1
|
Waerlop G, Leroux-Roels G, Pagnon A, Begue S, Salaun B, Janssens M, Medaglini D, Pettini E, Montomoli E, Gianchecchi E, Lambe T, Godfrey L, Bull M, Bellamy D, Amdam H, Bredholt G, Cox RJ, Clement F. Proficiency tests to evaluate the impact on assay outcomes of harmonized influenza-specific Intracellular Cytokine Staining (ICS) and IFN-ɣ Enzyme-Linked ImmunoSpot (ELISpot) protocols. J Immunol Methods 2023; 523:113584. [PMID: 37918618 DOI: 10.1016/j.jim.2023.113584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 09/30/2023] [Accepted: 10/28/2023] [Indexed: 11/04/2023]
Abstract
The magnitude and quality of cell-mediated immune responses elicited by natural infection or vaccination are commonly measured by Interferon-ɣ (IFN-ɣ) Enzyme-Linked ImmunoSpot (ELISpot) and Intracellular Cytokine Staining (ICS). To date, laboratories apply a variety of in-house procedures which leads to diverging results, complicates interlaboratory comparisons and hampers vaccine evaluations. During the FLUCOP project, efforts have been made to develop harmonized Standard Operating Procedures (SOPs) for influenza-specific IFN-ɣ ELISpot and ICS assays. Exploratory pilot studies provided information about the interlaboratory variation before harmonization efforts were initiated. Here we report the results of two proficiency tests organized to evaluate the impact of the harmonization effort on assay results and the performance of participating FLUCOP partners. The introduction of the IFN-ɣ ELISpot SOP reduced variation of both background and stimulated responses. Post-harmonization background responses were all lower than an arbitrary threshold of 50 SFU/million cells. When stimulated with A/California and B/Phuket, a statistically significant reduction in variation (p < 0.0001) was observed and CV values were strongly reduced, from 148% to 77% for A/California and from 126% to 73% for B/Phuket. The harmonizing effect of applying an ICS SOP was also confirmed by an increased homogeneity of data obtained by the individual labs. The application of acceptance criteria on cell viability and background responses further enhanced the data homogeneity. Finally, as the same set of samples was analyzed by both the IFN-ɣ ELISpot and the ICS assays, a method comparison was performed. A clear correlation between the two methods was observed, but they cannot be considered interchangeable. In conclusion, proficiency tests show that a limited harmonization effort consisting of the introduction of SOPs and the use of the same in vitro stimulating antigens leads to a reduction of the interlaboratory variation of IFN-ɣ ELISpot data and demonstrate that substantial improvements for the ICS assay are achieved as comparable laboratory datasets could be generated. Additional steps to further reduce the interlaboratory variation of ICS data can consist of standardized gating templates and detailed data reporting instructions as well as further efforts to harmonize reagent and instrument use.
Collapse
Affiliation(s)
- Gwenn Waerlop
- Center for Vaccinology (CEVAC), Ghent University and University Hospital, Ghent, Belgium.
| | - Geert Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University and University Hospital, Ghent, Belgium
| | - Anke Pagnon
- Sanofi, Research Global Immunology, Marcy l'Etoile, France
| | - Sarah Begue
- Sanofi, Research Global Immunology, Marcy l'Etoile, France
| | | | | | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elena Pettini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy; VisMederi srl, 53100 Siena, Italy
| | | | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, United Kingdom
| | - Leila Godfrey
- Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford, UK
| | - Maireid Bull
- Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, United Kingdom
| | - Duncan Bellamy
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Håkon Amdam
- Influenza Centre, Department of Clinical Science, University of Bergen, N5021 Bergen, Norway
| | - Geir Bredholt
- Influenza Centre, Department of Clinical Science, University of Bergen, N5021 Bergen, Norway
| | - Rebecca Jane Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, N5021 Bergen, Norway
| | - Frédéric Clement
- Center for Vaccinology (CEVAC), Ghent University and University Hospital, Ghent, Belgium
| |
Collapse
|
2
|
Li X, He S, Thomas J, Wu B, Yang TY, Swanson M. Optimization of Peripheral Blood Mononuclear Cell Processing for Improved Clinical ELISpot Assay Performance. AAPS J 2023; 25:93. [PMID: 37770755 DOI: 10.1208/s12248-023-00861-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/07/2023] [Indexed: 09/30/2023] Open
Abstract
Cell and gene therapies have demonstrated impressive therapeutic efficacy in various human diseases. Nevertheless, cellular immune response directed against these therapeutic agents is an obstacle for achieving long-lasting clinical efficacy. Therefore, it is crucial to develop robust assays to accurately monitor cellular immunogenicity towards these therapies. Enzyme-linked immunospot (ELISpot) assay is one of the primarily used methods for measuring cellular immune response in clinical programs, which requires isolation of the peripheral blood mononuclear cells (PBMCs). The quality of this clinical material is one of the most critical factors that impact the robust assessment of cellular immune responses. The optimal blood sample processing conditions, however, remain poorly understood. In this study, we examined the impact of blood sample processing time on the performance characteristics of ELISpot to measure antigen-specific cellular responses. Blood samples that were processed after overnight delay resulted in a loss of ELISpot signals. We subsequently optimized several parameters of sample processing, and successfully recovered ELISpot signals for the blood samples that are processed within 32 h. Furthermore, several mitigation strategies were employed that would potentially address the impact of granulocyte contamination on detection of antigen-specific cellular responses. Our investigation provides an extension of sample processing window for clinical studies and is significant for resolving the logistical challenge of whole blood sample shipment for timely PBMC preparation in cell/gene therapy clinical studies.
Collapse
Affiliation(s)
- Xinyuan Li
- Janssen Research & Development LLC., 1400 McKean Road, Spring House, Pennsylvania, 19477, USA.
| | - Shan He
- Janssen Research & Development LLC., 1400 McKean Road, Spring House, Pennsylvania, 19477, USA
| | - Jaya Thomas
- Janssen Research & Development LLC., 1400 McKean Road, Spring House, Pennsylvania, 19477, USA
| | - Bonnie Wu
- Janssen Research & Development LLC., 1400 McKean Road, Spring House, Pennsylvania, 19477, USA
| | - Tong-Yuan Yang
- Janssen Research & Development LLC., 1400 McKean Road, Spring House, Pennsylvania, 19477, USA
| | - Michael Swanson
- Janssen Research & Development LLC., 1400 McKean Road, Spring House, Pennsylvania, 19477, USA
| |
Collapse
|
3
|
Berliner KE, Ezzelle T, Klenk T, Dunn G, Sischo J, Campbell D, McKee KT. Rapid Establishment of a Biospecimen Resource To Study the Global Impact of COVID-19 Vaccines. Microbiol Spectr 2023; 11:e0211723. [PMID: 37367491 PMCID: PMC10434269 DOI: 10.1128/spectrum.02117-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 06/28/2023] Open
Abstract
The emergence and explosive spread of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 highlighted the need to rapidly develop curated biobanks to inform the etiology, diagnosis, and treatment options for global outbreaks of communicable diseases. Recently, we undertook efforts to develop a repository of biospecimens from individuals aged 12 and older who were to be vaccinated against coronavirus disease 19 (COVID-19) with vaccines developed with support from the United States Government. We planned to establish 40 or more clinical study sites in at least six countries to collect biospecimens from 1,000 individuals, 75% of whom were to be SARS-CoV-2 naive at the time of enrollment. Specimens would be used to (i) ensure quality control of future diagnostic tests, (ii) understand immune responses to multiple COVID-19 vaccines, and (iii) provide reference reagents for the development of new drugs, biologics, and vaccines. Biospecimens included serum, plasma, whole blood, and nasal secretions. Large-volume collections of peripheral blood mononuclear cells (PBMCs) and defibrinated plasma were also planned for a subset of subjects. Participant sampling was planned at intervals prior to and following vaccination over a 1-year period. Here, we describe the selection of clinical sites for specimen collection and processing, standard operating procedure (SOP) development, design of a training program for tracking specimen quality, and specimen transport to a repository for interim storage. This approach allowed us to enroll our first participants within 21 weeks from the study's initiation. Lessons learned from this experience should benefit the development of biobanks in response to future global epidemics. IMPORTANCE The ability to rapidly create a biobank of high-quality specimens in response to emergent infectious diseases is critical to allow for the development of prevention and treatment, as well as to effectively monitor the spread of the disease. In this paper, we report on a novel approach to getting global clinical sites up and running within a short time frame and to monitor the quality of specimens collected to ensure their value in future research efforts. Our results have important implications for the monitoring of the quality of biospecimens collected and to design effective interventions to address shortcomings, where needed.
Collapse
Affiliation(s)
| | | | - T. Klenk
- Allucent, Cary, North Carolina, USA
| | - G. Dunn
- Allucent, Cary, North Carolina, USA
| | | | | | | |
Collapse
|
4
|
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
|
5
|
Waerlop G, Leroux-Roels G, Lambe T, Bellamy D, Medaglini D, Pettini E, Cox RJ, Trieu MC, Davies R, Bredholt G, Montomoli E, Gianchecchi E, Clement F. Harmonization and qualification of an IFN-γ Enzyme-Linked ImmunoSpot assay (ELISPOT) to measure influenza-specific cell-mediated immunity within the FLUCOP consortium. Front Immunol 2022; 13:984642. [PMID: 36159843 PMCID: PMC9493492 DOI: 10.3389/fimmu.2022.984642] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Influenza continues to be the most important cause of viral respiratory disease, despite the availability of vaccines. Today’s evaluation of influenza vaccines mainly focuses on the quantitative and functional analyses of antibodies to the surface proteins haemagglutinin (HA) and neuraminidase (NA). However, there is an increasing interest in measuring cellular immune responses targeting not only mutation-prone surface HA and NA but also conserved internal proteins as these are less explored yet potential correlates of protection. To date, laboratories that monitor cellular immune responses use a variety of in-house procedures. This generates diverging results, complicates interlaboratory comparisons, and hampers influenza vaccine evaluation. The European FLUCOP project aims to develop and standardize assays for the assessment of influenza vaccine correlates of protection. This report describes the harmonization and qualification of the influenza-specific interferon-gamma (IFN-γ) Enzyme-Linked ImmunoSpot (ELISpot) assay. Initially, two pilot studies were conducted to identify sources of variability during sample analysis and spot enumeration in order to develop a harmonized Standard Operating Procedure (SOP). Subsequently, an assay qualification study was performed to investigate the linearity, intermediate precision (reproducibility), repeatability, specificity, Lower and Upper Limits of Quantification (LLOQ-ULOQ), Limit of Detection (LOD) and the stability of signal over time. We were able to demonstrate that the FLUCOP harmonized IFN-γ ELISpot assay procedure can accurately enumerate IFN-γ secreting cells in the analytical range of 34.4 Spot Forming Units (SFU) per million cells up to the technical limit of the used reader and in the linear range from 120 000 to 360 000 cells per well, in plates stored up to 6 weeks after development. This IFN-γ ELISpot procedure will hopefully become a useful and reliable tool to investigate influenza-specific cellular immune responses induced by natural infection or vaccination and can be an additional instrument in the search for novel correlates of protection.
Collapse
Affiliation(s)
- Gwenn Waerlop
- Center for Vaccinology (CEVAC), University Hospital, Ghent University, Ghent, Belgium
- *Correspondence: Gwenn Waerlop,
| | - Geert Leroux-Roels
- Center for Vaccinology (CEVAC), University Hospital, Ghent University, Ghent, Belgium
| | - Teresa Lambe
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Duncan Bellamy
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elena Pettini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Rebecca Jane Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Mai-Chi Trieu
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Richard Davies
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Geir Bredholt
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VisMederi srl, Siena, Italy
| | | | - Frédéric Clement
- Center for Vaccinology (CEVAC), University Hospital, Ghent University, Ghent, Belgium
| |
Collapse
|
6
|
Browne DJ, Kelly AM, Brady JL, Doolan DL. A high-throughput screening RT-qPCR assay for quantifying surrogate markers of immunity from PBMCs. Front Immunol 2022; 13:962220. [PMID: 36110843 PMCID: PMC9469018 DOI: 10.3389/fimmu.2022.962220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Immunoassays that quantitate cytokines and other surrogate markers of immunity from peripheral blood mononuclear cells (PBMCs), such as flow cytometry or Enzyme-Linked Immunosorbent Spot (ELIspot), allow highly sensitive measurements of immune effector function. However, those assays consume relatively high numbers of cells and expensive reagents, precluding comprehensive analyses and high-throughput screening (HTS). To address this issue, we developed a sensitive and specific reverse transcription-quantitative PCR (RT-qPCR)-based HTS assay, specifically designed to quantify surrogate markers of immunity from very low numbers of PBMCs. We systematically evaluated the volumes and concentrations of critical reagents within the RT-qPCR protocol, miniaturizing the assay and ultimately reducing the cost by almost 90% compared to current standard practice. We assessed the suitability of this cost-optimized RT-qPCR protocol as an HTS tool and determined the assay exceeds HTS uniformity and signal variance testing standards. Furthermore, we demonstrate this technique can effectively delineate a hierarchy of responses from as little as 50,000 PBMCs stimulated with CD4+ or CD8+ T cell peptide epitopes. Finally, we establish that this HTS-optimized protocol has single-cell analytical sensitivity and a diagnostic sensitivity equivalent to detecting 1:10,000 responding cells (i.e., 100 Spot Forming Cells/106 PBMCs by ELIspot) with over 90% accuracy. We anticipate this assay will have widespread applicability in preclinical and clinical studies, especially when samples are limited, and cost is an important consideration.
Collapse
|
7
|
De Rosa SC, Cohen KW, Bonaparte M, Fu B, Garg S, Gerard C, Goepfert PA, Huang Y, Larocque D, McElrath MJ, Morris D, Van der Most R, de Bruyn G, Pagnon A. Whole-blood cytokine secretion assay as a high-throughput alternative for assessing the cell-mediated immunity profile after two doses of an adjuvanted SARS-CoV-2 recombinant protein vaccine candidate. Clin Transl Immunology 2022; 11:e1360. [PMID: 35035955 PMCID: PMC8752373 DOI: 10.1002/cti2.1360] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/09/2021] [Accepted: 11/25/2021] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES We previously described the Phase I-II evaluation of SARS-CoV-2 recombinant protein candidate vaccine, CoV2-PreS-dTM, with AF03- or AS03-adjuvant systems (ClinicalTrials.gov, NCT04537208). Here, we further characterise the cellular immunogenicity profile of this vaccine candidate using a whole-blood secretion assay in parallel to intracellular cytokine staining (ICS) of cryopreserved peripheral blood mononuclear cells (PBMCs). METHODS A randomly allocated subset of 90 healthy, SARS-CoV-2-seronegative adults aged ≥ 18 years who had received (random allocation) one or two separate injections (on study day [D]1 and D22) of saline placebo or CoV2-PreS-dTM formulated with AS03 or AF03 were included. Cytokine secretion was assessed using a TruCulture® whole-blood stimulation system in combination with multiplex bead array, and intracellular cytokine profiles were evaluated on thawed PBMCs following ex vivo stimulation with recombinant S protein at pre-vaccination (D1), post-dose 1 (D22) and post-dose 2 (D36). RESULTS Both methods detected similar vaccine-induced responses after the first and second doses. We observed a Th1 bias (Th1/Th2 ratio > 1.0) for most treatment groups when analysed in whole blood, mainly characterised by increased IFN-γ, IL-2 and TNF-α secretion. Among participants aged ≥ 50 years, the Th1/Th2 ratio was higher for those who received vaccine candidate with AS03 versus AF03 adjuvant. ICS revealed that this higher Th1/Th2 ratio resulted from higher levels of IFN-γ expression and that the vaccine induced polyfunctional CD4+ T cells. CONCLUSIONS The whole-blood cytokine secretion assay is a high-throughput alternative for assessing the quantity and character of vaccine-induced cellular responses.
Collapse
Affiliation(s)
- Stephen C De Rosa
- Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Kristen W Cohen
- Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | | | - Bo Fu
- BiostaticsSanofi PasteurSwiftwaterPAUSA
| | | | | | - Paul A Goepfert
- Department of MedicineUniversity of Alabama at BirminghamBirminghamALUSA
| | - Ying Huang
- Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | | | - M. Juliana McElrath
- Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Daryl Morris
- Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | | | - Guy de Bruyn
- Global Clinical DevelopmentSanofi PasteurSwiftwaterPAUSA
| | - Anke Pagnon
- Research DepartmentSanofi PasteurMarcy l’ÉtoileFrance
| |
Collapse
|
8
|
Hope CM, Huynh D, Wong YY, Oakey H, Perkins GB, Nguyen T, Binkowski S, Bui M, Choo AYL, Gibson E, Huang D, Kim KW, Ngui K, Rawlinson WD, Sadlon T, Couper JJ, Penno MAS, Barry SC. Optimization of Blood Handling and Peripheral Blood Mononuclear Cell Cryopreservation of Low Cell Number Samples. Int J Mol Sci 2021; 22:ijms22179129. [PMID: 34502038 PMCID: PMC8431655 DOI: 10.3390/ijms22179129] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/14/2021] [Accepted: 08/19/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Rural/remote blood collection can cause delays in processing, reducing PBMC number, viability, cell composition and function. To mitigate these impacts, blood was stored at 4 °C prior to processing. Viable cell number, viability, immune phenotype, and Interferon-γ (IFN-γ) release were measured. Furthermore, the lowest protective volume of cryopreservation media and cell concentration was investigated. Methods: Blood from 10 individuals was stored for up to 10 days. Flow cytometry and IFN-γ ELISPOT were used to measure immune phenotype and function on thawed PBMC. Additionally, PBMC were cryopreserved in volumes ranging from 500 µL to 25 µL and concentration from 10 × 106 cells/mL to 1.67 × 106 cells/mL. Results: PBMC viability and viable cell number significantly reduced over time compared with samples processed immediately, except when stored for 24 h at RT. Monocytes and NK cells significantly reduced over time regardless of storage temperature. Samples with >24 h of RT storage had an increased proportion in Low-Density Neutrophils and T cells compared with samples stored at 4 °C. IFN-γ release was reduced after 24 h of storage, however not in samples stored at 4 °C for >24 h. The lowest protective volume identified was 150 µL with the lowest density of 6.67 × 106 cells/mL. Conclusion: A sample delay of 24 h at RT does not impact the viability and total viable cell numbers. When long-term delays exist (>4 d) total viable cell number and cell viability losses are reduced in samples stored at 4 °C. Immune phenotype and function are slightly altered after 24 h of storage, further impacts of storage are reduced in samples stored at 4 °C.
Collapse
Affiliation(s)
- Christopher M. Hope
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (C.M.H.); (D.H.); (Y.Y.W.); (H.O.); (G.B.P.); (T.N.); (T.S.); (J.J.C.); (M.A.S.P.)
- Women’s and Children’s Hospital, Adelaide, SA 5006, Australia
| | - Dao Huynh
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (C.M.H.); (D.H.); (Y.Y.W.); (H.O.); (G.B.P.); (T.N.); (T.S.); (J.J.C.); (M.A.S.P.)
| | - Ying Ying Wong
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (C.M.H.); (D.H.); (Y.Y.W.); (H.O.); (G.B.P.); (T.N.); (T.S.); (J.J.C.); (M.A.S.P.)
| | - Helena Oakey
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (C.M.H.); (D.H.); (Y.Y.W.); (H.O.); (G.B.P.); (T.N.); (T.S.); (J.J.C.); (M.A.S.P.)
| | - Griffith Boord Perkins
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (C.M.H.); (D.H.); (Y.Y.W.); (H.O.); (G.B.P.); (T.N.); (T.S.); (J.J.C.); (M.A.S.P.)
| | - Trung Nguyen
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (C.M.H.); (D.H.); (Y.Y.W.); (H.O.); (G.B.P.); (T.N.); (T.S.); (J.J.C.); (M.A.S.P.)
| | - Sabrina Binkowski
- Children’s Diabetes Centre, Telethon Kids Institute, The University of Western Australia, Perth, WA 6009, Australia; (S.B.); (A.Y.L.C.)
| | - Minh Bui
- Child Health Research Unit, Barwon Health, Geelong, VIC 3220, Australia;
| | - Ace Y. L. Choo
- Children’s Diabetes Centre, Telethon Kids Institute, The University of Western Australia, Perth, WA 6009, Australia; (S.B.); (A.Y.L.C.)
| | - Emily Gibson
- School of Women’s and Children’s Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia; (E.G.); (K.W.K.); (W.D.R.)
| | - Dexing Huang
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia; (D.H.); (K.N.)
| | - Ki Wook Kim
- School of Women’s and Children’s Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia; (E.G.); (K.W.K.); (W.D.R.)
- Virology Research Laboratory, Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, NSW 2031, Australia
| | - Katrina Ngui
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia; (D.H.); (K.N.)
| | - William D. Rawlinson
- School of Women’s and Children’s Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia; (E.G.); (K.W.K.); (W.D.R.)
- Virology Research Laboratory, Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, NSW 2031, Australia
| | - Timothy Sadlon
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (C.M.H.); (D.H.); (Y.Y.W.); (H.O.); (G.B.P.); (T.N.); (T.S.); (J.J.C.); (M.A.S.P.)
| | - Jennifer J. Couper
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (C.M.H.); (D.H.); (Y.Y.W.); (H.O.); (G.B.P.); (T.N.); (T.S.); (J.J.C.); (M.A.S.P.)
- Women’s and Children’s Hospital, Adelaide, SA 5006, Australia
| | - Megan A. S. Penno
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (C.M.H.); (D.H.); (Y.Y.W.); (H.O.); (G.B.P.); (T.N.); (T.S.); (J.J.C.); (M.A.S.P.)
| | - Simon C. Barry
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (C.M.H.); (D.H.); (Y.Y.W.); (H.O.); (G.B.P.); (T.N.); (T.S.); (J.J.C.); (M.A.S.P.)
- Women’s and Children’s Hospital, Adelaide, SA 5006, Australia
- Correspondence:
| | | |
Collapse
|
9
|
Langat RK, Farah B, Indangasi J, Ogola S, Omosa-Manyonyi G, Anzala O, Bizimana J, Tekirya E, Ngetsa C, Silwamba M, Muyanja E, Chetty P, Jangano M, Hills N, Gilmour J, Dally L, Cox JH, Hayes P. Performance of International AIDS Vaccine Initiative African clinical research laboratories in standardised ELISpot and peripheral blood mononuclear cell processing in support of HIV vaccine clinical trials. Afr J Lab Med 2021; 10:1056. [PMID: 33833946 PMCID: PMC8014752 DOI: 10.4102/ajlm.v10i1.1056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/23/2020] [Indexed: 11/28/2022] Open
Abstract
Background Standardisation of procedures for performing cellular functional assays across laboratories participating in multicentre clinical trials is key for generating comparable and reliable data. Objective This article describes the performance of accredited laboratories in Africa and Europe on testing done in support of clinical trials. Methods For enzyme-linked immunospot assay (ELISpot) proficiency, characterised peripheral blood mononuclear cells (PBMCs) obtained from 48 HIV-negative blood donors in Johannesburg, South Africa, were sent to participating laboratories between February 2010 and February 2014. The PBMCs were tested for responses against cytomegalovirus, Epstein Barr and influenza peptide pools in a total of 1751 assays. In a separate study, a total of 1297 PBMC samples isolated from healthy HIV-negative participants in clinical trials of two prophylactic HIV vaccine candidates in Kenya, Uganda, Rwanda and Zambia were analysed for cell viability, cell yield and cell recovery from frozen PBMCs. Results Most (99%) of the 1751 ELISpot proficiency assays had data within acceptable ranges with low responses to mock stimuli. No significant statistical difference were observed in ELISpot responses at the five laboratories actively conducting immunological analyses. Of the 1297 clinical trial PBMCs processed, 94% had cell viability above 90% and 96% had cell yield above 0.7 million per mL of blood in freshly isolated cells. All parameters were within the predefined acceptance criteria. Conclusion We demonstrate that multiple laboratories can generate reliable, accurate and comparable data by using standardised procedures, having regular training, having regular equipment maintenance and using centrally sourced reagents.
Collapse
Affiliation(s)
- Robert K Langat
- Kenya AIDS Vaccine Initiative, Institute of Clinical Research, University of Nairobi, Nairobi, Kenya.,International AIDS Vaccine Initiative (IAVI), Human Immunology Laboratory, Imperial College, London, United Kingdom
| | - Bashir Farah
- Kenya AIDS Vaccine Initiative, Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Jackton Indangasi
- Kenya AIDS Vaccine Initiative, Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Simon Ogola
- Kenya AIDS Vaccine Initiative, Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Gloria Omosa-Manyonyi
- Kenya AIDS Vaccine Initiative, Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Omu Anzala
- Kenya AIDS Vaccine Initiative, Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | | | | | - Caroline Ngetsa
- Kenya Medical Research Institute Centre for Geographical Medicine Research Coast, Mombasa, Kenya
| | | | - Enoch Muyanja
- Ugandan Virus Research Institute-IAVI, Entebbe, Uganda
| | - Paramesh Chetty
- International AIDS Vaccine Initiative, Johannesburg, South Africa
| | | | - Nancy Hills
- School of Medicine, University of California, San Francisco, California, United States
| | - Jill Gilmour
- International AIDS Vaccine Initiative (IAVI), Human Immunology Laboratory, Imperial College, London, United Kingdom
| | - Len Dally
- Emmes Corporation, Rockville, Maryland, United States
| | - Josephine H Cox
- Clinical Trials Program, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Peter Hayes
- International AIDS Vaccine Initiative (IAVI), Human Immunology Laboratory, Imperial College, London, United Kingdom
| |
Collapse
|
10
|
Bonilauri B, Santos MDM, Camillo-Andrade AC, Bispo S, Nogueira FCS, Carvalho PC, Zanchin NIT, Fischer JDSDG. The impact of blood-processing time on the proteome of human peripheral blood mononuclear cells. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1869:140581. [PMID: 33301959 DOI: 10.1016/j.bbapap.2020.140581] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/12/2022]
Abstract
Human peripheral blood mononuclear cells (PBMC) are key to several diagnostics assays and basic science research. Blood pre-analytical variations that occur before obtaining the PBMC fraction can significantly impact the assays results, including viability, composition, integrity, and gene expression changes of immune cells. With this as motivation, we performed a quantitative shotgun proteomics analysis using Isobaric Tag for Relative and Absolute Quantitation (iTRAQ 8plex) labeling to compare PBMC obtained from 24 h-stored blood at room temperature versus freshly isolated. We identified a total of 3195 proteins, of which 245 were differentially abundant (101 upregulated and 144 downregulated). Our results revealed enriched pathways of downregulated proteins related to exocytosis, localization, vesicle-mediated transport, cell activation, and secretion. In contrast, pathways related to exocytosis, neutrophil degranulation and activation, granulocyte activation, leukocyte degranulation, and myeloid leukocyte activation involved in immune response were enriched in upregulated proteins, which may indicate probable granulocyte contamination and activation due to blood storage time and temperature. Examples of upregulated proteins in the 24 h-PBMC samples are CAMP, S100A8, LTA4H, RASAL3, and S100A6, which are involved in an adaptive immune system and antimicrobial activity, proinflammatory mediation, aminopeptidase activities, and naïve T cells survival. Moreover, examples of downregulated proteins are NDUFA5, TAGLN2, H3C1, TUBA8, and CCT2 that are related to the cytoskeleton, cell junction, mitochondrial respiratory chain. In conclusion, the delay in blood-processing time directly impacts the proteomic profile of human PBMC, possibly through granulocyte contamination and activation.
Collapse
Affiliation(s)
- Bernardo Bonilauri
- Laboratory of Basic Biology of Stem Cells, Carlos Chagas Institute, Fiocruz-PR, Brazil
| | - Marlon D M Santos
- Laboratory for Structural and Computational Proteomics, Carlos Chagas Institute, Fiocruz-PR, Brazil
| | | | - Saloê Bispo
- Laboratory for Structural and Computational Proteomics, Carlos Chagas Institute, Fiocruz-PR, Brazil
| | - Fabio C S Nogueira
- Proteomic Unit, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo C Carvalho
- Laboratory for Structural and Computational Proteomics, Carlos Chagas Institute, Fiocruz-PR, Brazil
| | - Nilson I T Zanchin
- Laboratory for Structural Biology and Protein Engineering, Carlos Chagas Institute, Fiocruz-PR, Brazil.
| | - Juliana de S da G Fischer
- Laboratory for Structural and Computational Proteomics, Carlos Chagas Institute, Fiocruz-PR, Brazil.
| |
Collapse
|
11
|
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
|
12
|
Hermansen JU, Tjønnfjord GE, Munthe LA, Taskén K, Skånland SS. Cryopreservation of primary B cells minimally influences their signaling responses. Sci Rep 2018; 8:17651. [PMID: 30518828 PMCID: PMC6281576 DOI: 10.1038/s41598-018-36121-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023] Open
Abstract
Phospho flow is a powerful approach to detect cell signaling aberrations, identify biomarkers and assess pharmacodynamics, and can be performed using cryopreserved samples. The effects of cryopreservation on signaling responses and the reproducibility of phospho flow measurements are however unknown in many cell systems. Here, B lymphocytes were isolated from healthy donors and patients with the B cell malignancy chronic lymphocytic leukemia and analyzed by phospho flow using phospho-specific antibodies targeting 20 different protein epitopes. Cells were analyzed both at basal conditions and after activation of cluster of differentiation 40 (CD40) or the B cell receptor. Pharmacodynamics of the novel pathway inhibitor ibrutinib was also assessed. At all conditions, fresh cells were compared to cryopreserved cells. Minimal variation between fresh and frozen samples was detected. Reproducibility was tested by running samples from the same donors in different experiments. The results demonstrate reproducibility across different phospho flow runs and support the use of cryopreserved samples in future phospho flow studies of B lymphocytes.
Collapse
Affiliation(s)
- Johanne U Hermansen
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Geir E Tjønnfjord
- Department of Haematology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway
| | - Ludvig A Munthe
- K. G. Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway.,Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Kjetil Taskén
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway.,K. G. Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway.,K. G. Jebsen Centre for Cancer Immunotherapy, University of Oslo, Oslo, Norway.,Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Sigrid S Skånland
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway. .,K. G. Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway. .,K. G. Jebsen Centre for Cancer Immunotherapy, University of Oslo, Oslo, Norway. .,Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
13
|
Thurgood LA, Lower KM, Macardle C, Kuss BJ. Aberrant determination of phenotypic markers in chronic lymphocytic leukemia (CLL) lymphocytes after cryopreservation. Exp Hematol 2018; 63:28-32.e1. [PMID: 29705268 DOI: 10.1016/j.exphem.2018.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 02/01/2023]
Abstract
The cryopreservation of peripheral blood mononuclear cells (PBMCs) is a routine research laboratory process, enabling long-term storage of primary patient blood samples. Retrospective analysis of these samples has the potential to identify markers that may be associated with prognosis and response to treatment. To draw valid biological conclusions from this type of analysis, it is essential to ensure that any observed changes are directly related to the pathology of the disease rather than the preservation process itself. Therefore, we have investigated 15 cell surface markers that are relevant to chronic lymphocytic leukemia (CLL) on matched fresh and thawed samples to determine the effect of cryopreservation on their detection. We found that the number of CLL cells positive for the markers CD22, CD40, CD49d, CD54, CD69, and CXCR3 was decreased significantly after cryopreservation. In addition, the mean fluorescence intensity of 10 of the 15 markers changed significantly after cryopreservation. These findings demonstrate that care must be taken when interpreting this type of analysis on thawed samples.
Collapse
Affiliation(s)
- Lauren A Thurgood
- Discipline of Molecular Medicine and Pathology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.
| | - Karen M Lower
- Discipline of Molecular Medicine and Pathology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Cindy Macardle
- Department of Immunology, Allergy and Arthritis, SA Pathology, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Bryone J Kuss
- Discipline of Molecular Medicine and Pathology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia; Hematology, Molecular Medicine and Pathology, SA Pathology, Flinders Medical Centre, Adelaide, South Australia, Australia
| |
Collapse
|
14
|
Luo Y, Wang P, Liu H, Zhu Z, Li C, Gao Y. The state of T cells before cryopreservation: Effects on post-thaw proliferation and function. Cryobiology 2017; 79:65-70. [PMID: 28863950 DOI: 10.1016/j.cryobiol.2017.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/23/2017] [Accepted: 08/29/2017] [Indexed: 02/07/2023]
Abstract
AIM We aim to assess the effect of the state of T cells before cryopreservation on the post-thaw proliferative capacity, phenotype and functional response. METHODS Peripheral blood mononuclear cells (PBMCs) were isolated from a hepatocellular carcinoma (HCC) patient, and the T cells were frozen during cell culture according to our experimental design. After a period of re-culture, the proliferative capacity of the cryopreserved cells, the expression of T cell surface markers and the secretion of IFN-γ and IL-10 were assayed. RESULTS There was >90% cell viability after thaw in every group. Lymphocytes cryopreserved at day 4, 8 or 12 during the cell culture were allowed to recover for 24 h, whereas lymphocytes cryopreserved while freshly isolated were allowed to recover for 72 h. After the period of re-culture, cryopreservation at day 4, 8 or 12 during T cell culture was not found to alter the T cell subpopulation. The proportions of NKT and Treg cells were unchanged when cells were cryopreserved at day 12 during T cell culture. IFN-γ secretion was not impacted by cryopreservation, and IL-10 secretion was significantly decreased when cells were cryopreserved at day 8 or 12 during T cell culture. CONCLUSION The state of T cells before cryopreservation has effects on the post-thaw proliferation capacity, the phenotype and the secretion of IFN-γ and IL-10. Cryopreservation of lymphocytes at day 8 or 12 during the cell culture may be the best choice for T cell immunotherapy.
Collapse
Affiliation(s)
- Ying Luo
- Tianjin Key Laboratory of Artificial Cell, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Third Central Hospital of Tianjin, Tianjin, 300170, China.
| | - Peng Wang
- Third Central Clinical College of Tianjin Medical University, Tianjin, 300170, China.
| | - Hui Liu
- Tianjin Key Laboratory of Artificial Cell, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Third Central Hospital of Tianjin, Tianjin, 300170, China.
| | - Zhengyan Zhu
- Tianjin Key Laboratory of Artificial Cell, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Third Central Hospital of Tianjin, Tianjin, 300170, China.
| | - Chenglong Li
- Tianjin Key Laboratory of Artificial Cell, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Third Central Hospital of Tianjin, Tianjin, 300170, China.
| | - Yingtang Gao
- Tianjin Key Laboratory of Artificial Cell, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Third Central Hospital of Tianjin, Tianjin, 300170, China; Third Central Clinical College of Tianjin Medical University, Tianjin, 300170, China.
| |
Collapse
|
15
|
Optimizing recovery of frozen human peripheral blood mononuclear cells for flow cytometry. PLoS One 2017; 12:e0187440. [PMID: 29091947 PMCID: PMC5665600 DOI: 10.1371/journal.pone.0187440] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/19/2017] [Indexed: 11/28/2022] Open
Abstract
Introduction Live peripheral blood mononuclear cells (PBMCs) can be frozen and thawed for later analyses by adding and removing a cryoprotectant, such as dimethyl sulfoxide (DMSO). Laboratories across the world use various procedures, but published evidence of optimal thawing procedures is scarce. Materials and methods PBMCs were separated from blood collected from healthy Danish blood donors, and stored at -80°C after adding of DMSO. The essential steps in the thawing procedure were modified and performance was evaluated by flow cytometry with respect to the percentage and total yield of viable PMBCs. Results The best-performing washing medium was Roswell Park Memorial Institute (RPMI) 1640 at 37°C with 20% fetal bovine serum. When using 10 mL washing medium in a 15-mL Falcon tube, samples should be centrifuged for at least 10 minutes at 500 g. We failed to detect any differences between the tested methods of mixing PBMCs with washing medium. Likewise, neither the thawing duration nor centrifugation temperature (20°C and 37°C) had any effect. PBMCs could be incubated (rested) for up to eight hours in a 37°C 5% CO2 incubator without affecting cell counts, but incubating PBMCs for 16 hours significantly decreased viability and recovery. In general, high viability was not necessarily associated with high recovery. Conclusion Changing the thawing procedure significantly impacted PBMC viability and live cell recovery. Evaluating both viability and live PBMC recovery are necessary to evaluate method performance. Investigation of differential loss of PBMC subtypes and phenotypic changes during thawing and incubation requires further evaluation.
Collapse
|
16
|
Stroncek DF, Butterfield LH, Cannarile MA, Dhodapkar MV, Greten TF, Grivel JC, Kaufman DR, Kong HH, Korangy F, Lee PP, Marincola F, Rutella S, Siebert JC, Trinchieri G, Seliger B. Systematic evaluation of immune regulation and modulation. J Immunother Cancer 2017; 5:21. [PMID: 28331613 PMCID: PMC5359947 DOI: 10.1186/s40425-017-0223-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 02/10/2017] [Indexed: 02/06/2023] Open
Abstract
Cancer immunotherapies are showing promising clinical results in a variety of malignancies. Monitoring the immune as well as the tumor response following these therapies has led to significant advancements in the field. Moreover, the identification and assessment of both predictive and prognostic biomarkers has become a key component to advancing these therapies. Thus, it is critical to develop systematic approaches to monitor the immune response and to interpret the data obtained from these assays. In order to address these issues and make recommendations to the field, the Society for Immunotherapy of Cancer reconvened the Immune Biomarkers Task Force. As a part of this Task Force, Working Group 3 (WG3) consisting of multidisciplinary experts from industry, academia, and government focused on the systematic assessment of immune regulation and modulation. In this review, the tumor microenvironment, microbiome, bone marrow, and adoptively transferred T cells will be used as examples to discuss the type and timing of sample collection. In addition, potential types of measurements, assays, and analyses will be discussed for each sample. Specifically, these recommendations will focus on the unique collection and assay requirements for the analysis of various samples as well as the high-throughput assays to evaluate potential biomarkers.
Collapse
Affiliation(s)
- David F Stroncek
- Department of Transfusion Medicine, National Institutes of Health, 10 Center Drive, Building 10, Room 3C720, Bethesda, MD 20892 USA
| | - Lisa H Butterfield
- Department of Medicine, Surgery and Immunology, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| | - Michael A Cannarile
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Nonnenwald 2, 82377 Penzberg, Germany
| | - Madhav V Dhodapkar
- Department of Hematology & Immunobiology, Yale University, 333 Cedar Street, Box 208021, New Haven, CT 06510 USA
| | - Tim F Greten
- GI-Malignancy Section, Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 Room 12 N226, 9000 Rockville, Bethesda, MD 20892 USA
| | - Jean Charles Grivel
- Division of Translational Medicine, Sidra Medical and Research Center, PO Box 26999, Al Luqta Street, Doha, Qatar
| | - David R Kaufman
- Merck Research Laboratories, PO Box 1000, UG 3CD28, North Wales, PA 19454 USA
| | - Heidi H Kong
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, MSC 1908, Bethesda, MD 20892-1908 USA
| | - Firouzeh Korangy
- GI-Malignancy Section, Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 Room 12 N226, 9000 Rockville, Bethesda, MD 20892 USA
| | - Peter P Lee
- Department of Immuno-Oncology, City of Hope, 1500 East Duarte Road, Duarte, CA 91010 USA
| | - Francesco Marincola
- Division of Translational Medicine, Sidra Medical and Research Center, PO Box 26999, Al Luqta Street, Doha, Qatar
| | - Sergio Rutella
- The John van Geest Cancer Research Centre, Nottingham Trent University, Clifton Campus, Nottingham, NG11 8NS UK
| | - Janet C Siebert
- CytoAnalytics, 3500 South Albion Street, Cherry Hills Village, CO 80113 USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37/Room 4146, Bethesda, MD 20892 USA
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, Halle, Germany
| |
Collapse
|
17
|
Pacholewska A, Marti E, Leeb T, Jagannathan V, Gerber V. LPS-induced modules of co-expressed genes in equine peripheral blood mononuclear cells. BMC Genomics 2017; 18:34. [PMID: 28056766 PMCID: PMC5217269 DOI: 10.1186/s12864-016-3390-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/07/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Lipopolysaccharide (endotoxin, LPS) is a strong inducer of the innate immune response. It is widespread in our environment, e.g. in house dust and contributes to asthma. Compared to humans, horses are even more sensitive to LPS. However, data on LPS effects on the equine transcriptome are very limited. Using RNA-seq we analysed LPS-induced differences in the gene expression in equine peripheral blood mononuclear cells at the gene and gene-network level in two half-sib families and one group of unrelated horses. RESULTS 24 h-LPS challenge of equine immune cells resulted in substantial changes in the transcriptomic profile (1,265 differentially expressed genes) showing partial overlap with human data. One of the half-sib families showed a specific response different from the other two groups of horses. We also identified co-expressed gene modules that clearly differentiated 24 h-LPS- from non-stimulated samples. These modules consisted of 934 highly interconnected genes and included genes involved in the immune response (e.g. IL6, CCL22, CXCL6, CXCL2), however, none of the top ten hub genes of the modules have been annotated as responsive to LPS in gene ontology. CONCLUSIONS Using weighted gene co-expression network analysis we identified ten co-expressed gene modules significantly regulated by in vitro stimulation with LPS. Apart from 47 genes (5%) all other genes highly interconnected within the most up- and down-regulated modules were also significantly differentially expressed (FDR < 0.05). The LPS-regulated module hub genes have not yet been described as having a role in the immune response to LPS (e.g. VAT1 and TTC25).
Collapse
Affiliation(s)
- Alicja Pacholewska
- Department of Clinical Veterinary Medicine, Swiss Institute of Equine Medicine, Vetsuisse Faculty, University of Bern, and Agroscope, Länggassstrasse 124, 3012, Bern, Switzerland. .,Department of Clinical Research and Veterinary Public Health, Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109A, 3012, Bern, Switzerland.
| | - Eliane Marti
- Department of Clinical Research and Veterinary Public Health, Division of Experimental Clinical Research, Vetsuisse Faculty, University of Bern, Länggassstrasse 124, 3012, Bern, Switzerland
| | - Tosso Leeb
- Department of Clinical Research and Veterinary Public Health, Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109A, 3012, Bern, Switzerland
| | - Vidhya Jagannathan
- Department of Clinical Research and Veterinary Public Health, Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109A, 3012, Bern, Switzerland
| | - Vincent Gerber
- Department of Clinical Veterinary Medicine, Swiss Institute of Equine Medicine, Vetsuisse Faculty, University of Bern, and Agroscope, Länggassstrasse 124, 3012, Bern, Switzerland
| |
Collapse
|
18
|
Masucci GV, Cesano A, Hawtin R, Janetzki S, Zhang J, Kirsch I, Dobbin KK, Alvarez J, Robbins PB, Selvan SR, Streicher HZ, Butterfield LH, Thurin M. Validation of biomarkers to predict response to immunotherapy in cancer: Volume I - pre-analytical and analytical validation. J Immunother Cancer 2016; 4:76. [PMID: 27895917 PMCID: PMC5109744 DOI: 10.1186/s40425-016-0178-1] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/20/2016] [Indexed: 12/31/2022] Open
Abstract
Immunotherapies have emerged as one of the most promising approaches to treat patients with cancer. Recently, there have been many clinical successes using checkpoint receptor blockade, including T cell inhibitory receptors such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death-1 (PD-1). Despite demonstrated successes in a variety of malignancies, responses only typically occur in a minority of patients in any given histology. Additionally, treatment is associated with inflammatory toxicity and high cost. Therefore, determining which patients would derive clinical benefit from immunotherapy is a compelling clinical question. Although numerous candidate biomarkers have been described, there are currently three FDA-approved assays based on PD-1 ligand expression (PD-L1) that have been clinically validated to identify patients who are more likely to benefit from a single-agent anti-PD-1/PD-L1 therapy. Because of the complexity of the immune response and tumor biology, it is unlikely that a single biomarker will be sufficient to predict clinical outcomes in response to immune-targeted therapy. Rather, the integration of multiple tumor and immune response parameters, such as protein expression, genomics, and transcriptomics, may be necessary for accurate prediction of clinical benefit. Before a candidate biomarker and/or new technology can be used in a clinical setting, several steps are necessary to demonstrate its clinical validity. Although regulatory guidelines provide general roadmaps for the validation process, their applicability to biomarkers in the cancer immunotherapy field is somewhat limited. Thus, Working Group 1 (WG1) of the Society for Immunotherapy of Cancer (SITC) Immune Biomarkers Task Force convened to address this need. In this two volume series, we discuss pre-analytical and analytical (Volume I) as well as clinical and regulatory (Volume II) aspects of the validation process as applied to predictive biomarkers for cancer immunotherapy. To illustrate the requirements for validation, we discuss examples of biomarker assays that have shown preliminary evidence of an association with clinical benefit from immunotherapeutic interventions. The scope includes only those assays and technologies that have established a certain level of validation for clinical use (fit-for-purpose). Recommendations to meet challenges and strategies to guide the choice of analytical and clinical validation design for specific assays are also provided.
Collapse
Affiliation(s)
- Giuseppe V Masucci
- Department of Oncology-Pathology, Karolinska Institutet, 171 76 Stockholm, Sweden
| | | | - Rachael Hawtin
- Nodality, Inc, 170 Harbor Way, South San Francisco, 94080 CA USA
| | - Sylvia Janetzki
- ZellNet Consulting, Inc, 555 North Avenue, Fort Lee, 07024 NJ USA
| | - Jenny Zhang
- Covaris Inc, 14 Gill St, Woburn, MA 01801 USA
| | - Ilan Kirsch
- Adaptive Biotechnologies, Inc, 1551 Eastlake Ave. E, Seattle, WA 98102 USA
| | - Kevin K Dobbin
- Department of Epidemiology and Biostatistics, College of Public Health, The University of Georgia, 101 Buck Road, Athens, 30602 GA USA
| | - John Alvarez
- Janssen Research & Development, LLC, Spring House, PA 19477 USA
| | | | - Senthamil R Selvan
- Omni Array Biotechnology, 15601 Crabbs Branch Way, Rockville, 20855 MD USA
| | - Howard Z Streicher
- National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, 20892 MD USA
| | - Lisa H Butterfield
- Department of Medicine, Surgery and Immunology, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| | - Magdalena Thurin
- National Cancer Institute, Cancer Diagnosis Program, DCTD, National Institutes of Health, 9609 Medical Center Drive, Bethesda, 20892 MD USA ; Adaptive Biotechnologies, Inc, 1551 Eastlake Ave. E, Seattle, WA 98102 USA
| |
Collapse
|
19
|
Angel S, von Briesen H, Oh YJ, Baller MK, Zimmermann H, Germann A. Toward Optimal Cryopreservation and Storage for Achievement of High Cell Recovery and Maintenance of Cell Viability and T Cell Functionality. Biopreserv Biobank 2016; 14:539-547. [PMID: 27792414 PMCID: PMC5180082 DOI: 10.1089/bio.2016.0046] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Cryopreservation of biological materials such as cells, tissues, and organs is a prevailing topic of high importance. It is employed not only in many research fields but also in the clinical area. Cryopreservation is of great importance for reproductive medicine and clinical studies, as well as for the development of vaccines. Peripheral blood mononuclear cells (PBMCs) are commonly used in vaccine research where comparable and reliable results between different research institutions and laboratories are of high importance. Whereas freezing and thawing processes are well studied, controlled, and standardized, storage conditions are often disregarded. To close this gap, we investigated the influence of suboptimal storage conditions during low-temperature storage on PBMC viability, recovery, and T cell functionality. For this purpose, PBMCs were isolated and exposed with help of a robotic system in a low-temperature environment from 0 up to 350 temperature fluctuation cycles in steps of 50 cycles to simulate storage conditions in large biorepositories with sample storage, removal, and sorting functions. After the simulation, the viability, recovery, and T cell functionality were analyzed to determine the number of temperature rises, which ultimately lead to significant cell damage. All studied parameters decreased with increasing number of temperature cycles. Sometimes after as little as only 50 temperature cycles, a significant effect was observed. These results are very important for all fields in which cell cryopreservation is employed, particularly for clinical and multicenter studies wherein the comparability and reproducibility of results play a crucial role. To obtain reliable results and to maintain the quality of the cells, not only the freezing and thawing processes but also the storage conditions should be controlled and standardized, and any deviations should be documented.
Collapse
Affiliation(s)
- Stephanie Angel
- 1 Fraunhofer Institute for Biomedical Engineering , Sulzbach, Germany
| | - Hagen von Briesen
- 1 Fraunhofer Institute for Biomedical Engineering , Sulzbach, Germany
| | - Young-Joo Oh
- 1 Fraunhofer Institute for Biomedical Engineering , Sulzbach, Germany
| | - Marko K Baller
- 2 University of Applied Sciences Kaiserslautern , Amerikastraße, Zweibruecken, Germany
| | - Heiko Zimmermann
- 1 Fraunhofer Institute for Biomedical Engineering , Sulzbach, Germany .,3 Department of Molecular and Cellular Biotechnology, Saarland University , Saarbruecken, Germany
| | - Anja Germann
- 1 Fraunhofer Institute for Biomedical Engineering , Sulzbach, Germany
| |
Collapse
|
20
|
Calleros-Basilio L, Cortés MA, García-Jerez A, Luengo-Rodríguez A, Orozco-Agudo A, Valdivielso JM, Rodríguez-Puyol D, Rodríguez-Puyol M. Quality Assurance of Samples and Processes in the Spanish Renal Research Network (REDinREN) Biobank. Biopreserv Biobank 2016; 14:499-510. [PMID: 27541936 DOI: 10.1089/bio.2015.0095] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Biobanks are useful platforms to build bridges between basic, translational, and clinical research and clinical care. They are repositories of high-quality human biological samples ideal for evaluating their histological characteristics and also their genome, transcriptome, and proteome. The Spanish Renal Research Network Biobank contains more than 76,500 well-preserved frozen samples of a wide variety of kidney diseases, collected from 5450 patients seen by over 70 nephrology services throughout the Spanish territory. OBJECTIVE To determine and to report the results of the quality control of samples and processes conducted in our biobank, implemented in accordance with the requirements of the ISO 9001:2008 international standard. STUDY DESIGN Two types of quality controls were performed: (1) systematic, that is, measurement of viable peripheral blood mononuclear cells (PBMCs) obtained and purity of nucleic acids and (2) ad-hoc, that is, viability of thawed PBMC, DNA extraction process reproducibility, and the integrity and functionality of nucleic acids, implemented on a routine basis. METHODS AND RESULTS PBMC isolation by Ficoll yielded reproducible results and its cryopreserved viability was >90%. Acceptable A260/A280 ratios were obtained for the vast majority of the DNA (n = 2328) and RNA (n = 78) samples analyzed. DNA integrity was demonstrated by agarose gels and by β-globulin gene polymerase chain reaction (PCR) amplification of 1327 and 989 bp fragments. DNA of acceptable quality had at least three bands of β-globulin amplified obtained (n = 26/30). RNA integrity number (RIN) determinations obtained RIN numbers ≥7 (n = 87/96). The amplifiability of nucleic acids was confirmed by qPCR and RT-qPCR of β-actin and GAPDH genes. Long storage or delayed processing time did not affect the quality of the samples analyzed. The processes of DNA extraction also yielded reproducible results. CONCLUSIONS These results clearly indicate that our PBMC, DNA, and RNA stored samples meet the required quality standards to be used for biomedical research, ensuring their long-term preservation.
Collapse
Affiliation(s)
- Laura Calleros-Basilio
- 1 Physiology Unit, Department of Systems Biology, Medicine School, Alcala University , Madrid, Spain .,2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain
| | - María Alicia Cortés
- 3 CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Medicine School, Universidad Nacional del Nordeste , Corrientes, Argentina
| | - Andrea García-Jerez
- 1 Physiology Unit, Department of Systems Biology, Medicine School, Alcala University , Madrid, Spain .,2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain
| | - Alicia Luengo-Rodríguez
- 1 Physiology Unit, Department of Systems Biology, Medicine School, Alcala University , Madrid, Spain .,2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain
| | - Ana Orozco-Agudo
- 1 Physiology Unit, Department of Systems Biology, Medicine School, Alcala University , Madrid, Spain .,2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain
| | - José Manuel Valdivielso
- 2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain .,4 Department of Experimental Nephrology, Institut de Recerca Biomédica de Lleida, Universitat de Lleida , Lleida, Spain
| | - Diego Rodríguez-Puyol
- 2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain .,5 Nephrology Section and Research Unit, Hospital Universitario Príncipe de Asturias , Alcalá de Henares, Madrid, Spain
| | - Manuel Rodríguez-Puyol
- 1 Physiology Unit, Department of Systems Biology, Medicine School, Alcala University , Madrid, Spain .,2 IRSIN and REDinREN (Instituto de Salud Carlos III), Madrid, Spain
| |
Collapse
|
21
|
Lama JR, Karuna ST, Grant SP, Swann EM, Ganoza C, Segura P, Montano SM, Lacherre M, De Rosa SC, Buchbinder S, Sanchez J, McElrath MJ, Lemos MP. Transient Peripheral Immune Activation follows Elective Sigmoidoscopy or Circumcision in a Cohort Study of MSM at Risk of HIV Infection. PLoS One 2016; 11:e0160487. [PMID: 27536938 PMCID: PMC4990246 DOI: 10.1371/journal.pone.0160487] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 07/20/2016] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Rectal and genital sampling in HIV prevention trials permits assessments at the site of HIV entry. Yet the safety and acceptability of circumcision and sigmoidoscopy (and associated abstinence recommendations) are unknown in uncircumcised men who have sex with men (MSM) at high risk of HIV infection. METHODS Twenty-nine HIV-seronegative high-risk Peruvian MSM agreed to elective sigmoidoscopy biopsy collections (weeks 2 and 27) and circumcision (week 4) in a 28-week cohort study designed to mimic an HIV vaccine study mucosal collection protocol. We monitored adherence to abstinence recommendations, procedure-related complications, HIV infections, peripheral immune activation, and retention. RESULTS Twenty-three (79.3%) underwent a first sigmoidoscopy, 21 (72.4%) were circumcised, and 16 (55.2%) completed a second sigmoidoscopy during the study period. All who underwent procedures completed the associated follow-up safety visits. Those completing the procedures reported they were well tolerated, and complication rates were similar to those reported in the literature. Immune activation was detected during the healing period (1 week post-sigmoidoscopy, 6 weeks post-circumcision), including increases in CCR5+CD4+T cells and α4β7+CD4+T cells. Most participants adhered to post-circumcision abstinence recommendations whereas reduced adherence occurred post-sigmoidoscopy. CONCLUSION Rectosigmoid mucosal and genital tissue collections were safe in high-risk MSM. Although the clinical implications of the post-procedure increase in peripheral immune activation markers are unknown, they reinforce the need to provide ongoing risk reduction counseling and support for post-procedure abstinence recommendations. Future HIV vaccine studies should also consider the effects of mucosal and tissue collections on peripheral blood endpoints in trial design and analysis. TRIAL REGISTRATION ClinicalTrials.gov NCT02630082.
Collapse
Affiliation(s)
| | - Shelly T. Karuna
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Shannon P. Grant
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Edith M. Swann
- Vaccine Clinical Research Branch, Division of AIDS, National Institutes of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, Maryland, United States of America
| | | | | | | | | | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Susan Buchbinder
- San Francisco Department of Health, San Francisco, California, United States of America
| | - Jorge Sanchez
- Asociacion Civil Impacta Salud y Educacion, Lima, Peru
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Maria P. Lemos
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | | |
Collapse
|
22
|
Sei JJ, Cox KS, Dubey SA, Antonello JM, Krah DL, Casimiro DR, Vora KA. Effector and Central Memory Poly-Functional CD4(+) and CD8(+) T Cells are Boosted upon ZOSTAVAX(®) Vaccination. Front Immunol 2015; 6:553. [PMID: 26579128 PMCID: PMC4629102 DOI: 10.3389/fimmu.2015.00553] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 10/16/2015] [Indexed: 11/13/2022] Open
Abstract
ZOSTAVAX(®) is a live attenuated varicella-zoster virus (VZV) vaccine that is licensed for the protection of individuals ≥50 years against shingles and its most common complication, postherpetic neuralgia. While IFNγ responses increase upon vaccination, the quality of the T cell response has not been elucidated. By using polychromatic flow cytometry, we characterized the breadth, magnitude, and quality of ex vivo CD4(+) and CD8(+) T cell responses induced 3-4 weeks after ZOSTAVAX vaccination of healthy adults. We show, for the first time that the highest frequencies of VZV-specific CD4(+) T cells were poly-functional CD154(+)IFNγ(+)IL-2(+)TNFα(+) cells, which were boosted upon vaccination. The CD4(+) T cells were broadly reactive to several VZV proteins, with immediate early (IE) 63 ranking the highest among them in the fold rise of poly-functional cells, followed by IE62, gB, open reading frame (ORF) 9, and gE. We identified a novel poly-functional ORF9-specific CD8(+) T cell population in 62% of the subjects, and these were boosted upon vaccination. Poly-functional CD4(+) and CD8(+) T cells produced significantly higher levels of IFNγ, IL-2, and TNFα compared to mono-functional cells. After vaccination, a boost in the expression of IFNγ by poly-functional IE63- and ORF9-specific CD4(+) T cells and IFNγ, IL-2, and TNFα by ORF9-specific poly-functional CD8(+) T cells was observed. Responding poly-functional T cells exhibited both effector (CCR7(-)CD45RA(-)CD45RO(+)), and central (CCR7(+)CD45RA(-)CD45RO(+)) memory phenotypes, which expressed comparable levels of cytokines. Altogether, our studies demonstrate that a boost in memory poly-functional CD4(+) T cells and ORF9-specific CD8(+) T cells may contribute toward ZOSTAVAX efficacy.
Collapse
Affiliation(s)
- Janet J Sei
- Merck Research Laboratories, Department Vaccine Analytical Development, Merck & Co., Inc. , Kenilworth, NJ , USA
| | - Kara S Cox
- Merck Research Laboratories, Department of Infectious Diseases and Vaccines, Merck & Co., Inc. , Kenilworth, NJ , USA
| | - Sheri A Dubey
- Merck Research Laboratories, Department of Infectious Diseases and Vaccines, Merck & Co., Inc. , Kenilworth, NJ , USA
| | - Joseph M Antonello
- Merck Research Laboratories, Department of Infectious Diseases and Vaccines, Merck & Co., Inc. , Kenilworth, NJ , USA
| | - David L Krah
- Merck Research Laboratories, Department Vaccine Analytical Development, Merck & Co., Inc. , Kenilworth, NJ , USA
| | - Danilo R Casimiro
- Merck Research Laboratories, Department of Infectious Diseases and Vaccines, Merck & Co., Inc. , Kenilworth, NJ , USA
| | - Kalpit A Vora
- Merck Research Laboratories, Department of Infectious Diseases and Vaccines, Merck & Co., Inc. , Kenilworth, NJ , USA
| |
Collapse
|
23
|
Trück J, Mitchell R, Thompson AJ, Morales-Aza B, Clutterbuck EA, Kelly DF, Finn A, Pollard AJ. Effect of cryopreservation of peripheral blood mononuclear cells (PBMCs) on the variability of an antigen-specific memory B cell ELISpot. Hum Vaccin Immunother 2015; 10:2490-6. [PMID: 25424961 DOI: 10.4161/hv.29318] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The ELISpot assay is used in vaccine studies for the quantification of antigen-specific memory B cells (B(MEM)), and can be performed using cryopreserved samples. The effects of cryopreservation on B(MEM) detection and the consistency of cultured ELISpot assays when performed by different operators or laboratories are unknown. In this study, blood was taken from healthy volunteers, and a cultured ELISpot assay was used to count B(MEM) specific for 2 routine vaccine antigens (diphtheria and tetanus toxoid). Results were assessed for intra- and inter-operator variation, and the effects of cryopreservation. Cryopreserved samples were shipped to a second laboratory in order to assess inter-laboratory variation. B(MEM) frequencies were very strongly correlated when comparing fresh and frozen samples processed by the same operator, and were also very strongly correlated when comparing 2 operators in the same laboratory. Results were slightly less consistent when samples were processed in different laboratories but correlation between the 2 measurements was still very strong. Although cell viability was reduced in some cryopreserved samples due to higher temperatures during transportation, B(MEM) could still be quantified. These results demonstrate the reproducibility of the ELISpot assay across operators and laboratories, and support the use of cryopreserved samples in future B(MEM) studies.
Collapse
Affiliation(s)
- Johannes Trück
- a Oxford Vaccine Group; Department of Paediatrics; University of Oxford and the NIHR Oxford Biomedical Research Centre; Oxford, UK
| | | | | | | | | | | | | | | |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Bourguignon P, Clément F, Renaud F, Le Bras V, Koutsoukos M, Burny W, Moris P, Lorin C, Collard A, Leroux-Roels G, Roman F, Janssens M, Vandekerckhove L. Processing of blood samples influences PBMC viability and outcome of cell-mediated immune responses in antiretroviral therapy-naïve HIV-1-infected patients. J Immunol Methods 2014; 414:1-10. [PMID: 25224748 DOI: 10.1016/j.jim.2014.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 09/01/2014] [Accepted: 09/05/2014] [Indexed: 12/22/2022]
Abstract
Intracellular cytokine staining (ICS) assay is increasingly used in vaccine clinical trials to measure antigen-specific T-cell mediated immune (CMI) responses in cryopreserved peripheral blood mononuclear cells (PBMCs) and whole blood. However, recent observations indicate that several parameters involved in blood processing can impact PBMC viability and CMI responses, especially in antiretroviral therapy (ART)-naïve HIV-1-infected individuals. In this phase I study (NCT01610427), we collected blood samples from 22 ART-naïve HIV-1-infected adults. PBMCs were isolated and processed for ICS assay. The individual and combined effects of the following parameters were investigated: time between blood collection and PBMC processing (time-to-process: 2, 7 or 24 h); time between PBMC thawing and initiation of in vitro stimulation with HIV-1 antigens (resting-time: 0, 2, 6 and 18 h); and duration of antigen-stimulation in PBMC cultures (stimulation-time: 6h or overnight). The cell recovery after thawing, cell viability after ICS and magnitude of HIV-specific CD8(+) T-cell responses were considered to determine the optimal combination of process conditions. The impact of time-to-process (2 or 4 h) on HIV-specific CD8(+) T-cell responses was also assessed in a whole blood ICS assay. A higher quality of cells in terms of recovery and viability (up to 81% and >80% respectively) was obtained with shorter time-to-process (less than 7 h) and resting-time (less than 2 h) intervals. Longer (overnight) rather than shorter (6 h) stimulation-time intervals increased the frequency of CD8(+)-specific T-cell responses using ICS in PBMCs without change of the functionality. The CD8(+) specific T-cell responses detected using fresh whole blood showed a good correlation with the responses detected using frozen PBMCs. Our results support the need of standardized procedures for the evaluation of CMI responses, especially in HIV-1-infected, ART-naïve patients.
Collapse
Affiliation(s)
| | - Frédéric Clément
- Center for Vaccinology, Ghent University and Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| | - Frédéric Renaud
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Vivien Le Bras
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | | | - Wivine Burny
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Philippe Moris
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Clarisse Lorin
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Alix Collard
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Geert Leroux-Roels
- Center for Vaccinology, Ghent University and Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| | - François Roman
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Michel Janssens
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Linos Vandekerckhove
- ARC (AIDS Reference Center), Department of Internal Medicine, Ghent University and Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| |
Collapse
|
27
|
The Center for HIV/AIDS Vaccine Immunology (CHAVI) multi-site quality assurance program for cryopreserved human peripheral blood mononuclear cells. J Immunol Methods 2014; 409:21-30. [PMID: 24910414 DOI: 10.1016/j.jim.2014.05.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 05/23/2014] [Accepted: 05/28/2014] [Indexed: 01/17/2023]
Abstract
The Center for HIV/AIDS Vaccine Immunology (CHAVI) consortium was established to determine the host and virus factors associated with HIV transmission, infection and containment of virus replication, with the goal of advancing the development of an HIV protective vaccine. Studies to meet this goal required the use of cryopreserved Peripheral Blood Mononuclear Cell (PBMC) specimens, and therefore it was imperative that a quality assurance (QA) oversight program be developed to monitor PBMC samples obtained from study participants at multiple international sites. Nine site-affiliated laboratories in Africa and the USA collected and processed PBMCs, and cryopreserved PBMC were shipped to CHAVI repositories in Africa and the USA for long-term storage. A three-stage program was designed, based on Good Clinical Laboratory Practices (GCLP), to monitor PBMC integrity at each step of this process. The first stage evaluated the integrity of fresh PBMCs for initial viability, overall yield, and processing time at the site-affiliated laboratories (Stage 1); for the second stage, the repositories determined post-thaw viability and cell recovery of cryopreserved PBMC, received from the site-affiliated laboratories (Stage 2); the third stage assessed the long-term specimen storage at each repository (Stage 3). Overall, the CHAVI PBMC QA oversight program results highlight the relative importance of each of these stages to the ultimate goal of preserving specimen integrity from peripheral blood collection to long-term repository storage.
Collapse
|
28
|
Benefits of a comprehensive quality program for cryopreserved PBMC covering 28 clinical trials sites utilizing an integrated, analytical web-based portal. J Immunol Methods 2014; 409:9-20. [PMID: 24709391 DOI: 10.1016/j.jim.2014.03.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 02/28/2014] [Accepted: 03/28/2014] [Indexed: 11/20/2022]
Abstract
The HIV Vaccine Trials Network (HVTN) is a global network of 28 clinical trial sites dedicated to identifying an effective HIV vaccine. Cryopreservation of high-quality peripheral blood mononuclear cells (PBMC) is critical for the assessment of vaccine-induced cellular immune functions. The HVTN PBMC Quality Management Program is designed to ensure that viable PBMC are processed, stored and shipped for clinical trial assays from all HVTN clinical trial sites. The program has evolved by developing and incorporating best practices for laboratory and specimen quality and implementing automated, web-based tools. These tools allow the site-affiliated processing laboratories and the central Laboratory Operations Unit to rapidly collect, analyze and report PBMC quality data. The HVTN PBMC Quality Management Program includes five key components: 1) Laboratory Assessment, 2) PBMC Training and Certification, 3) Internal Quality Control, 4) External Quality Control (EQC), and 5) Assay Specimen Quality Control. Fresh PBMC processing data is uploaded from each clinical site processing laboratory to a central HVTN Statistical and Data Management Center database for access and analysis on a web portal. Samples are thawed at a central laboratory for assay or specimen quality control and sample quality data is uploaded directly to the database by the central laboratory. Four year cumulative data covering 23,477 blood draws reveals an average fresh PBMC yield of 1.45×10(6)±0.48 cells per milliliter of useable whole blood. 95% of samples were within the acceptable range for fresh cell yield of 0.8-3.2×10(6) cells/ml of usable blood. Prior to full implementation of the HVTN PBMC Quality Management Program, the 2007 EQC evaluations from 10 international sites showed a mean day 2 thawed viability of 83.1% and a recovery of 67.5%. Since then, four year cumulative data covering 3338 specimens used in immunologic assays shows that 99.88% had acceptable viabilities (>66%) for use in cellular assays (mean, 91.46% ±4.5%), and 96.2% had acceptable recoveries (50%-130%) with a mean of recovery of 85.8% ±19.12% of the originally cryopreserved cells. EQC testing revealed that since August 2009, failed recoveries dropped from 4.1% to 1.6% and failed viabilities dropped from 1.0% to 0.3%. The HVTN PBMC quality program provides for laboratory assessment, training and tools for identifying problems, implementing corrective action and monitoring for improvements. These data support the benefits of implementing a comprehensive, web-based PBMC quality program for large clinical trials networks.
Collapse
|
29
|
Kurzweil V, Tang R, Galinski M, Wang K, Zuo F, Cherukuri A, Gasser RA, Malkin E, Sifakis F, Mendel DB, Esser MT. Translational sciences approach to RSV vaccine development. Expert Rev Vaccines 2014; 12:1047-60. [PMID: 24053398 DOI: 10.1586/14760584.2013.824706] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections in infants and the elderly. Despite its relatively low degree of antigenic variation, it causes frequent reinfection throughout life. Clinical manifestations of RSV disease and the immune response to infection differ in infants and the elderly, suggesting that vaccines designed to protect these two populations may require different attributes. Here, the authors describe the translational approach of utilizing data from epidemiology studies performed in these populations, the use of RSV diagnostics in clinical practice, lessons learned from previous vaccine clinical trials and the success of palivizumab in prevention of RSV disease in premature and high-risk infants to aid the development of safe and effective RSV vaccines.
Collapse
Affiliation(s)
- Vanessa Kurzweil
- Cell and Molecular Biology Group, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kutscher S, Dembek CJ, Deckert S, Russo C, Körber N, Bogner JR, Geisler F, Umgelter A, Neuenhahn M, Albrecht J, Cosma A, Protzer U, Bauer T. Overnight resting of PBMC changes functional signatures of antigen specific T- cell responses: impact for immune monitoring within clinical trials. PLoS One 2013; 8:e76215. [PMID: 24146841 PMCID: PMC3795753 DOI: 10.1371/journal.pone.0076215] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/21/2013] [Indexed: 11/19/2022] Open
Abstract
Polyfunctional CD4 or CD8 T cells are proposed to represent a correlate of immune control for persistent viruses as well as for vaccine mediated protection against infection. A well-suited methodology to study complex functional phenotypes of antiviral T cells is the combined staining of intracellular cytokines and phenotypic marker expression using polychromatic flow cytometry. In this study we analyzed the effect of an overnight resting period at 37°C on the quantity and functionality of HIV-1, EBV, CMV, HBV and HCV specific CD4 and CD8 T-cell responses in a cohort of 21 individuals. We quantified total antigen specific T cells by multimer staining and used 10-color intracellular cytokine staining (ICS) to determine IFNγ, TNFα, IL2 and MIP1β production. After an overnight resting significantly higher numbers of functionally active T cells were detectable by ICS for all tested antigen specificities, whereas the total number of antigen specific T cells determined by multimer staining remained unchanged. Overnight resting shifted the quality of T-cell responses towards polyfunctionality and increased antigen sensitivity of T cells. Our data suggest that the observed effect is mediated by T cells rather than by antigen presenting cells. We conclude that overnight resting of PBMC prior to ex vivo analysis of antiviral T-cell responses represents an efficient method to increase sensitivity of ICS-based methods and has a prominent impact on the functional phenotype of T cells.
Collapse
Affiliation(s)
- Sarah Kutscher
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
| | - Claudia J. Dembek
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| | - Simone Deckert
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
| | - Carolina Russo
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
| | - Nina Körber
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
| | - Johannes R. Bogner
- Department of Infectious Diseases/Med. Klinik und Poliklinik, University Hospital of Munich/Ludwig Maximilians Universität, Munich, Germany
| | - Fabian Geisler
- Department of Internal Medicine II, Klinikum rechts der Isar/Technische Universität München, Munich, Germany
| | - Andreas Umgelter
- Department of Internal Medicine II, Klinikum rechts der Isar/Technische Universität München, Munich, Germany
| | - Michael Neuenhahn
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
- Institute of Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| | - Julia Albrecht
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
- Institute of Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| | | | - Ulrike Protzer
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| | - Tanja Bauer
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
- Cooperation Group ‘Immune Monitoring’, Helmholtz Zentrum München, Munich, Germany
- * E-mail:
| |
Collapse
|
31
|
Britten CM, Walter S, Janetzki S. Immunological Monitoring to Rationally Guide AAV Gene Therapy. Front Immunol 2013; 4:273. [PMID: 24062741 PMCID: PMC3770921 DOI: 10.3389/fimmu.2013.00273] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 08/27/2013] [Indexed: 12/12/2022] Open
Abstract
Recent successes with adeno-associated virus (AAV)-based gene therapies fuel the hope for new treatments for hereditary diseases. Pre-existing as well as therapy-induced immune responses against both AAV and the encoded transgenes have been described and may impact on safety and efficacy of gene therapy approaches. Consequently, monitoring of vector- and transgene-specific immunity is mandated and may rationally guide clinical development. Next to the humoral immune response, the cellular response is central in our understanding of the host reaction in gene therapy. But in contrast to the monitoring of antibodies, which has matured over many decades, sensitive and robust monitoring of T cells is a relatively new development. To make cellular immune assessments fit for purpose, investigators need to know, control and report the critical assay variables that influence the results. In addition, the quality of immune assays needs to be continuously adjusted to allow for exploratory hypothesis generation in early stages and confirmatory hypothesis validation in later stages of clinical development. The concept of immune assay harmonization which includes use of field-wide benchmarks, harmonization guidelines, and external quality control can support the context-specific evolution of immune assays. Multi-center studies pose particular challenges to sample logistics and quality control of sample specimens. Cooperative groups need to define if immune assessments should be performed in one central facility, in peripheral labs or including a combination of both. Finally, engineered reference samples that contain a defined number of antigen-specific T cells may become broadly applicable tools to control assay performance over time or across institutions.
Collapse
Affiliation(s)
- Cedrik Michael Britten
- Translational Oncology, University Medical Center, Johannes Gutenberg-University Mainz (TRON gGmbH) , Mainz , Germany ; Association for Cancer Immunotherapy (CIMT) , Mainz , Germany
| | | | | |
Collapse
|
32
|
Germann A, Oh YJ, Schmidt T, Schön U, Zimmermann H, von Briesen H. Temperature fluctuations during deep temperature cryopreservation reduce PBMC recovery, viability and T-cell function. Cryobiology 2013; 67:193-200. [PMID: 23850825 DOI: 10.1016/j.cryobiol.2013.06.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 06/24/2013] [Accepted: 06/26/2013] [Indexed: 10/26/2022]
Abstract
The ability to analyze cryopreserved peripheral blood mononuclear cell (PBMC) from biobanks for antigen-specific immunity is necessary to evaluate response to immune-based therapies. To ensure comparable assay results, collaborative research in multicenter trials needs reliable and reproducible cryopreservation that maintains cell viability and functionality. A standardized cryopreservation procedure is comprised of not only sample collection, preparation and freezing but also low temperature storage in liquid nitrogen without any temperature fluctuations, to avoid cell damage. Therefore, we have developed a storage approach to minimize suboptimal storage conditions in order to maximize cell viability, recovery and T-cell functionality. We compared the influence of repeated temperature fluctuations on cell health from sample storage, sample sorting and removal in comparison to sample storage without temperature rises. We found that cyclical temperature shifts during low temperature storage reduce cell viability, recovery and immune response against specific-antigens. We showed that samples handled under a protective hood system, to avoid or minimize such repeated temperature rises, have comparable cell viability and cell recovery rates to samples stored without any temperature fluctuations. Also T-cell functionality could be considerably increased with the use of the protective hood system compared to sample handling without such a protection system. This data suggests that the impact of temperature fluctuation on cell integrity should be carefully considered in future clinical vaccine trials and consideration should be given to optimal sample storage conditions.
Collapse
Affiliation(s)
- Anja Germann
- (a)Fraunhofer Institute for Biomedical Engineering, Ensheimerstr. 48, 66386 St. Ingbert, Germany
| | | | | | | | | | | |
Collapse
|
33
|
Kobayashi DT, Decker D, Zaworski P, Klott K, McGonigal J, Ghazal N, Sly L, Chung B, Vanderlugt J, Chen KS. Evaluation of peripheral blood mononuclear cell processing and analysis for Survival Motor Neuron protein. PLoS One 2012; 7:e50763. [PMID: 23226377 PMCID: PMC3511312 DOI: 10.1371/journal.pone.0050763] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 10/24/2012] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES Survival Motor Neuron (SMN) protein levels may become key pharmacodynamic (PD) markers in spinal muscular atrophy (SMA) clinical trials. SMN protein in peripheral blood mononuclear cells (PBMCs) can be quantified for trials using an enzyme-linked immunosorbent assay (ELISA). We developed protocols to collect, process, store and analyze these samples in a standardized manner for SMA clinical studies, and to understand the impact of age and intraindividual variability over time on PBMC SMN signal. METHODS Several variables affecting SMN protein signal were evaluated using an ELISA. Samples were from healthy adults, adult with respiratory infections, SMA patients, and adult SMA carriers. RESULTS Delaying PBMCs processing by 45 min, 2 hr or 24 hr after collection or isolation allows sensitive detection of SMN levels and high cell viability (>90%). SMN levels from PBMCs isolated by EDTA tubes/Lymphoprep gradient are stable with processing delays and have greater signal compared to CPT-collected samples. SMN signal in healthy individuals varies up to 8x when collected at intervals up to 1 month. SMN signals from individuals with respiratory infections show 3-5x changes, driven largely by the CD14 fraction. SMN signal in PBMC frozen lysates are relatively stable for up to 6 months. Cross-sectional analysis of PBMCs from SMA patients and carriers suggest SMN protein levels decline with age. CONCLUSIONS The sources of SMN signal variability in PBMCs need to be considered in the design and of SMA clinical trials, and interpreted in light of recent medical history. Improved normalization to DNA or PBMC subcellular fractions may mitigate signal variability and should be explored in SMA patients.
Collapse
Affiliation(s)
- Dione T Kobayashi
- Spinal Muscular Atrophy Foundation, New York, New York, United States of America.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW The purpose of this article is to describe the requirements for clinical laboratories supporting large-scale multinational trials of prophylactic AIDS vaccine trials and review the progress made. RECENT FINDINGS There is an increasing need for laboratories in Africa, Asia and the Caribbean to support internationally initiated and funded clinical trials of preventive HIV vaccine candidates. A number of qualified laboratories are currently supporting AIDS vaccine trials in these regions, although there remains a need to develop capacity further. The standardization of all tests is key in order that data can be pooled and compared across multiple sites and products. Significant progress has been made towards this aim. The recent development of quality programmes including good clinical laboratory practices are key to ensuring data are reliable and meet the requirements of regulatory bodies. In addition, HIV diagnostic tests are being developed to distinguish true HIV infection from vaccine-induced antibodies. SUMMARY Significant advances have been made to develop laboratories capable of supporting multinational AIDS vaccine trials.
Collapse
|
35
|
Kuerten S, Batoulis H, Recks MS, Karacsony E, Zhang W, Subbramanian RA, Lehmann PV. Resting of Cryopreserved PBMC Does Not Generally Benefit the Performance of Antigen-Specific T Cell ELISPOT Assays. Cells 2012; 1:409-27. [PMID: 24710483 PMCID: PMC3901103 DOI: 10.3390/cells1030409] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 07/12/2012] [Accepted: 07/14/2012] [Indexed: 12/15/2022] Open
Abstract
T cell monitoring is increasingly performed using cryopreserved PBMC. It has been suggested that resting of PBMC after thawing, that is, culturing them overnight in test medium, produces higher antigen-induced spot counts in ELISPOT assays. To evaluate the importance of overnight resting, we systematically tested cryopreserved PBMC from 25 healthy donors. CEF peptides (comprising CMV, EBV and flu antigens) were used to stimulate CD8 cells and mumps antigen to stimulate CD4 cells. The data show that resting significantly increased antigen-elicited T cell responses only for CEF high responder PBMC. The maximal gain observed was doubling of spot counts. For CEF low responders, and for mumps responders of either low- or high reactivity levels, resting had no statistically significant effect on the observed spot counts. Therefore, resting is not a generally applicable approach to improve ELISPOT assay performance, but can be recommended only for clinical subject cohorts and antigens for which it has a proven benefit. Because resting invariably leads to losing about half of the PBMC available for testing, and because doubling the PBMC numbers plated into the assay reliably doubles the antigen-induced spot counts, we suggest the latter approach as a simple and reliable alternative to resting for enhancing the performance of ELISPOT assays. Our data imply that resting is not required if PBMC were cryopreserved and thawed under conditions that minimize apoptosis of the cells. Therefore, this study should draw attention to the need to optimize freezing and thawing conditions for successful T cell work.
Collapse
Affiliation(s)
- Stefanie Kuerten
- Department of Anatomy I, University of Cologne, Joseph-Stelzmann-Str. 9, 50931 Cologne, Germany.
| | - Helena Batoulis
- Department of Anatomy I, University of Cologne, Joseph-Stelzmann-Str. 9, 50931 Cologne, Germany.
| | - Mascha S Recks
- Department of Anatomy I, University of Cologne, Joseph-Stelzmann-Str. 9, 50931 Cologne, Germany.
| | - Edith Karacsony
- Cellular Technology Ltd. (C.T.L), Shaker Heights, OH 44122, USA.
| | - Wenji Zhang
- Cellular Technology Ltd. (C.T.L), Shaker Heights, OH 44122, USA.
| | | | - Paul V Lehmann
- Cellular Technology Ltd. (C.T.L), Shaker Heights, OH 44122, USA.
| |
Collapse
|
36
|
Reynolds MR, Weiler AM, Piaskowski SM, Piatak M, Robertson HT, Allison DB, Bett AJ, Casimiro DR, Shiver JW, Wilson NA, Lifson JD, Koff WC, Watkins DI. A trivalent recombinant Ad5 gag/pol/nef vaccine fails to protect rhesus macaques from infection or control virus replication after a limiting-dose heterologous SIV challenge. Vaccine 2012; 30:4465-75. [PMID: 22569124 PMCID: PMC3372643 DOI: 10.1016/j.vaccine.2012.04.082] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 03/28/2012] [Accepted: 04/21/2012] [Indexed: 11/25/2022]
Abstract
It has been suggested that poor immunogenicity may explain the lack of vaccine efficacy in preventing or controlling HIV infection in the Step trial. To investigate this issue we vaccinated eight Indian rhesus macaques with a trivalent replication-incompetent adenovirus serotype 5 vaccine expressing SIV Gag, Pol, and Nef using a regimen similar to that employed in the Step trial. We detected broad vaccine-induced CD8(+) (2-7 pool-specific responses) and CD4(+) (5-19 pool-specific responses) T-cell responses in IFN-γ ELISPOT assays at one week post-boost using fresh PBMC. However, using cryopreserved cells at one and four weeks post-boost we observed a reduction in both the number and magnitude of most vaccine-induced responses. This demonstrates that the time points and conditions chosen to perform immune assays may influence the observed breadth and frequency of vaccine-induced T-cell responses. To evaluate protective efficacy, we challenged the immunized macaques, along with naïve controls, with repeated, limiting doses of the heterologous swarm isolate SIVsmE660. Vaccination did not significantly affect acquisition or control of virus replication in vaccinees compared to naïve controls. Post-infection we observed an average of only two anamnestic CD8(+) T-cell responses per animal, which may not have been sufficiently broad to control heterologous virus replication. While the trivalent vaccine regimen induced relatively broad T-cell responses in rhesus macaques, it failed to protect against infection or control viral replication. Our results are consistent with those observed in the Step trial and indicate that SIV immunization and challenge studies in macaque models of HIV infection can be informative in assessing pre-clinical HIV vaccines.
Collapse
Affiliation(s)
- Matthew R. Reynolds
- AIDS Vaccine Research Laboratory, 555 Science Drive, Madison, Wisconsin 53711, USA
| | - Andrea M. Weiler
- AIDS Vaccine Research Laboratory, 555 Science Drive, Madison, Wisconsin 53711, USA
| | - Shari M. Piaskowski
- AIDS Vaccine Research Laboratory, 555 Science Drive, Madison, Wisconsin 53711, USA
| | - Michael Piatak
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., National Cancer Institute, Frederick, Maryland 21702, USA
| | - Henry T. Robertson
- Department of Biostatistics, Section on Statistical Genetics, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - David B. Allison
- Department of Biostatistics, Section on Statistical Genetics, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Andrew J. Bett
- Department of Vaccine Basic Research, Merck Research Laboratories, West Point, PA 19486, USA
| | - Danilo R. Casimiro
- Department of Vaccine Basic Research, Merck Research Laboratories, West Point, PA 19486, USA
| | - John W. Shiver
- Department of Vaccine Basic Research, Merck Research Laboratories, West Point, PA 19486, USA
| | - Nancy A. Wilson
- AIDS Vaccine Research Laboratory, 555 Science Drive, Madison, Wisconsin 53711, USA
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., National Cancer Institute, Frederick, Maryland 21702, USA
| | - Wayne C. Koff
- International AIDS Vaccine Initiative, New York, New York 10038, USA
| | - David I. Watkins
- AIDS Vaccine Research Laboratory, 555 Science Drive, Madison, Wisconsin 53711, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
37
|
Schulz JC, Germann A, Kemp-Kamke B, Mazzotta A, von Briesen H, Zimmermann H. Towards a xeno-free and fully chemically defined cryopreservation medium for maintaining viability, recovery, and antigen-specific functionality of PBMC during long-term storage. J Immunol Methods 2012; 382:24-31. [PMID: 22580762 DOI: 10.1016/j.jim.2012.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/14/2011] [Accepted: 05/01/2012] [Indexed: 11/29/2022]
Abstract
Analysis of cryopreserved peripheral mononuclear cells (PBMC) is important for evaluating new vaccines in immune based therapies and in pathogenesis studies. To ensure comparable assay results from different laboratories and points of time, collaborative research in multicenter trials needs reliable and reproducible cryopreservation protocols that maintain cell viability and functionality. Current cryomedia consist largely of fetal bovine serum (FBS), a natural mix of growth factors, cytokines, and undefined compounds. Standardized procedures are not possible, as FBS can affect the antigen-specific T-cell response, the most important parameter in functionality assays. Also, worldwide sample exchange is complicated by the strict import restrictions on FBS, because of transfection risk. After establishing a serum-free cryopreservation protocol that maintains cell viability, recovery and antigen-specific T-cell response of PBMC comparably to FBS-based cryomedia (Germann et al., 2011), the aim of this study was the complete avoidance of animal proteins and products in combination with efficient cryopreservation. As long-term stability of the cryopreservation process is crucial for retrospective evaluation of samples at different points of time, PBMC were analyzed after storage for maximal four weeks and again after approximately six months. The cryopreservation efficiency of the protein-free and fully chemically defined cryomedium was comparable to FBS-medium after storage for few weeks and several months. Directly after thawing, this medium yielded viabilities over 97% and recovery values over 84%. Also, the specific T-cell functionality was preserved. Additionally, short-term and six month cryopreservation gave comparable results. The fully chemically defined medium presented here will increase standardization and reproducibility of analysis in multicenter-studies or in retrospective evaluation.
Collapse
Affiliation(s)
- Julia C Schulz
- Fraunhofer Institute for Biomedical Engineering, 66386 St. Ingbert, Germany
| | | | | | | | | | | |
Collapse
|
38
|
Detection of vaccinia virus-specific IFNγ and IL-10 secretion from human PBMCs and CD8⁺ T cells by ELISPOT. Methods Mol Biol 2012; 792:199-218. [PMID: 21956512 DOI: 10.1007/978-1-61779-325-7_16] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
High-throughput in vitro assays, which rapidly and succinctly assess the immune status of large cohorts of individuals, are essential tools for conducting population-based studies, including vaccine research. The enzyme-linked immunospot (ELISPOT) assay has emerged as a sensitive, reliable high-throughput tool to measure functional recall immunity by assessing the frequency of antigen-specific cytokine-secreting lymphocytes present in peripheral blood mononuclear cells (PBMCs). For the past 10 years, ELISPOT method has been the dominant platform and a standard for the cell-mediated immune (CMI) assays. ELISPOT assays are used extensively as a measure of CMI response to vaccines, including smallpox (vaccinia), following primary or secondary vaccination. Here, we present detailed methodology for using ELISPOT assays to detect the frequency of cytokine secreting vaccinia-specific lymphocytes including optimized protocols for growing, titrating, and inactivating vaccinia virus; isolating, cryopreserving, and thawing human PBMCs; and finally, detecting vaccinia-specific IL-10 and IFNγ secreting lymphocytes, as well as CD8(+) IFNγ T cells following in vitro stimulation of PBMCs with vaccinia virus. The methods presented below, although optimized for vaccinia virus, emphasize principles that can be generally applied to create ELISPOT assays capable of assessing the immune status as well as antiviral CD8(+) T cell response of individuals following primary or secondary vaccination with other licensed or novel vaccines.
Collapse
|
39
|
Butterfield LH, Potter DM, Kirkwood JM. Multiplex serum biomarker assessments: technical and biostatistical issues. J Transl Med 2011; 9:173. [PMID: 21989127 PMCID: PMC3200183 DOI: 10.1186/1479-5876-9-173] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 10/11/2011] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Identification of predictive and prognostic biomarkers for patients with disease and undergoing different therapeutic options is a very active area of investigation. Many of these studies seek biomarkers among circulating proteins accessed in blood. Many levels of standardization in materials and procedures have been identified which can impact the resulting data. METHODS Here, we have observed unexpected variability in levels of commonly tested analytes in serum which were processed and stored under standardized conditions. We have identified apparent changes in cytokine, chemokine and growth factor levels detected by multiplex Luminex assay in melanoma patient and healthy donor serum samples, over storage time at -80°C. Controls included Luminex kit standards, multiplexed cytokine standards and WHO cytokine controls. Data were analyzed by Wilcoxon rank-sum testing and Spearman's test for correlations. RESULTS The interpretation of these changes is confounded by lot-to-lot kit standard curve reagent changes made by a single manufacturer of Luminex kits. CONCLUSIONS This study identifies previously unknown sources of variation in a commonly used biomarker assay, and suggests additional levels of controls needed for identification of true changes in circulating protein levels.
Collapse
Affiliation(s)
- Lisa H Butterfield
- University of Pittsburgh Cancer Institute, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- University of Pittsburgh School of Medicine, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- Department of Surgery, University of Pittsburgh, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
| | - Douglas M Potter
- University of Pittsburgh Cancer Institute, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- University of Pittsburgh School of Medicine, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- Depament of Biostatistics, University of Pittsburgh, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
| | - John M Kirkwood
- University of Pittsburgh Cancer Institute, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- University of Pittsburgh School of Medicine, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh, 5117 Centre Avenue, suite 1.27, Pittsburgh, PA 15213, USA
| |
Collapse
|
40
|
A double-blind randomized phase I clinical trial targeting ALVAC-HIV vaccine to human dendritic cells. PLoS One 2011; 6:e24254. [PMID: 21949699 PMCID: PMC3174939 DOI: 10.1371/journal.pone.0024254] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 08/05/2011] [Indexed: 01/08/2023] Open
Abstract
Background We conducted a novel pilot study comparing different delivery routes of ALVAC-HIV (vCP205), a canarypox vaccine containing HIV gene inserts: env, gag and pol. We explored the concept that direct ex vivo targeting of human dendritic cells (DC) would enhance the immune response compared to either conventional intramuscular or intradermal injections of the vaccine alone. Methodology/Principal Findings Healthy HIV-1 uninfected volunteers were administered ALVAC-HIV or placebo by intramuscular injection (IM), intradermal injection (ID) or subcutaneous injection (SQ) of autologous ex vivo transfected DC at months 0, 1, 3 and 6. All vaccine delivery routes were well tolerated. Binding antibodies were observed to both the ALVAC vector and HIV-1 gp160 proteins. Modest cellular responses were observed in 2/7 individuals in the DC arm and 1/8 in the IM arm as determined by IFN-γ ELISPOT. Proliferative responses were most frequent in the DC arm where 4/7 individuals had measurable responses to multiple HIV-1 antigens. Loading DC after maturation resulted in lower gene expression, but overall better responses to both HIV-1 and control antigens, and were associated with better IL-2, TNF-α and IFN-γ production. Conclusions/Significance ALVAC-HIV delivered IM, ID or SQ with autologous ex vivo transfected DC proved to be safe. The DC arm was most immunogenic. Proliferative immune responses were readily detected with only modest cytotoxic CD8 T cell responses. Loading mature DC with the live viral vaccine induced stronger immune responses than loading immature DC, despite increased transgene expression with the latter approach. Volunteers who received the autologous vaccine loaded mature DC developed a broader and durable immune response compared to those vaccinated by conventional routes. Trial Registration ClinicalTrials.gov NCT00013572
Collapse
|
41
|
Butterfield LH, Palucka AK, Britten CM, Dhodapkar MV, Håkansson L, Janetzki S, Kawakami Y, Kleen TO, Lee PP, Maccalli C, Maecker HT, Maino VC, Maio M, Malyguine A, Masucci G, Pawelec G, Potter DM, Rivoltini L, Salazar LG, Schendel DJ, Slingluff CL, Song W, Stroncek DF, Tahara H, Thurin M, Trinchieri G, van Der Burg SH, Whiteside TL, Wigginton JM, Marincola F, Khleif S, Fox BA, Disis ML. Recommendations from the iSBTc-SITC/FDA/NCI Workshop on Immunotherapy Biomarkers. Clin Cancer Res 2011; 17:3064-76. [PMID: 21558394 DOI: 10.1158/1078-0432.ccr-10-2234] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To facilitate development of innovative immunotherapy approaches, especially for treatment concepts exploiting the potential benefits of personalized therapy, there is a need to develop and validate tools to identify patients who can benefit from immunotherapy. Despite substantial effort, we do not yet know which parameters of antitumor immunity to measure and which assays are optimal for those measurements. EXPERIMENTAL DESIGN The iSBTc-SITC (International Society for Biological Therapy of Cancer-Society for Immunotherapy of Cancer), FDA (Food and Drug Administration), and NCI (National Cancer Institute) partnered to address these issues for immunotherapy of cancer. Here, we review the major challenges, give examples of approaches and solutions, and present our recommendations. RESULTS AND CONCLUSIONS Although specific immune parameters and assays are not yet validated, we recommend following standardized (accurate, precise, and reproducible) protocols and use of functional assays for the primary immunologic readouts of a trial; consideration of central laboratories for immune monitoring of large, multi-institutional trials; and standardized testing of several phenotypic and functional potential potency assays specific to any cellular product. When reporting results, the full QA (quality assessment)/QC (quality control) should be conducted and selected examples of truly representative raw data and assay performance characteristics should be included. Finally, to promote broader analysis of multiple aspects of immunity, and gather data on variability, we recommend that in addition to cells and serum, RNA and DNA samples be banked (under standardized conditions) for later testing. We also recommend that sufficient blood be drawn to allow for planned testing of the primary hypothesis being addressed in the trial, and that additional baseline and posttreatment blood is banked for testing novel hypotheses (or generating new hypotheses) that arise in the field.
Collapse
Affiliation(s)
- Lisa H Butterfield
- Department of Medicine, University of Pittsburgh, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Nicholson O, DiCandilo F, Kublin J, Sun X, Quirk E, Miller M, Gray G, Pape J, Robertson MN, Mehrotra DV, Self S, Turner K, Sanchez J, Pitisuttithum P, Duerr A, Dubey S, Kierstead L, Casimiro D, Hammer For The Merck V/Hiv Vaccine Trials Network Study Team SM. Safety and Immunogenicity of the MRKAd5 gag HIV Type 1 Vaccine in a Worldwide Phase 1 Study of Healthy Adults. AIDS Res Hum Retroviruses 2011; 27:557-567. [PMID: 20854108 PMCID: PMC3422055 DOI: 10.1089/aid.2010.0151] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The safety and immunogenicity of the MRK adenovirus type 5 (Ad5) HIV-1 clade B gag vaccine was assessed in an international Phase I trial. Three-hundred and sixty healthy HIV-uninfected adults were enrolled on five continents. Subjects received placebo or 1 × 109 or 1 × 1010 viral particles (vp) per dose of the MRKAd5 HIV-1 gag vaccine at day 1, week 4, and week 26. Immunogenicity was evaluated using an IFN-γ ELISPOT gag 15-mer assay with positive responses defined as ≥55 SFC/106 PBMCs and ≥4-fold over mock control. The vaccine was well tolerated. The most common adverse events were injection site reactions, headache, pyrexia, diarrhea, fatigue, and myalgia. At week 30, geometric mean ELISPOT responses were 24, 114, and 226 SFC/106 PBMCs in the placebo, 1 × 109 vp/dose, and 1 × 1010 vp/dose groups, respectively. Overall, responses to 1 × 1010 vp were 85% and 68% in subjects with low (≤200) and high (>200) baseline Ad5 titers, respectively. The MRKAd5 HIV-1 gag vaccine was immunogenic in diverse geographic regions. Gag ELISPOT responses were greater in the 1 × 1010 vp/dose groups than in the 1 × 109 vp/dose groups. Data from this first international study indicate that adenovirus-vectored vaccines are well tolerated and may be immunogenic in subjects from regions with high prevalence of preexisting Ad5 immunity.
Collapse
Affiliation(s)
- Ouzama Nicholson
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Fay DiCandilo
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - James Kublin
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Xiao Sun
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Erin Quirk
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Michelle Miller
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Glenda Gray
- University of Witwatersrand, Johannesburg, South Africa
| | - Jean Pape
- Weill Cornell Medical College, New York, New York
| | - Michael N Robertson
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Devan V Mehrotra
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Steven Self
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Jorge Sanchez
- Asociacion Civil Impacta Salud y Educacion, Lima, Peru and Investigaciones Medicas en Salu, Immensa, Lima, Peru
| | | | - Ann Duerr
- Fred Hutchinson Cancer Research Center, Seattle, Washington
- University of Washington, Seattle, Washington
| | - Sheri Dubey
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Lisa Kierstead
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | - Danilo Casimiro
- Merck & Co., Inc., North Wales, Pennsylvania; current affiliation: GSK Biologicals, King of Prussia, Pennsylvania
| | | |
Collapse
|
43
|
Pine SO, Kublin JG, Hammer SM, Borgerding J, Huang Y, Casimiro DR, McElrath MJ. Pre-existing adenovirus immunity modifies a complex mixed Th1 and Th2 cytokine response to an Ad5/HIV-1 vaccine candidate in humans. PLoS One 2011; 6:e18526. [PMID: 21533229 PMCID: PMC3076372 DOI: 10.1371/journal.pone.0018526] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 03/03/2011] [Indexed: 11/30/2022] Open
Abstract
The results of the recent Step Study highlight a need to clarify the effects of pre-existing natural immunity to a vaccine vector on vaccine-induced T-cell responses. To investigate this interaction, we examined the relationship between pre-existing Ad5 immunity and T-cell cytokine response profiles in healthy, HIV-uninfected recipients of MRKAd5 HIV-1 gag vaccine (HVTN 050, ClinicalTrials.gov #NCT00849732). Participants were grouped by baseline Ad5 neutralizing antibody titer as either Ad5-seronegative (titer ≤18; n = 36) or Ad5-seropositive (titer >200; n = 34). Samples from vaccine recipients were analyzed for immune responses to either HIV-1 Gag peptide pools or Ad5 empty vector using an ex vivo assay that measures thirty cytokines in the absence of long-term culture. The overall profiles of cytokine responses to Gag and Ad5 had similar combinations of induced Th1- and Th2-type cytokines, including IFN-γ, IL-2, TNF-α, IP-10, IL-13, and IL-10, although the Ad5-specific responses were uniformly higher than the Gag-specific responses (p<0.0001 for 9 out of 11 significantly expressed analytes). At the peak response time point, PBMC from Ad5-seronegative vaccinees secreted significantly more IP-10 in response to Gag (p = 0.008), and significantly more IP-10 (p = 0.0009), IL-2 (p = 0.006) and IL-10 (p = 0.05) in response to Ad5 empty vector than PBMC from Ad5-seropositive vaccinees. Additionally, similar responses to the Ad5 vector prior to vaccination were observed in almost all subjects, regardless of Ad5 neutralizing antibody status, and the levels of secreted IFN-γ, IL-10, IL-1Ra and GM-CSF were blunted following vaccination. The cytokine response profile of Gag-specific T cells mirrored the Ad5-specific response present in all subjects before vaccination, and included a number of Th1- and Th2-associated cytokines not routinely assessed in current vaccine trials, such as IP-10, IL-10, IL-13, and GM-CSF. Together, these results suggest that vector-specific humoral responses may reduce vaccine-induced T-cell responses by previously undetected mechanisms.
Collapse
Affiliation(s)
- Samuel O. Pine
- Program in Pathobiology, Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - James G. Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Scott M. Hammer
- Division of Infectious Diseases, Columbia University College of Physicians and Surgeons, New York, New York, United States of America
| | - Joleen Borgerding
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Danilo R. Casimiro
- Vaccine Basic Research, Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - M. Juliana McElrath
- Program in Pathobiology, Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
44
|
Olson WC, Smolkin ME, Farris EM, Fink RJ, Czarkowski AR, Fink JH, Chianese-Bullock KA, Slingluff CL. Shipping blood to a central laboratory in multicenter clinical trials: effect of ambient temperature on specimen temperature, and effects of temperature on mononuclear cell yield, viability and immunologic function. J Transl Med 2011; 9:26. [PMID: 21385453 PMCID: PMC3063218 DOI: 10.1186/1479-5876-9-26] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 03/08/2011] [Indexed: 01/26/2023] Open
Abstract
Background Clinical trials of immunologic therapies provide opportunities to study the cellular and molecular effects of those therapies and may permit identification of biomarkers of response. When the trials are performed at multiple centers, transport and storage of clinical specimens become important variables that may affect lymphocyte viability and function in blood and tissue specimens. The effect of temperature during storage and shipment of peripheral blood on subsequent processing, recovery, and function of lymphocytes is understudied and represents the focus of this study. Methods Peripheral blood samples (n = 285) from patients enrolled in 2 clinical trials of a melanoma vaccine were shipped from clinical centers 250 or 1100 miles to a central laboratory at the sponsoring institution. The yield of peripheral blood mononuclear cells (PBMC) collected before and after cryostorage was correlated with temperatures encountered during shipment. Also, to simulate shipping of whole blood, heparinized blood from healthy donors was collected and stored at 15°C, 22°C, 30°C, or 40°C, for varied intervals before isolation of PBMC. Specimen integrity was assessed by measures of yield, recovery, viability, and function of isolated lymphocytes. Several packaging systems were also evaluated during simulated shipping for the ability to maintain the internal temperature in adverse temperatures over time. Results Blood specimen containers experienced temperatures during shipment ranging from -1 to 35°C. Exposure to temperatures above room temperature (22°C) resulted in greater yields of PBMC. Reduced cell recovery following cryo-preservation as well as decreased viability and immune function were observed in specimens exposed to 15°C or 40°C for greater than 8 hours when compared to storage at 22°C. There was a trend toward improved preservation of blood specimen integrity stored at 30°C prior to processing for all time points tested. Internal temperatures of blood shipping containers were maintained longer in an acceptable range when warm packs were included. Conclusions Blood packages shipped overnight by commercial carrier may encounter extreme seasonal temperatures. Therefore, considerations in the design of shipping containers should include protecting against extreme ambient temperature deviations and maintaining specimen temperature above 22°C or preferably near 30°C.
Collapse
Affiliation(s)
- Walter C Olson
- Human Immune Therapy Center, University of Virginia, Charlottesville, VA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Naranbhai V, Bartman P, Ndlovu D, Ramkalawon P, Ndung'u T, Wilson D, Altfeld M, Carr WH. Impact of blood processing variations on natural killer cell frequency, activation, chemokine receptor expression and function. J Immunol Methods 2011; 366:28-35. [PMID: 21255578 DOI: 10.1016/j.jim.2011.01.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 01/03/2011] [Indexed: 12/11/2022]
Abstract
Understanding the role of natural killer (NK) cells in human disease pathogenesis is crucial and necessitates study of patient samples directly ex vivo. Manipulation of whole blood by density gradient centrifugation or delays in sample processing due to shipping, however, may lead to artifactual changes in immune response measures. Here, we assessed the impact of density gradient centrifugation and delayed processing of both whole blood and peripheral blood mononuclear cells (PBMC) at multiple timepoints (2-24 h) on flow cytometric measures of NK cell frequency, activation status, chemokine receptor expression, and effector functions. We found that density gradient centrifugation activated the NK cells and modified the chemokine receptor expression. Delays in processing beyond 8h activated NK cells in PBMC but not in whole blood. Likewise, processing delays decreased chemokine receptor (CCR4 and CCR7) expression in both PBMC and whole blood. Finally, delays in processing PBMC were associated with a decreased ability of NK cells to degranulate (as measured by CD107a expression) or secrete cytokines (IFN-γ and TNF-α). In summary, our findings suggest that density gradient centrifugation and delayed processing of PBMC can alter measures of clinically relevant NK cell characteristics including effector functions; and therefore should be taken into account in designing clinical research studies.
Collapse
Affiliation(s)
- Vivek Naranbhai
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu Natal, Durban, South Africa
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Effects of blood sample age at time of separation on measured cytokine concentrations in human plasma. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 18:318-26. [PMID: 21159926 DOI: 10.1128/cvi.00465-10] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Measurement of peripheral blood cytokines and other immunomodulatory proteins is a useful and popular tool for assessing human immune responses to a wide range of assaults. A common challenge in this work is obtaining fresh, high-quality samples and limiting the time between blood collection and the separation of plasma or serum from cells. In this study we sought to determine the effect of sample age at the time of processing on the measured levels of 41 soluble immune mediators. Two cohorts were examined: healthy lab donors and trauma patients, who have significant immune perturbation. Whole-blood samples were aliquoted, and plasma was isolated, at days 0, 1, 2, and 3 after collection. Multiplexing techniques were used to measure protein concentrations, and general estimating equations were used to determine if there was a significant change over time. Over the 3-day period examined, only 15 of the 41 proteins showed no significant change in either cohort. Among the remaining proteins both increases and decreases were observed, with changes ranging from 2.4% per day to 325% per day. Proteins with significant changes in one cohort did not always show significant changes in the other group. These results support the need to separate plasma or serum from whole blood as quickly as possible and/or to standardize the length of time to processing within a given study of peripheral blood protein concentrations. When this is not possible, care should be taken to account for differences due to sample age.
Collapse
|
47
|
Gill DK, Huang Y, Levine GL, Sambor A, Carter DK, Sato A, Kopycinski J, Hayes P, Hahn B, Birungi J, Tarragona-Fiol T, Wan H, Randles M, Cooper AR, Ssemaganda A, Clark L, Kaleebu P, Self SG, Koup R, Wood B, McElrath MJ, Cox JH, Hural J, Gilmour J. Equivalence of ELISpot assays demonstrated between major HIV network laboratories. PLoS One 2010; 5:e14330. [PMID: 21179404 PMCID: PMC3001861 DOI: 10.1371/journal.pone.0014330] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Accepted: 11/22/2010] [Indexed: 12/29/2022] Open
Abstract
Background The Comprehensive T Cell Vaccine Immune Monitoring Consortium (CTC-VIMC) was created to provide standardized immunogenicity monitoring services for HIV vaccine trials. The ex vivo interferon-gamma (IFN-γ) ELISpot is used extensively as a primary immunogenicity assay to assess T cell-based vaccine candidates in trials for infectious diseases and cancer. Two independent, GCLP-accredited central laboratories of CTC-VIMC routinely use their own standard operating procedures (SOPs) for ELISpot within two major networks of HIV vaccine trials. Studies are imperatively needed to assess the comparability of ELISpot measurements across laboratories to benefit optimal advancement of vaccine candidates. Methods We describe an equivalence study of the two independently qualified IFN-g ELISpot SOPs. The study design, data collection and subsequent analysis were managed by independent statisticians to avoid subjectivity. The equivalence of both response rates and positivity calls to a given stimulus was assessed based on pre-specified acceptance criteria derived from a separate pilot study. Findings Detection of positive responses was found to be equivalent between both laboratories. The 95% C.I. on the difference in response rates, for CMV (−1.5%, 1.5%) and CEF (−0.4%, 7.8%) responses, were both contained in the pre-specified equivalence margin of interval [−15%, 15%]. The lower bound of the 95% C.I. on the proportion of concordant positivity calls for CMV (97.2%) and CEF (89.5%) were both greater than the pre-specified margin of 70%. A third CTC-VIMC central laboratory already using one of the two SOPs also showed comparability when tested in a smaller sub-study. Interpretation The described study procedure provides a prototypical example for the comparison of bioanalytical methods in HIV vaccine and other disease fields. This study also provides valuable and unprecedented information for future vaccine candidate evaluations on the comparison and pooling of ELISpot results generated by the CTC-VIMC central core laboratories.
Collapse
Affiliation(s)
- Dilbinder K Gill
- International AIDS Vaccine Initiative Human Immunology Laboratory, Imperial College, London, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Paris RM, Kim JH, Robb ML, Michael NL. Prime-boost immunization with poxvirus or adenovirus vectors as a strategy to develop a protective vaccine for HIV-1. Expert Rev Vaccines 2010; 9:1055-69. [PMID: 20822348 DOI: 10.1586/erv.10.106] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Challenges in the development of an effective HIV-1 vaccine are myriad with significant hurdles posed by viral diversity, the lack of a human correlate of protection and difficulty in creating immunogens capable of eliciting broadly neutralizing antibodies. The implicit requirement for novel approaches to these problems has resulted in vaccine candidates designed to elicit cellular and/or humoral immune responses, to include recombinant DNA, viral and bacterial vectors, and subunit proteins. Here, we review data from clinical studies primarily of poxvirus and adenovirus vector vaccines, used in a heterologous prime-boost combination strategy. Currently, this strategy appears to hold the most promise for an effective vaccine based on results from immunogenicity testing and nonhuman primate challenge models, as well as the modest efficacy recently observed in the Thai prime-boost trial.
Collapse
Affiliation(s)
- Robert M Paris
- US Military HIV Research Program (MHRP), Department of Retrovirology, Armed Forces Research Institute of Medical Sciences, 315/6 Rajvithi Road, Bangkok, 10400, Thailand.
| | | | | | | |
Collapse
|
49
|
Mallone R, Mannering SI, Brooks-Worrell BM, Durinovic-Belló I, Cilio CM, Wong FS, Schloot NC. Isolation and preservation of peripheral blood mononuclear cells for analysis of islet antigen-reactive T cell responses: position statement of the T-Cell Workshop Committee of the Immunology of Diabetes Society. Clin Exp Immunol 2010; 163:33-49. [PMID: 20939860 DOI: 10.1111/j.1365-2249.2010.04272.x] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Autoimmune T cell responses directed against insulin-producing β cells are central to the pathogenesis of type 1 diabetes (T1D). Detection of such responses is therefore critical to provide novel biomarkers for T1D 'immune staging' and to understand the mechanisms underlying the disease. While different T cell assays are being developed for these purposes, it is important to optimize and standardize methods for processing human blood samples for these assays. To this end, we review data relevant to critical parameters in peripheral blood mononuclear cell (PBMC) isolation, (cryo)preservation, distribution and usage for detecting antigen-specific T cell responses. Based on these data, we propose recommendations on processing blood samples for T cell assays and identify gaps in knowledge that need to be addressed. These recommendations may be relevant not only for the analysis of T cell responses in autoimmune disease, but also in cancer and infectious disease, particularly in the context of clinical trials.
Collapse
Affiliation(s)
- R Mallone
- INSERM U986, DeAR Lab Avenir, Saint Vincent de Paul Hospital, 82 avenue Denfert Rochereau, Paris cedex 14, France.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Weinberg A, Song LY, Wilkening CL, Fenton T, Hural J, Louzao R, Ferrari G, Etter PE, Berrong M, Canniff JD, Carter D, Defawe OD, Garcia A, Garrelts TL, Gelman R, Lambrecht LK, Pahwa S, Pilakka-Kanthikeel S, Shugarts DL, Tustin NB. Optimization of storage and shipment of cryopreserved peripheral blood mononuclear cells from HIV-infected and uninfected individuals for ELISPOT assays. J Immunol Methods 2010; 363:42-50. [PMID: 20888337 DOI: 10.1016/j.jim.2010.09.032] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 09/10/2010] [Accepted: 09/23/2010] [Indexed: 11/16/2022]
Abstract
Functional immunologic assays using cryopreserved peripheral blood mononuclear cells (PBMC) are influenced by blood processing, storage and shipment. The objective of this study was to compare the viability, recovery and ELISPOT results of PBMC stored and shipped in liquid nitrogen (LN/LN) or stored in LN and shipped on dry ice (LN/DI) or stored at -70°C for 3 to 12 weeks and shipped on DI (70/DI 3 to 12); and to assess the effect of donor HIV infection status on the interaction between storage/shipment and the outcome measures. PBMC from 12 HIV-infected and 12 uninfected donors showed that LN/LN conferred higher viability and recovery than LN/DI or 70/DI 3, 6, 9 or 12. LN/DI PBMC had higher viability than any 70/DI PBMC. The PBMC viability and recovery linearly decreased with the duration of storage at -70°C from 3 to 12 weeks. This effect was more pronounced in samples from HIV-infected than uninfected donors. Results of ELISPOT assays using CMV pp65, CEF and Candida albicans antigens were qualitatively and quantitatively similar across LN/LN, LN/DI and 70/DI 3. However, ELISPOT values significantly decreased with the duration of storage at -70°C both in HIV-infected and uninfected donors. ELISPOT results also decreased with PBMC viability <70%.
Collapse
Affiliation(s)
- Adriana Weinberg
- Infectious Diseases, University of Colorado Denver, Aurora, CO, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|