1
|
Tai Y, Chen M, Wang F, Fan Y, Zhang J, Cai B, Yan L, Luo Y, Li Y. The role of dendritic cells in cancer immunity and therapeutic strategies. Int Immunopharmacol 2024; 128:111548. [PMID: 38244518 DOI: 10.1016/j.intimp.2024.111548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
Dendritic cells (DCs) are asserted as the most potent antigen-presenting cells (APCs) that orchestrate both innate and adaptive immunity, being extremely effective in the induction of robust anti-cancer T cell responses. Hence, the modulation of DCs function represents an attractive target for improving cancer immunotherapy efficacy. A better understanding of the immunobiology of DCs, the interaction among DCs, immune effector cells and tumor cells in tumor microenvironment (TME) and the latest advances in biomedical engineering technology would be required for the design of optimal DC-based immunotherapy. In this review, we focus on elaborating the immunobiology of DCs in healthy and cancer environments, the recent advances in the development of enhancing endogenous DCs immunocompetence via immunomodulators as well as DC-based vaccines. The rapidly developing field of applying nanotechnology to improve DC-based immunotherapy is also highlighted.
Collapse
Affiliation(s)
- Yunze Tai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Man Chen
- Hebei Yanda Lu Daopei Hospital, Langfang 065201, China
| | - Fang Wang
- Department of Medical Laboratory, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou 556000, China
| | - Yu Fan
- Department of Urology, National Clinical Research Center for Geriatrics and Organ Transplantation Center, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu 610041, China
| | - Junlong Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bei Cai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Yan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yao Luo
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Wang Y, Song Y, He Y, Wang Y, Maurer J, Kiessling F, Lammers T, Wang F, Shi Y. Direct immunoactivation by chemotherapeutic drugs in cancer treatment. ADVANCED THERAPEUTICS 2023; 6:2300209. [PMID: 38249990 PMCID: PMC7615547 DOI: 10.1002/adtp.202300209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Indexed: 01/23/2024]
Abstract
The immune system plays a crucial role in recognizing and eliminating pathogenic substances and malignant cells in the body. For cancer treatment, immunotherapy is becoming the standard treatment for many types of cancer and is often combined with chemotherapy. Although chemotherapeutic agents are often reported to have adverse effects, including immunosuppression, they can also play a positive role in immunotherapy by directly stimulating the immune system. This has been demonstrated in preclinical and clinical studies in the past decades. Chemotherapeutics can activate immune cells through different immune receptors and signaling pathways depending on their chemical structure and formulation. In this review, we summarize and discuss the direct immunoactivation effects of chemotherapeutics and possible mechanisms behind these effects. Finally, we prospect chemo-immunotherapeutic combinations for the more effective and safer treatment of cancer.
Collapse
Affiliation(s)
- Yurui Wang
- Department of Polymer Therapeutics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Yiran Song
- Department of Gastroenterology, Shanghai 10th People's Hospital, School of Medicine, Tongji University, Shanghai 200040, PR China
| | - Yazhi He
- Department of Gastroenterology, Shanghai 10th People's Hospital, School of Medicine, Tongji University, Shanghai 200040, PR China
| | - Yang Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, School of Medicine, Tongji University, Shanghai 200040, PR China
| | - Jochen Maurer
- Department of Gynecology and Obstetrics, Uniklinik RWTH Aachen, Aachen 52074, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Feng Wang
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, PR China
| | - Yang Shi
- Department of Polymer Therapeutics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| |
Collapse
|
3
|
Dendritic Cells: The Long and Evolving Road towards Successful Targetability in Cancer. Cells 2022; 11:cells11193028. [PMID: 36230990 PMCID: PMC9563837 DOI: 10.3390/cells11193028] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/19/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Dendritic cells (DCs) are a unique myeloid cell lineage that play a central role in the priming of the adaptive immune response. As such, they are an attractive target for immune oncology based therapeutic approaches. However, targeting these cells has proven challenging with many studies proving inconclusive or of no benefit in a clinical trial setting. In this review, we highlight the known and unknown about this rare but powerful immune cell. As technologies have expanded our understanding of the complexity of DC development, subsets and response features, we are now left to apply this knowledge to the design of new therapeutic strategies in cancer. We propose that utilization of these technologies through a multiomics approach will allow for an improved directed targeting of DCs in a clinical trial setting. In addition, the DC research community should consider a consensus on subset nomenclature to distinguish new subsets from functional or phenotypic changes in response to their environment.
Collapse
|
4
|
Challagundla N, Shah D, Yadav S, Agrawal-Rajput R. Saga of monokines in shaping tumour-immune microenvironment: Origin to execution. Cytokine 2022; 157:155948. [PMID: 35764025 DOI: 10.1016/j.cyto.2022.155948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 11/19/2022]
Abstract
Cellular communication mediated by cytokines is an important mechanism dictating immune responses, their cross talk and final immune output. Cytokines play a major role in dictating the immune outcome to cancer by regulating the events of development, differentiation and activation of innate immune cells. Cytokines are pleiotropic in nature, hence understanding their role individually or as member of network cytokines is critical to delineate their role in tumour immunity. Tumour systemically manipulates the immune system to evade and escape immune recognition for their uncontrollable growth and metastasis. The developing tumour comprise a large and diverse set of myeloid cells which are vulnerable to manipulation by the tumour-microenvironment. The innate immune cells of the monocytic lineage skew the fate of the adaptive immune cells and thus dictating cancer elimination or progression. Targeting cells at tumour cite is preposterous owing to their tight network, poor reach and abundance of immunosuppressive mechanisms. Monocytic lineage-derived cytokines (monokines) play crucial role in tumour regression or progression by either directly killing the tumour cells with TNFα or promoting its growth by TGFβ. In addition, the monokines like IL-12, IL-1β, IL-6, IL-10 and TGFβ direct the adaptive immune cells to secrete anti-tumour cytokines, TNFα, IFNγ, perforin and granzyme or pro-tumour cytokines, IL-10 and TGFβ. In this review, we elucidate the roles of monokines in dictating the fate of tumour by regulating responses at various stages of generation, differentiation and activation of immune cells along with the extensive cross talk. We have attempted to delineate the synergy and antagonism of major monokines among themselves or with tumour-derived or adaptive immune cytokines. The review provides an update on the possibilities of placing monokines to potential practical use as cytokine therapy against cancer.
Collapse
Affiliation(s)
- Naveen Challagundla
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat 382426, India
| | - Dhruvi Shah
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat 382426, India
| | - Shivani Yadav
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat 382426, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat 382426, India.
| |
Collapse
|
5
|
Dendritic cell-based cancer immunotherapy in the era of immune checkpoint inhibitors: From bench to bedside. Life Sci 2022; 297:120466. [PMID: 35271882 DOI: 10.1016/j.lfs.2022.120466] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 12/18/2022]
Abstract
Dendritic cells (DCs) can present tumoral antigens to T-cells and stimulate T-cell-mediated anti-tumoral immune responses. In addition to uptaking, processing, and presenting tumoral antigens to T-cells, co-stimulatory signals have to be established between DCs with T-cells to develop anti-tumoral immune responses. However, most of the tumor-infiltrated immune cells are immunosuppressive in the tumor microenvironment (TME), paving the way for immune evasion of tumor cells. This immunosuppressive TME has also been implicated in suppressing the DC-mediated anti-tumoral immune responses, as well. Various factors, i.e., immunoregulatory cells, metabolic factors, tumor-derived immunosuppressive factors, and inhibitory immune checkpoint molecules, have been implicated in developing the immunosuppressive TME. Herein, we aimed to review the biology of DCs in developing T-cell-mediated anti-tumoral immune responses, the significance of immunoregulatory cells in the TME, metabolic barriers contributing to DCs dysfunction in the TME, tumor-derived immunosuppressive factors, and inhibitory immune checkpoint molecules in DC-based cell therapy outcomes. With reviewing the ongoing clinical trials, we also proposed a novel therapeutic strategy to increase the efficacy of DC-based cell therapy. Indeed, the combination of DC-based cell therapy with monoclonal antibodies against novel immune checkpoint molecules can be a promising strategy to increase the response rate of patients with cancers.
Collapse
|
6
|
El-Sayes N, Vito A, Salem O, Workenhe ST, Wan Y, Mossman K. A Combination of Chemotherapy and Oncolytic Virotherapy Sensitizes Colorectal Adenocarcinoma to Immune Checkpoint Inhibitors in a cDC1-Dependent Manner. Int J Mol Sci 2022; 23:1754. [PMID: 35163675 PMCID: PMC8915181 DOI: 10.3390/ijms23031754] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/13/2022] [Accepted: 01/18/2022] [Indexed: 02/04/2023] Open
Abstract
Immune checkpoint therapy has shown great promise in the treatment of cancers with a high mutational burden, such as mismatch repair-deficient colorectal carcinoma (dMMR CRC). However, many patients fail to respond to immune checkpoint therapy. Using a mouse model of dMMR CRC, we demonstrated that tumors can be further sensitized to immune checkpoint therapy by using a combination of low-dose chemotherapy and oncolytic HSV-1. This combination induced the infiltration of CD8+ and CD4+ T cells into the tumor and the upregulation of gene signatures associated with the chemoattraction of myeloid cell subsets. When combined with immune checkpoint therapy, the combination promoted the infiltration of activated type 1 conventional dendritic cells (cDC1s) into the tumor. Furthermore, we found this combination strategy to be dependent on cDC1s, and its therapeutic efficacy to be abrogated in cDC1-deficient Batf3-/- mice. Thus, we demonstrated that the adjuvanticity of dMMR CRCs can be improved by combining low-dose chemotherapy and oncolytic HSV-1 in a cDC1-dependent manner.
Collapse
Affiliation(s)
- Nader El-Sayes
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada; (N.E.-S.); (O.S.); (Y.W.)
- Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Alyssa Vito
- Department of Clinical Translation, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada;
| | - Omar Salem
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada; (N.E.-S.); (O.S.); (Y.W.)
| | - Samuel Tekeste Workenhe
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Yonghong Wan
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada; (N.E.-S.); (O.S.); (Y.W.)
| | - Karen Mossman
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada; (N.E.-S.); (O.S.); (Y.W.)
| |
Collapse
|
7
|
Subtil B, Cambi A, Tauriello DVF, de Vries IJM. The Therapeutic Potential of Tackling Tumor-Induced Dendritic Cell Dysfunction in Colorectal Cancer. Front Immunol 2021; 12:724883. [PMID: 34691029 PMCID: PMC8527179 DOI: 10.3389/fimmu.2021.724883] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is the third most diagnosed malignancy and the second leading cause of cancer-related deaths worldwide. Locally advanced and metastatic disease exhibit resistance to therapy and are prone to recurrence. Despite significant advances in standard of care and targeted (immuno)therapies, the treatment effects in metastatic CRC patients have been modest. Untreatable cancer metastasis accounts for poor prognosis and most CRC deaths. The generation of a strong immunosuppressive tumor microenvironment (TME) by CRC constitutes a major hurdle for tumor clearance by the immune system. Dendritic cells (DCs), often impaired in the TME, play a critical role in the initiation and amplification of anti-tumor immune responses. Evidence suggests that tumor-mediated DC dysfunction is decisive for tumor growth and metastasis initiation, as well as for the success of immunotherapies. Unravelling and understanding the complex crosstalk between CRC and DCs holds promise for identifying key mechanisms involved in tumor progression and spread that can be exploited for therapy. The main goal of this review is to provide an overview of the current knowledge on the impact of CRC-driven immunosuppression on DCs phenotype and functionality, and its significance for disease progression, patient prognosis, and treatment response. Moreover, present knowledge gaps will be highlighted as promising opportunities to further understand and therapeutically target DC dysfunction in CRC. Given the complexity and heterogeneity of CRC, future research will benefit from the use of patient-derived material and the development of in vitro organoid-based co-culture systems to model and study DCs within the CRC TME.
Collapse
Affiliation(s)
- Beatriz Subtil
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Alessandra Cambi
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Daniele V. F. Tauriello
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
8
|
Ohe G, Kudo Y, Kamada K, Mouri Y, Takamaru N, Kudoh K, Kurio N, Miyamoto Y. The Soluble Factor from Oral Cancer Cell Lines Inhibits Interferon-γ Production by OK-432 via the CD40/CD40 Ligand Pathway. Cancers (Basel) 2021; 13:cancers13133301. [PMID: 34209347 PMCID: PMC8269085 DOI: 10.3390/cancers13133301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/20/2021] [Accepted: 06/28/2021] [Indexed: 11/16/2022] Open
Abstract
(1) Background: OK-432 is a penicillin-killed, lyophilized formulation of a low-toxicity strain (Su) of Streptococcus pyogenes (Group A). It is a potent immunotherapy agent for several types of cancer, including oral cancer. We previously showed that (i) OK-432 treatment induces a high amount of IFN-? production from peripheral blood mononuclear cells (PBMCs), and (ii) conditioned medium (CM) from oral cancer cells suppresses both the IFN-? production and cytotoxic activity of PBMCs driven by OK-432. The aim of this study was to determine the inhibitory mechanism of OK-432-induced IFN-? production from PBMCs by CM. (2) Methods: We performed cDNA microarray analysis, quantitative RT-PCR, and ELISA to reveal the inhibitory mechanism of CM. (3) Results: We found that CD40 plays a key role in IFN-? production via IL-12 production. Although OK-432 treatment upregulated the expression levels of the IL-12p40, p35, and CD40 genes, CM from oral cancer cells downregulate these genes. The amount of IFN-? production by OK-432 treatment was decreased by an anti-CD40 neutralizing antibody. (4) Conclusions: Our study suggests that uncertain soluble factor(s) produced from oral cancer cells may inhibit IFN-? production from PBMCs via suppressing the CD40/CD40L-IL-12 axis.
Collapse
Affiliation(s)
- Go Ohe
- Department of Oral Surgery, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (K.K.); (N.T.); (K.K.); (N.K.); (Y.M.)
- Dentistry and Oral Surgery, Takamatsu Municipal Hospital, 847-1 Ko Busshozan-cho, Takamatsu 761-8538, Japan
- Correspondence:
| | - Yasusei Kudo
- Department of Oral Bioscience, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (Y.K.); (Y.M.)
| | - Kumiko Kamada
- Department of Oral Surgery, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (K.K.); (N.T.); (K.K.); (N.K.); (Y.M.)
| | - Yasuhiro Mouri
- Department of Oral Bioscience, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (Y.K.); (Y.M.)
| | - Natsumi Takamaru
- Department of Oral Surgery, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (K.K.); (N.T.); (K.K.); (N.K.); (Y.M.)
| | - Keiko Kudoh
- Department of Oral Surgery, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (K.K.); (N.T.); (K.K.); (N.K.); (Y.M.)
| | - Naito Kurio
- Department of Oral Surgery, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (K.K.); (N.T.); (K.K.); (N.K.); (Y.M.)
| | - Youji Miyamoto
- Department of Oral Surgery, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (K.K.); (N.T.); (K.K.); (N.K.); (Y.M.)
| |
Collapse
|
9
|
Zhu S, Yang N, Wu J, Wang X, Wang W, Liu YJ, Chen J. Tumor microenvironment-related dendritic cell deficiency: a target to enhance tumor immunotherapy. Pharmacol Res 2020; 159:104980. [PMID: 32504832 DOI: 10.1016/j.phrs.2020.104980] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/07/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs), as specialized antigen-presenting cells, are essential for the initiation of specific T cell responses in innate antitumor immunity and, in certain cases, support humoral responses to inhibit tumor development. Mounting evidence suggests that the DC system displays a broad spectrum of dysfunctional status in the tumor microenvironment (TME), which ultimately affects antitumor immune responses. DC-based therapy can restore the function of DCs in the TME, thus showing a promising potential in tumor therapy. In this review, we provide an overview of the DC deficiency caused by various factors in the TME and discuss proposed strategies to reverse DC deficiency and the applications of novel combinatorial DC-based therapy for immune normalization of the tumor.
Collapse
Affiliation(s)
- Shan Zhu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Ning Yang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jing Wu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xue Wang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Wan Wang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | | | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
10
|
Schlitzer A, Zhang W, Song M, Ma X. Recent advances in understanding dendritic cell development, classification, and phenotype. F1000Res 2018; 7. [PMID: 30345015 PMCID: PMC6173131 DOI: 10.12688/f1000research.14793.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/19/2018] [Indexed: 12/14/2022] Open
Abstract
Dendritic cells (DCs) play an essential role in the induction of adaptive immune responses against infectious agents and in the generation of tolerance to self-antigens. In this mini-review, we summarize new evidence suggesting that the tissue of residence significantly shapes the last developmental steps of DCs into locally adapted cellular entities, enabling them to perform tissue-specific tasks while maintaining the core DC properties. We also discuss recent advances that have highlighted DCs’ rather complex phenotypic and functional heterogeneity in the tumor microenvironment, based on their physical characteristics, such as activation status, maturity, and polarization, illustrating a key role for DCs in the induction of anti-tumor immunity.
Collapse
Affiliation(s)
| | - Wei Zhang
- Shanghai Institute of Cancer Research, Shanghai, China
| | - Mei Song
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Xiaojing Ma
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA.,Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
11
|
Zekri ARN, El Deeb S, Bahnassy AA, Badr AM, Abdellateif MS, Esmat G, Salama H, Mohanad M, El-dien AE, Rabah S, Abd Elkader A. Role of relevant immune-modulators and cytokines in hepatocellular carcinoma and premalignant hepatic lesions. World J Gastroenterol 2018; 24:1228-1238. [PMID: 29568203 PMCID: PMC5859225 DOI: 10.3748/wjg.v24.i11.1228] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 12/24/2017] [Accepted: 01/16/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To assess the levels of different immune modulators in patients with hepatocellular carcinoma (HCC), in relation to other hepatic diseases.
METHODS Eighty-eight patients were included in the current study and represented patients with HCC (20), liver cirrhosis (28) and chronic hepatitis (CH; 25), and normal controls (NC; 15). Peripheral blood was isolated for immunophenotyping of active myeloid dendritic cells (mDCs; CD1c and CD40), mature inactive myeloid cells (CD1c and HLA), active plasmacytoid cells (pDCs; CD303 and CD40), mature inactive pDCs (CD30 and HLA), active natural killer (NK) cells (CD56 and CD161), active NK cells (CD56 and CD314) and inactive NK cells (CD56 and CD158) was done by flow cytometry. Serum levels of interleukin (IL)-2, IL-10, IL-12, IL-1β, interferon (IFN)-α, IFN-γ and tumor necrosis factor (TNF)-αR2 were assessed by ELISA.
RESULTS Active mDCs (CD1C+/CD40+) and inactive mDCs (CD1c+/HLA+) were significantly decreased in HCC patients in relation to NC (P < 0.001). CD40+ expression on active pDCs was decreased in HCC patients (P < 0.001), and its level was not significantly changed among other groups. Inactive pDCs (CD303+/HLA+), inactive NKs (CD56+/CD158+) and active NKs (CD56+/CD161+) were not statistically changed among the four groups studied; however, the latter was increased in CH (P < 0.05). NKG2D was statistically decreased in HCC, CH and cirrhosis (P < 0.001), and it was not expressed in 63% (12/20) of HCC patients. There was significant decrease of IL-2, IFN-α and IFN-γ (P < 0.001), and a significant increase in IL-10, IL-1β, and TNF-αR2 (P <0.01, P < 0.001 and P < 0.001; respectively) in HCC patients. There was inverted correlation between IL-12 and IL-1β in HCC (r = -0.565, P < 0.01), with a strong correlation between pDCs (CD303+/CD40+) and NKs (CD56+/CD161+; r = 0.512, P < 0.05) as well as inactive mDCs (CD1c+/HLA+) and inactive NK cells (CD56+/CD158+; r = 0.945, P < 0.001).
CONCLUSION NKG2D, CD40, IL-2 and IL-10 are important modulators in the development and progression of HCC.
Collapse
Affiliation(s)
- Abdel-Rahman N Zekri
- Molecular Virology and Immunology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo 11976, Egypt
| | - Somaya El Deeb
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Abeer A Bahnassy
- Department of Pathology, National Cancer Institute, Cairo University, Cairo 11976, Egypt
| | - Abeer M Badr
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo 11976, Egypt
| | - Gamal Esmat
- Department of Hepatology and Tropical Medicine, Faculty of Medicine, Cairo University, Cairo 11441, Egypt
| | - Hosny Salama
- Department of Hepatology and Tropical Medicine, Faculty of Medicine, Cairo University, Cairo 11441, Egypt
| | - Marwa Mohanad
- Department of Biochemistry, Misr University for Science and Technology, 6th October 12945, Giza Governorate, Egypt
| | - Ahmed Esam El-dien
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Shimaa Rabah
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Assmaa Abd Elkader
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| |
Collapse
|
12
|
Sultan M, Coyle KM, Vidovic D, Thomas ML, Gujar S, Marcato P. Hide-and-seek: the interplay between cancer stem cells and the immune system. Carcinogenesis 2017; 38:107-118. [PMID: 27866156 DOI: 10.1093/carcin/bgw115] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 11/15/2016] [Indexed: 12/26/2022] Open
Abstract
The enhanced ability of cancer stem cells (CSCs) to give rise to new tumors suggests that these cells may also have an advantage in evading immune detection and elimination. This tumor-forming ability, combined with the known plasticity of the immune system, which can play both protumorigenic and antitumorigenic roles, has motivated investigations into the interaction between CSCs and the immune system. Herein, we review the interplay between host immunity and CSCs by examining the immune-related mechanisms that favor CSCs and the CSC-mediated expansion of protumorigenic immune cells. Furthermore, we discuss immune cells, such as natural killer cells, that preferentially target CSCs and the strategies used by CSCs to evade immune detection and destruction. An increased understanding of these interactions and the pathways that regulate them may allow us to harness immune system components to create new adjuvant therapies that eradicate CSCs and improve patient survival.
Collapse
Affiliation(s)
| | | | | | | | - Shashi Gujar
- Department of Pathology and.,Department of Microbiology and Immunology, Dalhousie University, 5850 College Street, Halifax, Nova Scotia B3H 4R2, Canada
| | - Paola Marcato
- Department of Pathology and.,Department of Microbiology and Immunology, Dalhousie University, 5850 College Street, Halifax, Nova Scotia B3H 4R2, Canada
| |
Collapse
|
13
|
Cancer acidity: An ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin Cancer Biol 2017; 43:74-89. [PMID: 28267587 DOI: 10.1016/j.semcancer.2017.03.001] [Citation(s) in RCA: 377] [Impact Index Per Article: 53.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/22/2017] [Accepted: 03/01/2017] [Indexed: 12/21/2022]
Abstract
The link between cancer metabolism and immunosuppression, inflammation and immune escape has generated major interest in investigating the effects of low pH on tumor immunity. Indeed, microenvironmental acidity may differentially impact on diverse components of tumor immune surveillance, eventually contributing to immune escape and cancer progression. Although the molecular pathways underlying acidity-related immune dysfunctions are just emerging, initial evidence indicates that antitumor effectors such as T and NK cells tend to lose their function and undergo a state of mostly reversible anergy followed by apoptosis, when exposed to low pH environment. At opposite, immunosuppressive components such as myeloid cells and regulatory T cells are engaged by tumor acidity to sustain tumor growth while blocking antitumor immune responses. Local acidity could also profoundly influence bioactivity and distribution of antibodies, thus potentially interfering with the clinical efficacy of therapeutic antibodies including immune checkpoint inhibitors. Hence tumor acidity is a central regulator of cancer immunity that orchestrates both local and systemic immunosuppression and that may offer a broad panel of therapeutic targets. This review outlines the fundamental pathways of acidity-driven immune dysfunctions and sheds light on the potential strategies that could be envisaged to potentiate immune-mediated tumor control in cancer patients.
Collapse
|
14
|
Gujar R, Maurya N, Yadav V, Gupta M, Arora S, Khatri N, Sen P. c-Src Suppresses Dendritic Cell Antitumor Activity via T Cell Ig and Mucin Protein-3 Receptor. THE JOURNAL OF IMMUNOLOGY 2016; 197:1650-62. [PMID: 27439518 DOI: 10.4049/jimmunol.1600104] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/17/2016] [Indexed: 11/19/2022]
Abstract
The enhanced expression of T cell Ig and mucin protein-3 (TIM-3) on tumor-associated dendritic cells (DCs) attenuates antitumor effects of DNA vaccines. To identify a potential target (or targets) for reducing TIM-3 expression on tumor-associated DCs, we explored the molecular mechanisms regulating TIM-3 expression. In this study, we have identified a novel signaling pathway (c-Src→Bruton's tyrosine kinase→transcription factors Ets1, Ets2, USF1, and USF2) necessary for TIM-3 upregulation on DCs. Both IL-10 and TGF-β, which are produced in the tumor microenvironment, upregulated TIM-3 expression on DCs via this pathway. Suppressed expression of c-Src or downstream Bruton's tyrosine kinase, Ets1, Ets2, USF1, or USF2 blocked IL-10- and TGF-β-induced TIM-3 upregulation on DCs. Notably, in vivo knockdown of c-Src in mice reduced TIM-3 expression on tumor-associated DCs. Furthermore, adoptive transfer of c-Src-silenced DCs in mouse tumors enhanced the in vivo antitumor effects of immunostimulatory CpG DNA; however, TIM-3 overexpression in c-Src-silenced DCs blocked this effect. Collectively, our data reveal the molecular mechanism regulating TIM-3 expression in DCs and identify c-Src as a target for improving the efficacy of nucleic acid-mediated anticancer therapy.
Collapse
Affiliation(s)
- Ravindra Gujar
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh 160036, India; and
| | - Neeraj Maurya
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh 160036, India; and
| | - Vinod Yadav
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh 160036, India; and
| | - Mamta Gupta
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh 160036, India; and
| | - Saurabh Arora
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh 160036, India; and
| | - Neeraj Khatri
- Division of Animal Facility, Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh 160036, India
| | - Pradip Sen
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh 160036, India; and
| |
Collapse
|
15
|
Zong J, Keskinov AA, Shurin GV, Shurin MR. Tumor-derived factors modulating dendritic cell function. Cancer Immunol Immunother 2016; 65:821-33. [PMID: 26984847 PMCID: PMC11028482 DOI: 10.1007/s00262-016-1820-y] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/26/2016] [Indexed: 12/22/2022]
Abstract
Dendritic cells (DC) play unique and diverse roles in the tumor occurrence, development, progression and response to therapy. First of all, DC can actively uptake tumor-associated antigens, process them and present antigenic peptides to T cells inducing and maintaining tumor-specific T cell responses. DC interaction with different immune effector cells may also support innate antitumor immunity, as well as humoral responses also known to inhibit tumor development in certain cases. On the other hand, DC are recruited to the tumor site by specific tumor-derived and stroma-derived factors, which may also impair DC maturation, differentiation and function, thus resulting in the deficient formation of antitumor immune response or development of DC-mediated tolerance and immune suppression. Identification of DC-stimulating and DC-suppressing/polarizing factors in the tumor environment and the mechanism of DC modulation are important for designing effective DC-based vaccines and for recovery of immunodeficient resident DC responsible for maintenance of clinically relevant antitumor immunity in patients with cancer. DC-targeting tumor-derived factors and their effects on resident and administered DC in the tumor milieu are described and discussed in this review.
Collapse
Affiliation(s)
- Jinbao Zong
- Department of Pathology, University of Pittsburgh Medical Center, Scaife Hall S735, 3550 Terrace Street, Pittsburgh, PA, 15261, USA
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao City, China
| | - Anton A Keskinov
- Department of Pathology, University of Pittsburgh Medical Center, Scaife Hall S735, 3550 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Galina V Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Scaife Hall S735, 3550 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Michael R Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Scaife Hall S735, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
16
|
Sensitivity of Dendritic Cells to Microenvironment Signals. J Immunol Res 2016; 2016:4753607. [PMID: 27088097 PMCID: PMC4819111 DOI: 10.1155/2016/4753607] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/03/2016] [Accepted: 03/06/2016] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells are antigen-presenting cells capable of either activating the immune response or inducing and maintaining immune tolerance. They do this by integrating stimuli from the environment and changing their functional status as a result of plasticity. The modifications suffered by these cells have consequences in the way the organism may respond. In the present work two opposing situations known to affect dendritic cells are analyzed: tumor growth, leading to a microenvironment that favors the induction of a tolerogenic profile, and organ transplantation, which leads to a proinflammatory profile. Lessons learned from these situations may help to understand the mechanisms of modulation resulting not only from the above circumstances, but also from other pathologies.
Collapse
|
17
|
T Lymphocyte Inhibition by Tumor-Infiltrating Dendritic Cells Involves Ectonucleotidase CD39 but Not Arginase-1. BIOMED RESEARCH INTERNATIONAL 2015; 2015:891236. [PMID: 26491691 PMCID: PMC4605267 DOI: 10.1155/2015/891236] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/18/2015] [Accepted: 08/25/2015] [Indexed: 12/14/2022]
Abstract
T lymphocytes activated by dendritic cells (DC) which present tumor antigens play a key role in the antitumor immune response. However, in patients suffering from active cancer, DC are not efficient at initiating and supporting immune responses as they participate to T lymphocyte inhibition. DC in the tumor environment are functionally defective and exhibit a characteristic of immature phenotype, different to that of DC present in nonpathological conditions. The mechanistic bases underlying DC dysfunction in cancer responsible for the modulation of T-cell responses and tumor immune escape are still being investigated. Using two different mouse tumor models, we showed that tumor-infiltrating DC (TIDC) are constitutively immunosuppressive, exhibit a semimature phenotype, and impair responder T lymphocyte proliferation and activation by a mechanism involving CD39 ectoenzyme.
Collapse
|
18
|
Motta JM, Sperandio A, Castelo-Branco MTL, Rumjanek VM. Induction of suppressive phenotype in monocyte-derived dendritic cells by leukemic cell products and IL-1β. Hum Immunol 2014; 75:641-9. [PMID: 24768898 DOI: 10.1016/j.humimm.2014.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 03/11/2014] [Accepted: 04/11/2014] [Indexed: 12/28/2022]
Abstract
Professional antigen-presenting cells, dendritic cells (DCs) play an important role in controlling tumors. It is known that solid tumor cell products inhibit DC differentiation. Recently a similar effect produced by leukemic cell products has been demonstrated. In this case, leukemic cell products induced the secretion of IL-1β by monocytes undergoing differentiation. The aim of the present work was to characterize and to compare the development of monocyte-derived DCs under the influence of leukemic cell products (K562 supernatant) or exogenous IL-1β. It became clear that leukemic cell products and IL-1β differentially modulate some of the parameters studied on monocytes stimulated to differentiate into DCs. In the presence of K562 supernatant, the expression of the macrophage markers CD16 and CD68 were higher than in immature DCs control. Contrasting with IL-1β, leukemic cell products possibly favor the development of cells with macrophage markers. In addition, CD80 and CD83 expressions were also higher in the presence of tumor supernatant whereas HLA-DR was lower. In the presence of IL-1β, only CD80 was increased. Furthermore, it was observed that when monocytes were induced to differentiate into DCs in the presence of tumor supernatant and then activated, they expressed less CD80 and CD83 than activated DCs control. A reduced expression of CD83 following activation was also seen in cells differentiated with IL-1β. TGF-β and VEGF were found in the tumor supernatants. Moreover, the exposure to tumor supernatant or IL-1β stimulated IL-10 production while decreased IL-12 production by activated DCs. Finally, these results suggest that the addition of products released by leukemic cells or, more discreetly, the addition of IL-1β affects DC differentiation, inducing a suppressive phenotype.
Collapse
Affiliation(s)
- Juliana Maria Motta
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aline Sperandio
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Vivian Mary Rumjanek
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
19
|
Horton JS, Stokes AJ. The transmembrane channel-like protein family and human papillomaviruses: Insights into epidermodysplasia verruciformis and progression to squamous cell carcinoma. Oncoimmunology 2014; 3:e28288. [PMID: 24800179 PMCID: PMC4006860 DOI: 10.4161/onci.28288] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 02/13/2014] [Accepted: 02/19/2014] [Indexed: 12/25/2022] Open
Abstract
Epidermodysplasia verruciformis (EV) is a rare genodermatosis characterized by increased sensitivity to infection by the β-subtype of human papillomaviruses (β-HPVs), causing persistent, tinea versicolor-like dermal lesions. In a majority of affected individuals, these macular lesions progress to invasive cutaneous squamous cell carcinoma (CSCC) in sun-exposed areas. While mutations in transmembrane channel-like 6 (TMC6 / EVER1) and 8 (TMC8 / EVER2) have been causally linked to EV, their molecular functions are unclear. It is likely that their protective effects involve regulation of the β-HPV life cycle, host keratinocyte apoptosis vs. survival balance and/or T-cell interaction with infected host cells.
Collapse
Affiliation(s)
- Jaime S Horton
- Laboratory of Experimental Medicine; John A. Burns School of Medicine; Honolulu, HI USA ; Department of Cell and Molecular Biology; John A. Burns School of Medicine; Honolulu, HI USA
| | - Alexander J Stokes
- Laboratory of Experimental Medicine; John A. Burns School of Medicine; Honolulu, HI USA ; Department of Cell and Molecular Biology; John A. Burns School of Medicine; Honolulu, HI USA ; Chaminade University; Honolulu, HI USA
| |
Collapse
|
20
|
Zhong H, Gutkin DW, Han B, Ma Y, Keskinov AA, Shurin MR, Shurin GV. Origin and pharmacological modulation of tumor-associated regulatory dendritic cells. Int J Cancer 2014; 134:2633-45. [PMID: 24443321 DOI: 10.1002/ijc.28590] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 10/28/2013] [Accepted: 10/30/2013] [Indexed: 12/29/2022]
Abstract
Protumorigenic activity of immune regulatory cells has been proven to play a major role in precluding immunosurveillance and limiting the efficacy of anticancer therapies. Although several approaches have been offered to deplete myeloid-derived suppressor cells (MDSC) and regulatory T cells, there are no data on how to control suppressive dendritic cell (DC) accumulation or function in the tumor environment. Although immunosuppressive function of DC in cancer was implicated to immature and plasmacytoid DC, details of how conventional DC (cDC) develop immunosuppressive properties remain less understood. Here, we show that the development of lung cancer in mice was associated with fast accumulation of regulatory DC (regDC) prior to the appearance of MDSC. Using the in vitro and in vivo approaches, we demonstrated that (i)both cDC and MDSC could be polarized into protumor regDC in the lung cancer environment; (ii) cDC → regDC polarization was mediated by the small Rho GTPase signaling, which could be controlled by noncytotoxic doses of paclitaxel; and (iii) prevention of regDC appearance increased the antitumor potential of DC vaccine in lung cancer. These findings not only bring new players to the family of myeloid regulatory cells and provide new targets for cancer therapy, but offer novel insights into the immunomodulatory capacity of chemotherapeutic agents used in low, noncytotoxic doses.
Collapse
Affiliation(s)
- Hua Zhong
- Department of Pulmonary Disease, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Dumas G, Dufresne M, Asselin É, Girouard J, Carrier C, Reyes-Moreno C. CD40 pathway activation reveals dual function for macrophages in human endometrial cancer cell survival and invasion. Cancer Immunol Immunother 2013; 62:273-83. [PMID: 22903346 PMCID: PMC11028733 DOI: 10.1007/s00262-012-1333-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 07/31/2012] [Indexed: 11/28/2022]
Abstract
Reproductive malignancies are a major cause of cancer death in women worldwide. CD40 is a TNF receptor family member, which upon activation may mediate tumor regression. However, despite the great potential of CD40 agonists, their use as a therapeutic option for reproductive cancers has never been investigated. Because CD40 ligation is a potent pathway of macrophage activation, an in vitro model of pro-inflammatory type-1 (Mϕ-1) and anti-inflammatory type-2 (Mϕ-2) macrophages was developed to determine whether and how macrophage CD40 pathway activation might influence endometrial tumor cell behavior. Analysis of tumor growth kinetic in the endometrial cancer xenograft model indicates that, when injected once into the growing tumors, CD40-activated Mϕ-1 greatly reduced, while CD40-activated Mϕ-2 increased tumor size when compared to control isotype-activated Mϕ-1 and Mϕ-2, respectively. In vitro assays indicated that CD40-activated Mϕ-2 increased cell viability but failed to promote cell invasion. CD40-activated Mϕ-1, in contrast, decreased cell survival but greatly increased cell invasion in tumor cells less susceptible to cell death by apoptosis; they also induced the expression of some pro-inflammatory genes, such as IL-6, LIF, and TNF-α, known to be involved in tumor promotion and metastasis. The presence of IFN-γ is minimally required for CD40-activated Mϕ-1 to promote tumor cell invasion, a process that is mediated in part through the activation of the PI3K/Akt2 signaling pathway in tumor cells. From these results, we speculate that some functions of CD40 in tumor-associated Mϕs might limit the therapeutic development of CD40 agonists in endometrial cancer malignancies.
Collapse
Affiliation(s)
- Geneviève Dumas
- Research Group in Molecular Oncology and Endocrinology, University of Quebec at Trois-Rivieres, Trois-Rivières, PQ G9A 5H7 Canada
| | - Mathieu Dufresne
- Research Group in Molecular Oncology and Endocrinology, University of Quebec at Trois-Rivieres, Trois-Rivières, PQ G9A 5H7 Canada
| | - Éric Asselin
- Research Group in Molecular Oncology and Endocrinology, University of Quebec at Trois-Rivieres, Trois-Rivières, PQ G9A 5H7 Canada
| | - Julie Girouard
- Research Group in Molecular Oncology and Endocrinology, University of Quebec at Trois-Rivieres, Trois-Rivières, PQ G9A 5H7 Canada
| | - Christian Carrier
- Haemato-oncologic Service, Regional Hospital of Trois-Rivieres, Trois-Rivières, PQ G8Z 3R9 Canada
| | - Carlos Reyes-Moreno
- Research Group in Molecular Oncology and Endocrinology, University of Quebec at Trois-Rivieres, Trois-Rivières, PQ G9A 5H7 Canada
- Department of Chemistry-Biology, University of Quebec at Trois-Rivieres, 3351 boul. des Forges, C.P. 500, Trois-Rivieres, G9A 5H7 Canada
| |
Collapse
|
22
|
Ma Y, Shurin GV, Peiyuan Z, Shurin MR. Dendritic cells in the cancer microenvironment. J Cancer 2012; 4:36-44. [PMID: 23386903 PMCID: PMC3564245 DOI: 10.7150/jca.5046] [Citation(s) in RCA: 244] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 12/01/2012] [Indexed: 01/01/2023] Open
Abstract
The complexity of the tumor immunoenvironment is underscored by the emergence and discovery of different subsets of immune effectors and regulatory cells. Tumor-induced polarization of immune cell differentiation and function makes this unique environment even more intricate and variable. Dendritic cells (DCs) represent a special group of cells that display different phenotype and activity at the tumor site and exhibit differential pro-tumorigenic and anti-tumorigenic functions. DCs play a key role in inducing and maintaining the antitumor immunity, but in the tumor environment their antigen-presenting function may be lost or inefficient. DCs might be also polarized into immunosuppressive/tolerogenic regulatory DCs, which limit activity of effector T cells and support tumor growth and progression. Although various factors and signaling pathways have been described to be responsible for abnormal functioning of DCs in cancer, there are still no feasible therapeutic modalities available for preventing or reversing DC malfunction in tumor-bearing hosts. Thus, better understanding of DC immunobiology in cancer is pivotal for designing novel or improved therapeutic approaches that will allow proper functioning of DCs in patients with cancer.
Collapse
Affiliation(s)
- Yang Ma
- 1. Departments of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
23
|
Kim JW, Bilusic M, Heery CJ, Madan RA. Therapeutic cancer vaccines in prostate cancer: the quest for intermediate markers of response. Cancers (Basel) 2012; 4:1229-46. [PMID: 24213505 PMCID: PMC3712729 DOI: 10.3390/cancers4041229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/09/2012] [Accepted: 11/14/2012] [Indexed: 11/17/2022] Open
Abstract
Despite recent advances in cancer immunotherapy, no prospectively validated intermediate biomarkers exist to predict response. These biomarkers are highly desirable given modern immunotherapy's paradoxical pattern of clinical benefit; that is, improvement in overall survival without short-term change in progression. Immunotherapy clinical trials have evaluated biomarkers that may correlate with clinical outcomes. Many of them are performed on peripheral blood to evaluate the systemic response, such as tumor-targeted humoral and cellular immunity, and cytokine responses. Accumulating evidence suggests that immune infiltrates in tumors may suggest evidence for the therapy's mechanism of action, and have greater potential for providing prognostic and predictive information. In addition, a non-immunologic biomarker, such as tumor growth kinetics, may explain this paradoxical pattern of clinical benefit, and predict survival in patients treated with an immunotherapy. Prospective assessment and validation of these and other intermediate markers would be required to better understand their potential clinical role.
Collapse
Affiliation(s)
- Joseph W Kim
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
24
|
Abstract
Receptor internalization is a common mechanism underlying surface receptor down-regulation (and thus receptor signaling) upon its engagement with the cognate ligand. Tight regulation of surface CD40 expression is critical in regulating different functional properties of dendritic cell (DC). Engagement of CD40 on mature DC and the cognate CD40 ligand on T cell activates c-Jun N-terminal MAPK, p38 and ERK1/2 MAPK pathways in mature DC. JNK-associated leucine zipper protein (JLP) is a scaffolding protein that interacted with p38 and JNK. The molecular mechanism underlying CD40 internalization and its physiological impact on DC functions remained unclear. Here we reported that the engagement of CD40 on the LPS-activated DC down-regulated the surface expression of CD40. We examined the role of the JLP protein in DC differentiation, and in the regulation of DC function(s) in vitro. In contrast to the abundant JLP expression observed in immortal cell lines, primary immature DC expressed low levels of the JLP proteins. The induction of the JLP protein expression was observed in the LPS-mature DC that were activated by CD40 ligation, and also in the poly I:C stimulated DC. JLP-silenced DC was impaired in regulating CD40 surface expression upon LPS stimulation and CD40 induced receptor internalization. Such aberrant change in the regulation of surface CD40 expression was associated with an augmented capacity of the JLP-silenced DC in IL-12 production. Collectively, our data identified a novel role of a scaffolding protein JLP in the regulation of surface CD40 expression and fine-tuning of DC function.
Collapse
|
25
|
Nars MS, Kaneno R. Immunomodulatory effects of low dose chemotherapy and perspectives of its combination with immunotherapy. Int J Cancer 2012; 132:2471-8. [PMID: 22927096 DOI: 10.1002/ijc.27801] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/29/2012] [Accepted: 08/16/2012] [Indexed: 02/06/2023]
Abstract
Given that cancer is one of the main causes of death worldwide, many efforts have been directed toward discovering new treatments and approaches to cure or control this group of diseases. Chemotherapy is the main treatment for cancer; however, a conventional schedule based on maximum tolerated dose (MTD) shows several side effects and frequently allows the development of drug resistance. On the other side, low dose chemotherapy involves antiangiogenic and immunomodulatory processes that help host to fight against tumor cells, with lower grade of side effects. In this review, we present evidence that metronomic chemotherapy, based on the frequent administration of low or intermediate doses of chemotherapeutics, can be better than or as efficient as MTD. Finally, we present some data indicating that noncytotoxic concentrations of antineoplastic agents are able to both up-regulate the immune system and increase the susceptibility of tumor cells to cytotoxic T lymphocytes. Taken together, data from the literature provides us with sufficient evidence that low concentrations of selected chemotherapeutic agents, rather than conventional high doses, should be evaluated in combination with immunotherapy.
Collapse
Affiliation(s)
- Mariana S Nars
- Department of Microbiology and Immunology, Institute of Biosciences, UNESP-Univ Estadual Paulista, Botucatu, São Paulo, Brazil
| | | |
Collapse
|
26
|
Gao J, Luo SM, Peng ML, Deng T. Enhanced immunity against hepatoma induced by dendritic cells pulsed with Hsp70-H22 peptide complexes and CD40L. J Cancer Res Clin Oncol 2012; 138:917-26. [PMID: 22327301 DOI: 10.1007/s00432-012-1166-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 01/25/2012] [Indexed: 11/24/2022]
Abstract
PURPOSE Dendritic cell (DC)-based cancer vaccines have become an attractive antitumour therapeutic approach. However, clinical application of current DC-based cancer vaccines has been limited by their ineffectiveness. Heat shock protein 70 from Mycobacterium tuberculosis (TBhsp70) is known to have a potent adjuvant capability to induce maturation of DCs and thus acts as an alternative ligand to the CD40 ligand (CD40L) on T cells to induce a T-cell response. The aim of this study is to investigate whether the combination of TBhsp70-H22 tumour-peptide complexes and CD40L might improve the antitumour efficacy for development of therapeutic DC-based vaccines against hepatoma. METHODS The CD40, CD80, CD86 and HLA-DR expression on DCs pulsed with TBhsp70-H22 tumour-peptide complexes and soluble CD40L was studied by flow cytometric analysis, and T-helper type 1 cytokine secretion, such as IL-12p70 secretion, was tested by ELISA. The H22-specific cytotoxic T-lymphocytes (CTLs) were detected by a (51)Cr-release assay, and the in vivo antitumour immunity against hepatoma was measured by utilising H22-tumour-bearing mice after therapeutic administration. RESULTS Up-regulation of CD40, CD80, CD86 and HLA-DR expression on DCs pulsed with TBhsp70-H22 tumour-peptide complexes and CD40L was found, which stimulated a high level of T-helper type 1 cytokine secretion, such as IL-12p70, and resulted in the induction of H22-specific CTLs. The therapeutic administration of DCs pulsed in vitro with TBhsp70-H22 tumour-peptide complexes and CD40L significantly reduced the progression of H22 tumours in mice compared with DC-Hsp70-H22 peptide complexes or DC-CD40L alone. CONCLUSIONS Our findings demonstrate that DCs pulsed with Hsp70-H22-peptide complexes and CD40L enhance the antitumour immunity against hepatoma, which provides a novel immunotherapeutic approach against cancer.
Collapse
Affiliation(s)
- Jian Gao
- Department of Gastroenterology, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China.
| | | | | | | |
Collapse
|
27
|
Shurin GV, Ouellette CE, Shurin MR. Regulatory dendritic cells in the tumor immunoenvironment. Cancer Immunol Immunother 2012; 61:223-230. [PMID: 22065047 PMCID: PMC3314382 DOI: 10.1007/s00262-011-1138-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 10/21/2011] [Indexed: 12/13/2022]
Abstract
The tumor microenvironment is a pivotal factor in tumorigenesis, and especially in progression, as the pathogenesis of cancer critically depends on the complex interactions between various microenvironmental components. A key component of the tumor immunoenvironment is the infiltration of immune cells, which has been proven to play a dual role in tumor growth and progression. This Janus two-faced function of the tumor immunoenvironment is seen in tumor infiltration by T cells, which correlates with improved patient survival, but also with the homing of multiple subsets of immunoregulatory cells that inhibit the antitumor immune response. Regulatory dendritic cells (regDCs) have recently been shown to be induced by tumor-derived factors and represent a new and potentially important player in supporting tumor progression and suppressing the development of antitumor immune responses. Our recent data reveal that different tumor cell lines produce soluble factors that induce polarization of conventional DCs into regDCs, both in vitro and in vivo. These regDCs can suppress the proliferation of pre-activated T cells and are phenotypically and functionally different from their precursors as well as the classical immature conventional DCs. Understanding the biology of regDCs and the mechanisms of their formation in the tumor immunoenvironment will provide a new therapeutic target for re-polarizing protumorigenic immunoregulatory cells into proimmunogenic effector cells able to induce and support effective antitumor immunity.
Collapse
Affiliation(s)
- Galina V Shurin
- Division of Experimental Pathology, Department of Pathology and Immunology, University of Pittsburgh Medical Center, 3550 Terrace Street, Scaife Hall S733, Pittsburgh, PA, 15261, USA.
| | - Camille E Ouellette
- Division of Experimental Pathology, Department of Pathology and Immunology, University of Pittsburgh Medical Center, 3550 Terrace Street, Scaife Hall S733, Pittsburgh, PA, 15261, USA
| | - Michael R Shurin
- Division of Experimental Pathology, Department of Pathology and Immunology, University of Pittsburgh Medical Center, 3550 Terrace Street, Scaife Hall S733, Pittsburgh, PA, 15261, USA
| |
Collapse
|
28
|
Mahipal A, Terai M, Berd D, Chervoneva I, Patel K, Mastrangelo MJ, Sato T. Tumor-derived interleukin-10 as a prognostic factor in stage III patients undergoing adjuvant treatment with an autologous melanoma cell vaccine. Cancer Immunol Immunother 2011; 60:1039-45. [PMID: 21519827 PMCID: PMC11029583 DOI: 10.1007/s00262-011-1019-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 04/07/2011] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Interleukin-10 (IL-10) downregulates T-cell-mediated immune responses. We studied the association between IL-10 production by freshly isolated melanoma cell suspensions in vitro and overall survival in patients undergoing adjuvant treatment with a vaccine prepared from the same autologous melanoma cells modified with a hapten, dinitrophenyl (DNP). METHODS Forty-four patients with cutaneous melanoma (29 stage III and 15 stage IV) were prospectively evaluated. Tumor cells were extracted from metastatic deposits for production of DNP-modified autologous melanoma cell vaccine. Small aliquots of the melanoma cell suspensions were separated prior to vaccine processing and cultured overnight for IL-10 production. Based on a blind assessment of the distribution of IL-10 levels in the culture supernatants, a cutoff of 200 pg/ml was used to define high versus low IL-10 producers. Cox regression model was used for multivariate analysis. Overall survival was calculated using the Kaplan-Meier method, and survival curves were compared with the log-rank test. RESULTS Out of 44 patients, 29 were low and 15 were high IL-10 producers. The median OS was significantly worse for high compared with low IL-10 producers (10.5 months vs. 42 months; P = 0.022). In stage III patients, the multivariate hazard ratio for high versus low IL-10 producers was 2.92 (95% CI, 1.04-8.20; P = 0.041). The corresponding hazard ratio in stage IV patients was 0.92 (95% CI, 1.04-8.20; P = 0.888). CONCLUSIONS High IL-10 production in the tumor microenvironment could be a determinant of clinical outcomes in stage III melanoma patients receiving autologous melanoma cell vaccine.
Collapse
Affiliation(s)
- Amit Mahipal
- Department of Medical Oncology, Thomas Jefferson University, 1025 Walnut Street, Suite 1024, Philadelphia, PA 19107 USA
| | - Mizue Terai
- Department of Medical Oncology, Thomas Jefferson University, 1025 Walnut Street, Suite 1024, Philadelphia, PA 19107 USA
- Department of Molecular and Tumor Pathology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - David Berd
- Cancer Treatment Centers of America, Eastern Regional Medical Center, Philadelphia, PA USA
| | - Inna Chervoneva
- Division of Biostatistics, Thomas Jefferson University, Philadelphia, PA USA
| | - Kashyap Patel
- Carolina Blood and Cancer Care Associates, Rock Hill, SC USA
| | - Michael J. Mastrangelo
- Department of Medical Oncology, Thomas Jefferson University, 1025 Walnut Street, Suite 1024, Philadelphia, PA 19107 USA
| | - Takami Sato
- Department of Medical Oncology, Thomas Jefferson University, 1025 Walnut Street, Suite 1024, Philadelphia, PA 19107 USA
| |
Collapse
|
29
|
Motta JM, Nascimento CR, Rumjanek VM. Leukemic cell products down-regulate human dendritic cell differentiation. Cancer Immunol Immunother 2010; 59:1645-53. [PMID: 20607236 PMCID: PMC11031054 DOI: 10.1007/s00262-010-0890-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Accepted: 06/21/2010] [Indexed: 12/25/2022]
Abstract
The microenvironment produced by solid tumors is inhibitory to the immune system, inducing dendritic cell (DC) alterations, but there is a paucity of information regarding haematological malignances. The aim of this study was to investigate DC differentiation under the influence of leukemic cell products. Monocytes from healthy volunteers were cultured in the presence of IL-4 and GM-CSF for the generation of immature DCs. Supernatants from leukemic cultures were added to monocyte cultures during differentiation. The lineages used were K562, a chronic myeloid leukemia, HL-60, a promyelocytic leukemia and DAUDI, originated from Burkitt lymphoma. It was observed that the expression of CD14 remained high and the CD1a was low in the presence of tumor supernatants, while non-malignant supernatants did not affect these parameters. Furthermore, IL-1beta and TNF-alpha production by monocytes during differentiation was increased by the presence of tumor supernatants. The modifications on CD14 and CD1a expressions could be mimicked by the addition of exogenous IL-1beta and partially inhibited by the neutralization of IL-1beta. These results suggest that soluble products from leukemic cells interfere with DC differentiation and, in the present work, this effect could be mediated by monocyte-derived IL-1beta in response to tumor supernatants.
Collapse
Affiliation(s)
- Juliana Maria Motta
- Laboratório de Imunologia Tumoral, Instituto de Bioquímica Médica, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Bloco H2, Sala 003, Rio de Janeiro, 21941-590 Brazil
| | - Clarissa Rodrigues Nascimento
- Laboratório de Imunologia Tumoral, Instituto de Bioquímica Médica, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Bloco H2, Sala 003, Rio de Janeiro, 21941-590 Brazil
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivian Mary Rumjanek
- Laboratório de Imunologia Tumoral, Instituto de Bioquímica Médica, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Bloco H2, Sala 003, Rio de Janeiro, 21941-590 Brazil
| |
Collapse
|
30
|
Whiteside TL. Inhibiting the inhibitors: evaluating agents targeting cancer immunosuppression. Expert Opin Biol Ther 2010; 10:1019-35. [PMID: 20415597 DOI: 10.1517/14712598.2010.482207] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
IMPORTANCE OF THE FIELD Immunotherapy of cancer has not improved disease-free or overall patient survival. The lack of concordance between immunological and clinical responses in cancer immunotherapy trials is thought to result from the pervasive presence of tumor-driven immune suppression that allows tumor to escape and that has not been adequately targeted by current therapies. AREAS COVERED IN THIS REVIEW Because multiple mechanisms of tumor induced suppression have been identified and shown to contribute to tumor escape, the opportunity arises to interfere with these mechanisms. A range of known tumor-derived inhibitors can now be blocked or neutralized by biologic or metabolic agents. Used alone or in combination with each other or with conventional cancer therapies, these agents offer novel therapeutic strategies for the control of tumor escape. WHAT THE READER WILL GAIN This review deals with currently available inhibitors for counteracting tumor immune escape. The restoration of effective anti-tumor immunity in patients with cancer will require new approaches aiming at: i) protection of immune cells from adverse effects of myeloid-derived suppressor cells, regulatory T cells or inhibitory factors thus enhancing effector functions; and ii) prolonging survival of central memory T cells, thus ensuring long-term protection. TAKE HOME MESSAGE Inhibitors of mechanisms responsible for tumor escape could restore anti-tumor immune responses in patients with cancer.
Collapse
Affiliation(s)
- Theresa L Whiteside
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pathology, 5117 Centre Avenue, Pittsburgh, 15213 USA.
| |
Collapse
|
31
|
Increase of in vivo antitumoral activity by CD40L (CD154) gene transfer into pancreatic tumor cell-dendritic cell hybrids. Pancreas 2009; 38:758-65. [PMID: 19546834 DOI: 10.1097/mpa.0b013e3181ae5e1a] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Fusion of dendritic cells (DC) with tumor cells is an approach in immunotherapy combining antigenicity and capacity of antigen presentation to activate T cells for the induction of tumor-specific cytotoxic immunity. Although there have been reports of clinical benefit, response rates have been limited and further improvements are warranted. METHODS We used murine DC and a novel protocol for an effective fusion of those cells with the murine pancreatic cell line Panc02. RESULTS We observed 2 events: only moderate in vitro and in vivo cytotoxicity of tumor cell/DC hybrids and a down-regulation of costimulatory molecules on fused cells. Therefore, we transfected tumor cell/DC hybrids with an adenovirus expressing CD154 to improve DC activation and generating antitumor immune response without the need of CD4 T cells. High CD154 expression could be obtained by transfection of DC and Panc02 cells prior fusion. Furthermore, vaccination with CD154-transfected tumor cell/DC hybrid led to a significantly increased induction of cytotoxic T cells in vitro and to an improved antitumoral effect in an orthotopic in vivo mouse model. CONCLUSIONS CD154-transfected tumor cell/DC hybrids are a promising approach to increase the efficiency of antitumoral response.
Collapse
|
32
|
Murugaiyan G, Saha B. Protumor vs Antitumor Functions of IL-17. THE JOURNAL OF IMMUNOLOGY 2009; 183:4169-75. [DOI: 10.4049/jimmunol.0901017] [Citation(s) in RCA: 281] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
33
|
Kaneno R, Shurin GV, Tourkova IL, Shurin MR. Chemomodulation of human dendritic cell function by antineoplastic agents in low noncytotoxic concentrations. J Transl Med 2009; 7:58. [PMID: 19591684 PMCID: PMC2716306 DOI: 10.1186/1479-5876-7-58] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Accepted: 07/10/2009] [Indexed: 11/19/2022] Open
Abstract
The dose-delivery schedule of conventional chemotherapy, which determines its efficacy and toxicity, is based on the maximum tolerated dose. This strategy has lead to cure and disease control in a significant number of patients but is associated with significant short-term and long-term toxicity. Recent data demonstrate that moderately low-dose chemotherapy may be efficiently combined with immunotherapy, particularly with dendritic cell (DC) vaccines, to improve the overall therapeutic efficacy. However, the direct effects of low and ultra-low concentrations on DCs are still unknown. Here we characterized the effects of low noncytotoxic concentrations of different classes of chemotherapeutic agents on human DCs in vitro. DCs treated with antimicrotubule agents vincristine, vinblastine, and paclitaxel or with antimetabolites 5-aza-2-deoxycytidine and methotrexate, showed increased expression of CD83 and CD40 molecules. Expression of CD80 on DCs was also stimulated by vinblastine, paclitaxel, azacytidine, methotrexate, and mitomycin C used in low nontoxic concentrations. Furthermore, 5-aza-2-deoxycytidine, methotrexate, and mitomycin C increased the ability of human DCs to stimulate proliferation of allogeneic T lymphocytes. Thus, our data demonstrate for the first time that in low noncytotoxic concentrations chemotherapeutic agents do not induce apoptosis of DCs, but directly enhance DC maturation and function. This suggests that modulation of human DCs by noncytotoxic concentrations of antineoplastic drugs, i.e. chemomodulation, might represent a novel approach for up-regulation of functional activity of resident DCs in the tumor microenvironment or improving the efficacy of DCs prepared ex vivo for subsequent vaccinations.
Collapse
Affiliation(s)
- Ramon Kaneno
- Department of Microbiology and Immunology, Institute of Biosciences, São Paulo State University, Botucatu, SP, Brazil.
| | | | | | | |
Collapse
|
34
|
Mees ST, Mardin WA, Sielker S, Willscher E, Senninger N, Schleicher C, Colombo-Benkmann M, Haier J. Involvement of CD40 targeting miR-224 and miR-486 on the progression of pancreatic ductal adenocarcinomas. Ann Surg Oncol 2009; 16:2339-50. [PMID: 19475450 DOI: 10.1245/s10434-009-0531-4] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 04/30/2009] [Accepted: 04/30/2009] [Indexed: 01/10/2023]
Abstract
BACKGROUND Genetic and epigenetic alterations during development of pancreatic ductal adenocarcinomas (PDAC) are well known. Genetic and epigenetic data were correlated with tumor biology to find specific alterations responsible for invasion and metastasis in pancreatic ductal adenocarcinomas. METHODS A total of 16 human PDAC cell lines were used in murine orthotopic PDAC models. By means of standardized dissemination scores, local invasion and metastatic spread were assessed. mRNA and microRNA expression were studied by microarray and TaqMan low-density array. Quantitative real-time-polymerase chain reaction and flow cytometry were used for expression validation. RESULTS CD40 was detected as a relevant target gene for differentially expressed miRNAs observed in highly invasive and metastatic PDAC only. A significant overexpression (P < .05) of CD40-related miRNAs miR-224 and miR-486 was detected in highly invasive and metastatic PDAC, whereas CD40 mRNA expression was not significantly altered. Instead, CD40 protein expression at cell surfaces of these highly invasive and metastatic PDAC was significantly reduced (P < .01). CONCLUSIONS Epigenetic alterations with upregulated CD40-targeting miR-224 and miR-486 are related to downregulated CD40 protein expression at cell surfaces in highly invasive and metastatic PDAC. Thus, miRNA-regulated CD40 expression seems to play an important role in progression of PDAC. These data suggest a diagnostic and therapeutic potential for CD40 and/or its targeting miRNAs in PDAC.
Collapse
Affiliation(s)
- Soeren Torge Mees
- Department of General and Visceral Surgery, University Hospital of Muenster, Muenster, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Morse MA, Hall JR, Plate JMD. Countering tumor-induced immunosuppression during immunotherapy for pancreatic cancer. Expert Opin Biol Ther 2009; 9:331-9. [PMID: 19216622 DOI: 10.1517/14712590802715756] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Vaccines for pancreatic cancer have been challenged by a number of factors, especially the immunosuppressive microenvironment within the tumor that allows for escape from immune surveillance. OBJECTIVE/METHODS We sought to identify results that define mechanisms of pancreatic-cancer-associated immunosuppression and strategies that might be useful to overcome them thereby resulting in effective immune responses to cancer vaccines capable of deleting pancreatic cancer cells. RESULTS/CONCLUSION Immunosuppressive tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSC), and regulatory T cells (Treg) reside in tumors, and their products along with tumor derived products (such as VEGF, TGFbeta and IL-10), create a microenvironment that counters immune activation and attack. Immunotherapy with cancer vaccines must include strategies to modulate these immunosuppressive cell types and tumor byproducts. Clinical trials are beginning to test these strategies.
Collapse
|
36
|
Rogers HD, Macgregor JL, Nord KM, Tyring S, Rady P, Engler DE, Grossman ME. Acquired epidermodysplasia verruciformis. J Am Acad Dermatol 2009; 60:315-20. [PMID: 19150275 DOI: 10.1016/j.jaad.2008.08.035] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Revised: 08/15/2008] [Accepted: 08/25/2008] [Indexed: 11/28/2022]
Abstract
Epidermodysplasia verruciformis (EV) is a rare autosomal recessive genodermatosis with an increased susceptibility to specific human papillomavirus (HPV) genotypes. Classically, this viral infection leads to the development of tinea versicolor-like macules on the trunk, neck, arms, and face during childhood, and over time, these lesions can progress to squamous cell carcinoma. More recently, an EV-like syndrome has been described in patients with impaired cell-mediated immunity. We describe two cases of EV-like syndrome in HIV-positive patients, review all previously reported cases of EV in patients with impaired cell-mediated immunity, introduce the term "acquired epidermodysplasia verruciformis" to describe EV developing in the immunocompromised host and examine the limited treatment options for these patients.
Collapse
Affiliation(s)
- Heather D Rogers
- Department of Dermatology, Columbia University Medical Center, New York, New York, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Gonzalez-Carmona MA, Lukacs-Kornek V, Timmerman A, Shabani S, Kornek M, Vogt A, Yildiz Y, Sievers E, Schmidt-Wolf IGH, Caselmann WH, Sauerbruch T, Schmitz V. CD40ligand-expressing dendritic cells induce regression of hepatocellular carcinoma by activating innate and acquired immunity in vivo. Hepatology 2008; 48:157-68. [PMID: 18537185 DOI: 10.1002/hep.22296] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
UNLABELLED Dendritic cells (DCs) are professional antigen-presenting cells able to prime T-cells against tumor-associated antigens (TAA), but their potential to induce hepatocellular carcinoma (HCC) regression is still limited. CD40/CD40L interaction is essential for DC activation and induction of antigen-specific T-cells. In this study, transduction of TAA-pulsed DC with a CD40L-encoding adenovirus (Ad-CD40L) was used to improve the immune response induced by DC toward HCC. Bone marrow-derived DC from C3H/HeNcrl mice were cultured with granulocyte-macrophage colony-stimulating factor and interleukin-4. On day 6, tumor-lysate pulsed DCs were infected with adenoviruses. HCCs were induced by inoculation of mice with Hepa129-cells subcutaneously. When tumor-volume was 100 to 400 mm(3), DCs were injected intratumorally, subcutaneously, or intravenously. Ad-CD40L transduction exerted CD40/CD40L interactions between DCs, increasing DC immunostimulation with up-regulation of CD80/CD86- and interleukin-12 (IL-12) expression. Intratumoral injection of CD40L-DC was superior to intravenous or subcutaneous treatments, yielding tumor elimination in almost 70% of mice. Moreover, all tumor-free animals were protected against hepatic tumor cell rechallenge. In a preventive setting, subcutaneous injection of CD40L-expressing DCs protected 50% of mice for more than 3 months toward tumor cell challenge. The induced immune response seemed to be dependent on cross-priming with Th1-lymphocytes in the lymph nodes, because transduced DCs were redetected in lymphoid tissues. In addition, immunohistochemistry of tumors indicated a significant tumor infiltration with CD4+, CD8+ T cells and natural killer (NK) cells. Tumor-infiltrating lymphocytes were tumor-specific, as shown in interferon-gamma (IFN-gamma) enzyme-linked immunosorbent spot and T-cell proliferation assays. CONCLUSION Transduction of DCs with Ad-CD40L increases significantly the stimulatory capacity of DCs. Intratumoral injection of DCs activates both acquired and innate immunity, inducing complete regression of established tumors and long-term immunity against tumor recurrence. This approach improves the antitumoral potential of DCs.
Collapse
|
38
|
Abstract
Inflammatory cell infiltration of tumors contributes either positively or negatively to tumor invasion, growth, metastasis, and patient outcomes, creating a Dr. Jekyll or Mr. Hyde conundrum when examining mechanisms of action. This is due to tumor heterogeneity and the diversity of the inflammatory cell phenotypes that infiltrate primary and metastatic lesions. Tumor infiltration by macrophages is generally associated with neoangiogenesis and negative outcomes, whereas dendritic cell (DC) infiltration is typically associated with a positive clinical outcome in association with their ability to present tumor antigens (Ags) and induce Ag-specific T cell responses. Myeloid-derived suppressor cells (MDSCs) also infiltrate tumors, inhibiting immune responses and facilitating tumor growth and metastasis. In contrast, T cell infiltration of tumors provides a positive prognostic surrogate, although subset analyses suggest that not all infiltrating T cells predict a positive outcome. In general, infiltration by CD8(+) T cells predicts a positive outcome, while CD4(+) cells predict a negative outcome. Therefore, the analysis of cellular phenotypes and potentially spatial distribution of infiltrating cells are critical for an accurate assessment of outcome. Similarly, cellular infiltration of metastatic foci is also a critical parameter for inducing therapeutic responses, as well as establishing tumor dormancy. Current strategies for cellular, gene, and molecular therapies are focused on the manipulation of infiltrating cellular populations. Within this review, we discuss the role of tumor infiltrating, myeloid-monocytic cells, and T lymphocytes, as well as their potential for tumor control, immunosuppression, and facilitation of metastasis.
Collapse
Affiliation(s)
- James E Talmadge
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, 987660 Nebraska Medical Center, Omaha, NE 68198-7660, USA.
| | | | | |
Collapse
|
39
|
Murugaiyan G, Martin S, Saha B. CD40-induced countercurrent conduits for tumor escape or elimination? Trends Immunol 2007; 28:467-73. [DOI: 10.1016/j.it.2007.08.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Revised: 08/08/2007] [Accepted: 08/08/2007] [Indexed: 12/14/2022]
|
40
|
Tourkova IL, Shurin GV, Wei S, Shurin MR. Small rho GTPases mediate tumor-induced inhibition of endocytic activity of dendritic cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:7787-93. [PMID: 17548616 DOI: 10.4049/jimmunol.178.12.7787] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The generation, maturation, and function of dendritic cells (DC) have been shown to be markedly compromised in the tumor microenvironment in animals and humans. However, the molecular mechanisms and intracellular pathways involved in the regulation of the DC system in cancer are not yet fully understood. Recently, we have reported on the role of the small Rho GTPase family members Cdc42, Rac1, and RhoA in regulating DC adherence, motility, and Ag presentation. To investigate involvement of small Rho GTPases in dysregulation of DC function by tumors, we next evaluated how Cdc42, Rac1, and RhoA regulated endocytic activity of DC in the tumor microenvironment. We revealed a decreased uptake of dextran 40 and polystyrene beads by DC generated in the presence of different tumor cell lines, including RM1 prostate, MC38 colon, 3LL lung, and B7E3 oral squamous cell carcinomas in vitro and by DC prepared from tumor-bearing mice ex vivo. Impaired endocytic activity of DC cocultured with tumor cells was associated with decreased levels of active Cdc42 and Rac1. Transduction of DC with the dominant negative Cdc42 and Rac1 genes also led to reduced phagocytosis and receptor-mediated endocytosis. Furthermore, transduction of DC with the constitutively active Cdc42 and Rac1 genes restored endocytic activity of DC that was inhibited by the tumors. Thus, our results suggest that tumor-induced dysregulation of endocytic activity of DC is mediated by reduced activity of several members of the small Rho GTPase family, which might serve as new targets for improving the efficacy of DC vaccines.
Collapse
Affiliation(s)
- Irina L Tourkova
- Department of Pathology, University of Pittsburgh Medical Center, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
41
|
Murugaiyan G, Martin S, Saha B. Levels of CD40 expression on dendritic cells dictate tumour growth or regression. Clin Exp Immunol 2007; 149:194-202. [PMID: 17488293 PMCID: PMC1942037 DOI: 10.1111/j.1365-2249.2007.03407.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Tumour regression requires activation of T cells. It has been shown that the interaction between T cell-expressed CD40-ligand (CD40-L) and antigen-presenting cell-expressed CD40 plays a crucial role in T cell activation. CD40-L- or CD40-deficient mice are susceptible to tumour growth. CD40-based therapies are also shown to control tumour growth significantly, suggesting that CD40-CD40-L interaction induces anti-tumour T cell responses and tumour regression. We demonstrate that the anti-tumour T cell response can be modulated reciprocally as a function of the levels of CD40 expression. At low expression levels, CD40 promotes tumour growth; at higher expression levels, CD40 induces tumour-regressing T cell response. Dendritic cells (DC) sorted onto major histocompatibility complex (MHC)-II expression are found to be similar in CD40 and CD80 expression. The MHC-II(hi)/CD40(hi) DC induce interleukin (IL)-12-dominated and T helper 1 (Th1)-type response, whereas MHC-II(lo)/CD40(lo) DC promote high IL-10 and Th2-type T cells. The T cells induced by these DC also differ in terms of regulatory T cell markers, lymphocyte activation gene-3 (LAG-3) and glucocorticoid-induced tumour necrosis factor (TNF) receptor family-related gene (GITR). Thus, we report for the first time that CD40-induced effector T cell response depends on CD40 expression levels in vivo.
Collapse
Affiliation(s)
- G Murugaiyan
- National Centre for Cell Science, Ganeshkhind, Pune, India
| | | | | |
Collapse
|
42
|
Shurin MR, Shurin GV, Chatta GS. Aging and the dendritic cell system: implications for cancer. Crit Rev Oncol Hematol 2007; 64:90-105. [PMID: 17446082 PMCID: PMC2084365 DOI: 10.1016/j.critrevonc.2007.03.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Revised: 03/14/2007] [Accepted: 03/15/2007] [Indexed: 10/23/2022] Open
Abstract
The immune system shows a decline in responsiveness to antigens both with aging, as well as in the presence of tumors. The malfunction of the immune system with age can be attributed to developmental and functional alterations in several cell populations. Previous studies have shown defects in humoral responses and abnormalities in T cell function in aged individuals, but have not distinguished between abnormalities in antigen presentation and intrinsic T cell or B cell defects in aged individuals. Dendritic cells (DC) play a pivotal role in regulating immune responses by presenting antigens to naïve T lymphocytes, modulating Th1/Th2/Th3/Treg balance, producing numerous regulatory cytokines and chemokines, and modifying survival of immune effectors. DC are receiving increased attention due to their involvement in the immunobiology of tolerance and autoimmunity, as well as their potential role as biological adjuvants in tumor vaccines. Recent advances in the molecular and cell biology of different DC populations allow for addressing the issue of DC and aging both in rodents and humans. Since DC play a crucial role in initiating and regulating immune responses, it is reasonable to hypothesize that they are directly involved in altered antitumor immunity in aging. However, the results of studies focusing on DC in the elderly are conflicting. The present review summarizes the available human and experimental animal data on quantitative and qualitative alterations of DC in aging and discusses the potential role of the DC system in the increased incidence of cancer in the elderly.
Collapse
Affiliation(s)
- Michael R Shurin
- University of Pittsburgh Cancer Institute, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| | | | | |
Collapse
|
43
|
Shurin MR, Shurin GV, Lokshin A, Yurkovetsky ZR, Gutkin DW, Chatta G, Zhong H, Han B, Ferris RL. Intratumoral cytokines/chemokines/growth factors and tumor infiltrating dendritic cells: friends or enemies? Cancer Metastasis Rev 2007; 25:333-56. [PMID: 17029028 DOI: 10.1007/s10555-006-9010-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The tumor microenvironment consists of a variable combination of tumor cells, stromal fibroblasts, endothelial cells and infiltrating leukocytes, such as macrophages, T lymphocytes, and dendritic cells. A variety of cytokines, chemokines and growth factors are produced in the local tumor environment by different cells accounting for a complex cell interaction and regulation of differentiation, activation, function and survival of multiple cell types. The interaction between cytokines, chemokines, growth factors and their receptors forms a comprehensive network at the tumor site, which is primary responsible for overall tumor progression and spreading or induction of antitumor immune responses and tumor rejection. Although the general thought is that dendritic cells are among the first cells migrating to the tumor site and recognizing tumor cells for the induction of specific antitumor immunity, the clinical relevance of dendritic cells at the site of the tumor remains a matter of debate regarding their role in the generation of successful antitumor immune responses in human cancers. While several lines of evidence suggest that intratumoral dendritic cells play an important role in antitumor immune responses, understanding the mechanisms of dendritic cell/tumor cell interaction and modulation of activity and function of different dendritic cell subtypes at the tumor site is incomplete. This review is limited to discussing the role of intratumoral cytokine network in the understanding immunobiology of tumor-associated dendritic cells, which seems to possess different regulatory functions at the tumor site.
Collapse
Affiliation(s)
- Michael R Shurin
- Department of Pathology, University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Murugaiyan G, Agrawal R, Mishra GC, Mitra D, Saha B. Differential CD40/CD40L Expression Results in Counteracting Antitumor Immune Responses. THE JOURNAL OF IMMUNOLOGY 2007; 178:2047-55. [PMID: 17277108 DOI: 10.4049/jimmunol.178.4.2047] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Establishment of host-protective memory T cells against tumors is the objective of an antitumor immunoprophylactic strategy such as reinforcing T cell costimulation via CD40-CD40L interaction. Previous CD40-targeted strategies assumed that T cell costimulation is an all-or-none phenomenon. It was unknown whether different levels of CD40L expression induce quantitatively and qualitatively different effector T cell responses. Using mice expressing different levels of CD40L, we demonstrated that the greater the T cell CD40L expression the less tumor growth occurred; the antitumor T cell response was host-protective. Lower levels of CD40L expression on T cells induced IL-10-mediated suppression of tumor-regressing effector CD8(+) T cells and higher productions of IL-4 and IL-10. Using mice expressing different levels of CD40 or by administering different doses of anti-CD40 Ab, similar observations were recorded implying that the induction of protumor or antitumor T cell responses was a function of the extent of CD40 cross-linking. IL-10 neutralization during priming with tumor Ags resulted in a stronger tumor-regressing effector T cell response. Using IL-10(-/-) DC for priming of mice expressing different levels of CD40L and subsequent transfer of the T cells from the primed mice to nu/nu mice, we demonstrated the protumor role of IL-10 in the induction of tumor-promoting T cells. Our results demonstrate that a dose-dependent cross-linking of a costimulatory molecule dictates the functional phenotype of the elicited effector T cell response. The T cell costimulation is a continuum of a function that induces not only graded T cell responses but also two counteracting responses at two extremes.
Collapse
|
45
|
Sevko AL, Barysik N, Perez L, Shurin MR, Gerein V. Differences in dendritic cell activation and distribution after intravenous, intraperitoneal, and subcutaneous injection of lymphoma cells in mice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 601:257-64. [PMID: 17713013 DOI: 10.1007/978-0-387-72005-0_27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Dendritic cells (DCs) are key antigen-presenting cells (APCs) for initiating immune responses. However, in recent years, several groups have shown the defective function of DCs in tumor-bearing mice and in cancer patients. Our aim was to study the effects of lymphoma on DC differentiation and maturation and to assess the input of the tumor microenvironment and intravasation of tumor cells on DC precursors. EL-4 lymphoma cells were administrated via different routes (intraperitoneal, subcutaneous, and intravenous) and DC phenotype was investigated. Bone marrow-derived DCs and APCs obtained from the spleen were examined by flow cytometry, and immunohistochemical analysis of lymphoma, lungs, livers, and spleens was also performed. Intravenous administration of lymphoma cells induced suppression of DC differentiation and maturation assessed as a significant decrease of the IAb, CD80, CD86, CD11b, and CD11c expression on DCs and IAb on splenic APCs. Upregulation of APC differentiation was observed in animals after subcutaneous and intraperitoneal administration of lymphoma cells determined as increased expression of CD40 and CD86 in spleen APCs. These data suggest that the development of antitumor immune response might differ in the host receiving tumor vaccines via different injection routes.
Collapse
Affiliation(s)
- Alexandra L Sevko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, Ukrainian Academy of Sciences, Kyiv, Ukraine
| | | | | | | | | |
Collapse
|
46
|
Newland A, Russ G, Krishnan R. Natural killer cells prime the responsiveness of autologous CD4+ T cells to CTLA4-Ig and interleukin-10 mediated inhibition in an allogeneic dendritic cell-mixed lymphocyte reaction. Immunology 2006; 118:216-23. [PMID: 16771856 PMCID: PMC1782277 DOI: 10.1111/j.1365-2567.2006.02359.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Cytotoxic T-lymphocyte antigen 4 immunoglobulin (CTLA4-Ig) and interleukin (IL)-10 are immunomodulatory molecules which target CD28 costimulation by acting either directly or indirectly on the CD80/86 receptors on dendritic cells (DCs). This study examined the effect of combined treatment with CTLA4-Ig and IL-10 on T-cell responsiveness in a dendritic cell-mixed lymphocyte reaction (DC-MLR). T cells derived from nylon wool enrichment (NWT cells) demonstrated 15% (P = 0.006) and 10% (P = 0.0015) inhibition of proliferation with suboptimal doses of IL-10 (5 ng/ml) and CTLA4-Ig (20 ng/ml), respectively. Combined treatment with both agents resulted in 38% inhibition (P = 0.004) of the MLR response compared with untreated controls. In contrast to NWT cells, which consisted of CD4+, CD8+ and CD56+ (NK) cells, purified CD4+ T cells were less responsive to immunomodulation by CTLA4-Ig and IL-10. Repletion of the CD4+ T cells with NK cells restored IL-10 and CTLA4-Ig mediated immunomodulation, suggesting a role for NK cells in the regulation of DC-T-cell interactions. The specific effect of NK cells on DC activation was demonstrated by CD80 up-regulation on DCs in the absence of T cells. However, in the absence of DCs, NK cells augmented the proliferation of autologous CD4+ T cells stimulated by anti-CD3 monoclonal antibody (mAb), which was blocked by CTLA4-Ig. It is proposed that, in the MLR, immunomodulation by suboptimal CTLA4-Ig and IL-10 is influenced by cellular interactions of NK cells with DCs and T cells involving DC lysis and costimulation. Thus, NK cells prime both DCs and T cells to low doses of CTLA4-Ig and IL-10 during alloimmune responses, providing evidence for the potential interaction between innate and adaptive immunity.
Collapse
Affiliation(s)
- Ashley Newland
- Transplantation Immunology Laboratory, Basil Hetzel Institute, The Queen Elizabeth HospitalWoodville
- Department of Medicine, University of AdelaideSouth Australia, Australia
| | - Graeme Russ
- Transplantation Immunology Laboratory, Basil Hetzel Institute, The Queen Elizabeth HospitalWoodville
- Department of Medicine, University of AdelaideSouth Australia, Australia
| | - Ravi Krishnan
- Transplantation Immunology Laboratory, Basil Hetzel Institute, The Queen Elizabeth HospitalWoodville
- Department of Medicine, University of AdelaideSouth Australia, Australia
| |
Collapse
|
47
|
Berhanu A, Huang J, Alber SM, Watkins SC, Storkus WJ. Combinational FLt3 ligand and granulocyte macrophage colony-stimulating factor treatment promotes enhanced tumor infiltration by dendritic cells and antitumor CD8(+) T-cell cross-priming but is ineffective as a therapy. Cancer Res 2006; 66:4895-903. [PMID: 16651446 DOI: 10.1158/0008-5472.can-05-2384] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Dendritic cells play significant roles in the development and maintenance of antitumor immune responses. Therapeutic recruitment of dendritic cells into the tumor microenvironment has the potential to result in enhanced antitumor T-cell cross-priming against a broad array of naturally processed and presented tumor-associated antigens. We have observed that the treatment of BALB/c mice bearing syngeneic CMS4 sarcomas with the combination of recombinant Flt3 ligand and recombinant granulocyte-macrophage colony-stimulating factor (GM-CSF) for five sequential days is sufficient to optimize the number of tumor-infiltrating dendritic cells (TIDC). However, despite the significant increase in the number of TIDCs, the therapeutic benefit of Flt3 ligand and GM-CSF treatment is minimal. Therapy-associated TIDCs do not exhibit a "suppressed" or "suppressor" phenotype in vitro, and their enhanced numbers in cytokine-treated mice were associated with increased levels of peripheral antitumor CD8(+) T effector cells and with an augmented population of CD8(+) tumor-infiltrating lymphocytes (TIL). These data suggest that Flt3 ligand + GM-CSF therapy of murine tumors fails at a mechanistic point that is downstream of specific T-cell priming by therapy-induced TIDCs and the recruitment of these T cells into the tumor microenvironment. Based on the enhanced infiltration of tumors by CD4(+)CD25(+) TIL in Flt3 ligand + GM-CSF-treated mice, this could reflect the dominant influence of regulatory T cells in situ.
Collapse
Affiliation(s)
- Aklile Berhanu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | |
Collapse
|
48
|
Errington F, Jones J, Merrick A, Bateman A, Harrington K, Gough M, O'Donnell D, Selby P, Vile R, Melcher A. Fusogenic membrane glycoprotein-mediated tumour cell fusion activates human dendritic cells for enhanced IL-12 production and T-cell priming. Gene Ther 2006; 13:138-49. [PMID: 16136162 DOI: 10.1038/sj.gt.3302609] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Fusogenic membrane glycoproteins (FMG) are a family of viral genes that, when expressed in tumour cells, trigger extensive cell to cell fusion and subsequent cell death. Gene therapy approaches using FMG are also potentially immunogenic, since syncitia generated ex vivo can be therapeutic as antitumour vaccines in murine models. This study has addressed the mechanisms responsible for the immunogenicity of FMG-mediated cell death, and its applicability to human immune priming. We show that fusion of human Mel888 melanoma cells following transfection with FMG can reverse the suppressive effects of Mel888 on dendritic cells (DC) phenotype, and potentiate IL-12 production by DC on activation in a cell contact-dependent manner. DC loaded with fusing, but not intact, tumour cells primed a naive, tumour-specific cytotoxic T-cell response, which was MHC class I-restricted and associated with production of high levels of IFNgamma and, later, IL-5. Fusing cells were an effective source of antigen for DC cross-priming and presentation of the melanoma-specific antigen gp100 to a specific T-cell clone. These data show, in a human system, that FMG represent an immunogenic, as well as cytotoxic, gene therapy for cancer, reversing the inhibitory effects of tumour cells on DC to potentiate IL-12 production and naive T-cell priming.
Collapse
Affiliation(s)
- F Errington
- Cancer Research UK Clinical Center, St James's University Hospital, Leeds
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Metharom P, Velten FW, Goerdt S. Highly phagocytic, CD4hi, CD14hi and CD16hi antigen-presenting cells modulated by tumour-conditioned media retain the capacity to mature and induce TH1 T-cell proliferation. Mol Immunol 2006; 43:2070-82. [PMID: 16455138 DOI: 10.1016/j.molimm.2005.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2005] [Revised: 12/14/2005] [Accepted: 12/20/2005] [Indexed: 11/16/2022]
Abstract
The tumour microenvironment down-modulates antigen-presentation by dendritic cells (DC), presumably due to inhibition of DC maturation. Here, we sought to examine (1) whether monocyte-derived cells cultivated with tumour-conditioned media under conditions that are conducive to DC generation (APCTCM) resemble immature DC (iDC), IL-10-induced regulatory DC (DCIL10) or display other distinctive features; (2) whether APCTCM are convertible to immunostimulatory DC (DCims) upon proper activation and (3) whether APCTCM and activated APCTCM are functionally defective. Four tumour cell lines expressing different cytokines were used to mimic different tumour microenvironments. As compared to iDC, DCims or DCIL10, APCTCM exhibited the highest levels of expression for CD14, CD16 and CD4. These markers and a high phagocytic capacity were unique features of these cells. When APCTCM were activated by a maturation cocktail, CD83, CD86, HLA-DR and CD25 were up-regulated to levels considerably higher than in DCIL10 and comparable to DCims while CD14, CD16, CD4 and dextran-uptake were down-modulated. Activated APCTCM induced 50-60% of the proliferative response of DCims in the allogeneic T-cell proliferation assay while DCIL10 mounted a 20-30% response (iDC elicited approximately 10%). Activated APCTCM induced secretion of almost equal amounts of IFN-gamma, TNF-alpha and IL-2 as DCims indicating induction of Th1 differentiation. When mature DCims were exposed to TCM, their immunostimulatory function was not significantly altered. However, when TCM were added to the co-cultures of DCims and CD4 T-cells the proliferative outcome was dependent on the TCM. In summary, APCTCM display special features but can mature into DCims-like cells.
Collapse
Affiliation(s)
- P Metharom
- Department of Dermatology, University Medical Centre Mannheim, Ruprecht Karls University of Heidelberg, Mannheim 68167, Germany.
| | | | | |
Collapse
|
50
|
Gerlini G, Romagnoli P, Pimpinelli N. Skin cancer and immunosuppression. Crit Rev Oncol Hematol 2005; 56:127-36. [PMID: 15978830 DOI: 10.1016/j.critrevonc.2004.11.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2004] [Revised: 10/30/2004] [Accepted: 11/04/2004] [Indexed: 11/24/2022] Open
Abstract
All immunosuppressive treatments, either pharmacological or physical, have the potential to impair the skin immune system network of cells and cytokines, thus leading to an increased incidence of skin cancer. Since skin cancer in transplant recipients may show uncommon clinical features and have an unusually aggressive course, transplant patients should be strictly followed up by experienced dermatologists in order to diagnose and treat properly any skin cancer in an early phase. Importantly, due to the fact that sun exposure increases immunosuppression in the skin, patients should be clearly informed about the additional risk of sun exposure and the preventive measures to be taken.
Collapse
Affiliation(s)
- Gianni Gerlini
- Department of Dermatological Sciences, University of Florence Medical School, Via degli Alfani, 37-50121 Florence, Italy
| | | | | |
Collapse
|