1
|
Coombs RS, Overacre-Delgoffe AE, Bhattacharjee A, Hand TW, Boyle JP. Mouse innate resistance to Neospora caninum infection is driven by early production of IFNγ by NK cells in response to parasite ligands. mSphere 2024; 9:e0025524. [PMID: 39445806 PMCID: PMC11580461 DOI: 10.1128/msphere.00255-24] [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: 03/28/2024] [Accepted: 05/03/2024] [Indexed: 10/25/2024] Open
Abstract
Toxoplasma gondii is capable of being transmitted by nearly all warm-blooded animals, and rodents are a major source of parasite dissemination, yet mechanisms driving its broad host range are poorly understood. Although a phylogenetically close relative of T. gondii, Neospora caninum differs from T. gondii in that it does not infect mice and only infects a small number of ruminant and canine species. We recently showed that T. gondii and N. caninum grow similarly in mice during the first 24 h post-infection, but only N. caninum induces an IFNγ-driven response within hours that controls the infection. The goal of the present study was to understand the cellular basis of this rapid response to N. caninum. To do this, we compared immune cell populations at the site of infection 4 h after T. gondii or N. caninum infection in mice. We found that both parasites induced similar frequencies of peritoneal monocytes, while macrophages and dendritic cell populations were not increased compared to uninfected mice. Through a series of knockout mouse experiments, we show that B, T, and NKT cells are not required for immediate IFNγ production and ultimate control of N. caninum infection, suggesting that natural killer (NK) cells are the primary inducers of immediate IFNγ in response to N. caninum. N. caninum infections exhibited significantly more IFNγ+ NK cells in the peritoneum compared with T. gondii-infected and uninfected mice. Finally, we demonstrate that differences in early IFNγ production during N. caninum and T. gondii infections in mice are at least partly due to differences in soluble antigen(s) produced by tachyzoites. IMPORTANCE Pathogen differences in host range are poorly understood at the molecular level even though even closely related pathogen species can have dramatically distinct host ranges. Here, we study two related parasite species that have a dramatic difference in their ability to infect mice. Here, we show that soluble proteins from these species determine one driver of this difference: induction of interferon gamma by cells of the innate immune system.
Collapse
Affiliation(s)
- R. S. Coombs
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - A. Bhattacharjee
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - T. W. Hand
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - J. P. Boyle
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
2
|
Radziszewska A, Peckham H, de Gruijter NM, Restuadi R, Wu WH, Jury EC, Rosser EC, Ciurtin C. Active juvenile systemic lupus erythematosus is associated with distinct NK cell transcriptional and phenotypic alterations. Sci Rep 2024; 14:13074. [PMID: 38844784 PMCID: PMC11156641 DOI: 10.1038/s41598-024-62325-3] [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: 07/07/2023] [Accepted: 05/15/2024] [Indexed: 06/09/2024] Open
Abstract
While adaptive immune responses have been studied extensively in SLE (systemic lupus erythematosus), there is limited and contradictory evidence regarding the contribution of natural killer (NK) cells to disease pathogenesis. There is even less evidence about the role of NK cells in the more severe phenotype with juvenile-onset (J)SLE. In this study, analysis of the phenotype and function of NK cells in a large cohort of JSLE patients demonstrated that total NK cells, as well as perforin and granzyme A expressing NK cell populations, were significantly diminished in JSLE patients compared to age- and sex-matched healthy controls. The reduction in NK cell frequency was associated with increased disease activity, and transcriptomic analysis of NK populations from active and low disease activity JSLE patients versus healthy controls confirmed that disease activity was the main driver of differential NK cell gene expression. Pathway analysis of differentially expressed genes revealed an upregulation of interferon-α responses and a downregulation of exocytosis in active disease compared to healthy controls. Further gene set enrichment analysis also demonstrated an overrepresentation of the apoptosis pathway in active disease. This points to increased propensity for apoptosis as a potential factor contributing to NK cell deficiency in JSLE.
Collapse
Affiliation(s)
- Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK.
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK.
| | - Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Nina M de Gruijter
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Restuadi Restuadi
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK
| | - Wing Han Wu
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK
- NHS North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Elizabeth C Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Elizabeth C Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK.
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK.
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK.
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK.
| |
Collapse
|
3
|
Bennett-Boehm MM, Mahr AR, Hartwell ST, Regan AK, Weber IS, Blackmon A, Bisson CR, Truong AN, Circo BA, Nienhueser J, Rogers DR, Booher N, Rajagopalan N, Martens JW, Denton PW. Development and implementation of natural killer cell simultaneous ADCC and direct killing assay. Heliyon 2023; 9:e22991. [PMID: 38125417 PMCID: PMC10731071 DOI: 10.1016/j.heliyon.2023.e22991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/09/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Assays to quantify natural killer (NK) cell killing efficacy have traditionally focused on assessing either direct killing or antibody dependent cell-mediated cytotoxicity (ADCC) independently. Due to the probability that immunotherapeutic interventions affect NK cell-mediated direct killing and NK cell-mediated ADCC differently, we developed an assay with the capacity to measure NK cell-mediated direct killing and ADCC simultaneously with cells from the same human donor. Specifically, this design allows for a single NK cell population to be split into several experimental conditions (e.g., direct killing, ADCC), thus controlling for potential confounders associated with human-to-human variation when assessing immunotherapy impacts. Our Natural Killer cell Simultaneous ADCC and Direct Killing Assay (NK-SADKA) allows researchers to reproducibly quantify both direct killing and ADCC by human NK cells. Furthermore, this optimized experimental design allows for concurrent analysis of the NK cells via flow cytometric immunophenotyping of NK cell populations which will facilitate the identification of relationships between NK cell phenotype and the subsequent killing potential. This assay will be valuable for assessing the broader impact(s) of immunotherapy strategies on both modes of NK cell killing.
Collapse
Affiliation(s)
- Maia M.C. Bennett-Boehm
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
- Department of Information Science and Technology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Anna R. Mahr
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Sean T. Hartwell
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Alexander K. Regan
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Isabelle S. Weber
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Arriana Blackmon
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Cami R. Bisson
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Angela N. Truong
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Bella A. Circo
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Jaden Nienhueser
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Donald R. Rogers
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Nathan Booher
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Nithya Rajagopalan
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Jacob W.S. Martens
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Paul W. Denton
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| |
Collapse
|
4
|
Motais B, Charvátová S, Walek Z, Hájek R, Bagó JR. NK92 Expressing Anti-BCMA CAR and Secreted TRAIL for the Treatment of Multiple Myeloma: Preliminary In Vitro Assessment. Cells 2023; 12:2748. [PMID: 38067177 PMCID: PMC10706019 DOI: 10.3390/cells12232748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/03/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Multiple myeloma (MM) has witnessed improved patient outcomes through advancements in therapeutic approaches. Notably, allogeneic stem cell transplantation, proteasome inhibitors, immunomodulatory drugs, and monoclonal antibodies have contributed to enhanced quality of life. Recently, a promising avenue has emerged with chimeric antigen receptor (CAR) T cells targeting B-cell maturation antigen (BCMA), expressed widely on MM cells. To mitigate risks associated with allogenic T cells, we investigated the potential of BCMA CAR expression in natural killer cells (NKs), known for potent cytotoxicity and minimal side effects. Using the NK-92 cell line, we co-expressed BCMA CAR and soluble tumor necrosis factor-related apoptosis-inducing ligand (sTRAIL) employing the piggyBac transposon system. Engineered NK cells (CAR-NK-92-TRAIL) demonstrated robust cytotoxicity against a panel of MM cell lines and primary patient samples, outperforming unmodified NK-92 cells with a mean difference in viability of 45.1% (±26.1%, depending on the target cell line). Combination therapy was explored with the proteasome inhibitor bortezomib (BZ) and γ-secretase inhibitors (GSIs), leading to a significant synergistic effect in combination with CAR-NK-92-TRAIL cells. This synergy was evident in cytotoxicity assays where a notable decrease in MM cell viability was observed in combinatorial therapy compared to single treatment. In summary, our study demonstrates the therapeutic potential of the CAR-NK-92-TRAIL cells for the treatment of MM. The synergistic impact of combining these engineered NK cells with BZ and GSI supports further development of allogeneic CAR-based products for effective MM therapy.
Collapse
Affiliation(s)
- Benjamin Motais
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (Z.W.); (R.H.)
- Faculty of Science, University of Ostrava, 701 00 Ostrava, Czech Republic
| | - Sandra Charvátová
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (Z.W.); (R.H.)
- Faculty of Science, University of Ostrava, 701 00 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 00 Ostrava, Czech Republic
| | - Zuzana Walek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (Z.W.); (R.H.)
| | - Roman Hájek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (Z.W.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 00 Ostrava, Czech Republic
| | - Juli R. Bagó
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (Z.W.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 00 Ostrava, Czech Republic
| |
Collapse
|
5
|
Charvátová S, Motais B, Czapla J, Cichoń T, Smolarczyk R, Walek Z, Giebel S, Hájek R, Bagó JR. Novel Local "Off-the-Shelf" Immunotherapy for the Treatment of Myeloma Bone Disease. Cells 2023; 12:cells12030448. [PMID: 36766789 PMCID: PMC9914109 DOI: 10.3390/cells12030448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/13/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Myeloma bone disease (MBD) is one of the major complications in multiple myeloma (MM)-the second most frequent hematologic malignancy. It is characterized by the formation of bone lesions due to the local action of proliferating MM cells, and to date, no effective therapy has been developed. In this study, we propose a novel approach for the local treatment of MBD with a combination of natural killer cells (NKs) and mesenchymal stem cells (MSCs) within a fibrin scaffold, altogether known as FINM. The unique biological properties of the NKs and MSCs, joined to the injectable biocompatible fibrin, permitted to obtain an efficient "off-the-shelf" ready-to-use composite for the local treatment of MBD. Our in vitro analyses demonstrate that NKs within FINM exert a robust anti-tumor activity against MM cell lines and primary cells, with the capacity to suppress osteoclast activity (~60%) within in vitro 3D model of MBD. Furthermore, NKs' post-thawing cytotoxic activity is significantly enhanced (~75%) in the presence of MSCs, which circumvents the decrease of NKs cytotoxicity after thawing, a well-known issue in the cryopreservation of NKs. To reduce the tumor escape, we combined FINM with other therapeutic agents (bortezomib (BZ), and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)), observing a clear therapeutic synergistic effect in vitro. Finally, the therapeutic efficacy of FINM in combination with BZ and TRAIL was assessed in a mouse model of MM, achieving 16-fold smaller tumors compared to the control group without treatment. These results suggest the potential of FINM to serve as an allogeneic "off-the-shelf" approach to improve the outcomes of patients suffering from MBD.
Collapse
Affiliation(s)
- Sandra Charvátová
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Benjamin Motais
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Justyna Czapla
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44102 Gliwice, Poland
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44102 Gliwice, Poland
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44102 Gliwice, Poland
| | - Zuzana Walek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Sebastian Giebel
- Department of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44102 Gliwice, Poland
| | - Roman Hájek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Juli R. Bagó
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
- Correspondence: ; Tel.: +42-(05)-97372092
| |
Collapse
|
6
|
Newton HS, Dobrovolskaia MA. Immunophenotyping: Analytical approaches and role in preclinical development of nanomedicines. Adv Drug Deliv Rev 2022; 185:114281. [PMID: 35405297 PMCID: PMC9164149 DOI: 10.1016/j.addr.2022.114281] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/18/2022] [Accepted: 04/05/2022] [Indexed: 12/17/2022]
Abstract
Pharmaceutical products can activate immune cells, suppress their function, or change the immune responses to traditional immunologically active agonists such as those present in microbes. Therefore, the assessment of immunostimulation, immunosuppression, and immunomodulation comprises the backbone of immunotoxicity studies of new drug entities. Depending on physicochemical properties (e.g., size, charge, surface functionalities, hydrophobicity), nanoparticles can be immunostimulatory, immunosuppressive, and immunomodulatory. Various methods and experimental frameworks have been established to support preclinical translational studies of nanotechnology-based drug products. Immunophenotyping after the exposure of cells or preclinical animal models to nanoparticles can provide critical information about the changes in both the numbers of immune cells and their activation status. However, this methodology is underutilized in preclinical studies of engineered nanomaterials. Herein, we review current literature about varieties of instrumentation and methods utilized for immunophenotyping, discuss their advantages and limitations, and propose a roadmap for applying immunophenotyping to support preclinical immunological characterization of nanotechnology-based formulations.
Collapse
Affiliation(s)
- Hannah S Newton
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick MD, USA
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research, Frederick MD, USA.
| |
Collapse
|
7
|
Johnson CDL, Zale NE, Frary ED, Lomakin JA. Feeder-Cell-Free and Serum-Free Expansion of Natural Killer Cells Using Cloudz Microspheres, G-Rex6M, and Human Platelet Lysate. Front Immunol 2022; 13:803380. [PMID: 35320938 PMCID: PMC8934851 DOI: 10.3389/fimmu.2022.803380] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/26/2022] [Indexed: 11/18/2022] Open
Abstract
The versatility of natural killer cells has ignited growing interest in their therapeutic use for cancer and other immunotherapy treatments. However, NK cells compose a small portion of peripheral blood mononuclear cells (5%–20% of PBMCs) and clinical doses require billions of cells. Manufacturing suitable doses of NK cells remains a major challenge for NK immunotherapy. The current standard for expanding NK cells relies on feeder cells and fetal bovine serum to achieve large expansion, but both encounter regulatory concerns. We developed NK Cloudz, a dissolvable polymer-based microsphere platform, as an alternative to a feeder cell approach to expand NK cells. We demonstrated that a combination of NK Cloudz, a G-Rex6M culture vessel, and GMP Human Platelet Lysate expanded NK cells 387 ± 100-fold in 10 days from a PBMC starting population. The NK purity, viability, and cytotoxicity were similar to both a feeder cell protocol and an FBS-based protocol. Additionally, we found no significant differences between FBS and GMP Human Platelet Lysate and concluded that platelet lysate is a good xeno-free alternative to FBS for NK expansion. Overall, we demonstrated a feeder-cell-free and FBS-free protocol that leverages NK Cloudz as a promising step toward a commercial GMP manufacturing method to expand NK cells for therapeutic use.
Collapse
|
8
|
Pro- and Anti-Inflammatory Cytokines in the Context of NK Cell-Trophoblast Interactions. Int J Mol Sci 2022; 23:ijms23042387. [PMID: 35216502 PMCID: PMC8878424 DOI: 10.3390/ijms23042387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 02/04/2023] Open
Abstract
During pregnancy, uterine NK cells interact with trophoblast cells. In addition to contact interactions, uterine NK cells are influenced by cytokines, which are secreted by the cells of the decidua microenvironment. Cytokines can affect the phenotypic characteristics of NK cells and change their functional activity. An imbalance of pro- and anti-inflammatory signals can lead to the development of reproductive pathology. The aim of this study was to assess the effects of cytokines on NK cells in the presence of trophoblast cells in an in vitro model. We used TNFα, IFNγ, TGFβ and IL-10; the NK-92 cell line; and peripheral blood NK cells (pNKs) from healthy, non-pregnant women. For trophoblast cells, the JEG-3 cell line was used. In the monoculture of NK-92 cells, TNFα caused a decrease in CD56 expression. In the coculture of NK cells with JEG-3 cells, TNFα increased the expression of NKG2C and NKG2A by NK-92 cells. Under the influence of TGFβ, the expression of CD56 increased and the expression of NKp30 decreased in the monoculture. After the preliminary cultivation of NK-92 cells in the presence of TGFβ, their cytotoxicity increased. In the case of adding TGFβ to the PBMC culture, as well as coculturing PBMCs and JEG-3 cells, the expression of CD56 and NKp44 by pNK cells was reduced. The differences in the effects of TGFβ in the model using NK-92 cells and pNK cells may be associated with the possible influence of monocytes or other lymphoid cells from the mononuclear fraction.
Collapse
|
9
|
Floerchinger A, Engeland CE. NK Cell Effector Functions and Bystander Tumor Cell Killing in Immunovirotherapy. Methods Mol Biol 2022; 2521:233-248. [PMID: 35733001 DOI: 10.1007/978-1-0716-2441-8_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Oncolytic virotherapy is a compelling strategy to combine cancer gene therapy with immunotherapy. Lytic virus replication in malignant cells not only enables localized transgene expression based on engineered vectors but also triggers immunogenic tumor cell death and elicits inflammation in the tumor microenvironment. Modified oncolytic viruses encoding immunomodulators have been developed to enhance antitumor immune effects and therapeutic efficacy. As one example, bispecific molecules that engage immune cells to exert antitumor cytotoxicity can be encoded within the viral vector. This chapter describes an in vitro coculture experiment to study functionality and antitumor efficacy of engineered measles vaccine strain virus encoding natural killer cell engagers. In a flow cytometry-based analysis, target cell death of noninfected bystander cancer cells and effector functions of primary human natural killer cells are investigated. This methodology can facilitate assessment of advanced oncolytic viral vectors for cancer immunotherapy.
Collapse
Affiliation(s)
- Alessia Floerchinger
- Center for Biomedical Research and Education (ZBAF), School of Medicine, Institute of Virology and Microbiology, Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Christine E Engeland
- Center for Biomedical Research and Education (ZBAF), School of Medicine, Institute of Virology and Microbiology, Faculty of Health, Witten/Herdecke University, Witten, Germany.
- Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT) and Department of Medical Oncology, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
10
|
Choi H, Ho M, Adeniji OS, Giron L, Bordoloi D, Kulkarni AJ, Puchalt AP, Abdel-Mohsen M, Muthumani K. Development of Siglec-9 Blocking Antibody to Enhance Anti-Tumor Immunity. Front Oncol 2021; 11:778989. [PMID: 34869028 PMCID: PMC8640189 DOI: 10.3389/fonc.2021.778989] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Sialic acid-binding Immunoglobulin-like lectin-9 (Siglec-9) is a glyco-immune negative checkpoint expressed on several immune cells. Siglec-9 exerts its inhibitory effects by binding to sialoglycan ligands expressed on cancer cells, enabling them to evade immunosurveillance. We developed a panel of human anti-Siglec-9 hybridoma clones by immunizing mice with Siglec-9-encoding DNA and Siglec-9 protein. The lead antibodies, with high specificity and functionality against Siglec-9, were identified through screening of clones. The in vitro cytotoxicity assays showed that our lead antibody enhances anti-tumor immune activity. Further, in vivo testing utilizing ovarian cancer humanized mouse model showed a drastic reduction in tumor volume. Together, we developed novel antibodies that augment anti-tumor immunity through interference with Siglec-9-mediated immunosuppression.
Collapse
Affiliation(s)
- Hyeree Choi
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Michelle Ho
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Opeyemi S Adeniji
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Leila Giron
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Devivasha Bordoloi
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Abhijeet J Kulkarni
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | | | - Mohamed Abdel-Mohsen
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Kar Muthumani
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| |
Collapse
|
11
|
Toffoli EC, Sheikhi A, Lameris R, King LA, van Vliet A, Walcheck B, Verheul HMW, Spanholtz J, Tuynman J, de Gruijl TD, van der Vliet HJ. Enhancement of NK Cell Antitumor Effector Functions Using a Bispecific Single Domain Antibody Targeting CD16 and the Epidermal Growth Factor Receptor. Cancers (Basel) 2021; 13:cancers13215446. [PMID: 34771609 PMCID: PMC8582566 DOI: 10.3390/cancers13215446] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Strategies to enhance the preferential accumulation and activation of Natural Killer (NK) cells in the tumor microenvironment can be expected to increase the efficacy of NK cell-based cancer immunotherapy. In this study, we report that a bispecific single domain antibody (VHH) that targets CD16 (FcRγIII) on NK cells and the epidermal growth factor receptor (EGFR) on tumor cells can be used to target and enhance cytolysis of cancer cells. The bispecific VHH enhanced NK cell activation and cytotoxicity in an EGFR- and CD16-dependent and KRAS-independent manner. Moreover, the bispecific VHH induced stronger activity of cancer patient-derived NK cells and resulted in tumor control in a co-culture of metastatic colorectal cancer cells and either autologous peripheral blood mononuclear cells or allogeneic CD16+ NK cells. We believe that this novel approach could represent a valid therapeutic strategy either alone or in combination with other NK cell-based therapies. Abstract The ability to kill tumor cells while maintaining an acceptable safety profile makes Natural Killer (NK) cells promising assets for cancer therapy. Strategies to enhance the preferential accumulation and activation of NK cells in the tumor microenvironment can be expected to increase the efficacy of NK cell-based therapies. In this study, we show binding of a novel bispecific single domain antibody (VHH) to both CD16 (FcRγIII) on NK cells and the epidermal growth factor receptor (EGFR) on tumor cells of epithelial origin. The bispecific VHH triggered CD16- and EGFR-dependent activation of NK cells and subsequent lysis of tumor cells, regardless of the KRAS mutational status of the tumor. Enhancement of NK cell activation by the bispecific VHH was also observed when NK cells of colorectal cancer (CRC) patients were co-cultured with EGFR expressing tumor cells. Finally, higher levels of cytotoxicity were found against patient-derived metastatic CRC cells in the presence of the bispecific VHH and autologous peripheral blood mononuclear cells or allogeneic CD16 expressing NK cells. The anticancer activity of CD16-EGFR bispecific VHHs reported here merits further exploration to assess its potential therapeutic activity either alone or in combination with adoptive NK cell-based therapeutic approaches.
Collapse
Affiliation(s)
- Elisa C. Toffoli
- Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (E.C.T.); (A.S.); (R.L.); (L.A.K.); (T.D.d.G.)
| | - Abdolkarim Sheikhi
- Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (E.C.T.); (A.S.); (R.L.); (L.A.K.); (T.D.d.G.)
- School of Medicine, Dezful University of Medical Sciences, Department of Immunology, Dezful 64616-43993, Iran
| | - Roeland Lameris
- Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (E.C.T.); (A.S.); (R.L.); (L.A.K.); (T.D.d.G.)
| | - Lisa A. King
- Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (E.C.T.); (A.S.); (R.L.); (L.A.K.); (T.D.d.G.)
| | - Amanda van Vliet
- Glycostem Therapeutics, 5349 AB Oss, The Netherlands; (A.v.V.); (J.S.)
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN 55108, USA;
| | - Henk M. W. Verheul
- Radboud Institute for Health Sciences, Department of Medical Oncology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Jan Spanholtz
- Glycostem Therapeutics, 5349 AB Oss, The Netherlands; (A.v.V.); (J.S.)
| | - Jurriaan Tuynman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, 1081 HV Amsterdam, The Netherlands;
| | - Tanja D. de Gruijl
- Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (E.C.T.); (A.S.); (R.L.); (L.A.K.); (T.D.d.G.)
| | - Hans J. van der Vliet
- Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (E.C.T.); (A.S.); (R.L.); (L.A.K.); (T.D.d.G.)
- Lava Therapeutics, 3584 CM Utrecht, The Netherlands
- Correspondence:
| |
Collapse
|
12
|
Using Immunohistochemistry Without Linkers to Determine the Optimum Concentrations of Primary Antibodies for Immunofluorescence Staining of Formalin-fixed Paraffin-embedded Tissue Sections. Appl Immunohistochem Mol Morphol 2021; 28:249-257. [PMID: 32197004 DOI: 10.1097/pai.0000000000000718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The use of immunofluorescence (IF) technique to detect and evaluate expression levels and localization of cellular proteins and other antigens of interest through the antibodies in their cellular or tissue context has become a standard approach among researchers. Optimizing primary antibody concentrations/dilutions is an essential step in the fluorescent antibody staining protocol. The steps in IF staining are similar to those of the immunohistochemistry (IHC) technique. The use of IHC technique to determine the optimal working dilutions of primary antibodies for IF staining of formalin-fixed paraffin-embedded (FFPE) tissues sections can minimize time wasting and cumbersome approach of using direct IF single labeling using variable dilutions of both primary and secondary antibodies. We used IHC staining technique to determine the working dilutions of the respective primary antibodies by staining 3-µm sections of recommended positive FFPE tissue sections using 3 different dilutions of the primary antibodies and an isotype control (used at the highest concentration). Digital images of sections stained were reviewed in ImageScope by a Consultant Pathologist for positivity, intensity, and histologic distribution. We adopted the IHC predetermined optimal dilutions of primary antibodies to CD4, CD8, CD16, CD21, CD56, CD68, CD163, FOXP3, and PD1 to carry out IF staining of FFPE tissue sections. This approach has helped to remove the complexities associated with grappling with 2 unknown to optimize for both the primary and secondary antibodies using IF technique.
Collapse
|
13
|
Selection, Expansion, and Unique Pretreatment of Allogeneic Human Natural Killer Cells with Anti-CD38 Monoclonal Antibody for Efficient Multiple Myeloma Treatment. Cells 2021; 10:cells10050967. [PMID: 33919155 PMCID: PMC8143171 DOI: 10.3390/cells10050967] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023] Open
Abstract
Cellular immunotherapy is becoming a new pillar in cancer treatment after recent striking results in different clinical trials with chimeric antigen receptor T cells. However, this innovative therapy is not exempt from challenges such as off-tumor toxicity, tumor recurrence in heterogeneous tumors, and affordability. To surpass these limitations, we exploit the unique anti-tumor characteristics of natural killer (NK) cells. In this study, we aimed to obtain a clinically relevant number of allogeneic NK cells derived from peripheral blood (median of 14,050 million cells from a single donor) to target a broad spectrum of solid and liquid tumor types. To boost their anti-tumor activity, we combined allogeneic NK cells with the approved anti-cluster of differentiation 38 (CD-38) monoclonal antibody Daratumumab to obtain a synergistic therapeutic effect against incurable multiple myeloma. The combination therapy was refined with CD16 polymorphism donor selection and uncomplicated novel in vitro pretreatment to avoid undesired fratricide, increasing the in vitro therapeutic effect against the CD-38 positive multiple myeloma cell line by more than 20%. Time-lapse imaging of mice with established human multiple myeloma xenografts revealed that combination therapy of selected and pretreated NK cells with Daratumumab presented tumor volumes 43-fold smaller than control ones. Combination therapy with an allogeneic source of fully functional NK cells could be beneficial in future clinical settings to circumvent monoclonal antibodies' low therapeutic efficiency due to NK cell dysfunctionality in MM patients.
Collapse
|
14
|
Dumitrascu B, Villar S, Mixon DG, Engelhardt BE. Optimal marker gene selection for cell type discrimination in single cell analyses. Nat Commun 2021; 12:1186. [PMID: 33608535 PMCID: PMC7895823 DOI: 10.1038/s41467-021-21453-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 01/27/2021] [Indexed: 11/17/2022] Open
Abstract
Single-cell technologies characterize complex cell populations across multiple data modalities at unprecedented scale and resolution. Multi-omic data for single cell gene expression, in situ hybridization, or single cell chromatin states are increasingly available across diverse tissue types. When isolating specific cell types from a sample of disassociated cells or performing in situ sequencing in collections of heterogeneous cells, one challenging task is to select a small set of informative markers that robustly enable the identification and discrimination of specific cell types or cell states as precisely as possible. Given single cell RNA-seq data and a set of cellular labels to discriminate, scGeneFit selects gene markers that jointly optimize cell label recovery using label-aware compressive classification methods. This results in a substantially more robust and less redundant set of markers than existing methods, most of which identify markers that separate each cell label from the rest. When applied to a data set given a hierarchy of cell types as labels, the markers found by our method improves the recovery of the cell type hierarchy with fewer markers than existing methods using a computationally efficient and principled optimization. The selection of a small set of cellular labels to distinguish a subpopulation of cells from a complex mixture is an important task in cell biology. Here the authors propose a method for supervised genetic marker selection using linear programming and provides a Python package scGeneFit that implements this approach.
Collapse
Affiliation(s)
- Bianca Dumitrascu
- Department of Computer Science and Technology, University of Cambridge, Cambridge, UK
| | - Soledad Villar
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA.,Mathematical Institute for Data Science, Johns Hopkins University, Baltimore, MD, USA
| | - Dustin G Mixon
- Department of Mathematics, The Ohio State University, Columbus, OH, USA
| | - Barbara E Engelhardt
- Department of Computer Science, Princeton University, Princeton, NJ, USA. .,Center for Statistics and Machine Learning, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
15
|
Gómez-Mora E, Carrillo J, Urrea V, Rigau J, Alegre J, Cabrera C, Oltra E, Castro-Marrero J, Blanco J. Impact of Long-Term Cryopreservation on Blood Immune Cell Markers in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Implications for Biomarker Discovery. Front Immunol 2020; 11:582330. [PMID: 33329554 PMCID: PMC7732598 DOI: 10.3389/fimmu.2020.582330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/20/2020] [Indexed: 12/11/2022] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a complex neuroimmune disorder characterized by numerous symptoms of unknown etiology. The ME/CFS immune markers reported so far have failed to generate a clinical consensus, perhaps partly due to the limitations of biospecimen biobanking. To address this issue, we performed a comparative analysis of the impact of long-term biobanking on previously identified immune markers and also explored additional potential immune markers linked to infection in ME/CFS. A correlation analysis of marker cryostability across immune cell subsets based on flow cytometry immunophenotyping of fresh blood and frozen PBMC samples collected from individuals with ME/CFS (n = 18) and matched healthy controls (n = 18) was performed. The functionality of biobanked samples was assessed on the basis of cytokine production assay after stimulation of frozen PBMCs. T cell markers defining Treg subsets and the expression of surface glycoprotein CD56 in T cells and the frequency of the effector CD8 T cells, together with CD57 expression in NK cells, appeared unaltered by biobanking. By contrast, NK cell markers CD25 and CD69 were notably increased, and NKp46 expression markedly reduced, by long-term cryopreservation and thawing. Further exploration of Treg and NK cell subsets failed to identify significant differences between ME/CFS patients and healthy controls in terms of biobanked PBMCs. Our findings show that some of the previously identified immune markers in T and NK cell subsets become unstable after cell biobanking, thus limiting their use in further immunophenotyping studies for ME/CFS. These data are potentially relevant for future multisite intervention studies and cooperative projects for biomarker discovery using ME/CFS biobanked samples. Further studies are needed to develop novel tools for the assessment of biomarker stability in cryopreserved immune cells from people with ME/CFS.
Collapse
Affiliation(s)
- Elisabet Gómez-Mora
- IrsiCaixa AIDS Research Institute, Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Víctor Urrea
- IrsiCaixa AIDS Research Institute, Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | | | - José Alegre
- Division of Rheumatology, ME/CFS Clinical Unit, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cecilia Cabrera
- IrsiCaixa AIDS Research Institute, Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Elisa Oltra
- School of Medicine, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Jesús Castro-Marrero
- Division of Rheumatology, ME/CFS Research Unit, Vall d’Hebron Hospital Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- Chair in Infectious Diseases and Immunity, Centre for Health and Social Care Research (CESS), Faculty of Medicine, University of Vic, Central University of Catalonia (UVic–UCC), Vic, Spain
| |
Collapse
|
16
|
Wieckowski S, Avenal C, Orjalo AV, Gygax D, Cymer F. Toward a Better Understanding of Bioassays for the Development of Biopharmaceuticals by Exploring the Structure-Antibody-Dependent Cellular Cytotoxicity Relationship in Human Primary Cells. Front Immunol 2020; 11:552596. [PMID: 33193318 PMCID: PMC7658677 DOI: 10.3389/fimmu.2020.552596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/28/2020] [Indexed: 01/02/2023] Open
Abstract
Pharmaceutical manufacturing relies on rigorous methods of quality control of drugs and in particular of the physico-chemical and functional characterizations of monoclonal antibodies. To that end, robust bioassays are very often limited to reporter gene assays and the use of immortalized cell lines that are supposed to mimic immune cells such as natural killer (NK) cells to the detriment of primary materials, which are appreciated for their biological validity but are also difficult to exploit due to the great diversity between individuals. Here, we characterized the phenotype of the peripheral blood circulating cytotoxic cells of 30 healthy donors, in particular the repertoire of cytotoxic markers, using flow cytometry. In parallel, we characterized the antibody-dependent cellular cytotoxicity (ADCC) effector functions of these primary cells by measuring their cytolytic activity against a cancer cell-line expressing HER2 in the presence of trastuzumab and with regards to FCGR3A genotype. We could not establish a correlation or grouping of individuals using the data generated from whole peripheral blood mononuclear cells, however the isolation of the CD56-positive population, which is composed not only of NK cells but also of natural killer T (NKT) and γδ-T cells, as well as subsets of activated cytotoxic T cells, monocytes and dendritic cells, made it possible to standardize the parameters of the ADCC and enhance the overall functional avidity without however eliminating the inter-individual diversity. Finally, the use of primary CD56+ cells in ADCC experiments comparing glycoengineered variants of trastuzumab was conclusive to test the limits of this type of ex vivo system. Although the effector functions of CD56+ cells reflected to some extent the in vitro receptor binding properties and cytolytic activity data using NK92 cells, as previously published, reaching a functional avidity plateau could limit their use in a quality control framework.
Collapse
Affiliation(s)
- Sébastien Wieckowski
- School of Life Sciences, Institute for Chemistry and Bioanalytics, University of Applied Life Sciences and Arts Northwestern Switzerland (FHNW), Muttenz, Switzerland
| | - Cécile Avenal
- Department PTDE-A, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Arturo V. Orjalo
- Biological Technologies, Genentech, Inc., South San Francisco, CA, United States
| | - Daniel Gygax
- School of Life Sciences, Institute for Chemistry and Bioanalytics, University of Applied Life Sciences and Arts Northwestern Switzerland (FHNW), Muttenz, Switzerland
| | - Florian Cymer
- Department PTDE-A, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
17
|
Mfarrej B, Gaude J, Couquiaud J, Calmels B, Chabannon C, Lemarie C. Validation of a flow cytometry-based method to quantify viable lymphocyte subtypes in fresh and cryopreserved hematopoietic cellular products. Cytotherapy 2020; 23:77-87. [PMID: 32718876 DOI: 10.1016/j.jcyt.2020.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/27/2020] [Accepted: 06/22/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND AIMS Adoptive cellular therapy with immune effector cells (IECs) has shown promising efficacy against some neoplastic diseases as well as potential in immune regulation. Both inherent variability in starting material and variations in cell composition produced by the manufacturing process must be thoroughly evaluated with a validated method established to quantify viable lymphocyte subtypes. Currently, commercialized immunophenotyping methods determine cell viability with significant errors in thawed products since they do not include any viability staining. We hereby report on the validation of a flow cytometry-based method for quantifying viable lymphocyte immunophenotypes in fresh and cryopreserved hematopoietic cellular products. METHODS Using fresh or frozen cellular products and stabilized blood, we report on the validation parameters accuracy, uncertainty, precision, sensitivity, robustness and contamination between samples for quantification of viable CD3+, CD4+ T cells, CD8+ T cells, CD3-CD56+CD16+/- NK cells, CD19+ B cells and CD14+ monocytes of relevance to fresh and cryopreserved hematopoietic cellular products using the Cytomics FC500 cytometer (Beckman Coulter). RESULTS The acceptance criteria set in the validation plan were all met. The method is able to accommodate the variability in absolute numbers of cells in starting materials collected or cryopreserved from patients or healthy donors (uncertainty of ≤20% at three different concentrations), stability over time (compliance over 3 years during regular inter-laboratory comparisons) and confidence in meaningful changes during cell processing and manufacturing (intra-assay and intermediate precision of 10% coefficient of variation). Furthermore, the method can accurately report on the efficacy of cell depletion since the lower limit of quantification was established (CD3+, CD4+ and CD8+ cells at 9, 8 and 8 cells/µL, respectively). The method complies with Foundation for the Accreditation of Cellular Therapy (FACT) standards for IEC, FACT-Joint Accreditation Committee of ISCT-EBMT (JACIE) hematopoietic cell therapy standards, International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use Q2(R1) and International Organization for Standardization 15189 standards. Furthermore, it complies with Ligand Binding Assay Bioanalytical Focus Group/American Association of Pharmaceutical Scientists, International Council for Standardization of Hematology/International Clinical Cytometry Society and European Bioanalysis Forum recommendations for validating such methods. CONCLUSIONS The implications of this effort include standardization of viable cell immunophenotyping of starting material for cell manufacturing, cell selection and in-process quality controls or dosing of IECs. This method also complies with all relevant standards, particularly FACT-JACIE standards, in terms of enumerating and reporting on the viability of the "clinically relevant cell populations."
Collapse
Affiliation(s)
- Bechara Mfarrej
- Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France.
| | - Julie Gaude
- Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France
| | - Jerome Couquiaud
- Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France
| | - Boris Calmels
- Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France
| | | | - Claude Lemarie
- Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France
| |
Collapse
|
18
|
Standardization procedure for flow cytometry data harmonization in prospective multicenter studies. Sci Rep 2020; 10:11567. [PMID: 32665668 PMCID: PMC7360585 DOI: 10.1038/s41598-020-68468-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 06/22/2020] [Indexed: 01/26/2023] Open
Abstract
One of the most challenging objective for clinical cytometry in prospective multicenter immunomonitoring trials is to compare frequencies, absolute numbers of leukocyte populations and further the mean fluorescence intensities of cell markers, especially when the data are generated from different instruments. Here, we describe an innovative standardization workflow to compare all data to carry out any large-scale, prospective multicentric flow cytometry analysis whatever the duration, the number or type of instruments required for the realization of such projects.
Collapse
|
19
|
Phelps DS, Chinchilli VM, Weisz J, Shearer D, Zhang X, Floros J. Using toponomics to characterize phenotypic diversity in alveolar macrophages from male mice treated with exogenous SP-A1. Biomark Res 2020; 8:5. [PMID: 32082572 PMCID: PMC7020580 DOI: 10.1186/s40364-019-0181-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/30/2019] [Indexed: 01/12/2023] Open
Abstract
Background We used the Toponome Imaging System (TIS) to identify “patterns of marker expression”, referred to here as combinatorial molecular phenotypes (CMPs) in alveolar macrophages (AM) in response to the innate immune molecule, SP-A1. Methods We compared 114 AM from male SP-A deficient mice. One group (n = 3) was treated with exogenous human surfactant protein A1 (hSP-A1) and the other with vehicle (n = 3). AM obtained by bronchoalveolar lavage were plated onto slides and analyzed using TIS to study the AM toponome, the spatial network of proteins within intact cells. With TIS, each slide is sequentially immunostained with multiple FITC-conjugated antibodies. Images are analyzed pixel-by-pixel identifying all of the proteins within each pixel, which are then designated as CMPs. CMPs represent organized protein clusters postulated to contribute to specific functions. Results 1) We compared identical CMPs in KO and SP-A1 cells and found them to differ significantly (p = 0.0007). Similarities between pairs of markers in the two populations also differed significantly (p < 0.0001). 2) Focusing on the 20 most abundant CMPs for each cell, we developed a method to generate CMP “signatures” that characterized various groups of cells. Phenotypes were defined as cells exhibiting similar signatures of CMPs. i) AM were extremely diverse and each group contained cells with multiple phenotypes. ii) Among the 114 AM analyzed, no two cells were identical. iii) However, CMP signatures could distinguish among cell subpopulations within and between groups. iv) Some cell populations were enriched with SP-A1 treatment, some were more common without SP-A1, and some seemed not to be influenced by the presence of SP-A1. v) We also found that AM were more diverse in mice treated with SP-A1 compared to those treated with vehicle. Conclusions AM diversity is far more extensive than originally thought. The increased diversity of SP-A1-treated mice points to the possibility that SP-A1 enhances or activates several pathways in the AM to better prepare it for its innate immune functions and other functions shown previously to be affected by SP-A treatment. Future studies may identify key protein(s) responsible for CMP integrity and consequently for a given function, and target it for therapeutic purposes.
Collapse
Affiliation(s)
- David S Phelps
- 1Penn State Center for Host defense, Inflammation, and Lung Disease (CHILD) Research and Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Vernon M Chinchilli
- 2Public Health Sciences; and Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Judith Weisz
- 3Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Debra Shearer
- 3Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Xuesheng Zhang
- 1Penn State Center for Host defense, Inflammation, and Lung Disease (CHILD) Research and Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Joanna Floros
- 1Penn State Center for Host defense, Inflammation, and Lung Disease (CHILD) Research and Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA.,3Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| |
Collapse
|
20
|
Liu Y, Guo J, Huang L. Modulation of tumor microenvironment for immunotherapy: focus on nanomaterial-based strategies. Am J Cancer Res 2020; 10:3099-3117. [PMID: 32194857 PMCID: PMC7053194 DOI: 10.7150/thno.42998] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/19/2020] [Indexed: 02/07/2023] Open
Abstract
Recent advances in the field of immunotherapy have profoundly opened up the potential for improved cancer therapy and reduced side effects. However, the tumor microenvironment (TME) is highly immunosuppressive, therefore, clinical outcomes of currently available cancer immunotherapy are still poor. Recently, nanomaterial-based strategies have been developed to modulate the TME for robust immunotherapeutic responses. In this review, the immunoregulatory cell types (cells relating to the regulation of immune responses) inside the TME in terms of stimulatory and suppressive roles are described, and the technologies used to identify and quantify these cells are provided. In addition, recent examples of nanomaterial-based cancer immunotherapy are discussed, with particular emphasis on those designed to overcome barriers caused by the complexity and diversity of TME.
Collapse
|
21
|
van Dongen JJM, van der Burg M, Kalina T, Perez-Andres M, Mejstrikova E, Vlkova M, Lopez-Granados E, Wentink M, Kienzler AK, Philippé J, Sousa AE, van Zelm MC, Blanco E, Orfao A. EuroFlow-Based Flowcytometric Diagnostic Screening and Classification of Primary Immunodeficiencies of the Lymphoid System. Front Immunol 2019; 10:1271. [PMID: 31263462 PMCID: PMC6585843 DOI: 10.3389/fimmu.2019.01271] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 05/17/2019] [Indexed: 12/16/2022] Open
Abstract
Guidelines for screening for primary immunodeficiencies (PID) are well-defined and several consensus diagnostic strategies have been proposed. These consensus proposals have only partially been implemented due to lack of standardization in laboratory procedures, particularly in flow cytometry. The main objectives of the EuroFlow Consortium were to innovate and thoroughly standardize the flowcytometric techniques and strategies for reliable and reproducible diagnosis and classification of PID of the lymphoid system. The proposed EuroFlow antibody panels comprise one orientation tube and seven classification tubes and corresponding databases of normal and PID samples. The 8-color 12-antibody PID Orientation tube (PIDOT) aims at identification and enumeration of the main lymphocyte and leukocyte subsets; this includes naïve pre-germinal center (GC) and antigen-experienced post-GC memory B-cells and plasmablasts. The seven additional 8(-12)-color tubes can be used according to the EuroFlow PID algorithm in parallel or subsequently to the PIDOT for more detailed analysis of B-cell and T-cell subsets to further classify PID of the lymphoid system. The Pre-GC, Post-GC, and immunoglobulin heavy chain (IgH)-isotype B-cell tubes aim at identification and enumeration of B-cell subsets for evaluation of B-cell maturation blocks and specific defects in IgH-subclass production. The severe combined immunodeficiency (SCID) tube and T-cell memory/effector subset tube aim at identification and enumeration of T-cell subsets for assessment of T-cell defects, such as SCID. In case of suspicion of antibody deficiency, PIDOT is preferably directly combined with the IgH isotype tube(s) and in case of SCID suspicion (e.g., in newborn screening programs) the PIDOT is preferably directly combined with the SCID T-cell tube. The proposed ≥8-color antibody panels and corresponding reference databases combined with the EuroFlow PID algorithm are designed to provide fast, sensitive and cost-effective flowcytometric diagnosis of PID of the lymphoid system, easily applicable in multicenter diagnostic settings world-wide.
Collapse
Affiliation(s)
- Jacques J M van Dongen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Mirjam van der Burg
- Department of Immunology, Erasmus MC, Rotterdam, Netherlands.,Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Tomas Kalina
- Department of Pediatric Hematology and Oncology, University Hospital Motol, Charles University, Prague, Czechia
| | - Martin Perez-Andres
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), University of Salamanca (USAL), Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), CB/16/12/00233, Instituto Carlos III, Madrid, Spain
| | - Ester Mejstrikova
- Department of Pediatric Hematology and Oncology, University Hospital Motol, Charles University, Prague, Czechia
| | - Marcela Vlkova
- Institute of Clinical Immunology and Allergology, St. Anne's University Hospital Brno, Masaryk University, Brno, Czechia
| | | | | | - Anne-Kathrin Kienzler
- Experimental Medicine Division, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jan Philippé
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Ana E Sousa
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Menno C van Zelm
- Department of Immunology, Erasmus MC, Rotterdam, Netherlands.,Department of Immunology and Pathology, Central Clinical School, Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Elena Blanco
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), University of Salamanca (USAL), Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), CB/16/12/00233, Instituto Carlos III, Madrid, Spain
| | - Alberto Orfao
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), University of Salamanca (USAL), Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), CB/16/12/00233, Instituto Carlos III, Madrid, Spain
| |
Collapse
|
22
|
Neo SY, O'Reilly A, Pico de Coaña Y. Immune Monitoring of Cancer Patients by Multi-color Flow Cytometry. Methods Mol Biol 2019; 1913:49-65. [PMID: 30666598 DOI: 10.1007/978-1-4939-8979-9_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The irruption of immune-activating therapies to treat cancer has created a need for evaluating both the response and possible adverse events related to these novel treatments. Multicolor flow cytometry is a powerful tool that enables tumor immunologists to characterize the immune system of patients before and in response to immunotherapy. We present here a protocol for purifying human peripheral blood mononuclear cells and staining them with a set of six multicolor panels that allow for a thorough characterization of the immune system of healthy donors as well as patients that are undergoing treatments that may modify the immune system.
Collapse
Affiliation(s)
- Shi Yong Neo
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Aine O'Reilly
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Yago Pico de Coaña
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
23
|
Villarrubia N, Rodríguez-Martín E, Alari-Pahissa E, Aragón L, Castillo-Triviño T, Eixarch H, Ferrer JM, Martínez-Rodríguez JE, Massot M, Pinto-Medel MJ, Prada Á, Rodríguez-Acevedo B, Urbaneja P, Gascón-Gimenez F, Herrera G, Hernández-Clares R, Salgado MG, Oterino A, San Segundo D, Cuello JP, Gil-Herrera J, Cámara C, Gómez-Gutiérrez M, Martínez-Hernández E, Meca-Lallana V, Moga E, Muñoz-Calleja C, Querol L, Presas-Rodríguez S, Teniente-Serra A, Vlagea A, Muriel A, Roldán E, Villar LM. Multi-centre validation of a flow cytometry method to identify optimal responders to interferon-beta in multiple sclerosis. Clin Chim Acta 2018; 488:135-142. [PMID: 30408481 DOI: 10.1016/j.cca.2018.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/23/2018] [Accepted: 11/03/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVES Percentages of blood CD19+CD5+ B cells and CD8+perforin+ T lymphocytes can predict response to Interferon (IFN)-beta treatment in relapsing-remitting multiple sclerosis (RRMS) patients. We aimed to standardize their detection in a multicenter study, prior to their implementation in clinical practice. METHODS Fourteen hospitals participated in the study. A reference centre was established for comparison studies. Peripheral blood cells of 105 untreated RRMS patients were studied. Every sample was analyzed in duplicate in the participating centre and in the reference one by flow cytometry. When needed, participating centres corrected fluorescence compensations and negative cut-off position following reference centre suggestions. Concordance between results obtained by participating centres and by reference one was evaluated by intraclass correlation coefficients (ICC) and Spearman correlation test. Centre performance was measured by using z-scores values. RESULTS After results review and corrective actions implementation, overall ICC was 0.86 (CI: 0.81-0.91) for CD19+CD5+ B cell and 0.89 (CI: 0.85-0.93) for CD8+ perforin+ T cell quantification; Spearman r was 0.92 (0.89-0.95; p <0.0001) and 0.92 (0.88-0.95; p <0.0001) respectively. All centres obtained z-scores≤0.5 for both biomarkers. CONCLUSION Homogenous percentages of CD19+CD5+ B cells and CD8 perforin+ T lymphocytes can be obtained if suitable compensation values and negative cut-off are pre-established.
Collapse
Affiliation(s)
- Noelia Villarrubia
- Immunology Dpt. and Biostatistic Unit, Hospital Universitario Ramón y Cajal, IRYCIS, Ctra. de Colmenar Viejo km 9.100, 28034 Madrid, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Eulalia Rodríguez-Martín
- Immunology Dpt. and Biostatistic Unit, Hospital Universitario Ramón y Cajal, IRYCIS, Ctra. de Colmenar Viejo km 9.100, 28034 Madrid, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Elisenda Alari-Pahissa
- Immunology and Neurology Dpt., Universitat Pompeu Fabra, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Carrer del Dr. Aiguader 88, 08003 Barcelona, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Larraitz Aragón
- Immunology and Neurology Dpt., Hospital Universitario de Donostia, Biodonostia, P° Dr. Beguiristain 107-111, 20014 San Sebastián, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Tamara Castillo-Triviño
- Immunology and Neurology Dpt., Hospital Universitario de Donostia, Biodonostia, P° Dr. Beguiristain 107-111, 20014 San Sebastián, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Herena Eixarch
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Cemcat, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Passeig Vall d'Hebron, 119-129, 08035 Barcelona, Spain. Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Joana María Ferrer
- Immunology and Neurology Dpt., Hospital Universitari Son Espases, Instituto de Investigación Sanitaria Illes Balears, IdISBa, Ctra. Valldemossa 79, Palma 07010, Spain
| | - José Enrique Martínez-Rodríguez
- Immunology and Neurology Dpt., Universitat Pompeu Fabra, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Carrer del Dr. Aiguader 88, 08003 Barcelona, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Margarita Massot
- Immunology and Neurology Dpt., Hospital Universitari Son Espases, Instituto de Investigación Sanitaria Illes Balears, IdISBa, Ctra. Valldemossa 79, Palma 07010, Spain.
| | - María Jesús Pinto-Medel
- Clinical Management Unit of Neurosciences, Instituto de Biomedicina de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Plaza del Hospital Civil s/n., Málaga, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Álvaro Prada
- Immunology and Neurology Dpt., Hospital Universitario de Donostia, Biodonostia, P° Dr. Beguiristain 107-111, 20014 San Sebastián, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Breogán Rodríguez-Acevedo
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Cemcat, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Passeig Vall d'Hebron, 119-129, 08035 Barcelona, Spain. Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Patricia Urbaneja
- Clinical Management Unit of Neurosciences, Instituto de Biomedicina de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Plaza del Hospital Civil s/n., Málaga, Spain; Red Española de Esclerosis Múltiple (REEM), Spain
| | - Francisco Gascón-Gimenez
- Neuroimmunology Unit, Hospital Clínic Universitari de València, Av. de Blasco Ibáñez 17, 46010 València, Spain; Red Española de Esclerosis Múltiple (REEM), Spain
| | - Guadalupe Herrera
- Flow Cytometry Unit, UCIM, INCLIVA-Universidad de Valencia, Avda Blasco Ibañez 13, 46010 València, Spain
| | - Rocío Hernández-Clares
- Immunology and Neurology Dpt., Hospital Universitario Virgen de la Arrixaca, Ctra. Madrid-Cartagena, s/n, 30120 El Palmar, Murcia, Spain
| | - María Gema Salgado
- Immunology and Neurology Dpt., Hospital Universitario Virgen de la Arrixaca, Ctra. Madrid-Cartagena, s/n, 30120 El Palmar, Murcia, Spain
| | - Agustín Oterino
- Immunology and Neurology Dpt., IDIVAL, Hospital Universitario Marqués de Valdecilla, Avda. Valdecilla 25, 39008 Santander, Spain; Red Española de Esclerosis Múltiple (REEM), Spain
| | - David San Segundo
- Immunology and Neurology Dpt., IDIVAL, Hospital Universitario Marqués de Valdecilla, Avda. Valdecilla 25, 39008 Santander, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Juan Pablo Cuello
- Immunology and Neurology Dpt., Hospital General Universitario and Instituto de Investigación Sanitaria "Gregorio Marañón", C/ Dr Esquerdo 46, 28007 Madrid, Spain.
| | - Juana Gil-Herrera
- Immunology and Neurology Dpt., Hospital General Universitario and Instituto de Investigación Sanitaria "Gregorio Marañón", C/ Dr Esquerdo 46, 28007 Madrid, Spain.
| | - Carmen Cámara
- Immunology and Neurology Dpt., Hospital San Pedro de Alcántara, Avda. Pablo Naranjo s/n, 10003 Cáceres, Spain.
| | - Montserrat Gómez-Gutiérrez
- Immunology and Neurology Dpt., Hospital San Pedro de Alcántara, Avda. Pablo Naranjo s/n, 10003 Cáceres, Spain
| | - Eugenia Martínez-Hernández
- Immunology and Neurology Dpt., Hospital Clínic de Barcelona, Carrer de Villarroel, 170, 08036 Barcelona, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Virginia Meca-Lallana
- Immunology and Neurology Dpt., Instituto de Investigación Sanitaria La Princesa, IIS-IP, Demyelinating Diseases Unit, Hospital Universitario de La Princesa, C/ Diego de León 62, 28006 Madrid, Spain
| | - Esther Moga
- Immunology and Neurology Dpt., Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, C/ Sant Antoni Maria Claret 167, 08025 Barcelona, Ciberer, Madrid, Spain.
| | - Cecilia Muñoz-Calleja
- Immunology and Neurology Dpt., Instituto de Investigación Sanitaria La Princesa, IIS-IP, Demyelinating Diseases Unit, Hospital Universitario de La Princesa, C/ Diego de León 62, 28006 Madrid, Spain.
| | - Luis Querol
- Immunology and Neurology Dpt., Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, C/ Sant Antoni Maria Claret 167, 08025 Barcelona, Ciberer, Madrid, Spain.
| | - Silvia Presas-Rodríguez
- Immunology and Neurology Dpt., Multiple Sclerosis Unit, Hospital Universitario e Instituto de Investigación Germans Trias i Pujol, Campus Can Ruti, 08916 Badalona, Barcelona, Spain; Red Española de Esclerosis Múltiple (REEM), Spain
| | - Aina Teniente-Serra
- Immunology and Neurology Dpt., Multiple Sclerosis Unit, Hospital Universitario e Instituto de Investigación Germans Trias i Pujol, Campus Can Ruti, 08916 Badalona, Barcelona, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Alexandru Vlagea
- Immunology and Neurology Dpt., Hospital Clínic de Barcelona, Carrer de Villarroel, 170, 08036 Barcelona, Spain.
| | - Alfonso Muriel
- Immunology Dpt. and Biostatistic Unit, Hospital Universitario Ramón y Cajal, IRYCIS, Ctra. de Colmenar Viejo km 9.100, 28034 Madrid, Spain.
| | - Ernesto Roldán
- Immunology Dpt. and Biostatistic Unit, Hospital Universitario Ramón y Cajal, IRYCIS, Ctra. de Colmenar Viejo km 9.100, 28034 Madrid, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| | - Luisa María Villar
- Immunology Dpt. and Biostatistic Unit, Hospital Universitario Ramón y Cajal, IRYCIS, Ctra. de Colmenar Viejo km 9.100, 28034 Madrid, Spain; Red Española de Esclerosis Múltiple (REEM), Spain.
| |
Collapse
|
24
|
Desforges JP, Jasperse L, Jensen TH, Grøndahl C, Bertelsen MF, Guise SD, Sonne C, Dietz R, Levin M. Immune function in arctic mammals: Natural killer (NK) cell-like activity in polar bear, muskox and reindeer. Vet Immunol Immunopathol 2017; 195:72-75. [PMID: 29249321 DOI: 10.1016/j.vetimm.2017.11.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/31/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022]
Abstract
Natural killer (NK) cells are a vital part of the rapid and non-specific immune defense against invading pathogens and tumor cells. This study evaluated NK cell-like activity by flow cytometry for the first time in three ecologically and culturally important Arctic mammal species: polar bear (Ursus maritimus), muskox (Ovibos moschatus) and reindeer (Rangifer tarandus). NK cell-like activity for all three species was most effective against the mouse lymphoma cell line YAC-1, compared to the human leukemia cell line K562; NK cell response displayed the characteristic increase in cytotoxic activity when the effector:target cell ratio increased. Comparing NK activity between fresh and cryopreserved mouse lymphocytes revealed little to no difference in function, highlighting the applicability of cryopreserving cells in field studies. The evaluation of this important innate immune function in Arctic mammals can contribute to future population health assessments, especially as pollution-induced suppression of immune function may increase infectious disease susceptibility.
Collapse
Affiliation(s)
- Jean-Pierre Desforges
- Department of Bioscience, Arctic Research Centre, Aarhus University, Frederiksborgvej 399, PO Box 358, DK-4000, Roskilde, Denmark.
| | - Lindsay Jasperse
- Department of Pathobiology and Veterinary Science, University of Connecticut, 61 North Eagleville Road, Storrs, CT, 06269-3089, United States
| | | | - Carsten Grøndahl
- Center for Zoo and Wild Animal Health, Copenhagen Zoo, Roskildevej 38, DK-2000, Frederiksberg, Denmark
| | - Mads F Bertelsen
- Center for Zoo and Wild Animal Health, Copenhagen Zoo, Roskildevej 38, DK-2000, Frederiksberg, Denmark
| | - Sylvain De Guise
- Department of Pathobiology and Veterinary Science, University of Connecticut, 61 North Eagleville Road, Storrs, CT, 06269-3089, United States
| | - Christian Sonne
- Department of Bioscience, Arctic Research Centre, Aarhus University, Frederiksborgvej 399, PO Box 358, DK-4000, Roskilde, Denmark
| | - Rune Dietz
- Department of Bioscience, Arctic Research Centre, Aarhus University, Frederiksborgvej 399, PO Box 358, DK-4000, Roskilde, Denmark
| | - Milton Levin
- Department of Pathobiology and Veterinary Science, University of Connecticut, 61 North Eagleville Road, Storrs, CT, 06269-3089, United States
| |
Collapse
|
25
|
Delso-Vallejo M, Kollet J, Koehl U, Huppert V. Influence of Irradiated Peripheral Blood Mononuclear Cells on Both Ex Vivo Proliferation of Human Natural Killer Cells and Change in Cellular Property. Front Immunol 2017; 8:854. [PMID: 28791015 PMCID: PMC5522833 DOI: 10.3389/fimmu.2017.00854] [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: 02/22/2017] [Accepted: 07/06/2017] [Indexed: 11/13/2022] Open
Abstract
Clinical studies with adoptive immunotherapy using allogeneic natural killer (NK) cells showed feasibility, but also limitation regarding the transfused absolute cell numbers. First promising results with peripheral blood mononuclear cells (PBMCs) as feeder cells to improve the final cell number need further optimization and investigation of the unknown controlling mechanism in the cross-talk to NK cells. We investigated the influence of irradiated autologous PBMCs to boost NK cell proliferation in the presence of OKT3 and IL-2. Our findings demonstrate a requirement for receptor-ligand interactions between feeders and NK cells to produce soluble factors that can sustain NK cell proliferation. Thus, both physical contact between feeder and NK cells, and soluble factors produced in consequence, are required to fully enhance NK cell ex vivo proliferation. This occurred with an indispensable role of the cross-talk between T cells, monocytes, and NK cells, while B cells had no further influence in supporting NK cell proliferation under these co-culture conditions. Moreover, gene expression analysis of highly proliferating and non-proliferating NK cells revealed important phenotypic changes on 5-day cultured NK cells. Actively proliferating NK cells have reduced Siglec-7 and -9 expression compared with non-proliferating and resting NK cells (day 0), independently of the presence of feeder cells. Interestingly, proliferating NK cells cultured with feeder cells contained increased frequencies of cells expressing RANKL, B7-H3, and HLA class II molecules, particularly HLA-DR, compared with resting NK cells or expanded with IL-2 only. A subset of HLA-DR expressing NK cells, co-expressing RANKL, and B7-H3 corresponded to the most proliferative population under the established co-culture conditions. Our results highlight the importance of the crosstalk between T cells, monocytes, and NK cells in autologous feeder cell-based ex vivo NK cell expansion protocols, and reveal the appearance of a highly proliferative subpopulation of NK cells (HLA-DR+RANKL+B7-H3+) with promising characteristics to extend the therapeutic potential of NK cells.
Collapse
Affiliation(s)
| | - Jutta Kollet
- Miltenyi Biotec GmbH, Bergisch-Gladbach, Germany
| | - Ulrike Koehl
- Hannover Medical School, Institute for Cellular Therapeutics, IFB-Tx, Hannover, Germany
| | | |
Collapse
|