201
|
Kalantari T, Ciric B, Kamali-Sarvestani E, Rostami A. Bone marrow dendritic cells deficient for CD40 and IL-23p19 are tolerogenic in vitro. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:287-292. [PMID: 32440313 PMCID: PMC7229508 DOI: 10.22038/ijbms.2020.36160.8615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Objective(s): In addition to pro-inflammatory role, dendritic cells (DCs) can also be anti-inflammatory when they acquire tolerogenic phenotype. In this study we tested the role of CD40 and IL-23p19 in antigen presenting function of bone marrow-derived DCs (BMDCs) by comparing BMDCs derived from CD40 knockout (CD40KO-DCs) and IL-23p19 (IL-23p19KO-DCs) knockout mice with those from C57BL/6 mice (Cont-DCs). We have focused on CD40 and IL-23, as these molecules have well established pro-inflammatory roles in a number of autoimmune and inflammatory diseases. Materials and Methods: The expression of maturation markers MHC II and co-stimulatory molecules CD40, CD80, and CD86 was analyzed by flow cytometry, while the expression of CD40 and IL-23p19 was measured by RT-PCR. The capacity of BMDCs to activate CD4+ T cells was evaluated by 3H-thymidine incorporation, and the capacity of BMDCs to uptake antigen was evaluated using fluorescent OVA and flow cytometry. Results: The lack of CD40 or IL-23p19 had no effect on uptake of FITC-OVA by the DCs, confirming their immature phenotype. Moreover, CD40KO-DCs had significantly reduced capacity to stimulate proliferation of CD4+ T cells. CD4+ T cells activated by CD40KO-DCs and IL-23p19KO-DCs produced significantly less IFN-γ (P-value ≤0.05), while CD4+ T cells stimulated by IL-23p19KO-DCs produced less GM-CSF and more IL-10 than Cont-DCs. Conclusion: This study shows that CD40KO-DCs and IL-23p19KO-DCs have a marked tolerogenic potency in vitro. Future in vivo studies should determine if and to what extent DCs lacking CD40 or IL-23 have a potential to be useful in therapy of autoimmune inflammation.
Collapse
Affiliation(s)
- Tahereh Kalantari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
202
|
Srinivasan S, Babensee JE. Controlled Delivery of Immunomodulators from a Biomaterial Scaffold Niche to Induce a Tolerogenic Phenotype in Human Dendritic Cells. ACS Biomater Sci Eng 2020; 6:4062-4076. [DOI: 10.1021/acsbiomaterials.0c00439] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Sangeetha Srinivasan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Julia E. Babensee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
203
|
Kishimoto TK. Development of ImmTOR Tolerogenic Nanoparticles for the Mitigation of Anti-drug Antibodies. Front Immunol 2020; 11:969. [PMID: 32508839 PMCID: PMC7251066 DOI: 10.3389/fimmu.2020.00969] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/24/2020] [Indexed: 12/19/2022] Open
Abstract
The development of anti-drug antibodies (ADAs) is a common cause for treatment failure and hypersensitivity reactions for many biologics. The focus of this review is the development of ImmTOR, a platform technology designed to prevent the formation of ADAs that can be applied broadly across a wide variety of biologics by inducing immunological tolerance with ImmTOR nanoparticles encapsulating rapamycin. The induction of tolerance is antigen-specific and dependent on the incorporation of rapamycin in nanoparticles and the presence of the antigen at the time of administration of ImmTOR. Evidence for the induction of specific immune tolerance vs. general immune suppression is supported by the findings that: (1) ImmTOR induces regulatory T cells specific to the co-administered antigen; (2) tolerance can be transferred by adoptive transfer of splenocytes from treated animals to naïve recipients; (3) the tolerance is durable to subsequent challenge with antigen alone; and (4) animals tolerized to a specific antigen are capable of responding to an unrelated antigen. ImmTOR nanoparticles can be added to new or existing biologics without the need to modify or reformulate the biologic drug. The ability of ImmTOR to mitigate the formation of ADAs has been demonstrated for coagulation factor VIII in a mouse model of hemophilia A, an anti-TNFα monoclonal antibody in a mouse model of inflammatory arthritis, pegylated uricase in hyperuricemic mice and in non-human primates, acid alpha-glucosidase in a mouse model of Pompe disease, recombinant immunotoxin in a mouse model of mesothelioma, and adeno-associated vectors in a model of repeat dosing of gene therapy vectors in mice and in non-human primates. Human proof-of concept for the mitigation of ADAs has been demonstrated with SEL-212, a combination product consisting of ImmTOR + pegadricase, a highly immunogenic enzyme therapy for the treatment of gout. ImmTOR represents a promising approach to preventing the formation of ADAs to a broad range of biologic drugs.
Collapse
|
204
|
Zhang X, Wang Y, Zhang D, Li H, Zhou Z, Yang R. CD70‐silenced dendritic cells induce immune tolerance in immune thrombocytopenia patients. Br J Haematol 2020; 191:466-475. [PMID: 32419211 DOI: 10.1111/bjh.16689] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/03/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Xian Zhang
- The Hematology Department of Zhongnan Hospital of Wuhan University Wuhan City Hubei China
- State Key Laboratory of Experimental Hematology National Clinical Research Center for Hematological Disorders Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College China
| | - Yunlong Wang
- Department of Rheumatology and Immunology Minda Hospital of Hubei University for Nationalities Enshi China
- Department of Hematology Second Affiliated Hospital of Kunming Medical University Kunming China
| | - Donglei Zhang
- State Key Laboratory of Experimental Hematology National Clinical Research Center for Hematological Disorders Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College China
| | - Huiyuan Li
- State Key Laboratory of Experimental Hematology National Clinical Research Center for Hematological Disorders Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College China
| | - Zeping Zhou
- Department of Hematology Second Affiliated Hospital of Kunming Medical University Kunming China
| | - Renchi Yang
- State Key Laboratory of Experimental Hematology National Clinical Research Center for Hematological Disorders Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College China
| |
Collapse
|
205
|
Liu A, Ferretti C, Shi FD, Cohen IR, Quintana FJ, La Cava A. DNA Vaccination With Hsp70 Protects Against Systemic Lupus Erythematosus in (NZB × NZW)F1 Mice. Arthritis Rheumatol 2020; 72:997-1002. [PMID: 31943822 DOI: 10.1002/art.41202] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/07/2020] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To address whether a targeted modulation of the abnormal expression of Hsp70 and autoantibodies against this molecule in systemic lupus erythematosus can influence disease. METHODS Lupus-prone (NZB × NZW)F1 mice that had been DNA-vaccinated with plasmids encoding Hsp70 and controls were monitored for lupus disease parameters including anti-double stranded DNA (anti-dsDNA) autoantibodies and cytokines using enzyme-linked immunosorbent assay, and for kidney function and pathology. The phenotypic and numerical changes in relevant immune cells were evaluated by flow cytometry, and cell function was assessed. RESULTS Mice that had been DNA-vaccinated with Hsp70 displayed marked suppression of anti-dsDNA antibody production, reduced renal disease, and antiinflammatory responses that are associated with a significantly extended survival, compared to controls. These protective effects in Hsp70-vaccinated mice were associated with an induction of tolerogenic immune responses and an expansion of functional Treg cells. CONCLUSION DNA vaccination with Hsp70 suppresses murine lupus by inducing tolerogenic immune responses and antiinflammatory immune responses associated with reduced disease manifestations and increased mouse survival.
Collapse
Affiliation(s)
| | | | - Fu-Dong Shi
- Barrow Neurological Institute, Phoenix, Arizona
| | - Irun R Cohen
- The Weizmann Institute of Science, Rehovot, Israel
| | | | | |
Collapse
|
206
|
Schönberg A, Hamdorf M, Bock F. Immunomodulatory Strategies Targeting Dendritic Cells to Improve Corneal Graft Survival. J Clin Med 2020; 9:E1280. [PMID: 32354200 PMCID: PMC7287922 DOI: 10.3390/jcm9051280] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/08/2020] [Accepted: 04/21/2020] [Indexed: 12/16/2022] Open
Abstract
Even though the cornea is regarded as an immune-privileged tissue, transplantation always comes with the risk of rejection due to mismatches between donor and recipient. It is common sense that an alternative to corticosteroids as the current gold standard for treatment of corneal transplantation is needed. Since blood and lymphatic vessels have been identified as a severe risk factor for corneal allograft survival, much research has focused on vessel regression or inhibition of hem- and lymphangiogenesis in general. However, lymphatic vessels have been identified as required for the inflammation's resolution. Therefore, targeting other players of corneal engraftment could reveal new therapeutic strategies. The establishment of a tolerogenic microenvironment at the graft site would leave the recipient with the ability to manage pathogenic conditions independent from transplantation. Dendritic cells (DCs) as the central player of the immune system represent a target that allows the induction of tolerogenic mechanisms by many different strategies. These strategies are reviewed in this article with regard to their success in corneal transplantation.
Collapse
Affiliation(s)
- Alfrun Schönberg
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (A.S.); (M.H.)
| | - Matthias Hamdorf
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (A.S.); (M.H.)
| | - Felix Bock
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (A.S.); (M.H.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50937 Cologne, Germany
| |
Collapse
|
207
|
Anselmi G, Vaivode K, Dutertre CA, Bourdely P, Missolo-Koussou Y, Newell E, Hickman O, Wood K, Saxena A, Helft J, Ginhoux F, Guermonprez P. Engineered niches support the development of human dendritic cells in humanized mice. Nat Commun 2020; 11:2054. [PMID: 32345968 PMCID: PMC7189247 DOI: 10.1038/s41467-020-15937-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/18/2020] [Indexed: 12/24/2022] Open
Abstract
Classical dendritic cells (cDCs) are rare sentinel cells specialized in the regulation of adaptive immunity. Modeling cDC development is crucial to study cDCs and harness their therapeutic potential. Here we address whether cDCs could differentiate in response to trophic cues delivered by mesenchymal components of the hematopoietic niche. We find that mesenchymal stromal cells engineered to express membrane-bound FLT3L and stem cell factor (SCF) together with CXCL12 induce the specification of human cDCs from CD34+ hematopoietic stem and progenitor cells (HSPCs). Engraftment of engineered mesenchymal stromal cells (eMSCs) together with CD34+ HSPCs creates an in vivo synthetic niche in the dermis of immunodeficient mice driving the differentiation of cDCs and CD123+AXL+CD327+ pre/AS-DCs. cDC2s generated in vivo display higher levels of resemblance with human blood cDCs unattained by in vitro-generated subsets. Altogether, eMSCs provide a unique platform recapitulating the full spectrum of cDC subsets enabling their functional characterization in vivo.
Collapse
Affiliation(s)
- Giorgio Anselmi
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immmunobiology, King's College London, London, UK.,Cancer Research UK, King's Health Partners Cancer Centre, King's College London, London, UK.,MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Kristine Vaivode
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immmunobiology, King's College London, London, UK.,Cancer Research UK, King's Health Partners Cancer Centre, King's College London, London, UK
| | | | - Pierre Bourdely
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immmunobiology, King's College London, London, UK.,Cancer Research UK, King's Health Partners Cancer Centre, King's College London, London, UK
| | - Yoann Missolo-Koussou
- Paris-Sciences-Lettres University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, Paris, France
| | - Evan Newell
- Singapore Immunology Network (SIgN), A*STAR, Singapore, Singapore
| | - Oliver Hickman
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immmunobiology, King's College London, London, UK.,Cancer Research UK, King's Health Partners Cancer Centre, King's College London, London, UK.,Drug Target Discovery Team, Division of Breast Cancer Research, Institute of Cancer Research, London, UK
| | - Kristie Wood
- National Institute of Health Research Biomedical Research Centre at Guy's and St Thomas' Hospital and King's College London, London, UK.,Labcyte Ltd, Norton Canes, Cannock, Staffordshire, UK
| | - Alka Saxena
- National Institute of Health Research Biomedical Research Centre at Guy's and St Thomas' Hospital and King's College London, London, UK
| | - Julie Helft
- Paris-Sciences-Lettres University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, Paris, France
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), A*STAR, Singapore, Singapore
| | - Pierre Guermonprez
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immmunobiology, King's College London, London, UK. .,Cancer Research UK, King's Health Partners Cancer Centre, King's College London, London, UK. .,Université de Paris, Centre for Inflammation Research, CNRS ERL8252, INSERM1149, Paris, France.
| |
Collapse
|
208
|
Zhang H, Wang Y, Wang QT, Sun SN, Li SY, Shang H, He YW. Enhanced Human T Lymphocyte Antigen Priming by Cytokine-Matured Dendritic Cells Overexpressing Bcl-2 and IL-12. Front Cell Dev Biol 2020; 8:205. [PMID: 32292785 PMCID: PMC7118208 DOI: 10.3389/fcell.2020.00205] [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: 01/19/2020] [Accepted: 03/10/2020] [Indexed: 01/08/2023] Open
Abstract
Dendritic cell (DC)-based vaccination is a promising immunotherapeutic strategy for cancer. However, clinical trials have shown only limited efficacy, suggesting the need to optimize protocols for human DC vaccine preparation. In this study, we systemically compared five different human DC vaccine maturation protocols used in clinical trials: (1) a four-cytokine cocktail (TNF-α, IL-6, IL-1β, and PGE2); (2) an α-DC-cytokine cocktail (TNF-α, IL-1β, IFN-α, IFN-γ, and poly I:C); (3) lipopolysaccharide (LPS)/IFN-γ; (4) TNF-α and PGE2; and (5) TriMix (mRNAs encoding CD40L, CD70, and constitutively active Toll-like receptor 4 electroporated into immature DCs). We found that the four-cytokine cocktail induced high levels of costimulatory and HLA molecules, as well as CCR7, in DCs. Mature DCs (mDCs) matured with the four-cytokine cocktail had higher viability than those obtained with the other protocols. Based on these features, we chose the four-cytokine cocktail protocol to further improve the immunizing capability of DCs by introducing exogenous genes. We showed that introducing exogenous Bcl-2 increased DC survival. Furthermore, introducing IL-12p70 rescued the inhibition of IL-12 secretion by PGE2 without impairing the DC phenotype. Introducing both Bcl-2 and IL-12p70 mRNAs into DCs induced enhanced cytomegalovirus pp65-specific CD8+ T cells secreting IFN-γ and TNF-α. Taken together, our data suggest that DC matured by the four-cytokine cocktail combined with exogenous Bcl-2 and IL-12p70 gene expression represents a promising approach for clinical applications in cancer immunotherapy.
Collapse
Affiliation(s)
- Hui Zhang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yu Wang
- Life Science Institute, Jinzhou Medical University, Jinzhou, China
| | | | - Sheng-Nan Sun
- Beijing Tricision Biotherapeutics Inc., Beijing, China
| | - Shi-You Li
- Beijing Tricision Biotherapeutics Inc., Beijing, China
| | - Hong Shang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - You-Wen He
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
209
|
I-a lowCD11b high DC Regulates the Immune Response in the Eyes of Experimental Autoimmune Uveitis. Mediators Inflamm 2020; 2020:6947482. [PMID: 32256194 PMCID: PMC7085850 DOI: 10.1155/2020/6947482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 12/30/2019] [Indexed: 12/23/2022] Open
Abstract
Regulatory dendritic cells (DCreg) have been reported to be a negative regulator in the immune response. These cells are widely distributed in the liver, spleen, and lung. However, the status and function of DCreg in the eyes and disease are still not very clear. Herein, we found that the number of I-alowCD11bhigh DC increased in the eye and spleen at the recovery stage of experimental autoimmune uveitis (EAU), which is a mouse model for autoimmune uveitis. These cells expressed lower levels of CD80, CD86, and CD54 than the mature DCs and expressed interleukin 10 (IL-10), indoleamine 2,3-dioxygenase (IDO), and transforming growth factor beta (TGF-β) as well. Moreover, these DCreg can regulate the development of EAU by promoting CD4+CD25+Foxp3+ regulatory T cells. The increased interferon-gamma (IFN-γ) in the aqueous humor of EAU participates in inducing DCreg to alleviate the symptom of EAU. Furthermore, DCreg was found to exist in the eyes of normal mice. Aqueous humor, containing a certain concentration of IL-10, TGF-β, prostaglandin E2 (PGE2), IDO, and nitric oxide (NO), induced the tolerance of DCreg in normal eyes. It can be concluded that DCreg exists in the eyes and plays a protective role in inflamed eyes. These DCreg induced by IFN-γ might be used as a strategy to develop therapy for EAU management.
Collapse
|
210
|
Nazimek K, Bryniarski K. Approaches to inducing antigen-specific immune tolerance in allergy and autoimmunity: Focus on antigen-presenting cells and extracellular vesicles. Scand J Immunol 2020; 91:e12881. [PMID: 32243636 DOI: 10.1111/sji.12881] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022]
Abstract
Increasing prevalence of allergic and autoimmune diseases urges clinicians and researchers to search for new and efficient treatments. Strategies that activate antigen-specific immune tolerance and simultaneously maintain immune reactivity to all other antigens deserve special attention. Accordingly, antigen-presenting cells (APCs) seem to be the best suited for orchestrating these mechanisms by directing T cell immune responses towards a tolerant subtype. Recent advances in understanding cell-to-cell communication via extracellular vesicles (EVs) make the latter promising candidates for reprogramming APCs towards a tolerant phenotype, and for mediating tolerogenic APC function. Thus, comprehensive studies have been undertaken to describe the interactions of APCs and EVs naturally occurring during immune tolerance induction, as well as to develop EV-based manoeuvres enabling the induction of immune tolerance in an antigen-specific manner. In this review, we summarize the findings of relevant studies, with a special emphasis on future perspectives on their translation to clinical practice.
Collapse
Affiliation(s)
- Katarzyna Nazimek
- Jagiellonian University Medical College, Department of Immunology, Krakow, Poland
| | - Krzysztof Bryniarski
- Jagiellonian University Medical College, Department of Immunology, Krakow, Poland
| |
Collapse
|
211
|
Suekane S, Yutani S, Yamada A, Sasada T, Matsueda S, Takamori S, Toh U, Kawano K, Yoshiyama K, Sakamoto S, Sugawara S, Komatsu N, Yamada T, Naito M, Terasaki M, Mine T, Itoh K, Shichijo S, Noguchi M. Identification of biomarkers for personalized peptide vaccination in 2,588 cancer patients. Int J Oncol 2020; 56:1479-1489. [PMID: 32236612 PMCID: PMC7170040 DOI: 10.3892/ijo.2020.5019] [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: 11/29/2019] [Accepted: 03/05/2020] [Indexed: 01/08/2023] Open
Abstract
Peptide-based cancer vaccines have failed to provide sufficient clinical benefits in order to be approved in clinical trials since the 1990s. To understand the mechanisms underlying this failure, the present study investigated biomarkers associated with the lower overall survival (OS) among 2,588 patients receiving personalized peptide vaccination (PPV). Survival data were obtained from a database of 2,588 cancer patients including 399 patients with lung, 354 with prostate and 344 with colon cancer. They entered into phase II clinical trials of PPV in which 2 to 4 of 31 warehouse peptides were selected for vaccination on an individual patient basis based on human leukocyte antigen (HLA) class IA-types and pre-existing peptide-specific IgG levels. Higher pre-vaccination neutrophil, monocyte and platelet counts, and lower pre-vaccination lymphocyte and red blood cell counts were inversely associated with OS, with higher sensitivities in the proportions of neutrophils and lymphocytes, respectively. The most potent unfavorable and favorable factors for OS were the median percentage of neutrophils (>64.8%) or percentage of lymphocytes (>25.1%) with correlation coefficients (R2) of 0.98 and 0.92, respectively. Higher pre-vaccination levels of c-reactive protein and other inflammatory soluble factors were inversely associated with OS. Pre-vaccination peptide-specific immunity levels had no effect on OS, although lower immune boosting levels were inversely associated with OS. None of the 31 peptides was inversely associated with OS, although a few peptides were positively associated with it. On the whole, the findings of the present study suggested that pre-vaccination inflammatory signatures, but not those of post-vaccination immune induction, were associated with lower clinical benefits of PPV.
Collapse
Affiliation(s)
- Shigetaka Suekane
- Department of Urology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Shigeru Yutani
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839‑0863, Japan
| | - Akira Yamada
- Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Fukuoka 830‑0011, Japan
| | - Tetsuro Sasada
- Cancer Vaccine Center, Kanagawa Cancer Center, Yokohama, Kanagawa 241‑8515, Japan
| | - Satoko Matsueda
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Shinzo Takamori
- Department of Surgery, Kurume University, Kurume, Fukuoka 830‑0011, Japan
| | - Uhi Toh
- Department of Surgery, Kurume University, Kurume, Fukuoka 830‑0011, Japan
| | - Kouichiro Kawano
- Department of Obstetrics and Gynecology, Kurume University, Kurume, Fukuoka 830‑0011, Japan
| | - Koichi Yoshiyama
- Department of Surgery, Kurume University, Kurume, Fukuoka 830‑0011, Japan
| | - Shinjiro Sakamoto
- Department of Molecular and Internal Medicine School of Medicine, Hiroshima University, Hiroshima, Hiroshima 734‑8551, Japan
| | - Shunichi Sugawara
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Miyagi 980‑0873, Japan
| | - Nobukazu Komatsu
- Department of Immunology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Teppei Yamada
- Department of Gastroenterological Surgery, Fukuoka University School of Medicine, Fukuoka, Fukuoka 814‑0180, Japan
| | - Masayasu Naito
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839‑0863, Japan
| | | | - Takashi Mine
- Department of Clinical Oncology, Nagasaki Harbor Medical Center, Nagasaki, Nagasaki 850‑8555, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839‑0863, Japan
| | - Shigeki Shichijo
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839‑0863, Japan
| | - Masanori Noguchi
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839‑0863, Japan
| |
Collapse
|
212
|
Extracellular Vesicles and Tumor-Immune Escape: Biological Functions and Clinical Perspectives. Int J Mol Sci 2020; 21:ijms21072286. [PMID: 32225076 PMCID: PMC7177226 DOI: 10.3390/ijms21072286] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023] Open
Abstract
The modulation of the immune system is one of the hallmarks of cancer. It is now widely described that cancer cells are able to evade the immune response and thus establish immune tolerance. The exploration of the mechanisms underlying this ability of cancer cells has always attracted the scientific community and is the basis for the development of new promising cancer therapies. Recent evidence has highlighted how extracellular vesicles (EVs) represent a mechanism by which cancer cells promote immune escape by inducing phenotypic changes on different immune cell populations. In this review, we will discuss the recent findings on the role of tumor-derived extracellular vesicles (TEVs) in regulating immune checkpoints, focusing on the PD-L1/PD-1 axis.
Collapse
|
213
|
Maier B, Leader AM, Chen ST, Tung N, Chang C, LeBerichel J, Chudnovskiy A, Maskey S, Walker L, Finnigan JP, Kirkling ME, Reizis B, Ghosh S, D'Amore NR, Bhardwaj N, Rothlin CV, Wolf A, Flores R, Marron T, Rahman AH, Kenigsberg E, Brown BD, Merad M. A conserved dendritic-cell regulatory program limits antitumour immunity. Nature 2020; 580:257-262. [PMID: 32269339 DOI: 10.1038/s41586-020-2134-y] [Citation(s) in RCA: 481] [Impact Index Per Article: 120.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 01/17/2020] [Indexed: 11/09/2022]
Abstract
Checkpoint blockade therapies have improved cancer treatment, but such immunotherapy regimens fail in a large subset of patients. Conventional type 1 dendritic cells (DC1s) control the response to checkpoint blockade in preclinical models and are associated with better overall survival in patients with cancer, reflecting the specialized ability of these cells to prime the responses of CD8+ T cells1-3. Paradoxically, however, DC1s can be found in tumours that resist checkpoint blockade, suggesting that the functions of these cells may be altered in some lesions. Here, using single-cell RNA sequencing in human and mouse non-small-cell lung cancers, we identify a cluster of dendritic cells (DCs) that we name 'mature DCs enriched in immunoregulatory molecules' (mregDCs), owing to their coexpression of immunoregulatory genes (Cd274, Pdcd1lg2 and Cd200) and maturation genes (Cd40, Ccr7 and Il12b). We find that the mregDC program is expressed by canonical DC1s and DC2s upon uptake of tumour antigens. We further find that upregulation of the programmed death ligand 1 protein-a key checkpoint molecule-in mregDCs is induced by the receptor tyrosine kinase AXL, while upregulation of interleukin (IL)-12 depends strictly on interferon-γ and is controlled negatively by IL-4 signalling. Blocking IL-4 enhances IL-12 production by tumour-antigen-bearing mregDC1s, expands the pool of tumour-infiltrating effector T cells and reduces tumour burden. We have therefore uncovered a regulatory module associated with tumour-antigen uptake that reduces DC1 functionality in human and mouse cancers.
Collapse
Affiliation(s)
- Barbara Maier
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew M Leader
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven T Chen
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Navpreet Tung
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christie Chang
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica LeBerichel
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aleksey Chudnovskiy
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Shrisha Maskey
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura Walker
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John P Finnigan
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Margaret E Kirkling
- Department of Pathology and Department of Medicine, New York University School of Medicine, New York, NY, USA.,Graduate Program in Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Boris Reizis
- Department of Pathology and Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - Sourav Ghosh
- Department of Neurology & Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Nina Bhardwaj
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Carla V Rothlin
- Department of Immunobiology & Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Andrea Wolf
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raja Flores
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas Marron
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adeeb H Rahman
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ephraim Kenigsberg
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian D Brown
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
214
|
Quercetin induces an immunoregulatory phenotype in maturing human dendritic cells. Immunobiology 2020; 225:151929. [PMID: 32115260 DOI: 10.1016/j.imbio.2020.151929] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 02/20/2020] [Accepted: 02/22/2020] [Indexed: 12/22/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is an environmental sensor and ligand-activated transcription factor that is critically involved in the regulation of inflammatory responses and the induction of tolerance by modulating immune cells. As dendritic cells (DCs) express high AhR levels, they are efficient to induce immunomodulatory effects after being exposed to AhR-activating compounds derived from the environment or diet. To gain new insights into the molecular targets following AhR-activation in human monocyte-derived (mo)DCs, we investigated whether the natural AhR ligand quercetin or the synthetic ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) modulates the function of human moDCs regarding their capability to prime naïve T cells or to migrate. As only quercetin, but not TCDD, impaired T cell activation and migration of LPS-matured DCs (LPS-DCs), we analyzed the mode of action of quercetin on moDCs in more detail. Here, we found a specific down-regulation of the immunomodulatory molecule CD83 through the direct binding of the activated AhR to the CD83 promoter. Furthermore, treatment of LPS-DCs with quercetin resulted in a reduced production of the pro-inflammatory cytokine IL-12p70 and in an increased expression of the immunoregulatory molecules disabled adaptor protein (Dab) 2, immunoglobulin-like transcript (ILT)-3, ILT4, ILT5 as well as ectonucleotidases CD39 and CD73, thereby inducing a tolerogenic phenotype in quercetin-treated maturing DCs. Overall, these data demonstrate that quercetin represents a potent immunomodulatory agent to alter human DC phenotype and function, shifting the immune balance from inflammation to resolution.
Collapse
|
215
|
Li P, Zhao R, Fan K, Iwanowycz S, Fan H, Li Z, Liu B. Regulation of dendritic cell function improves survival in experimental sepsis through immune chaperone. Innate Immun 2020; 25:235-243. [PMID: 31018807 PMCID: PMC6830886 DOI: 10.1177/1753425919840423] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells (DCs) are professional Ag-presenting cells that play a critical
role in both innate and adaptive immune responses. DCs recognize and respond to
bacteria through multiple PRRs, including TLRs. Heat shock protein gp96/grp94 is
a master essential chaperone for TLRs in the endoplasmic reticulum. We generated
DC-specific gp96-knockout (KO) mice and showed that gp96 KO DCs were unable to
respond to multiple TLR ligands. TLR-mediated hyperinflammatory response can
lead to sepsis. However, the roles of neither DCs nor the DC-intrinsic gp96 in
the process are completely understood. In a LPS-induced sepsis model, we hereby
found that deletion of gp96 in DCs significantly reduced serum TNF-α levels and
improved survival. Furthermore, using the well-defined polymicrobial sepsis
model of cecal ligation and puncture, we found that DC-specific ablation of gp96
improved survival with significantly attenuated liver and renal injuries,
decreased circulating inflammatory cytokines, altered DC maturation and
activation, and increased serum Ig. Collectively, we demonstrate that deletion
of gp96 in DCs is beneficial in protecting mice against sepsis induced by both
endotoxemia and polymicrobial infections. We conclude that targeting gp96 in DCs
may provide a potential novel approach for reducing the morbidity and mortality
of sepsis.
Collapse
Affiliation(s)
- Pengfei Li
- 1 Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Ran Zhao
- 2 Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Kevin Fan
- 2 Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Stephen Iwanowycz
- 2 Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Hongkuan Fan
- 1 Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Zihai Li
- 2 Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Bei Liu
- 2 Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
216
|
Shao S, Cui D, Ma C, Chen P, Zhou B, Tao R, Wang J. Transcriptome profiling of tolerogenic dendritic cells conditioned with dual mTOR kinase inhibitor, AZD8055. Int Immunopharmacol 2020; 81:106241. [PMID: 32058927 DOI: 10.1016/j.intimp.2020.106241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 01/07/2020] [Accepted: 01/21/2020] [Indexed: 01/16/2023]
Abstract
Dendritic cells (DCs) can initiate and regulate adaptive immunity depending on their maturation status. Many pharmacological and genetic means have been used in the generation of immature/tolerogenic DCs. However, the key factors controlling DCs tolerogenicity remain obscure. In this work, we demonstrated that AZD8055, an ATP-competitive inhibitor of mammalian target of rapamycin (mTOR), could also lead to a tolerogenic DC phenotype from several lines of evidence, such as suppression of T cell proliferation, promoting the generation of Tregs, and inducing allogeneic T cell apoptosis. Further studies using RNA-seq method identified 430, 1172 and 1436 differentially expressed genes (DEGs) between AZD-DCs vs. Control-DCs, LPS-DCs vs. Control-DCs, and AZD-DCs vs. LPS-DCs, respectively. The 5 most differentially expressed transcripts identified by RNA-seq expression profiles were validated by quantitative RT-PCR assays. NF-κB, p38MAPK, the ribosome and PPAR signaling pathways may be involved in the induction of tolerogenic DCs by AZD8055. Functional annotation showed some genes like MGL2, Cadherin-1, 4-1BB, RhoB and Pdpn, were quite different between AZD-DCs and Control-DCs/LPS-DCs, which might be related to the tolerogenic properties of AZD-DCs. Our work provided the potential underlying molecular mechanisms involved in the generation of tolerogenic DCs. Further functional characterization of individual target gene in DC tolerogenicity will help to develop novel therapeutic modalities in circumstances like transplant tolerance induction and autoimmunity.
Collapse
Affiliation(s)
- Su Shao
- Department of General Surgery, Chunan 1st People's Hospital, Hangzhou, China
| | - Di Cui
- Center for Clinical Medical Research, Affiliated Zhejiang Provincial People's Hospital, Hangzhou Medical School, Hangzhou, China
| | - Chenyang Ma
- Center for Clinical Medical Research, Affiliated Zhejiang Provincial People's Hospital, Hangzhou Medical School, Hangzhou, China
| | - Ping Chen
- Center for Clinical Medical Research, Affiliated Zhejiang Provincial People's Hospital, Hangzhou Medical School, Hangzhou, China; Department of Gynecology, Shaoxing 2nd People's Hospital, Shaoxing, China
| | - Bing Zhou
- Center for Clinical Medical Research, Affiliated Zhejiang Provincial People's Hospital, Hangzhou Medical School, Hangzhou, China; Department of Cardiothoracic Surgery, Affiliated Zhejiang Provincial People's Hospital, Hangzhou Medical School, Hangzhou, China
| | - Ran Tao
- Center for Clinical Medical Research, Affiliated Zhejiang Provincial People's Hospital, Hangzhou Medical School, Hangzhou, China; Department of Hepatobiliary-Pancreatic Surgery, Affiliated Zhejiang Provincial People's Hospital, Hangzhou Medical School, Hangzhou, China.
| | - Jianjun Wang
- Department of General Surgery, Chunan 1st People's Hospital, Hangzhou, China.
| |
Collapse
|
217
|
Ibrutinib treatment inhibits breast cancer progression and metastasis by inducing conversion of myeloid-derived suppressor cells to dendritic cells. Br J Cancer 2020; 122:1005-1013. [PMID: 32025027 PMCID: PMC7109110 DOI: 10.1038/s41416-020-0743-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 12/26/2019] [Accepted: 01/17/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Ibrutinib is a Bruton's tyrosine kinase (BTK) and interleukin-2-inducible kinase (ITK) inhibitor used for treating chronic lymphocytic leukaemia (CLL) and other cancers. Although ibrutinib is known to inhibit the growth of breast cancer cell growth in vitro, its impact on the treatment and metastasis of breast cancer is unclear. METHODS Using an orthotopic mouse breast cancer model, we show that ibrutinib inhibits the progression and metastasis of breast cancer. RESULTS Ibrutinib inhibited proliferation of cancer cells in vitro, and Ibrutinib-treated mice displayed significantly lower tumour burdens and metastasis compared to controls. Furthermore, the spleens and tumours from Ibrutinib-treated mice contained more mature DCs and lower numbers of myeloid-derived suppressor cells (MDSCs), which promote disease progression and are linked to poor prognosis. We also confirmed that ex vivo treatment of MDSCs with ibrutinib switched their phenotype to mature DCs and significantly enhanced MHCII expression. Further, ibrutinib treatment promoted T cell proliferation and effector functions leading to the induction of antitumour TH1 and CTL immune responses. CONCLUSIONS Ibrutinib inhibits tumour development and metastasis in breast cancer by promoting the development of mature DCs from MDSCs and hence could be a novel therapeutic agent for the treatment of breast cancer.
Collapse
|
218
|
Darvekar S, Juzenas P, Oksvold M, Kleinauskas A, Holien T, Christensen E, Stokke T, Sioud M, Peng Q. Selective Killing of Activated T Cells by 5-Aminolevulinic Acid Mediated Photodynamic Effect: Potential Improvement of Extracorporeal Photopheresis. Cancers (Basel) 2020; 12:cancers12020377. [PMID: 32041351 PMCID: PMC7072277 DOI: 10.3390/cancers12020377] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 01/28/2020] [Accepted: 02/04/2020] [Indexed: 01/08/2023] Open
Abstract
Extracorporeal photopheresis (ECP), a modality that exposes isolated leukocytes to the photosensitizer 8-methoxypsoralen (8-MOP) and ultraviolet-A (UV-A) light, is used to treat conditions such as cutaneous T-cell lymphoma and graft-versus-host disease. However, the current procedure of ECP has limited selectivity and efficiency; and produces only partial response in the majority of treated patients. Additionally, the treatment is expensive and time-consuming, so the improvement for this modality is needed. In this study, we used the concept of photodynamic therapy (PDT) with 5-aminolevulinic acid (ALA), a precursor of an endogenously synthesized photosensitizer protoporphyrin IX (PpIX) in combination with blue light to explore the possibility of targeting activated human blood T cells ex vivo. With various T-cell activation protocols, a high ALA-induced PpIX production took place in activated CD3+, CD4+CD25+, and CD8+ T cell populations with their subsequent killing after blue light exposure. By contrast, resting T cells were much less damaged by the treatment. The selective and effective killing effect on the activated cells was also seen after co-cultivating activated and resting T cells. Under our clinically relevant experimental conditions, ALA-PDT killed activated T cells more selectively and efficiently than 8-MOP/UV-A. Monocyte-derived dendritic cells (DCs) were not affected by the treatment. Incubation of ALA-PDT damaged T cells with autologous DCs induced a downregulation of the co-stimulatory molecules CD80/CD86 and also upregulation of interleukin 10 (IL-10) and indoleamine 2,3-dioxygenase expression, two immunosuppressive factors that may account for the generation of tolerogenic DCs. Overall, the data support the potential use of ALA-PDT strategy for improving ECP by selective and effective killing of activated T cells and induction of immune tolerance.
Collapse
Affiliation(s)
- Sagar Darvekar
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, N-0379 Oslo, Norway; (S.D.); (P.J.); (M.O.); (A.K.); (T.H.); (E.C.)
| | - Petras Juzenas
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, N-0379 Oslo, Norway; (S.D.); (P.J.); (M.O.); (A.K.); (T.H.); (E.C.)
| | - Morten Oksvold
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, N-0379 Oslo, Norway; (S.D.); (P.J.); (M.O.); (A.K.); (T.H.); (E.C.)
| | - Andrius Kleinauskas
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, N-0379 Oslo, Norway; (S.D.); (P.J.); (M.O.); (A.K.); (T.H.); (E.C.)
| | - Toril Holien
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, N-0379 Oslo, Norway; (S.D.); (P.J.); (M.O.); (A.K.); (T.H.); (E.C.)
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, N-7491 Trondheim, Norway
- Department of Hematology, St. Olavs University Hospital HF, N-7491 Trondheim, Norway
| | - Eidi Christensen
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, N-0379 Oslo, Norway; (S.D.); (P.J.); (M.O.); (A.K.); (T.H.); (E.C.)
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, N-7491 Trondheim, Norway
- Department of Dermatology, St. Olavs University Hospital HF, N-7491 Trondheim, Norway
| | - Trond Stokke
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, N-0379 Oslo, Norway;
| | - Mouldy Sioud
- Department of Cancer Immunology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, N-0379 Oslo, Norway
- Correspondence: (M.S.); (Q.P.); Tel.: +47-22781414 (M.S.); +47-22782353 (Q.P.)
| | - Qian Peng
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, N-0379 Oslo, Norway; (S.D.); (P.J.); (M.O.); (A.K.); (T.H.); (E.C.)
- Department of Optical Science and Engineering, The School of Information Science and Technology, Fudan University, Shanghai 200433, China
- Correspondence: (M.S.); (Q.P.); Tel.: +47-22781414 (M.S.); +47-22782353 (Q.P.)
| |
Collapse
|
219
|
Kimura TFE, Romera LMD, de Almeida SR. Fonsecaea pedrosoi Conidia Induces Activation of Dendritic Cells and Increases CD11c + Cells in Regional Lymph Nodes During Experimental Chromoblastomycosis. Mycopathologia 2020; 185:245-256. [PMID: 32008205 DOI: 10.1007/s11046-020-00429-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/21/2020] [Indexed: 10/20/2022]
Abstract
The chromoblastomycosis is a subcutaneous mycosis with a high morbidity rate, Fonsecaea pedrosoi being the largest etiologic agent of this mycosis, usually confined to the skin and subcutaneous tissues. Rarely people get the cure, because the therapies shown to be deficient and few studies report the host-parasite relationship. Dendritic cells (DCs) are specialized in presenting antigens to naïve T lymphocytes inducing primary immune responses. Therefore, we propose to study the migratory capacity of DCs after infection with conidia of F. pedrosoi. The phenotype of DCs was evaluated using cells obtained from footpad and lymph nodes of BALB/c mice after 12, 24 and 72 h of infection. After 24 and 72 h of infection, we found a significant decrease in DCs in footpad and a significant increase in the lymph nodes after 72 h. The expression of surface markers and co-stimulatory molecules were reduced in cells obtained from footpad. To better assess the migratory capacity of DCs migration from footpad, CFSE-stained conidia were injected subcutaneously. We found that after 12 and 72 h, CD11c+ cells were increased in regional lymph nodes, leading us to believe that DCs (CD11c+) were able to phagocytic conidia present in footpad and migrated to regional lymph nodes.
Collapse
Affiliation(s)
- Telma Fátima Emidio Kimura
- Laboratory of Mycology, Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Scienses, Universidade de Sao Paulo, São Paulo, Brazil
| | - Lavínia Maria Dal'Mas Romera
- Laboratory of Mycology, Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Scienses, Universidade de Sao Paulo, São Paulo, Brazil.
| | - Sandro Rogério de Almeida
- Laboratory of Mycology, Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Scienses, Universidade de Sao Paulo, São Paulo, Brazil
| |
Collapse
|
220
|
|
221
|
Madelon N, Montanari E, Gruaz L, Pimenta J, Muller YD, Bühler LH, Puga Yung GL, Seebach JD. Prolongation of rat-to-mouse islets xenograft survival by co-transplantation of autologous IL-10 differentiated murine tolerogenic dendritic cells. Xenotransplantation 2020; 27:e12584. [PMID: 31984564 DOI: 10.1111/xen.12584] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/06/2019] [Accepted: 01/05/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Tolerogenic dendritic cells (DCs) represent a promising approach to promote transplantation tolerance. In this study, the potential of autologous bone marrow (BM)-derived murine DC to protect rat-to-mouse islets xenografts was analyzed. METHODS Tolerogenic DCs were generated by differentiating BM cells in the presence of granulocyte-macrophage colony-stimulating factor and interleukin 10 (IL-10, IL-10 DC). The phenotype of IL-10 DC was characterized in vitro by expression of costimulatory/inhibitory molecules (flow cytometry) and cytokines (Luminex and ELISA), their function by phagocytosis and T-cell stimulation assays. To study transplant tolerance in vivo, rat islets were transplanted alone or in combination with autologous murine IL-10 DC under the kidney capsule of streptozotocin-induced diabetic C57BL/6 mice. Xenograft survival was evaluated by monitoring glycemia, cellular infiltration of xenografts by microscopy and flow cytometry 10 days post-transplantation. RESULTS Compared with control DC, IL-10 DC exhibited lower levels of major histocompatibility complex class II, costimulatory molecules (CD40, CD86, CD205), lower production of pro-inflammatory cytokines (IL-12p70, TNF, IL-6), and higher production of IL-10. Phagocytosis of xenogeneic rat splenocytes was not impaired in IL-10 DC, whereas stimulation of T-cell proliferation was reduced in the presence of IL-10 DC. Xenograft survival of rat islets in diabetic mice co-transplanted with autologous murine IL-10 DC was significantly prolonged from 12 to 21 days, without additional immunosuppressive treatment. Overall, infiltration of xenografts by T cells and myeloid cells was not different in IL-10 DC recipient mice, but enriched for CD8+ T cells and myeloid cells with suppressor-associated phenotype. CONCLUSIONS Autologous IL-10-differentiated DC with tolerogenic properties prolong rat-to-mouse islets xenograft survival, potentially by locally inducing immune regulatory cells, indicating their potential for regulatory immune cell therapy in xenotransplantation.
Collapse
Affiliation(s)
- Natacha Madelon
- Laboratory of Translational Immunology, Division of Immunology and Allergology, Department of Medical Specialties, Medical Faculty, Geneva University Hospitals, Geneva, Switzerland
| | - Elisa Montanari
- Department of Surgery, Medical Faculty, Cell Isolation and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Lyssia Gruaz
- Laboratory of Translational Immunology, Division of Immunology and Allergology, Department of Medical Specialties, Medical Faculty, Geneva University Hospitals, Geneva, Switzerland
| | - Joel Pimenta
- Department of Surgery, Medical Faculty, Cell Isolation and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Yannick D Muller
- Laboratory of Translational Immunology, Division of Immunology and Allergology, Department of Medical Specialties, Medical Faculty, Geneva University Hospitals, Geneva, Switzerland
| | - Leo H Bühler
- Department of Surgery, Medical Faculty, Cell Isolation and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Gisella L Puga Yung
- Laboratory of Translational Immunology, Division of Immunology and Allergology, Department of Medical Specialties, Medical Faculty, Geneva University Hospitals, Geneva, Switzerland
| | - Jörg D Seebach
- Laboratory of Translational Immunology, Division of Immunology and Allergology, Department of Medical Specialties, Medical Faculty, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
222
|
Link C, Bujupi F, Krammer PH, Weyd H. Annexin-coated particles induce antigen-specific immunosuppression. Autoimmunity 2020; 53:86-94. [PMID: 31933381 DOI: 10.1080/08916934.2019.1710134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Apoptotic cells mediate the development of tolerogenic dendritic cells (DC) and thus facilitate induction and maintenance of peripheral tolerance. Following the identification of the evolutionary conserved annexin core domain (Anx) as a specific signal on apoptotic cells which antagonises Toll-like receptor (TLR) signalling, we examined whether the tolerogenic capacity of Anx can be exploited to downregulate antigen-specific immune responses. The treatment of bone marrow-derived dendritic cells (BMDC) with particles harbouring Anx as well as the model antigen ovalbumin (OVA) attenuated the response of OVA-specific OT-II T cells. The co-culture of Anx-particle-treated DC and T cells resulted in an anergy-like phenotype characterized by reduced proliferation and cytokine secretion. Here we demonstrate that the anti-inflammatory effects of Anx which are mediated through DC can be used as a tool to generate a particle-based antigen delivery system that promotes antigen-specific immunosuppression. Such Anx-particles may be a new therapeutic approach for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Corinna Link
- Division of Immunogenetics, German Cancer Research Center, Heidelberg, Germany.,Faculty of Biosciences, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Fatmire Bujupi
- Division of Immunogenetics, German Cancer Research Center, Heidelberg, Germany.,Faculty of Biosciences, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Peter H Krammer
- Division of Immunogenetics, German Cancer Research Center, Heidelberg, Germany
| | - Heiko Weyd
- Division of Immunogenetics, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
223
|
Salminen A. Activation of immunosuppressive network in the aging process. Ageing Res Rev 2020; 57:100998. [PMID: 31838128 DOI: 10.1016/j.arr.2019.100998] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/29/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022]
Abstract
Chronic low-grade inflammation has a key role in the aging process, a state called inflammaging. It is known that the chronic inflammatory condition generates counteracting immunosuppressive state in many diseases. Inflammaging is also associated with an immune deficiency; generally termed as immunosenescence, although it is not known whether it represents the senescence of immune cells or the active remodeling of immune system. Evidence has accumulated since the 1970's indicating that immunosenescence might be caused by an increased activity of immunosuppressive cells rather than cellular senescence. Immune cells display remarkable plasticity; many of these cells can express both proinflammatory and immunosuppressive phenotypes in a context-dependent manner. The immunosuppressive network involves the regulatory subtypes of T (Treg) and B (Breg) cells as well as regulatory phenotypes of macrophages (Mreg), dendritic (DCreg), natural killer (NKreg), and type II natural killer T (NKT) cells. The immunosuppressive network also includes monocytic (M-MDSC) and polymorphonuclear (PMN-MDSC) myeloid-derived suppressor cells which are immature myeloid cells induced by inflammatory mediators. This co-operative network is stimulated in chronic inflammatory conditions preventing excessive inflammatory responses but at the same time they exert harmful effects on the immune system and tissue homeostasis. Recent studies have revealed that the aging process is associated with the activation of immunosuppressive network, especially the functions of MDSCs, Tregs, and Mregs are increased. I will briefly review the properties of the regulatory phenotypes of immune cells and examine in detail the evidences for an activation of immunosuppressive network with aging.
Collapse
|
224
|
Sioud M. Unleashing the Therapeutic Potential of Dendritic and T Cell Therapies Using RNA Interference. Methods Mol Biol 2020; 2115:259-280. [PMID: 32006406 DOI: 10.1007/978-1-0716-0290-4_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Therapeutic dendritic cell (DC) cancer vaccines work to boost the body's immune system to fight a cancer. Although this type of immunotherapy often leads to the activation of tumor-specfic T cells, clinical responses are fairly low, arguing for the need to improve the design of DC-based vaccines. Recent studies revealed a promising strategy of combining DC vaccines with small interfering RNAs (siRNAs) targeting immunosuppressive signals such as checkpoint receptors. Similarly, incorporating checkpoint siRNA blockers in adoptive T-cell therapy to amplify cytotoxic T lymphocyte responses is now being tested in the clinic. The development of the next generation of cancer immunotherapies using siRNA technology will hopefuly benefit patients with various cancer types including those who did not respond to current therapies. This review highlights the latest advances in RNA interference technology to improve the therapeutic efficacy of DC cancer vaccines and T cell therapy.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Ullernchausseen 70, Oslo, Norway.
| |
Collapse
|
225
|
E-Cadherin is Dispensable to Maintain Langerhans Cells in the Epidermis. J Invest Dermatol 2020; 140:132-142.e3. [DOI: 10.1016/j.jid.2019.06.132] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/28/2019] [Accepted: 06/17/2019] [Indexed: 11/18/2022]
|
226
|
Baxevanis CN, Fortis SP, Perez SA. The balance between breast cancer and the immune system: Challenges for prognosis and clinical benefit from immunotherapies. Semin Cancer Biol 2019; 72:76-89. [PMID: 31881337 DOI: 10.1016/j.semcancer.2019.12.018] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
Abstract
Cancer evolution is a complex process influenced by genetic factors and extracellular stimuli that trigger signaling pathways to coordinate the continuous and dynamic interaction between tumor cells and the elements of the immune system. For over 20 years now, the immune mechanisms controlling cancer progression have been the focus of intensive research. It is well established that the immune system conveys protective antitumor immunity by destroying immunogenic tumor variants, but also facilitates tumor progression by shaping tumor immunogenicity in a process called "immunoediting". It is also clear that immune-guided tumor editing is associated with tumor evasion from immune surveillance and therefore reinforcing the endogenous antitumor immunity is a desired goal in the context of cancer therapies. The tumor microenvironment (TME) is a complex network which consists of various cell types and factors having important roles regarding tumor development and progression. Tumor infiltrating lymphocytes (TILs) and other tumor infiltrating immune cells (TIICs) are key to our understanding of tumor immune surveillance based on tumor immunogenicity, whereby the densities and location of TILs and TIICs in the tumor regions, as well as their functional programs (comprising the "immunoscore") have a prominent role for prognosis and prediction for several cancers. The presence of tertiary lymphoid structures (TLS) in the TME or in peritumoral areas has an influence on the locally produced antitumor immune response, and therefore also has a significant prognostic impact. The cross-talk between elements of the immune system with tumor cells in the TME is greatly influenced by hypoxia, the gut and/or the local microbiota, and several metabolic elements, which, in a dynamic interplay, have a crucial role for tumor cell heterogeneity and reprogramming of immune cells along their activation and differentiation pathways. Taking into consideration the recent clinical success with the application immunotherapies for the treatment of several cancer types, increasing endeavors have been made to gain better insights into the mechanisms underlying phenotypic and metabolic profiles in the context of tumor progression and immunotherapy. In this review we will address (i) the role of TILs, TIICs and TLS in breast cancer (BCa); (ii) the different metabolic-based pathways used by immune and breast cancer cells; and (iii) implications for immunotherapy-based strategies in BCa.
Collapse
Affiliation(s)
- Constantin N Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras Ave., 11522, Athens, Greece.
| | - Sotirios P Fortis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras Ave., 11522, Athens, Greece
| | - Sonia A Perez
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras Ave., 11522, Athens, Greece
| |
Collapse
|
227
|
Nataf S, Guillen M, Pays L. Common Neurodegeneration-Associated Proteins Are Physiologically Expressed by Human B Lymphocytes and Are Interconnected via the Inflammation/Autophagy-Related Proteins TRAF6 and SQSTM1. Front Immunol 2019; 10:2704. [PMID: 31824497 PMCID: PMC6886494 DOI: 10.3389/fimmu.2019.02704] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
There is circumstantial evidence that, under neurodegenerative conditions, peptides deriving from aggregated or misfolded specific proteins elicit adaptive immune responses. On another hand, several genes involved in familial forms of neurodegenerative diseases exert key innate immune functions. However, whether or not such observations are causally linked remains unknown. To start addressing this issue, we followed a systems biology strategy based on the mining of large proteomics and immunopeptidomics databases. First, we retrieved the expression patterns of common neurodegeneration-associated proteins in two professional antigen-presenting cells, namely B lymphocytes and dendritic cells. Surprisingly, we found that under physiological conditions, numerous neurodegeneration-associated proteins are abundantly expressed by human B lymphocytes. A survey of the human proteome allowed us to map a unique protein-protein interaction network linking common neurodegeneration-associated proteins and their first shell interactors in human B lymphocytes. Interestingly, network connectivity analysis identified two major hubs that both relate with inflammation and autophagy, namely TRAF6 (TNF Receptor Associated Factor 6) and SQSTM1 (Sequestosome-1). Moreover, the mapped network in B lymphocytes comprised two additional hub proteins involved in both inflammation and autoimmunity: HSPA8 (Heat Shock Protein Family A Member 8 also known as HSC70) and HSP90AA1 (Heat Shock Protein 90 Alpha Family Class A Member 1). Based on these results, we then explored the Immune Epitope Database "IEDB-AR" and actually found that a large share of neurodegeneration-associated proteins were previously reported to provide endogenous MHC class II-binding peptides in human B lymphocytes. Of note, peptides deriving from amyloid beta A4 protein, sequestosome-1 or profilin-1 were reported to bind multiple allele-specific MHC class II molecules. In contrast, peptides deriving from microtubule-associated protein tau, presenilin 2 and serine/threonine-protein kinase TBK1 were exclusively reported to bind MHC molecules encoded by the HLA-DRB1 1501 allele, a recently-identified susceptibility gene for late onset Alzheimer's disease. Finally, we observed that the whole list of proteins reported to provide endogenous MHC class II-binding peptides in human B lymphocytes is specifically enriched in neurodegeneration-associated proteins. Overall, our work indicates that immunization against neurodegeneration-associated proteins might be a physiological process which is shaped, at least in part, by B lymphocytes.
Collapse
Affiliation(s)
- Serge Nataf
- CarMeN Laboratory, INSERM U1060, INRA U1397, INSA de Lyon, Lyon-Sud Faculty of Medicine, University of Lyon, Pierre-Bénite, France
- Faculté de Médecine Lyon-Est, University of Lyon 1, Lyon, France
- Banque de Tissus et de Cellules des Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon, France
| | - Marine Guillen
- Faculté de Médecine Lyon-Est, University of Lyon 1, Lyon, France
| | - Laurent Pays
- CarMeN Laboratory, INSERM U1060, INRA U1397, INSA de Lyon, Lyon-Sud Faculty of Medicine, University of Lyon, Pierre-Bénite, France
- Faculté de Médecine Lyon-Est, University of Lyon 1, Lyon, France
- Banque de Tissus et de Cellules des Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon, France
| |
Collapse
|
228
|
Wei Y, Lan Y, Zhong Y, Yu K, Xu W, Zhu R, Sun H, Ding Y, Wang Y, Zeng Q. Interleukin-38 alleviates cardiac remodelling after myocardial infarction. J Cell Mol Med 2019; 24:371-384. [PMID: 31746138 PMCID: PMC6933378 DOI: 10.1111/jcmm.14741] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/12/2019] [Accepted: 09/23/2019] [Indexed: 12/21/2022] Open
Abstract
Excessive immune‐mediated inflammatory reaction plays a deleterious role in ventricular remodelling after myocardial infarction (MI). Interleukin (IL)‐38 is a newly characterized cytokine of the IL‐1 family and has been reported to exert a protective effect in some autoimmune diseases. However, its role in cardiac remodelling post‐MI remains unknown. In this study, we found that the expression of IL‐38 was increased in infarcted heart after MI induced in C57BL/6 mice by permanent ligation of the left anterior descending artery. In addition, our data showed that ventricular remodelling after MI was significantly ameliorated after recombinant IL‐38 injection in mice. This amelioration was demonstrated by better cardiac function, restricted inflammatory response, attenuated myocardial injury and decreased myocardial fibrosis. Our results in vitro revealed that IL‐38 affects the phenotype of dendritic cells (DCs) and IL‐38 plus troponin I (TNI)‐treated tolerogenic DCs dampened adaptive immune response when co‐cultured with CD4+T cells. In conclusion, IL‐38 plays a protective effect in ventricular remodelling post‐MI, one possibility by influencing DCs to attenuate inflammatory response. Therefore, targeting IL‐38 may hold a new therapeutic potential in treating MI.
Collapse
Affiliation(s)
- Yuzhen Wei
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Yin Lan
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Yucheng Zhong
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Kunwu Yu
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Wenbin Xu
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Ruirui Zhu
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Haitao Sun
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Yan Ding
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Yue Wang
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Qiutang Zeng
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| |
Collapse
|
229
|
Han P, Hanlon D, Sobolev O, Chaudhury R, Edelson RL. Ex vivo dendritic cell generation-A critical comparison of current approaches. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:251-307. [PMID: 31759433 DOI: 10.1016/bs.ircmb.2019.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells, required for the initiation of naïve and memory T cell responses and regulation of adaptive immunity. The discovery of DCs in 1973, which culminated in the Nobel Prize in Physiology or Medicine in 2011 for Ralph Steinman and colleagues, initially focused on the identification of adherent mononuclear cell fractions with uniquely stellate dendritic morphology, followed by key discoveries of their critical immunologic role in initiating and maintaining antigen-specific immunity and tolerance. The medical promise of marshaling these key capabilities of DCs for therapeutic modulation of antigen-specific immune responses has guided decades of research in hopes to achieve genuine physiologic partnership with the immune system. The potential uses of DCs in immunotherapeutic applications include cancer, infectious diseases, and autoimmune disorders; thus, methods for rapid and reliable large-scale production of DCs have been of great academic and clinical interest. However, difficulties in obtaining DCs from lymphoid and peripheral tissues, low numbers and poor survival in culture, have led to advancements in ex vivo production of DCs, both for probing molecular details of DC function as well as for experimenting with their clinical utility. Here, we review the development of a diverse array of DC production methodologies, ranging from cytokine-based strategies to genetic engineering tools devised for enhancing DC-specific immunologic functions. Further, we explore the current state of DC therapies in clinic, as well as emerging insights into physiologic production of DCs inspired by existing therapies.
Collapse
Affiliation(s)
- Patrick Han
- Department of Chemical and Environmental Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT, United States
| | - Douglas Hanlon
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT, United States
| | - Olga Sobolev
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT, United States
| | - Rabib Chaudhury
- Department of Chemical and Environmental Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT, United States
| | - Richard L Edelson
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT, United States.
| |
Collapse
|
230
|
Deslyper G, Doherty DG, Carolan JC, Holland CV. The role of the liver in the migration of parasites of global significance. Parasit Vectors 2019; 12:531. [PMID: 31703729 PMCID: PMC6842148 DOI: 10.1186/s13071-019-3791-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 11/04/2019] [Indexed: 02/08/2023] Open
Abstract
Many parasites migrate through different tissues during their life-cycle, possibly with the aim to enhance their fitness. This is true for species of three parasite genera of global importance, Ascaris, Schistosoma and Plasmodium, which cause significant global morbidity and mortality. Interestingly, these parasites all incorporate the liver in their life-cycle. The liver has a special immune status being able to preferentially induce tolerance over immunity. This function may be exploited by parasites to evade host immunity, with Plasmodium spp. in particular using this organ for its multiplication. However, hepatic larval attrition occurs in both ascariasis and schistosomiasis. A better understanding of the molecular mechanisms involved in hepatic infection could be useful in developing novel vaccines and therapies for these parasites.
Collapse
Affiliation(s)
- Gwendoline Deslyper
- Department of Zoology, School of Natural Sciences, Trinity College Dublin, Dublin 2, Ireland.
| | - Derek G Doherty
- School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - James C Carolan
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Celia V Holland
- Department of Zoology, School of Natural Sciences, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
231
|
Sun XM, Guo K, Hao CY, Zhan B, Huang JJ, Zhu X. Trichinella spiralis Excretory-Secretory Products Stimulate Host Regulatory T Cell Differentiation through Activating Dendritic Cells. Cells 2019; 8:cells8111404. [PMID: 31703440 PMCID: PMC6912532 DOI: 10.3390/cells8111404] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/05/2019] [Accepted: 10/24/2019] [Indexed: 01/08/2023] Open
Abstract
Trichinella spiralis maintains chronic infections within its host, involving a variety of immunomodulatory properties, the mechanisms of which have not been completely elucidated. In this study, we found that T. spiralis infection induced strong regulatory T cell responses through parasite excretory-secretory (ES) products, characterized by increase of CD4+CD25+Foxp3+ and CD4+CD25-Foxp3+ Treg cells accompanied by high levels of IL-10 and TGF-β. T. spiralis adult worm excretory-secretory products (AES) and muscle larvae excretory-secretory products (MES) were both able to activate BMDCs in vitro to facilitate their maturation and to create regulatory cytokines IL-10 and TGF-β. The T. spiralis AES- and MES-pulsed dendritic cells (DCs) possessed abilities not only to present antigens to sensitized CD4+ T cell to stimulate their proliferation but also to induce naive CD4+ T cells to differentiate to Treg cells secreting IL-10 and TGF-β. The passive transfer of T. spiralis AES- and MES-pulsed bone marrow-derived dendritic cells (BMDCs) conferred the naive mice to acquire the differentiation of Treg cells. T. spiralis AES possesses a better ability to induce Treg cells than did MES, although the latter has the ability to induce CD4+CD25-Foxp3+ Treg cells. The results obtained in this study suggested that T. spiralis ES products stimulate the differentiation of host Treg cells possibly through activating dendritic cells to create a regulatory environment that benefits the survival of the parasite in the host.
Collapse
Affiliation(s)
- Xi-Meng Sun
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Kai Guo
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Chun-Yue Hao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Bin Zhan
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jing-Jing Huang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xinping Zhu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Correspondence:
| |
Collapse
|
232
|
Lesch S, Benmebarek MR, Cadilha BL, Stoiber S, Subklewe M, Endres S, Kobold S. Determinants of response and resistance to CAR T cell therapy. Semin Cancer Biol 2019; 65:80-90. [PMID: 31705998 DOI: 10.1016/j.semcancer.2019.11.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/28/2019] [Accepted: 11/03/2019] [Indexed: 12/27/2022]
Abstract
The remarkable success of chimeric antigen receptor (CAR)-engineered T cells in pre-B cell acute lymphoblastic leukemia (ALL) and B cell lymphoma led to the approval of anti-CD19 CAR T cells as the first ever CAR T cell therapy in 2017. However, with the number of CAR T cell-treated patients increasing, observations of tumor escape and resistance to CAR T cell therapy with disease relapse are demonstrating the current limitations of this therapeutic modality. Mechanisms hampering CAR T cell efficiency include limited T cell persistence, caused for example by T cell exhaustion and activation-induced cell death (AICD), as well as therapy-related toxicity. Furthermore, the physical properties, antigen heterogeneity and immunosuppressive capacities of solid tumors have prevented the success of CAR T cells in these entities. Herein we review current obstacles of CAR T cell therapy and propose strategies in order to overcome these hurdles and expand CAR T cell therapy to a broader range of cancer patients.
Collapse
Affiliation(s)
- Stefanie Lesch
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Mohamed-Reda Benmebarek
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Bruno L Cadilha
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Stefan Stoiber
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Marion Subklewe
- German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany; Department of Medicine III, Klinikum der Universität München, LMU Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany; German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany; German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany.
| |
Collapse
|
233
|
Keratinocytes Share Gene Expression Fingerprint with Epidermal Langerhans Cells via mRNA Transfer. J Invest Dermatol 2019; 139:2313-2323.e8. [DOI: 10.1016/j.jid.2019.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 01/13/2023]
|
234
|
Stojić-Vukanić Z, Pilipović I, Bufan B, Stojanović M, Leposavić G. Age and sex determine CD4+ T cell stimulatory and polarizing capacity of rat splenic dendritic cells. Biogerontology 2019; 21:83-107. [PMID: 31646402 DOI: 10.1007/s10522-019-09845-y] [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] [Received: 08/06/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022]
Abstract
The study investigated influence of sex and age on splenic myeloid dendritic cells (DCs) from Dark Agouti rats. Freshly isolated DCs from young males exhibited less mature phenotype and greater endocytic capacity compared with those from age-matched females. Upon LPS stimulation in vitro they were less potent in stimulating allogeneic CD4+ cells in mixed leukocyte reaction (MLR), due to lower expression of MHC II, and greater NO and IL-10 production. In accordance with higher TGF-β production, young male rat DCs were less potent in stimulating IL-17 production in MLR than those from young females. Irrespective of sex, endocytic capacity and responsiveness of DCs to LPS stimulation in culture, judging by their allostimulatory capacity in MLR decreased with age, reflecting decline in MHC II surface density followed by their greater NO production; the effects more prominent in females. Additionally, compared with LPS-stimulated DCs from young rats, those from sex-matched aged rats were more potent in stimulating IL-10 production in MLR, whereas capacity of DCs from aged female and male rats to stimulate IL-17 production remained unaltered and decreased, respectively. This reflected age-related shift in IL-6/TGF-β production level ratio in LPS-stimulated DC cultures towards TGF-β, and sex-specific age-related remodeling CD4+ cell cytokine pathways. Additionally, compared with LPS-stimulated DCs from young rats, those cells from sex-matched aged rats were less potent in stimulating IFN-γ production in MLR, the effect particularly prominent in MLRs encompassing male rat DCs. The study showed that stimulatory and polarizing capacity of DCs depends on rat sex and age.
Collapse
Affiliation(s)
- Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, Belgrade, 11221, Serbia
| | - Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, Belgrade, 11221, Serbia
| | - Biljana Bufan
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, Belgrade, 11221, Serbia
| | - Marija Stojanović
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, Belgrade, 11221, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, Belgrade, 11221, Serbia.
| |
Collapse
|
235
|
Verbeek JS, Hirose S, Nishimura H. The Complex Association of FcγRIIb With Autoimmune Susceptibility. Front Immunol 2019; 10:2061. [PMID: 31681256 PMCID: PMC6803437 DOI: 10.3389/fimmu.2019.02061] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 08/15/2019] [Indexed: 12/20/2022] Open
Abstract
FcγRIIb is the only inhibitory Fc receptor and controls many aspects of immune and inflammatory responses. The observation 19 years ago that Fc γ RIIb -/- mice generated by gene targeting in 129 derived ES cells developed severe lupus like disease when backcrossed more than 7 generations into C57BL/6 background initiated extensive research on the functional understanding of this strong autoimmune phenotype. The genomic region in the distal part of Chr1 both in human and mice in which the Fc γ R gene cluster is located shows a high level of complexity in relation to the susceptibility to SLE. Specific haplotypes of closely linked genes including the Fc γ RIIb and Slamf genes are associated with increased susceptibility to SLE both in mice and human. Using forward and reverse genetic approaches including in human GWAS and in mice congenic strains, KO mice (germline and cell type specific, on different genetic background), knockin mice, overexpressing transgenic mice combined with immunological models such as adoptive transfer of B cells from Ig transgenic mice the involved genes and the causal mutations and their associated functional alterations were analyzed. In this review the results of this 19 years extensive research are discussed with a focus on (genetically modified) mouse models.
Collapse
Affiliation(s)
- J Sjef Verbeek
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| | - Sachiko Hirose
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| | - Hiroyuki Nishimura
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| |
Collapse
|
236
|
IL-37 Plays a Beneficial Role in Patients with Acute Coronary Syndrome. Mediators Inflamm 2019; 2019:9515346. [PMID: 31686988 PMCID: PMC6803729 DOI: 10.1155/2019/9515346] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 08/10/2019] [Accepted: 08/22/2019] [Indexed: 12/13/2022] Open
Abstract
Background Interleukin-37 (IL-37) acts as an inhibitor of innate and adaptive immunity. However, the exact role of IL-37 in the patients with acute coronary syndrome (ACS) remains to be elucidated. Methods Patients were classified into 4 groups: normal coronary artery (NCA), stable angina (SA), unstable angina (UA), and acute myocardial infarction (AMI). The circulating Treg, Th1, and Th17 frequencies were measured. The effect of IL-37 on stimulated peripheral blood mononuclear cells (PBMCs) and the influence of IL-37 on DCs were explored. In addition, the role of IL-37-treated tDCs on Treg cell expansion and the stability of these tDCs were also tested. Results Our results showed that the circulating Treg frequencies were decreased, while Th1 and Th17 frequencies were increased in ACS patients, and that IL-37 expanded Tregs but suppressed Th1 and Th17 cells in activated PBMCs derived from ACS patients. Of note, IL-37-treated human DCs obtained a tolerogenic phenotype, and such tDCs promoted expansion of Tregs and decreased the Th1 and Th17 populations when cocultured with CD4+ T cells. Interestingly, IL-37-treated DCs from patients with ACS are phenotypically and functionally comparable to IL-37-treated DCs from NCA patients, and tolerogenic properties of IL-37-treated DCs were highly stable. Conclusion In conclusion, our results reveal a beneficial role of IL-37 in the patients with ACS and suggest that autologous IL-37-treated tDCs may be a novel therapeutic strategy for the patients with ACS.
Collapse
|
237
|
Farag MM, Peschel G, Müller M, Weigand K. Characterization Of The Interaction Between Subviral Particles Of Hepatitis B Virus And Dendritic Cells - In Vitro Study. Infect Drug Resist 2019; 12:3125-3135. [PMID: 31632101 PMCID: PMC6789970 DOI: 10.2147/idr.s221294] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 08/28/2019] [Indexed: 12/21/2022] Open
Abstract
Background During an infection with hepatitis B virus (HBV), infectious particles (Dane particles) can be detected in addition to aggregates of the subviral particles (SVP) which is considered an immune escaping mechanism for the virus. Dendritic cells (DCs) are a specialized type of antigen-presenting cell (APC) that can activate native T-cells to prime an immune response controlling HBV infection. The aim of this study was to characterize the interaction between HBVsvp and DCs in vitro. Methods HBVsvp that comprises surface and core proteins were produced in vitro by HepG2.2.15 as a culturing system; DCs derived from the bone marrow of mice were pulsed by HBVsvp. A different pattern of cytokines secreted by bone-marrow-derived dendritic cells from C56BL/6 mice pulsed with HBVsvp were analyzed. The interactions between HBVsvp and DCs were characterized using FACS analysis, protein assay, Western blot, and immunofluorescence staining. Results Pulsation of DCs with HBVsvp resulted in strong activation and higher secretion of DC cytokines including INF-α, INF-γ, TNF-α, IL-1α, IL-10, and IL-12; but not for IL-1β, IL-2, IL-6, and IL-15. The production of CXCL-10/IP-10 was increased during the observation period and reached the maximal secretion after 24 hrs (p < 0.001). In total protein assay, we found significantly higher protein concentration in HBVsvp stimulated DC groups compared to not activated DCs (p < 0.001). Both 24 kDa small surface antigen (HBVs) and the 21 kDa core protein (HBVc) were detected in activated DCs. For DCs immunofluorescence staining, our data showed clear differences in the morphology of DCs between negative control and those pulsed with HBVsvp. Conclusion Result demonstrates a significant complex interaction between HBVsvp and DCs, in vitro.
Collapse
Affiliation(s)
- Mohamed Ms Farag
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, Egypt
| | - Georg Peschel
- Department of Gastroenterology, Endocrinology, Rheumatology and Infectious Diseases, University Hospital Regensburg, Regensburg 93053, Germany
| | - Martina Müller
- Department of Gastroenterology, Endocrinology, Rheumatology and Infectious Diseases, University Hospital Regensburg, Regensburg 93053, Germany
| | - Kilian Weigand
- Department of Gastroenterology, Endocrinology, Rheumatology and Infectious Diseases, University Hospital Regensburg, Regensburg 93053, Germany
| |
Collapse
|
238
|
Dika IE, Khalil DN, Abou-Alfa GK. Immune checkpoint inhibitors for hepatocellular carcinoma. Cancer 2019; 125:3312-3319. [PMID: 31290997 PMCID: PMC7944520 DOI: 10.1002/cncr.32076] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 01/12/2019] [Accepted: 01/18/2019] [Indexed: 12/20/2022]
Abstract
The position of immunotherapy as a pillar of systemic cancer treatment has been firmly established over the past decade. Immune checkpoint inhibitors are a welcome option for patients with different malignancies. This is in part because they offer the possibility of durable benefit, even for patients who have failed other treatment modalities. The recent demonstration that immunotherapy is effective for patients with hepatocellular carcinoma (HCC) is a milestone in the history of this recalcitrant disease. The treatment of HCC has been a challenge, and for many years was limited to the tyrosine kinase inhibitor sorafenib and to several novel tyrosine kinase inhibitors that have shown efficacy and have been approved. The current role of immune checkpoint inhibitors in the management of HCC, and how this role is likely to evolve in the years ahead, are key. Other than efforts evaluating single checkpoint inhibitors, potential combination strategies, including combinations with existing local and systemic approaches, including novel therapies are evolving. This is understandably of special interest considering the potential unique immune system of the liver, which may impact the use of immunotherapy in patients with HCC going forward, and how can it be enhanced further.
Collapse
Affiliation(s)
- Imane El Dika
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| | - Danny N. Khalil
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ghassan K. Abou-Alfa
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| |
Collapse
|
239
|
Choi HG, Oh DJ, Kim M, Kim S, Min C, Kong IG. Appendectomy and rheumatoid arthritis: A longitudinal follow-up study using a national sample cohort. Medicine (Baltimore) 2019; 98:e17153. [PMID: 31577706 PMCID: PMC6783172 DOI: 10.1097/md.0000000000017153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The present study evaluated the association between appendectomy and rheumatoid arthritis (RA) using a national sample cohort of the Korean population. In this cohort study, the Korean National Health Insurance Service-National Sample Cohort of individuals ≥20 years old was collected from 2002 to 2013. A total of 14,995 appendectomy participants were 1:4 matched with 59,980 control subjects for age, group, sex, income group, region of residence, hypertension, diabetes, and dyslipidemia. We analyzed the occurrence of RA in both the appendectomy and control groups. Appendectomies were identified using operation codes for appendicitis only. RA was defined by International Classification of Disease-10 codes (M05 or M06) and medication histories. Crude and adjusted hazard ratios (HRs) were analyzed using a stratified Cox proportional hazard model. Subgroup analyses were performed on groups stratified by age and sex. The adjusted HR for RA was 1.02 (95% confidence interval = 0.76-1.38) in the appendectomy group (P = .883). In all of the subgroup analyses according to age and sex, the adjusted HRs for RA were not higher in the appendectomy group than those in the control group. We could not identify any significant relationship between appendectomy and RA.
Collapse
Affiliation(s)
- Hyo Geun Choi
- Department of Otorhinolaryngology-Head & Neck Surgery, Hallym University College of Medicine, Anyang
| | - Dong Jun Oh
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul
| | - Miyoung Kim
- Department of Laboratory Medicine, Hallym University College of Medicine, Anyang
| | - Sunmi Kim
- Department of Family Medicine, Kangwon National University College of Medicine, Chuncheon, Gangwon-do
| | - Chanyang Min
- Hallym Data Science Laboratory, Hallym University College of Medicine, Anyang
- Graduate School of Public Health, Seoul National University
| | - Il Gyu Kong
- Department of Otorhinolaryngology-Head & Neck Surgery, Hallym University College of Medicine, Anyang
- Department of Otorhinolaryngology-Head and Neck Surgery, National Medical Center, Seoul, Korea
| |
Collapse
|
240
|
Shtylla B, Gee M, Do A, Shabahang S, Eldevik L, de Pillis L. A Mathematical Model for DC Vaccine Treatment of Type I Diabetes. Front Physiol 2019; 10:1107. [PMID: 31555144 PMCID: PMC6742690 DOI: 10.3389/fphys.2019.01107] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 08/12/2019] [Indexed: 01/28/2023] Open
Abstract
Type I diabetes (T1D) is an autoimmune disease that can be managed, but for which there is currently no cure. Recent discoveries, particularly in mouse models, indicate that targeted modulation of the immune response has the potential to move an individual from a diabetic to a long-term, if not permanent, healthy state. In this paper we develop a single compartment mathematical model that captures the dynamics of dendritic cells (DC and tDC), T cells (effector and regulatory), and macrophages in the development of type I diabetes. The model supports the hypothesis that differences in macrophage clearance rates play a significant role in determining whether or not an individual is likely to become diabetic subsequent to a significant immune challenge. With this model we are able to explore the effects of strengthening the anti-inflammatory component of the immune system in a vulnerable individual. Simulations indicate that there are windows of opportunity in which treatment intervention is more likely to be beneficial in protecting an individual from entering a diabetic state. This model framework can be used as a foundation for modeling future T1D treatments as they are developed.
Collapse
Affiliation(s)
- Blerta Shtylla
- Mathematics Department, Pomona College, Claremont, CA, United States
| | - Marissa Gee
- Mathematics Department, Harvey Mudd College, Claremont, CA, United States
| | - An Do
- Institute of Mathematical Sciences, Claremont Graduate University, Claremont, CA, United States
| | | | - Leif Eldevik
- Aditx Therapeutics, Inc., Loma Linda, CA, United States
| | - Lisette de Pillis
- Mathematics Department, Harvey Mudd College, Claremont, CA, United States
| |
Collapse
|
241
|
Li H, Burgueño-Bucio E, Xu S, Das S, Olguin-Alor R, Elmets CA, Athar M, Raman C, Soldevila G, Xu H. CD5 on dendritic cells regulates CD4+ and CD8+ T cell activation and induction of immune responses. PLoS One 2019; 14:e0222301. [PMID: 31491023 PMCID: PMC6730919 DOI: 10.1371/journal.pone.0222301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 08/26/2019] [Indexed: 12/20/2022] Open
Abstract
The role of CD5 as a regulator of T cell signaling and tolerance is well recognized. Recent data show expression of CD5 on different subtypes of human dendritic cells, however its functional relevance in modulating DC mediated responses remains poorly understood. In this study, we show CD5 is expressed on CD11c+ DC from murine thymus, lymph node, spleen, skin and lung. Although the development of DC subpopulations in CD5-/- mice was normal, CD5-deficient DC produced significantly higher levels of IL-12 than wild type DC in response to LPS. CD5-/- DC, in comparison to CD5+/+ DC, enhanced the activation of CD4+ and CD8+ T cells in vitro and in vivo and induced significantly higher production of IL-2 and IFN-gamma by T cells. Consequently, CD5-/- DC were significantly more potent than wild type DC in the induction of anti-tumor immunity and contact hypersensitivity responses in mice. Restoration of CD5 expression in CD5-/- DC reduced IL-12 production and inhibited their capacity to stimulate T cells. Collectively, these data demonstrate that the specific expression of CD5 on DC inhibits the production of inflammatory cytokines and has a regulatory effect on their activity to stimulate T cells and induce immune responses. This study reveals a previously unrecognized regulatory role for CD5 on DC and provides novel insights into mechanisms for DC biology in immune responses.
Collapse
Affiliation(s)
- Hui Li
- Department of Dermatology, University of Alabama at Birmingham. Birmingham, Alabama, United States of America
| | - Erica Burgueño-Bucio
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Shin Xu
- Department of Dermatology, University of Alabama at Birmingham. Birmingham, Alabama, United States of America
| | - Shaonli Das
- Department of Dermatology, University of Alabama at Birmingham. Birmingham, Alabama, United States of America
| | - Roxana Olguin-Alor
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Craig A. Elmets
- Department of Dermatology, University of Alabama at Birmingham. Birmingham, Alabama, United States of America
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham. Birmingham, Alabama, United States of America
| | - Chander Raman
- Department of Medicine, University of Alabama at Birmingham. Birmingham, Alabama, United States of America
| | - Gloria Soldevila
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Hui Xu
- Department of Dermatology, University of Alabama at Birmingham. Birmingham, Alabama, United States of America
| |
Collapse
|
242
|
Xiong A, Wang J, Mao XL, Jiang Y, Fan Y. MiR‐199a‐3p modulates the function of dendritic cells involved in transplantation tolerance by targeting CD86. HLA 2019; 94:493-503. [PMID: 31448543 DOI: 10.1111/tan.13677] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 08/06/2019] [Accepted: 08/23/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Ali Xiong
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| | - Jing Wang
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| | - Xiao Li Mao
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| | - Yi Jiang
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| | - Yue Fan
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| |
Collapse
|
243
|
Intradermal vaccination prevents anti-MOG autoimmune encephalomyelitis in macaques. EBioMedicine 2019; 47:492-505. [PMID: 31492559 PMCID: PMC6796575 DOI: 10.1016/j.ebiom.2019.08.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/21/2019] [Accepted: 08/23/2019] [Indexed: 12/28/2022] Open
Abstract
Background Autoimmune demyelinating diseases (ADD) are a major cause of neurological disability due to autoreactive cellular and humoral immune responses against brain antigens. A cure for chronic ADD could be obtained by appropriate immunomodulation. Methods We implemented a preclinical scheme to foster immune tolerance to myelin oligodendrocyte glycoprotein (MOG), in a cynomolgus-macaque model of experimental autoimmune encephalomyelitis (EAE), in which administration of recombinant human MOG (rhMOG) elicits brain inflammation mediated by MOG-autoreactive CD4+ lymphocytes and anti-MOG IgG. For immunotherapy, we used a recombinant antibody (Ab) directed against the dendritic cell-asialoglycoprotein receptor (DC-ASGPR) fused either to MOG or a control antigen PSA (prostate-specific antigen). Findings rhMOG and the anti-DC-ASGPR-MOG were respectively detected in CD1a+ DCs or CD163+ cells in the skin of macaques. Intradermal administration of anti-DC-ASGPR-MOG, but not control anti-DC-ASGPR-PSA, was protective against EAE. The treatment prevented the CD4+ T cell activation and proinflammatory cytokine production observed in controls. Moreover, the administration of anti-DC-ASGPR-MOG induced MOG-specific CD4+CD25+FOXP3+CD39+ regulatory lymphocytes and favoured an upsurge in systemic TGFβ and IL-8 upon rhMOG re-administration in vivo. Interpretation We show that the delivery of an anti-DC-ASGPR-MOG allows antigen-specific adaptive immune modulation to prevent the breach of immune tolerance to MOG. Our findings pave the way for therapeutic vaccines for long-lasting remission to grave encephalomyelitis with identified autoantigens, such as ADD associated with anti-MOG autoantibodies. Fund Work supported by the French ANR (ANR-11-INBS-0008 and ANR-10-EQPX-02-01), NIH (NIH 1 R01 AI 105066), the Baylor Scott and White Healthcare System funding and Roche Research Collaborative grants.
Collapse
|
244
|
Cannarile L, Delfino DV, Adorisio S, Riccardi C, Ayroldi E. Implicating the Role of GILZ in Glucocorticoid Modulation of T-Cell Activation. Front Immunol 2019; 10:1823. [PMID: 31440237 PMCID: PMC6693389 DOI: 10.3389/fimmu.2019.01823] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/18/2019] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoid-induced leucine zipper (GILZ) is a protein with multiple biological roles that is upregulated by glucocorticoids (GCs) in both immune and non-immune cells. Importantly, GCs are immunosuppressive primarily due to their regulation of cell signaling pathways that are crucial for immune system activity. GILZ, which is transcriptionally induced by the glucocorticoid receptor (GR), mediates part of these immunosuppressive, and anti-inflammatory effects, thereby controlling immune cell proliferation, survival, and differentiation. The primary immune cells targeted by the immunosuppressive activity of GCs are T cells. Importantly, the effects of GCs on T cells are partially mediated by GILZ. In fact, GILZ regulates T-cell activation, and differentiation by binding and inhibiting factors essential for T-cell function. For example, GILZ associates with nuclear factor-κB (NF-κB), c-Fos, and c-Jun and inhibits NF-κB-, and AP-1-dependent transcription. GILZ also binds Raf and Ras, inhibits activation of Ras/Raf downstream targets, including mitogen-activated protein kinase 1 (MAPK1). In addition GILZ inhibits forkhead box O3 (FoxO3) without physical interaction. GILZ also promotes the activity of regulatory T cells (Tregs) by activating transforming growth factor-β (TGF-β) signaling. Ultimately, these actions inhibit T-cell activation and modulate the differentiation of T helper (Th)-1, Th-2, Th-17 cells, thereby mediating the immunosuppressive effects of GCs on T cells. In this mini-review, we discuss how GILZ mediates GC activity on T cells, focusing mainly on the therapeutic potential of this protein as a more targeted anti-inflammatory/immunosuppressive GC therapy.
Collapse
Affiliation(s)
- Lorenza Cannarile
- Section of Pharmacology, Department of Medicine, Medical School, University of Perugia, Perugia, Italy
| | - Domenico V Delfino
- Section of Pharmacology, Department of Medicine, Medical School, University of Perugia, Perugia, Italy
| | - Sabrina Adorisio
- Section of Pharmacology, Department of Medicine, Medical School, University of Perugia, Perugia, Italy
| | - Carlo Riccardi
- Section of Pharmacology, Department of Medicine, Medical School, University of Perugia, Perugia, Italy
| | - Emira Ayroldi
- Section of Pharmacology, Department of Medicine, Medical School, University of Perugia, Perugia, Italy
| |
Collapse
|
245
|
Das T, Bergen IM, Koudstaal T, van Hulst JA, van Loo G, Boonstra A, Vanwolleghem T, Leung PS, Gershwin ME, Hendriks RW, Kool M. DNGR1-mediated deletion of A20/Tnfaip3 in dendritic cells alters T and B-cell homeostasis and promotes autoimmune liver pathology. J Autoimmun 2019; 102:167-178. [DOI: 10.1016/j.jaut.2019.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/27/2019] [Accepted: 05/05/2019] [Indexed: 02/06/2023]
|
246
|
Hinshaw DC, Shevde LA. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res 2019; 79:4557-4566. [PMID: 31350295 DOI: 10.1158/0008-5472.can-18-3962] [Citation(s) in RCA: 1843] [Impact Index Per Article: 368.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/28/2019] [Accepted: 05/21/2019] [Indexed: 12/14/2022]
Abstract
Cancer development and progression occurs in concert with alterations in the surrounding stroma. Cancer cells can functionally sculpt their microenvironment through the secretion of various cytokines, chemokines, and other factors. This results in a reprogramming of the surrounding cells, enabling them to play a determinative role in tumor survival and progression. Immune cells are important constituents of the tumor stroma and critically take part in this process. Growing evidence suggests that the innate immune cells (macrophages, neutrophils, dendritic cells, innate lymphoid cells, myeloid-derived suppressor cells, and natural killer cells) as well as adaptive immune cells (T cells and B cells) contribute to tumor progression when present in the tumor microenvironment (TME). Cross-talk between cancer cells and the proximal immune cells ultimately results in an environment that fosters tumor growth and metastasis. Understanding the nature of this dialog will allow for improved therapeutics that simultaneously target multiple components of the TME, increasing the likelihood of favorable patient outcomes.
Collapse
Affiliation(s)
- Dominique C Hinshaw
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Lalita A Shevde
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama. .,O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
247
|
Liu Y, Zhao JJ, Zhou ZQ, Pan QZ, Zhu Q, Tang Y, Xia JC, Weng DS. IL-37 induces anti-tumor immunity by indirectly promoting dendritic cell recruitment and activation in hepatocellular carcinoma. Cancer Manag Res 2019; 11:6691-6702. [PMID: 31410060 PMCID: PMC6646800 DOI: 10.2147/cmar.s200627] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 06/11/2019] [Indexed: 12/15/2022] Open
Abstract
Introduction IL-37 is a cytokine of IL-1 family that plays an important role in innate immunity and inflammation, and has been studied as a tumor suppressor in many cancers. However, it remains unclear whether IL-37 plays a regulatory role in tumor-infiltrating dendritic cells (DCs) in hepatocellular carcinoma (HCC). Materials and methods We evaluated the relationship between IL-37 expression and tumor infiltration by DCs in 155 HCC samples through immunohistochemical analysis and Kaplan–Meier survival analysis. The effects of IL-37 on the anti-tumor activity of DCs were investigated by ELISA, flow cytometry, real-time quantitative PCR, cytotoxicity assays and tumorigenicity assays. Results The expression level of IL-37 in HCC samples was positively correlated with the degree of CD1a+ DCs infiltration. The survival rates of patients with both a high expression of IL-37 and a high infiltration by CD1a+ DCs were significantly higher than those of patients with a low expression of IL-37 and a low infiltration by CD1a+ DCs. In vitro chemotaxis analysis indicated that HCC cells overexpressing IL-37 recruited more DCs by secreting higher levels of specific chemokines (eg, CCL3 and CCL20). In addition, IL-37 indirectly up-regulated the expression of major histocompatibility class II molecules, CD86 and CD40 on DCs by acting on tumor cells; IL-37 also indirectly enhanced the anti-tumor effect of T lymphocytes by stimulating DCs to secrete cytokines such as IL-2, IL-12, IL-12p70, interferon-α (IFN-α) and IFN-γ. Finally, overexpression IL-37 in HCC cells significantly delayed tumor growth and increased recruitment of CD11c+ DCs to tumor tissues was also revealed in vivo mouse model. Conclusion DCs play an important role in IL-37 mediated anti-tumor immune responses in HCC, which may contribute to the development of novel cancer immunotherapeutic strategies.
Collapse
Affiliation(s)
- Yuan Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jing-Jing Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Zi-Qi Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Qiu-Zhong Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Qian Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yan Tang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jian-Chuan Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - De-Sheng Weng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| |
Collapse
|
248
|
Sprooten J, Ceusters J, Coosemans A, Agostinis P, De Vleeschouwer S, Zitvogel L, Kroemer G, Galluzzi L, Garg AD. Trial watch: dendritic cell vaccination for cancer immunotherapy. Oncoimmunology 2019; 8:e1638212. [PMID: 31646087 PMCID: PMC6791419 DOI: 10.1080/2162402x.2019.1638212] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
Dendritic- cells (DCs) have received considerable attention as potential targets for the development of anticancer vaccines. DC-based anticancer vaccination relies on patient-derived DCs pulsed with a source of tumor-associated antigens (TAAs) in the context of standardized maturation-cocktails, followed by their reinfusion. Extensive evidence has confirmed that DC-based vaccines can generate TAA-specific, cytotoxic T cells. Nonetheless, clinical efficacy of DC-based vaccines remains suboptimal, reflecting the widespread immunosuppression within tumors. Thus, clinical interest is being refocused on DC-based vaccines as combinatorial partners for T cell-targeting immunotherapies. Here, we summarize the most recent preclinical/clinical development of anticancer DC vaccination and discuss future perspectives for DC-based vaccines in immuno-oncology.
Collapse
Affiliation(s)
- Jenny Sprooten
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jolien Ceusters
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
- Department of Gynecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
- Center for Cancer Biology (CCB), VIB, Leuven, Belgium
| | - Steven De Vleeschouwer
- Research Group Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China
- Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
- Université de Paris Descartes, Paris, France
| | - Abhishek D. Garg
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
249
|
Significant body mass increase by oral administration of a cascade of shIL21-MSTN yeast-based DNA vaccine in mice. Biomed Pharmacother 2019; 118:109147. [PMID: 31302418 DOI: 10.1016/j.biopha.2019.109147] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/31/2022] Open
Abstract
Base on the practical of MSTN-specific yeast-based protein vaccine in mice as described previously, this research was designed for developing a better DNA vaccine (a cascade of shIL21-MSTN yeast-based DNA vaccine) than solely MSTN yeast-based DNA vaccine to block the endogenous MSTN in the murine model. We first constructed the target vectors, including CMV-driven MSTN expression vector and a combined shIL21-MSTN vector which containing MSTN expression cassette and shIL21 (short hairpin RNA-IL21) expression cassette. After necessary validation, recombinant yeast vaccines harboring different vectors were well prepared. Subsequently, after 2-month administration, the MSTN-specific immune response was detected with western blots. The commercial ELISA assays indicated that the production of IL21 and IL6 were decreased compared with control groups. More importantly, the MSTN-specific antibody titer was much higher in the shIL21-MSTN group than MSTN group, which was consistent with the western blots result. The most important finding was significant body mass increased after oral administration of these yeast-based DNA vaccines, in which the shIL21-MSTN vaccine is slightly higher than the sole MSTN vaccine in mice. In this study, we confirmed the role of different MSTN-specific yeast-based DNA vaccines on increasing body mass in mice, to provide a good inspiration for livestock breeding through the new type of immunoregulatory method. On the other hand, we also detected the possible modulating role of shIL21 on the dendritic cell-mediated immune function which needs more practical application and deeper exploration.
Collapse
|
250
|
STAT3 Activation in Combination with NF-KappaB Inhibition Induces Tolerogenic Dendritic Cells with High Therapeutic Potential to Attenuate Collagen-Induced Arthritis. J Immunol Res 2019; 2019:1982570. [PMID: 31355296 PMCID: PMC6636450 DOI: 10.1155/2019/1982570] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/02/2019] [Accepted: 06/10/2019] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DCs) have the ability to induce tolerance or inflammation in response to self-antigens, which makes them fundamental players in autoimmunity. In this regard, immunogenic DCs produce IL-12 and IL-23 favouring the acquisition of Th1 and Th17 inflammatory phenotypes, respectively, by autoreactive CD4+ T-cells, thus promoting autoimmunity. Conversely, tolerogenic DCs produce IL-10 and TGF-β, inducing the generation of CD4+ T-cells with suppressive activity (Treg), which promote tolerance to self-constituents. Previous studies have shown that STAT3 signalling in DCs attenuates the production of proinflammatory cytokines, whilst NF-κB activation promotes it. In this study, we aimed to generate DCs displaying strong and constitutive tolerogenic profile to be used as immunotherapy in autoimmunity. To this end, we transduced bone marrow-derived DCs with lentiviral particles codifying for a constitutively active version of STAT3 (constitutively active STAT3 (STAT3ca)) or with a constitutive repressor of NF-κB (IκBα superrepressor (IκBαSR)), and their therapeutic potential was evaluated in a mouse model of arthritis induced by collagen (CIA). Our results show that STAT3ca transduction favoured the production of the anti-inflammatory mediator IL-10, whereas IκBαSR transduction attenuated the expression of the proinflammatory cytokine IL-23 in DCs. Moreover, both STAT3ca-transduced and IκBαSR-transduced DCs separately exerted a mild but significant therapeutic effect reducing the severity of CIA development. Furthermore, when DCs were transduced with both STAT3ca and IκBαSR together, they reduced CIA manifestation significantly stronger than when transduced with only STAT3ca or IκBαSR separately. These results show STAT3 and NF-κB as two important and complementary regulators of the tolerogenic behaviour of DCs, which should be considered as molecular targets in the design of DC-based suppressive immunotherapies for the treatment of autoimmune disorders.
Collapse
|