1
|
Hartley C, Davila PV, Cole E, Karnsakul W. Varicella and Zoster Vaccination Strategies in Immunosuppressed Pediatric Transplant Recipients. Vaccines (Basel) 2025; 13:534. [PMID: 40432143 PMCID: PMC12115575 DOI: 10.3390/vaccines13050534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/12/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
The varicella vaccine has prevented varicella in hundreds of thousands of patients since its establishment in 1974. It stimulates both humoral and cell-mediated immunity to produce an immune response that helps protect against the disease (not necessarily the infection). Serious sequala of varicella including pneumonia, hepatitis, and encephalitis can occur, with higher incidence in immunosuppressed individuals than in the general population. Patients who are not immunosuppressed should receive routine varicella vaccinations. For those who have not completed the series or are significantly distant from their last immunization, serologic testing may be considered. In pre-transplant patients, live-attenuated vaccines should ideally be administered at least four weeks before transplantation. Case studies have documented instances of patients requiring treatment for varicella after receiving a transplant within four weeks of vaccination.
Collapse
Affiliation(s)
- Christopher Hartley
- The Department of Pharmacy, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Priscila Villalba Davila
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Johns Hopkins University of Medicine, Baltimore, MD 21287, USA (W.K.)
| | - Emma Cole
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Johns Hopkins University of Medicine, Baltimore, MD 21287, USA (W.K.)
| | - Wikrom Karnsakul
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Johns Hopkins University of Medicine, Baltimore, MD 21287, USA (W.K.)
| |
Collapse
|
2
|
Balusamy SR, Balamurugan M, Purushothaman S, Somasundaram S, Elsadek MF, Sohn D, Almutairi SM, Mijakovic I, Rahimi S, Perumalsamy H. Apoptotic cell death of stomach cancer lines (AGS) induced by Co-NTB complex through cellular organelles and DNA damage. RSC Adv 2025; 15:739-747. [PMID: 39802467 PMCID: PMC11711993 DOI: 10.1039/d4ra06377e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 12/02/2024] [Indexed: 01/16/2025] Open
Abstract
Given that stomach cancer is the fourth leading cause of cancer-related death, there is a need to develop new drugs. Among various methods, metal-based coordination compounds are considered as an efficient strategy against this type of cancer. Similarly, the benzimidazole moiety plays a crucial role in biology; thus, various benzimidazole-based compounds have been found to be active as potential anticancer drugs and are currently used in clinical trials. In this study, we explored the benzimidazole-based cobalt(ii) complex as an anticancer agent against AGS stomach cancer cell lines. Interestingly, the MTT assay of the Co-NTB complex shows a lower IC50 value of 4.25 μg mL-1 compared to cisplatin, which has an IC50 of 7.5 μg mL-1 against AGS cell lines. Light microscopy and Hoechst/propidium iodide dye staining clearly indicate that the complex damages DNA, leading to cell death through an apoptotic pathway. The apoptotic cell death pathway was further complemented by Lysotracker and Mitotracker staining, as well as transmission electron microscopy (TEM) imaging. Overall, the Co-NTB complex acts as an effective anticancer agent against AGS stomach cancer cell lines, with apoptotic cell death induced by targeting cellular organelles and DNA.
Collapse
Affiliation(s)
- Sri Renukadevi Balusamy
- Department of Food Science and Biotechnology, Sejong University Gwangjin-gu Seoul Republic of Korea
| | - Mani Balamurugan
- Department of Materials Science and Engineering, Seoul National University (SNU) 1 Gwanak ro Seoul 08826 Republic of Korea
| | - Sumitha Purushothaman
- Department of Microbiology, Bioprocess Engineering Division, Smykon Biotech Kanniyakumari India
| | - Sivaraman Somasundaram
- Department of Chemistry, Saveetha School of Engineering, SIMATS Chennai Tamil Nadu 600124 India
| | - Mohamed Farouk Elsadek
- Department of Biochemistry, College of Science, King Saud University P.O. 2455 Riyadh 11451 Saudi Arabia
| | - Daewon Sohn
- Department of Chemistry, College of Natural Sciences, Hanyang University Seoul 04763 Republic of Korea
| | - Saeedah Musaed Almutairi
- Department of Botany and Microbiology, College of Science, King Saud University P.O. 2455 Riyadh 11451 Saudi Arabia
| | - Ivan Mijakovic
- Systems and Synthetic Biology Division, Department of Life Sciences, Chalmers University of Technology Gothenburg SE-412 96 Sweden
- The Novo Nordisk Foundation, Center for Biosustainability, Technical University of Denmark Kongens Lyngby DK-2800 Denmark
| | - Shadi Rahimi
- Systems and Synthetic Biology Division, Department of Life Sciences, Chalmers University of Technology Gothenburg SE-412 96 Sweden
| | - Haribalan Perumalsamy
- Research Institute for Convergence of Basic Science, Hanyang University Seoul 04763 Republic of Korea
- Center for Creative Convergence Education, Hanyang University Seoul 04763 Republic of Korea
| |
Collapse
|
3
|
Pan HX, Qiu LX, Liang Q, Chen Z, Zhang ML, Liu S, Zhong GH, Zhu KX, Liao MJ, Hu JL, Li JX, Xu JB, Fan Y, Huang Y, Su YY, Huang SJ, Wang W, Han JL, Jia JZ, Zhu H, Cheng T, Ye XZ, Li CG, Wu T, Zhu FC, Zhang J, Xia NS. Immunogenicity and safety of an ORF7-deficient skin-attenuated and neuro-attenuated live vaccine for varicella: a randomised, double-blind, controlled, phase 2a trial. THE LANCET. INFECTIOUS DISEASES 2024; 24:922-934. [PMID: 38614117 DOI: 10.1016/s1473-3099(24)00159-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND The Oka varicella vaccine strain remains neurovirulent and can establish lifelong latent infection, raising safety concerns about vaccine-related herpes zoster. In this study, we aimed to evaluate the immunogenicity and safety of a skin-attenuated and neuro-attenuated varicella vaccine candidate (v7D vaccine). METHODS We did this randomised, double-blind, controlled, phase 2a clinical trial in Jiangsu, China. Healthy children aged 3-12 years with no history of varicella infection or vaccination were enrolled and randomly assigned (1:1:1:1) to receive a single subcutaneous injection of the v7D vaccine at 3·3 log10 plaque forming units (PFU; low-dose v7D group), 3·9 log10 PFU (medium-dose v7D group), and 4·2 log10 PFU (high-dose v7D group), or the positive control varicella vaccine (vOka vaccine group). All the participants, laboratory personnel, and investigators other than the vaccine preparation and management staff were masked to the vaccine allocation. The primary outcome was assessment of the geometric mean titres (GMTs) and seroconversion rates of anti-varicella zoster virus immunoglobulin G (IgG) induced by different dose groups of v7D vaccine at 0, 42, 60, and 90 days after vaccination in the per-protocol set for humoral immune response analysis. Safety was a secondary outcome, focusing on adverse events within 42 days post-vaccination, and serious adverse events within 6 months after vaccination. This study was registered on Chinese Clinical Trial Registry, ChiCTR2000034434. FINDINGS On Aug 18-21, 2020, 842 eligible volunteers were enrolled and randomly assigned treatment. After three participants withdrew, 839 received a low dose (n=211), middle dose (n=210), or high dose (n=210) of v7D vaccine, or the vOka vaccine (n=208). In the per-protocol set for humoral immune response analysis, the anti-varicella zoster virus IgG antibody response was highest at day 90. At day 90, the seroconversion rates of the low-dose, medium-dose, and high-dose groups of v7D vaccine and the positive control vOka vaccine group were 100·0% (95% CI 95·8-100·0; 87 of 87 participants), 98·9% (93·8-100·0; 87 of 88 participants), 97·8% (92·4-99·7; 91 of 93 participants), and 96·4% (89·8-99·2; 80 of 83 participants), respectively; the GMTs corresponded to values of 30·8 (95% CI 26·2-36·0), 31·3 (26·7-36·6), 28·2 (23·9-33·2), and 38·5 (31·7-46·7). The v7D vaccine, at low dose and medium dose, elicited a humoral immune response similar to that of the vOka vaccine. However, the high-dose v7D vaccine induced a marginally lower GMT compared with the vOka vaccine at day 90 (p=0·027). In the per-protocol set, the three dose groups of the v7D vaccine induced a similar humoral immune response at each timepoint, with no statistically significant differences. The incidence of adverse reactions in the low-dose, medium-dose, and high-dose groups of v7D vaccine was significantly lower than that in the vOka vaccine group (17% [35 of 211 participants], 20% [41 of 210 participants], and 13% [27 of 210 participants] vs 24% [50 of 208 participants], respectively; p=0·025), especially local adverse reactions (10% [22 of 211 participants], 14% [30 of 210 participants] and 9% [18 of 210 participants] vs 18% [38 of 208 participants], respectively; p=0·016). None of the serious adverse events were vaccine related. INTERPRETATION The three dose groups of the candidate v7D vaccine exhibit similar humoral immunogenicity to the vOka vaccine and are well tolerated. These findings encourage further investigations on two-dose vaccination schedules, efficacy, and the potential safety benefit of v7D vaccine in the future. FUNDING The National Natural Science Foundation of China, CAMS Innovation Fund for Medical Sciences, the Fundamental Research Funds for the Central Universities, and Beijing Wantai. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Hong-Xing Pan
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Ling-Xian Qiu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, China
| | - Qi Liang
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Zhen Chen
- National Institute for Food and Drug Control, Beijing, China
| | - Ming-Lei Zhang
- Ganyu County Center for Disease Control and Prevention, Ganyu County, Lianyungang, China
| | - Sheng Liu
- Ganyu County Center for Disease Control and Prevention, Ganyu County, Lianyungang, China
| | - Guo-Hua Zhong
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, China
| | - Kong-Xin Zhu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, China
| | - Meng-Jun Liao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, China
| | - Jia-Lei Hu
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jia-Xue Li
- Ganyu County Center for Disease Control and Prevention, Ganyu County, Lianyungang, China
| | - Jin-Bo Xu
- Ganyu County Center for Disease Control and Prevention, Ganyu County, Lianyungang, China
| | - Yong Fan
- Ganyu County Center for Disease Control and Prevention, Ganyu County, Lianyungang, China
| | - Yue Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, China
| | - Ying-Ying Su
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, China
| | - Shou-Jie Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, China
| | - Wei Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, China
| | - Jin-Le Han
- Beijing Wantai Biological Pharmacy Enterprise CO., LTD., Beijing, China
| | - Ji-Zong Jia
- Beijing Wantai Biological Pharmacy Enterprise CO., LTD., Beijing, China
| | - Hua Zhu
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Tong Cheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, China
| | - Xiang-Zhong Ye
- Beijing Wantai Biological Pharmacy Enterprise CO., LTD., Beijing, China
| | - Chang-Gui Li
- National Institute for Food and Drug Control, Beijing, China
| | - Ting Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, China.
| | - Feng-Cai Zhu
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, China.
| | - Ning-Shao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, China; The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
4
|
Browne DJ, Miller CM, Doolan DL. Technical pitfalls when collecting, cryopreserving, thawing, and stimulating human T-cells. Front Immunol 2024; 15:1382192. [PMID: 38812513 PMCID: PMC11133553 DOI: 10.3389/fimmu.2024.1382192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/29/2024] [Indexed: 05/31/2024] Open
Abstract
The collection, cryopreservation, thawing, and culture of peripheral blood mononuclear cells (PBMCs) can profoundly influence T cell viability and immunogenicity. Gold-standard PBMC processing protocols have been developed by the Office of HIV/AIDS Network Coordination (HANC); however, these protocols are not universally observed. Herein, we have explored the current literature assessing how technical variation during PBMC processing can influence cellular viability and T cell immunogenicity, noting inconsistent findings between many of these studies. Amid the mounting concerns over scientific replicability, there is growing acknowledgement that improved methodological rigour and transparent reporting is required to facilitate independent reproducibility. This review highlights that in human T cell studies, this entails adopting stringent standardised operating procedures (SOPs) for PBMC processing. We specifically propose the use of HANC's Cross-Network PBMC Processing SOP, when collecting and cryopreserving PBMCs, and the HANC member network International Maternal Pediatric Adolescent AIDS Clinical Trials (IMPAACT) PBMC Thawing SOP when thawing PBMCs. These stringent and detailed protocols include comprehensive reporting procedures to document unavoidable technical variations, such as delayed processing times. Additionally, we make further standardisation and reporting recommendations to minimise and document variability during this critical experimental period. This review provides a detailed overview of the challenges inherent to a procedure often considered routine, highlighting the importance of carefully considering each aspect of SOPs for PBMC collection, cryopreservation, thawing, and culture to ensure accurate interpretation and comparison between studies.
Collapse
Affiliation(s)
- Daniel J. Browne
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Catherine M. Miller
- College of Medicine and Dentistry, James Cook University, Cairns, QLD, Australia
| | - Denise L. Doolan
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
5
|
Janetzki S. Important Considerations for ELISpot Validation. Methods Mol Biol 2024; 2768:1-13. [PMID: 38502384 DOI: 10.1007/978-1-0716-3690-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
The ELISpot assay has a solid place in the immune monitoring field for over 40 years. It is an assay that can assess the function of single immune cells in a straightforward and easy-to-learn approach. Its use in basic research, translational, and clinical work has been documented in countless publications. Harmonization guidelines and invaluable tools for optimal assay performance and evaluation exist. However, the validation of an established ELISpot protocol has been left to diverse opinions about how to interpret and tackle typical validation parameters. This chapter addresses important considerations for ELISpot validation, including the interpretations of validation parameters for a meaningful description of assay performance.
Collapse
|
6
|
Emami MR, Espinoza A, Young CS, Ma F, Farahat PK, Felgner PL, Chamberlain JS, Xu X, Pyle AD, Pellegrini M, Villalta SA, Spencer MJ. Innate and adaptive AAV-mediated immune responses in a mouse model of Duchenne muscular dystrophy. Mol Ther Methods Clin Dev 2023; 30:90-102. [PMID: 37746243 PMCID: PMC10512012 DOI: 10.1016/j.omtm.2023.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/08/2023] [Indexed: 09/26/2023]
Abstract
High systemic doses of adeno-associated viruses (AAVs) have been associated with immune-related serious adverse events (SAEs). Although AAV was well tolerated in preclinical models, SAEs were observed in clinical trials, indicating the need for improved preclinical models to understand AAV-induced immune responses. Here, we show that mice dual-dosed with AAV9 at 4-week intervals better recapitulate aspects of human immunity to AAV. In the model, anti-AAV9 immunoglobulin G (IgGs) increased in a linear fashion between the first and second AAV administrations. Complement activation was only observed in the presence of high levels of both AAV and anti-AAV IgG. Myeloid-derived pro-inflammatory cytokines were significantly induced in the same pattern as complement activation, suggesting that myeloid cell activation to AAV may rely on the presence of both AAV and anti-AAV IgG complexes. Single-cell RNA sequencing of peripheral blood mononuclear cells confirmed that activated monocytes were a primary source of pro-inflammatory cytokines and chemokines, which were significantly increased after a second AAV9 exposure. The same activated monocyte clusters expressed both Fcγ and complement receptors, suggesting that anti-AAV-mediated activation of myeloid cells through Fcγ receptors and/or complement receptors is one mechanism by which anti-AAV antigen complexes may prime antigen-presenting cells and amplify downstream immunity.
Collapse
Affiliation(s)
- Michael R. Emami
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alejandro Espinoza
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences – The Collaboratory, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Feiyang Ma
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Philip K. Farahat
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Philip L. Felgner
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Jeffrey S. Chamberlain
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, USA
- Department of Biochemistry, University of Washington School of Medicine, Seattle, WA, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - April D. Pyle
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
- Institute for Genomics and Proteomics, University of California, Los Angeles, Los Angeles, CA, USA
| | - S. Armando Villalta
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
- Institute of Immunology, University of California, Irvine, Irvine, CA, USA
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Melissa J. Spencer
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
7
|
Essawi K, Hakami W, Naeem Khan MB, Martin R, Zeng J, Chu R, Uchida N, Bonifacino AC, Krouse AE, Linde NS, Donahue RE, Blobel GA, Gerdemann U, Kean LS, Maitland SA, Wolfe SA, Metais JY, Gottschalk S, Bauer DE, Tisdale JF, Demirci S. Pre-existing immunity does not impair the engraftment of CRISPR-Cas9-edited cells in rhesus macaques conditioned with busulfan or radiation. Mol Ther Methods Clin Dev 2023; 29:483-493. [PMID: 37273902 PMCID: PMC10236215 DOI: 10.1016/j.omtm.2023.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/17/2023] [Indexed: 06/06/2023]
Abstract
CRISPR-Cas9-based therapeutic genome editing approaches hold promise to cure a variety of human diseases. Recent findings demonstrate pre-existing immunity for the commonly used Cas orthologs from Streptococcus pyogenes (SpCas9) and Staphylococcus aureus (SaCas9) in humans, which threatens the success of this powerful tool in clinical use. Thus, a comprehensive investigation and potential risk assessment are required to exploit the full potential of the system. Here, we investigated existence of immunity to SpCas9 and SaCas9 in control rhesus macaques (Macaca mulatta) alongside monkeys transplanted with either lentiviral transduced or CRISPR-SpCas9 ribonucleoprotein (RNP)-edited cells. We observed significant levels of Cas9 antibodies in the peripheral blood of all transplanted and non-transplanted control animals. Transplantation of ex vivo transduced or SpCas9-mediated BCL11A enhancer-edited cells did not alter the levels of Cas9 antibodies in rhesus monkeys. Following stimulation of peripheral blood cells with SpCas9 or SaCas9, neither Cas9-specific T cells nor cytokine induction were detected. Robust and durable editing frequencies and expression of high levels of fetal hemoglobin in BCL11A enhancer-edited rhesus monkeys with no evidence of an immune response (>3 years) provide an optimistic outlook for the use of ex vivo CRISPR-SpCas9 (RNP)-edited cells.
Collapse
Affiliation(s)
- Khaled Essawi
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| | - Waleed Hakami
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| | - Muhammad Behroz Naeem Khan
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Reid Martin
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Jing Zeng
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA, USA
| | - Rebecca Chu
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Naoya Uchida
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | | | - Allen E. Krouse
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD, USA
| | | | - Robert E. Donahue
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Gerd A. Blobel
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ulrike Gerdemann
- Boston Children’s Hospital, Department of Pediatric Oncology, Dana Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Leslie S. Kean
- Boston Children’s Hospital, Department of Pediatric Oncology, Dana Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Stacy A. Maitland
- Department of Molecular, Cell and Cancer Biology, Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Scot A. Wolfe
- Department of Molecular, Cell and Cancer Biology, Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jean-Yves Metais
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Daniel E. Bauer
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA, USA
| | - John F. Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Selami Demirci
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
8
|
Gorovits B, Azadeh M, Buchlis G, Fiscella M, Harrison T, Havert M, Janetzki S, Jawa V, Long B, Mahnke YD, McDermott A, Milton M, Nelson R, Vettermann C, Wu B. Evaluation of Cellular Immune Response to Adeno-Associated Virus-Based Gene Therapy. AAPS J 2023; 25:47. [PMID: 37101079 PMCID: PMC10132926 DOI: 10.1208/s12248-023-00814-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/14/2023] [Indexed: 04/28/2023] Open
Abstract
The number of approved or investigational late phase viral vector gene therapies (GTx) has been rapidly growing. The adeno-associated virus vector (AAV) technology continues to be the most used GTx platform of choice. The presence of pre-existing anti-AAV immunity has been firmly established and is broadly viewed as a potential deterrent for successful AAV transduction with a possibility of negative impact on clinical efficacy and a connection to adverse events. Recommendations for the evaluation of humoral, including neutralizing and total antibody based, anti-AAV immune response have been presented elsewhere. This manuscript aims to cover considerations related to the assessment of anti-AAV cellular immune response, including review of correlations between humoral and cellular responses, potential value of cellular immunogenicity assessment, and commonly used analytical methodologies and parameters critical for monitoring assay performance. This manuscript was authored by a group of scientists involved in GTx development who represent several pharma and contract research organizations. It is our intent to provide recommendations and guidance to the industry sponsors, academic laboratories, and regulatory agencies working on AAV-based GTx viral vector modalities with the goal of achieving a more consistent approach to anti-AAV cellular immune response assessment.
Collapse
Affiliation(s)
| | - Mitra Azadeh
- Ultragenyx Pharmaceutical Inc, Novato, California, USA
| | - George Buchlis
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Mike Havert
- Gene Therapy Partners, San Diego, California, USA
| | | | - Vibha Jawa
- Bristol Myers Squibb Pharmaceutical, Princeton, New Jersey, USA
| | - Brian Long
- BioMarin Pharmaceutical Inc, Novato, California, USA
| | | | - Andrew McDermott
- Labcorp Early Development Laboratories Inc, Indianapolis, Indiana, USA
| | - Mark Milton
- Lake Boon Pharmaceutical Consulting LLC, Hudson, New York, USA
| | | | | | - Bonnie Wu
- Janssen Pharmaceuticals, Raritan, New Jersey, USA
| |
Collapse
|
9
|
Rongkard P, Dunachie SJ, Kronsteiner B. Impact of shipping temperature on cell viability and T cell responses to bacterial antigens. Wellcome Open Res 2023; 8:188. [PMID: 38903244 PMCID: PMC11187529 DOI: 10.12688/wellcomeopenres.18822.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 06/22/2024] Open
Abstract
Background: Interferon-γ (IFN-γ) secretion by T cells is a key correlate of immune protection against many pathogens including tuberculosis and the neglected tropical disease melioidosis. Clinical studies in tropical regions of immune responses to pathogens and vaccine monitoring studies require the collection of samples in resource-limited rural areas and subsequent shipment to central laboratories for downstream assays and long-term storage. Here, we studied the impact of two different shipping temperatures on the viability, composition and function of peripheral blood mononuclear cells (PBMC) using multi-colour flow cytometry and IFN-γ enzyme-linked immunospot assay (IFN-γ ELISpot), in order to provide guidance on sample shipment conditions for future clinical studies. Methods: Paired peripheral blood mononuclear cell (PBMC) samples from recovered melioidosis patients were stored in liquid nitrogen (-196°C) and then shipped from Bangkok, Thailand to Oxford, UK at either -80°C (dry ice) or -196°C (dry shipper). After thawing, cell viability and composition were assessed by flow cytometry and antigen specific responses to Burkholderia pseudomallei (BP) were measured using IFN-γ ELISpot. Results: We observed modest lowering of viability in the majority of samples and a reduction in IFN-γ responses to BP which correlated to a decrease of monocytes and natural killer cells in samples shipped at -80°C compared to -196°C. Despite being lower in magnitude antigen-specific responses remained detectable in the majority of samples. Conclusions: Here we demonstrate that shipment of cryopreserved PBMC at -196°C has a benefit on cell viability, recovery and T cell responses to bacterial antigens, although useful information can still be obtained from samples shipped at -80°C, thus providing important guidance for sample management in future clinical trials.
Collapse
Affiliation(s)
- Patpong Rongkard
- Faculty of Tropical Medicine, Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7LG, UK
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, OX1 3SY, UK
| | - Susanna J. Dunachie
- Faculty of Tropical Medicine, Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7LG, UK
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, OX1 3SY, UK
| | - Barbara Kronsteiner
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7LG, UK
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, OX1 3SY, UK
| |
Collapse
|
10
|
Mouton W, Conrad A, Alcazer V, Boccard M, Bodinier M, Oriol G, Subtil F, Labussière-Wallet H, Ducastelle-Lepretre S, Barraco F, Balsat M, Fossard G, Brengel-Pesce K, Ader F, Trouillet-Assant S. Distinct Immune Reconstitution Profiles Captured by Immune Functional Assays at 6 Months Post Allogeneic Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2023; 29:94.e1-94.e13. [PMID: 36336259 DOI: 10.1016/j.jtct.2022.10.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 11/05/2022]
Abstract
Immune reconstitution after allogeneic-hematopoietic-stem-cell transplantation (allo-HSCT) is a complex and individual process. In this cross-sectional study, whole-blood (WB) immune functional assay (IFA) was used to characterize immune function by assessing immune-related gene/pathway alterations. The usefulness of this tool in the context of infection, 6 months after transplantation, was evaluated. Sixty allo-HSCT recipients at 6 months after transplantation and 10 healthy volunteers (HV) were included. WB was stimulated in standardized TruCulture tubes using lipopolysaccharides and Staphylococcal enterotoxin B. Gene expression was quantified using a custom 144-gene panel using NanoString nCounter technology and analyzed using Ingenuity Pathway Analysis. The relationships between immune function and clinical characteristics, immune cell counts, and post-transplantation infections were assessed. Allo-HSCT recipients were able to activate similar networks of the innate and adaptive immune response compared to HV, with, nevertheless, a lower intensity. A reduced number and a lower expression of genes associated with immunoregulatory and inflammatory processes were observed in allo-HSCT recipients. The use of immunosuppressive treatments was associated with a protracted immune reconstitution revealed by transcriptomic immunoprofiling. No difference in immune cell counts was observed among patients receiving or not receiving immunosuppressive treatments using a large immunophenotyping panel. Moreover, the expression of a set of genes, including CCL3/CCL4, was significantly lower in patients with Herpesviridae reactivation (32%, 19/60), which once again was not identified using classical immune cell counts. Transcriptional IFA revealed the heterogeneity among allo-HSCT recipients with a reduced immune function, a result that could not be captured by circulating immune cell counts. This highlights the potential added value of this tool for the personalized care of immunocompromised patients.
Collapse
Affiliation(s)
- William Mouton
- Joint Research Unit Hospices Civils de Lyon-bioMérieux, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France; Virology and Human Pathology - Virpath Team, International Centre for Research in Infectiology (CIRI), Claude Bernard Lyon 1 University, Lyon, France
| | - Anne Conrad
- Legionella Pathogenesis Team, International Centre for Research in Infectiology (CIRI), Claude Bernard Lyon 1 University, Lyon, France; Infectious and Tropical Diseases Department, Hospices Civils de Lyon, Croix-Rousse Hospital, Lyon, France; Claude Bernard Lyon I University, Villeurbanne, France
| | - Vincent Alcazer
- Clinical Hematology Department, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France; LIB TEAM, International Centre for Research in Infectiology (CIRI), Oullins, France
| | - Mathilde Boccard
- Joint Research Unit Hospices Civils de Lyon-bioMérieux, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France; Legionella Pathogenesis Team, International Centre for Research in Infectiology (CIRI), Claude Bernard Lyon 1 University, Lyon, France; Infectious and Tropical Diseases Department, Hospices Civils de Lyon, Croix-Rousse Hospital, Lyon, France
| | - Maxime Bodinier
- Joint Research Unit Hospices Civils de Lyon-bioMérieux, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | - Guy Oriol
- Joint Research Unit Hospices Civils de Lyon-bioMérieux, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | - Fabien Subtil
- Biostatistics Department, Hospices Civils de Lyon, Lyon France, Lyon 1 University, Villeurbanne, France; CNRS, Biometrics and Evolutionary Biology Laboratory UMR, Villeurbanne, France
| | - Hélène Labussière-Wallet
- Clinical Hematology Department, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | | | - Fiorenza Barraco
- Clinical Hematology Department, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | - Marie Balsat
- Clinical Hematology Department, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | - Gaëlle Fossard
- Clinical Hematology Department, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | - Karen Brengel-Pesce
- Joint Research Unit Hospices Civils de Lyon-bioMérieux, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | - Florence Ader
- Legionella Pathogenesis Team, International Centre for Research in Infectiology (CIRI), Claude Bernard Lyon 1 University, Lyon, France; Infectious and Tropical Diseases Department, Hospices Civils de Lyon, Croix-Rousse Hospital, Lyon, France; Claude Bernard Lyon I University, Villeurbanne, France.
| | - Sophie Trouillet-Assant
- Joint Research Unit Hospices Civils de Lyon-bioMérieux, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France; Virology and Human Pathology - Virpath Team, International Centre for Research in Infectiology (CIRI), Claude Bernard Lyon 1 University, Lyon, France
| |
Collapse
|
11
|
Ikeda D, Imano H, Mori Y, Asada H, Yamanishi K, Okuno Y, Iso H. Longitudinal changes in cell-mediated immunity after varicella-zoster virus skin test in the general population; Shozu Herpes Zoster Study: SHEZ study. J Med Virol 2023; 95:e28336. [PMID: 36418204 DOI: 10.1002/jmv.28336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/28/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Varicella-zoster virus-specific cell-mediated immunity has been associated with the onset and severity of herpes zoster (HZ), and the administration of the HZ vaccine enhanced the immunity. However, limited data is available on the duration of cell-mediated immunity enhancement by soluble antigen of varicella-zoster virus (VZV) skin test. A prospective, community-based cohort study was conducted in Shozu County, Kagawa Prefecture, Japan. Repeated VZV skin tests containing inactivated VZV antigen and blood tests were performed on 365 subjects aged 60 years and older at baseline, 1, 2, and 3 years later. The differential immunity indices of VZV over time for cell-mediated and humoral immunity were evaluated. VZV skin test reaction and ELISpot counts increased significantly at 1, 2, and 3 years later compared to the baseline. However, humoral immunity indices did not change materially over time. Soluble antigen by VZV skin test enhanced VZV-specific cell-mediated immunity, and it persisted for at least 1 year. In addition, the inoculation with inactivated antigens every year by VZV skin test continued to enhance VZV-specific cell-mediated immunity after 2 and 3 years.
Collapse
Affiliation(s)
- Daisuke Ikeda
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hironori Imano
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine, Osaka, Japan.,Department of Public Health, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Yasuko Mori
- Division of Clinical Virology, Department of Microbiology and Infectious Diseases, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Hideo Asada
- Department of Dermatology, Nara Medical University School of Medicine, Nara, Japan
| | - Koichi Yamanishi
- The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
| | - Yoshinobu Okuno
- The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
| | - Hiroyasu Iso
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine, Osaka, Japan.,Institute of Global Health Policy Research (iGHP), Bureau of International Health Cooperation, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
12
|
Koldehoff M, Horn PA, Lindemann M. Cellular Immune Response after Vaccination with an Adjuvanted, Recombinant Zoster Vaccine in Allogeneic Hematopoietic Stem Cell Transplant Recipients. Vaccines (Basel) 2022; 10:809. [PMID: 35632565 PMCID: PMC9143460 DOI: 10.3390/vaccines10050809] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/15/2022] Open
Abstract
Hematopoietic stem cell transplant (HSCT) recipients have a high risk of developing primary varicella-zoster virus (VZV) infection and reactivation. VZV vaccination may prevent infection and reactivation. In the current study, recipients of allogeneic HSCT (34 females, 45 males) were vaccinated with adjuvanted, recombinant zoster vaccine Shingrix™, which contains the VZV glycoprotein E. Cellular immunity against various VZV antigens was analyzed by interferon-gamma ELISpot. Peripheral blood mononuclear cells (PBMC) of recipients with versus without prior shingles (n = 36 and n = 43, respectively) showed approximately twofold higher VZV-specific responses prior to and post vaccination. After the first and second vaccination, ELISpot responses towards the glycoprotein E were significantly higher in males versus females (median of spots increment 18 versus 1 and 17 versus 4, respectively, p ≤ 0.02 each). Multivariate analysis showed that shingles and sex both impacts significantly on VZV immunity. Whereas vaccination-induced changes could hardly be detected after stimulation with a whole VZV antigen, there was a significant increase in responses towards glycoprotein E after vaccination (p < 0.005). These data indicate that vaccination with Shingrix™ augmented cellular, VZV-specific immunity in HSCT recipients. Shingles and male sex could both be identified as factors leading to increased immunity.
Collapse
Affiliation(s)
- Michael Koldehoff
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany;
- Department of Hygiene and Environmental Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Peter A. Horn
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany;
| | - Monika Lindemann
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany;
| |
Collapse
|
13
|
Otani N, Shima M, Yamamoto T, Okuno T. Effect of Routine Varicella Immunization on the Epidemiology and Immunogenicity of Varicella and Shingles. Viruses 2022; 14:v14030588. [PMID: 35336994 PMCID: PMC8954496 DOI: 10.3390/v14030588] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 12/30/2022] Open
Abstract
Varicella-zoster virus (VZV) causes varicella as a primary infection and remains latent in the ganglia until it becomes reactivated to cause herpes zoster. Individuals with varicella develop adaptive humoral and cell-mediated immunity. Compromised cell-mediated immunity is thought to contribute to the development of herpes zoster. Recent evidence suggests that changes in the epidemiology of varicella have affected the epidemiology of herpes zoster. The incidence of herpes zoster is higher in older adults; thus, the herpes zoster vaccine is recommended for older adults. However, the incidence of herpes zoster is expected to rise among younger individuals; hence, vaccination with the varicella vaccine should also be considered in younger adults. In order to determine the need for vaccination in different populations, it is important to establish methods to accurately assess the activity of cell-mediated immunity and humoral immunity.
Collapse
Affiliation(s)
- Naruhito Otani
- Department of Public Health, Hyogo College of Medicine, Nishinomiya 663-8501, Japan;
- Correspondence: ; Tel.: +81-798-45-6566
| | - Masayuki Shima
- Department of Public Health, Hyogo College of Medicine, Nishinomiya 663-8501, Japan;
| | - Takuma Yamamoto
- Department of Legal Medicine, Hyogo College of Medicine, Nishinomiya 663-8501, Japan;
| | - Toshiomi Okuno
- Department of Microbiology, Hyogo College of Medicine, Nishinomiya 663-8501, Japan;
| |
Collapse
|
14
|
Mak WA, Koeleman JG, Ong DS. Development of an in-house SARS-CoV-2 interferon-gamma ELISpot and plate reader-free spot detection method. J Virol Methods 2022; 300:114398. [PMID: 34863782 PMCID: PMC8634702 DOI: 10.1016/j.jviromet.2021.114398] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 11/30/2022]
Abstract
Coronavirus disease 2019 (COVID-19) vaccination programs rolled out in an attempt to stop the COVID-19 pandemic. Besides neutralising antibodies, effective T cell responses are also crucial for protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and COVID-19 disease severity. To assess SARS-CoV-2-specific T cell immunity, we developed an interferon-gamma (IFN-γ) enzyme-linked immunospot (ELISpot) that can be deployed in research and diagnostic settings. We optimised our ELISpot by testing multiple antigen concentrations to stimulate peripheral blood mononuclear cells of SARS-CoV-2-unexposed, COVID-19 convalescent and COVID-19 vaccinated volunteers. Also, we developed an ELISpot plate reader-free method to detect and quantify spots, which we compared to manual spot counting and automated analysis by an ELISpot plate reader. We observed strong SARS-CoV-2-reactive T cell responses in COVID-19 convalescent, and COVID-19 vaccinated volunteers but absent or only weak responses in unexposed volunteers. Overall, antigens with concentrations from 0.1 to 5.0 μg/mL per peptide elicited similar T cell responses. Also, our plate reader-free detection method reliably detected and quantified SARS-CoV-2-specific T cells, demonstrated by an excellent reliability when compared to manual analysis and automated analysis by an ELISpot plate reader.
Collapse
Affiliation(s)
- Willem A. Mak
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Kleiweg 500, 3045PM Rotterdam, the Netherlands
| | - Johannes G.M. Koeleman
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Kleiweg 500, 3045PM Rotterdam, the Netherlands
| | - David S.Y. Ong
- Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Kleiweg 500, 3045PM Rotterdam, the Netherlands,Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Universiteitsweg 100, 3584GC Utrecht, the Netherlands,Corresponding author at: Department of Medical Microbiology and Infection Control, Franciscus Gasthuis & Vlietland, Kleiweg 500, 3045PM Rotterdam, the Netherlands
| |
Collapse
|
15
|
Endonuclease increases efficiency of osteoblast isolation from murine calvariae. Sci Rep 2021; 11:8502. [PMID: 33875686 PMCID: PMC8055883 DOI: 10.1038/s41598-021-87716-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/25/2021] [Indexed: 11/08/2022] Open
Abstract
Bone is a highly dynamic organ that undergoes remodeling equally regulated by osteoblast-mediated bone formation and osteoclast-mediated bone resorption. To clarify the regulation of osteoblastogenesis, primary murine osteoblasts are required for an in vitro study. Primary osteoblasts are isolated from neonatal calvariae through digestion with collagenase. However, the number of cells collected from one pup is not sufficient for further in vitro experiments, leading to an increase in the use of euthanized pups. We hypothesized that the viscosity of digested calvariae and digestion solution supplemented with collagenase results in cell clumping and reduction of isolated cells from bones. We simply added Benzonase, a genetically engineered endonuclease that shears all forms of DNAs/RNAs, in order to reduce nucleic acid-mediated viscosity. We found that addition of Benzonase increased the number of collected osteoblasts by three fold compared to that without Benzonase through reduction of viscosity. Additionally, Benzonase has no effect on cellular identity and function. The new osteoblast isolation protocol with Benzonase minimizes the number of neonatal pups required for an in vitro study and expands the concept that isolation of other populations of cells including osteocytes that are difficult to be purified could be modified by Benzonase.
Collapse
|
16
|
Chun JY, Kim K, Lee MK, Kang CK, Koh Y, Shin DY, Hong J, Choe PG, Kim NJ, Yoon SS, Park WB, Kim I, Oh MD. Immunogenicity and safety of a live herpes zoster vaccine in hematopoietic stem cell transplant recipients. BMC Infect Dis 2021; 21:117. [PMID: 33499826 PMCID: PMC7836155 DOI: 10.1186/s12879-021-05806-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/15/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Herpes zoster (HZ) infection of hematopoietic stem cell transplant (HSCT) patients is of clinical concern. Vaccination could help restore immunity to varicella zoster virus (VZV); however, temporal changes in immunogenicity and safety of live HZ vaccines after HSCT is still unclear. The aim of this study was to elucidate the temporal immunogenicity and safety of the HZ vaccine according to time since HSCT and to determine optimal timing of vaccination. METHODS Live HZ vaccine was administered to patients 2-5 years or > 5 years post-HSCT. Control groups comprised patients with a hematologic malignancy who received cytotoxic chemotherapy and healthy volunteers. Humoral and cellular immunogenicity were measured using a glycoprotein enzyme-linked immunosorbent assay (gpELISA) and an interferon-γ (IFN-γ) enzyme-linked immunospot (ELISPOT) assay. Vaccine-related adverse events were also monitored. RESULTS Fifty-six patients with hematologic malignancy (41 in the HSCT group and 15 in the chemotherapy group) along with 30 healthy volunteers were enrolled. The geometric mean fold rises (GMFRs) in humoral immune responses of the 2-5 year and > 5 year HSCT groups, and the healthy volunteer group, were comparable and significantly higher than that of the chemotherapy group (3.15, 95% CI [1.96-5.07] vs 5.05, 95% CI [2.50-10.20] vs 2.97, 95% CI [2.30-3.83] vs 1.42, 95% CI [1.08-1.86]). The GMFR of cellular immune responses was highest in the HSCT 2-5 year group and lowest in the chemotherapy group. No subject suffered clinically significant adverse events or reactivation of VZV within the follow-up period. CONCLUSION Our findings demonstrate that a live HZ vaccine is immunogenic and safe when administered 2 years post-HSCT.
Collapse
Affiliation(s)
- June Young Chun
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Present affiliation: Department of Internal Medicine, National Cancer Center, Goyang, South Korea
| | - Kichun Kim
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Min Kyeong Lee
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Chang Kyung Kang
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Dong-Yeop Shin
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Junshik Hong
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Pyoeng Gyun Choe
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Nam Joong Kim
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Wan Beom Park
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Inho Kim
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Myoung-Don Oh
- Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| |
Collapse
|
17
|
Vakkilainen S, Kleino I, Honkanen J, Salo H, Kainulainen L, Gräsbeck M, Kekäläinen E, Mäkitie O, Klemetti P. The Safety and Efficacy of Live Viral Vaccines in Patients With Cartilage-Hair Hypoplasia. Front Immunol 2020; 11:2020. [PMID: 32849667 PMCID: PMC7432140 DOI: 10.3389/fimmu.2020.02020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/27/2020] [Indexed: 12/17/2022] Open
Abstract
Background Live viral vaccines are generally contraindicated in patients with combined immunodeficiency including cartilage-hair hypoplasia (CHH); however, they may be tolerated in milder syndromes. We evaluated the safety and efficacy of live viral vaccines in patients with CHH. Methods We analyzed hospital and immunization records of 104 patients with CHH and measured serum antibodies to measles, mumps, rubella, and varicella zoster virus (VZV) in all patients who agreed to blood sampling (n = 50). We conducted a clinical trial (ClinicalTrials.gov identifier: NCT02383797) of live VZV vaccine on five subjects with CHH who lacked varicella history, had no clinical symptoms of immunodeficiency, and were seronegative for VZV; humoral and cellular immunologic responses were assessed post-immunization. Results A large proportion of patients have been immunized with live viral vaccines, including measles-mumps-rubella (MMR) (n = 40, 38%) and VZV (n = 10, 10%) vaccines, with no serious adverse events. Of the 50 patients tested for antibodies, previous immunization has been documented with MMR (n = 22), rubella (n = 2) and measles (n = 1) vaccines. Patients with CHH demonstrated seropositivity rates of 96%/75%/91% to measles, mumps and rubella, respectively, measured at a medium of 24 years post-immunization. Clinical trial participants developed humoral and cellular responses to VZV vaccine. One trial participant developed post-immunization rash and knee swelling, both resolved without treatment. Conclusion No serious adverse events have been recorded after immunization with live viral vaccines in Finnish patients with CHH. Patients generate humoral and cellular immune response to live viral vaccines. Immunization with live vaccines may be considered in selected CHH patients with no or clinically mild immunodeficiency.
Collapse
Affiliation(s)
- Svetlana Vakkilainen
- Children’s Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Iivari Kleino
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Jarno Honkanen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Harri Salo
- Children’s Hospital, Clinicum, University of Helsinki, Helsinki, Finland
| | - Leena Kainulainen
- Department of Pediatrics and Adolescents, Turku University Hospital, University of Turku, Turku, Finland
| | - Michaela Gräsbeck
- Department of Pediatrics, Kymenlaakso Central Hospital, Kotka, Finland
| | - Eliisa Kekäläinen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital, HUSLAB, Division of Clinical Microbiology, Helsinki, Finland
| | - Outi Mäkitie
- Children’s Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Paula Klemetti
- Children’s Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
18
|
Boeckh MJ, Arvin AM, Mullane KM, Camacho LH, Winston DJ, Morrison VA, Hurtado K, Durrand Hall J, Pang L, Su SC, Kaplan SS, Annunziato PW, Popmihajlov Z. Immunogenicity of Inactivated Varicella Zoster Vaccine in Autologous Hematopoietic Stem Cell Transplant Recipients and Patients With Solid or Hematologic Cancer. Open Forum Infect Dis 2020; 7:ofaa172. [PMID: 32665955 PMCID: PMC7336559 DOI: 10.1093/ofid/ofaa172] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/27/2020] [Indexed: 11/13/2022] Open
Abstract
Background In phase 3 trials, inactivated varicella zoster virus (VZV) vaccine (ZVIN) was well tolerated and efficacious against herpes zoster (HZ) in autologous hematopoietic stem cell transplant (auto-HSCT) recipients and patients with solid tumor malignancies receiving chemotherapy (STMc) but did not reduce HZ incidence in patients with hematologic malignancies (HMs). Here, we describe ZVIN immunogenicity from these studies. Methods Patients were randomized to ZVIN or placebo (4 doses). Immunogenicity was assessed by glycoprotein enzyme-linked immunosorbent assay (gpELISA) and VZV interferon (IFN)-γ enzyme-linked immunospot (ELISPOT) assay in patients receiving all 4 doses without developing HZ at the time of blood sampling. Results Estimated geometric mean fold rise ratios (ZVIN/placebo) by gpELISA and IFN-y ELISPOT ~28 days post-dose 4 were 2.02 (95% confidence interval [CI], 1.53-2.67) and 5.41 (95% CI, 3.60-8.12) in auto-HSCT recipients; 1.88 (95% CI, 1.79-1.98) and 2.10 (95% CI, 1.69-2.62) in patients with STMc; and not assessed and 2.35 (95% CI, 1.81-3.05) in patients with HM. Conclusions ZVIN immunogenicity was directionally consistent with clinical efficacy in auto-HSCT recipients and patients with STMc even though HZ protection and VZV immunity were not statistically correlated. Despite a lack of clinical efficacy in patients with HM, ZVIN immunogenicity was observed in this population. Immunological results did not predict vaccine efficacy in these 3 populations. Clinical trial registration NCT01229267, NCT01254630.
Collapse
Affiliation(s)
- Michael J Boeckh
- Vaccine and Infectious Disease & Clinical Research Divisions, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Ann M Arvin
- Microbiology & Immunology Departments, Stanford University School of Medicine, Stanford, California, USA
| | | | - Luis H Camacho
- Medical Oncology, Oncology Consultants, Houston, Texas, USA
| | - Drew J Winston
- Department of Medicine, University of California Los Angeles Medical Center, Los Angeles, California, USA
| | - Vicki A Morrison
- Hematology Oncology Division, Hennepin County Medical Center, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | - Lei Pang
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Shu-Chih Su
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| | | | | | | | | |
Collapse
|
19
|
Rubio-Carrasquilla M, Santa CD, Rendón JP, Botero-Garcés J, Guimarães AJ, Moreno E, Cano LE. An interferon gamma release assay specific for Histoplasma capsulatum to detect asymptomatic infected individuals: A proof of concept study. Med Mycol 2020; 57:724-732. [PMID: 30534945 DOI: 10.1093/mmy/myy131] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/14/2018] [Accepted: 11/07/2018] [Indexed: 12/17/2022] Open
Abstract
Histoplasmosis is the most common endemic mycosis in the Americas. Currently, there is no laboratory test capable to detect subclinical or latent infections by Histoplasma capsulatum (Hc), which might develop as severe infections in immunocompromised individuals. For the first time to our knowledge, we explore the suitability of an interferon gamma release assay (IGRA) to detect latent Hc infection in asymptomatic individuals. A cohort of 126 volunteers was enrolled in the study, 13 of which underwent a Hc infection in the past, and 93 of them showing risk factors for this infection. The remaining 20 participants did not refer any risk factors of Hc infection, but eight of them showed evidences of infection with Mycobacterium tuberculosis. All participants were recruited in Medellin, Colombia, between January 2014 and December 2017. Whole blood samples were cultured with four different Hc crude antigens and phytohemaglutinin as positive control. The interferon (IFN)-γ released by T lymphocytes upon antigen stimulation was quantified by ELISA. A defined cutoff value of 20 pg/ml for the IFN-γ concentration allowed us to distinguish between the group with documented past infections and the group of noninfected individuals with high sensitivity (70-92%) and specificity (85-95%), for the four tested antigens. Positive 82-95% and negative 77-92% predictive values were also very high, comparable to those reported for commercially available IGRAs. The new test constitutes a promising screening method to detect individuals with latent Hc infection, even decades after the primary infection, as evidenced in this study.
Collapse
Affiliation(s)
- Marcela Rubio-Carrasquilla
- Grupo de Micología Médica y Experimental, Corporación para Investigaciones Biológicas (CIB), Medellín, Colombia.,Instituto de Biología, Universidad de Antioquia, Medellín, Colombia
| | | | - Juan Pablo Rendón
- Grupo de Micología Médica y Experimental, Corporación para Investigaciones Biológicas (CIB), Medellín, Colombia
| | | | - Allan J Guimarães
- Depto de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói-RJ, Brasil
| | - Ernesto Moreno
- Facultad de Ciencias Básicas, Universidad de Medellín, Medellín, Colombia
| | - Luz Elena Cano
- Grupo de Micología Médica y Experimental, Corporación para Investigaciones Biológicas (CIB), Medellín, Colombia.,Escuela de Microbiología, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
20
|
Smith C, Moraka NO, Ibrahim M, Moyo S, Mayondi G, Kammerer B, Leidner J, Gaseitsiwe S, Li S, Shapiro R, Lockman S, Weinberg A. Human Immunodeficiency Virus Exposure but Not Early Cytomegalovirus Infection Is Associated With Increased Hospitalization and Decreased Memory T-Cell Responses to Tetanus Vaccine. J Infect Dis 2020; 221:1167-1175. [PMID: 31711179 PMCID: PMC7075416 DOI: 10.1093/infdis/jiz590] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/07/2019] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV)-exposed, uninfected (HEU) infants experience high rates of infectious morbidity. We hypothesized that early cytomegalovirus (CMV) infection was associated with increased hospitalization rates and decreased vaccine responses in HEU compared with HIV-unexposed (HUU) infants. METHODS Among infants enrolled in the Tshipidi study in Botswana, we determined CMV infection status by 6 months of age and compared hospitalization rates and responses to tetanus and Bacille Calmette-Guérin vaccines among HEU and HUU vaccinees. RESULTS Fifteen of 226 (6.6%) HEU infants and 17 (19.3%) of 88 HUU infants were CMV-infected by 6 months. The HEU infants were approximately 3 times as likely to be hospitalized compared with HUU infants (P = .02). The HEU peripheral blood cells produced less interleukin (IL)-2 (P = .004), but similar amounts of interferon-γ, after stimulation with tetanus toxoid. Antitetanus immunoglobulin G titers were similar between groups. Cellular responses to purified protein derivative stimulation did not differ between groups. Maternal receipt of 3-drug antiretroviral therapy compared with zidovudine was associated with increased IL-2 expression after tetanus toxoid stimulation. The infants' CMV infection status was not associated with clinical or vaccine response outcomes. CONCLUSIONS We observed that increased rates of hospitalization and decreased memory T-cell responses to tetanus vaccine were associated with HIV exposure and incomplete treatment of maternal HIV infection, but not early CMV infection.
Collapse
Affiliation(s)
- Christiana Smith
- University of Colorado School of Medicine, Denver, Colorado, USA
| | - Natasha O Moraka
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Stellenbosch University, Stellenbosch, South Africa
| | | | - Sikhulile Moyo
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Gloria Mayondi
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Betsy Kammerer
- Boston Children’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Jean Leidner
- Goodtables Data Consulting, Norman, Oklahoma, USA
| | - Simani Gaseitsiwe
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Shaobing Li
- University of Colorado School of Medicine, Denver, Colorado, USA
| | - Roger Shapiro
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Shahin Lockman
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Adriana Weinberg
- University of Colorado School of Medicine, Denver, Colorado, USA
| |
Collapse
|
21
|
Bacterial non-specific nucleases of the phospholipase D superfamily and their biotechnological potential. Appl Microbiol Biotechnol 2020; 104:3293-3304. [PMID: 32086594 DOI: 10.1007/s00253-020-10459-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 12/19/2022]
Abstract
Bacterial non-specific nucleases are ubiquitously distributed and involved in numerous intra- and extracellular processes. Although all nucleases share the basic chemistry for the hydrolysis of phosphodiester bonds in nucleic acid molecules, the catalysis comprises diverse modes of action, which offers great potential for versatile biotechnological applications. A major criterium for their differentiation is substrate specificity. Specific endonucleases are widely used as restriction enzymes in molecular biology approaches, whereas the main applications of non-specific nucleases (NSNs) are the removal of nucleic acids from crude extracts in industrial downstream processing and the prevention of cell clumping in microfabricated channels. In nature, the predominant role of NSNs is the acquisition of nutrient sources such as nucleotides and phosphates. The number of extensively characterized NSNs and available structures is limited. Moreover, their applicability is mostly challenged by the presence of metal chelators that impede the hydrolysis of nucleic acids in a metal ion-dependent manner. However, a few metal ion-independent NSNs that tolerate the presence of metal chelators have been characterized in recent years with none being commercially available to date. The classification and biotechnological potential of bacterial NSNs with a special focus on metal ion-independent nucleases are presented and discussed.Key Points • Bacterial phospholipases (PLD-family) exhibit nucleolytic activity. • Bacterial nucleases of the PLD-family are metal ion-independent. • NSNs can be used in downstream processing approaches.
Collapse
|
22
|
West Nile or Usutu Virus? A Three-Year Follow-Up of Humoral and Cellular Response in a Group of Asymptomatic Blood Donors. Viruses 2020; 12:v12020157. [PMID: 32013152 PMCID: PMC7077259 DOI: 10.3390/v12020157] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/17/2020] [Accepted: 01/26/2020] [Indexed: 11/23/2022] Open
Abstract
West Nile virus (WNV) and Usutu virus (USUV) are two related arboviruses (genus Flavivirus, family Flaviviridae), with birds as a reservoir and mosquitoes as transmitting vectors. In recent years, WNV epidemiology changed in many European countries with increased frequency of outbreaks posing the issue of virus transmission risks by blood transfusion. USUV emerged for the first time in birds of the Tuscany region (Italy) in 1996 and in 2001 in Austria. While WNV is responsible for both mild and neuroinvasive diseases, USUV infection is usually asymptomatic and neuroinvasive symptoms are rare. Since WNV and USUV co-circulate, the surveillance of WNV allows also the detection of USUV. Due to the great similarity in amino-acid sequence of major surface proteins of the two viruses, a high cross-reactivity can lead to misinterpretation of serological results. Here, we report the results obtained from 54 asymptomatic blood donors during a three-year follow-up showing an unexpected high positivity (46.3%) for USUV. The major obstacle encountered in the differential diagnosis between these two viruses was the high cross-reactivity found in neutralizing antibodies (NT Abs) and, in some cases, a long follow-up was mandatory for a correct diagnosis. Moreover, two new ELISpot assays were developed for a more rapid and specific differential diagnosis, especially in those cases in which NT Abs were not determinant. Using a combination of Enzyme-linked immunospot (ELISpot), molecular, and serological tests, we could identify 25 true positive WNV and 25 true positive USUV blood donors. Our data highlight the importance of raising awareness for increasing USUV infections in endemic countries involved in blood transfusion and organ donation.
Collapse
|
23
|
Rubio-Carrasquilla M, Ochoa R, Santa C, Guimarães AJ, Cano LE, Moreno E. Identifying molecularly defined antigens for a Histoplasma capsulatum-specific interferon gamma release assay. Rev Iberoam Micol 2019; 36:186-191. [PMID: 31757594 DOI: 10.1016/j.riam.2019.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 05/05/2019] [Accepted: 06/14/2019] [Indexed: 10/25/2022] Open
Abstract
BACKGROUND In a previous work we showed the feasibility of an interferon gamma release assay (IGRA) for detecting latent infection by Histoplasma capsulatum. While in that proof-of-concept study we used crude fungal extracts as antigens, the newest IGRAs developed for other infections are based on molecularly defined antigens, mostly on mixtures of immunogenic peptides. AIMS To identify proteins in H. capsulatum that might serve as molecularly defined antigens for an IGRA test. METHODS We surveyed the literature looking for known H. capsulatum-immunogenic proteins and assayed two of them as antigens in an IGRA test, in a study that involved 80 volunteers. Furthermore, we used several bioinformatics tools to identify specific H. capsulatum proteins and to analyze possible strategies for the design of H. capsulatum-specific immunogenic peptides. RESULTS Seven H. capsulatum-immunogenic proteins were retrieved from the literature. IGRA tests using either the heat shock protein 60 or the M antigen showed high sensitivities but low specificities, most likely due to the high sequence similarity with the corresponding orthologs in other pathogenic microorganisms. We identified around 2000 H. capsulatum-specific proteins, most of which remain unannotated. Class II T-cell epitope predictions for a small number of these proteins showed a great variability among different alleles, prompting for a "brute force" approach for peptide design. CONCLUSIONS The H. capsulatum genome encodes a large number of distinctive proteins, which represent a valuable source of potential specific antigens for an IGRA test. Among them, the Cfp4 protein stands out as a very attractive candidate.
Collapse
Affiliation(s)
- Marcela Rubio-Carrasquilla
- Grupo de Micología Médica y Experimental, Corporación para Investigaciones Biológicas (CIB), Medellín, Colombia; Instituto de Biología, Universidad de Antioquia, Medellín, Colombia
| | - Rodrigo Ochoa
- Programa de Estudio y Control de Enfermedades Tropicales - PECET, Universidad de Antioquia, Medellín, Colombia
| | - Cristian Santa
- Universidad Nacional de Colombia, Sede Medellín, Medellín, Colombia
| | - Allan J Guimarães
- Depto de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Luz Elena Cano
- Grupo de Micología Médica y Experimental, Corporación para Investigaciones Biológicas (CIB), Medellín, Colombia; Escuela de Microbiología, Universidad de Antioquia, Medellín, Colombia
| | - Ernesto Moreno
- Facultad de Ciencias Básicas, Universidad de Medellín, Medellín, Colombia.
| |
Collapse
|
24
|
Mullane KM, Morrison VA, Camacho LH, Arvin A, McNeil SA, Durrand J, Campbell B, Su SC, Chan ISF, Parrino J, Kaplan SS, Popmihajlov Z, Annunziato PW, Cerana S, Dictar MO, Bonvehi P, Tregnaghi JP, Fein L, Ashley D, Singh M, Hayes T, Playford G, Morrissey O, Thaler J, Kuehr T, Greil R, Pecherstorfer M, Duck L, Van Eygen K, Aoun M, De Prijck B, Franke FA, Barrios CHE, Mendes AVA, Serrano SV, Garcia RF, Moore F, Camargo JFC, Pires LA, Alves RS, Radinov A, Oreshkov K, Minchev V, Hubenova AI, Koynova T, Ivanov I, Rabotilova B, Minchev V, Petrov PA, Chilingirov P, Karanikolov S, Raynov J, Grimard D, McNeil S, Kumar D, Larratt LM, Weiss K, Delage R, Diaz-Mitoma FJ, Cano PO, Couture F, Carvajal P, Yepes A, Torres Ulloa R, Fardella P, Caglevic C, Rojas C, Orellana E, Gonzalez P, Acevedo A, Galvez KM, Gonzalez ME, Franco S, Restrepo JG, Rojas CA, Bonilla C, Florez LE, Ospina AV, Manneh R, Zorica R, Vrdoljak DV, Samarzija M, Petruzelka L, Vydra J, Mayer J, Cibula D, Prausova J, Paulson G, Ontaneda M, Palk K, Vahlberg A, Rooneem R, Galtier F, Postil D, Lucht F, Laine F, Launay O, Laurichesse H, Duval X, Cornely OA, et alMullane KM, Morrison VA, Camacho LH, Arvin A, McNeil SA, Durrand J, Campbell B, Su SC, Chan ISF, Parrino J, Kaplan SS, Popmihajlov Z, Annunziato PW, Cerana S, Dictar MO, Bonvehi P, Tregnaghi JP, Fein L, Ashley D, Singh M, Hayes T, Playford G, Morrissey O, Thaler J, Kuehr T, Greil R, Pecherstorfer M, Duck L, Van Eygen K, Aoun M, De Prijck B, Franke FA, Barrios CHE, Mendes AVA, Serrano SV, Garcia RF, Moore F, Camargo JFC, Pires LA, Alves RS, Radinov A, Oreshkov K, Minchev V, Hubenova AI, Koynova T, Ivanov I, Rabotilova B, Minchev V, Petrov PA, Chilingirov P, Karanikolov S, Raynov J, Grimard D, McNeil S, Kumar D, Larratt LM, Weiss K, Delage R, Diaz-Mitoma FJ, Cano PO, Couture F, Carvajal P, Yepes A, Torres Ulloa R, Fardella P, Caglevic C, Rojas C, Orellana E, Gonzalez P, Acevedo A, Galvez KM, Gonzalez ME, Franco S, Restrepo JG, Rojas CA, Bonilla C, Florez LE, Ospina AV, Manneh R, Zorica R, Vrdoljak DV, Samarzija M, Petruzelka L, Vydra J, Mayer J, Cibula D, Prausova J, Paulson G, Ontaneda M, Palk K, Vahlberg A, Rooneem R, Galtier F, Postil D, Lucht F, Laine F, Launay O, Laurichesse H, Duval X, Cornely OA, Camerer B, Panse J, Zaiss M, Derigs HG, Menzel H, Verbeek M, Georgoulias V, Mavroudis D, Anagnostopoulos A, Terpos E, Cortes D, Umanzor J, Bejarano S, Galeano RW, Wong RSM, Hui P, Pedrazzoli P, Ruggeri L, Aversa F, Bosi A, Gentile G, Rambaldi A, Contu A, Marei L, Abbadi A, Hayajneh W, Kattan J, Farhat F, Chahine G, Rutkauskiene J, Marfil Rivera LJ, Lopez Chuken YA, Franco Villarreal H, Lopez Hernandez J, Blacklock H, Lopez RI, Alvarez R, Gomez AM, Quintana TS, Moreno Larrea MDC, Zorrilla SJ, Alarcon E, Samanez FCA, Caguioa PB, Tiangco BJ, Mora EM, Betancourt-Garcia RD, Hallman-Navarro D, Feliciano-Lopez LJ, Velez-Cortes HA, Cabanillas F, Ganea DE, Ciuleanu TE, Ghizdavescu DG, Miron L, Cebotaru CL, Cainap CI, Anghel R, Dvorkin MV, Gladkov OA, Fadeeva NV, Kuzmin AA, Lipatov ON, Zbarskaya II, Akhmetzyanov FS, Litvinov IV, Afanasyev BV, Cherenkova M, Lioznov D, Lisukov IA, Smirnova YA, Kolomietz S, Halawani H, Goh YT, Drgona L, Chudej J, Matejkova M, Reckova M, Rapoport BL, Szpak WM, Malan DR, Jonas N, Jung CW, Lee DG, Yoon SS, Lopez Jimenez J, Duran Martinez I, Rodriguez Moreno JF, Solano Vercet C, de la Camara R, Batlle Massana M, Yeh SP, Chen CY, Chou HH, Tsai CM, Chiu CH, Siritanaratkul N, Norasetthada L, Sriuranpong V, Seetalarom K, Akan H, Dane F, Ozcan MA, Ozsan GH, Kalayoglu Besisik SF, Cagatay A, Yalcin S, Peniket A, Mullan SR, Dakhil KM, Sivarajan K, Suh JJG, Sehgal A, Marquez F, Gomez EG, Mullane MR, Skinner WL, Behrens RJ, Trevarthe DR, Mazurczak MA, Lambiase EA, Vidal CA, Anac SY, Rodrigues GA, Baltz B, Boccia R, Wertheim MS, Holladay CS, Zenk D, Fusselman W, Wade III JL, Jaslowsk AJ, Keegan J, Robinson MO, Go RS, Farnen J, Amin B, Jurgens D, Risi GF, Beatty PG, Naqvi T, Parshad S, Hansen VL, Ahmed M, Steen PD, Badarinath S, Dekker A, Scouros MA, Young DE, Graydon Harker W, Kendall SD, Citron ML, Chedid S, Posada JG, Gupta MK, Rafiyath S, Buechler-Price J, Sreenivasappa S, Chay CH, Burke JM, Young SE, Mahmood A, Kugler JW, Gerstner G, Fuloria J, Belman ND, Geller R, Nieva J, Whittenberger BP, Wong BMY, Cescon TP, Abesada-Terk G, Guarino MJ, Zweibach A, Ibrahim EN, Takahashi G, Garrison MA, Mowat RB, Choi BS, Oliff IA, Singh J, Guter KA, Ayrons K, Rowland KM, Noga SJ, Rao SB, Columbie A, Nualart MT, Cecchi GR, Campos LT, Mohebtash M, Flores MR, Rothstein-Rubin R, O'Connor BM, Soori G, Knapp M, Miranda FG, Goodgame BW, Kassem M, Belani R, Sharma S, Ortiz T, Sonneborn HL, Markowitz AB, Wilbur D, Meiri E, Koo VS, Jhangiani HS, Wong L, Sanani S, Lawrence SJ, Jones CM, Murray C, Papageorgiou C, Gurtler JS, Ascensao JL, Seetalarom K, Venigalla ML, D'Andrea M, De Las Casas C, Haile DJ, Qazi FU, Santander JL, Thomas MR, Rao VP, Craig M, Garg RJ, Robles R, Lyons RM, Stegemoller RK, Goel S, Garg S, Lowry P, Lynch C, Lash B, Repka T, Baker J, Goueli BS, Campbell TC, Van Echo DA, Lee YJ, Reyes EA, Senecal FM, Donnelly G, Byeff P, Weiss R, Reid T, Roeland E, Goel A, Prow DM, Brandt DS, Kaplan HG, Payne JE, Boeckh MG, Rosen PJ, Mena RR, Khan R, Betts RF, Sharp SA, Morrison VA, Fitz-Patrick D, Congdon J, Erickson N, Abbasi R, Henderson S, Mehdi A, Wos EJ, Rehmus E, Beltzer L, Tamayo RA, Mahmood T, Reboli AC, Moore A, Brown JM, Cruz J, Quick DP, Potz JL, Kotz KW, Hutchins M, Chowhan NM, Devabhaktuni YD, Braly P, Berenguer RA, Shambaugh SC, O'Rourke TJ, Conkright WA, Winkler CF, Addo FEK, Duic JP, High KP, Kutner ME, Collins R, Carrizosa DR, Perry DJ, Kailath E, Rosen N, Sotolongo R, Shoham S, Chen T. Safety and efficacy of inactivated varicella zoster virus vaccine in immunocompromised patients with malignancies: a two-arm, randomised, double-blind, phase 3 trial. THE LANCET. INFECTIOUS DISEASES 2019; 19:1001-1012. [DOI: 10.1016/s1473-3099(19)30310-x] [Show More Authors] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 12/25/2022]
|
25
|
Haredy AM, Takei M, Iwamoto SI, Ohno M, Kosaka M, Hirota K, Koketsu R, Okuno T, Ikuta K, Yamanishi K, Ebina H. Quantification of a cell-mediated immune response against varicella zoster virus by assessing responder CD4 high memory cell proliferation in activated whole blood cultures. Vaccine 2019; 37:5225-5232. [PMID: 31358406 DOI: 10.1016/j.vaccine.2019.07.068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 11/18/2022]
Abstract
BACKGROUND Herpes zoster (HZ) is caused by reactivation of a latent varicella zoster virus (VZV). The potential to develop HZ increases with age due to waning of memory cell-mediated immunity (CMI), mainly the CD4 response. Therefore, VZV-CD4-memory T cells (CD4-M) count in blood could serve as a barometer for HZ protection. However, direct quantification of these cells is known to be difficult because they are few in number in the blood. We thus developed a method to measure the proliferation level of CD4-M cells responding to VZV antigen in whole blood culture. METHODS Blood samples were collected from 32 children (2-15 years old) with or without a history of varicella infection, 18 young adults (28-45 years old), and 80 elderly (50-86 years old) with a history of varicella infection. The elderly group was vaccinated, and blood samples were taken 2 months and 1 year after VZV vaccination. Then, 1 mL of blood was mixed with VZV, diluted 1/10 in medium, and cultured. CD4-M cells were identified and measured by flow cytometry. RESULTS There was distinct proliferation of CD3+CD4highCD45RA-RO+ (CD4high-M) cells specific to VZV antigen at day 9. The majority of CD4high-M cells had the effector memory phenotype CCR7- and was granzyme B-positive. CD4high-M cells were detected in blood culture from varicella-immune but not varicella-non-immune children. Meanwhile, a higher level of CD4high-M proliferation was observed in young adults than in the elderly. The CD4high-M proliferation level was boosted 2 months after VZV vaccination and maintained for at least 1 year in the elderly. CONCLUSION Quantifying VZV responder CD4high -M cell proliferation is a convenient way to measure VZV CMI using small blood volumes. Our method can be applied to measure VZV vaccine-induced CMI in the elderly. Clinical study registry numbers: (www.clinicaltrials.jp) 173532 and 183985.
Collapse
Affiliation(s)
- Ahmad M Haredy
- Biken Center for Innovative Vaccine Research and Development, The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan.
| | | | | | | | - Mitsuyo Kosaka
- Biken Center for Innovative Vaccine Research and Development, The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan
| | - Kazue Hirota
- Biken Center for Innovative Vaccine Research and Development, The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan
| | - Ritsuko Koketsu
- Biken Center for Innovative Vaccine Research and Development, The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan
| | - Toshiomi Okuno
- Department of Microbiology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Kazuyoshi Ikuta
- Biken Center for Innovative Vaccine Research and Development, The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan
| | - Koichi Yamanishi
- Biken Center for Innovative Vaccine Research and Development, The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan
| | - Hirotaka Ebina
- Biken Center for Innovative Vaccine Research and Development, The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Osaka, Japan
| |
Collapse
|
26
|
The Effect of Age on the Immunogenicity of the Live Attenuated Zoster Vaccine Is Predicted by Baseline Regulatory T Cells and Varicella-Zoster Virus-Specific T Cell Immunity. J Virol 2019; 93:JVI.00305-19. [PMID: 31092579 DOI: 10.1128/jvi.00305-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/07/2019] [Indexed: 12/30/2022] Open
Abstract
Older age is associated with increased infectious morbidity and decreased immune responses to vaccines, but the mechanisms that mediate this effect are incompletely understood. The efficacy and immunogenicity of the live attenuated zoster vaccine (ZVL) have a very-well-described negative association with the age of the vaccinee. In a study of 600 ZVL recipients 50 to >80 years of age, we investigated immunological factors that might explain the effect of age on the immunogenicity of ZVL. Using FluoroSpot assays and flow cytometry, we determined that varicella-zoster virus (VZV)-specific peak T helper 1 (VZV-Th1) responses to ZVL were independently predicted by prevaccination VZV-Th1 responses, regulatory T cells (Treg), and PD1-expressing immune checkpoint T cells (Tcheck) but not by the age of the vaccinee. Persistence of VZV-Th1 1 year after vaccination was independently predicted by the factors mentioned above, by peak VZV-Th1 responses to ZVL, and by the age of the vaccinee. We further demonstrated by ex vivo blocking experiments the mechanistic role of PD1 and CTLA4 as modulators of decreased VZV-Th1 responses in the study participants. VZV-specific cytotoxic T cell (VZV-CTL) and T follicular helper responses to ZVL did not correlate with age, but similar to other Th1 responses, VZV-CTL peak and baseline responses were independently correlated. These data expand our understanding of the factors affecting the magnitude and kinetics of T cell responses to ZVL in older adults and show the importance of prevaccination Treg and Tcheck in modulating the immunogenicity of ZVL. This presents new potential interventions to increase vaccine responses in older adults.IMPORTANCE Vaccination is the most effective method to protect older adults against viral infections. However, the immunogenicity of viral vaccines in older adults is notoriously poor. The live attenuated zoster vaccine (ZVL) provides the best example of a gradual decrease of vaccine immunogenicity with every 10-year age increase above 50 years. Here we show that the abundance of regulatory T cells before vaccine administration to older adults has a significant inhibitory effect on immune responses to ZVL and, together with baseline immunity to varicella-zoster virus, explains the effect of age on the immunogenicity of ZVL. Moreover, in vitro blockade of regulatory T cell mechanisms of action with biologic modulators restores immune responses to varicella-zoster virus in vaccinees. Collectively, these observations suggest that immune modulators that block regulatory T cell activity may increase responses to viral attenuated vaccines in older adults.
Collapse
|
27
|
Abstract
Much has been written about Elispot and how to optimally run the assay for a wide variety of applications. But only a limited number of articles exist addressing the analysis step, the plate evaluation. Comparing that fact with the vast amount of analysis advise available for other single cell immune assay, for example, intracellular cytokine staining, the overall impression may be that Elispot evaluation is just simple enough to not require extensive elaboration and guidance. At first thought this appears reasonable because how difficult can it be counting colored spots on a white background. In addition, automated Elispot readers were already introduced more than 20 years ago (Herr et al., J Immunol Methods 203, 141-152, 1997), easing the strenuous load of manual counting and providing means to decrease the subjectivity in Elispot analysis. Just shortly thereafter however, the first report was published about the subjectivity and operator-dependency of plate evaluation even when using automated reader systems (Janetzki et al., J Immunol Methods 291, 175-183, 2004). Later, the plate evaluation was identified as a main factor causing variability in Elispot results, triggering the inclusion of recommendations on handling of artifacts and the audits of plate reading results in the Initial Elispot Harmonization guidelines (Janetzki et al., Cancer Immunol Immunother 57, 303-315, 2008; Britten et al., Cancer Immunol Immunother 57, 289-302, 2008). In follow-up, a large international study with 75 laboratories was conducted to address the current approaches taken to evaluate Elispot plates and to establish consensus guidelines for plate evaluation (Janetzki et al., Nat Protoc 10, 1098-1115, 2015). This article addresses the special challenges of plate evaluation, gives explanations for unusual observation, and provides overall recommendations on how to work through the labyrinth of available algorithms and reader settings to obtain reliable Elispot data.
Collapse
|
28
|
Yang P, Chen Z, Zhang J, Li W, Zhu C, Qiu P, Quan Y, Cui X, Yuan L, Jiang C. Evaluation of Varicella-zoster virus-specific cell-mediated immunity by interferon-γ Enzyme-Linked Immunosorbent Assay in adults ≥50 years of age administered a herpes zoster vaccine. J Med Virol 2019; 91:829-835. [PMID: 30613990 DOI: 10.1002/jmv.25391] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/29/2018] [Indexed: 11/11/2022]
Abstract
Varicella-zoster virus (VZV)-specific cell-mediated immunity (CMI) is critical for preventing and controlling the onset of herpes zoster (HZ). To assess VZV CMI, an interferon-γ (IFN-γ) enzyme-linked immunosorbent assay (ELISA) was validated by examining the influence of VZV-specific antigen content, incubation time, and interval from whole blood collection on the assay. In phase II clinical trial, VZV-specific CMI in adults ≥50 years of age administered an HZ vaccine were evaluated by IFN-γ ELISA, as determined by measuring IFN-γ production in the whole blood in response to stimulation with ultraviolet light-inactivated VZV. The VZV-specific IFN-γ levels varied among individuals from prevaccination (baseline) to 6 weeks postvaccination. In most subjects, VZV-specific CMI was increased at 6 weeks postvaccination. The HZ vaccine elicited a significant increase in the VZV-specific CMI response as measured by ELISA; the geometric mean fold-rises from baseline to 6 weeks postvaccination were 3.50, 4.22, and 5.24 in the 4.3, 4.7, and 4.9 log plaque-forming unit vaccine groups, respectively, which was significantly higher than in the placebo group (P < 0.05). These results indicate that vaccination enhances the VZV-specific CMI responses in subjects; IFN-γ ELISA is an effective method for evaluating the CMI response and may be useful for identifying individuals at a high risk of HZ infection.
Collapse
Affiliation(s)
- Ping Yang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Zhen Chen
- National Institutes for Food and Drug Control, Beijing, China
| | | | - Wei Li
- Changchun BCHT Biotechnology Company, Changchun, China
| | - Changlin Zhu
- Changchun BCHT Biotechnology Company, Changchun, China
| | - Ping Qiu
- National Institutes for Food and Drug Control, Beijing, China
| | - Yaru Quan
- National Institutes for Food and Drug Control, Beijing, China
| | - Xiaoyu Cui
- National Institutes for Food and Drug Control, Beijing, China
| | - Liyong Yuan
- National Institutes for Food and Drug Control, Beijing, China
| | - Chunlai Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology & Engineering, Ministry of Education, Jilin University, Changchun, China
| |
Collapse
|
29
|
Levin MJ, Kroehl ME, Johnson MJ, Hammes A, Reinhold D, Lang N, Weinberg A. Th1 memory differentiates recombinant from live herpes zoster vaccines. J Clin Invest 2018; 128:4429-4440. [PMID: 30024861 PMCID: PMC6159998 DOI: 10.1172/jci121484] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/13/2018] [Indexed: 12/14/2022] Open
Abstract
The adjuvanted varicella-zoster virus (VZV) glycoprotein E (gE) subunit herpes zoster vaccine (HZ/su) confers higher protection against HZ than the live attenuated zoster vaccine (ZV). To understand the immunologic basis for the different efficacies of the vaccines, we compared immune responses to the vaccines in adults 50 to 85 years old. gE-specific T cells were very low/undetectable before vaccination when analyzed by FluoroSpot and flow cytometry. Both ZV and HZ/su increased gE-specific responses, but at peak memory response (PMR) after vaccination (30 days after ZV or after the second dose of HZ/su), gE-specific CD4+ and CD8+ T cell responses were 10-fold or more higher in HZ/su compared with ZV recipients. Comparing the vaccines, T cell memory responses, including gE-IL-2+ and VZV-IL-2+ spot-forming cells (SFCs), were higher in HZ/su recipients and cytotoxic and effector responses were lower. At 1 year after vaccination, all gE-Th1 and VZV-IL-2+ SFCs remained higher in HZ/su compared with ZV recipients. Mediation analyses showed that IL-2+ PMR were necessary for the persistence of Th1 responses to either vaccine and VZV-IL-2+ PMR explained 73% of the total effect of HZ/su on persistence. This emphasizes the biological importance of the memory responses, which were clearly superior in HZ/su compared with ZV participants.
Collapse
Affiliation(s)
- Myron J. Levin
- Department of Pediatrics, and
- Department of Medicine, University of Colorado School of Medicine
| | - Miranda E. Kroehl
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, and
| | | | - Andrew Hammes
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, and
| | - Dominik Reinhold
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, and
| | | | - Adriana Weinberg
- Department of Pediatrics, and
- Department of Medicine, University of Colorado School of Medicine
- Department of Pathology, University of Colorado School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Denver, Colorado USA
| |
Collapse
|
30
|
Winston DJ, Mullane KM, Cornely OA, Boeckh MJ, Brown JW, Pergam SA, Trociukas I, Žák P, Craig MD, Papanicolaou GA, Velez JD, Panse J, Hurtado K, Fernsler DA, Stek JE, Pang L, Su SC, Zhao Y, Chan ISF, Kaplan SS, Parrino J, Lee I, Popmihajlov Z, Annunziato PW, Arvin A. Inactivated varicella zoster vaccine in autologous haemopoietic stem-cell transplant recipients: an international, multicentre, randomised, double-blind, placebo-controlled trial. Lancet 2018; 391:2116-2127. [PMID: 29856344 DOI: 10.1016/s0140-6736(18)30631-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/28/2018] [Accepted: 03/06/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Recipients of autologous haemopoietic stem-cell transplants (auto-HSCT) have an increased risk of herpes zoster and herpes zoster-related complications. The aim of this study was to establish the efficacy and safety of an inactivated varicella zoster vaccine for the prevention of herpes zoster after auto-HSCT. METHODS In this randomised, double-blind, placebo-controlled phase 3 trial, participants were recruited from 135 medical centres (ie, stem-cell transplant centres and hospitals) in North America, South America, Europe, and Asia. Patients were eligible if they were aged 18 years or older, scheduled to receive an auto-HSCT within 60 days of enrolment, and had a history of varicella infection or were seropositive for antibodies to varicella zoster virus, or both. Exclusion criteria included a history of herpes zoster within the previous year of enrolment, and intended antiviral prophylaxis for longer than 6 months after transplantation. Participants were randomly assigned according to a central randomisation schedule generated by the trial statistician, to receive either the inactivated-virus vaccine from one of three consistency lots, a high-antigen lot, or placebo, stratified by age (<50 vs ≥50 years) and intended duration of antiviral prophylaxis after transplantation (≤3 months vs >3 to ≤6 months). Participants, investigators, trial staff, and the funder's clinical and laboratory personnel were masked to group assignment. Participants were given four doses of inactivated vaccine or placebo, with the first dose 5-60 days before auto-HSCT, and the second, third, and fourth doses at about 30, 60, and 90 days after transplantation. The primary efficacy endpoint was the incidence of herpes zoster, confirmed by PCR or adjudication by a masked clinical committee, or both, assessed in all participants randomly assigned to the vaccine consistency lot group or placebo group who received at least one dose of vaccine and had auto-HSCT. Safety was assessed in all randomised participants who received at least one dose of vaccine and had follow-up data. A prespecified vaccine efficacy success criterion required the lower bound of the 95% CI be higher than 25% for the relative reduction of the hazard ratio of herpes zoster infection in participants given the vaccine from one of the consistency lots compared with those given placebo. This trial is registered on ClinicalTrials.gov (NCT01229267) and EudraCT (2010-020150-34). FINDINGS Between Dec 7, 2010, and April 25, 2013, 560 participants were randomly assigned to the vaccine consistency lot group, 106 to the high-antigen lot group, and 564 to the placebo group. 249 (44%) of patients in the vaccine consistency lot group, 35 (33%) in the high-antigen lot group, and 220 (39%) in the placebo group discontinued before study end, mostly because of death or withdrawal. 51 participants were excluded from the primary efficacy endpoint analyses because they did not undergo auto-HSCT or were not vaccinated, or both (22 [4%] in the vaccine consistency lot group, and 29 [5%] in the placebo group). Mean follow-up for efficacy was 2·4 years (SD 1·3) in the vaccine consistency lot group and 2·3 years (SD 1·3) in the placebo group. 42 (8%) of 538 participants in the vaccine consistency lot group (32·9 per 1000 person-years) and 113 (21%) of 535 in the placebo group (91·9 per 1000 person-years) had a confirmed case of herpes zoster. The estimated vaccine efficacy was 63·8% (95% CI 48·4-74·6), meeting the pre-specified success criterion. For the combined vaccine groups versus the placebo group, the proportion of patients with serious adverse events (216 [33%] of 657 vs 181 [33%] of 554; risk difference 0·2%, 95% CI -5·1 to 5·5) and serious vaccine-related adverse events (five [1%] vs five [1%]; risk difference 0·1%, -1·4 to 1·1) were similar. Vaccine-related injection-site adverse events occurred more frequently in participants given vaccine than those given placebo (191 [29%] vs 36 [7%]; risk difference 22·6%, 95% CI 18·5-26·6; p<0·0001). INTERPRETATION This study shows for the first time in a large phase 3 trial that early vaccination of auto-HSCT recipients during the peri-transplant period can be effective for the prevention of an opportunistic infection like herpes zoster and that the vaccine is well tolerated. FUNDING Merck & Co., Inc.
Collapse
Affiliation(s)
- Drew J Winston
- Department of Medicine, University of California Los Angeles Medical Center, Los Angeles, CA, USA.
| | | | - Oliver A Cornely
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Department I of Internal Medicine, Clinical Trials Centre Cologne (ZKS Köln), German Centre for Infection Research (DZIF), University of Cologne, Cologne, Germany
| | | | | | | | | | - Pavel Žák
- Department of Medicine, Fakultní nemocnice Hradec Králové, Hradec Králové, Czech Republic
| | | | | | | | - Jens Panse
- Department of Oncology, Hematology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, University Hospital RWTH Aachen, Aachen, Germany
| | | | | | | | - Lei Pang
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | | | | | | | - Ingi Lee
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | - Ann Arvin
- Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
31
|
Welter A, Sundararaman S, Li R, Zhang T, Karulin AY, Lehmann A, Naeem V, Roen DR, Kuerten S, Lehmann PV. High-Throughput GLP-Capable Target Cell Visualization Assay for Measuring Cell-Mediated Cytotoxicity. Cells 2018; 7:cells7050035. [PMID: 29695103 PMCID: PMC5981259 DOI: 10.3390/cells7050035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/19/2018] [Accepted: 04/21/2018] [Indexed: 11/17/2022] Open
Abstract
One of the primary effector functions of immune cells is the killing of virus-infected or malignant cells in the body. Natural killer (NK) and CD8 effector T cells are specialized for this function. The gold standard for measuring such cell-mediated cytolysis has been the chromium release assay, in which the leakage of the radioactive isotope from damaged target cells is being detected. Flow cytometry-based single cell analysis of target cells has recently been established as a non-radioactive alternative. Here we introduce a target cell visualization assay (TVA) that applies similar target cell staining approaches as used in flow cytometry but based on single cell computer image analysis. Two versions of TVA are described here. In one, the decrease in numbers of calcein-stained, i.e., viable, target cells is assessed. In the other, the CFSE/PI TVA, the increase in numbers of dead target cells is established in addition. TVA assays are shown to operate with the same sensitivity as standard chromium release assays, and, leaving data audit trails in form of scanned (raw), analyzed, and quality-controlled images, thus meeting requirements for measuring cell-mediated cytolysis in a regulated environment.
Collapse
Affiliation(s)
- Anna Welter
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Srividya Sundararaman
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Ruliang Li
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Ting Zhang
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Alexey Y Karulin
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Alexander Lehmann
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Villian Naeem
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Diana R Roen
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Paul V Lehmann
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA.
| |
Collapse
|
32
|
Pinna RA, Dos Santos AC, Perce-da-Silva DS, da Silva LA, da Silva RNR, Alves MR, Santos F, de Oliveira Ferreira J, Lima-Junior JC, Villa-Verde DM, De Luca PM, Carvalho-Pinto CE, Banic DM. Correlation of APRIL with production of inflammatory cytokines during acute malaria in the Brazilian Amazon. IMMUNITY INFLAMMATION AND DISEASE 2018; 6:207-220. [PMID: 29314720 PMCID: PMC5946147 DOI: 10.1002/iid3.208] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 02/06/2023]
Abstract
INTRODUCTION A proliferation-inducing ligand (APRIL) and B cell activation factor (BAFF) are known to play a significant role in the pathogenesis of several diseases, including BAFF in malaria. The aim of this study was to investigate whether APRIL and BAFF plasma concentrations could be part of inflammatory responses associated with P. vivax and P. falciparum malaria in patients from the Brazilian Amazon. METHODS Blood samples were obtained from P. vivax and P. falciparum malaria patients (n = 52) resident in Porto Velho before and 15 days after the beginning of treatment and from uninfected individuals (n = 12). We investigated APRIL and BAFF circulating levels and their association with parasitaemia, WBC counts, and cytokine/chemokine plasma levels. The expression levels of transmembrane activator and calcium-modulating cyclophilin ligand interactor (TACI) on PBMC from a subset of 5 P. vivax-infected patients were analyzed by flow cytometry. RESULTS APRIL plasma levels were transiently increased during acute P. vivax and P. falciparum infections whereas BAFF levels were only increased during acute P. falciparum malaria. Although P. vivax and P. falciparum malaria patients have similar cytokine profiles during infection, in P. vivax acute phase malaria, APRIL but not BAFF levels correlated positively with IL-1, IL-2, IL-4, IL-6, and IL-13 levels. We did not find any association between P. vivax parasitaemia and APRIL levels, while an inverse correlation was found between P. falciparum parasitaemia and APRIL levels. The percentage of TACI positive CD4+ and CD8+ T cells were increased in the acute phase P. vivax malaria. CONCLUSION These findings suggest that the APRIL and BAFF inductions reflect different host strategies for controlling infection with each malaria species.
Collapse
Affiliation(s)
- Raquel A Pinna
- Laboratory of Clinical Immunology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ, Brazil, 21040-360
| | - Adriana C Dos Santos
- Laboratory of Experimental Pathology, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil, 24020-140
| | - Daiana S Perce-da-Silva
- Laboratory of Clinical Immunology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ, Brazil, 21040-360
| | - Luciene A da Silva
- Laboratory of Clinical Immunology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ, Brazil, 21040-360
| | - Rodrigo N Rodrigues da Silva
- Laboratory of Imunoparasitology Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ, Brazil, 21040-360
| | - Marcelo R Alves
- Laboratory of Research in Pharmacogenetics, National Institute of Infectology, Oswaldo Cruz Foundation, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ, Brazil, 21040-360
| | - Fátima Santos
- Laboratory of Entomology, LACEN/RO, Rua Anita Garibalde, 4130 - Costa e Silva, Porto Velho, RO, Brazil, 76803-620
| | - Joseli de Oliveira Ferreira
- Laboratory of Imunoparasitology Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ, Brazil, 21040-360
| | - Josué C Lima-Junior
- Laboratory of Imunoparasitology Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ, Brazil, 21040-360
| | - Déa M Villa-Verde
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ, Brazil, 21040-360
| | - Paula M De Luca
- Laboratory of Imunoparasitology Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ, Brazil, 21040-360
| | - Carla E Carvalho-Pinto
- Laboratory of Experimental Pathology, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil, 24020-140
| | - Dalma M Banic
- Laboratory of Clinical Immunology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ, Brazil, 21040-360
| |
Collapse
|
33
|
A Positive Control for Detection of Functional CD4 T Cells in PBMC: The CPI Pool. Cells 2017; 6:cells6040047. [PMID: 29215584 PMCID: PMC5753071 DOI: 10.3390/cells6040047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 12/29/2022] Open
Abstract
Testing of peripheral blood mononuclear cells (PBMC) for immune monitoring purposes requires verification of their functionality. This is of particular concern when the PBMC have been shipped or stored for prolonged periods of time. While the CEF (Cytomegalo-, Epstein-Barr and Flu-virus) peptide pool has become the gold standard for testing CD8 cell functionality, a positive control for CD4 cells is so far lacking. The latter ideally consists of proteins so as to control for the functionality of the antigen processing and presentation compartments, as well. Aiming to generate a positive control for CD4 cells, we first selected 12 protein antigens from infectious/environmental organisms that are ubiquitous: Varicella, Influenza, Parainfluenza, Mumps, Cytomegalovirus, Streptococcus, Mycoplasma, Lactobacillus, Neisseria, Candida, Rubella, and Measles. Of these antigens, three were found to elicited interferon (IFN)-γ-producing CD4 cells in the majority of human test subjects: inactivated cytomegalo-, parainfluenza-, and influenza virions (CPI). While individually none of these three antigens triggered a recall response in all donors, the pool of the three (the ‘CPI pool’), did. One hundred percent of 245 human donors tested were found to be CPI positive, including Caucasians, Asians, and African-Americans. Therefore, the CPI pool appears to be suitable to serve as universal positive control for verifying the functionality of CD4 and of antigen presenting cells.
Collapse
|
34
|
Zia A, Singh D, Saxena S, Umrao J, Baluni M, Ghildiyal S, Fatima T, Shukla M, Agarwal V, Dhole TN. Detection of long term cellular immune response to Japanese encephalitis vaccination using IFN-γ ELIspot assay. J Med Virol 2017; 89:2235-2238. [PMID: 28671301 DOI: 10.1002/jmv.24893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/15/2017] [Indexed: 11/09/2022]
Abstract
Vaccine is the most effective preventive measure against Japanese Encephalitis infection. Role of IFN-γ expressing T cells for JE virus clearance has been described as a part of cellular immunity. Vaccine induced immunity also involve the cellular immune response, therefore the study was aimed to observe induction and persistence of IFN-γ expressing T cells by IFN-γ ELISpot assay. The cell count increased significantly after 28 (P < 0.0001) days post vaccination, and remained higher at all time points (day 28, day 180, day 360) when compared with prevaccination. This study will be helpful for designing future vaccination strategy and improving vaccine efficacy.
Collapse
Affiliation(s)
- Amreen Zia
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Dharamveer Singh
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Swati Saxena
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Jyoti Umrao
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Manjari Baluni
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Sneha Ghildiyal
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Tanzeem Fatima
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Mukti Shukla
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Vikas Agarwal
- Department of Clinical Immunology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Tapan N Dhole
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| |
Collapse
|
35
|
Shaw J, Halsey NA, Weinberg A, Scott Schmid D, George KS, Weldon WC, Jordan M, Bryant PW, LaRussa PS, Bradshaw DY, Harrington T, Gershon A. Arm Paralysis After Routine Childhood Vaccinations: Application of Advanced Molecular Methods to the Causality Assessment of an Adverse Event After Immunization. J Pediatric Infect Dis Soc 2017; 6:e161-e164. [PMID: 28339574 PMCID: PMC6251534 DOI: 10.1093/jpids/piw084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 12/02/2016] [Indexed: 12/27/2022]
Abstract
Post-licensure surveillance for adverse events following immunizations (AEFI) can identify rare complications of vaccinations and rigorous vaccine adverse event causality assessments can help to identify possible causal relationships. We report the development of arm paralysis after varicella vaccination in a 1-year-old child. Paralysis was initially presumed to be due to vOka because of the temporal relationship between vaccination and onset of arm weakness; however, molecular studies identified wild-type varicella zoster virus VZV (WT-VZV) in the CSF, leading the authors to conclude that WT-VZV was the probable cause. This case illustrates the complexity of assessing AEFI causality, and the importance of careful and complete evaluations when determining the most likely cause of an AEFI.
Collapse
Affiliation(s)
- Jana Shaw
- Department of Pediatrics, Division of Infectious Diseases, State University of New York Upstate Medical University, Golisano Children’s Hospital, Syracuse;,Correspondence: J. Shaw, MD, MPH, Associate Professor of Pediatrics, Department of Pediatrics, Division of Infectious Diseases, State University of New York Upstate Medical University, Golisano Children’s Hospital, 750 East Adams Street, Syracuse, NY 13210 ()
| | - Neal A Halsey
- Institute for Vaccine Safety, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Adriana Weinberg
- Departments of Pediatrics, Medicine and Pathology, Director, Molecular and Virology Clinical Laboratories, University of Colorado Denver, Aurora
| | - D Scott Schmid
- National Center for Immunizations and Respiratory Diseases, Division of Viral Diseases, Atlanta, Georgia
| | - Kirsten St George
- Laboratory of Viral Diseases, Wadsworth Center, New York State Department of Health, Clinical Professor, Department of Biomedical Sciences, University at Albany, SUNY
| | - William C Weldon
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Michael Jordan
- Pediatrics, Divisions of Immunobiology, and Bone Marrow Transplantation and Immune Deficiency Cincinnati Children’s Hospital/University of Cincinnati, Ohio
| | - Patrick W Bryant
- Laboratory of Viral Diseases, Wadsworth Center, New York State Department of Health, Albany
| | - Philip S LaRussa
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Columbia University, New York, New York
| | | | - Theresa Harrington
- Centers for Disease Control and Prevention, National Center for Emerging, Zoonotic, and Infectious Diseases, Division of Healthcare Quality Promotion, Immunization Safety Office, Clinical Immunization Safety Assessment Project, Atlanta, Georgia
| | - Anne Gershon
- Columbia University College of Physicians and Surgeons, New York, New York
| |
Collapse
|
36
|
Parrino J, McNeil SA, Lawrence SJ, Kimby E, Pagnoni MF, Stek JE, Zhao Y, Chan IS, Kaplan SS. Safety and immunogenicity of inactivated varicella-zoster virus vaccine in adults with hematologic malignancies receiving treatment with anti-CD20 monoclonal antibodies. Vaccine 2017; 35:1764-1769. [DOI: 10.1016/j.vaccine.2016.10.055] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/18/2016] [Accepted: 10/19/2016] [Indexed: 11/25/2022]
|
37
|
Herpes zoster: Risk and prevention during immunomodulating therapy. Joint Bone Spine 2017; 84:21-27. [DOI: 10.1016/j.jbspin.2016.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2016] [Indexed: 12/30/2022]
|
38
|
Lelic A, Verschoor CP, Lau VWC, Parsons R, Evelegh C, Bowdish DM, Bramson JL, Loeb MB. Immunogenicity of Varicella Vaccine and Immunologic Predictors of Response in a Cohort of Elderly Nursing Home Residents. J Infect Dis 2016; 214:1905-1910. [PMID: 27707807 DOI: 10.1093/infdis/jiw462] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 09/23/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Little is known about the immunogenicity of live-attenuated Oka/Merck varicella zoster virus (VZV)-containing vaccine (hereafter, "varicella vaccine") in frail nursing homes residents nor about immune phenotypes associated with a response. METHODS A cohort of 190 frail nursing home residents aged 80-102 years and a cohort of 50 community-dwelling seniors aged 60-75 years (a comparison group) received varicella vaccine. Interferon γ (IFN-γ) enzyme-linked immunospot assays were performed before and 6 weeks after vaccination. Cellular markers of immunosenescence were measured in the nursing home elderly. RESULTS The average number of IFN-γ spot-forming cells at baseline was significantly lower in the elderly nursing home residents than in the community-dwelling seniors. However, following vaccination, the VZV immune response increased in both cohorts, and no difference was noted in the fold difference of the response between the 2 cohorts. Upon further examination of the elderly nursing home residents, we found that higher frequencies of regulatory T cells and cytomegalovirus-specific CD4+ T cells correlated negatively with the magnitude of VZV-specific responses. CONCLUSIONS The Oka/Merck varicella vaccine induces VZV immunity in elderly nursing home residents that is similar to that produced in community-dwelling seniors. CLINICAL TRIALS REGISTRATION NCT01328548.
Collapse
Affiliation(s)
- Alina Lelic
- Department of Pathology and Molecular Medicine
| | - Chris P Verschoor
- Department of Pathology and Molecular Medicine.,Canadian Longitudinal Study on Aging
| | | | | | | | - Dawn M Bowdish
- Department of Pathology and Molecular Medicine.,Institute of Infectious Diseases Research
| | | | - Mark B Loeb
- Department of Pathology and Molecular Medicine.,Institute of Infectious Diseases Research.,Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Canada
| |
Collapse
|
39
|
De Paschale M, Clerici P. Microbiology laboratory and the management of mother-child varicella-zoster virus infection. World J Virol 2016; 5:97-124. [PMID: 27563537 PMCID: PMC4981827 DOI: 10.5501/wjv.v5.i3.97] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/08/2016] [Accepted: 07/22/2016] [Indexed: 02/05/2023] Open
Abstract
Varicella-zoster virus, which is responsible for varicella (chickenpox) and herpes zoster (shingles), is ubiquitous and causes an acute infection among children, especially those aged less than six years. As 90% of adults have had varicella in childhood, it is unusual to encounter an infected pregnant woman but, if the disease does appear, it can lead to complications for both the mother and fetus or newborn. The major maternal complications include pneumonia, which can lead to death if not treated. If the virus passes to the fetus, congenital varicella syndrome, neonatal varicella (particularly serious if maternal rash appears in the days immediately before or after childbirth) or herpes zoster in the early years of life may occur depending on the time of infection. A Microbiology laboratory can help in the diagnosis and management of mother-child infection at four main times: (1) when a pregnant woman has been exposed to varicella or herpes zoster, a prompt search for specific antibodies can determine whether she is susceptible to, or protected against infection; (2) when a pregnant woman develops clinical symptoms consistent with varicella, the diagnosis is usually clinical, but a laboratory can be crucial if the symptoms are doubtful or otherwise unclear (atypical patterns in immunocompromised subjects, patients with post-vaccination varicella, or subjects who have received immunoglobulins), or if there is a need for a differential diagnosis between varicella and other types of dermatoses with vesicle formation; (3) when a prenatal diagnosis of uterine infection is required in order to detect cases of congenital varicella syndrome after the onset of varicella in the mother; and (4) when the baby is born and it is necessary to confirm a diagnosis of varicella (and its complications), make a differential diagnosis between varicella and other diseases with similar symptoms, or confirm a causal relationship between maternal varicella and malformations in a newborn.
Collapse
|
40
|
Kantor AB, Moore WA, Meehan S, Parks DR. A Quantitative Method for Comparing the Brightness of Antibody-dye Reagents and Estimating Antibodies Bound per Cell. ACTA ACUST UNITED AC 2016; 77:1.30.1-1.30.23. [DOI: 10.1002/cpcy.6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Aaron B. Kantor
- Department of Genetics, Stanford University; Stanford California
| | - Wayne A. Moore
- Department of Genetics, Stanford University; Stanford California
| | - Stephen Meehan
- Department of Genetics, Stanford University; Stanford California
| | - David R. Parks
- Department of Genetics, Stanford University; Stanford California
| |
Collapse
|
41
|
Beals CR, Railkar RA, Schaeffer AK, Levin Y, Kochba E, Meyer BK, Evans RK, Sheldon EA, Lasseter K, Lang N, Weinberg A, Canniff J, Levin MJ. Immune response and reactogenicity of intradermal administration versus subcutaneous administration of varicella-zoster virus vaccine: an exploratory, randomised, partly blinded trial. THE LANCET. INFECTIOUS DISEASES 2016; 16:915-22. [PMID: 27061887 DOI: 10.1016/s1473-3099(16)00133-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 02/15/2016] [Accepted: 02/26/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND The licensed live, attenuated varicella-zoster virus vaccine prevents herpes zoster in adults older than 50 years. We aimed to determine whether intradermal administration of zoster vaccine could enhance vaccine immunogenicity compared with conventional needle subcutaneous administration. METHODS In this randomised, dose-ranging study, adults aged 50 years or older who had a history of varicella or who had resided in a country with endemic varicella-zoster virus infection for 30 years or more were eligible. Participants received the approved full or a 1/3 dose of zoster vaccine given subcutaneously or one of four intradermal doses (full, 1/3, 1/10, or 1/27 dose) using the MicronJet600 device. The two subcutaneous doses and the four intradermal doses were randomised (1·5:1:1:1:1:1) by computer generated sequence with randomisation stratified by age (50-59 years or 60 years or older). The primary immunogenicity endpoint was the change from baseline in IgG antibody to varicella-zoster virus-specific glycoproteins (gpELISA) measured at 6 weeks. All patients were included in the primary and safety analyses. This study is registered with ClinicalTrials.gov, number NCT01385566. FINDINGS Between Sept 2, 2011, and Jan 13, 2012, 224 participants were enrolled from three clinics in the USA and 223 were randomly assigned: 52 to receive the full dose subcutaneous zoster vaccine, 34 to receive the 1/3 dose subcutaneous zoster vaccine, 34 to receive the full dose intradermal zoster vaccine, 35 to receive the 1/3 dose intradermal zoster vaccine, 34 to receive the 1/10 dose intradermal zoster vaccine, and 34 to receive the 1/27 dose intradermal zoster vaccine. Full dose zoster vaccine given subcutaneously resulted in a gpELISA geometric mean fold-rise (GMFR) of 1·74 (90% CI 1·48-2·04) at 6 weeks post-vaccination compared with intradermal administration which resulted in a significantly higher gpELISA GMFR of 3·25 (2·68-3·94; p<0·0001), which also remained high at 18 months. An apparent dose-response relation was observed with intradermal administration (1/3 dose subcutaneous GMFR 1·64 [90% CI 1·36-1·99], 1/3 dose intradermal 2·58 (2·13-3·13), 1/10 dose intradermal 2·22 [1·83-2·69], and 1/27 dose intradermal 1·64 [1·35-2·00]). Each partial dose of zoster vaccine given intradermaly had a gpELISA GMFR comparable to that of full dose zoster vaccine given subcutaneously. Transient erythema and induration were more common after intradermal administration (31% erythema for full subcutaneous dose and 77% for intradermal dose). INTERPRETATION Intradermal zoster vaccine showed a greater increase in varicella-zoster virus gpELISA antibody compared with subcutaneous zoster vaccine at comparable doses. Larger and longer studies of intradermal administration of live, attenuated zoster vaccine are needed to provide convincing evidence of improved cell mediated immunity. FUNDING Merck & Co Inc.
Collapse
Affiliation(s)
| | | | | | - Yotam Levin
- NanoPass Technologies Ltd, Nes Ziona, Israel
| | | | | | | | | | | | - Nancy Lang
- Pediatric Infectious Diseases, University of Colorado, Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Adriana Weinberg
- Pediatric Infectious Diseases, University of Colorado, Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer Canniff
- Pediatric Infectious Diseases, University of Colorado, Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Myron J Levin
- Pediatric Infectious Diseases, University of Colorado, Denver Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
42
|
Abstract
Membranes are widely used as protein blotting matrices for a large variety of research applications including western blotting and enzyme-linked immunospot assay (ELISPOT). The largest advantage of using membranes versus solid plastic support is the porosity of membranes allowing for immobilization of high concentrations of proteins and antibodies which, in turn, increases the sensitivity of detection. Similar to plastic surfaces, polyvinylidene difluoride (PVDF) and nitrocellulose membranes create good microenvironment for live cells cultured in vitro and do not interfere with cellular physiology. It appears that PVDF-backed microplates are a golden standard for ELISPOT assays: such plates are inexpensive, easy to use and after assay development, membranes can be removed from the plates and archived. Given the convenience and reliability of membrane microplates, they are widely used in ELISPOT assays for basic research and clinical trials. The ELISPOT assay is an antibody "sandwich" technique aimed at trapping cell-secreted molecules between capture and detection antibodies, followed by either chromogenic enzymatic or fluorescence detection. This review covers the principles of the ELISPOT assay on membrane microplates including single-color and two-color detection techniques with the emphasis on assay design, choosing membrane microplates, and troubleshooting protocols.
Collapse
|
43
|
Kim JW, Min CK, Mun YC, Park Y, Kim BS, Nam SH, Koh Y, Kwon JH, Choe PG, Park WB, Kim I. Varicella-zoster virus-specific cell-mediated immunity and herpes zoster development in multiple myeloma patients receiving bortezomib- or thalidomide-based chemotherapy. J Clin Virol 2015; 73:64-69. [DOI: 10.1016/j.jcv.2015.10.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 09/25/2015] [Accepted: 10/21/2015] [Indexed: 10/22/2022]
|
44
|
Hayashida K, Ozaki T, Nishimura N, Gotoh K, Funahashi K, Nakane K, Gomi Y, Manabe S, Ishikawa T, Yamanishi K. Evaluation of varicella zoster virus-specific cell-mediated immunity by using an interferon-γ enzyme-linked immunosorbent assay. J Immunol Methods 2015; 426:50-5. [DOI: 10.1016/j.jim.2015.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 07/27/2015] [Accepted: 07/27/2015] [Indexed: 11/30/2022]
|
45
|
Rekers NV, von Herrath MG, Wesley JD. Immunotherapies and immune biomarkers in Type 1 diabetes: A partnership for success. Clin Immunol 2015; 161:37-43. [PMID: 26122172 DOI: 10.1016/j.clim.2015.05.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/13/2015] [Accepted: 05/17/2015] [Indexed: 12/16/2022]
Abstract
The standard of care (SoC) for Type 1 diabetes (T1D) today is much the same as it was in the early 1920s, simply with more insulin options-fast-acting, slow-acting, injectable, and inhalable insulins. However, these well-tolerated treatments only manage the symptoms and complications, but do nothing to halt the underlying immune response. There is an unmet need for better treatment options for T1D that address all aspects of the disease. For decades, we have successfully treated T1D in preclinical animal models with immune-modifying therapies that have not demonstrated comparable efficacy in humans. The path to bringing such options to the clinic will depend on the implementation and standard inclusion of biomarkers of immune and therapeutic efficacy in T1D clinical trials, and dictate if we can create a new SoC that treats the underlying autoimmunity as well as the symptoms it causes.
Collapse
Affiliation(s)
- Niels V Rekers
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, WA, USA; Pacific Northwest Diabetes Research Institute, Seattle, WA, USA
| | | | - Johnna D Wesley
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, WA, USA.
| |
Collapse
|
46
|
Measuring Cellular Immunity to Influenza: Methods of Detection, Applications and Challenges. Vaccines (Basel) 2015; 3:293-319. [PMID: 26343189 PMCID: PMC4494351 DOI: 10.3390/vaccines3020293] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 12/11/2022] Open
Abstract
Influenza A virus is a respiratory pathogen which causes both seasonal epidemics and occasional pandemics; infection continues to be a significant cause of mortality worldwide. Current influenza vaccines principally stimulate humoral immune responses that are largely directed towards the variant surface antigens of influenza. Vaccination can result in an effective, albeit strain-specific antibody response and there is a need for vaccines that can provide superior, long-lasting immunity to influenza. Vaccination approaches targeting conserved viral antigens have the potential to provide broadly cross-reactive, heterosubtypic immunity to diverse influenza viruses. However, the field lacks consensus on the correlates of protection for cellular immunity in reducing severe influenza infection, transmission or disease outcome. Furthermore, unlike serological methods such as the standardized haemagglutination inhibition assay, there remains a large degree of variation in both the types of assays and method of reporting cellular outputs. T-cell directed immunity has long been known to play a role in ameliorating the severity and/or duration of influenza infection, but the precise phenotype, magnitude and longevity of the requisite protective response is unclear. In order to progress the development of universal influenza vaccines, it is critical to standardize assays across sites to facilitate direct comparisons between clinical trials.
Collapse
|
47
|
Crivello P, Zito L, Sizzano F, Zino E, Maiers M, Mulder A, Toffalori C, Naldini L, Ciceri F, Vago L, Fleischhauer K. The Impact of Amino Acid Variability on Alloreactivity Defines a Functional Distance Predictive of Permissive HLA-DPB1 Mismatches in Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2015; 21:233-41. [DOI: 10.1016/j.bbmt.2014.10.017] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/19/2014] [Indexed: 01/11/2023]
|
48
|
Abstract
The ELISpot, a heterogeneous immunoassay, is widely used for detection of low abundant analytes. It is a reliable and robust assay to monitor responses of the immune system at the single-cell level by capturing secreted molecules of interest with specific, membrane-bound antibodies. Those molecules are then made visible by a cascade of ELISA-related development steps. The final results are distinct spots on the membrane as an imprint of the cell secreting the captured molecules, not only allowing their quantification but also providing insight on the kinetics and strength of secretion. This chapter describes the optimized protocol steps of the ELISpot technique, important improvements and tools available for the community, and the current expansion of the technique into polyfunctional cell analysis.
Collapse
Affiliation(s)
- Sylvia Janetzki
- ZellNet Consulting, Inc., 555 North Avenue, Suite 25-S, Fort Lee, NJ, 07024, USA.
| | - Rachel Rabin
- ZellNet Consulting, Inc., 555 North Avenue, Suite 25-S, Fort Lee, NJ, 07024, USA
| |
Collapse
|
49
|
Improvement of IFNg ELISPOT Performance Following Overnight Resting of Frozen PBMC Samples Confirmed Through Rigorous Statistical Analysis. Cells 2014; 4:1-18. [PMID: 25546016 PMCID: PMC4381205 DOI: 10.3390/cells4010001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/16/2014] [Indexed: 01/18/2023] Open
Abstract
Immune monitoring of functional responses is a fundamental parameter to establish correlates of protection in clinical trials evaluating vaccines and therapies to boost antigen-specific responses. The IFNg ELISPOT assay is a well-standardized and validated method for the determination of functional IFNg-producing T-cells in peripheral blood mononuclear cells (PBMC); however, its performance greatly depends on the quality and integrity of the cryopreserved PBMC. Here, we investigate the effect of overnight (ON) resting of the PBMC on the detection of CD8-restricted peptide-specific responses by IFNg ELISPOT. The study used PBMC from healthy donors to evaluate the CD8 T-cell response to five pooled or individual HLA-A2 viral peptides. The results were analyzed using a modification of the existing distribution free resampling (DFR) recommended for the analysis of ELISPOT data to ensure the most rigorous possible standard of significance. The results of the study demonstrate that ON resting of PBMC samples prior to IFNg ELISPOT increases both the magnitude and the statistical significance of the responses. In addition, a comparison of the results with a 13-day preculture of PBMC with the peptides before testing demonstrates that ON resting is sufficient for the efficient evaluation of immune functioning.
Collapse
|
50
|
Terada K, Itoh Y, Fujii A, Kitagawa S, Ogita S, Ouchi K. Varicella-zoster virus-specific, cell-mediated immunity with interferon-gamma release assay after vaccination of college students with no or intermediate IgG antibody response. J Med Virol 2014; 87:350-6. [DOI: 10.1002/jmv.24031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2014] [Indexed: 02/03/2023]
|