1
|
Hansen SB, Højgaard LD, Kastrup J, Ekblond A, Follin B, Juhl M. Optimizing an immunomodulatory potency assay for Mesenchymal Stromal Cell. Front Immunol 2022; 13:1085312. [PMID: 36578497 PMCID: PMC9791065 DOI: 10.3389/fimmu.2022.1085312] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/25/2022] [Indexed: 12/14/2022] Open
Abstract
The expeditious progress of Mesenchymal Stromal Cells (MSC) for therapeutic intervention calls for means to compare differences in potency of cell products. The differences may be attributed to innumerable sources including tissue origin, production methods, or even between batches. While the immunomodulatory potential of MSC is recognized and well-documented by an expansive body of evidence, the methodologies and findings vary markedly. In this study, we utilized flowcytometric analysis of lymphocyte proliferation based on cryopreserved peripheral blood mononuclear cells for quantification of the inhibitory effect of MSC. Technical aspects of fluorescent staining and cryopreservation of peripheral blood mononuclear cells were evaluated to obtain optimal results and increase feasibility. A range of common specific and unspecific mitogens was titrated to identify the conditions, in which the effects of Adipose tissue-derived Stromal Cells (ASC; a type of MSC) were most pronounced. Specific stimulation by antibody-mediated activation of CD3 and CD28 via TransAct and Dynabeads lead to substantial proliferation of lymphocytes, which was inhibited by ASC. These results were closely mirrored when applying unspecific stimulation in form of phytohemagglutinin (PHA), but not concanavalin A or pokeweed mitogen. The mixed lymphocyte reaction is a common assay which exploits alloreactivity between donors. While arguably more physiologic, the output of the assay often varies substantially, and the extent of proliferation is limited since the frequency of alloreactive cells is low, as opposed to the mitogens. To heighten the proliferative response and robustness, combinations of 2-5 donors were tested. Maximum proliferation was observed when combining 4 or more donors, which was efficiently suppressed by ASC. Several desirable and unfavorable traits can be attributed to the tested stimuli in the form of keywords. The importance of these traits should be scored on a laboratory-level to identify the ideal mitogen. In our case the ranking listed PHA as the most suited candidate. Developing robust assays is no trivial feat. By disclosing the full methodological framework in the present study, we hope to aid others in establishing functional metrics on the road to potency assays.
Collapse
Affiliation(s)
- Stine Bangsgaard Hansen
- Cell2Cure, Cardiology Stem Cell Centre, The Heart Centre, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Lisbeth Drozd Højgaard
- Cell2Cure, Cardiology Stem Cell Centre, The Heart Centre, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Jens Kastrup
- Cell2Cure, Cardiology Stem Cell Centre, The Heart Centre, University Hospital Rigshospitalet, Copenhagen, Denmark
- Cell2Cure, Birkerød, Denmark
| | - Annette Ekblond
- Cell2Cure, Cardiology Stem Cell Centre, The Heart Centre, University Hospital Rigshospitalet, Copenhagen, Denmark
- Cell2Cure, Birkerød, Denmark
| | - Bjarke Follin
- Cell2Cure, Cardiology Stem Cell Centre, The Heart Centre, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Morten Juhl
- Cell2Cure, Cardiology Stem Cell Centre, The Heart Centre, University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
2
|
Rusconi G, Cusumano G, Mariotta L, Canevascini R, Gola M, Gornati R, Soldati G. Upgrading Monocytes Therapy for Critical Limb Ischemia Patient Treatment: Pre-Clinical and GMP-Validation Aspects. Int J Mol Sci 2022; 23:ijms232012669. [PMID: 36293525 PMCID: PMC9604444 DOI: 10.3390/ijms232012669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2022] Open
Abstract
Advanced cell therapy medicinal products (ATMP) are at the forefront of a new range of biopharmaceuticals. The use of ATMP has evolved and increased in the last decades, representing a new approach to treating diseases that are not effectively managed with conventional treatments. The standard worldwide recognized for drug production is the Good Manufacturing Practices (GMP), widely used in the pharma production of synthesized drugs but applying also to ATMP. GMP guidelines are worldwide recognized standards to manufacture medicinal products to guarantee high quality, safety, and efficacy. In this report, we describe the pre-clinical and the GMP upgrade of peripheral blood mononuclear cell (PBMC) preparation, starting from peripheral blood and ending up with a GMP-grade clinical product ready to be used in patients with critical limb ischemia (CLI). We also evaluated production in hypoxic conditions to increase PBMC functional activity and angiogenic potential. Furthermore, we extensively analyzed the storage and transport conditions of the final product as required by the regulatory body for ATMPs. Altogether, results suggest that the whole manufacturing process can be performed for clinical application. Peripheral blood collected by a physician should be transported at room temperature, and PBMCs should be isolated in a clean room within 8 h of venipuncture. Frozen cells can be stored in nitrogen vapors and thawed for up to 12 months. PBMCs resuspended in 5% human albumin solution should be stored and transported at 4 °C before injection in patients within 24 h to thawing. Hypoxic conditioning of PBMCs should be implemented for clinical application, as it showed a significant enhancement of PBMC functional activity, in particular with increased adhesion, migration, and oxidative stress resistance. We demonstrated the feasibility and the quality of a GMP-enriched suspension of monocytes as an ATMP, tested in a clean room facility for all aspects related to production in respect of all the GMP criteria that allow its use as an ATMP. We think that these results could ease the way to the clinical application of ATMPs.
Collapse
Affiliation(s)
| | | | - Luca Mariotta
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland
| | - Reto Canevascini
- Department of Surgery, Service of Angiology, Lugano Regional Hospital, 6900 Lugano, Switzerland
| | - Mauro Gola
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland
| | - Rosalba Gornati
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Gianni Soldati
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland
- Correspondence:
| |
Collapse
|
3
|
Cryopreservation of NK and T Cells Without DMSO for Adoptive Cell-Based Immunotherapy. BioDrugs 2021; 35:529-545. [PMID: 34427899 DOI: 10.1007/s40259-021-00494-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 10/20/2022]
Abstract
Dimethylsufoxide (DMSO) being universally used as a cryoprotectant in clinical adoptive cell-therapy settings to treat hematological malignancies and solid tumors is a growing concern, largely due to its broad toxicities. Its use has been associated with significant clinical side effects-cardiovascular, neurological, gastrointestinal, and allergic-in patients receiving infusions of cell-therapy products. DMSO has also been associated with altered expression of natural killer (NK) and T-cell markers and their in vivo function, not to mention difficulties in scaling up DMSO-based cryoprotectants, which introduce manufacturing challenges for autologous and allogeneic cellular therapies, including chimeric antigen receptor (CAR)-T and CAR-NK cell therapies. Interest in developing alternatives to DMSO has resulted in the evaluation of a variety of sugars, proteins, polymers, amino acids, and other small molecules and osmolytes as well as modalities to efficiently enable cellular uptake of these cryoprotectants. However, the DMSO-free cryopreservation of NK and T cells remains difficult. They represent heterogeneous cell populations that are sensitive to freezing and thawing. As a result, clinical use of cryopreserved cell-therapy products has not moved past the use of DMSO. Here, we present the state of the art in the development and use of cryopreservation options that do not contain DMSO toward clinical solutions to enable the global deployment of safer adoptively transferred cell-based therapies.
Collapse
|
4
|
van der Walle CF, Godbert S, Saito G, Azhari Z. Formulation Considerations for Autologous T Cell Drug Products. Pharmaceutics 2021; 13:pharmaceutics13081317. [PMID: 34452278 PMCID: PMC8400304 DOI: 10.3390/pharmaceutics13081317] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/29/2021] [Accepted: 08/18/2021] [Indexed: 11/16/2022] Open
Abstract
Genetically modified autologous T cells have become an established immunotherapy in the fight against cancer. The manufacture of chimeric antigen receptor (CAR) and αβ-T cell receptor (TCR) transduced T cells poses unique challenges, including the formulation, cryopreservation and fill-finish steps, which are the focus of this review. With an increasing number of marketing approvals for CAR-T cell therapies, comparison of their formulation design and presentation for administration can be made. These differences will be discussed alongside the emergence of automated formulation and fill-finish processes, the formulation design space, Monte Carlo simulation applied to risk analysis, primary container selection, freezing profiles and thaw and the use of dimethyl sulfoxide and alternative solvents/excipients as cryopreservation agents. The review will conclude with a discussion of the pharmaceutical solutions required to meet the simplification of manufacture and flexibility in dosage form for clinical treatment.
Collapse
|
5
|
Xu Y, Zou Q, Gao F, Wang D, Xue S, Lin H, Guo H, He X, Yang H, Gao D. Effect of Warming Process on the Survival of Cryopreserved Human Peripheral Blood Mononuclear Cells. Biopreserv Biobank 2021; 19:318-323. [PMID: 34061624 DOI: 10.1089/bio.2020.0058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
It is well known that the warming process is a critical step in cell cryopreservation, affecting the survival rate of the cryopreserved cells. However, there is a lack of understanding and optimization of the warming process for the cryopreserved human peripheral blood mononuclear cells (PBMCs) that are greatly needed for the cellular/immune therapies worldwide. In this study, the effect of the warming process on cryosurvival of the PBMCs was investigated, resulting in a recommendation of an optimal warming method. In the experiments, all PBMC samples were cooled by a fixed slow cooling process and stored in a liquid nitrogen tank. The frozen samples were then warmed in water baths with stirring at various temperatures, 37°C, 42°C, and 65°C, respectively. After thawing, PBMC's viability as well as phenotypic and functional analyses were performed and evaluated. It was shown that a relatively rapid warming process at 65°C in a water bath with stirring generated a significant improvement of cell viability, recovery, and functionality of the cryopreserved PBMCs. In addition, interferon-γ and interleukin-2 secretion were much higher in PBMCs thawed at 65°C than that in 42°C and 37°C, respectively. This study suggests that a rapid warming process at 65°C in a water bath should be used to replace the current conventional warming approach at 37°C.
Collapse
Affiliation(s)
- Yanhong Xu
- Department of Technology R&D, Origincell Technology Group, Shanghai, P.R. China.,Department of Engineering Technology Research Center of Cell Therapy and Clinical Translation. Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Qiongna Zou
- Department of Technology R&D, Origincell Technology Group, Shanghai, P.R. China
| | - Frankliu Gao
- Department of School of Public Affairs, University of Science and Technology of China, Hefei, P.R. China
| | - Daimeng Wang
- Department of Technology R&D, Origincell Technology Group, Shanghai, P.R. China
| | - Suxia Xue
- Department of Technology R&D, Origincell Technology Group, Shanghai, P.R. China
| | - Hebei Lin
- Department of Technology R&D, Origincell Technology Group, Shanghai, P.R. China
| | - Hao Guo
- Department of Technology R&D, Origincell Technology Group, Shanghai, P.R. China
| | - Xiaowen He
- Department of Technology R&D, Origincell Technology Group, Shanghai, P.R. China
| | - Huanfeng Yang
- Department of Technology R&D, Origincell Technology Group, Shanghai, P.R. China.,Department of School of Life Sciences and Technology, Tongji University, Shanghai, P.R. China
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, USA
| |
Collapse
|
6
|
Pasha R, Howell A, Turner TR, Halpenny M, Elmoazzen H, Acker JP, Pineault N. Transient warming affects potency of cryopreserved cord blood units. Cytotherapy 2020; 22:690-697. [PMID: 32591113 DOI: 10.1016/j.jcyt.2020.04.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/04/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND AIMS Cryopreserved cord blood units (CBUs) can be exposed to transient warming events (TWEs) during routine banking operations, which may affect their potency. NetCord-FACT guidelines recommend removal of these CBUs from inventory. The objective of this work was to evaluate warming kinetics of frozen CBUs in different settings to determine the optimal working environment and define the impact of different TWE scenarios on CB post-thaw quality and potency. METHODS The warming kinetics of frozen CBUs was influenced by both working surfaces and ambient working temperature, with cold plates providing better protection than vinyl or metal surfaces. Measurement of time for required operational activities revealed that CBUs are probably exposed to core temperatures greater than -150°C even when cold plates are used to reduce warming rates. RESULTS On the basis of the warming kinetics and observed operational activities, three TWE causing scenarios (control, typical, worst case) were investigated using a pool-and-split design and cell viability, recovery and potency (colony-forming unit [CFU]) assays were performed. TWEs were found to have little impact on the recovery of total nucleated cells or on the viability of CD34+ cells. In contrast, the viability and recovery of CD45+ cells in the smaller CBU compartments were reduced by TWEs. Moreover, the worst-case TWE reduced CFU recovery from CBUs, whereas the typical-scenario TWE had little effect. CONCLUSIONS Our results demonstrate that the distal segment underestimates the viability and potency of CBUs and that TWEs can affect the post-thaw viability and potency of CBUs. Although TWEs are almost inevitable during cord-blood banking operations, their effects must be diminished by reducing exposure time, using cold plates and strict operational protocols, to prevent worst-case TWEs.
Collapse
Affiliation(s)
- Roya Pasha
- Canadian Blood Services, Centre for Innovation, Ottawa, Ontario, Canada
| | - Anita Howell
- Canadian Blood Services, Centre for Innovation, Edmonton, Alberta, Canada
| | - Tracey R Turner
- Canadian Blood Services, Centre for Innovation, Edmonton, Alberta, Canada
| | - Mike Halpenny
- Canadian Blood Services, Cord Blood Bank and Stem Cell Manufacturing, Ottawa, Canada
| | - Heidi Elmoazzen
- Canadian Blood Services, Cord Blood Bank and Stem Cell Manufacturing, Ottawa, Canada
| | - Jason P Acker
- University of Alberta, Department of Laboratory Medicine and Pathology, Edmonton, Alberta, Canada
| | - Nicolas Pineault
- University of Ottawa, Biochemistry, Microbiology and Immunology Department, Ottawa, Canada.
| |
Collapse
|
7
|
Martikainen MV, Roponen M. Cryopreservation affected the levels of immune responses of PBMCs and antigen-presenting cells. Toxicol In Vitro 2020; 67:104918. [PMID: 32565220 DOI: 10.1016/j.tiv.2020.104918] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 10/24/2022]
Abstract
The effect of cryopreservation on antigen-presenting cells (APCs) is understudied. It is important to understand the effects of cryopreservation on these cells as they play a major role in immune responses, and they could be utilized in different clinical applications. In this study, we compared fresh and cryopreserved PBMCs in regards of their general immune responsiveness and, furthermore, the effect of cryopreservation on the circulating APCs among PBMCs. We stimulated fresh and cryopreserved PBMCs (N = 6) with LPS or Poly(I:C).Cytokine production of PBMCs and expression of functional markers CD80 and ILT4 on major types of APCs, dendritic cells (DCs) and monocytes, were analysed. We also analysed whether cryopreservation affects different subtypes of DCs (plasmacytoid and myeloid DCs) differently. Cryopreserved PBMCs produced less cytokines than fresh cells in response to stimulation, but the response profiles were comparable. Cryopreservation had also an effect on the relative proportions of APCs. Stimuli-induced responses were somewhat parallel but weaker than those observed in fresh cells. This study suggests that the use of cryopreserved cells is more suitable in studies that assess general responses to stimuli instead of measuring exact levels of reactions. Thus, the interpretation and comparison of the results of different studies should not be done without considering the differences in cryopreservation techniques and their effects on PBMCs and, more specifically, on APCs.
Collapse
Affiliation(s)
- Maria-Viola Martikainen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Marjut Roponen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
8
|
Hesler M, Kohl Y, Wagner S, von Briesen H. Non-pooled Human Platelet Lysate: A Potential Serum Alternative for In Vitro Cell Culture. Altern Lab Anim 2019; 47:116-127. [PMID: 31698922 DOI: 10.1177/0261192919882516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Serum supplementation is crucial in in vitro cell culture to provide all the essential nutrients needed for cellular processes. Fetal bovine serum (FBS) is considered the 'gold standard', but its production raises serious ethical concerns. Human-derived alternatives to FBS exist in the form of human platelet lysates (hPLs) or human AB serum (ABS). However, these serum products are usually pooled from several donors, in order to have a standardised product without patient-specific deviations. Nevertheless, the use of patient-specific serum in cell culture might be the key to successful transplantation of the cultured cells in medical applications, particularly as it avoids the transmission of infectious components or xenogenic proteins. In addition, the production of non-pooled hPL from single donors is likely to be a cost-effective and time-saving method. The current study used hPL units isolated from single donors and tested their performance as medium supplements for cell culture in comparison with FBS or ABS. This proof-of-concept study aimed to assess the potential of non-pooled hPL for personalised serum supplementation, and thus optimise in vitro models by making them more relevant to human physiology. We showed that A549, HepG2 and Caco-2 human cell lines were generally able to adapt to the new culture conditions and maintain viability, morphology and certain cell-specific characteristics. These results indicate that non-pooled, single patient-derived hPL could be a suitable alternative for in vitro serum supplementation.
Collapse
Affiliation(s)
- Michelle Hesler
- Department of Bioprocessing & Bioanalytics, Fraunhofer Institute for Biomedical Engineering, Sulzbach/Saar, Germany
| | - Yvonne Kohl
- Department of Bioprocessing & Bioanalytics, Fraunhofer Institute for Biomedical Engineering, Sulzbach/Saar, Germany
| | - Sylvia Wagner
- Department of Bioprocessing & Bioanalytics, Fraunhofer Institute for Biomedical Engineering, Sulzbach/Saar, Germany
| | - Hagen von Briesen
- Department of Bioprocessing & Bioanalytics, Fraunhofer Institute for Biomedical Engineering, Sulzbach/Saar, Germany
| |
Collapse
|
9
|
The Impact of Varying Cooling and Thawing Rates on the Quality of Cryopreserved Human Peripheral Blood T Cells. Sci Rep 2019; 9:3417. [PMID: 30833714 PMCID: PMC6399228 DOI: 10.1038/s41598-019-39957-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 02/06/2019] [Indexed: 12/11/2022] Open
Abstract
For the clinical delivery of immunotherapies it is anticipated that cells will be cryopreserved and shipped to the patient where they will be thawed and administered. An established view in cellular cryopreservation is that following freezing, cells must be warmed rapidly (≤5 minutes) in order to maintain high viability. In this study we examine the interaction between the rate of cooling and rate of warming on the viability, and function of T cells formulated in a conventional DMSO based cryoprotectant and processed in conventional cryovials. The data obtained show that provided the cooling rate is -1 °C min-1 or slower, there is effectively no impact of warming rate on viable cell number within the range of warming rates examined (1.6 °C min-1 to 113 °C min-1). It is only following a rapid rate of cooling (-10 °C min-1) that a reduction in viable cell number is observed following slow rates of warming (1.6 °C min-1 and 6.2 °C min-1), but not rapid rates of warming (113 °C min-1 and 45 °C min-1). Cryomicroscopy studies revealed that this loss of viability is correlated with changes in the ice crystal structure during warming. At high cooling rates (-10 °C min-1) the ice structure appeared highly amorphous, and when subsequently thawed at slow rates (6.2 °C min-1 and below) ice recrystallization was observed during thaw suggesting mechanical disruption of the frozen cells. This data provides a fascinating insight into the crystal structure dependent behaviour during phase change of frozen cell therapies and its effect on live cell suspensions. Furthermore, it provides an operating envelope for the cryopreservation of T cells as an emerging industry defines formulation volumes and cryocontainers for immunotherapy products.
Collapse
|
10
|
Consuegra I, Rodríguez-Aierbe C, Santiuste I, Bosch A, Martínez-Marín R, Fortuto MA, Díaz T, Martí S, Muñoz-Fernández MÁ. Isolation Methods of Peripheral Blood Mononuclear Cells in Spanish Biobanks: An Overview. Biopreserv Biobank 2017; 15:305-309. [DOI: 10.1089/bio.2016.0105] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Irene Consuegra
- Spanish HIV HGM BioBank, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | | | - Inés Santiuste
- Instituto de Investigación Marqués de Valdecilla, Biobanco Valdecilla, Santander, Spain
| | - Anna Bosch
- Biobanco Hospital Clínic–IDIBAPS, Institut d'Investigacions Biomèdiques August Pi i Sunyer, IDIBAPS, Barcelona, Spain
| | | | - M Antonia Fortuto
- Clínica Universidad de Navarra, Biobanco de la Universidad de Navarra, Pamplona, Spain
| | - Tatiana Díaz
- Hospital Regional Universitario Carlos Haya, Málaga, Spain
| | - Salvador Martí
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER BIOBANK, Valencia, Spain
| | - M Ángeles Muñoz-Fernández
- Spanish HIV HGM BioBank, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
11
|
Lauterboeck L, Saha D, Chatterjee A, Hofmann N, Glasmacher B. Xeno-Free Cryopreservation of Bone Marrow-Derived Multipotent Stromal Cells from Callithrix jacchus. Biopreserv Biobank 2016; 14:530-538. [DOI: 10.1089/bio.2016.0038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Lothar Lauterboeck
- Institute for Multiphase Processes, Leibniz Universität Hannover, Hannover, Germany
| | - Debapriya Saha
- Institute for Multiphase Processes, Leibniz Universität Hannover, Hannover, Germany
| | - Anamika Chatterjee
- Institute for Multiphase Processes, Leibniz Universität Hannover, Hannover, Germany
| | - Nicola Hofmann
- Institute for Multiphase Processes, Leibniz Universität Hannover, Hannover, Germany
| | - Birgit Glasmacher
- Institute for Multiphase Processes, Leibniz Universität Hannover, Hannover, Germany
| |
Collapse
|
12
|
Neubauer JC, Sébastien I, Germann A, Müller SC, Meyerhans A, von Briesen H, Zimmermann H. Towards standardized automated immunomonitoring: an automated ELISpot assay for safe and parallelized functionality analysis of immune cells. Cytotechnology 2016; 69:57-73. [PMID: 27896556 PMCID: PMC5264623 DOI: 10.1007/s10616-016-0037-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/26/2016] [Indexed: 01/06/2023] Open
Abstract
The ELISpot assay is used for the detection of T cell responses in clinical trials and vaccine evaluations. Standardization and reproducibility are necessary to compare the results worldwide, inter- and intra-assay variability being critical factors. To assure operator safety as well as high-quality experiment performance, the ELISpot assay was implemented on an automated liquid handling platform, a Tecan Freedom EVO. After validation of the liquid handling, automated loading of plates with cells and reagents was investigated. With step by step implementation of the manual procedure and liquid dispensing optimization on the robot platform, a fully automated ELISpot assay was accomplished with plates remaining in the system from the plate blocking step to spot development. The mean delta difference amounted to a maximum of 6%, and the mean dispersion was smaller than in the manual assay. Taken together, we achieved with this system not only a lower personnel attendance but also higher throughput and a more precise and parallelized analysis. This platform has the potential to guarantee validated, safe, fast, reproducible and cost-efficient immunological and toxicological assays in the future.
Collapse
Affiliation(s)
- J C Neubauer
- Department of Medical Biotechnology, Fraunhofer Institute for Biomedical Engineering, Sulzbach, Germany
| | - I Sébastien
- Department of Medical Biotechnology, Fraunhofer Institute for Biomedical Engineering, Sulzbach, Germany
| | - A Germann
- Department of Medical Biotechnology, Fraunhofer Institute for Biomedical Engineering, Sulzbach, Germany
| | - S C Müller
- Department of Medical Biotechnology, Fraunhofer Institute for Biomedical Engineering, Sulzbach, Germany
| | - A Meyerhans
- Infection Biology Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - H von Briesen
- Department of Medical Biotechnology, Fraunhofer Institute for Biomedical Engineering, Sulzbach, Germany
| | - H Zimmermann
- Department of Medical Biotechnology, Fraunhofer Institute for Biomedical Engineering, Sulzbach, Germany. .,Department of Molecular and Cellular Biotechnology, Saarland University, Saarbrücken, Germany.
| |
Collapse
|
13
|
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: 24] [Impact Index Per Article: 3.0] [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
|
14
|
Frese L, Dijkman PE, Hoerstrup SP. Adipose Tissue-Derived Stem Cells in Regenerative Medicine. Transfus Med Hemother 2016; 43:268-274. [PMID: 27721702 DOI: 10.1159/000448180] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/04/2016] [Indexed: 12/15/2022] Open
Abstract
In regenerative medicine, adult stem cells are the most promising cell types for cell-based therapies. As a new source for multipotent stem cells, human adipose tissue has been introduced. These so called adipose tissue-derived stem cells (ADSCs) are considered to be ideal for application in regenerative therapies. Their main advantage over mesenchymal stem cells derived from other sources, e.g. from bone marrow, is that they can be easily and repeatable harvested using minimally invasive techniques with low morbidity. ADSCs are multipotent and can differentiate into various cell types of the tri-germ lineages, including e.g. osteocytes, adipocytes, neural cells, vascular endothelial cells, cardiomyocytes, pancreatic β-cells, and hepatocytes. Interestingly, ADSCs are characterized by immunosuppressive properties and low immunogenicity. Their secretion of trophic factors enforces the therapeutic and regenerative outcome in a wide range of applications. Taken together, these particular attributes of ADSCs make them highly relevant for clinical applications. Consequently, the therapeutic potential of ADSCs is enormous. Therefore, this review will provide a brief overview of the possible therapeutic applications of ADSCs with regard to their differentiation potential into the tri-germ lineages. Moreover, the relevant advancements made in the field, regulatory aspects as well as other challenges and obstacles will be highlighted.
Collapse
Affiliation(s)
- Laura Frese
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Petra E Dijkman
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Simon P Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Wyss Translational Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
Hughes SM, Shu Z, Levy CN, Ferre AL, Hartig H, Fang C, Lentz G, Fialkow M, Kirby AC, Adams Waldorf KM, Veazey RS, Germann A, von Briesen H, McElrath MJ, Dezzutti CS, Sinclair E, Baker CAR, Shacklett BL, Gao D, Hladik F. Cryopreservation of Human Mucosal Leukocytes. PLoS One 2016; 11:e0156293. [PMID: 27232996 PMCID: PMC4883784 DOI: 10.1371/journal.pone.0156293] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/11/2016] [Indexed: 01/09/2023] Open
Abstract
Background Understanding how leukocytes in the cervicovaginal and colorectal mucosae respond to pathogens, and how medical interventions affect these responses, is important for developing better tools to prevent HIV and other sexually transmitted infections. An effective cryopreservation protocol for these cells following their isolation will make studying them more feasible. Methods and Findings To find an optimal cryopreservation protocol for mucosal mononuclear leukocytes, we compared cryopreservation media and procedures using human vaginal leukocytes and confirmed our results with endocervical and colorectal leukocytes. Specifically, we measured the recovery of viable vaginal T cells and macrophages after cryopreservation with different cryopreservation media and handling procedures. We found several cryopreservation media that led to recoveries above 75%. Limiting the number and volume of washes increased the fraction of cells recovered by 10–15%, possibly due to the small cell numbers in mucosal samples. We confirmed that our cryopreservation protocol also works well for both endocervical and colorectal leukocytes. Cryopreserved leukocytes had slightly increased cytokine responses to antigenic stimulation relative to the same cells tested fresh. Additionally, we tested whether it is better to cryopreserve endocervical cells on the cytobrush or in suspension. Conclusions Leukocytes from cervicovaginal and colorectal tissues can be cryopreserved with good recovery of functional, viable cells using several different cryopreservation media. The number and volume of washes has an experimentally meaningful effect on the percentage of cells recovered. We provide a detailed, step-by-step protocol with best practices for cryopreservation of mucosal leukocytes.
Collapse
Affiliation(s)
- Sean M. Hughes
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Zhiquan Shu
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, United States of America
- School of Mechanical and Materials Engineering, Washington State University, Everett, Washington, United States of America
| | - Claire N. Levy
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - April L. Ferre
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Heather Hartig
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Cifeng Fang
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, United States of America
| | - Gretchen Lentz
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Michael Fialkow
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Anna C. Kirby
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Kristina M. Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Ronald S. Veazey
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
| | - Anja Germann
- Division of Medical Biotechnology, Fraunhofer Institute for Biomedical Engineering IBMT, Sulzbach/Saar, Germany
| | - Hagen von Briesen
- Division of Medical Biotechnology, Fraunhofer Institute for Biomedical Engineering IBMT, Sulzbach/Saar, Germany
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Charlene S. Dezzutti
- School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
| | - Elizabeth Sinclair
- Division of Experimental Medicine, Department of Medicine, San Francisco General Hospital, University of California San Francisco, San Francisco, California, United States of America
| | - Chris A. R. Baker
- Division of Experimental Medicine, Department of Medicine, San Francisco General Hospital, University of California San Francisco, San Francisco, California, United States of America
- Core Immunology Lab, University of California San Francisco, San Francisco, California, United States of America
| | - Barbara L. Shacklett
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
- Department of Medicine, Division of Infectious Diseases, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, United States of America
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
- * E-mail: (DG); (FH)
| | - Florian Hladik
- Department of Obstetrics and Gynecology, 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
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail: (DG); (FH)
| |
Collapse
|
16
|
Daniele N, Campus M, Pellegrini C, Shkembi E, Zinno F. Biobanks and Clinical Research: An "Interesting" Connection. ACTA ACUST UNITED AC 2016. [DOI: 10.17352/acp.000005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
Abstract
This article first discusses some fundamentals of cryobiology and challenges for cell and tissue cryopreservation. Then, the results of cryopreservation of adipose cells and tissues, including adipose-derived stem cells, in the last decade are reviewed. In addition, from the viewpoint of cryobiology, some desired future work in fat cryopreservation is proposed that would benefit the optimization, standardization, and better application of such techniques.
Collapse
Affiliation(s)
- Zhiquan Shu
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Lee L Q Pu
- Division of Plastic Surgery, University of California Davis, 2221 Stockton Boulevard, Suite 2123, Sacramento, CA 95817, USA.
| |
Collapse
|
18
|
Vasson MP, Farges MC, Goncalves-Mendes N, Talvas J, Ribalta J, Winklhofer-Roob B, Rock E, Rossary A. Does aging affect the immune status? A comparative analysis in 300 healthy volunteers from France, Austria and Spain. IMMUNITY & AGEING 2013; 10:38. [PMID: 24010581 PMCID: PMC3848737 DOI: 10.1186/1742-4933-10-38] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 08/11/2013] [Indexed: 01/10/2023]
Abstract
Background As the European population is getting older, there is growing need in scientific data on how to achieve healthy and successful aging. A decline in immune function with age is unanimously supported by many epidemiological and clinical observations, with a decrease in T-cell mediated function encompassing a large part of this alteration. In the EU-funded VITAGE project, the effects of aging on biomarkers of immune status are being studied in three European countries. According to strict inclusion/exclusion criteria, a cohort of 300 healthy male non-smoking 20–75 years old volunteers were enrolled in France (n = 99), Spain (n = 100) and Austria (n = 101). In each country, the volunteers were classified as a function of age (one age group per decade). Biomarkers of immune status were determined including delayed-type hypersensitivity tests, measurement of lymphocyte surface markers, and serum determinations of interleukin-2, complement fractions and immunoglobulins. Results There were moderate differences in the biomarkers of immune status of the VITAGE study volunteers among the three European centres. The percentage of Natural Killer (NK) cells was 156% and 142% higher in Spain as compared to France and Austria, respectively (p < 0.0001), and this increase was observed at any age group above 30 years. Comparison between age-groups showed that in Spain, but not in France or Austria, older individuals had significantly a lower B lymphocyte distribution and conversely, a higher NK cell distribution. Moreover, the CD4/CD8 ratio was positively correlated with age in Austrian subjects (p < 0.0001). Conclusion Our results provide evidence of an increased NK cell distribution in the elderly, especially in the Spanish population. NK cell status may predict morbidity and mortality in the elderly, emphasizing the importance of innate as well as adaptive immunity in ensuring healthy longevity and cancer resistance, possibly in link with the Mediterranean diet.
Collapse
Affiliation(s)
- Marie-Paule Vasson
- Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, F-63000, Clermont-Ferrand, France.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
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: 5.2] [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
|
20
|
Herrmann M, Abdullah S, Alabi A, Alonso P, Friedrich AW, Fuhr G, Germann A, Kern WV, Kremsner PG, Mandomando I, Mellmann AC, Pluschke G, Rieg S, Ruffing U, Schaumburg F, Tanner M, Peters G, von Briesen H, von Eiff C, von Müller L, Grobusch MP. Staphylococcal disease in Africa: another neglected ‘tropical’ disease. Future Microbiol 2013; 8:17-26. [DOI: 10.2217/fmb.12.126] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The term ‘neglected tropical diseases’ predominantly refers to single-entity, mostly parasitic diseases. However, a considerable morbidity and mortality burden is carried by patients infected with Gram-positive cocci and Gram-negative bacilli that are prevalent all over the world, yet have impact in tropical and developing countries, particularly in children, with much higher incidence rates than those reported from developed countries. Staphylococcus aureus is among these pathogens. The African–German StaphNet consortium uses microbiological characterization of African S. aureus isolates, including identification of virulence factors, alongside the gathering of epidemiological and clinical data in an innovative research network between a European country (Germany) and several African partners. By creating an accessible strain repository and by implementing personnel training and capacity building, this network aims to put staphylococcal disease on the international agenda as a truly neglected condition with a major global impact on public health.
Collapse
Affiliation(s)
- Mathias Herrmann
- Institute of Medical Microbiology & Hygiene, University of Saarland Medical Center, Homburg, Germany
| | - Salim Abdullah
- Ifakara Health Research & Development Center, Bagamoyo, Tanzania
| | - Abraham Alabi
- Medical Research Unit, Albert Schweitzer Hospital, Lambaréné, Gabon
| | - Pedro Alonso
- Barcelona Centre for International Health Research (Hospital Clínic-Universitat de Barcelona), Barcelona, Spain
- Manhiça Health & Research Center, Maputo, Mozambique
| | - Alexander W Friedrich
- Department of Medical Microbiology, University Hospital Groningen, Groningen, The Netherlands
| | - Günther Fuhr
- Fraunhofer Institute for Biomedical Engineering, St Ingbert, Germany
| | - Anja Germann
- Fraunhofer Institute for Biomedical Engineering, St Ingbert, Germany
| | - Winfried V Kern
- Division of Infectious Diseases, Department of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter G Kremsner
- Medical Research Unit, Albert Schweitzer Hospital, Lambaréné, Gabon
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Inacio Mandomando
- Manhiça Health & Research Center, Maputo, Mozambique
- Instituto Nacional de Saúde, Ministério da Saúde, Maputo, Mozambique
| | | | - Gerd Pluschke
- Swiss Tropical & Public Health Institute & University of Basel, Switzerland
| | - Siegbert Rieg
- Division of Infectious Diseases, Department of Medicine, University of Freiburg, Freiburg, Germany
| | - Ulla Ruffing
- Institute of Medical Microbiology & Hygiene, University of Saarland Medical Center, Homburg, Germany
| | - Frieder Schaumburg
- Medical Research Unit, Albert Schweitzer Hospital, Lambaréné, Gabon
- Institute of Medical Microbiology, University of Münster, Münster, Germany
| | - Marcel Tanner
- Swiss Tropical & Public Health Institute & University of Basel, Switzerland
| | - Georg Peters
- Institute of Medical Microbiology, University of Münster, Münster, Germany
| | - Hagen von Briesen
- Fraunhofer Institute for Biomedical Engineering, St Ingbert, Germany
| | - Christof von Eiff
- Institute of Medical Microbiology, University of Münster, Münster, Germany
| | - Lutz von Müller
- Institute of Medical Microbiology & Hygiene, University of Saarland Medical Center, Homburg, Germany
| | - Martin P Grobusch
- Center for Tropical Medicine & Travel Medicine, Department of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|