human NK cell-mediated antibody-dependent cell-mediated cytotoxicity (ADCC) per IgG FcR-positive NK cell without purification of NK cells. J Immunol Methods 2017; 452:63-72. [PMID: 29113954 DOI: 10.1016/j.jim.2017.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 10/31/2017] [Accepted: 11/02/2017] [Indexed: 12/12/2022]
Abstract
Natural killer (NK) lymphocyte ADCC supports anti-viral protection and monoclonal antibody (mAb) anti-tumor therapies. To predict in vivo ADCC therapeutic responses of different individuals, measurement of both ADCC cellular lytic capacity and their NK cellular receptor recognition of antibodies on 'target' cells are needed, using clinically available amounts of blood. Twenty ml of blood provides sufficient peripheral blood mononuclear cells (PBMCs) for the new assay for lytic capacity described here and for an antibody EC50 assay for Fc-receptor recognition. For the lytic capacity assay, we employed flow cytometry to quantify the CD16A IgG Fc-receptor positive NK effector cells from PBMCs to avoid loss of NKs during isolation. Targets were 51Cr-labeled Daudi B cells pretreated with excess obinutuzumab type 2 anti-CD20 mAb and washed; remaining free mAb was insufficient to convert B cells in the PBMCs into 'targets'. We calculated: the percentage Daudis killed at a 1:1 ratio of CD16A-positive NK cells to Daudis (CX1:1); lytic slopes; and ADCC50 lytic units. Among 27 donors, we detected wide ranges in CX1:1 (16-73% targets killed) and in lytic slopes. Slope variations prevented application of lytic units. We recommend CX1:1 to compare individuals' ADCC capacity. CX1:1 was similar for purified NK cells vs. PBMCs and independent of CD16A V & F genotypes and antibody EC50s. With high mAb bound onto targets and the high affinity of obinutuzumab Fc for CD16A, CX1:1 measurements discern ADCC lytic capacity rather than antibody recognition. This assay allows ADCC to be quantified without NK cell isolation and avoids distortion associated with lytic units.
Collapse
Affiliation(s)
- Alexander P Sung
- University of Nevada Reno School of Medicine, Department of Microbiology and Immunology Reno, Nevada, 1664 N. Virginia St., Reno, NV 89557, United States
| | - Jennifer J-J Tang
- University of Nevada Reno School of Medicine, Department of Microbiology and Immunology Reno, Nevada, 1664 N. Virginia St., Reno, NV 89557, United States
| | - Michael J Guglielmo
- University of Nevada Reno School of Medicine, Department of Microbiology and Immunology Reno, Nevada, 1664 N. Virginia St., Reno, NV 89557, United States
| | - Doug Redelman
- University of Nevada Reno School of Medicine, Department of Physiology and Cell Biology Reno, Nevada, 1664 N. Virginia St., Reno, NV 89557, United States
| | - Julie Smith-Gagen
- University of Nevada Reno School of Medicine, School of Community Health Sciences Reno, Nevada, 1664 N. Virginia St., Reno, NV 89557, United States
| | - Lucinda Bateman
- Bateman Horne Center, Salt Lake City, UT 84102, United States
| | - Dorothy Hudig
- University of Nevada Reno School of Medicine, Department of Microbiology and Immunology Reno, Nevada, 1664 N. Virginia St., Reno, NV 89557, United States.
| |
Collapse
|
64
|
Tóth G, Szöllősi J, Vereb G. Quantitating ADCC against adherent cells: Impedance-based detection is superior to release, membrane permeability, or caspase activation assays in resolving antibody dose response. Cytometry A 2017; 91:1021-1029. [DOI: 10.1002/cyto.a.23247] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 08/13/2017] [Accepted: 09/02/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Gábor Tóth
- Department of Biophysics and Cell Biology; University of Debrecen; Debrecen Hungary
| | - János Szöllősi
- Department of Biophysics and Cell Biology; University of Debrecen; Debrecen Hungary
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine; University of Debrecen; Debrecen Hungary
| | - György Vereb
- Department of Biophysics and Cell Biology; University of Debrecen; Debrecen Hungary
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine; University of Debrecen; Debrecen Hungary
- Faculty of Pharmacy; University of Debrecen; Debrecen Hungary
| |
Collapse
|
65
|
Monoclonal Antibodies, Derived from Humans Vaccinated with the RV144 HIV Vaccine Containing the HVEM Binding Domain of Herpes Simplex Virus (HSV) Glycoprotein D, Neutralize HSV Infection, Mediate Antibody-Dependent Cellular Cytotoxicity, and Protect Mice from Ocular Challenge with HSV-1. J Virol 2017; 91:JVI.00411-17. [PMID: 28701403 DOI: 10.1128/jvi.00411-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/29/2017] [Indexed: 01/29/2023] Open
Abstract
The RV144 HIV vaccine trial included a recombinant HIV glycoprotein 120 (gp120) construct fused to a small portion of herpes simplex virus 1 (HSV-1) glycoprotein D (gD) so that the first 40 amino acids of gp120 were replaced by the signal sequence and the first 27 amino acids of the mature form of gD. This region of gD contains most of the binding site for HVEM, an HSV receptor important for virus infection of epithelial cells and lymphocytes. RV144 induced antibodies to HIV that were partially protective against infection, as well as antibodies to HSV. We derived monoclonal antibodies (MAbs) from peripheral blood B cells of recipients of the RV144 HIV vaccine and showed that these antibodies neutralized HSV-1 infection in cells expressing HVEM, but not the other major virus receptor, nectin-1. The MAbs mediated antibody-dependent cellular cytotoxicity (ADCC), and mice that received the MAbs and were then challenged by corneal inoculation with HSV-1 had reduced eye disease, shedding, and latent infection. To our knowledge, this is the first description of MAbs derived from human recipients of a vaccine that specifically target the HVEM binding site of gD. In summary, we found that monoclonal antibodies derived from humans vaccinated with the HVEM binding domain of HSV-1 gD (i) neutralized HSV-1 infection in a cell receptor-specific manner, (ii) mediated ADCC, and (iii) reduced ocular disease in virus-infected mice.IMPORTANCE Herpes simplex virus 1 (HSV-1) causes cold sores and neonatal herpes and is a leading cause of blindness. Despite many trials, no HSV vaccine has been approved. Nectin-1 and HVEM are the two major cellular receptors for HSV. These receptors are expressed at different levels in various tissues, and the role of each receptor in HSV pathogenesis is not well understood. We derived human monoclonal antibodies from persons who received the HIV RV144 vaccine that contained the HVEM binding domain of HSV-1 gD fused to HIV gp120. These antibodies were able to specifically neutralize HSV-1 infection in vitro via HVEM. Furthermore, we showed for the first time that HVEM-specific HSV-1 neutralizing antibodies protect mice from HSV-1 eye disease, indicating the critical role of HVEM in HSV-1 ocular infection.
Collapse
|
66
|
Messaoudene M, Frazao A, Gavlovsky PJ, Toubert A, Dulphy N, Caignard A. Patient's Natural Killer Cells in the Era of Targeted Therapies: Role for Tumor Killers. Front Immunol 2017; 8:683. [PMID: 28659921 PMCID: PMC5466965 DOI: 10.3389/fimmu.2017.00683] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/26/2017] [Indexed: 12/12/2022] Open
Abstract
Natural killer (NK) cells are potent antitumor effectors, involved in hematological malignancies and solid tumor immunosurveillance. They infiltrate various solid tumors, and their numbers are correlated with good outcome. The function of NK cells extends their lytic capacities toward tumor cells expressing stress-induced ligands, through secretion of immunoregulatory cytokines, and interactions with other immune cells. Altered NK cell function due to tumor immune escape is frequent in advanced tumors; however, strategies to release the function of NK infiltrating tumors are emerging. Recent therapies targeting specific oncogenic mutations improved the treatment of cancer patients, but patients often relapse. The actual development consists in combined therapeutic strategies including agents targeting the proliferation of tumor cells and others restorating functional antitumor immune effectors for efficient and durable efficacy of anticancer treatment. In that context, we discuss the recent results of the literature to propose hypotheses concerning the potential use of NK cells, potent antitumor cytotoxic effectors, to design novel antitumor strategies.
Collapse
Affiliation(s)
- Meriem Messaoudene
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France
| | - Alexandra Frazao
- INSERM U1160, Institut Universitaire d'Hématologie, Hôpital Saint Louis, Paris, France
| | - Pierre Jean Gavlovsky
- INSERM U1160, Institut Universitaire d'Hématologie, Hôpital Saint Louis, Paris, France
| | - Antoine Toubert
- INSERM U1160, Institut Universitaire d'Hématologie, Hôpital Saint Louis, Paris, France
| | - Nicolas Dulphy
- INSERM U1160, Institut Universitaire d'Hématologie, Hôpital Saint Louis, Paris, France
| | - Anne Caignard
- INSERM U1160, Institut Universitaire d'Hématologie, Hôpital Saint Louis, Paris, France
| |
Collapse
|
67
|
Natural killer cell immunotherapies against cancer: checkpoint inhibitors and more. Semin Immunol 2017; 31:55-63. [DOI: 10.1016/j.smim.2017.08.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/03/2017] [Indexed: 12/17/2022]
|
68
|
Zhang C, Oberoi P, Oelsner S, Waldmann A, Lindner A, Tonn T, Wels WS. Chimeric Antigen Receptor-Engineered NK-92 Cells: An Off-the-Shelf Cellular Therapeutic for Targeted Elimination of Cancer Cells and Induction of Protective Antitumor Immunity. Front Immunol 2017; 8:533. [PMID: 28572802 PMCID: PMC5435757 DOI: 10.3389/fimmu.2017.00533] [Citation(s) in RCA: 221] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 04/21/2017] [Indexed: 12/27/2022] Open
Abstract
Significant progress has been made in recent years toward realizing the potential of natural killer (NK) cells for cancer immunotherapy. NK cells can respond rapidly to transformed and stressed cells and have the intrinsic potential to extravasate and reach their targets in almost all body tissues. In addition to donor-derived primary NK cells, also the established NK cell line NK-92 is being developed for adoptive immunotherapy, and general safety of infusion of irradiated NK-92 cells has been established in phase I clinical trials with clinical responses observed in some of the cancer patients treated. To enhance their therapeutic utility, NK-92 cells have been modified to express chimeric antigen receptors (CARs) composed of a tumor-specific single chain fragment variable antibody fragment fused via hinge and transmembrane regions to intracellular signaling moieties such as CD3ζ or composite signaling domains containing a costimulatory protein together with CD3ζ. CAR-mediated activation of NK cells then bypasses inhibitory signals and overcomes NK resistance of tumor cells. In contrast to primary NK cells, CAR-engineered NK-92 cell lines suitable for clinical development can be established from molecularly and functionally well-characterized single cell clones following good manufacturing practice-compliant procedures. In preclinical in vitro and in vivo models, potent antitumor activity of NK-92 variants targeted to differentiation antigens expressed by hematologic malignancies, and overexpressed or mutated self-antigens associated with solid tumors has been found, encouraging further development of CAR-engineered NK-92 cells. Importantly, in syngeneic mouse tumor models, induction of endogenous antitumor immunity after treatment with CAR-expressing NK-92 cells has been demonstrated, resulting in cures and long-lasting immunological memory protecting against tumor rechallenge at distant sites. Here, we summarize the current status and future prospects of CAR-engineered NK-92 cells as off-the-shelf cellular therapeutics, with special emphasis on ErbB2 (HER2)-specific NK-92 cells that are approaching clinical application.
Collapse
Affiliation(s)
- Congcong Zhang
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pranav Oberoi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Sarah Oelsner
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Anja Waldmann
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Aline Lindner
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Torsten Tonn
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Red Cross Blood Donation Service North-East, Institute for Transfusion Medicine, Dresden, Germany.,Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
| | - Winfried S Wels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
69
|
Lopez-Lastra S, Di Santo JP. Modeling Natural Killer Cell Targeted Immunotherapies. Front Immunol 2017; 8:370. [PMID: 28405194 PMCID: PMC5370275 DOI: 10.3389/fimmu.2017.00370] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/14/2017] [Indexed: 01/01/2023] Open
Abstract
Animal models have extensively contributed to our understanding of human immunobiology and to uncover the underlying pathological mechanisms occurring in the development of diseases. However, mouse models do not reproduce the genetic and molecular complexity inherent in human disease conditions. Human immune system (HIS) mouse models that are susceptible to human pathogens and can recapitulate human hematopoiesis and tumor immunobiology provide one means to bridge the interspecies gap. Natural killer cells are the founding member of the innate lymphoid cell family. They exert a rapid and strong immune response against tumor and pathogen-infected cells. Their antitumor features have long been exploited for therapeutic purposes in the context of cancer. In this review, we detail the development of highly immunodeficient mouse strains and the models currently used in cancer research. We summarize the latest improvements in adoptive natural killer (NK) cell therapies and the development of novel NK cell sources. Finally, we discuss the advantages of HIS mice to study the interactions between human NK cells and human cancers and to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Silvia Lopez-Lastra
- Innate Immunity Unit, Institut Pasteur, Paris, France
- Inserm U1223, Paris, France
- Université Paris-Sud (Paris-Saclay), Paris, France
| | - James P. Di Santo
- Innate Immunity Unit, Institut Pasteur, Paris, France
- Inserm U1223, Paris, France
| |
Collapse
|
70
|
Antibody-dependent cell cytotoxicity: immunotherapy strategies enhancing effector NK cells. Immunol Cell Biol 2017; 95:347-355. [PMID: 28138156 DOI: 10.1038/icb.2017.6] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 01/24/2017] [Accepted: 01/24/2017] [Indexed: 02/07/2023]
Abstract
Antibody-dependent cellular cytotoxicity (ADCC) is a set of mechanisms that target cells coated with IgG antibodies of the proper subclasses (IgG1 in the human) to be the prey of cell-to-cell cytolysis executed by immune cells expressing FcRIIIA (CD16A). These effectors include not only natural killer (NK) cells but also other CD16+ subsets such as monocyte/macrophages, NKT cells or γδ T cells. In cancer therapy, ADCC is exploited by antibodies that selectively recognize proteins on the surface of malignant cells. An approach to enhance antitumor activity is to act on effector cells so they are increased in their numbers or enhanced in their individual (on a cell per cell basis) ADCC performance. This enhancement can be therapeutically attained by cytokines (that is, interleukin (IL)-15, IL-21, IL-18, IL-2); immunostimulatory monoclonal antibodies (that is, anti-CD137, anti-CD96, anti-TIGIT, anti-KIR, anti-PD-1); TLR agonists or by adoptive infusions of ex vivo expanded NK cells which can be genetically engineered to become more efficient effectors. In conjunction with approaches optimizing IgG1 Fc affinity to CD16, acting on effector cells offers hope to achieve synergistic immunotherapy strategies.
Collapse
|
71
|
Donahue RN, Lepone LM, Grenga I, Jochems C, Fantini M, Madan RA, Heery CR, Gulley JL, Schlom J. Analyses of the peripheral immunome following multiple administrations of avelumab, a human IgG1 anti-PD-L1 monoclonal antibody. J Immunother Cancer 2017; 5:20. [PMID: 28239472 PMCID: PMC5320726 DOI: 10.1186/s40425-017-0220-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 02/01/2017] [Indexed: 12/11/2022] Open
Abstract
Background Multiple anti-PD-L1/PD-1 checkpoint monoclonal antibodies (MAb) have shown clear evidence of clinical benefit. All except one have been designed or engineered to omit the possibility to mediate antibody-dependent cell-mediated cytotoxicity (ADCC) as a second potential mode of anti-tumor activity; the reason for this is the concern of lysis of PD-L1 positive immune cells. Avelumab is a fully human IgG1 MAb which has been shown in prior in vitro studies to mediate ADCC versus a range of human tumor cells, and clinical studies have demonstrated anti-tumor activity versus a range of human cancers. This study was designed to investigate the effect on immune cell subsets in the peripheral blood of cancer patients prior to and following multiple administrations of avelumab. Methods One hundred twenty-three distinct immune cell subsets in the peripheral blood of cancer patients (n = 28) in a phase I trial were analyzed by flow cytometry prior to and following one, three, and nine cycles of avelumab. Changes in soluble (s) CD27 and sCD40L in plasma were also evaluated. In vitro studies were also performed to determine if avelumab would mediate ADCC of PBMC. Results No statistically significant changes in any of the 123 immune cell subsets analyzed were observed at any dose level, or number of doses, of avelumab. Increases in the ratio of sCD27:sCD40L were observed, suggesting potential immune activation. Controlled in vitro studies also showed lysis of tumor cells by avelumab versus no lysis of PBMC from five donors. Conclusions These studies demonstrate the lack of any significant effect on multiple immune cell subsets, even those expressing PD-L1, following multiple cycles of avelumab. These results complement prior studies showing anti-tumor effects of avelumab and comparable levels of adverse events with avelumab versus other anti-PD-1/PD-L1 MAbs. These studies provide the rationale to further exploit the potential ADCC mechanism of action of avelumab as well as other human IgG1 checkpoint inhibitors. Trial registration ClinicalTrials.gov identifier: NCT01772004 (first received: 1/14/13; start date: January 2013) and NCT00001846 (first received date: 11/3/99; start date: August 1999). Electronic supplementary material The online version of this article (doi:10.1186/s40425-017-0220-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Renee N Donahue
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| | - Lauren M Lepone
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| | - Italia Grenga
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| | - Caroline Jochems
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| | - Massimo Fantini
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| | - Ravi A Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | - Christopher R Heery
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| | - James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD USA
| |
Collapse
|
72
|
Hoseini SS, Cheung NK. Acute myeloid leukemia targets for bispecific antibodies. Blood Cancer J 2017; 7:e522. [PMID: 28157217 PMCID: PMC5386336 DOI: 10.1038/bcj.2017.2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/22/2016] [Indexed: 12/31/2022] Open
Abstract
Despite substantial gains in our understanding of the genomics of acute myelogenous leukemia (AML), patient survival remains unsatisfactory especially among the older age group. T cell-based therapy of lymphoblastic leukemia is rapidly advancing; however, its application in AML is still lagging behind. Bispecific antibodies can redirect polyclonal effector cells to engage chosen targets on leukemia blasts. When the effector cells are natural-killer cells, both antibody-dependent and antibody-independent mechanisms could be exploited. When the effectors are T cells, direct tumor cytotoxicity can be engaged followed by a potential vaccination effect. In this review, we summarize the AML-associated tumor targets and the bispecific antibodies that have been studied. The potentials and limitations of each of these systems will be discussed.
Collapse
Affiliation(s)
- S S Hoseini
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - N K Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
73
|
Hsu HT, Mace EM, Carisey AF, Viswanath DI, Christakou AE, Wiklund M, Önfelt B, Orange JS. NK cells converge lytic granules to promote cytotoxicity and prevent bystander killing. J Cell Biol 2016; 215:875-889. [PMID: 27903610 PMCID: PMC5166499 DOI: 10.1083/jcb.201604136] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 09/04/2016] [Accepted: 10/28/2016] [Indexed: 12/16/2022] Open
Abstract
Natural killer (NK) cell activation triggers sequential cellular events leading to destruction of diseased cells. We previously identified lytic granule convergence, a dynein- and integrin signal-dependent movement of lysosome-related organelles to the microtubule-organizing center, as an early step in the cell biological process underlying NK cell cytotoxicity. Why lytic granules converge during NK cell cytotoxicity, however, remains unclear. We experimentally controlled the availability of human ligands to regulate NK cell signaling and promote granule convergence with either directed or nondirected degranulation. By the use of acoustic trap microscopy, we generated specific effector-target cell arrangements to define the impact of the two modes of degranulation. NK cells with converged granules had greater targeted and less nonspecific "bystander" killing. Additionally, NK cells in which dynein was inhibited or integrin blocked under physiological conditions demonstrated increased nondirected degranulation and bystander killing. Thus, NK cells converge lytic granules and thereby improve the efficiency of targeted killing and prevent collateral damage to neighboring healthy cells.
Collapse
Affiliation(s)
- Hsiang-Ting Hsu
- Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030
- Department of Pediatrics, Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
| | - Emily M Mace
- Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030
- Department of Pediatrics, Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
| | - Alexandre F Carisey
- Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030
- Department of Pediatrics, Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
| | - Dixita I Viswanath
- Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030
- Rice University, Houston, TX 77005
| | - Athanasia E Christakou
- Department of Applied Physics, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Martin Wiklund
- Department of Applied Physics, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Björn Önfelt
- Department of Applied Physics, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Jordan S Orange
- Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030
- Department of Pediatrics, Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
- Rice University, Houston, TX 77005
| |
Collapse
|
74
|
Burga RA, Nguyen T, Zulovich J, Madonna S, Ylisastigui L, Fernandes R, Yvon E. Improving efficacy of cancer immunotherapy by genetic modification of natural killer cells. Cytotherapy 2016; 18:1410-1421. [PMID: 27421740 DOI: 10.1016/j.jcyt.2016.05.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 11/17/2022]
Abstract
Natural killer (NK) cells are members of the innate immune system that recognize target cells via activating and inhibitory signals received through cell receptors. Derived from the lymphoid lineage, NK cells are able to produce cytokines and exert a cytotoxic effect on viral infected and malignant cells. It is their unique ability to lyse target cells rapidly and without prior education that renders NK cells a promising effector cell for adoptive cell therapy. However, both viruses and tumors employ evasion strategies to avoid attack by NK cells, which represent biological challenges that need to be harnessed to fully exploit the cytolytic potential of NK cells. Using genetic modification, the function of NK cells can be enhanced to improve their homing, cytolytic activity, in vivo persistence and safety. Examples include gene modification to express chemokine, high-affinity Fc receptor and chimeric antigen receptors, suicide genes and the forced expression of cytokines such as interleukin (IL)-2 and IL-15. Preclinical studies have clearly demonstrated that such approaches are effective in improving NK-cell function, homing and safety. In this review, we summarize the recent advances in the genetic manipulations of NK cells and their application for cellular immunotherapeutic strategies.
Collapse
Affiliation(s)
- Rachel A Burga
- Institute for Biomedical Sciences, The George Washington University, Washington, DC, USA; Children's National Health System, Washington, DC, USA
| | - Tuongvan Nguyen
- The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jane Zulovich
- The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Sarah Madonna
- The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Loyda Ylisastigui
- The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Rohan Fernandes
- Institute for Biomedical Sciences, The George Washington University, Washington, DC, USA; Children's National Health System, Washington, DC, USA
| | - Eric Yvon
- The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
75
|
Tóth G, Szöőr Á, Simon L, Yarden Y, Szöllősi J, Vereb G. The combination of trastuzumab and pertuzumab administered at approved doses may delay development of trastuzumab resistance by additively enhancing antibody-dependent cell-mediated cytotoxicity. MAbs 2016; 8:1361-1370. [PMID: 27380003 PMCID: PMC5058622 DOI: 10.1080/19420862.2016.1204503] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Although the recently concluded CLEOPATRA trial showed clinical benefits of combining trastuzumab and pertuzumab for treating HER2-positive metastatic breast cancer, trastuzumab monotherapy is still the mainstay in adjuvant settings. Since trastuzumab resistance occurs in over half of these cancers, we examined the mechanisms by which treatment of intrinsically trastuzumab-resistant and -sensitive tumors can benefit from the combination of these antibodies. F(ab′)2 of both trastuzumab and pertuzumab were generated and validated in order to separately analyze antibody-dependent cell-mediated cytotoxicity (ADCC)-based and direct biological effects of the antibodies. Compared to monotherapy, combination of the two antibodies at clinically permitted doses enhanced the recruitment of natural killer cells responsible for ADCC, and significantly delayed the outgrowth of xenografts from intrinsically trastuzumab-resistant JIMT-1 cells. Antibody dose-response curves of in vitro ADCC showed that antibody-mediated killing can be saturated, and the two antibodies exert an additive effect at sub-saturation doses. Thus, the additive effect in vivo indicates that therapeutic tissue levels likely do not saturate ADCC. Additionally, isobole studies with the in vitro trastuzumab-sensitive BT-474 cells showed that the direct biological effect of combined treatment is additive, and surpasses the maximum effect of either monotherapy. Our results suggest the combined therapy is expected to give results that are superior to monotherapy, whatever the type of HER2-positive tumor may be. The combination of both antibodies at maximum clinically approved doses should thus be administered to patients to recruit maximum ADCC and cause maximum direct biological growth inhibition.
Collapse
Affiliation(s)
- Gábor Tóth
- a Department of Biophysics and Cell Biology , Faculty of Medicine, University of Debrecen , Debrecen , Hungary
| | - Árpád Szöőr
- a Department of Biophysics and Cell Biology , Faculty of Medicine, University of Debrecen , Debrecen , Hungary
| | - László Simon
- a Department of Biophysics and Cell Biology , Faculty of Medicine, University of Debrecen , Debrecen , Hungary
| | - Yosef Yarden
- b Department of Biological Regulation , The Weizmann Institute of Science , Rehovot , Israel
| | - János Szöllősi
- a Department of Biophysics and Cell Biology , Faculty of Medicine, University of Debrecen , Debrecen , Hungary.,c MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine
| | - György Vereb
- a Department of Biophysics and Cell Biology , Faculty of Medicine, University of Debrecen , Debrecen , Hungary.,c MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine.,d Faculty of Pharmacy, University of Debrecen , Debrecen , Hungary
| |
Collapse
|
76
|
Combination of NK Cells and Cetuximab to Enhance Anti-Tumor Responses in RAS Mutant Metastatic Colorectal Cancer. PLoS One 2016; 11:e0157830. [PMID: 27314237 PMCID: PMC4912059 DOI: 10.1371/journal.pone.0157830] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 06/06/2016] [Indexed: 01/28/2023] Open
Abstract
The ability of Natural Killer (NK) cells to kill tumor targets has been extensively studied in various hematological malignancies. However, NK cell therapy directed against solid tumors is still in early development. Epidermal Growth Factor Receptor (EGFR) targeted therapies using monoclonal antibodies (mAbs) such as cetuximab and panitumumab are widely used for the treatment of metastatic colorectal cancer (mCRC). Still, the clinical efficacy of this treatment is hampered by mutations in RAS gene, allowing tumors to escape from anti-EGFR mAb therapy. It is well established that NK cells kill tumor cells by natural cytotoxicity and can in addition be activated upon binding of IgG1 mAbs through Fc receptors (CD16/FcγRIIIa) on their surface, thereby mediating antibody dependent cellular cytotoxicity (ADCC). In the current study, activated Peripheral Blood NK cells (PBNK) were combined with anti-EGFR mAbs to study their effect on the killing of EGFR+/- cancer cell lines, including those with RAS mutations. In vitro cytotoxicity experiments using colon cancer primary tumors and cell lines COLO320, Caco-2, SW620, SW480 and HT-29, demonstrated that PBNK cells are cytotoxic for a range of tumor cells, regardless of EGFR, RAS or BRAF status and at low E:T ratios. Cetuximab enhanced the cytotoxic activity of NK cells on EGFR+ tumor cells (either RASwt, RASmut or BRAFmut) in a CD16 dependent manner, whereas it could not increase the killing of EGFR- COLO320. Our study provides a rationale to strengthen NK cell immunotherapy through a combination with cetuximab for RAS and BRAF mutant mCRC patients.
Collapse
|
77
|
Antibody Responses with Fc-Mediated Functions after Vaccination of HIV-Infected Subjects with Trivalent Influenza Vaccine. J Virol 2016; 90:5724-5734. [PMID: 27053553 DOI: 10.1128/jvi.00285-16] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/30/2016] [Indexed: 01/24/2023] Open
Abstract
UNLABELLED This study seeks to assess the ability of seasonal trivalent inactivated influenza vaccine (TIV) to induce nonneutralizing antibodies (Abs) with Fc-mediated functions in HIV-uninfected and HIV-infected subjects. Functional influenza-specific Ab responses were studied in 30 HIV-negative and 27 HIV-positive subjects immunized against seasonal influenza. All 57 subjects received the 2015 TIV. Fc-mediated antihemagglutinin (anti-HA) Ab activity was measured in plasma before and 4 weeks after vaccination using Fc-receptor-binding assays, NK cell activation assays, and phagocytosis assays. At baseline, the HIV-positive group had detectable but reduced functional Ab responses to both vaccine and nonvaccine influenza antigens. TIV enhanced Fc-mediated Ab responses in both HIV-positive and HIV-negative groups. A larger rise was generally observed in the HIV-positive group, such that there was no difference in functional Ab responses between the two groups after vaccination. The 2015 TIV enhanced functional influenza-specific Ab responses in both HIV-negative and HIV-positive subjects to a range of influenza HA proteins. The increase in functional Ab responses in the HIV-positive group supports recommendations to immunize this at-risk group. IMPORTANCE Infection with HIV is associated with increasing disease severity following influenza infections, and annual influenza vaccinations are recommended for this target group. However, HIV-infected individuals respond relatively poorly to vaccination compared to healthy individuals, particularly if immunodeficient. There is therefore a need to increase our understanding of immunity to influenza in the context of underlying HIV infection. While antibodies can mediate direct virus neutralization, interactions with cellular Fc receptors may be important for anti-influenza immunity in vivo by facilitating antibody-dependent cellular cytotoxicity (ADCC) and/or antibody-dependent phagocytosis (ADP). The ability of seasonal influenza vaccines to induce antibody responses with potent Fc-mediated antiviral activity is currently unclear. Probing the ADCC and ADP responses to influenza vaccination has provided important new information in the quest to improve immunity to influenza.
Collapse
|
78
|
Donor KIR B Genotype Improves Progression-Free Survival of Non-Hodgkin Lymphoma Patients Receiving Unrelated Donor Transplantation. Biol Blood Marrow Transplant 2016; 22:1602-1607. [PMID: 27220262 DOI: 10.1016/j.bbmt.2016.05.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/13/2016] [Indexed: 11/20/2022]
Abstract
Donor killer immunoglobulin-like receptor (KIR) genotypes are associated with relapse protection and survival after allotransplantation for acute myelogenous leukemia. We examined the possibility of a similar effect in a cohort of 614 non-Hodgkin lymphoma (NHL) patients receiving unrelated donor (URD) T cell-replete marrow or peripheral blood grafts. Sixty-four percent (n = 396) of donor-recipient pairs were 10/10 allele HLA matched and 26% were 9/10 allele matched. Seventy percent of donors had KIR B/x genotype; the others had KIR A/A genotype. NHL patients receiving 10/10 HLA-matched URD grafts with KIR B/x donors experienced significantly lower relapse at 5 years (26%; 95% confidence interval [CI], 21% to 32% versus 37%; 95% CI, 27% to 46%; P = .05) compared with KIR A/A donors, resulting in improved 5-year progression-free survival (PFS) (35%; 95% CI, 26% to 44% versus 22%; 95% CI, 11% to 35%; P = .007). In multivariate analysis, use of KIR B/x donors was associated with significantly reduced relapse risk (relative risk [RR], .63, P = .02) and improved PFS (RR, .71, P = .008). The relapse protection afforded by KIR B/x donors was not observed in HLA-mismatched transplantations and was not specific to any particular KIR-B gene. Selecting 10/10 HLA-matched and KIR B/x donors should benefit patients with NHL receiving URD allogeneic transplantation.
Collapse
|
79
|
Zhong W, Liu F, Wilson JR, Holiday C, Li ZN, Bai Y, Tzeng WP, Stevens J, York IA, Levine MZ. Antibody-Dependent Cell-Mediated Cytotoxicity to Hemagglutinin of Influenza A Viruses After Influenza Vaccination in Humans. Open Forum Infect Dis 2016; 3:ofw102. [PMID: 27419174 PMCID: PMC4943536 DOI: 10.1093/ofid/ofw102] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/16/2016] [Indexed: 11/13/2022] Open
Abstract
Background. Detection of neutralizing antibodies (nAbs) to influenza A virus hemagglutinin (HA) antigens by conventional serological assays is currently the main immune correlate of protection for influenza vaccines However, current prepandemic avian influenza vaccines are poorly immunogenic in inducing nAbs despite considerable protection conferred. Recent studies show that Ab-dependent cell-mediated cytotoxicity (ADCC) to HA antigens are readily detectable in the sera of healthy individuals and patients with influenza infection. Methods. Virus neutralization and ADCC activities of serum samples from individuals who received either seasonal or a stock-piled H5N1 avian influenza vaccine were evaluated by hemagglutination inhibition assay, microneutralization assay, and an improved ADCC natural killer (NK) cell activation assay. Results. Immunization with inactivated seasonal influenza vaccine led to strong expansion of both nAbs and ADCC-mediating antibodies (adccAbs) to H3 antigen of the vaccine virus in 24 postvaccination human sera. In sharp contrast, 18 individuals vaccinated with the adjuvanted H5N1 avian influenza vaccine mounted H5-specific antibodies with strong ADCC activities despite moderate virus neutralization capacity. Strength of HA-specific ADCC activities is largely associated with the titers of HA-binding antibodies and not with the fine antigenic specificity of anti-HA nAbs. Conclusions. Detection of both nAbs and adccAbs may better reflect protective capacity of HA-specific antibodies induced by avian influenza vaccines.
Collapse
Affiliation(s)
- Weimin Zhong
- Influenza Division , National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Feng Liu
- Influenza Division , National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Jason R Wilson
- Influenza Division , National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Crystal Holiday
- Influenza Division , National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Zhu-Nan Li
- Influenza Division , National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Yaohui Bai
- Influenza Division , National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Wen-Pin Tzeng
- Influenza Division , National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - James Stevens
- Influenza Division , National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Ian A York
- Influenza Division , National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Min Z Levine
- Influenza Division , National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| |
Collapse
|
80
|
Suck G, Linn YC, Tonn T. Natural Killer Cells for Therapy of Leukemia. Transfus Med Hemother 2016; 43:89-95. [PMID: 27226791 DOI: 10.1159/000445325] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/07/2016] [Indexed: 12/18/2022] Open
Abstract
Clinical application of natural killer (NK) cells against leukemia is an area of intense investigation. In human leukocyte antigen-mismatched allogeneic hematopoietic stem cell transplantations (HSCT), alloreactive NK cells exert powerful anti-leukemic activity in preventing relapse in the absence of graft-versus-host disease, particularly in acute myeloid leukemia patients. Adoptive transfer of donor NK cells post-HSCT or in non-transplant scenarios may be superior to the currently widely used unmanipulated donor lymphocyte infusion. This concept could be further improved through transfusion of activated NK cells. Significant progress has been made in good manufacturing practice (GMP)-compliant large-scale production of stimulated effectors. However, inherent limitations remain. These include differing yields and compositions of the end-product due to donor variability and inefficient means for cryopreservation. Moreover, the impact of the various novel activation strategies on NK cell biology and in vivo behavior are barely understood. In contrast, reproduction of the third-party NK-92 drug from a cryostored GMP-compliant master cell bank is straightforward and efficient. Safety for the application of this highly cytotoxic cell line was demonstrated in first clinical trials. This novel 'off-the-shelf' product could become a treatment option for a broad patient population. For specific tumor targeting chimeric-antigen-receptor-engineered NK-92 cells have been designed.
Collapse
Affiliation(s)
- Garnet Suck
- Institute for Transfusion Medicine Berlin, German Red Cross Blood Donation Service North-East, Berlin, Germany
| | - Yeh Ching Linn
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Torsten Tonn
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany; Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
81
|
|
82
|
Dulphy N, Chrétien AS, Khaznadar Z, Fauriat C, Nanbakhsh A, Caignard A, Chouaib S, Olive D, Toubert A. Underground Adaptation to a Hostile Environment: Acute Myeloid Leukemia vs. Natural Killer Cells. Front Immunol 2016; 7:94. [PMID: 27014273 PMCID: PMC4783386 DOI: 10.3389/fimmu.2016.00094] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 02/26/2016] [Indexed: 12/31/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of malignancies which incidence increases with age. The disease affects the differentiation of hematopoietic stem or precursor cells in the bone marrow and can be related to abnormal cytogenetic and/or specific mutational patterns. AML blasts can be sensitive to natural killer (NK) cell antitumor response. However, NK cells are frequently defective in AML patients leading to tumor escape. NK cell defects affect not only the expression of the activating NK receptors, including the natural cytotoxicity receptors, the NK group 2, member D, and the DNAX accessory molecule-1, but also cytotoxicity and IFN-γ release. Such perturbations in NK cell physiology could be related to the adaptation of the AML to the immune pressure and more generally to patient’s clinical features. Various mechanisms are potentially involved in the inhibition of NK-cell functions in AML, including defects in the normal lymphopoiesis, reduced expression of activating receptors through cell-to-cell contacts, and production of immunosuppressive soluble agents by leukemic blasts. Therefore, the continuous cross-talk between AML and NK cells participates to the leukemia immune escape and eventually to patient’s relapse. Methods to restore or stimulate NK cells seem to be attractive strategies to treat patients once the complete remission is achieved. Moreover, our capacity in stimulating the NK cell functions could lead to the development of preemptive strategies to eliminate leukemia-initiating cells before the emergence of the disease in elderly individuals presenting preleukemic mutations in hematopoietic stem cells.
Collapse
Affiliation(s)
- Nicolas Dulphy
- UMRS-1160, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France; U 1160, Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Anne-Sophie Chrétien
- Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Immunité et Cancer, INSERM, U1068, Institut Paoli-Calmettes, Aix-Marseille Université, UM 105, CNRS, UMR7258 , Marseille , France
| | - Zena Khaznadar
- UMRS-1160, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France; U 1160, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Cyril Fauriat
- Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Immunité et Cancer, INSERM, U1068, Institut Paoli-Calmettes, Aix-Marseille Université, UM 105, CNRS, UMR7258 , Marseille , France
| | | | - Anne Caignard
- UMRS-1160, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France; U 1160, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | - Daniel Olive
- Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Immunité et Cancer, INSERM, U1068, Institut Paoli-Calmettes, Aix-Marseille Université, UM 105, CNRS, UMR7258 , Marseille , France
| | - Antoine Toubert
- UMRS-1160, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France; U 1160, Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| |
Collapse
|
83
|
Zhong W, Gross FL, Holiday C, Jefferson SN, Bai Y, Liu F, Katz JM, Levine MZ. Vaccination with 2014-15 Seasonal Inactivated Influenza Vaccine Elicits Cross-Reactive Anti-HA Antibodies with Strong ADCC Against Antigenically Drifted Circulating H3N2 Virus in Humans. Viral Immunol 2016; 29:259-62. [PMID: 26950058 DOI: 10.1089/vim.2016.0003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
It is well established that virus neutralizing (VN) antibodies to hemagglutinin (HA) antigens of influenza A viruses provide optimal protection against antigenically matched strains of influenza A viruses. In contrast, little is known about the potential role of HA-specific, non-neutralizing antibodies in protection against human influenza illness at present. In this study, we show that individuals vaccinated with the 2014-15 seasonal inactivated influenza vaccine displayed strong A/H3N2 HA-specific antibody-dependent cell-mediated cytotoxicity (ADCC) activities against an antigenically drifted H3N2 virus, despite poor induction of cross-reactive neutralizing antibodies against the antigenic variant. Given that passive transfer of influenza HA-monospecific immune sera with negligible levels of HA-specific VN antibodies can often confer considerable cross protection against lethal challenge with heterologous influenza viruses in animal models, it is conceivable that HA-specific, non-neutralizing antibodies may provide certain degree of cross protection against antigenically drifted influenza A viruses through ADCC in case of influenza vaccine mismatches. This may have important implications for public health.
Collapse
Affiliation(s)
- Weimin Zhong
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - F Liaini Gross
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Crystal Holiday
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Stacie N Jefferson
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Yaohui Bai
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Feng Liu
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Jacqueline M Katz
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| | - Min Z Levine
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention , Atlanta, Georgia
| |
Collapse
|
84
|
Selection and expansion of natural killer cells for NK cell-based immunotherapy. Cancer Immunol Immunother 2016; 65:477-84. [PMID: 26810567 PMCID: PMC4826432 DOI: 10.1007/s00262-016-1792-y] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 01/01/2016] [Indexed: 01/19/2023]
Abstract
Natural killer (NK) cells have been used in several clinical trials as adaptive immunotherapy. The low numbers of these cells in peripheral blood mononuclear cells (PBMC) have resulted in various approaches to preferentially expand primary NK cells from PBMC. While some clinical trials have used the addition of interleukin 2 (IL-2) to co-stimulate the expansion of purified NK cells from allogeneic donors, recent studies have shown promising results in achieving in vitro expansion of NK cells to large numbers for adoptive immunotherapy. NK cell expansion requires multiple cell signals for survival, proliferation and activation. Thus, expansion strategies have been focused either to substitute these factors using autologous feeder cells or to use genetically modified allogeneic feeder cells. Recent developments in the clinical use of genetically modified NK cell lines with chimeric antigen receptors, the development of expansion protocols for the clinical use of NK cell from human embryonic stem cells and induced pluripotent stem cells are challenging improvements for NK cell-based immunotherapy. Transfer of several of these protocols to clinical-grade production of NK cells necessitates adaptation of good manufacturing practice conditions, and the development of freezing conditions to establish NK cell stocks will require some effort and, however, should enhance the therapeutic options of NK cells in clinical medicine.
Collapse
|
85
|
McCulley KJ, Somanchi SS. Assessment of Natural Killer Cell Cytotoxicity Using Image Cytometry Method. Methods Mol Biol 2016; 1441:107-16. [PMID: 27177660 DOI: 10.1007/978-1-4939-3684-7_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Although natural killer (NK) cells produce various cytokines that regulate other lymphocytes of the immune system, the primary effector function of NK cells is the direct cytolysis of their targets. Hence analyzing the cytotoxic potential of these lymphocytes is fundamental to understanding their biology and their clinical impact. We have previously shown that release-based cytotoxicity assays, such as calcein release assay, could potentially underestimate percent specific lysis if the entrapped reporter is not completely released and demonstrated that an Image cytometry method can overcome this caveat. In this chapter, we describe a detailed methodology to quantitate NK cell cytotoxicity using the Cellometer Vision Image Cytometry system.
Collapse
Affiliation(s)
- Kelsey J McCulley
- Department of Technology R&D, Nexcelom Bioscience LLC, 360 Merrimack St. Building 9, Lawrence, MA, 01843, USA
| | - Srinivas S Somanchi
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit # 853, Houston, TX, 77030, USA.
| |
Collapse
|
86
|
Rezvani K, Rouce RH. The Application of Natural Killer Cell Immunotherapy for the Treatment of Cancer. Front Immunol 2015; 6:578. [PMID: 26635792 PMCID: PMC4648067 DOI: 10.3389/fimmu.2015.00578] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 10/29/2015] [Indexed: 12/17/2022] Open
Abstract
Natural killer (NK) cells are essential components of the innate immune system and play a critical role in host immunity against cancer. Recent progress in our understanding of NK cell immunobiology has paved the way for novel NK cell-based therapeutic strategies for the treatment of cancer. In this review, we will focus on recent advances in the field of NK cell immunotherapy, including augmentation of antibody-dependent cellular cytotoxicity, manipulation of receptor-mediated activation, and adoptive immunotherapy with ex vivo-expanded, chimeric antigen receptor (CAR)-engineered, or engager-modified NK cells. In contrast to T lymphocytes, donor NK cells do not attack non-hematopoietic tissues, suggesting that an NK-mediated antitumor effect can be achieved in the absence of graft-vs.-host disease. Despite reports of clinical efficacy, a number of factors limit the application of NK cell immunotherapy for the treatment of cancer, such as the failure of infused NK cells to expand and persist in vivo. Therefore, efforts to enhance the therapeutic benefit of NK cell-based immunotherapy by developing strategies to manipulate the NK cell product, host factors, and tumor targets are the subject of intense research. In the preclinical setting, genetic engineering of NK cells to express CARs to redirect their antitumor specificity has shown significant promise. Given the short lifespan and potent cytolytic function of mature NK cells, they are attractive candidate effector cells to express CARs for adoptive immunotherapies. Another innovative approach to redirect NK cytotoxicity towards tumor cells is to create either bispecific or trispecific antibodies, thus augmenting cytotoxicity against tumor-associated antigens. These are exciting times for the study of NK cells; with recent advances in the field of NK cell biology and translational research, it is likely that NK cell immunotherapy will move to the forefront of cancer immunotherapy over the next few years.
Collapse
Affiliation(s)
- Katayoun Rezvani
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Rayne H Rouce
- Department of Pediatrics, Texas Children's Cancer and Hematology Centers, Baylor College of Medicine , Houston, TX , USA ; Center for Cell and Gene Therapy, Baylor College of Medicine Houston Methodist Hospital and Texas Children's Hospital , Houston, TX , USA
| |
Collapse
|
87
|
A Novel Method for Assessment of Natural Killer Cell Cytotoxicity Using Image Cytometry. PLoS One 2015; 10:e0141074. [PMID: 26492577 PMCID: PMC4619620 DOI: 10.1371/journal.pone.0141074] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 10/04/2015] [Indexed: 11/28/2022] Open
Abstract
Natural killer (NK) cells belong to the innate arm of the immune system and though activated NK cells can modulate immune responses through the secretion of cytokines, their primary effector function is through target cell lysis. Accordingly, cytotoxicity assays are central to studying NK cell function. The 51Chromium release assay, is the “gold standard” for cytotoxicity assay, however, due to concerns over toxicity associated with the use and disposal of radioactive compounds there is a significant interest in non-radioactive methods. We have previously used the calcein release assay as a non-radioactive alternative for studying NK cell cytotoxicity. In this study, we show that the calcein release assay varies in its dynamic range for different tumor targets, and that the entrapped calcein could remain unreleased within apoptotic bodies of lysed tumor targets or incompletely released resulting in underestimation of percent specific lysis. To overcome these limitations, we developed a novel cytotoxicity assay using the Cellometer Vision Image Cytometer and compared this method to standard calcein release assay for measuring NK cell cytotoxicity. Using tumor lines K562, 721.221, and Jurkat, we demonstrate here that image cytometry shows significantly higher percent specific lysis of the target cells compared to the standard calcein release assay within the same experimental setup. Image cytometry is able to accurately analyze live target cells by excluding dimmer cells and smaller apoptotic bodies from viable target cell counts. The image cytometry-based cytotoxicity assay is a simple, direct and sensitive method and is an appealing option for routine cytotoxicity assay.
Collapse
|
88
|
A molecular perspective on rituximab: A monoclonal antibody for B cell non Hodgkin lymphoma and other affections. Crit Rev Oncol Hematol 2015; 97:275-90. [PMID: 26443686 DOI: 10.1016/j.critrevonc.2015.09.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 08/04/2015] [Accepted: 09/24/2015] [Indexed: 12/20/2022] Open
Abstract
Rituximab (a chimeric anti-CD20 monoclonal antibody) is the first Food and Drug Administration approved anti-tumor antibody. Immunotherapy by rituximab, especially in combination-therapy, is a mainstay for a vast variety of B-cell malignancies therapy. Its therapeutic value is unquestionable, yet the mechanisms of action responsible for anti-tumor activity of rituximab and rituximab resistance mechanisms are not completely understood. Investigation of the mechanisms of action that contribute to the rituximab activity have eventually directed to a suite of novel combinations and novel treatment schedules, and also have resulted new generations of antibodies with more desired effects. Although, further investigations are needed to define the mechanisms of rituximab resistance and prominent effector activity of the altered next generation anti-CD20 to improve their efficacies and develop new anti-CD20 monoclonal antibodies in NHL treatment. This article focuses on the properties of CD20 which led scientists to select it as an effective therapeutic target and the molecular details of mechanisms of rituximab action and resistance. We also discuss about the impact of rituximab in monotherapy and in combination with chemotherapy regimens. Finally, we comparatively summarize the next generations of anti CD20 monoclonal antibodies to highlight their advantages relative to their ancestor: Rituximab.
Collapse
|
89
|
Battella S, Cox MC, Santoni A, Palmieri G. Natural killer (NK) cells and anti-tumor therapeutic mAb: unexplored interactions. J Leukoc Biol 2015; 99:87-96. [PMID: 26136506 DOI: 10.1189/jlb.5vmr0415-141r] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/16/2015] [Indexed: 12/11/2022] Open
Abstract
Tumor-targeting mAb are widely used in the treatment of a variety of solid and hematopoietic tumors and represent the first immunotherapeutic approach successfully arrived to the clinic. Nevertheless, the role of distinct immune mechanisms in contributing to their therapeutic efficacy is not completely understood and may vary depending on tumor- or antigen/antibody-dependent characteristics. Availability of next-generation, engineered, tumor-targeting mAb, optimized in their capability to recruit selected immune effectors, re-enforces the need for a deeper understanding of the mechanisms underlying anti-tumor mAb functionality. NK cells participate with a major role to innate anti-tumor responses, by exerting cytotoxic activity and producing a vast array of cytokines. As the CD16 (low-affinity FcγRIIIA)-activating receptor is expressed on the majority of NK cells, its effector functions can be ideally recruited against therapeutic mAb-opsonized tumor cells. The exact role of NK cells in determining therapeutic efficacy of tumor-targeting mAb is still unclear and much sought after. This knowledge will be instrumental to design innovative combination schemes with newly validated immunomodulatory agents. We will summarize what is known about the role of NK cells in therapeutic anti-tumor mAb therapy, with particular emphasis on RTX chimeric anti-CD20 mAb, the first one used in clinical practice for treating B cell malignancies.
Collapse
Affiliation(s)
- Simone Battella
- Departments of *Experimental Medicine and Molecular Medicine, Hematology Unit, Sant'Andrea Hospital, and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Maria Christina Cox
- Departments of *Experimental Medicine and Molecular Medicine, Hematology Unit, Sant'Andrea Hospital, and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Angela Santoni
- Departments of *Experimental Medicine and Molecular Medicine, Hematology Unit, Sant'Andrea Hospital, and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Gabriella Palmieri
- Departments of *Experimental Medicine and Molecular Medicine, Hematology Unit, Sant'Andrea Hospital, and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| |
Collapse
|
90
|
Carlsten M, Childs RW. Genetic Manipulation of NK Cells for Cancer Immunotherapy: Techniques and Clinical Implications. Front Immunol 2015; 6:266. [PMID: 26113846 PMCID: PMC4462109 DOI: 10.3389/fimmu.2015.00266] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/13/2015] [Indexed: 11/13/2022] Open
Abstract
Given their rapid and efficient capacity to recognize and kill tumor cells, natural killer (NK) cells represent a unique immune cell to genetically reprogram in an effort to improve the outcome of cell-based cancer immunotherapy. However, technical and biological challenges associated with gene delivery into NK cells have significantly tempered this approach. Recent advances in viral transduction and electroporation have now allowed detailed characterization of genetically modified NK cells and provided a better understanding for how these cells can be utilized in the clinic to optimize their capacity to induce tumor regression in vivo. Improving NK cell persistence in vivo via autocrine IL-2 and IL-15 stimulation, enhancing tumor targeting by silencing inhibitory NK cell receptors such as NKG2A, and redirecting tumor killing via chimeric antigen receptors, all represent approaches that hold promise in preclinical studies. This review focuses on available methods for genetic reprograming of NK cells and the advantages and challenges associated with each method. It also gives an overview of strategies for genetic reprograming of NK cells that have been evaluated to date and an outlook on how these strategies may be best utilized in clinical protocols. With the recent advances in our understanding of the complex biological networks that regulate the ability of NK cells to target and kill tumors in vivo, we foresee genetic engineering as an obligatory pathway required to exploit the full potential of NK-cell based immunotherapy in the clinic.
Collapse
Affiliation(s)
- Mattias Carlsten
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health , Bethesda, MD , USA
| | - Richard W Childs
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
91
|
Kellner C, Günther A, Humpe A, Repp R, Klausz K, Derer S, Valerius T, Ritgen M, Brüggemann M, van de Winkel JG, Parren PW, Kneba M, Gramatzki M, Peipp M. Enhancing natural killer cell-mediated lysis of lymphoma cells by combining therapeutic antibodies with CD20-specific immunoligands engaging NKG2D or NKp30. Oncoimmunology 2015; 5:e1058459. [PMID: 26942070 DOI: 10.1080/2162402x.2015.1058459] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 02/06/2023] Open
Abstract
Antibody-dependent cell-mediated cytotoxicity (ADCC) mediated through the IgG Fc receptor FcγRIIIa represents a major effector function of many therapeutic antibodies. In an attempt to further enhance natural killer (NK) cell-mediated ADCC, we combined therapeutic antibodies against CD20 and CD38 with recombinant immunoligands against the stimulatory NK cell receptors NKG2D or NKp30. These immunoligands, respectively designated as ULBP2:7D8 and B7-H6:7D8, contained the CD20 scFv 7D8 as a targeting moiety and a cognate ligand for either NKG2D or NKp30 (i.e. ULBP2 and B7-H6, respectively). Both the immunoligands synergistically augmented ADCC in combination with the CD20 antibody rituximab and the CD38 antibody daratumumab. Combinations with ULBP2:7D8 resulted in higher cytotoxicity compared to combinations with B7-H6:7D8, suggesting that coligation of FcγRIIIa with NKG2D triggered NK cells more efficiently than with NKp30. Addition of B7-H6:7D8 to ULBP2:7D8 and rituximab in a triple combination did not further increase the extent of tumor cell lysis. Importantly, immunoligand-mediated enhancement of ADCC was also observed for tumor cells and autologous NK cells from patients with hematologic malignancies, in which, again, ULBP2:7D8 was particularly active. In summary, co-targeting of NKG2D was more effective in promoting rituximab or daratumumab-mediated ADCC by NK cells than co-ligation of NKp30. The observed increase in the ADCC activity of these therapeutic antibodies suggests promise for a 'dual-dual-targeting' approach in which tumor cell surface antigens are targeted in concert with two distinct activating NK cell receptors (i.e. FcγRIIIa and NKG2D or B7-H6).
Collapse
Affiliation(s)
- Christian Kellner
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Andreas Günther
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Andreas Humpe
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Roland Repp
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Katja Klausz
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Stefanie Derer
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Matthias Ritgen
- 2nd Department of Medicine; Christian-Albrechts-University Kiel ; Kiel, Germany
| | - Monika Brüggemann
- 2nd Department of Medicine; Christian-Albrechts-University Kiel ; Kiel, Germany
| | - Jan Gj van de Winkel
- Genmab; Utrecht, the Netherlands; Department of Immunology; University Medical Center Utrecht; Utrecht, the Netherlands
| | - Paul Whi Parren
- Genmab; Utrecht, the Netherlands; Department of Cancer and Inflammation Research; Institute of Molecular Medicine; University of Southern Denmark; Odense, Denmark; Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden, the Netherlands
| | - Michael Kneba
- 2nd Department of Medicine; Christian-Albrechts-University Kiel ; Kiel, Germany
| | - Martin Gramatzki
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Matthias Peipp
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| |
Collapse
|
92
|
Lim O, Jung MY, Hwang YK, Shin EC. Present and Future of Allogeneic Natural Killer Cell Therapy. Front Immunol 2015; 6:286. [PMID: 26089823 PMCID: PMC4453480 DOI: 10.3389/fimmu.2015.00286] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/18/2015] [Indexed: 01/04/2023] Open
Abstract
Natural killer (NK) cells are innate lymphocytes that are capable of eliminating tumor cells and are therefore used for cancer therapy. Although many early investigators used autologous NK cells, including lymphokine-activated killer cells, the clinical efficacies were not satisfactory. Meanwhile, human leukocyte antigen (HLA)-haploidentical hematopoietic stem cell transplantation revealed the antitumor effect of allogeneic NK cells, and HLA-haploidentical, killer cell immunoglobulin-like receptor ligand-mismatched allogeneic NK cells are currently used for many protocols requiring NK cells. Moreover, allogeneic NK cells from non-HLA-related healthy donors have been recently used in cancer therapy. The use of allogeneic NK cells from non-HLA-related healthy donors allows the selection of donor NK cells with higher flexibility and to prepare expanded, cryopreserved NK cells for instant administration without delay for ex vivo expansion. In cancer therapy with allogeneic NK cells, optimal matching of donors and recipients is important to maximize the efficacy of the therapy. In this review, we summarize the present state of allogeneic NK cell therapy and its future directions.
Collapse
Affiliation(s)
- Okjae Lim
- Virology and Immunology Team, MOGAM Biotechnology Institute , Yongin , South Korea
| | - Mi Young Jung
- Virology and Immunology Team, MOGAM Biotechnology Institute , Yongin , South Korea
| | - Yu Kyeong Hwang
- Cell Therapy Center, GreenCross LabCell , Yongin , South Korea
| | - Eui-Cheol Shin
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, KAIST , Daejeon , South Korea
| |
Collapse
|
93
|
NK cell function triggered by multiple activating receptors is negatively regulated by glycogen synthase kinase-3β. Cell Signal 2015; 27:1731-41. [PMID: 26022178 DOI: 10.1016/j.cellsig.2015.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/15/2015] [Accepted: 05/19/2015] [Indexed: 12/20/2022]
Abstract
Activation of NK cells is triggered by combined signals from multiple activating receptors that belong to different families. Several NK cell activating receptors have been identified, but their role in the regulation of effector functions is primarily understood in the context of their individual engagement. Therefore, little is known about the signaling pathways broadly implicated by the multiple NK cell activation cues. Here we provide evidence pointing to glycogen synthase kinase (GSK)-3β as a negative regulator of multiple NK cell activating signals. Using an activation model that combines NKG2D and 2B4 and tests different signaling molecules, we found that GSK-3 undergoes inhibitory phosphorylation at regulatory serine residues by the engagement of NKG2D and 2B4, either individually or in combination. The extent of such phosphorylation was closely correlated with the degree of NK cell activation. NK cell functions, such as cytokine production and cytotoxicity, were consistently enhanced by the knockdown of GSK-3β or its inhibition with different pharmacological inhibitors, whereas inhibition of the GSK-3α isoform had no effect. In addition, NK cell function was augmented by the overexpression of a catalytically inactive form of GSK-3β. Importantly, the regulation of NK cell function by GSK-3β was common to diverse activating receptors that signal through both ITAM and non-ITAM pathways. Thus, our results suggest that GSK-3β negatively regulates NK cell activation and that modulation of GSK-3β function could be used to enhance NK cell activation.
Collapse
|
94
|
Zhang Q, Liu XY, Zhang T, Zhang XF, Zhao L, Long F, Liu ZK, Wang EH. The dual-functional capability of cytokine-induced killer cells and application in tumor immunology. Hum Immunol 2015; 76:385-91. [DOI: 10.1016/j.humimm.2014.09.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 09/27/2014] [Accepted: 09/27/2014] [Indexed: 01/18/2023]
|
95
|
Roberti MP, Juliá EP, Rocca YS, Amat M, Bravo AI, Loza J, Coló F, Loza CM, Fabiano V, Maino M, Podhorzer A, Fainboim L, Barrio MM, Mordoh J, Levy EM. Overexpression of CD85j in TNBC patients inhibits Cetuximab-mediated NK-cell ADCC but can be restored with CD85j functional blockade. Eur J Immunol 2015; 45:1560-9. [PMID: 25726929 DOI: 10.1002/eji.201445353] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/15/2015] [Accepted: 02/25/2015] [Indexed: 01/09/2023]
Abstract
Clinical studies suggest that triple negative breast cancer (TNBC) patients with epidermal growth factor receptor (EGFR)-expressing tumors could benefit from therapy with Cetuximab, which targets EGFR. NK cells are the primary effectors of antibody (Ab)-dependent cell-mediated cytotoxicity (ADCC) and thus play a role in Ab-based therapies. We have previously described diminished levels of Cetuximab-mediated ADCC in vitro in patients with advanced breast cancer. Here, we investigated the potential causes of this NK-cell functional deficiency. We characterized NK-cell activating/inhibitory receptors in the peripheral blood of breast cancer patients and found CD85j inhibitory receptor overexpression. The capacity of NK cells to perform Cetuximab-triggered ADCC against TNBC cells correlated inversely with CD85j expression, even in the presence of the stimulatory cytokines IL-2 or IL-15. Hence, patients expressing high levels of CD85j had an impaired ability to lyse TNBC cells in the presence of Cetuximab. We also found that CD85j overexpression was associated with HLA-I and soluble HLA-G expression by tumors. A CD85j functional blockade with a CD85j antagonist Ab restored ADCC levels in breast cancer patients and reverted this negative effect. Our data suggest that strategies that overcome the hurdles of immune activation could improve Cetuximab clinical efficacy.
Collapse
Affiliation(s)
- María P Roberti
- Centro de Investigaciones Oncológicas CIO-FUCA, Ciudad de Buenos Aires, Argentina
| | - Estefanía P Juliá
- Centro de Investigaciones Oncológicas CIO-FUCA, Ciudad de Buenos Aires, Argentina
| | - Yamila S Rocca
- Fundación Instituto Leloir-IIBBA, Ciudad de Buenos Aires, Argentina
| | - Mora Amat
- Instituto Alexander Fleming, Ciudad de Buenos Aires, Argentina
| | - Alicia I Bravo
- Hospital Eva Perón, San Martín, Provincia de Buenos Aires, Argentina
| | - José Loza
- Instituto Alexander Fleming, Ciudad de Buenos Aires, Argentina
| | - Federico Coló
- Instituto Alexander Fleming, Ciudad de Buenos Aires, Argentina
| | - Carlos M Loza
- Instituto Alexander Fleming, Ciudad de Buenos Aires, Argentina
| | | | - Mercedes Maino
- Instituto Alexander Fleming, Ciudad de Buenos Aires, Argentina
| | - Ariel Podhorzer
- Hospital de Clínicas José de San Martín, Ciudad de Buenos Aires, Argentina
| | - Leonardo Fainboim
- Hospital de Clínicas José de San Martín, Ciudad de Buenos Aires, Argentina
| | - María M Barrio
- Centro de Investigaciones Oncológicas CIO-FUCA, Ciudad de Buenos Aires, Argentina
| | - José Mordoh
- Centro de Investigaciones Oncológicas CIO-FUCA, Ciudad de Buenos Aires, Argentina.,Fundación Instituto Leloir-IIBBA, Ciudad de Buenos Aires, Argentina.,Instituto Alexander Fleming, Ciudad de Buenos Aires, Argentina
| | - Estrella M Levy
- Centro de Investigaciones Oncológicas CIO-FUCA, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
96
|
Jing Y, Ni Z, Wu J, Higgins L, Markowski TW, Kaufman DS, Walcheck B. Identification of an ADAM17 cleavage region in human CD16 (FcγRIII) and the engineering of a non-cleavable version of the receptor in NK cells. PLoS One 2015; 10:e0121788. [PMID: 25816339 PMCID: PMC4376770 DOI: 10.1371/journal.pone.0121788] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 02/09/2015] [Indexed: 12/12/2022] Open
Abstract
CD16a and CD16b are IgG Fc receptors expressed by human natural killer (NK) cells and neutrophils, respectively. Both CD16 isoforms undergo a rapid down-regulation in expression by ADAM17-mediated proteolytic cleavage upon cell activation by various stimuli. We examined soluble CD16 released from activated NK cells and neutrophils by mass spectrometric analysis, and identified three separate cleavage sites in close proximity at P1/P1′ positions alanine195/valine196, valine196/serine197, and threonine198/isoleucine199, revealing a membrane proximal cleavage region in CD16. Substitution of the serine at position 197 in the middle of the cleavage region for a proline (S197P) effectively blocked CD16a and CD16b cleavage in cell-based assays. We also show that CD16a/S197P was resistant to cleavage when expressed in the human NK cell line NK92 and primary NK cells derived from genetically-engineered human induced pluripotent stem cells. CD16a is a potent activating receptor and despite blocking CD16a shedding, the S197P mutation did not disrupt IgG binding by the receptor or its activation of NK92 cells by antibody-treated tumor cells. Our findings provide further characterization of CD16 cleavage by ADAM17 and they demonstrate that a non-cleavable version of CD16a can be expressed in engineered NK cells.
Collapse
Affiliation(s)
- Yawu Jing
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minneapolis and St. Paul, Minnesota, United States of America
| | - Zhenya Ni
- Department of Medicine, Stem Cell Institute, University of Minnesota, Minneapolis and St. Paul, Minnesota, United States of America
| | - Jianming Wu
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minneapolis and St. Paul, Minnesota, United States of America
| | - LeeAnn Higgins
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis and St. Paul, Minnesota, United States of America
| | - Todd W. Markowski
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis and St. Paul, Minnesota, United States of America
| | - Dan S. Kaufman
- Department of Medicine, Stem Cell Institute, University of Minnesota, Minneapolis and St. Paul, Minnesota, United States of America
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minneapolis and St. Paul, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
97
|
Natural killer cells can inhibit the transmission of human cytomegalovirus in cell culture by using mechanisms from innate and adaptive immune responses. J Virol 2014; 89:2906-17. [PMID: 25540381 DOI: 10.1128/jvi.03489-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED Human cytomegalovirus (HCMV) transmission within the host is important for the pathogenesis of HCMV diseases. Natural killer (NK) cells are well known to provide a first line of host defense against virus infections. However, the role of NK cells in the control of HCMV transmission is still unknown. Here, we provide the first experimental evidence that NK cells can efficiently control HCMV transmission in different cell types. NK cells engage different mechanisms to control the HCMV transmission both via soluble factors and by cell contact. NK cell-produced interferon gamma (IFN-γ) suppresses HCMV production and induces resistance of bystander cells to HCMV infection. The UL16 viral gene contributes to an immune evasion from the NK cell-mediated control of HCMV transmission. Furthermore, the efficacy of the antibody-dependent NK cell-mediated control of HCMV transmission is dependent on a CD16-158V/F polymorphism. Our findings indicate that NK cells may have a clinical relevance in HCMV infection and highlight the need to consider potential therapeutic strategies based on the manipulation of NK cells. IMPORTANCE Human cytomegalovirus (HCMV) infects 40% to 100% of the human population worldwide. After primary infection, mainly in childhood, the virus establishes a lifelong persistence with possible reactivations. Most infections remain asymptomatic; however, HCMV represents a major health problem since it is the most frequent cause of infection-induced birth defects and is responsible for high morbidity and mortality in immunocompromised patients. The immune system normally controls the infection by antibodies and immune effector cells. One type of effector cells are the natural killer (NK) cells, which provide a rapid response to virus-infected cells. NK cells participate in viral clearance by inducing the death of infected cells. NK cells also secrete antiviral cytokines as a consequence of the interaction with an infected cell. In this study, we investigated the mechanisms by which NK cells control HCMV transmission, from the perspectives of immune surveillance and immune evasion.
Collapse
|
98
|
Cheng ZJ, Garvin D, Paguio A, Moravec R, Engel L, Fan F, Surowy T. Development of a robust reporter-based ADCC assay with frozen, thaw-and-use cells to measure Fc effector function of therapeutic antibodies. J Immunol Methods 2014; 414:69-81. [DOI: 10.1016/j.jim.2014.07.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 11/16/2022]
|
99
|
Murray JC, Aldeghaither D, Wang S, Nasto RE, Jablonski SA, Tang Y, Weiner LM. c-Abl modulates tumor cell sensitivity to antibody-dependent cellular cytotoxicity. Cancer Immunol Res 2014; 2:1186-98. [PMID: 25300860 DOI: 10.1158/2326-6066.cir-14-0083] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Monoclonal antibodies (mAb) can modulate cancer cell signal transduction and recruit antitumor immune effector mechanisms-including antibody-dependent cellular cytotoxicity (ADCC). Although several clinically effective antibodies can promote ADCC, therapeutic resistance is common. We hypothesized that oncogenic signaling networks within tumor cells affect their sensitivity to ADCC. We developed a screening platform and targeted 60 genes derived from an EGFR gene network using RNAi in an in vitro ADCC model system. Knockdown of GRB7, PRKCE, and ABL1 enhanced ADCC by primary and secondary screens. ABL1 knockdown also reduced cell proliferation, independent of its ADCC enhancement effects. c-Abl overexpression decreased ADCC sensitivity and rescued the effects of ABL1 knockdown. Imatinib inhibition of c-Abl kinase activity also enhanced ADCC-phenocopying ABL1 knockdown-against several EGFR-expressing head-and-neck squamous cell carcinoma cell lines by ex vivo primary natural killer cells. Our findings suggest that combining c-Abl inhibition with ADCC-promoting antibodies, such as cetuximab, could translate into increased therapeutic efficacy of mAbs.
Collapse
Affiliation(s)
- Joseph C Murray
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Dalal Aldeghaither
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Shangzi Wang
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Rochelle E Nasto
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC. School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania. Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Sandra A Jablonski
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Yong Tang
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Louis M Weiner
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC.
| |
Collapse
|
100
|
Sta Maria NS, Barnes SR, Jacobs RE. In vivo monitoring of natural killer cell trafficking during tumor immunotherapy. MAGNETIC RESONANCE INSIGHTS 2014; 7:15-21. [PMID: 25114550 PMCID: PMC4122546 DOI: 10.4137/mri.s13145] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/29/2014] [Accepted: 04/29/2014] [Indexed: 12/19/2022]
Abstract
Natural killer (NK) cells are a crucial part of the innate immune system and play critical roles in host anti-viral, anti-microbial, and antitumor responses. The elucidation of NK cell biology and their therapeutic use are actively being pursued with 200 clinical trials currently underway. In this review, we outline the role of NK cells in cancer immunotherapies and summarize current noninvasive imaging technologies used to track NK cells in vivo to investigate mechanisms of action, develop new therapies, and evaluate efficacy of adoptive transfer.
Collapse
Affiliation(s)
- Naomi S Sta Maria
- Biology and Biological Engineering, Beckman Institute, California Institute of Technology, Pasadena, CA, USA
| | - Samuel R Barnes
- Biology and Biological Engineering, Beckman Institute, California Institute of Technology, Pasadena, CA, USA
| | - Russell E Jacobs
- Biology and Biological Engineering, Beckman Institute, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|