1
|
Yu Q, Zhang T, He T, Yang Y, Zhang W, Kang Y, Wu Z, Xie W, Zheng J, Qian Q, Li G, Zhang D, Mao Q, Gao Z, Wang X, Shi X, Huang S, Guo H, Zhang H, Chen L, Li X, Deng D, Zhang L, Tong Y, Yao W, Gao X, Tian H. Altered epitopes enhance macrophage-mediated anti-tumour immunity to low-immunogenic tumour mutations. Immunology 2024; 173:654-671. [PMID: 39174487 DOI: 10.1111/imm.13854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/02/2024] [Indexed: 08/24/2024] Open
Abstract
Personalized neoantigen therapy has shown long-term and stable efficacy in specific patient populations. However, not all patients have sufficient levels of neoantigens for treatment. Although somatic mutations are commonly found in tumours, a significant portion of these mutations do not trigger an immune response. Patients with low mutation burdens continue to exhibit unresponsiveness to this treatment. We propose a design paradigm for neoantigen vaccines by utilizing the highly immunogenic unnatural amino acid p-nitrophenylalanine (pNO2Phe) for sequence alteration of somatic mutations that failed to generate neoepitopes. This enhances the immunogenicity of the mutations and transforms it into a suitable candidate for immunotherapy. The nitrated altered epitope vaccines designed according to this paradigm is capable of activating circulating CD8+ T cells and inducing immune cross-reactivity against autologous mutated epitopes in different MHC backgrounds (H-2Kb, H-2Kd, and human HLA-A02:01), leading to the elimination of tumour cells carrying the mutation. After immunization with the altered epitopes, tumour growth was significantly inhibited. It is noteworthy that nitrated epitopes induce tumour-infiltrating macrophages to differentiate into the M1 phenotype, surprisingly enhancing the MHC II molecule presenting pathway of macrophages. Nitrated epitope-treated macrophages have the potential to cross-activate CD4+ and CD8+ T cells, which may explain why pNO2Phe can enhance the immunogenicity of epitopes. Meanwhile, the immunosuppressive microenvironment of the tumour is altered due to the activation of macrophages. The nitrated neoantigen vaccine strategy enables the design of vaccines targeting non-immunogenic tumour mutations, expanding the pool of potential peptides for personalized and shared novel antigen therapy. This approach provides treatment opportunities for patients previously ineligible for new antigen vaccine therapy.
Collapse
Affiliation(s)
- Qiumin Yu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Tingran Zhang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Tiandi He
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yifan Yang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wanli Zhang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yanliang Kang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zijie Wu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenbin Xie
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jiaxue Zheng
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qianqian Qian
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Guozhi Li
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Di Zhang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qiuli Mao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zheng Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaoning Wang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xupeiyao Shi
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shitong Huang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Hanlin Guo
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Haoyu Zhang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Lingxiao Chen
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ximing Li
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Danni Deng
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, China
| | - Li Zhang
- Department of General Internal Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yue Tong
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenbing Yao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Hong Tian
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
2
|
Lazarski CA, Hanley PJ. Review of flow cytometry as a tool for cell and gene therapy. Cytotherapy 2024; 26:103-112. [PMID: 37943204 PMCID: PMC10872958 DOI: 10.1016/j.jcyt.2023.10.005] [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: 08/16/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Quality control testing and analytics are critical for the development and manufacture of cell and gene therapies, and flow cytometry is a key quality control and analytical assay that is used extensively. However, the technical scope of characterization assays and safety assays must keep apace as the breadth of cell therapy products continues to expand beyond hematopoietic stem cell products into producing novel adoptive immune therapies and gene therapy products. Flow cytometry services are uniquely positioned to support the evolving needs of cell therapy facilities, as access to flow cytometers, new antibody clones and improved fluorochrome reagents becomes more egalitarian. This report will outline the features, logistics, limitations and the current state of flow cytometry within the context of cellular therapy.
Collapse
Affiliation(s)
- Christopher A Lazarski
- Program for Cell Enhancement and Technology for Immunotherapy, Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA; The George Washington University, Washington, DC, USA.
| | - Patrick J Hanley
- Program for Cell Enhancement and Technology for Immunotherapy, Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA; The George Washington University, Washington, DC, USA.
| |
Collapse
|
3
|
Koukoulias K, Papayanni PG, Jones J, Kuvalekar M, Watanabe A, Velazquez Y, Gilmore S, Papadopoulou A, Leen AM, Vasileiou S. Assessment of the cytolytic potential of a multivirus-targeted T cell therapy using a vital dye-based, flow cytometric assay. Front Immunol 2023; 14:1299512. [PMID: 38187380 PMCID: PMC10766817 DOI: 10.3389/fimmu.2023.1299512] [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: 09/22/2023] [Accepted: 12/05/2023] [Indexed: 01/09/2024] Open
Abstract
Reliable and sensitive characterization assays are important determinants of the successful clinical translation of immunotherapies. For the assessment of cytolytic potential, the chromium 51 (51Cr) release assay has long been considered the gold standard for testing effector cells. However, attaining the approvals to access and use radioactive isotopes is becoming increasingly complex, while technical aspects [i.e. sensitivity, short (4-6 hours) assay duration] may lead to suboptimal performance. This has been the case with our ex vivo expanded, polyclonal (CD4+ and CD8+) multivirus-specific T cell (multiVST) lines, which recognize 5 difficult-to-treat viruses [Adenovirus (AdV), BK virus (BKV), cytomegalovirus (CMV), Epstein Barr virus (EBV), and human herpes virus 6 (HHV6)] and when administered to allogeneic hematopoietic stem cell (HCT) or solid organ transplant (SOT) recipients have been associated with clinical benefit. However, despite mediating potent antiviral effects in vivo, capturing in vitro cytotoxic potential has proven difficult in a traditional 51Cr release assay. Now, in addition to cytotoxicity surrogates, including CD107a and Granzyme B, we report on an alternative, vital dye -based, flow cytometric platform in which superior sensitivity and prolonged effector:target co-culture duration enabled the reliable detection of both CD4- and CD8-mediated in vitro cytolytic activity against viral targets without non-specific effects.
Collapse
Affiliation(s)
- Kiriakos Koukoulias
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Penelope G. Papayanni
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Julia Jones
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Manik Kuvalekar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Ayumi Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Yovana Velazquez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | | | - Anastasia Papadopoulou
- Hematology Department- Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, Thessaloniki, Greece
| | - Ann M. Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Spyridoula Vasileiou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
4
|
Activated T cell therapy targeting glioblastoma cancer stem cells. Sci Rep 2023; 13:196. [PMID: 36604465 PMCID: PMC9814949 DOI: 10.1038/s41598-022-27184-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
Naïve T cells become effector T cells following stimulation by antigen-loaded dendritic cells (DCs) and sequential cytokine activation. We aimed to develop procedures to efficiently activate T cells with tumor-associated antigens (TAAs) to glioblastoma (GBM) stem cells. To remove antigen presentation outside of the immunosuppressive tumor milieu, three different glioma stem cell (GSC) specific antigen sources to load DCs were compared in their ability to stimulate lymphocytes. An activated T cell (ATC) protocol including cytokine activation and expansion in culture to target GSCs was generated and optimized for a planned phase I clinical trial. We compared three different antigen-loading methods on DCs to effectively activate T cells, which were GBM patient-derived GSC-lysate, acid-eluate of GSCs and synthetic peptides derived from proteins expressed in GSCs. DCs derived from HLA-A2 positive blood sample were loaded with TAAs. Autologous T cells were activated by co-culturing with loaded DCs. Efficiency and cytotoxicity of ATCs were evaluated by targeting TAA-pulsed DCs or T2 cells, GSCs, or autologous PHA-blasts. Characteristics of ATCs were evaluated by Flow Cytometry and ELISpot assay, which showed increased number of ATCs secreting IFN-γ targeting GSCs as compared with non-activated T cells and unloaded target cells. Neither GSC-lysate nor acid-eluate loading showed enhancement in response of ATCs but the synthetic peptide pool showed significantly increased IFN-γ secretion and increased cytotoxicity towards target cells. These results demonstrate that ATCs activated using a TAA synthetic peptide pool efficiently enhance cytotoxicity specifically to target cells including GSC.
Collapse
|
5
|
BST2, a Novel Inhibitory Receptor, Is Involved in NK Cell Cytotoxicity through Its Cytoplasmic Tail Domain. Int J Mol Sci 2022; 23:ijms231911395. [PMID: 36232695 PMCID: PMC9570199 DOI: 10.3390/ijms231911395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Bone Marrow Stromal Cell Antigen 2 (BST2) is a type II transmembrane protein expressed on various cell types that tethers the release of viruses. Natural killer (NK) cells express low levels of BST2 under normal conditions but exhibit increased expression of BST2 upon activation. In this study, we show for the first time that murine BST2 can control the cytotoxicity of NK cells. The cytoplasmic tail of murine BST2 contains an immunoreceptor tyrosine-based inhibitory motif (ITIM). The absence of BST2 on NK cells can enhance their cytotoxicity against tumor cells compared to wild type NK cells. NK cells isolated from NZW mice, which express ITIM-deficient BST2, also showed higher cytotoxicity than wild type NK cells. In addition, we found that galectin-8 and galectin-9 were ligands of BST2, since blocking galectin-8 or -9 with monoclonal antibodies enhanced the cytotoxicity of NK cells. These results suggested that BST2 might be a novel NK cell inhibitory receptor as it was involved in regulating NK cell cytotoxicity through its interaction with galectins.
Collapse
|
6
|
Rodríguez‐Izquierdo I, Sepúlveda‐Crespo D, Lasso JM, Resino S, Muñoz‐Fernández MÁ. Baseline and time-updated factors in preclinical development of anionic dendrimers as successful anti-HIV-1 vaginal microbicides. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1774. [PMID: 35018739 PMCID: PMC9285063 DOI: 10.1002/wnan.1774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/05/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022]
Abstract
Although a wide variety of topical microbicides provide promising in vitro and in vivo efficacy, most of them failed to prevent sexual transmission of human immunodeficiency virus type 1 (HIV-1) in human clinical trials. In vitro, ex vivo, and in vivo models must be optimized, considering the knowledge acquired from unsuccessful and successful clinical trials to improve the current gaps and the preclinical development protocols. To date, dendrimers are the only nanotool that has advanced to human clinical trials as topical microbicides to prevent HIV-1 transmission. This fact demonstrates the importance and the potential of these molecules as microbicides. Polyanionic dendrimers are highly branched nanocompounds with potent activity against HIV-1 that disturb HIV-1 entry. Herein, the most significant advancements in topical microbicide development, trying to mimic the real-life conditions as closely as possible, are discussed. This review also provides the preclinical assays that anionic dendrimers have passed as microbicides because they can improve current antiviral treatments' efficacy. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
| | - Daniel Sepúlveda‐Crespo
- Unidad de Infección Viral e Inmunidad, Centro Nacional de MicrobiologíaInstituto de Salud Carlos IIIMadridSpain
| | | | - Salvador Resino
- Unidad de Infección Viral e Inmunidad, Centro Nacional de MicrobiologíaInstituto de Salud Carlos IIIMadridSpain
| | - Ma Ángeles Muñoz‐Fernández
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM)MadridSpain
- Spanish HIV HGM BioBankMadridSpain
- Section of Immunology, Laboratorio InmunoBiología MolecularHospital General Universitario Gregorio Marañón (HGUGM)MadridSpain
| |
Collapse
|
7
|
Liu C, Skorupinska-Tudek K, Eriksson SG, Parmryd I. Potentiating Vγ9Vδ2 T cell proliferation and assessing their cytotoxicity towards adherent cancer cells at the single cell level. Biol Open 2022; 11:274281. [PMID: 34994391 PMCID: PMC8822357 DOI: 10.1242/bio.059049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/09/2021] [Indexed: 11/20/2022] Open
Abstract
Vγ9Vδ2 T cells is the dominant γδ T cell subset in human blood. They are cytotoxic and activated by phosphoantigens whose concentrations are increased in cancer cells, making the cancer cells targets for Vγ9Vδ2 T cell immunotherapy. For successful immunotherapy, it is important both to characterise Vγ9Vδ2 T cell proliferation and optimise the assessment of their cytotoxic potential, which is the aim of this study. We found that supplementation with freshly-thawed human serum potentiated Vγ9Vδ2 T cell proliferation from peripheral mononuclear cells (PBMCs) stimulated with (E)-4-Hydroxy-3-methyl-but-2-enyl diphosphate (HMBPP) and consistently enabled Vγ9Vδ2 T cell proliferation from cryopreserved PBMCs. In cryopreserved PBMCs the proliferation was higher than in freshly prepared PBMCs. In a panel of short-chain prenyl alcohols, monophosphates and diphosphates, most diphosphates and also dimethylallyl monophosphate stimulated Vγ9Vδ2 T cell proliferation. We developed a method where the cytotoxicity of Vγ9Vδ2 T cells towards adherent cells is assessed at the single cell level using flow cytometry, which gives more clear-cut results than the traditional bulk release assays. Moreover, we found that HMBPP enhances the Vγ9Vδ2 T cell cytotoxicity towards colon cancer cells. In summary we have developed an easily interpretable method to assess the cytotoxicity of Vγ9Vδ2 T cells towards adherent cells, found that Vγ9Vδ2 T cell proliferation can be potentiated media-supplementation and how misclassification of non-responders may be avoided. Our findings will be useful in the further development of Vγ9Vδ2 T cell immunotherapy.
Collapse
Affiliation(s)
- Chenxiao Liu
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Sven-Göran Eriksson
- Department of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ingela Parmryd
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.,Department of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
8
|
Activated naïve γδ T cells accelerate deep molecular response to BCR-ABL inhibitors in patients with chronic myeloid leukemia. Blood Cancer J 2021; 11:182. [PMID: 34785653 PMCID: PMC8595379 DOI: 10.1038/s41408-021-00572-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/23/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) that target BCR-ABL are the frontline treatments in chronic myeloid leukemia (CML). Growing evidence has shown that TKIs also enhance immunity. Since gamma-delta T (γδT) cells possess the potent anticancer capability, here we investigated the potential involvement of γδT cells in TKI treatments for CML. We characterized γδT cells isolated from chronic-phase CML patients before and during TKI treatments. γδT expression increased significantly in CML patients who achieved major molecular response (MMR) and deep molecular response (DMR). Their Vδ2 subset of γδT also expanded, and increased expression of activating molecules, namely IFN-γ, perforin, and CD107a, as well as γδT cytotoxicity. Mechanistically, TKIs augmented the efflux of isopentenyl pyrophosphate (IPP) from CML cells, which stimulated IFN-γ production and γδT expansion. Notably, the size of the IFN-γ+ naïve γδT population in TKI-treated CML patients was strongly correlated with their rates to reach DMR and with the duration on DMR. Statistical analysis suggests that a cutoff of 7.5% IFN-γ+ naïve subpopulation of γδT in CML patients could serve as a determinant for MR4.0 sustainability. Our results highlight γδT cells as a positive regulator for TKI responses in CML patients.
Collapse
|
9
|
A mechanism of cooling hot tumors: Lactate attenuates inflammation in dendritic cells. iScience 2021; 24:103067. [PMID: 34541473 PMCID: PMC8441070 DOI: 10.1016/j.isci.2021.103067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/02/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022] Open
Abstract
Turning non-inflamed (cold) tumors into inflamed (hot) tumors is important for maximizing the effect of immune checkpoint inhibitors (ICIs) against malignancies. We showed that lactate, a product of the Warburg effect, inhibited the efficacy of ICIs and suppressed IL-12 p40 expression in dendritic cells (DCs) through reducing NF-κB p65, p50, and c-Rel DNA-binding activity to the IL-12 p40 promoter. Additionally, lactate promoted the expression of early growth response protein 1 (EGR1), whose expression was increased in human invasive melanoma compared with non-invasive melanoma. We also found that EGR1 interacts with serum response factor (SRF) and represses the expression of CD80 in DCs. These findings suggest that lactate and its induced EGR1 are key factors that turn hot tumors into cold tumors and may represent targets in cancer treatment with ICIs. Lactate suppressed IL-12 p40 expression in dendritic cells Lactate promoted the expression of early growth response protein 1 (EGR1) EGR1 interacts with serum response factor (SRF) and represses the expression of CD80 Lactate and EGR1 switch inflamed (hot) tumors to non-inflamed (cold) tumors
Collapse
|
10
|
AuYeung AWK, Mould RC, Stegelmeier AA, van Vloten JP, Karimi K, Woods JP, Petrik JJ, Wood GA, Bridle BW. Mechanisms that allow vaccination against an oncolytic vesicular stomatitis virus-encoded transgene to enhance safety without abrogating oncolysis. Sci Rep 2021; 11:15290. [PMID: 34315959 PMCID: PMC8316323 DOI: 10.1038/s41598-021-94483-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/09/2021] [Indexed: 11/26/2022] Open
Abstract
Vaccination can prevent viral infections via virus-specific T cells, among other mechanisms. A goal of oncolytic virotherapy is replication of oncolytic viruses (OVs) in tumors, so pre-existing T cell immunity against an OV-encoded transgene would seem counterproductive. We developed a treatment for melanomas by pre-vaccinating against an oncolytic vesicular stomatitis virus (VSV)-encoded tumor antigen. Surprisingly, when the VSV-vectored booster vaccine was administered at the peak of the primary effector T cell response, oncolysis was not abrogated. We sought to determine how oncolysis was retained during a robust T cell response against the VSV-encoded transgene product. A murine melanoma model was used to identify two mechanisms that enable this phenomenon. First, tumor-infiltrating T cells had reduced cytopathic potential due to immunosuppression. Second, virus-induced lymphopenia acutely removed virus-specific T cells from tumors. These mechanisms provide a window of opportunity for replication of oncolytic VSV and rationale for a paradigm change in oncolytic virotherapy, whereby immune responses could be intentionally induced against a VSV-encoded melanoma-associated antigen to improve safety without abrogating oncolysis.
Collapse
Affiliation(s)
- Amanda W K AuYeung
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Robert C Mould
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Ashley A Stegelmeier
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Jacob P van Vloten
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Khalil Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - J Paul Woods
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - James J Petrik
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Geoffrey A Wood
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada. .,Department of Pathobiology, Ontario Veterinary College, University of Guelph, Rm. 4834, Bldg. 89, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
11
|
Bassani B, Tripodo C, Portararo P, Gulino A, Botti L, Chiodoni C, Jachetti E, Bolli N, Ciciarello M, Joehrens K, Anagnostopoulos I, Na IK, Curti A, Colombo MP, Sangaletti S. CD40 Activity on Mesenchymal Cells Negatively Regulates OX40L to Maintain Bone Marrow Immune Homeostasis Under Stress Conditions. Front Immunol 2021; 12:662048. [PMID: 34084166 PMCID: PMC8168593 DOI: 10.3389/fimmu.2021.662048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/26/2021] [Indexed: 12/24/2022] Open
Abstract
Background Within the bone marrow (BM), mature T cells are maintained under homeostatic conditions to facilitate proper hematopoietic development. This homeostasis depends upon a peculiar elevated frequency of regulatory T cells (Tregs) and immune regulatory activities from BM-mesenchymal stem cells (BM-MSCs). In response to BM transplantation (BMT), the conditioning regimen exposes the BM to a dramatic induction of inflammatory cytokines and causes an unbalanced T-effector (Teff) and Treg ratio. This imbalance negatively impacts hematopoiesis, particularly in regard to B-cell lymphopoiesis that requires an intact cross-talk between BM-MSCs and Tregs. The mechanisms underlying the ability of BM-MSCs to restore Treg homeostasis and proper B-cell development are currently unknown. Methods We studied the role of host radio-resistant cell-derived CD40 in restoring Teff/Treg homeostasis and proper B-cell development in a murine model of BMT. We characterized the host cellular source of CD40 and performed radiation chimera analyses by transplanting WT or Cd40-KO with WT BM in the presence of T-reg and co-infusing WT or - Cd40-KO BM-MSCs. Residual host and donor T cell expansion and activation (cytokine production) and also the expression of Treg fitness markers and conversion to Th17 were analyzed. The presence of Cd40+ BM-MSCs was analyzed in a human setting in correlation with the frequency of B-cell precursors in patients who underwent HSCT and variably developed acute graft-versus-host (aGVDH) disease. Results CD40 expression is nearly undetectable in the BM, yet a Cd40-KO recipient of WT donor chimera exhibited impaired B-cell lymphopoiesis and Treg development. Lethal irradiation promotes CD40 and OX40L expression in radio-resistant BM-MSCs through the induction of pro-inflammatory cytokines. OX40L favors Teff expansion and activation at the expense of Tregs; however, the expression of CD40 dampens OX40L expression and restores Treg homeostasis, thus facilitating proper B-cell development. Indeed, in contrast to dendritic cells in secondary lymphoid organs that require CD40 triggers to express OX40L, BM-MSCs require CD40 to inhibit OX40L expression. Conclusions CD40+ BM-MSCs are immune regulatory elements within BM. Loss of CD40 results in uncontrolled T cell activation due to a reduced number of Tregs, and B-cell development is consequently impaired. GVHD provides an example of how a loss of CD40+ BM-MSCs and a reduction in B-cell precursors may occur in a human setting.
Collapse
Affiliation(s)
- Barbara Bassani
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | | | - Paola Portararo
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | | | - Laura Botti
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Claudia Chiodoni
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Elena Jachetti
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Niccolò Bolli
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan, Italy
| | - Marilena Ciciarello
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Institute of Hematology "Seràgnoli", Bologna, Italy
| | - Korinna Joehrens
- Charité-Universitätsmedizin Berlin, Institute of Pathology, Berlin, Germany
| | | | - Il-Kang Na
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Antonio Curti
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Institute of Hematology "Seràgnoli", Bologna, Italy
| | - Mario P Colombo
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Sabina Sangaletti
- Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
12
|
CRISPR/Cas9-Mediated GFP Reporter Knock-in in K562 and Raji Cell Lines for Tracking Immune Cell Killing Assay. Methods Mol Biol 2020. [PMID: 33336280 DOI: 10.1007/978-1-0716-0943-9_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
Cell-mediated cytotoxicity plays an important role in several fundamental immunological processes and is crucial for biological evaluation in in vitro studies. In order to determine the immunological activities of the cells, an assay should be safe, reproducible, and cost-effective. Here, we present a simple and cost-effective approach for evaluation of natural killer (NK) cell-mediated cytotoxicity by generating a CRISPR/Cas9-mediated GFP reporter knock-in in the target cell line, K562, and the non-target cell line, Raji, using a plasmid-based transfection method. The GFP+ target cells facilitate tracking of the immune cell killing assay, which avoids the need for multiple cell labeling with fluorescent dyes. Our approach is also applicable to the genome editing of other target cell types for functional analysis of effector cells.
Collapse
|
13
|
Lasso P, Gomez-Cadena A, Urueña C, Donda A, Martinez-Usatorre A, Romero P, Barreto A, Fiorentino S. An Immunomodulatory Gallotanin-Rich Fraction From Caesalpinia spinosa Enhances the Therapeutic Effect of Anti-PD-L1 in Melanoma. Front Immunol 2020; 11:584959. [PMID: 33312174 PMCID: PMC7708328 DOI: 10.3389/fimmu.2020.584959] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/21/2020] [Indexed: 12/23/2022] Open
Abstract
PD-1/PD-L1 pathway plays a role in inhibiting immune response. Therapeutic antibodies aimed at blocking the PD-1/PD-L1 interaction have entered clinical development and have been approved for a variety of cancers. However, the clinical benefits are reduced to a group of patients. The research in combined therapies, which allow for a greater response, is strongly encouraging. We previously characterized a polyphenol-rich extract from Caesalpinia spinosa (P2Et) with antitumor activity in both melanoma and breast carcinoma, as well as immunomodulatory activity. We hypothesize that the combined treatment with P2Et and anti-PD-L1 can improve the antitumor response through an additive antitumor effect. We investigated the antitumor and immunomodulatory activity of P2Et and anti-PD-L1 combined therapy in B16-F10 melanoma and 4T1 breast carcinoma. We analyzed tumor growth, hematologic parameters, T cell counts, cytokine expression, and T cell cytotoxicity. In the melanoma model, combined P2Et and anti-PD-L1 therapy has the following effects: decrease in tumor size; increase in the number of activated CD4+ and CD8+ T cells; decrease in the number of suppressor myeloid cells; increase in PD-L1 expression; decrease in the frequency of CD8+ T cell expressing PD-1; improvement in the cytotoxic activity of T cells; and increase in the IFN γ secretion. In the breast cancer model, P2Et and PD-L1 alone or in combination show antitumor effect with no clear additive effect. This study shows that combined therapy of P2Et and anti-PD-L1 can improve antitumor response in a melanoma model by activating the immune response and neutralizing immunosuppressive mechanisms.
Collapse
Affiliation(s)
- Paola Lasso
- Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Alejandra Gomez-Cadena
- Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá, Colombia.,University of Geneva, Department of Pathology and Immunology, Geneva, Switzerland
| | - Claudia Urueña
- Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Alena Donda
- University of Lausanne, Department of Fundamental Oncology, Lausanne, Switzerland
| | - Amaia Martinez-Usatorre
- Swiss Federal Institute of Technology Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
| | - Pedro Romero
- University of Lausanne, Department of Fundamental Oncology, Lausanne, Switzerland
| | - Alfonso Barreto
- Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Susana Fiorentino
- Grupo de Inmunobiología y Biología Celular, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
14
|
Wu X, Zhang Y, Li Y, Schmidt‐Wolf IGH. Improvements in Flow Cytometry‐Based Cytotoxicity Assay. Cytometry A 2020; 99:680-688. [DOI: 10.1002/cyto.a.24242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/23/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Xiaolong Wu
- Department of Integrated Oncology, CIO Bonn University Hospital Bonn Bonn D‐53105 Germany
| | - Ying Zhang
- Department of Integrated Oncology, CIO Bonn University Hospital Bonn Bonn D‐53105 Germany
| | - Yutao Li
- Department of Integrated Oncology, CIO Bonn University Hospital Bonn Bonn D‐53105 Germany
| | | |
Collapse
|
15
|
Baruch EN, Ortenberg R, Avivi C, Anafi L, Dick-Necula D, Stossel C, Moshkovits Y, Itzhaki O, Besser MJ, Schachter J, Barshack I, Markel G. Immune co-culture cell microarray - a feasible tool for high-throughput functional investigation of lymphocyte-cancer interactions. Oncoimmunology 2020; 9:1741267. [PMID: 32373399 PMCID: PMC7194292 DOI: 10.1080/2162402x.2020.1741267] [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: 09/28/2019] [Revised: 01/09/2020] [Accepted: 01/09/2020] [Indexed: 12/05/2022] Open
Abstract
Omics analyses often result in dozens to hundreds of potential targets, requiring validation for their biological relevance. Current high-throughput functional investigation methods are frequently labor-intensive, expensive, and display low reproducibility. The Immune Co-Culture Cell Microarray (ICCM) is a formalin-fixed paraffin-embedded cell block microarray based on co-cultures of patient-derived tumor-infiltrating lymphocytes and their autologous melanoma cells. Each ICCM slide represents the same experiment and can be stained using standard immunohistochemistry and immunofluorescence techniques. Functional dynamics assessment of both proteins and microRNAs using ICCM stained slides demonstrated similar findings to flow cytometry assays and to previously published patient-derived biopsy reports.
Collapse
Affiliation(s)
- Erez Nissim Baruch
- Department of Clinical Immunology and Microbiology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Rona Ortenberg
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Camila Avivi
- Pathological Institute, Sheba Medical Center, Tel-HaShomer; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Liat Anafi
- Pathological Institute, Sheba Medical Center, Tel-HaShomer; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daniela Dick-Necula
- Pathological Institute, Sheba Medical Center, Tel-HaShomer; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chani Stossel
- Department of Clinical Immunology and Microbiology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Yonatan Moshkovits
- School of Of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orit Itzhaki
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Michal Judith Besser
- Department of Clinical Immunology and Microbiology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
| | - Jacob Schachter
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
- School of Of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Iris Barshack
- Pathological Institute, Sheba Medical Center, Tel-HaShomer; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- School of Of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gal Markel
- Department of Clinical Immunology and Microbiology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Tel-HaShomer, Israel
- Talpiot Medical Leadership Program, Sheba Medical Center, Tel HaShomer, Israel
| |
Collapse
|
16
|
|
17
|
Lasso P, Llano Murcia M, Sandoval TA, Urueña C, Barreto A, Fiorentino S. Breast Tumor Cells Highly Resistant to Drugs Are Controlled Only by the Immune Response Induced in an Immunocompetent Mouse Model. Integr Cancer Ther 2019; 18:1534735419848047. [PMID: 31056957 PMCID: PMC6505237 DOI: 10.1177/1534735419848047] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background: The tumor cells responsible for metastasis are highly
resistant to chemotherapy and have characteristics of stem cells, with a high
capacity for self-regeneration and the use of detoxifying mechanisms that
participate in drug resistance. In vivo models of highly resistant cells allow
us to evaluate the real impact of the immune response in the control of cancer.
Materials and Methods: A tumor population derived from the 4T1
breast cancer cell line that was stable in vitro and highly aggressive in vivo
was obtained, characterized, and determined to exhibit cancer stem cell (CSC)
phenotypes (CD44+, CD24+, ALDH+,
Oct4+, Nanog+, Sox2+, and high self-renewal
capacity). Orthotopic transplantation of these cells allowed us to evaluate
their in vivo susceptibility to chemo and immune responses induced after
vaccination. Results: The immune response induced after vaccination
with tumor cells treated with doxorubicin decreased the formation of tumors and
macrometastasis in this model, which allowed us to confirm the immune response
relevance in the control of highly chemotherapy-resistant ALDH+ CSCs
in an aggressive tumor model in immunocompetent animals.
Conclusions: The antitumor immune response was the main element
capable of controlling tumor progression as well as metastasis in a highly
chemotherapy-resistant aggressive breast cancer model.
Collapse
Affiliation(s)
- Paola Lasso
- 1 Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | | | | | | | | |
Collapse
|
18
|
Bai L, Peng H, Hao X, Tang L, Sun C, Zheng M, Liu F, Lian Z, Bai L, Wei H, Sun R, Tian Z. CD8 + T Cells Promote Maturation of Liver-Resident NK Cells Through the CD70-CD27 axis. Hepatology 2019; 70:1804-1815. [PMID: 31077406 DOI: 10.1002/hep.30757] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 05/08/2019] [Indexed: 01/12/2023]
Abstract
Liver-resident natural killer (LrNK) cells are a unique subset of NK cells that are distinct from conventional NK cells. However, little is known about the mechanisms by which LrNK cells mature. In this study, we discovered that LrNK cells exhibit a relatively immature phenotype and impaired cytotoxic capacity in the absence of CD8+ T cells. The provision of CD8+ T cells to Cd8-/- or Rag1-/- mice led to the restoration of LrNK cell maturation. Furthermore, co-culture with CD8+ T cells induced immature CD27+ LrNK cells to convert into mature CD27- LrNK cells, whereas blocking the interaction of CD70 and CD27 abrogated the ability of CD8+ T cells to promote the maturation of LrNK cells. Conclusion: Our findings indicate that CD8+ T cells promote the maturation of LrNK cells through the CD70-CD27 axis, and therefore highlight a previously unknown mechanism responsible for the mediation of LrNK cell maturation.
Collapse
Affiliation(s)
- Lu Bai
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Institue of Immunology, University of Science and Technology of China, Hefei, Anhui, China
| | - Hui Peng
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Institue of Immunology, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaolei Hao
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Institue of Immunology, University of Science and Technology of China, Hefei, Anhui, China
| | - Ling Tang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Institue of Immunology, University of Science and Technology of China, Hefei, Anhui, China
| | - Cheng Sun
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Institue of Immunology, University of Science and Technology of China, Hefei, Anhui, China.,Organ Transplant Center & Immunology Laboratory, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Meijuan Zheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fubao Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhexiong Lian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Institue of Immunology, University of Science and Technology of China, Hefei, Anhui, China
| | - Li Bai
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Institue of Immunology, University of Science and Technology of China, Hefei, Anhui, China
| | - Haiming Wei
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Institue of Immunology, University of Science and Technology of China, Hefei, Anhui, China
| | - Rui Sun
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Institue of Immunology, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Institue of Immunology, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
19
|
Zhang P, Zhao S, Wu C, Li J, Li Z, Wen C, Hu S, An G, Meng H, Zhang X, Yang L. Effects of CSF1R-targeted chimeric antigen receptor-modified NK92MI & T cells on tumor-associated macrophages. Immunotherapy 2019; 10:935-949. [PMID: 30149762 DOI: 10.2217/imt-2018-0012] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tumor immunotherapy has shown great progress for the treatment of cancer; however, both endogenous and exogenous T cells are inhibited by the immunosuppressive tumor microenvironment. Tumor-associated macrophages (TAMs) in the microenvironment play pivotal and complex roles in tumor development and progression. Macrophages are categorized as M1 and M2 types. Relevant studies suggest that M2 TAMs correlate with poor prognosis. Colony-stimulating factor 1 receptor (CSF1R) controls the formation, differentiation and function of M2 macrophages, which helps tumors grow, metastasize and secrete immunosuppressive cytokines. The objectives of this study were to establish two types of third-generation chimeric antigen receptors (CARs) that could specifically target human CSF1R, and to introduce the CARs into NK92MI cells and normal human peripheral blood T cells through lentiviral transduction to produce CAR-natural killer (NK) and -T cells. We then tested their cytotoxicity against cell lines and peripheral blood monocytes expressing CSF1R. In vitro experiments confirmed that third-generation CARs had good target specificity and cytotoxicity. It was expected that CAR-NK and -T cells could specifically kill M2 TAMs in the tumor microenvironment and remove their inhibitory effect. Therefore, CSF1R-targeting CAR-NK and -T cells could represent a novel cellular immunotherapy strategy in conjunction with other antibody-based drugs and targeted therapeutics.
Collapse
Affiliation(s)
- Ping Zhang
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, China
| | - Songbo Zhao
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, China
| | - Chao Wu
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, China
| | - Jialu Li
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, China
| | - Zixuan Li
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, China
| | - Chunmei Wen
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, China
| | - Siyi Hu
- Anhui Anke Biotechnology (Group) Co., Ltd, Hefei, Anhui, China
| | - Gangli An
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, China
| | - Huimin Meng
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, China
| | - Xingding Zhang
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lin Yang
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China.,State Key Laboratory of Radiation Medicine & Protection, Soochow University, Suzhou, Jiangsu, China.,Persongen Bio Therapeutics (Suzhou) Co., Ltd, Suzhou, Jiangsu, China
| |
Collapse
|
20
|
Chávez-Galán L, Illescas-Eugenio J, Alvarez-Sekely M, Baez-Saldaña R, Chávez R, Lascurain R. Tuberculosis patients display a high proportion of CD8 + T cells with a high cytotoxic potential. Microbiol Immunol 2019; 63:316-327. [PMID: 31254409 PMCID: PMC6772019 DOI: 10.1111/1348-0421.12724] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 06/10/2019] [Accepted: 06/23/2019] [Indexed: 01/23/2023]
Abstract
Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis (Mtb) and remains a major cause of morbidity and mortality worldwide. In the host's immune response system, T cells play a critical role in mediating protection against Mtb infection, but the role of CD8+ T cells is still controversial. We evaluated the phenotypical characterization and cytotoxic ability of CD8+ T cells by flow cytometry‐based assay. Cytokine levels in serum were measured by multiplex cytokine assay. Our data show that cells from TB patients have an increased percentage of peripheral blood CD8+αβ+ T (p = 0.02) and CD56+CD8+ T (p = 0.02) and a decreased frequency of NKG2D+CD8+ T (p = 0.02) compared with healthy donors. Unlike CD8+ T cells from healthy donors, CD8+ T cells from TB patients exhibit greater cytotoxicity, mediated by HLA class I molecules, on autologous monocytes in the presence of mycobacterial antigens (p = 0.005). Finally, TB patients have a proinflammatory profile characterized by serum high level of TNF‐α (p = 0.02) and IL‐8 (p = 0.0001), but, interestingly, IL‐4 (p = 0.002) was also increased compared with healthy donors. Our data show evidence regarding the highly cytotoxic status of CD8+ T cells in Mtb infection. These cytotoxic cells restricted to HLA‐A, B, and C could be used to optimize strategies for designing new TB vaccines or for identifying markers of disease progression.
Collapse
Affiliation(s)
- Leslie Chávez-Galán
- Integrative Immunology Laboratory, National Institute of Respiratory Diseases "Ismael Cosío Villegas" (INER), Mexico City, Mexico
| | | | - Magaly Alvarez-Sekely
- Department of Hematology, National Institute of Cancerology (INCAN), Mexico City, Mexico
| | - Renata Baez-Saldaña
- Oncologic Pulmonology Clinic, National Institute of Respiratory Diseases "Ismael Cosío Villegas" (INER), Mexico City, Mexico
| | - Raúl Chávez
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Ricardo Lascurain
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico (UNAM), Mexico City, Mexico.,Homeopatic National Hospital, Chimalpopoca135, 06800, Mexico City, Mexico
| |
Collapse
|
21
|
Qin Y, Lee Y, Seo J, Kim T, Shin JH, Park SH. NIH3T3 Directs Memory-Fated CTL Programming and Represses High Expression of PD-1 on Antitumor CTLs. Front Immunol 2019; 10:761. [PMID: 31031760 PMCID: PMC6470252 DOI: 10.3389/fimmu.2019.00761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/21/2019] [Indexed: 11/15/2022] Open
Abstract
Memory CD8+ T cells have long been considered a promising population for adoptive cell therapy (ACT) due to their long-term persistence and robust re-stimulatory response. NIH3T3 is an immortalized mouse embryonic fibroblast cell line. We report that NIH3T3-conditioned medium (CM) can augment effector functions of CTLs following antigen priming and confer phenotypic and transcriptional properties of central memory cells. After NIH3T3-CM-educated CTLs were infused into naïve mice, they predominantly developed to central memory cells. A large number of NIH3T3-CM-educated CTLs with high functionality persisted and infiltrated to tumor mass. In addition, NIH3T3-CM inhibited CTLs expression of PD-1 in vitro and repressed their high expression of PD-1 in tumor microenvironment after adoptive transfer. Consequently, established tumor models showed that infusion of NIH3T3-CM-educated CTLs dramatically improved CTL mediated-antitumor immunity. Furthermore, NIH3T3-CM also promoted human CD8+ T cells differentiation into memory cells. These results suggest that NIH3T3-CM-programmed CTLs are good candidates for adoptive transfer in tumor therapy.
Collapse
Affiliation(s)
- Yingyu Qin
- Department of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Yuna Lee
- ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Jaeho Seo
- ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Taehyun Kim
- ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Jung Hoon Shin
- ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Se-Ho Park
- Department of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| |
Collapse
|
22
|
Chang MC, Cheng HI, Hsu K, Hsu YN, Kao CW, Chang YF, Lim KH, Chen CG. NKG2A Down-Regulation by Dasatinib Enhances Natural Killer Cytotoxicity and Accelerates Effective Treatment Responses in Patients With Chronic Myeloid Leukemia. Front Immunol 2019; 9:3152. [PMID: 30705677 PMCID: PMC6344416 DOI: 10.3389/fimmu.2018.03152] [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: 08/27/2018] [Accepted: 12/20/2018] [Indexed: 12/20/2022] Open
Abstract
Chronic myeloid leukemia (CML) is a hematological malignancy characterized by the presence of t(9;22) chromosomal translocation that results in BCR-ABL fusion gene. ABL tyrosine kinase inhibitors (TKIs), such as imatinib, nilotinib, and dasatinib, are currently the front-line treatment options for CML. Recently, natural killer (NK) cell activation and expansion have been shown to be associated with optimal treatment responses for CML. To investigate the effects and mechanisms of these TKIs on NK cells, here we characterized activating and inhibitory NK receptors in CD3-CD16+CD56dim NK cells isolated from CML patients in chronic phase (CP). The expressions of activating NK receptors, such as NKG2D, natural cytotoxicity receptor (NCR) and DNAM-1, rebounded after successful TKI treatments for CML. In contrast, among the three surveyed inhibitory receptors (NKG2A, KIR2DL1, and KIR3DL1), only the expression of NKG2A was reverted and suppressed to a very low level by dasatinib, and not by imatinib or nilotinib. CML patients treated with dasatinib indeed expressed fewer NKG2A+ NK cells, which send negative signals for induction of NK cytotoxicity. For these dasatinib-treated patients, the duration to reach major molecular response (MMR) was shorter, and significantly correlated with individual's NKG2A+ NK cell number. This clinical relevance to NKG2A was not observed in treatments with imatinib or nilotinib. In line with dasatinib-specific down-regulation of NKG2A, NK cytotoxicity evaluated by the killing assay was also significantly higher in patients treated with dasatinib than in those treated with imatinib or nilotinib. The lower NK cytotoxicity from imatinib or nilotinib treatments could be reverted by NKG2A blockade using anti-NKG2A antibody. Further in vitro experiments revealed mechanistically that dasatinib could inactivate p38 mitogen-activated protein kinase (MAPK), and consequently affect nuclear import of GATA-3 and GATA-3 transcriptional activities for NKG2A. Our results highlight the dual effects of dasatinib in direct inhibition of ABL kinase and in immunomodulation through NKG2A down-regulation, contributing to accelerated molecular responses (MR) in CML.
Collapse
Affiliation(s)
- Ming-Chin Chang
- Department of Hematology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei, Taiwan
| | - Hung-I Cheng
- Department of Hematology, MacKay Memorial Hospital, Hsin-Chu, Taiwan
| | - Kate Hsu
- Department of Medical Research, Transfusion Medicine & Immunogenetics Laboratories, MacKay Memorial Hospital, Tamsui, Taiwan
| | - Yen-Ning Hsu
- Department of Hematology, MacKay Memorial Hospital, Hsin-Chu, Taiwan
| | - Chen-Wei Kao
- GCRC Laboratory, Department of Hematology, MacKay Memorial Hospital, New Taipei, Taiwan
| | - Yi-Fang Chang
- Department of Hematology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei, Taiwan.,GCRC Laboratory, Department of Hematology, MacKay Memorial Hospital, New Taipei, Taiwan
| | - Ken-Hong Lim
- Department of Hematology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei, Taiwan.,GCRC Laboratory, Department of Hematology, MacKay Memorial Hospital, New Taipei, Taiwan
| | - Caleb Gonshen Chen
- Department of Hematology, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei, Taiwan.,GCRC Laboratory, Department of Hematology, MacKay Memorial Hospital, New Taipei, Taiwan
| |
Collapse
|
23
|
Hoshino A, Yang X, Tanita K, Yoshida K, Ono T, Nishida N, Okuno Y, Kanzaki T, Goi K, Fujino H, Ohshima K, Shiraishi Y, Chiba K, Tanaka H, Miyano S, Ogawa S, Kojima S, Morio T, Kanegane H. Modification of cellular and humoral immunity by somatically reverted T cells in X-linked lymphoproliferative syndrome type 1. J Allergy Clin Immunol 2019; 143:421-424.e11. [DOI: 10.1016/j.jaci.2018.07.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 07/09/2018] [Accepted: 07/13/2018] [Indexed: 01/25/2023]
|
24
|
Lee SW, Park HJ, Cheon JH, Wu L, Van Kaer L, Hong S. iNKT Cells Suppress Pathogenic NK1.1 +CD8 + T Cells in DSS-Induced Colitis. Front Immunol 2018; 9:2168. [PMID: 30333822 PMCID: PMC6176072 DOI: 10.3389/fimmu.2018.02168] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/03/2018] [Indexed: 12/21/2022] Open
Abstract
T cells producing IFNγ play a pathogenic role in the development of inflammatory bowel disease (IBD). To investigate the functions of CD1d-dependent invariant natural killer T (iNKT) cells in experimental colitis induced in Yeti mice with dysregulated expression of IFNγ, we generated iNKT cell-deficient Yeti/CD1d KO mice and compared colitis among WT, CD1d KO, Yeti, and Yeti/CD1d KO mice following DSS treatment. We found that deficiency of iNKT cells exacerbated colitis and disease pathogenesis was mainly mediated by NK1.1+CD8+ T cells. Furthermore, the protective effects of iNKT cells correlated with up-regulation of regulatory T cells. Taken together, our results have demonstrated that CD1d-dependent iNKT cells and CD1d-independent NK1.1+CD8+ T cells reciprocally regulate the development of intestinal inflammatory responses mediated by IFNγ-dysregulation. These findings also identify NK1.1+CD8+ T cells as novel target cells for the development of therapeutics for human IBD.
Collapse
Affiliation(s)
- Sung Won Lee
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, South Korea
| | - Hyun Jung Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, South Korea
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Lan Wu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Seokmann Hong
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, South Korea
| |
Collapse
|
25
|
Kim SH, Lee SW, Park HJ, Lee SH, Im WK, Kim YD, Kim KH, Park SJ, Hong S, Jeon SH. Anti-cancer activity of Angelica gigas by increasing immune response and stimulating natural killer and natural killer T cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:218. [PMID: 30021579 PMCID: PMC6052638 DOI: 10.1186/s12906-018-2277-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 07/02/2018] [Indexed: 12/29/2022]
Abstract
Background The polysaccharide component of Angelica gigas induces immuno-stimulatory effects on innate immune cells. However, it is unclear whether A. gigas’ adjuvant activity on the immune system can elicit anti-cancer responses. Methods A water-soluble immuno-stimulatory component of A. gigas was prepared. How this ISAg modulated the activation of innate immune cells such as dendritic cells (DCs) was examined. ISAg-induced cytotoxic activity via natural killer (NK) and NKT cells was also tested using a tumor-bearing mouse model. Results ISAg treatment induced nitric oxide (NO) production and cytokine gene expression involved in innate immune responses. ISAg activated macrophages and DCs to secrete cytokine IL-12, through the TLR4 signaling pathway. IL-12 plays a crucial role in ISAg-mediated NK and NKT cell activation. Thus, the anti-cancer activity of NK and NKT cells induced ISAg-mediated cytotoxicity of B16 melanoma cells in mice. Conclusions These results indicated that the natural ingredient, ISAg, has adjuvant activity to induce strong anti-cancer activity of NK and NKT cells in vivo.
Collapse
|
26
|
Edri A, Shemesh A, Iraqi M, Matalon O, Brusilovsky M, Hadad U, Radinsky O, Gershoni-Yahalom O, Dye JM, Mandelboim O, Barda-Saad M, Lobel L, Porgador A. The Ebola-Glycoprotein Modulates the Function of Natural Killer Cells. Front Immunol 2018; 9:1428. [PMID: 30013549 PMCID: PMC6036185 DOI: 10.3389/fimmu.2018.01428] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/08/2018] [Indexed: 12/23/2022] Open
Abstract
The Ebola virus (EBOV) uses evasion mechanisms that directly interfere with host T-cell antiviral responses. By steric shielding of human leukocyte antigen class-1, the Ebola glycoprotein (GP) blocks interaction with T-cell receptors (TCRs), thus rendering T cells unable to attack virus-infected cells. It is likely that this mechanism could promote increased natural killer (NK) cell activity against GP-expressing cells by preventing the engagement of NK inhibitory receptors; however, we found that primary human NK cells were less reactive to GP-expressing HEK293T cells. This was manifested as reduced cytokine secretion, a reduction in NK degranulation, and decreased lysis of GP-expressing target cells. We also demonstrated reduced recognition of GP-expressing cells by recombinant NKG2D and NKp30 receptors. In accordance, we showed a reduced monoclonal antibody-based staining of NKG2D and NKp30 ligands on GP-expressing target cells. Trypsin digestion of the membrane-associated GP led to a recovery of the recognition of membrane-associated NKG2D and NKp30 ligands. We further showed that membrane-associated GP did not shield recognition by KIR2DL receptors; in accordance, GP expression by target cells significantly perturbed signal transduction through activating, but not through inhibitory, receptors. Our results suggest a novel evasion mechanism employed by the EBOV to specifically avoid the NK cell immune response.
Collapse
Affiliation(s)
- Avishay Edri
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Avishai Shemesh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Muhammed Iraqi
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Omri Matalon
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Michael Brusilovsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Uzi Hadad
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Olga Radinsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Orly Gershoni-Yahalom
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - John M Dye
- U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Ofer Mandelboim
- The Lautenberg Center for General and Tumor Immunology, The BioMedical Research Institute Israel Canada of the Faculty of Medicine (IMRIC), The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Mira Barda-Saad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Leslie Lobel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Department of Emerging and Reemerging Diseases and Special Pathogens Uganda Virus Research Institute (UVRI), Entebbe, Uganda
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
27
|
Desimio MG, Giuliani E, Ferraro AS, Adorno G, Doria M. In Vitro Exposure to Prostratin but Not Bryostatin-1 Improves Natural Killer Cell Functions Including Killing of CD4 + T Cells Harboring Reactivated Human Immunodeficiency Virus. Front Immunol 2018; 9:1514. [PMID: 30008723 PMCID: PMC6033996 DOI: 10.3389/fimmu.2018.01514] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 06/19/2018] [Indexed: 01/04/2023] Open
Abstract
In the attempt of purging the HIV-1 reservoir through the “shock-and-kill” strategy, it is important to select latency-reversing agents (LRAs) devoid of deleterious effects on the antiviral function of immune effector cells. Here, we investigated two LRAs with PKC agonist activity, prostratin (PRO) and bryostatin-1 (BRY), for their impact on the function of natural killer (NK) cells, the major effectors of innate immunity whose potential in HIV-1 eradication has emerged in recent clinical trials. Using NK cells of healthy donors, we found that exposure to either PRO or BRY potently activated NK cells, resulting in upmodulation of NKG2D and NKp44 activating receptors and matrix metalloprotease-mediated shedding of CD16 receptor. Despite PRO and BRY affected NK cell phenotype in the same manner, their impact on NK cell function was diverse and showed considerable donor-to-donor variation. Altogether, in most tested donors, the natural cytotoxicity and antibody-dependent cellular cytotoxicity (ADCC) of NK cells were either improved or maintained by PRO, while both activities were impaired by BRY. Moreover, we analyzed the effect of these drugs on the capacity of treated NK cells to kill autologous latently infected CD4+ T cells reactivated via the same treatment. First, we found that PRO but not BRY increased upmodulation of the ULBP2 ligand for NKG2D on reactivated p24+ cells. Importantly, we showed that clearance of reactivated p24+ cells by NK cells was enhanced when both targets and effectors were exposed to PRO but not to BRY. Overall, PRO had a superior potential compared with BRY as to the impact on key NK cell functions and on NK-cell-mediated clearance of the HIV-1 reservoir. Our results emphasize the importance of considering the effects on NK cells of candidate “shock-and-kill” interventions. With respect to combinative approaches, the impact on NK cells of each LRA should be re-evaluated upon combination with a second LRA, which may have analogous or opposite effects, or with immunotherapy targeting NK cells. In addition, avoiding co-administration of LRAs that negatively impact ADCC activity by NK cells might be essential for successful application of antibodies or vaccination to “shock-and-kill” strategies.
Collapse
Affiliation(s)
- Maria Giovanna Desimio
- Laboratory of Immunoinfectivology, Immune and Infectious Diseases Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Erica Giuliani
- Laboratory of Immunoinfectivology, Immune and Infectious Diseases Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Gaspare Adorno
- SIMT, Policlinico Tor Vergata, Rome, Italy.,Department of Biomedicine and Prevention, Università degli Studi di Roma Tor Vergata, Rome, Italy
| | - Margherita Doria
- Laboratory of Immunoinfectivology, Immune and Infectious Diseases Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
28
|
Qin Y, Shin JH, Yoon JH, Park SH. Embryonic Fibroblasts Promote Antitumor Cytotoxic Effects of CD8 + T Cells. Front Immunol 2018; 9:685. [PMID: 29706956 PMCID: PMC5908885 DOI: 10.3389/fimmu.2018.00685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/20/2018] [Indexed: 12/31/2022] Open
Abstract
Adoptive CD8+ T cell therapy has emerged as an important modality for the treatment of cancers. However, the significant drawback of transfused T cells is their poor survival and functionality in response to tumors. To overcome this limitation, an important consideration is exploring a culture condition to generate superior antitumor cytotoxic T lymphocytes (CTLs) for adoptive therapy. Here, we provide a novel approach to generate potent CTL clones in mouse embryonic fibroblast-conditioned medium (MEF-CM). We found CTLs derived with MEF-CM have higher potential in long-term persistence in tumor bearing and non-tumor-bearing mice. Importantly, adoptive transfer of MEF-CM-cultured CTLs dramatically regressed tumor growth and prolonged mice survival. Characterization of MEF-CM-cultured CTLs (effector molecules, phenotypes, and transcription factors) suggests that MEF-CM enhances the effector functions of CD8+ T cells in part by the upregulation of the T-box transcription factor eomesodermin. Consequently, MEF-CM enhances the intrinsic qualities of effector CD8+ T cells to augment antitumor immunity.
Collapse
Affiliation(s)
- Yingyu Qin
- Department of Life Sciences, Korea University, Seoul, South Korea.,ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Jung Hoon Shin
- ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Jeong-Ho Yoon
- ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Se-Ho Park
- Department of Life Sciences, Korea University, Seoul, South Korea
| |
Collapse
|
29
|
Zhang Y, Wang J, Wu D, Li M, Zhao F, Ren M, Cai Y, Dou J. IL-21-secreting hUCMSCs combined with miR-200c inhibit tumor growth and metastasis via repression of Wnt/β-catenin signaling and epithelial-mesenchymal transition in epithelial ovarian cancer. Onco Targets Ther 2018; 11:2037-2050. [PMID: 29692616 PMCID: PMC5901132 DOI: 10.2147/ott.s147855] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Epithelial ovarian cancer (EOC) with insidious characteristic manifests no symptoms in its early onset but most patients have advanced and distant cancer metastasis at diagnosis. Innovative early diagnosis and effective treatment of EOC are urgently needed. Methods In the study, we developed a novel agent of IL-21-secreting human umbilical cord mesenchymal stem cells (hUCMSCs) combined with miR-200c to evaluate its effects on SKOV3 EOC in vitro and in vivo. Results hUCMSCs-LV-IL-21 combined with miR-200c significantly inhibited the SKOV3 cell mobility and tumorigenesis compared with hUCMSCs-LV-IL-21, hUCMSCs-LV-vector, and hUCMSCs, respectively. These were reflected in decreasing the tumor sizes and elongating the tumor bearing nude mouse survival, accompanied with increasing the serum cytokine levels of IFN-γ, IL-21 and TNF-α as well as the splenocyte cytotoxicity. In addition, the expression of β-catenin, cyclin-D1, Gli1, Gli2, and ZEB1 was decreased but the E-cadherin expression was increased in tumor tissues of mice treated with hUCMSCs-LV-IL-21 plus miR-200c. Conclusion We demonstrated that the synergistic effect of fighting SKOV3 EOC is attributable to repression of Wnt/β-catenin signaling and epithelial-mesenchymal transition in SKOV3 EOC. The findings may provide a new strategy for therapy of EOC.
Collapse
Affiliation(s)
- Yunxia Zhang
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, People's Republic of China.,Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Jing Wang
- Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Di Wu
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Miao Li
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Fenshu Zhao
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Mulan Ren
- Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Yunlong Cai
- Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Jun Dou
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
30
|
de Wolf C, van de Bovenkamp M, Hoefnagel M. Regulatory perspective on in vitro potency assays for human T cells used in anti-tumor immunotherapy. Cytotherapy 2018; 20:601-622. [PMID: 29598903 DOI: 10.1016/j.jcyt.2018.01.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/25/2018] [Accepted: 01/27/2018] [Indexed: 02/06/2023]
Abstract
The adaptive immune system is known to play an important role in anti-neoplastic responses via induction of several effector pathways, resulting in tumor cell death. Because of their ability to specifically recognize and kill tumor cells, the potential use of autologous tumor-derived and genetically engineered T cells as adoptive immunotherapy for cancer is currently being explored. Because of the variety of potential T cell-based medicinal products at the level of starting material and manufacturing process, product-specific functionality assays are needed to ensure quality for individual products. In this review, we provide an overview of in vitro potency assays suggested for characterization and release of different T cell-based anti-tumor products. We discuss functional assays, as presented in scientific advices and literature, highlighting specific advantages and limitations of the various assays. Because the anticipated in vivo mechanism of action for anti-tumor T cells involves tumor recognition and cell death, in vitro potency assays based on the cytotoxic potential of antigen-specific T cells are most evident. However, assays based on other T cell properties may be appropriate as surrogates for cytotoxicity. For all proposed assays, biological relevance of the tests and correlation of the read-outs with in vivo functionality need to be substantiated with sufficient product-specific (non-)clinical data. Moreover, further unraveling the complex interaction of immune cells with and within the tumor environment is expected to lead to further improvement of the T cell-based products. Consequently, increased knowledge will allow further optimized guidance for potency assay development.
Collapse
Affiliation(s)
- Charlotte de Wolf
- Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands; Department of Infectious Diseases and Immunology, Utrecht University, The Netherlands
| | | | | |
Collapse
|
31
|
Gene-modified NK-92MI cells expressing a chimeric CD16-BB-ζ or CD64-BB-ζ receptor exhibit enhanced cancer-killing ability in combination with therapeutic antibody. Oncotarget 2018; 8:37128-37139. [PMID: 28415754 PMCID: PMC5514896 DOI: 10.18632/oncotarget.16201] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 03/04/2017] [Indexed: 12/27/2022] Open
Abstract
Natural killer (NK) cells play a pivotal role in monoclonal antibody-mediated immunotherapy through the antibody-dependent cell-mediated cytotoxicity (ADCC) mechanism. NK-92MI is an interleukin-2 (IL-2)-independent cell line, which was derived from NK-92 cells with superior cytotoxicity toward a wide range of tumor cells in vitro and in vivo. Nonetheless, the Fc-receptor (CD16) that usually mediates ADCC is absent in NK-92 and NK-92MI cells. To apply NK-92MI cell-based immunotherapy to cancer treatment, we designed and generated two chimeric receptors in NK-92MI cells that can bind the Fc portion of human immunoglobulins. The construct includes the low-affinity Fc receptor CD16 (158F) or the high-affinity Fc receptor CD64, with the addition of the CD8a extracellular domain, CD28 transmembrane domains, two costimulatory domains (CD28 and 4-1BB), and the signaling domain from CD3ζ. The resulting chimeric receptors, termed CD16-BB-ζ and CD64-BB-ζ, were used to generate modified NK-92MI cells expressing the chimeric receptor, which were named NK-92MIhCD16 and NK-92MIhCD64 cells, respectively. We found that NK-92MIhCD16 and NK-92MIhCD64 cells significantly improved cytotoxicity against CD20-positive non-Hodgkin's lymphoma cells in the presence of rituximab. These results suggest that the chimeric receptor-expressing NK-92MI cells may enhance the clinical responses to currently available anticancer monoclonal antibodies.
Collapse
|
32
|
Nonstructural proteins nsp2TF and nsp2N of porcine reproductive and respiratory syndrome virus (PRRSV) play important roles in suppressing host innate immune responses. Virology 2018; 517:164-176. [PMID: 29325778 PMCID: PMC5884420 DOI: 10.1016/j.virol.2017.12.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 12/24/2022]
Abstract
Recently, we identified a unique -2/-1 ribosomal frameshift mechanism in PRRSV, which yields two truncated forms of nonstructural protein (nsp) 2 variants, nsp2TF and nsp2N. Here, in vitro expression of individual PRRSV nsp2TF and nsp2N demonstrated their ability to suppress cellular innate immune responses in transfected cells. Two recombinant viruses were further analyzed, in which either nsp2TF was C-terminally truncated (vKO1) or expression of both nsp2TF and nsp2N was knocked out (vKO2). Host cellular mRNA profiling showed that a panel of cellular immune genes, in particular those involved in innate immunity, was upregulated in cells infected with vKO1 and vKO2. Compared to the wild-type virus, vKO1 and vKO2 expedited the IFN-α response and increased NK cell cytotoxicity, and subsequently enhanced T cell immune responses in infected pigs. Our data strongly implicate nsp2TF/nsp2N in arteriviral immune evasion and demonstrate that nsp2TF/nsp2N-deficient PRRSV is less capable of counteracting host innate immune responses. In vitro expression of PRRSV nsp2TF/nsp2N affects cellular innate immune response. Nsp2TF/nsp2N suppressed immune genes expression in PRRSV-infected cells. Nsp2TF/nsp2N-deficient mutants showed attenuated growth in vivo. Nsp2TF/nsp2N-deficient mutants induced augmented immune responses in infected pigs.
Collapse
|
33
|
Chheda ZS, Kohanbash G, Okada K, Jahan N, Sidney J, Pecoraro M, Yang X, Carrera DA, Downey KM, Shrivastav S, Liu S, Lin Y, Lagisetti C, Chuntova P, Watchmaker PB, Mueller S, Pollack IF, Rajalingam R, Carcaboso AM, Mann M, Sette A, Garcia KC, Hou Y, Okada H. Novel and shared neoantigen derived from histone 3 variant H3.3K27M mutation for glioma T cell therapy. J Exp Med 2017; 215:141-157. [PMID: 29203539 PMCID: PMC5748856 DOI: 10.1084/jem.20171046] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/01/2017] [Accepted: 10/23/2017] [Indexed: 12/11/2022] Open
Abstract
The median overall survival for children with diffuse intrinsic pontine glioma (DIPG) is less than one year. The majority of diffuse midline gliomas, including more than 70% of DIPGs, harbor an amino acid substitution from lysine (K) to methionine (M) at position 27 of histone 3 variant 3 (H3.3). From a CD8+ T cell clone established by stimulation of HLA-A2+ CD8+ T cells with synthetic peptide encompassing the H3.3K27M mutation, complementary DNA for T cell receptor (TCR) α- and β-chains were cloned into a retroviral vector. TCR-transduced HLA-A2+ T cells efficiently killed HLA-A2+H3.3K27M+ glioma cells in an antigen- and HLA-specific manner. Adoptive transfer of TCR-transduced T cells significantly suppressed the progression of glioma xenografts in mice. Alanine-scanning assays suggested the absence of known human proteins sharing the key amino acid residues required for recognition by the TCR, suggesting that the TCR could be safely used in patients. These data provide us with a strong basis for developing T cell-based therapy targeting this shared neoepitope.
Collapse
Affiliation(s)
- Zinal S Chheda
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Gary Kohanbash
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA.,Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kaori Okada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Naznin Jahan
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - John Sidney
- Center for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Matteo Pecoraro
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Xinbo Yang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA
| | - Diego A Carrera
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Kira M Downey
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Shruti Shrivastav
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Shuming Liu
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Yi Lin
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Chetana Lagisetti
- Department of Public Health, University of California, Berkeley, Berkeley, CA
| | - Pavlina Chuntova
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Payal B Watchmaker
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Sabine Mueller
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Ian F Pollack
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Raja Rajalingam
- Department of Surgery, Immunogenetics and Transplantation Laboratory, University of California, San Francisco, San Francisco, CA
| | | | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Alessandro Sette
- Center for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA.,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA
| | - Yafei Hou
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA .,Cancer Immunotherapy Program, University of California, San Francisco, San Francisco, CA.,The Parker Institute for Cancer Immunotherapy, San Francisco, CA
| |
Collapse
|
34
|
Zhao F, Zhang R, Wang J, Wu D, Pan M, Li M, Guo M, Dou J. Effective tumor immunity to melanoma mediated by B16F10 cancer stem cell vaccine. Int Immunopharmacol 2017; 52:238-244. [PMID: 28950176 DOI: 10.1016/j.intimp.2017.09.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/18/2017] [Accepted: 09/19/2017] [Indexed: 12/29/2022]
Abstract
Although tumor vaccines have been considered a promising immunotherapy approach, therapeutic tumor vaccines are mostly disappointing in the clinic due to vaccine weak immunogenicity. Cancer stem cells (CSCs) may broaden the antigenic breadth and effectively induce the immune responses against autologous cancer cells. Here we report on the development of the B16F10 CD133+CD44+CSCs (B16F10 CSCs) vaccine to induce tumor immunity to melanoma in mice. Efficacy of against melanoma was evaluated by analysis of tumor growth and mouse survival. Immunogenicity was assessed by ELISA and flow cytometric assays, including serum cytokines, cytotoxic activity of NK cells and splenocytes in the immunized mice. The results showed that the B16F10 CSC vaccine resulted in tumor shrinkage and mouse lifespan extension. The cytotoxic activity and IFN-γ level were significantly increased in mice immunized with B16F10 CSC vaccine compared with the mice immunized with control vaccines. Additionally, New York esophageal squamous cell carcinoma-1, an efficient tumor associated antigen over-expressed by B16F10 CSCs, was markedly reduced in expression in melanoma tissue, suggesting decrease of CSC subpopulation due to B16F10 CSC vaccination. Collectively, the findings may represent a new powerful approach for treatment of melanoma by B16F10 CSC vaccination.
Collapse
Affiliation(s)
- Fengshu Zhao
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Rong Zhang
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jing Wang
- Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Di Wu
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Meng Pan
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Miao Li
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Mei Guo
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jun Dou
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
35
|
Cencioni MT, Magliozzi R, Nicholas R, Ali R, Malik O, Reynolds R, Borsellino G, Battistini L, Muraro PA. Programmed death 1 is highly expressed on CD8 + CD57 + T cells in patients with stable multiple sclerosis and inhibits their cytotoxic response to Epstein-Barr virus. Immunology 2017; 152:660-676. [PMID: 28767147 DOI: 10.1111/imm.12808] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/27/2017] [Accepted: 07/20/2017] [Indexed: 02/06/2023] Open
Abstract
Growing evidence points to a deregulated response to Epstein-Barr virus (EBV) in the central nervous system of patients with multiple sclerosis (MS) as a possible cause of disease. We have investigated the response of a subpopulation of effector CD8+ T cells to EBV in 36 healthy donors and in 35 patients with MS in active and inactive disease. We have measured the expression of markers of degranulation, the release of cytokines, cytotoxicity and the regulation of effector functions by inhibitory receptors, such as programmed death 1 (PD-1) and human inhibitor receptor immunoglobulin-like transcript 2 (ILT2). We demonstrate that polyfunctional cytotoxic CD8+ CD57+ T cells are able to kill EBV-infected cells in healthy donors. In contrast, an anergic exhaustion-like phenotype of CD8+ CD57+ T cells with high expression of PD-1 was observed in inactive patients with MS compared with active patients with MS or healthy donors. Detection of CD8+ CD57+ T cells in meningeal inflammatory infiltrates from post-mortem MS tissue confirmed the association of this cell phenotype with the disease pathological process. The overall results suggest that ineffective immune control of EBV in patietns with MS during remission may be one factor preceding and enabling the reactivation of the virus in the central nervous system and may cause exacerbation of the disease.
Collapse
Affiliation(s)
- Maria T Cencioni
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK
| | - Roberta Magliozzi
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK.,Department of Neurosciences, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Richard Nicholas
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK.,Department of Neurosciences, Imperial College Healthcare NHS Trust, London, UK
| | - Rehiana Ali
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK.,Department of Neurosciences, Imperial College Healthcare NHS Trust, London, UK
| | - Omar Malik
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK.,Department of Neurosciences, Imperial College Healthcare NHS Trust, London, UK
| | - Richard Reynolds
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK
| | | | - Luca Battistini
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Paolo A Muraro
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK.,Department of Neurosciences, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
36
|
Kandarian F, Sunga GM, Arango-Saenz D, Rossetti M. A Flow Cytometry-Based Cytotoxicity Assay for the Assessment of Human NK Cell Activity. J Vis Exp 2017. [PMID: 28829424 DOI: 10.3791/56191] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Within the innate immune system, effector lymphocytes known as natural killer (NK) cells play an essential role in host defense against aberrant cells, specifically eliminating tumoral and virally infected cells. Approximately 30 known monogenic defects, together with a host of other pathological conditions, cause either functional or classic NK cell deficiency, manifesting in reduced or absent cytotoxic activity. Historically, cytotoxicity has been investigated with radioactive methods, which are cumbersome, expensive and potentially hazardous. This article describes a streamlined, clinically applicable flow cytometry-based method to quantify NK cell cytotoxic activity. In this assay, peripheral blood mononuclear cells (PBMCs) or purified NK cell preparations are co-incubated at different ratios with a target tumor cell line known to be sensitive to NK cell-mediated cytotoxicity (NKCC). The target cells are pre-labeled with a fluorescent dye to allow their discrimination from the effector cells (NK cells). After the incubation period, killed target cells are identified by a nucleic acid stain, which specifically permeates dead cells. This method is amenable to both diagnostic and research applications and, thanks to the multi-parameter capabilities of flow cytometry, has the added advantage of potentially enabling a deeper analysis of NK cell phenotype and function.
Collapse
Affiliation(s)
- Fadi Kandarian
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Gemalene M Sunga
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Diana Arango-Saenz
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Maura Rossetti
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles;
| |
Collapse
|
37
|
Desimio MG, Giuliani E, Doria M. The histone deacetylase inhibitor SAHA simultaneously reactivates HIV-1 from latency and up-regulates NKG2D ligands sensitizing for natural killer cell cytotoxicity. Virology 2017; 510:9-21. [PMID: 28689087 DOI: 10.1016/j.virol.2017.06.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/14/2017] [Accepted: 06/29/2017] [Indexed: 01/01/2023]
Abstract
In pilot HIV-1 eradication studies, patients' immune responses were ineffective at killing viral reservoirs reactivated through latency reversing agents (LRAs) like suberoylanilide hydroxamic acid (SAHA). We hypothesized that T cells harboring reactivated HIV-1 express MIC and ULBP ligands for the activating NKG2D receptor of natural killer (NK) cells. Here, we demonstrated that MICA/B and ULBP2 are induced by SAHA on primary T cells harboring reactivated virus. Using latently HIV-1-infected J-Lat 6.3/8.4/9.2 and J1.1 cell lines, we showed that SAHA reverts latency and, simultaneously, up-regulates MICA/B and ULBP2 acting at the transcriptional level and through ATR activation, thus sensitizing T cells with reactivated virus to NKG2D-mediated killing by NK cells. Moreover, IL-2 and IL-15 potently boosted NKG2D expression and cytotoxicity of NK cells against SAHA-reactivated p24+ target cells. Therefore, immunotherapy with cytokines enhancing NKG2D-mediated NK-cell cytotoxicity combined with administration of LRAs up-modulating NKG2D ligands, represents a promising approach towards HIV-1 eradication.
Collapse
Affiliation(s)
| | - Erica Giuliani
- Laboratory of Immunoinfectivology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Margherita Doria
- Laboratory of Immunoinfectivology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| |
Collapse
|
38
|
Yang ZZ, Kim HJ, Villasboas JC, Chen YP, Price-Troska T, Jalali S, Wilson M, Novak AJ, Ansell SM. Expression of LAG-3 defines exhaustion of intratumoral PD-1 + T cells and correlates with poor outcome in follicular lymphoma. Oncotarget 2017; 8:61425-61439. [PMID: 28977875 PMCID: PMC5617435 DOI: 10.18632/oncotarget.18251] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 04/26/2017] [Indexed: 11/25/2022] Open
Abstract
Exhausted T-cells in follicular lymphoma (FL) typically express PD-1, but expression of PD-1 is not limited to exhausted cells. Although expected to be functionally suppressed, we found that the population of intratumoral PD-1+ T cells were predominantly responsible for production of cytokines and granules. This surprising finding prompted us to explore the involvement of LAG-3 to specifically identify functionally exhausted T cells. We found that LAG-3 was expressed on a subset of intratumoral T cells from FL and LAG-3+ T cells almost exclusively came from PD-1+ population. CyTOF analysis revealed that intratumoral LAG-3+ T cells were phenotypically heterogeneous as LAG-3 was expressed on a variety of T cell subsets. In contrast to PD-1+LAG-3- cells, intratumoral PD-1+LAG-3+ T cells exhibited reduced capacity to produce cytokines and granules. LAG-3 expression could be substantially upregulated on CD4+ or CD8+ T cells by IL-12, a cytokine that has been shown to induce T-cell exhaustion and be increased in the serum of lymphoma patients. Furthermore, we found that blockade of both PD-1 and LAG-3 signaling enhanced the function of intratumoral CD8+ T cells resulting in increased IFN-γ and IL-2 production. Clinically, LAG-3 expression on intratumoral T cells correlated with a poor outcome in FL patients. Taken together, we find that LAG-3 expression is necessary to identify the population of intratumoral PD-1+ T cells that are functionally exhausted and, in contrast, find that PD-1+LAG-3- T cells are simply activated cells that are immunologically functional. These findings may have important implications for immune checkpoint therapy in FL.
Collapse
Affiliation(s)
- Zhi-Zhang Yang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hyo Jin Kim
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jose C Villasboas
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ya-Ping Chen
- Division of Oncology/Hematology and Internal Medicine, College of Medicine, National Cheng Kung University and Hospital, Tainan, Taiwan
| | - Tammy Price-Troska
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shahrzad Jalali
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Mara Wilson
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Anne J Novak
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Stephen M Ansell
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
39
|
Khanna S, Thomas A, Abate-Daga D, Zhang J, Morrow B, Steinberg SM, Orlandi A, Ferroni P, Schlom J, Guadagni F, Hassan R. Malignant Mesothelioma Effusions Are Infiltrated by CD3 + T Cells Highly Expressing PD-L1 and the PD-L1 + Tumor Cells within These Effusions Are Susceptible to ADCC by the Anti-PD-L1 Antibody Avelumab. J Thorac Oncol 2016; 11:1993-2005. [PMID: 27544053 DOI: 10.1016/j.jtho.2016.07.033] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 07/12/2016] [Accepted: 07/22/2016] [Indexed: 12/29/2022]
Abstract
INTRODUCTION The functional aspects of programmed death 1 (PD-1) and PD ligand 1 (PD-L1) immune checkpoints in malignant mesothelioma have not been studied. METHODS Tumor samples from 65 patients with mesothelioma were evaluated for PD-L1 expression by immunohistochemistry, and its prognostic significance was examined. Malignant effusions from patients with pleural and peritoneal mesothelioma were evaluated for PD-1-positive and PD-L1-positive infiltrating lymphocytes and their role in inducing PD-L1 expression in tumor cells. Antibody-dependent cellular cytotoxicity (ADCC) of avelumab, a fully humanized immunoglobulin G1 anti PD-L1 antibody against primary mesothelioma cell lines, was evaluated in presence of autologous and allogeneic natural killer cells. RESULTS Of 65 pleural and peritoneal mesothelioma tumors examined, 41 (63%) were PD-L1-positive, which was associated with slightly inferior overall survival compared to patients with PD-L1-negative tumors (median 23.0 versus 33.3 months, p = 0.35). The frequency of PD-L1 expression was similar in patients with pleural and peritoneal mesothelioma, with 62% and 64% of samples testing positive, respectively. In nine mesothelioma effusion samples evaluated, the fraction of cells expressing PD-L1 ranged from 12% to 83%. In seven patients with paired malignant effusion and peripheral blood mononuclear cell (PBMC) samples, PD-L1 expression was significantly higher on CD3-positive T cells present in malignant effusions as compared with PBMCs (p = 0.016). In addition, the numbers of CD14-positive PD-1-positive cells were increased in malignant effusions compared with PBMCs (p = 0.031). The lymphocytes present in malignant effusions recognized autologous tumor cells and induced interferon-γ-mediated PD-L1 expression on the tumor cell surface. Of the three primary mesothelioma cell lines tested, two were susceptible to avelumab-mediated ADCC in the presence of autologous natural killer cells. CONCLUSIONS Most pleural as well as peritoneal mesotheliomas express PD-L1. Malignant effusions in this disease are characterized by the presence of tumor cells and CD3-positive T cells that highly express PD-L1. In addition, mesothelioma tumor cells are susceptible to ADCC by the anti-PD-L1 antibody avelumab.
Collapse
Affiliation(s)
- Swati Khanna
- Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Anish Thomas
- Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Daniel Abate-Daga
- Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jingli Zhang
- Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Betsy Morrow
- Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University of Rome, Italy
| | - Patrizia Ferroni
- San Raffaele Roma Open University and BioBIM (Interinstitutional Multidisciplinary BioBank), SR Research Center, IRCCS San Raffaele Pisana, Rome, Italy
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Fiorella Guadagni
- San Raffaele Roma Open University and BioBIM (Interinstitutional Multidisciplinary BioBank), SR Research Center, IRCCS San Raffaele Pisana, Rome, Italy
| | - Raffit Hassan
- Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
40
|
Goplen NP, Saxena V, Knudson KM, Schrum AG, Gil D, Daniels MA, Zamoyska R, Teixeiro E. IL-12 Signals through the TCR To Support CD8 Innate Immune Responses. THE JOURNAL OF IMMUNOLOGY 2016; 197:2434-43. [PMID: 27521342 DOI: 10.4049/jimmunol.1600037] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 07/13/2016] [Indexed: 01/19/2023]
Abstract
CD8 T cells must integrate antigenic and inflammatory signals to differentiate into efficient effector and memory T cells able to protect us from infections. The mechanisms by which TCR signaling and proinflammatory cytokine receptor signaling cooperate in these processes are poorly defined. In this study, we show that IL-12 and other proinflammatory cytokines transduce signals through the TCR signalosome in a manner that requires Fyn activity and self-peptide-MHC (self-pMHC) interactions. This mechanism is crucial for CD8 innate T cell functions. Loss of Fyn activity or blockade of self-pMHC interactions severely impaired CD8 T cell IFN-γ and NKG2D expression, proliferation, and cytotoxicity upon cytokine-mediated bystander activation. Most importantly, in the absence of self-pMHC interactions, CD8 memory T cells fail to undergo bystander activation upon an unrelated infection. Thus, CD8 T cell bystander activation, although independent of cognate Ag, still requires self-pMHC and TCR signaling.
Collapse
Affiliation(s)
- Nicholas P Goplen
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Vikas Saxena
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Karin M Knudson
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Adam G Schrum
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905; and
| | - Diana Gil
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905; and
| | - Mark A Daniels
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Rose Zamoyska
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, Ashworth Laboratories, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
| | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212;
| |
Collapse
|
41
|
Developmental cell death programs license cytotoxic cells to eliminate histocompatible partners. Proc Natl Acad Sci U S A 2016; 113:6520-5. [PMID: 27217570 DOI: 10.1073/pnas.1606276113] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In a primitive chordate model of natural chimerism, one chimeric partner is often eliminated in a process of allogeneic resorption. Here, we identify the cellular framework underlying loss of tolerance to one partner within a natural Botryllus schlosseri chimera. We show that the principal cell type mediating chimeric partner elimination is a cytotoxic morula cell (MC). Proinflammatory, developmental cell death programs render MCs cytotoxic and, in collaboration with activated phagocytes, eliminate chimeric partners during the "takeover" phase of blastogenic development. Among these genes, the proinflammatory cytokine IL-17 enhances cytotoxicity in allorecognition assays. Cellular transfer of FACS-purified MCs from allogeneic donors into recipients shows that the resorption response can be adoptively acquired. Transfer of 1 × 10(5) allogeneic MCs eliminated 33 of 78 (42%) recipient primary buds and 20 of 76 (20.5%) adult parental adult organisms (zooids) by 14 d whereas transfer of allogeneic cell populations lacking MCs had only minimal effects on recipient colonies. Furthermore, reactivity of transferred cells coincided with the onset of developmental-regulated cell death programs and disproportionately affected developing tissues within a chimera. Among chimeric partner "losers," severe developmental defects were observed in asexually propagating tissues, reflecting a pathologic switch in gene expression in developmental programs. These studies provide evidence that elimination of one partner in a chimera is an immune cell-based rejection that operates within histocompatible pairs and that maximal allogeneic responses involve the coordination of both phagocytic programs and the "arming" of cytotoxic cells.
Collapse
|
42
|
Wang X, Zhao F, He X, Wang J, Zhang Y, Zhang H, Ni Y, Sun J, Wang X, Dou J. Combining TGF-β1 knockdown and miR200c administration to optimize antitumor efficacy of B16F10/GPI-IL-21 vaccine. Oncotarget 2016; 6:12493-504. [PMID: 25895132 PMCID: PMC4494953 DOI: 10.18632/oncotarget.3722] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 03/05/2015] [Indexed: 11/26/2022] Open
Abstract
TGF-β1 secreted abundantly by tumors cells as well as present in the local microenvironment promotes neoplasm invasion and metastasis by triggering the epithelial to mesenchymal transition (EMT). MiR200c has been shown to suppress EMT and to regulate the cellular epithelial and interstitial state conversion, whereas the tumor vaccines are intended to specifically initiate or amplify a host response against evolving tumor cells. Our study aimed at optimizing the antitumor effects of the B16F10/glycosylphosphatidylinositol-interleukin 21 (B16F10/GPI-IL-21) tumor vaccine on melanoma bearing mice by combining the TGF-β1 knockdown and the administration of miR200c agomir. The mice were subcutaneously vaccinated with inactivated B16F10/GPI-IL-21 vaccine and challenged by B16F10 cells transfected with shTGF-β1 (B16F10/shTGF-β1 cells) or B16F10/shTGF-β1 cells with the administration of miR200c agomir. The later combination showed that, when compared with the mice in the control group that received no vaccination, vaccinated mice significantly increased NK and CTL activities, enhanced levels of IFN-γ, and reduced expression of TGF-β1, N-cadherin, Vimentin, Gli1/2, P-Smad2/3 and others involved in promoting expression of EMT-related molecules in tumor areas, and inhibited the melanoma metastasis in lungs and lymph nodes. Altogether, our findings demonstrate that this synergistic anti-cancer regimen effectively induces strong immune response and diminishes the melanoma progression.
Collapse
Affiliation(s)
- Xiaoying Wang
- Department of Pathogenic Biology and Immunology, School of Medicine & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, China
| | - Fengshu Zhao
- Department of Pathogenic Biology and Immunology, School of Medicine & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, China
| | - Xiangfeng He
- Department of Pathogenic Biology and Immunology, School of Medicine & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, China.,Department of Medical Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Jing Wang
- Department of Gynecology and Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ying Zhang
- Department of Pathogenic Biology and Immunology, School of Medicine & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, China
| | - Hongyi Zhang
- Department of Pathogenic Biology and Immunology, School of Medicine & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, China
| | - Yaoyao Ni
- Department of Pathogenic Biology and Immunology, School of Medicine & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, China
| | - Jianan Sun
- Department of Pathogenic Biology and Immunology, School of Medicine & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, China
| | - Xiaobing Wang
- Department of Center for Experiment Animal, School of Medicine, Southeast University, Nanjing, China
| | - Jun Dou
- Department of Pathogenic Biology and Immunology, School of Medicine & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, China
| |
Collapse
|
43
|
Du VY, Bansal A, Carlson J, Salazar-Gonzalez JF, Salazar MG, Ladell K, Gras S, Josephs TM, Heath SL, Price DA, Rossjohn J, Hunter E, Goepfert PA. HIV-1-Specific CD8 T Cells Exhibit Limited Cross-Reactivity during Acute Infection. THE JOURNAL OF IMMUNOLOGY 2016; 196:3276-86. [PMID: 26983786 DOI: 10.4049/jimmunol.1502411] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/11/2016] [Indexed: 01/03/2023]
Abstract
Prior work has demonstrated that HIV-1-specific CD8 T cells can cross-recognize variant epitopes. However, most of these studies were performed in the context of chronic infection, where the presence of viral quasispecies makes it difficult to ascertain the true nature of the original antigenic stimulus. To overcome this limitation, we evaluated the extent of CD8 T cell cross-reactivity in patients with acute HIV-1 clade B infection. In each case, we determined the transmitted founder virus sequence to identify the autologous epitopes restricted by individual HLA class I molecules. Our data show that cross-reactive CD8 T cells are infrequent during the acute phase of HIV-1 infection. Moreover, in the uncommon instances where cross-reactive responses were detected, the variant epitopes were poorly recognized in cytotoxicity assays. Molecular analysis revealed that similar antigenic structures could be cross-recognized by identical CD8 T cell clonotypes mobilized in vivo, yet even subtle differences in a single TCR-accessible peptide residue were sufficient to disrupt variant-specific reactivity. These findings demonstrate that CD8 T cells are highly specific for autologous epitopes during acute HIV-1 infection. Polyvalent vaccines may therefore be required to provide optimal immune cover against this genetically labile pathogen.
Collapse
Affiliation(s)
- Victor Y Du
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Anju Bansal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | | | | | - Maria G Salazar
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Kristin Ladell
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Stephanie Gras
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Tracy M Josephs
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Sonya L Heath
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - David A Price
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom; Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Jamie Rossjohn
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom; Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Eric Hunter
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30329
| | - Paul A Goepfert
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294;
| |
Collapse
|
44
|
Wei FQ, Sun W, Wong TS, Gao W, Wen YH, Wei JW, Wei Y, Wen WP. Eliciting cytotoxic T lymphocytes against human laryngeal cancer-derived antigens: evaluation of dendritic cells pulsed with a heat-treated tumor lysate and other antigen-loading strategies for dendritic-cell-based vaccination. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:18. [PMID: 26795730 PMCID: PMC4722756 DOI: 10.1186/s13046-016-0295-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/17/2016] [Indexed: 11/10/2022]
Abstract
BACKGROUND Dendritic cells (DCs) have been used successfully in clinical pilot studies. However, tumor-specific immunity and clinical responses were only induced in certain cancer patients. It has been well documented that immunotherapy efficacy can be optimized for responses using antigen pulsing. METHODS The human laryngeal squamous cell cancer (LSCC) cell line SNU899 was used to evaluate the in vitro anti-tumor efficacy of three different preparations of dendritic cell (DC) vaccines consisting of either whole tumor cells or their derivatives including: i) DCs pulsed with a tumor cell supernatant (DC-TCS), ii) DCs pulsed with whole-cell tumor stressed lysate (DC-TSL), and iii) DCs pulsed with irradiated tumor cells (DC-ITC). RESULTS Our results showed that DC-TSL is an effective source of tumor-associated antigens (TAAs) for pulsing DCs. DC-TSL induced the highest expansion of TAA-specific T cells, the strongest Th1 cytokine response, and the most potent cytotoxic T lymphocyte (CTL) activity. DC-TCS and DC-ITC inhibited T cell activation but induced a certain extent of CTL activity. CONCLUSIONS These data suggest that DC-TSL is a more potent inducer of antitumor immunity against laryngeal cancer than other antigen-loading strategies using whole tumor cell materials. This strategy provides an alternative approach for DC-based immunotherapy for laryngeal cancer.
Collapse
Affiliation(s)
- Fan-Qin Wei
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Sun Yat-sen University, 2nd Zhongshan Road 58#, Guangzhou, 510080, Guangdong, P.R. China. .,Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-sen University, 2nd Zhongshan Road 58#, Guangzhou, 510080, Guangdong, P.R. China. .,Department of Otorhinolaryngology Head and Neck Surgery, the Sixth Affiliated Hospital of Sun Yat-Sen University, Yuancun Second Cross Road 26#, Guangzhou, 510655, Guangdong, P.R. China.
| | - Wei Sun
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Sun Yat-sen University, 2nd Zhongshan Road 58#, Guangzhou, 510080, Guangdong, P.R. China.
| | - Thian-Sze Wong
- Department of Surgery, The University of Hong Kong, Pokfulam Road 102#, Hong Kong, P.R. China.
| | - Wei Gao
- Department of Surgery, The University of Hong Kong, Pokfulam Road 102#, Hong Kong, P.R. China.
| | - Yi-Hui Wen
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Sun Yat-sen University, 2nd Zhongshan Road 58#, Guangzhou, 510080, Guangdong, P.R. China. .,Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-sen University, 2nd Zhongshan Road 58#, Guangzhou, 510080, Guangdong, P.R. China.
| | - Jia-Wei Wei
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Sun Yat-sen University, 2nd Zhongshan Road 58#, Guangzhou, 510080, Guangdong, P.R. China. .,Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-sen University, 2nd Zhongshan Road 58#, Guangzhou, 510080, Guangdong, P.R. China.
| | - Yi Wei
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Sun Yat-sen University, 2nd Zhongshan Road 58#, Guangzhou, 510080, Guangdong, P.R. China. .,Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-sen University, 2nd Zhongshan Road 58#, Guangzhou, 510080, Guangdong, P.R. China.
| | - Wei-Ping Wen
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Sun Yat-sen University, 2nd Zhongshan Road 58#, Guangzhou, 510080, Guangdong, P.R. China. .,Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-sen University, 2nd Zhongshan Road 58#, Guangzhou, 510080, Guangdong, P.R. China.
| |
Collapse
|
45
|
Mutations in a Highly Conserved Motif of nsp1β Protein Attenuate the Innate Immune Suppression Function of Porcine Reproductive and Respiratory Syndrome Virus. J Virol 2016; 90:3584-99. [PMID: 26792733 DOI: 10.1128/jvi.03069-15] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/11/2016] [Indexed: 01/31/2023] Open
Abstract
UNLABELLED Porcine reproductive and respiratory syndrome virus (PRRSV) nonstructural protein 1β (nsp1β) is a multifunctional viral protein, which is involved in suppressing the host innate immune response and activating a unique -2/-1 programmed ribosomal frameshifting (PRF) signal for the expression of frameshifting products. In this study, site-directed mutagenesis analysis showed that the R128A or R129A mutation introduced into a highly conserved motif ((123)GKYLQRRLQ(131)) reduced the ability of nsp1β to suppress interferon beta (IFN-β) activation and also impaired nsp1β's function as a PRF transactivator. Three recombinant viruses, vR128A, vR129A, and vRR129AA, carrying single or double mutations in the GKYLQRRLQ motif were characterized. In comparison to the wild-type (WT) virus, vR128A and vR129A showed slightly reduced growth abilities, while the vRR129AA mutant had a significantly reduced growth ability in infected cells. Consistent with the attenuated growth phenotype in vitro, pigs infected with nsp1β mutants had lower levels of viremia than did WT virus-infected pigs. Compared to the WT virus in infected cells, all three mutated viruses stimulated high levels of IFN-α expression and exhibited a reduced ability to suppress the mRNA expression of selected interferon-stimulated genes (ISGs). In pigs infected with nsp1β mutants, IFN-α production was increased in the lungs at early time points postinfection, which was correlated with increased innate NK cell function. Furthermore, the augmented innate response was consistent with the increased production of IFN-γ in pigs infected with mutated viruses. These data demonstrate that residues R128 and R129 are critical for nsp1β function and that modifying these key residues in the GKYLQRRLQ motif attenuates virus growth ability and improves the innate and adaptive immune responses in infected animals. IMPORTANCE PRRSV infection induces poor antiviral innate IFN and cytokine responses, which results in weak adaptive immunity. One of the strategies in next-generation vaccine construction is to manipulate viral proteins/genetic elements involved in antagonizing the host immune response. PRRSV nsp1β was identified to be a strong innate immune antagonist. In this study, two basic amino acids, R128 and R129, in a highly conserved GKYLQRRLQ motif were determined to be critical for nsp1β function. Mutations introduced into these two residues attenuated virus growth and improved the innate and adaptive immune responses of infected animals. Technologies developed in this study could be broadly applied to current commercial PRRSV modified live-virus (MLV) vaccines and other candidate vaccines.
Collapse
|
46
|
La Muraglia GM, O'Neil MJ, Madariaga ML, Michel SG, Mordecai KS, Allan JS, Madsen JC, Hanekamp IM, Preffer FI. A novel approach to measuring cell-mediated lympholysis using quantitative flow and imaging cytometry. J Immunol Methods 2015; 427:85-93. [PMID: 26516062 DOI: 10.1016/j.jim.2015.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 08/05/2015] [Accepted: 10/21/2015] [Indexed: 11/30/2022]
Abstract
In this study, we established a novel isotope-free approach for the detection of cell-mediated lympholysis (CML) in MHC defined peripheral blood mononuclear cells (PBMCs) using multiparameter flow and imaging cytometry. CML is an established in vitro assay to detect the presence of cytotoxic effector T-lymphocytes precursors (CTLp). Current methods employed in the identification of CTLp in the context of transplantation are based upon the quantification of chromium ((51)Cr) released from target cells. In order to adapt the assay to flow cytometry, primary porcine PBMC targets were labeled with eFluor670 and incubated with major histocompatibility complex (MHC) mismatched effector cytotoxic lymphocytes (CTLs). With this method, we were able to detect target-specific lysis that was comparable to that observed with the (51)Cr-based assay. In addition, the use of quantitative cell imaging demonstrates the presence of accessory cells involved in the cytotoxic pathway. This innovative technique improves upon the standard (51)Cr release assay by eliminating the need for radioisotopes and provides enhanced characterization of the interactions between effector and target cells. This technique has wide applicability to numerous experimental and clinical models involved with effector-cell interactions.
Collapse
Affiliation(s)
- G M La Muraglia
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - M J O'Neil
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - M L Madariaga
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - S G Michel
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - K S Mordecai
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - J S Allan
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - J C Madsen
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - I M Hanekamp
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - F I Preffer
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
47
|
Wu D, Wang J, Cai Y, Ren M, Zhang Y, Shi F, Zhao F, He X, Pan M, Yan C, Dou J. Effect of targeted ovarian cancer immunotherapy using ovarian cancer stem cell vaccine. J Ovarian Res 2015; 8:68. [PMID: 26497895 PMCID: PMC4620009 DOI: 10.1186/s13048-015-0196-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/12/2015] [Indexed: 12/21/2022] Open
Abstract
Background Accumulating evidence has shown that different immunotherapies for ovarian cancer might overcome barriers to resistance to standard chemotherapy. The vaccine immunotherapy may be a useful one addition to conditional chemotherapy regimens. The present study investigated the use of vaccine of ovarian cancer stem cells (CSCs) to inhibit ovarian cancer growth. Methods CD117+CD44+CSCs were isolated from human epithelial ovarian cancer (EOC) SKOV3 cell line by using a magnetic-activated cell sorting system. Pre-inactivated CD117+CD44+CSC vaccine was vacccinated into athymic nude mice three times, and then the mice were challenged subcutaneously with SKOV3 cells. The anti-tumor efficacy of CSC vaccine was envaluated by in vivo tumorigenicity, immune efficient analysis by flow cytometer, and enzyme-linked immunosorbent assays, respectively. Results The CD117+ CD44+CSC vaccine increased anti-ovarian cancer efficacy in that it depressed ovarian cancer growth in the athymic nude mice. Vaccination resulted in enhanced serum IFN-γ, decreased TGF-β levels, and increased cytotoxic activity of natural killer cells in the CD117+ CD44+CSC vaccine immunized mice. Moreover, the CSC-based vaccine significantly reduced the CD117+CD44+CSC as well as the aldehyde dehydrogenase 1 positive cell populations in the ovarian cancer tissues in the xenograft mice. Conclusion The present study provided the first evidence that human SKOV3 CD117+ CD44+CSC-based vaccine may induce the anti-ovarian cancer immunity against tumor growth by reducing the CD117+CD44+CSC population.
Collapse
Affiliation(s)
- Di Wu
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jing Wang
- Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yunlang Cai
- Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
| | - Mulan Ren
- Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yuxia Zhang
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, 210009, China.,Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Fangfang Shi
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, 210009, China.,Department of oncology, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Fengshu Zhao
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xiangfeng He
- Department of Medical Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, 226361, China
| | - Meng Pan
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Chunguang Yan
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jun Dou
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
48
|
Tang L, Peng H, Zhou J, Chen Y, Wei H, Sun R, Yokoyama WM, Tian Z. Differential phenotypic and functional properties of liver-resident NK cells and mucosal ILC1s. J Autoimmun 2015; 67:29-35. [PMID: 26422992 DOI: 10.1016/j.jaut.2015.09.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 09/15/2015] [Accepted: 09/18/2015] [Indexed: 12/16/2022]
Abstract
Group 1 innate lymphoid cells (ILCs) consist of conventional natural killer (cNK) cells, tissue-resident NK cells and mucosal ILC1s. Recently identified liver-resident NK cells, which can mount contact hypersensitivity responses, and mucosal ILC1s that are involved in pathogenesis of colitis are distinct from cNK cells in several aspects, but the issue of how they are related to each other has not been clearly clarified. Here, we show that liver-resident NK cells and mucosal ILC1s have different phenotypes, as evidenced by distinct expression patterns of homing-associated molecules. Moreover, mucosal ILC1s exhibit tissue residency akin to liver-resident NK cells. Importantly, liver-resident NK cells express relative high levels of cytotoxic effector molecules, which are poorly expressed by mucosal ILC1s, and exhibit stronger cytotoxic activity compared with mucosal ILC1s. These results demonstrate differential phenotypic and functional characteristics of liver-resident NK cells and mucosal ILC1s, shedding new light on the diversity of ILC family.
Collapse
Affiliation(s)
- Ling Tang
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Hui Peng
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Jing Zhou
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yongyan Chen
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Haiming Wei
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Rui Sun
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Wayne M Yokoyama
- Howard Hughes Medical Institute and Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhigang Tian
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China; Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China.
| |
Collapse
|
49
|
Pan QZ, Pan K, Wang QJ, Weng DS, Zhao JJ, Zheng HX, Zhang XF, Jiang SS, Lv L, Tang Y, Li YQ, He J, Liu Q, Chen CL, Zhang HX, Xia JC. Annexin A3 as a potential target for immunotherapy of liver cancer stem-like cells. Stem Cells 2015; 33:354-66. [PMID: 25267273 DOI: 10.1002/stem.1850] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 09/04/2014] [Indexed: 12/14/2022]
Abstract
Cancer stem-like cells/cancer-initiating cells (CSCs/CICs) are considered to represent a small population of cancer cells that is resistant to conventional cancer treatments and responsible for tumor recurrence and metastasis. The aim of this study was to establish CSC/CIC-targeting immunotherapy. In this study, we found that Annexin A3 (ANXA3) was preferentially expressed in CSCs/CICs derived from hepatocellular carcinoma (HCC) cells compared to non-CSCs/CICs. In HCC samples, high levels of ANXA3 correlated with expansion of CD133(+) tumor cells representing CSCs/CICs in HCC; the combination of high levels of ANXA3 and CD133 was associated with progression of HCC. Overexpression of ANXA3 increased the proportion of CD133(+) cells, enhancing their tumorigenicity. On the contrary, knockdown of ANXA3 decreased CD133(+) cells and inhibited tumorigenicity. The mechanistic study revealed that ANXA3-mediated maintenance of HCC CSCs/CICs activity was likely involved with the HIF1A/Notch pathway. Using ANXA3 as a target, ANXA3-transfected dendritic cells could induce more functionally active T cells and these effector T cells could superiorly kill CD133(+) HCC CSCs/CICs in vitro and in vivo. Taken together, our findings suggest that ANXA3 plays a role in HCC CSC/CIC maintenance, and that ANXA3 may represent a potential CSC/CIC-specific therapeutic target for improving the treatment of HCC.
Collapse
Affiliation(s)
- Qiu-Zhong Pan
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China; Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ayano M, Tsukamoto H, Kohno K, Ueda N, Tanaka A, Mitoma H, Akahoshi M, Arinobu Y, Niiro H, Horiuchi T, Akashi K. Increased CD226 Expression on CD8+ T Cells Is Associated with Upregulated Cytokine Production and Endothelial Cell Injury in Patients with Systemic Sclerosis. THE JOURNAL OF IMMUNOLOGY 2015; 195:892-900. [PMID: 26109642 DOI: 10.4049/jimmunol.1403046] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 05/26/2015] [Indexed: 02/06/2023]
Abstract
Systemic sclerosis (SSc) is an autoimmune disease characterized by vascular damage and fibrosis of the skin and internal organs. Because activated and oligoclonally expanded CD8(+) T cells can be detected in peripheral blood and lungs of SSc patients, effector memory CD8(+) T cells may play a critical role for organ involvement in SSc; however, the pathogenic functions of effector memory CD8(+) T cells remain incompletely understood. In this study, we performed DNA microarray analysis of the sort-purified effector memory CD8(+) T cells from SSc patients and healthy controls, and showed that the expression of genes related to immune response and cell adhesion, including CD226 (also known as DNAX accessory molecule-1 [DNAM-1]), was significantly altered. Moreover, detailed analysis of CD226 revealed that CD226(high)CD8(+) T cells were increased in SSc patients (mean, 50.7%) compared with healthy controls (32.9%) and were appreciably associated with the severity of skin sclerosis and interstitial lung disease. Furthermore, CD226(+)CD8(+) T cells produced higher amount of various cytokines than CD226(-) ones, and CD226(high)CD8(+) T cells from SSc patients showed upregulated IL-13 production and positive correlation with the cytotoxic capacity of CD8(+) T cells against HUVECs. Finally, the neutralization of CD226 in CD8(+) T cells impaired costimulation, cytokine productions, and cytolysis against HUVECs. These findings indicate that upregulated CD226 expression on CD8(+) T cells reflects disease severity and is involved in SSc pathogenesis via the production of various cytokines, including profibrotic IL-13 and endothelial cell injury, and that CD226 may be a useful target in the treatment of SSc.
Collapse
Affiliation(s)
- Masahiro Ayano
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiroshi Tsukamoto
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan;
| | - Kentaro Kohno
- Department of Hematology, National Kyushu Medical Center, Fukuoka 810-8563, Japan
| | - Naoyasu Ueda
- Department of Internal Medicine, Miyazaki Prefectural Miyazaki Hospital, Miyazaki 880-8510, Japan
| | - Atsushi Tanaka
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiroki Mitoma
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Mitsuteru Akahoshi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Yojiro Arinobu
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiroaki Niiro
- Clinical Education Center, Kyushu University Hospital, Fukuoka 812-8582, Japan; and
| | - Takahiko Horiuchi
- Department of Internal Medicine, Kyushu University Beppu Hospital, Beppu 874-0836, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| |
Collapse
|