1
|
Graydon EK, Conner TL, Dunham K, Olsen C, Goguet E, Coggins SA, Rekedal M, Samuels E, Jackson-Thompson B, Moser M, Lindrose A, Hollis-Perry M, Wang G, Maiolatesi S, Alcorta Y, Reyes A, Wong M, Ramsey K, Davies J, Parmelee E, Ortega O, Sanchez M, Moller S, Inglefield J, Tribble D, Burgess T, O’Connell R, Malloy AMW, Pollett S, Broder CC, Laing ED, Anderson SK, Mitre E. Natural killer cells and BNT162b2 mRNA vaccine reactogenicity and durability. Front Immunol 2023; 14:1225025. [PMID: 37711632 PMCID: PMC10497936 DOI: 10.3389/fimmu.2023.1225025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/27/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Natural killer (NK) cells can both amplify and regulate immune responses to vaccination. Studies in humans and animals have observed NK cell activation within days after mRNA vaccination. In this study, we sought to determine if baseline NK cell frequencies, phenotype, or function correlate with antibody responses or inflammatory side effects induced by the Pfizer-BioNTech COVID-19 vaccine (BNT162b2). Methods We analyzed serum and peripheral blood mononuclear cells (PBMCs) from 188 participants in the Prospective Assessment of SARS-CoV-2 Seroconversion study, an observational study evaluating immune responses in healthcare workers. Baseline serum samples and PBMCs were collected from all participants prior to any SARS-CoV-2 infection or vaccination. Spike-specific IgG antibodies were quantified at one and six months post-vaccination by microsphere-based multiplex immunoassay. NK cell frequencies and phenotypes were assessed on pre-vaccination PBMCs from all participants by multi-color flow cytometry, and on a subset of participants at time points after the 1st and 2nd doses of BNT162b2. Inflammatory side effects were assessed by structured symptom questionnaires, and baseline NK cell functionality was quantified by an in vitro killing assay on participants that reported high or low post-vaccination symptom scores. Results Key observations include: 1) circulating NK cells exhibit evidence of activation in the week following vaccination, 2) individuals with high symptom scores after 1st vaccination had higher pre-vaccination NK cytotoxicity indices, 3) high pre-vaccination NK cell numbers were associated with lower spike-specific IgG levels six months after two BNT162b2 doses, and 4) expression of the inhibitory marker NKG2A on immature NK cells was associated with higher antibody responses 1 and 6 months post-vaccination. Discussion These results suggest that NK cell activation by BNT162b2 vaccination may contribute to vaccine-induced inflammatory symptoms and reduce durability of vaccine-induced antibody responses.
Collapse
Affiliation(s)
- Elizabeth K. Graydon
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Tonia L. Conner
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
| | - Kim Dunham
- Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Cara Olsen
- Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Emilie Goguet
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Si’Ana A. Coggins
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Marana Rekedal
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Emily Samuels
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Belinda Jackson-Thompson
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Matthew Moser
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Alyssa Lindrose
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Monique Hollis-Perry
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
| | - Gregory Wang
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
- General Dynamics Information Technology, Silver Spring, MD, United States
| | - Santina Maiolatesi
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
| | - Yolanda Alcorta
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
- General Dynamics Information Technology, Silver Spring, MD, United States
| | - Anatalio Reyes
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
- General Dynamics Information Technology, Silver Spring, MD, United States
| | - Mimi Wong
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
- General Dynamics Information Technology, Silver Spring, MD, United States
| | - Kathy Ramsey
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
- General Dynamics Information Technology, Silver Spring, MD, United States
| | - Julian Davies
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Edward Parmelee
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Orlando Ortega
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Mimi Sanchez
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Sydney Moller
- Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Jon Inglefield
- Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - David Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Timothy Burgess
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Robert O’Connell
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Allison M. W. Malloy
- Department of Pediatrics, Uniformed Services University, Bethesda, MD, United States
| | - Simon Pollett
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Christopher C. Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
| | - Eric D. Laing
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
| | - Stephen K. Anderson
- Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Edward Mitre
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
2
|
Non-Assembled ORF2 Capsid Protein of Porcine Circovirus 2b Does Not Confer Protective Immunity. Pathogens 2021; 10:pathogens10091161. [PMID: 34578193 PMCID: PMC8466160 DOI: 10.3390/pathogens10091161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/28/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022] Open
Abstract
Porcine Circovirus 2 (PCV2) vaccines are based on either inactivated whole virion, or recombinant ORF2 capsid protein assembled into Virus-like Particles (VLPs). No data are available about the immunizing properties of free, non-assembled capsid protein. To investigate this issue, ORF2 of a reference PCV2b strain was expressed in a Baculovirus-based expression system without assembly into VLPs. The free purified protein was formulated into an oil vaccine at three distinct Ag payloads: 10.8/3.6/1.2 micrograms/dose. Each dose was injected intramuscularly into five, 37-day old piglets, carefully matched for maternally-derived antibody. Five control piglets were injected with sterile PBS in oil adjuvant. Twenty-eight days later, all the pigs were challenged intranasally with 105.3 TCID50 of PCV2b strain DV6503. After challenge infection, all the pigs remained in good clinical conditions. The recombinant vaccine did not induce significant antibody and PCV2-specific IFN-γ responses. ELISPOT and lymphocyte proliferation data confirmed poor induction of cell-mediated immunity. In terms of PCV2 viremia, there was no significant difference between vaccinated and control animals. The histological data indicated the absence of a detectable viral load and of PCVAD lesions in both vaccinated and control animals, as well as of histiocytes and multi-nucleated giant cells. We conclude that free, non-assembled ORF2 capsid protein does not induce protective immunity.
Collapse
|
3
|
Sanchez Vargas LA, Adam A, Masterson M, Smith M, Lyski ZL, Dowd KA, Pierson TC, Messer WB, Currier JR, Mathew A. Non-structural protein 1-specific antibodies directed against Zika virus in humans mediate antibody-dependent cellular cytotoxicity. Immunology 2021; 164:386-397. [PMID: 34056709 PMCID: PMC8442231 DOI: 10.1111/imm.13380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/18/2021] [Accepted: 05/23/2021] [Indexed: 12/14/2022] Open
Abstract
There is growing interest in understanding antibody (Ab) function beyond neutralization. The non-structural protein 1 (NS1) of Zika virus (ZIKV) is an attractive candidate for an effective vaccine as Abs against NS1, unlike the envelope or premembrane, do not carry the risk of mediating antibody-dependent enhancement. Our aim was to evaluate whether ZIKV NS1 Abs elicited following natural infection in humans can mediate antibody-dependent cellular cytotoxicity (ADCC). We evaluated the isotype specificity of ZIKV-specific Abs in immune sera and supernatants from stimulated immune PBMC and found that Abs against ZIKV NS1 and virus-like particles were predominantly of the IgG1 isotype. Using a recently developed FluoroSpot assay, we found robust frequencies of NS1-specific Ab-secreting cells in PBMC of individuals who were naturally infected with ZIKV. We developed assays to measure both natural killer cell activation by flow cytometry and target cell lysis of ZIKV NS1-expressing cells using an image cytometry assay in the presence of ZIKV NS1 Abs. Our data indicate efficient opsonization of ZIKV NS1-expressing CEM-NKR cell lines using ZIKV-immune but not ZIKV-naïve sera, a prerequisite of ADCC. Furthermore, sera from immune donors were able to induce both NK cell degranulation and lysis of ZIKV NS1 CEM-NKR cells in vitro. Our data suggest that ADCC is a possible mechanism for ZIKV NS1 Abs to eliminate virally infected target cells.
Collapse
Affiliation(s)
- Luis A Sanchez Vargas
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, USA
| | - Awadalkareem Adam
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, USA
| | - Mary Masterson
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Madison Smith
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, USA
| | - Zoe L Lyski
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | | | | | - William B Messer
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA.,Division of Infectious Diseases, Department of Medicine, Oregon Health & Science University, Portland, OR, USA.,OHSU-PSU School of Public Health, Program in Epidemiology, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey R Currier
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Anuja Mathew
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, USA
| |
Collapse
|
4
|
Dataset of immune responses induced in swine by an inactivated Porcine Circovirus 2b vaccine. Data Brief 2021; 35:106906. [PMID: 33997189 PMCID: PMC8100898 DOI: 10.1016/j.dib.2021.106906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/13/2021] [Accepted: 02/19/2021] [Indexed: 01/03/2023] Open
Abstract
A whole virus, inactivated, Porcine Circovirus 2b (PCV2b) vaccine was submitted to a quantal assay of potency, as explained in detail in our companion paper [1]. To this purpose, twenty, 45-day old piglets, checked for maternally-derived antibody (MDA), were allocated to four groups of 5 animals each; these were vaccinated with 800/266/88/0 nanograms, respectively, of an inactivated PCV2b strain, consisting of two distinct virion populations. Twenty-six days later, all the pigs were challenged intranasally with the homologous PCV2b strain. In the presence of a clear dose-dependent protection in terms of viremia, no such effect was observed in terms of weight gain after challenge. The 800 and 266-ng payloads were associated with neutralizing antibody titers above the MDA levels in oral fluids. Higher levels of viremia in control and 88-ng groups [1] coincided with a higher Natural Killer activity of tracheobronchial lymph node cells from PCV2-infected pigs. The PCV2 ORF2-specific ELISPOT assay for IFN-g– secreting cells showed very few (2–4) ORF2-specific cells/105 peripheral blood mononuclear cells beyond the basal levels under our experimental conditions (non-significant differences among groups). Also, no significant differences were observed in the degree of lymphoid tissue hyperplasia among the different groups.
Collapse
|
5
|
Xu Z, Zhu X, Su L, Zou C, Chen X, Hou Y, Gong C, Ng W, Ni Z, Wang L, Yan X, Zhu Y, Jiao X, Yao C, Zhu S. A high-throughput assay for screening natural products that boost NK cell-mediated killing of cancer cells. PHARMACEUTICAL BIOLOGY 2020; 58:357-366. [PMID: 32356467 PMCID: PMC7241510 DOI: 10.1080/13880209.2020.1748661] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Context: Natural killer (NK) cells can eliminate malignant cells and play a vital role in immunosurveillance. Administration of natural compounds represents a promising approach for antitumor immunotherapy, which may enhance the NK cell activity via multiple mechanisms.Objective: Establishing approaches to evaluate the effect of select natural products on NK cell-mediated cytotoxicity.Materials and methods: We selected a natural product library containing 2880 pure compounds, which was provided by the National Centre for Drug Screening of China. 0.1% DMSO was employed as a negative control, and 100 U/mL human recombinant IL-2 was employed as a positive control. To evaluate the % of tumour cells which were killed by NK cells, expanded NK cells were co-cultured with tumour cells and then treated with natural products at the concentration of 10 μM. After 24-h co-incubation, luminescent signal was detected and percent lysis was calculated.Results: We report on the results of a three-round high-throughput screening effort that identified 20-deoxyingenol 3-angelate (DI3A) and its analogue ingenol 3-angelate (I3A) as immuno enhancers which boosts NK cell-mediated killing of non-small cell lung cancer cells (NSCLCs). Biophotonic cytotoxicity assay and calcein release assay were used as two well-established NK cell cytotoxicity detection assays to validate the immuno-enhancing effects of DI3A and I3A, which was achieved by increasing degranulation and interferon-gamma secretion of NK cells.Conclusions: Our newly established ATP-based method was a valuable and information-rich screening tool to investigate the biological effects of natural products on both NK cells and tumour cells.
Collapse
Affiliation(s)
- Zihang Xu
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaowen Zhu
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lin Su
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunpu Zou
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao Chen
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifei Hou
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenyuan Gong
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wanyi Ng
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhongya Ni
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lixin Wang
- Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuewei Yan
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yangzhuangzhuang Zhu
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoning Jiao
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Yao
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- CONTACT Chao Yao
| | - Shiguo Zhu
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shiguo Zhu Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Tang G, Yuan X, Luo Y, Lin Q, Chen Z, Xing X, Song H, Wu S, Hou H, Yu J, Mao L, Liu W, Wang F, Sun Z. Establishing immune scoring model based on combination of the number, function, and phenotype of lymphocytes. Aging (Albany NY) 2020; 12:9328-9343. [PMID: 32396527 PMCID: PMC7288950 DOI: 10.18632/aging.103208] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 04/17/2020] [Indexed: 12/31/2022]
Abstract
Background: Quantitatively assessing host immunity remains a challenge in clinical practice. Results: Most parameters in lymphocyte number, function and phenotype were correlated with age. The reference ranges of these parameters were established in four age groups (children, adolescents, adults, and elders). The numbers of CD4+ T cells, CD8+ T cells, B cells, but not NK cells, were negatively correlated with age. However, the function of CD4+ T cells, CD8+ T cells and NK cells was positively correlated with age. The expression of CD28 on T cells gradually decreased with increasing age and was negatively correlated with their function. An opposite phenomenon was observed in the expressions of HLA-DR and CD45RO on T cells. An immune scoring model was established by using 8 parameters (CD4+ T cell number × function, CD28+CD4+ T cell number, HLA-DR+CD4+ T cell number, CD45RO+CD4+ T cell number, CD8+ T cell number × function, CD28+CD8+ T cell number, HLA-DR+CD8+ T cell number, NK cell number × function) from the results of lymphocyte number, function, and phenotype. This immune scoring model showed sensitivities of 70% and 71.4% in determining hyper-immune and hypo-immune status, respectively. Conclusions: An immune scoring model based on combination of lymphocyte number, function, and phenotype shows potential value in quantitatively assessing host immunity. Methods: 261 healthy individuals aged 1 to 82 years were recruited from Tongji Hospital. The number, function, and phenotype of CD4+ T cells, CD8+ T cells and NK cells were simultaneously determined.
Collapse
Affiliation(s)
- Guoxing Tang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Yuan
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Luo
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qun Lin
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhishui Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Beijing, China
| | - Xue Xing
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huijuan Song
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiji Wu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyan Hou
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Yu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liyan Mao
- Center for Cellular and Molecular Diagnosis, Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Weiyong Liu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyong Sun
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
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: 6.7] [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
|