1
|
Chen B, Mu C, Zhang Z, He X, Liu X. The Love-Hate Relationship Between TGF-β Signaling and the Immune System During Development and Tumorigenesis. Front Immunol 2022; 13:891268. [PMID: 35720407 PMCID: PMC9204485 DOI: 10.3389/fimmu.2022.891268] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Since TGF-β was recognized as an essential secreted cytokine in embryogenesis and adult tissue homeostasis a decade ago, our knowledge of the role of TGF-β in mammalian development and disease, particularly cancer, has constantly been updated. Mounting evidence has confirmed that TGF-β is the principal regulator of the immune system, as deprivation of TGF-β signaling completely abrogates adaptive immunity. However, enhancing TGF-β signaling constrains the immune response through multiple mechanisms, including boosting Treg cell differentiation and inducing CD8+ T-cell apoptosis in the disease context. The love-hate relationship between TGF-β signaling and the immune system makes it challenging to develop effective monotherapies targeting TGF-β, especially for cancer treatment. Nonetheless, recent work on combination therapies of TGF-β inhibition and immunotherapy have provide insights into the development of TGF-β-targeted therapies, with favorable outcomes in patients with advanced cancer. Hence, we summarize the entanglement between TGF-β and the immune system in the developmental and tumor contexts and recent progress on hijacking crucial TGF-β signaling pathways as an emerging area of cancer therapy.
Collapse
Affiliation(s)
- Baode Chen
- Department of Laboratory Medicine, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chenglin Mu
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Zhiwei Zhang
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Xuelin He
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xia Liu
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| |
Collapse
|
2
|
Lim KPH, Milne P, Poidinger M, Duan K, Lin H, McGovern N, Abhyankar H, Zinn D, Burke TM, Eckstein OS, Chakraborty R, Sengal A, Scull B, Newell E, Merad M, McClain KL, Man TK, Ginhoux F, Collin M, Allen CE. Circulating CD1c+ myeloid dendritic cells are potential precursors to LCH lesion CD1a+CD207+ cells. Blood Adv 2020; 4:87-99. [PMID: 31899802 PMCID: PMC6960472 DOI: 10.1182/bloodadvances.2019000488] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 10/24/2019] [Indexed: 12/17/2022] Open
Abstract
Langerhans cell histiocytosis (LCH) is a myeloproliferative disorder that is characterized by the inflammatory lesions with pathogenic CD1a+CD207+ dendritic cells (DCs). BRAFV600E and other somatic activating MAPK gene mutations have been identified in differentiating bone marrow and blood myeloid cells, but the origin of the LCH lesion CD1a+CD207+ DCs and mechanisms of lesion formation remain incompletely defined. To identify candidate LCH CD1a+CD207+ DC precursor populations, gene-expression profiles of LCH lesion CD1a+CD207+ DCs were first compared with established gene signatures from human myeloid cell subpopulations. Interestingly, the CD1c+ myeloid DC (mDC) gene signature was most enriched in the LCH CD1a+CD207+ DC transcriptome. Additionally, the BRAFV600E allele was not only localized to CD1a+CD207- DCs and CD1a+CD207+ DCs, but it was also identified in CD1c+ mDCs in LCH lesions. Transcriptomes of CD1a+CD207- DCs were nearly indistinguishable from CD1a+CD207+ DCs (both CD1a+CD207low and CD1a+CD207high subpopulations). Transcription profiles of LCH lesion CD1a+CD207+ DCs and peripheral blood CD1c+ mDCs from healthy donors were compared to identify potential LCH DC-specific biomarkers: HLA-DQB2 expression was significantly increased in LCH lesion CD1a+CD207+ DCs compared with circulating CD1c+ mDCs from healthy donors. HLA-DQB2 antigen was identified on LCH lesion CD1a+CD207- DCs and CD1a+CD207+ DCs as well as on CD1c+(CD1a+CD207-) mDCs, but it was not identified in any other lesion myeloid subpopulations. HLA-DQB2 expression was specific to peripheral blood of patients with BRAFV600E+ peripheral blood mononuclear cells, and HLA-DQB2+CD1c+ blood cells were highly enriched for the BRAFV600E in these patients. These data support a model in which blood CD1c+HLA-DQB2+ mDCs with activated ERK migrate to lesion sites where they differentiate into pathogenic CD1a+CD207+ DCs.
Collapse
Affiliation(s)
- Karen Phaik Har Lim
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
- Graduate Program in Translational Biology and Molecular Medicine, College of Medicine, Baylor University, Houston, TX
| | - Paul Milne
- Human Dendritic Cell Laboratory, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kaibo Duan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Howard Lin
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Naomi McGovern
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Harshal Abhyankar
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Daniel Zinn
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Thomas M Burke
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
- Graduate Program in Translational Biology and Molecular Medicine, College of Medicine, Baylor University, Houston, TX
- Medical Scientist Training Program, College of Medicine, Baylor University, Houston, TX; and
| | - Olive S Eckstein
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Rikhia Chakraborty
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Amel Sengal
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Brooks Scull
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Evan Newell
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Miriam Merad
- Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kenneth L McClain
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Tsz-Kwong Man
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Matthew Collin
- Human Dendritic Cell Laboratory, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Carl E Allen
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
- Graduate Program in Translational Biology and Molecular Medicine, College of Medicine, Baylor University, Houston, TX
| |
Collapse
|
3
|
Chrisikos TT, Zhou Y, Slone N, Babcock R, Watowich SS, Li HS. Molecular regulation of dendritic cell development and function in homeostasis, inflammation, and cancer. Mol Immunol 2019; 110:24-39. [PMID: 29549977 PMCID: PMC6139080 DOI: 10.1016/j.molimm.2018.01.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 01/04/2018] [Accepted: 01/25/2018] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are the principal antigen-presenting cells of the immune system and play key roles in controlling immune tolerance and activation. As such, DCs are chief mediators of tumor immunity. DCs can regulate tolerogenic immune responses that facilitate unchecked tumor growth. Importantly, however, DCs also mediate immune-stimulatory activity that restrains tumor progression. For instance, emerging evidence indicates the cDC1 subset has important functions in delivering tumor antigens to lymph nodes and inducing antigen-specific lymphocyte responses to tumors. Moreover, DCs control specific therapeutic responses in cancer including those resulting from immune checkpoint blockade. DC generation and function is influenced profoundly by cytokines, as well as their intracellular signaling proteins including STAT transcription factors. Regardless, our understanding of DC regulation in the cytokine-rich tumor microenvironment is still developing and must be better defined to advance cancer treatment. Here, we review literature focused on the molecular control of DCs, with a particular emphasis on cytokine- and STAT-mediated DC regulation. In addition, we highlight recent studies that delineate the importance of DCs in anti-tumor immunity and immune therapy, with the overall goal of improving knowledge of tumor-associated factors and intrinsic DC signaling cascades that influence DC function in cancer.
Collapse
Affiliation(s)
- Taylor T Chrisikos
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Yifan Zhou
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Natalie Slone
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rachel Babcock
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Stephanie S Watowich
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| | - Haiyan S Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
4
|
Strobl H, Krump C, Borek I. Micro-environmental signals directing human epidermal Langerhans cell differentiation. Semin Cell Dev Biol 2018; 86:36-43. [PMID: 29448069 DOI: 10.1016/j.semcdb.2018.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/12/2017] [Accepted: 02/10/2018] [Indexed: 01/11/2023]
Abstract
Human Langerhans cells (LC) can be generated ex vivo from hematopoietic precursor cells in response to cytokines and cell-membrane associated ligands. These in vitro differentiation models provided mechanistic insights into the molecular and cellular pathways underlying the development of this unique, epithelia-associated dendritic cell subset. Notably, the human epidermal microenvironment is fully sufficient to induce LC differentiation from hematopoietic progenitors. Hence, dissecting the molecular characteristics of the human epithelial/epidermal LC niche, and testing defined ligands for their capacity to induce LC differentiation, led to a refined molecular model of LC lineage commitment. During epidermal ontogeny, spatially and temporally regulated availability of TGF-β family members cooperate with other keratinocyte-derived signals, such as E-cadherin and Notch ligands, for instructing LC differentiation. In this review, we discuss the signals known to instruct human hematopoietic progenitor cells and myelomonocytic cells to undergo LC lineage commitment. Additionally, the current methods for generation of large numbers of human LC-like cells ex vivo in defined serum-free media are discussed.
Collapse
Affiliation(s)
- Herbert Strobl
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria.
| | - Corinna Krump
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Izabela Borek
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| |
Collapse
|
5
|
Kurotaki D, Sasaki H, Tamura T. Transcriptional control of monocyte and macrophage development. Int Immunol 2018; 29:97-107. [PMID: 28379391 DOI: 10.1093/intimm/dxx016] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 03/19/2017] [Indexed: 12/12/2022] Open
Abstract
Monocytes and macrophages play critical roles in immune responses, tissue homeostasis and disease progression. There are a number of functionally and phenotypically distinct subpopulations throughout the body. However, the mechanisms by which macrophage and monocyte heterogeneity is established remain unclear. Recent studies have suggested that most tissue-resident macrophages originate from fetal progenitors but not from hematopoietic stem cells, whereas some subpopulations are derived from adult monocytes. In addition, transcription factors specifically required for the development of each subpopulation have been identified. Interestingly, local environmental factors such as heme, retinoic acid and RANKL induce the expression and/or activation of tissue-specific transcription factors, thereby controlling transcriptional programs specific for the subpopulations. Thus, distinct differentiation pathways and local microenvironments appear to contribute to the determination of macrophage transcriptional identities. In this review, we highlight recent advances in our knowledge of the transcriptional control of macrophage and monocyte development.
Collapse
Affiliation(s)
- Daisuke Kurotaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Haruka Sasaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| |
Collapse
|
6
|
Distinct Murine Mucosal Langerhans Cell Subsets Develop from Pre-dendritic Cells and Monocytes. Immunity 2015; 43:369-81. [DOI: 10.1016/j.immuni.2015.06.017] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 04/16/2015] [Accepted: 06/23/2015] [Indexed: 11/21/2022]
|
7
|
The TGF-β superfamily in dendritic cell biology. Cytokine Growth Factor Rev 2015; 26:647-57. [PMID: 26115564 DOI: 10.1016/j.cytogfr.2015.06.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 06/15/2015] [Indexed: 12/18/2022]
Abstract
The TGF-β superfamily consists of a large group of pleiotropic cytokines that are involved in the regulation of many developmental, physiological and pathological processes. Dendritic cells are antigen-presenting cells that play a key role in innate and adaptive immune responses. Dendritic cells have a complex relationship with the TGF-β cytokine superfamily being both source and targets for many of these cytokines. Some TGF-β family members are expressed by dendritic cells and modulate immune responses, for instance through the induction of T cell polarization. Others play a crucial role in the development and function of the different dendritic cell subsets. This review summarizes the current knowledge on the role of TGF-β family cytokines in dendritic cell biology, focusing on TGF-β as well as on other, less characterized, members of these important immune mediators.
Collapse
|
8
|
Bloy N, Pol J, Aranda F, Eggermont A, Cremer I, Fridman WH, Fučíková J, Galon J, Tartour E, Spisek R, Dhodapkar MV, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Dendritic cell-based anticancer therapy. Oncoimmunology 2014; 3:e963424. [PMID: 25941593 DOI: 10.4161/21624011.2014.963424] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 09/05/2014] [Indexed: 02/06/2023] Open
Abstract
The use of patient-derived dendritic cells (DCs) as a means to elicit therapeutically relevant immune responses in cancer patients has been extensively investigated throughout the past decade. In this context, DCs are generally expanded, exposed to autologous tumor cell lysates or loaded with specific tumor-associated antigens (TAAs), and then reintroduced into patients, often in combination with one or more immunostimulatory agents. As an alternative, TAAs are targeted to DCs in vivo by means of monoclonal antibodies, carbohydrate moieties or viral vectors specific for DC receptors. All these approaches have been shown to (re)activate tumor-specific immune responses in mice, often mediating robust therapeutic effects. In 2010, the first DC-based preparation (sipuleucel-T, also known as Provenge®) has been approved by the US Food and Drug Administration (FDA) for use in humans. Reflecting the central position occupied by DCs in the regulation of immunological tolerance and adaptive immunity, the interest in harnessing them for the development of novel immunotherapeutic anticancer regimens remains high. Here, we summarize recent advances in the preclinical and clinical development of DC-based anticancer therapeutics.
Collapse
Key Words
- DC, dendritic cell
- DC-based vaccination
- FDA, Food and Drug Administration
- IFN, interferon
- MRC1, mannose receptor, C type 1
- MUC1, mucin 1
- TAA, tumor-associated antigen
- TLR, Toll-like receptor
- Toll-like receptor agonists
- Treg, regulatory T cell
- WT1, Wilms tumor 1
- antigen cross-presentation
- autophagy
- iDC, immature DC
- immunogenic cell death
- mDC, mature DC
- pDC, plasmacytoid DC
- regulatory T cells
Collapse
Affiliation(s)
- Norma Bloy
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France ; Université Paris-Sud/Paris XI ; Orsay, France
| | - Jonathan Pol
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France
| | - Fernando Aranda
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France
| | | | - Isabelle Cremer
- INSERM , U1138; Paris France ; Equipe 13; Centre de Recherche des Cordeliers ; Paris France ; Université Pierre et Marie Curie/Paris VI ; Paris France
| | - Wolf Hervé Fridman
- INSERM , U1138; Paris France ; Equipe 13; Centre de Recherche des Cordeliers ; Paris France ; Université Pierre et Marie Curie/Paris VI ; Paris France
| | - Jitka Fučíková
- Department of Immunology; 2nd Medical School Charles University and University Hospital Motol ; Prague, Czech Republic ; Sotio a.s. ; Prague, Czech Republic
| | - Jérôme Galon
- INSERM , U1138; Paris France ; Université Pierre et Marie Curie/Paris VI ; Paris France ; Laboratory of Integrative Cancer Immunology; Centre de Recherche des Cordeliers ; Paris France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris France
| | - Eric Tartour
- Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris France ; INSERM , U970; Paris France ; Pôle de Biologie; Hôpital Européen Georges Pompidou, AP-HP ; Paris France
| | - Radek Spisek
- Department of Immunology; 2nd Medical School Charles University and University Hospital Motol ; Prague, Czech Republic ; Sotio a.s. ; Prague, Czech Republic
| | - Madhav V Dhodapkar
- Department of Medicine; Immunobiology and Yale Cancer Center; Yale University ; New Haven, CT USA
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1015, CICBT507 ; Villejuif, France
| | - Guido Kroemer
- INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris France ; Pôle de Biologie; Hôpital Européen Georges Pompidou, AP-HP ; Paris France ; Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus ; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris France
| |
Collapse
|
9
|
Wu L, Yun Z, Tagawa T, De la Maza L, Wu MO, Yu J, Zhao Y, de Perrot M. Activation of CD1d-restricted natural killer T cells can inhibit cancer cell proliferation during chemotherapy by promoting the immune responses in murine mesothelioma. Cancer Immunol Immunother 2014; 63:1285-96. [PMID: 25183171 PMCID: PMC11029433 DOI: 10.1007/s00262-014-1597-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 08/07/2014] [Indexed: 12/31/2022]
Abstract
We studied the impact of natural killer T (NKT) cell activation by alpha-galactocysylceramide (α-GalCer, α-GC) on cancer cell repopulation during chemotherapy in murine mesothelioma. The number of NKT cells was found to be increased during the development of murine mesothelioma. NKT cells specifically recognize α-GC through CD1d resulting in their activation and expansion. Tumor-bearing mice were treated with chemotherapy once weekly, and α-GC was followed after each cycle of chemotherapy. Anti-tumor effect was evaluated on wild-type (WT) and CD1d knockout (CD1dKO) mice. Cancer cell proliferation and apoptosis were evaluated by Ki67 and TUNEL immunohistochemistry. CD4(+) and CD8(+) T cell proportion and activation in tumor, spleen, draining lymph node and peripheral blood were determined by flow cytometry, and gene expression of activated T cell-related cytokines was quantified by reverse transcription PCR. NKT cells were identified by CD1d-α-GC-tetramer staining. In WT mice, tumor growth delay was achieved by cisplatin (Cis), and this effect was improved in combination with α-GC, but α-GC alone had little effect. Cancer cell proliferation during chemotherapy was significantly inhibited by α-GC, while cancer cell death was significantly upregulated. α-GC following chemotherapy resulted in NKT cell expansion and an increase of interferon-γ production in the draining lymph node, blood and spleen. Gene expression of immune-associated cytokines was upregulated. Strikingly, the percentage of inducible T cell co-stimulator(+)CD4 T cells, Th17/Tc17 cells increased in splenocytes. In CD1d KO mice, however, Cis alone was less effective and Cis + α-GC provided no additional benefit over Cis alone. α-GC alone had minimal effect in both mice. NKT activation between cycles of chemotherapy could improve the outcome of mesothelioma treatment.
Collapse
Affiliation(s)
- Licun Wu
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Zhihong Yun
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Tetsuzo Tagawa
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Luis De la Maza
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Matthew Onn Wu
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Julie Yu
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Yidan Zhao
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Marc de Perrot
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
- Toronto Mesothelioma Research Program, Division of Thoracic Surgery, Toronto General Hospital, 9N-961, 200 Elizabeth St, Toronto, ON M5G 2C4 Canada
| |
Collapse
|
10
|
Vacchelli E, Vitale I, Eggermont A, Fridman WH, Fučíková J, Cremer I, Galon J, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2013; 2:e25771. [PMID: 24286020 PMCID: PMC3841205 DOI: 10.4161/onci.25771] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 07/16/2013] [Indexed: 12/26/2022] Open
Abstract
Dendritic cells (DCs) occupy a privileged position at the interface between innate and adaptive immunity, orchestrating a large panel of responses to both physiological and pathological cues. In particular, whereas the presentation of antigens by immature DCs generally results in the development of immunological tolerance, mature DCs are capable of priming robust, and hence therapeutically relevant, adaptive immune responses. In line with this notion, functional defects in the DC compartment have been shown to etiologically contribute to pathological conditions including (but perhaps not limited to) infectious diseases, allergic and autoimmune disorders, graft rejection and cancer. Thus, the possibility of harnessing the elevated immunological potential of DCs for anticancer therapy has attracted considerable interest from both researchers and clinicians over the last decade. Alongside, several methods have been developed not only to isolate DCs from cancer patients, expand them, load them with tumor-associated antigens and hence generate highly immunogenic clinical grade infusion products, but also to directly target DCs in vivo. This intense experimental effort has culminated in 2010 with the approval by the US FDA of a DC-based preparation (sipuleucel-T, Provenge®) for the treatment of asymptomatic or minimally symptomatic metastatic castration-refractory prostate cancer. As an update to the latest Trial Watch dealing with this exciting field of research (October 2012), here we summarize recent advances in DC-based anticancer regimens, covering both high-impact studies that have been published during the last 13 mo and clinical trials that have been launched in the same period to assess the antineoplastic potential of this variant of cellular immunotherapy.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM, U848; Villejuif, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|