1
|
Naji NS, Sathish M, Karantanos T. Inflammation and Related Signaling Pathways in Acute Myeloid Leukemia. Cancers (Basel) 2024; 16:3974. [PMID: 39682161 DOI: 10.3390/cancers16233974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic malignancy, and inflammatory signaling is involved in its pathogenesis. Cytokines exert a robust effect on the progression of AML and affect survival outcomes. The dysregulation in the cytokine network may foster a pro-tumorigenic microenvironment, increasing leukemic cell proliferation, decreasing survival and driving drug resistance. The dominance of pro-inflammatory mediators such as IL-11β, TNF-α and IL-6 over anti-inflammatory mediators such as TGF-β and IL-10 has been implicated in tumor progression. Additionally, inflammatory cytokines have favored certain populations of hematopoietic stem and progenitor cells with mutated clonal hematopoiesis genes. This article summarizes current knowledge about inflammatory cytokines and signaling pathways in AML, their modes of action and the implications for immune tolerance and clonal hematopoiesis, with the aim of finding potential therapeutic interventions to improve clinical outcomes in AML patients.
Collapse
Affiliation(s)
- Nour Sabiha Naji
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Mrudula Sathish
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Theodoros Karantanos
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
2
|
Wang D, Dou L, Sui L, Xue Y, Xu S. Natural killer cells in cancer immunotherapy. MedComm (Beijing) 2024; 5:e626. [PMID: 38882209 PMCID: PMC11179524 DOI: 10.1002/mco2.626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/18/2024] Open
Abstract
Natural killer (NK) cells, as innate lymphocytes, possess cytotoxic capabilities and engage target cells through a repertoire of activating and inhibitory receptors. Particularly, natural killer group 2, member D (NKG2D) receptor on NK cells recognizes stress-induced ligands-the MHC class I chain-related molecules A and B (MICA/B) presented on tumor cells and is key to trigger the cytolytic response of NK cells. However, tumors have developed sophisticated strategies to evade NK cell surveillance, which lead to failure of tumor immunotherapy. In this paper, we summarized these immune escaping strategies, including the downregulation of ligands for activating receptors, upregulation of ligands for inhibitory receptors, secretion of immunosuppressive compounds, and the development of apoptosis resistance. Then, we focus on recent advancements in NK cell immune therapies, which include engaging activating NK cell receptors, upregulating NKG2D ligand MICA/B expression, blocking inhibitory NK cell receptors, adoptive NK cell therapy, chimeric antigen receptor (CAR)-engineered NK cells (CAR-NK), and NKG2D CAR-T cells, especially several vaccines targeting MICA/B. This review will inspire the research in NK cell biology in tumor and provide significant hope for improving cancer treatment outcomes by harnessing the potent cytotoxic activity of NK cells.
Collapse
Affiliation(s)
- DanRu Wang
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - LingYun Dou
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - LiHao Sui
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - Yiquan Xue
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - Sheng Xu
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
- Shanghai Institute of Stem Cell Research and Clinical Translation Dongfang Hospital Shanghai China
| |
Collapse
|
3
|
Glucomannan as a Dietary Supplement for Treatment of Breast Cancer in a Mouse Model. Vaccines (Basel) 2022; 10:vaccines10101746. [PMID: 36298611 PMCID: PMC9608331 DOI: 10.3390/vaccines10101746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Konjac glucomannan (KGM) is a water-soluble polysaccharide derived from the Amorphophallus’s tuber and, as herbal medicine has shown, can suppress tumor growth or improve health. However, there has been no investigation into the effects of KGM on breast tumor-bearing mice. Therefore, in two cohort experiments, we assessed the effect of glucomannan at daily doses of 2 and 4 mg for 28 days as a dietary supplement and also glucomannan in combination with tumor lysate vaccine as an adjuvant. Tumor volume was monitored twice weekly. In addition, TNF-α cytokines and granzyme B (Gr–B) release were measured with ELISA kits, and IL-2, IL-4, IL-17, and IFN-γ were used as an index for cytotoxic T lymphocyte activity. Moreover, TGF-β and Foxp3 gene expression were assessed in a real-time PCR test. The results show that glucomannan as a dietary supplement increased the IFN-γ cytokine and Th1 responses to suppress tumor growth. Glucomannan as a dietary supplement at the 4 mg dose increased the IL-4 cytokine response compared to control groups. In addition, cell lysate immunization with 2 or 4 mg of glucomannan suppressed tumor growth. As an adjuvant, glucomannan at both doses showed 41.53% and 52.10% tumor suppression compared with the PBS group. Furthermore, the administration of glucomannan as a dietary supplement or adjuvant reduced regulatory T cell response through decreasing TGF-β and Foxp3 gene expression in the tumor microenvironment. In conclusion, glucomannan as a dietary supplement or adjuvant enhanced the immune responses of tumor-bearing mice and decreased immune response suppression in the tumor milieu, making it a potentially excellent therapeutic agent for lowering breast tumor growth.
Collapse
|
4
|
Ogasawara M, Yamasaki-Yashiki S, Hamada M, Yamaguchi-Miyamoto T, Kawasuji T, Honda H, Yanagibashi T, Ikutani M, Watanabe Y, Fujimoto R, Matsunaga T, Nakajima N, Nagai Y, Takatsu K. Betulin Attenuates TGF-β1- and PGE 2-Mediated Inhibition of NK Cell Activity to Suppress Tumor Progression and Metastasis in Mice. Biol Pharm Bull 2022; 45:339-353. [PMID: 35228400 DOI: 10.1248/bpb.b21-00921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Transforming growth factor (TGF)-β1 and prostaglandin E2 (PGE2) are humoral factors critically involved in the induction of immunosuppression in the microenvironment of various types of tumors, including melanoma. In this study, we identified a natural compound that attenuated TGF-β1- and PGE2-induced immunosuppression and examined its effect on B16 melanoma growth in mice. By screening 502 natural compounds for attenuating activity against TGF-β1- or PGE2-induced suppression of cytolysis in poly(I:C)-stimulated murine splenocytes, we found that betulin was the most potent compound. Betulin also reduced TGF-β1- and PGE2-induced downregulation of perforin and granzyme B mRNA expression and cell surface expression of NKG2D and CD69 in natural killer (NK) cells. Cell depletion and coculture experiments showed that NK cells, dendritic cells, B cells, and T cells were necessary for the attenuating effects of betulin. Structure-activity relationship analysis revealed that two hydroxyl groups at positions C3 and C28 of betulin, their cis-configuration, and methyl group at C30 played crucial roles in its attenuating activity. In a subcutaneous implantation model of B16 melanoma in mice, intratumor administration of betulin and LY2157299, a TGF-β1 type I receptor kinase inhibitor, significantly retarded the growth of B16 melanoma. Notably, betulin increased significantly the number of CD69 positive NK cells in tumor sites at early stages of post-tumor cell injection. Our data suggest that betulin inhibits the growth of B16 melanoma by enhancing NK cell activity through attenuating the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Masaru Ogasawara
- Toyama Prefectural Institute for Pharmaceutical Research.,Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama
| | | | - Masahiro Hamada
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University
| | | | - Toru Kawasuji
- Toyama Prefectural Institute for Pharmaceutical Research
| | - Hiroe Honda
- Toyama Prefectural Institute for Pharmaceutical Research.,Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama
| | - Tsutomu Yanagibashi
- Toyama Prefectural Institute for Pharmaceutical Research.,Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama
| | - Masashi Ikutani
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama.,Graduate School of Integrated Sciences for Life, Hiroshima University.,Department of Immune Regulation, Research Institute, National Center for Global Health and Medicine
| | - Yasuharu Watanabe
- Toyama Prefectural Institute for Pharmaceutical Research.,Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama
| | - Ryota Fujimoto
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University
| | | | - Noriyuki Nakajima
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University
| | - Yoshinori Nagai
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama.,Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University
| | - Kiyoshi Takatsu
- Toyama Prefectural Institute for Pharmaceutical Research.,Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama
| |
Collapse
|
5
|
Qiu Q, Li C, Yan X, Zhang H, Luo X, Gao X, Liu X, Song Y, Deng Y. Photodynamic/ photothermal therapy enhances neutrophil-mediated ibrutinib tumor delivery for potent tumor immunotherapy: More than one plus one? Biomaterials 2021; 269:120652. [PMID: 33450581 DOI: 10.1016/j.biomaterials.2021.120652] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/11/2020] [Accepted: 12/31/2020] [Indexed: 12/14/2022]
Abstract
Neutrophil-mediated drug-delivery systems have gained widespread attention owing to their superior efficacy in cancer therapy. Neutrophils, the most abundant white cells in peripheral blood, are known to migrate to inflamed tumors. Here, we elaborate on a novel strategy to enhance tumor infiltration of neutrophils by photodynamic/photothermal therapy (PDT/PTT) to deliver ibrutinib (IBR) nanocomplexes for cancer immunotherapy. DiR-loading liposomes (DiR-lipos) were administered to induce acute inflammation, and sialic acid (SA) derivative-coated IBR-loading nanocomplexes (SA-2@NCs) were fabricated for targeting activated peripheral blood neutrophils (PBNs). This in vitro and in vivo attempt, therefore, proved the hypothesis that inducing acute inflammation via PDT/PTT could facilitate the migration of PBNs, which could deliver SA-2@NCs into the tumor. The enhanced tumor delivery of SA-2@NCs was accompanied by enhanced antitumor T-cell immune responses in a mouse orthotopic breast cancer model. Our findings indicate that the combination of IBR-mediated immunotherapy with DiR-mediated PDT/PTT bring together two leading novel strategies, taking advantage of their synergistic mechanisms of action for a potent anti-tumor efficacy for breast cancer therapy.
Collapse
Affiliation(s)
- Qiujun Qiu
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| | - Cong Li
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| | - Xinyang Yan
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| | - Hongxia Zhang
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| | - Xiang Luo
- College of Chemistry and Chemical Engineering, Shaoxing University, No. 508 Huancheng West Road, Shaoxing, Zhejiang Province, 312000, PR China.
| | - Xin Gao
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| |
Collapse
|
6
|
Zhu L, Yang X, Zhong D, Xie S, Shi W, Li Y, Hou X, HuaYao, Zhou H, Zhao M, Ding Z, Zhao X, Mo F, Yin S, Liu A, Lu X. Single-Domain Antibody-Based TCR-Like CAR-T: A Potential Cancer Therapy. J Immunol Res 2020; 2020:2454907. [PMID: 32964055 PMCID: PMC7492946 DOI: 10.1155/2020/2454907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/30/2020] [Accepted: 08/14/2020] [Indexed: 12/17/2022] Open
Abstract
Retargeting the antigen-binding specificity of T cells to intracellular antigens that are degraded and presented on the tumor surface by engineering chimeric antigen receptor (CAR), also named TCR-like antibody CAR-T, remains limited. With the exception of the commercialized CD19 CAR-T for hematological malignancies and other CAR-T therapies aiming mostly at extracellular antigens achieving great success, the rareness and scarcity of TCR-like CAR-T therapies might be due to their current status and limitations. This review provides the probable optimized initiatives for improving TCR-like CAR-T reprogramming and discusses single-domain antibodies administered as an alternative to conventional scFvs and secreted by CAR-T cells, which might be of great value to the development of CAR-T immunotherapies for intracellular antigens.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- Epitopes, T-Lymphocyte/immunology
- Genetic Engineering
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Neoplasms/immunology
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Single-Chain Antibodies/immunology
- Single-Domain Antibodies/genetics
- Single-Domain Antibodies/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Treatment Outcome
Collapse
Affiliation(s)
- Lichen Zhu
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiaomei Yang
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Dani Zhong
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- Department of Chemotherapy, Affiliated Cancer Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Shenxia Xie
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Wei Shi
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yangzi Li
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiaoqiong Hou
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - HuaYao
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Huihui Zhou
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Minlong Zhao
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Ziqiang Ding
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xinyue Zhao
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Fengzhen Mo
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Shihua Yin
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Aiqun Liu
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiaoling Lu
- Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi 530021, China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi 530021, China
| |
Collapse
|
7
|
Li JY, Sun F, Zhou HF, Yang J, Huang C, Fan H. A Systematic Review Exploring the Anticancer Activity and Mechanisms of Glucomannan. Front Pharmacol 2019; 10:930. [PMID: 31507423 PMCID: PMC6715771 DOI: 10.3389/fphar.2019.00930] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/22/2019] [Indexed: 12/24/2022] Open
Abstract
Glucomannan, long recognized as the active ingredient of the traditional Chinese medicinal herb Konjac glucomannan, is a naturally occurring polysaccharide existing in certain plant species and fungi. Due to its special property to also serve as a dietary supplement, glucomannan has been widely applied in clinic to lower body weight and circulation cholesterol level and to treat constipation, diabetes, and arterial sclerosis. Besides the regulatory role engaged with gastroenterological and metabolic syndrome, recently, its therapeutic effect and the underlying mechanisms in treating cancerous diseases have been appreciated by mounting researches. The present review aims to emphasize the multifaceted aspects of how glucomannan exerts its anti-tumor function.
Collapse
Affiliation(s)
- Jun-Yi Li
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Sun
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hai-Feng Zhou
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Yang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Qiu Q, Li C, Song Y, Shi T, Luo X, Zhang H, Hu L, Yan X, Zheng H, Liu M, Liu M, Liu M, Yang S, Liu X, Chen G, Deng Y. Targeted delivery of ibrutinib to tumor-associated macrophages by sialic acid-stearic acid conjugate modified nanocomplexes for cancer immunotherapy. Acta Biomater 2019; 92:184-195. [PMID: 31108259 DOI: 10.1016/j.actbio.2019.05.030] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/02/2019] [Accepted: 05/10/2019] [Indexed: 12/12/2022]
Abstract
Ibrutinib (IBR), an irreversible Bruton's tyrosine kinase (BTK) inhibitor, is expected to be a potent therapeutic modality, given that BTK is overexpressed in tumor-associated macrophages (TAMs) and participates in promoting tumor progression, angiogenesis, and immunosuppression. However, rapid clearance in vivo and low tumor accumulation have rendered effective uptake of IBR by TAMs challenge. Herein, we designed and synthesized a sialic acid (SA)-stearic acid conjugate modified on the surface of nanocomplexes to encapsulate IBR (SA/IBR/EPG) for targeted immunotherapy. Amphiphilic egg phosphatidylglycerol (EPG) structure and strong IBR-EPG interactions render these nanocomplexes high IBR loading capacity, prolonged blood circulation, and optimal particle sizes (∼30 nm), which can effectively deliver IBR to the tumor, followed by subsequent internalization of IBR by TAMs through SA-mediated active targeting. In vitro and in vivo tests showed that the prepared SA/IBR/EPG nanocomplexes could preferentially accumulate in TAMs and exert potent antitumor activity. Immunofluorescence staining analysis further confirmed that SA/IBR/EPG remarkably inhibited angiogenesis and tumorigenic cytokines released by TAM and eventually suppressed tumor progression, without eliciting any unwanted effect. Thus, SA-decorated IBR nanocomplexes present a promising strategy for cancer immunotherapy. STATEMENT OF SIGNIFICANCE: Ibrutinib (IBR), an irreversible Bruton's tyrosine kinase (BTK) inhibitor, is expected to be a potent therapeutic modality, given that BTK is overexpressed in tumor-associated macrophages (TAMs) and participates in promoting tumor progression, angiogenesis, and immunosuppression. However, rapid clearance in vivo and low tumor accumulation have rendered effective uptake of IBR by TAMs challenge. Herein, we designed and synthesized a sialic acid (SA)-stearic acid conjugate modified on the surface of nanocomplexes to encapsulate IBR (SA/IBR/EPG) for targeted delivery of IBR to TAMs. The developed SA/IBR/EPG nanocomplexes exhibited high efficiency in targeting TAMs and inhibiting BTK activation, consequently inhibiting Th2 tumorigenic cytokine release, reducing angiogenesis, and suppressing tumor growth. These results implied that the SA/IBR/EPG nanocomplex could be a promising strategy for TAM-targeting immunotherapy with minimal systemic side effects.
Collapse
|
9
|
Wu N, Sun H, Sun Q, Cui M, Jiang R, Cong X. Associations Between IL-10 Polymorphisms and Susceptibility to Melanoma, Basal Cell Carcinoma, and Squamous Cell Carcinoma: A Meta-Analysis. Genet Test Mol Biomarkers 2018; 22:693-701. [PMID: 30427744 DOI: 10.1089/gtmb.2018.0172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND According to relevant reports, interleukin-10 (IL-10), as a multifunctional anti-inflammatory cytokine, has a critical influence in cancer development. A meta-analysis was carried out regarding the relationships among the -592 A/C, -1082 G/A, and -819 T/C polymorphisms as well as the susceptibility to skin squamous cell carcinoma (sSCC), melanoma, and basal cell carcinoma (BCC). MATERIALS AND METHODS A meta-analysis was carried out on the inter-relationships among the -592 A/C, IL-10-1082 G/A, and -819 T/C polymorphisms as well as the susceptibility to sSCC, melanoma, and BCC. RESULTS In this analysis, a total of 11 researches, involving 2149 controls and 2128 cases, were included. No association was found between skin cancer risk and the -592A/C or IL-10-1082G/A polymorphisms in any of the analyses. However, a moderately decreased skin cancer risk was found in the -819 TC versus CC model (odds ratio [OR] = 0.81 and 95% confidence interval [CI] = 0.67-0.99, p = 0.04). From the subgroup analysis, a decreased risk was found between the studies of nonmelanoma skin cancers and IL-10-819T/C in the dominant model (OR = 0.60, 95% CI = 0.43-0.85, p = 0.004 for TT+TC vs. CC). Egger's and Begg's tests demonstrated that there was no significant publication bias. CONCLUSION This meta-analysis showed that the -592A/C and 1082G/A IL-10 polymorphisms might not be risk factors for melanoma or for BCC and sSCC patients, but we obtained a correlation between skin cancer risk and the IL-10 -819T/C polymorphism.
Collapse
Affiliation(s)
- Nan Wu
- 1 Department of Dermatology, China-Japan Union Hospital of Jilin University , Changchun, Jilin, P.R. China
| | - Hongyan Sun
- 2 Tissue Bank, China-Japan Union Hospital of Jilin University , Changchun, Jilin, P.R. China
| | - Qian Sun
- 1 Department of Dermatology, China-Japan Union Hospital of Jilin University , Changchun, Jilin, P.R. China
| | - Mingchao Cui
- 3 Department of Orthopedics, China-Japan Union Hospital of Jilin University , Changchun, Jilin, P.R. China
| | - Rihua Jiang
- 1 Department of Dermatology, China-Japan Union Hospital of Jilin University , Changchun, Jilin, P.R. China
| | - Xianling Cong
- 1 Department of Dermatology, China-Japan Union Hospital of Jilin University , Changchun, Jilin, P.R. China
| |
Collapse
|
10
|
Zhao H, Yang J, Yu Z, Shen H, Huang X, Zhang M, Long T, Cailing A, Wang W. Synthetic analysis of associations between IL-10 polymorphisms and skin cancer risk. Oncotarget 2018; 9:6728-6736. [PMID: 29467923 PMCID: PMC5805509 DOI: 10.18632/oncotarget.23385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/04/2017] [Indexed: 12/01/2022] Open
Abstract
The current study was designed to quantitatively summarize the evidence for the strength of the associations between common IL-10 functional polymorphisms and skin cancer risk. Relevant publications concerning the associations between common IL-10 functional polymorphisms(−1082G>A, −819C>T and −592C>A) and skin cancer were retrieved by a comprehensive electronic literature search in PubMed, Web of Science, EBSCO, Embase, China National Knowledge Infrastructure, Wanfang, Chinese Biomedical Database (CBM). The odds ratio (OR) and 95% confidence interval (CI) were utilized to assess the strength of the relationship. A total of 26 studies including 4090 cases and 4133 controls (−1082G>A, 10 studies with 1809 cases and 1830 controls; −819C>T, 7 studies with 862 cases and 957 controls; −592C>A, 9 studies with 1419 cases and 1346 controls) were enrolled in the meta-analysis. Overall, the results revealed a borderline decreased risk of skin cancer in heterozygote model (OR = 0.82, 95CI = 0.67–1.00, p = 0.05). The subgroup analysis also presented similar association for non-melanoma skin cancer in heterozygote model (OR = 0.67, 95CI = 0.50–0.91, p = 0.01). Moreover, the further analysis based on the histological type of non-melanoma skin cancer indicated a significantly decreased risk of BCC in allele model (OR = 0.67, 95% CI = 0.50–0.91, p = 0.02) and dominant model (OR = 0.68, 95% CI = 0.48–0.98, p = 0.04). However, neither overall analysis nor subgroup analysis based on cancer subtype revealed a significant association of −1082G>A or −592C>A polymorphisms with skin cancer. The present study suggested a potential association between IL-10 −819C>T polymorphism and decreased risk of skin cancer, but a lack of association for −1082G>A and −592C>A polymorphisms. Further invalidation is urgently needed.
Collapse
Affiliation(s)
- Hongbo Zhao
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Jiaoli Yang
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Zhenzhen Yu
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Hui Shen
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Xinlin Huang
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Mi Zhang
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Teng Long
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - A Cailing
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Wenhui Wang
- Department of Traditional Chinese Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| |
Collapse
|
11
|
Pang L, Han S, Jiao Y, Jiang S, He X, Li P. Bu Fei Decoction attenuates the tumor associated macrophage stimulated proliferation, migration, invasion and immunosuppression of non-small cell lung cancer, partially via IL-10 and PD-L1 regulation. Int J Oncol 2017; 51:25-38. [PMID: 28534943 PMCID: PMC5467788 DOI: 10.3892/ijo.2017.4014] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/03/2017] [Indexed: 12/21/2022] Open
Abstract
Macrophages play a pivotal role in tumor microenvironment. Bu-Fei Decoction (BFD) is a classical formula of traditional Chinese medicine (TCM) to alleviate lung cancer related symptoms, whether it has antitumor effect or could influence cancer microenvironment deserves further study. The aim of the present study was to examine the antitumor effect of BFD on non-small cell lung cancer (NSCLC), and to investigate the underlying mechanisms through tumor associated macrophages (TAMs). M2-polarized TAMs were induced by Phorbol 12-myristate 13-acetate (PMA) and interleukin 4 (IL-4). The antitumor activity of BFD in vitro was investigated in A549 and H1975 cells using MTT assay. The in vivo anticancer effect of BFD was evaluated in athymic nude mouse xenograft model. The invasive and migration properties of NSCLC cells were measured using Transwell. The protein expression was assessed using western blotting, ELISA and immunohistochemistry. The gene expression was examined using RT-PCR. TAMs was successfully established. Conditioned medium from TAMs increased cell proliferation, migration and invasion in NSCLC cells (p<0.05). BFD showed dose-dependent inhibitory effect on cell proliferation, migration and invasion abilities induced by TAMs. TAMs and rhIL-10 promoted the mRNA and protein expression of PD-L1 in NSCLC cells (p<0.01). Anti-IL-10 antibodies inhibited the elevated PD-L1 expression induced by TAMs. In vitro, the expression of PD-L1 and IL-10 was inhibited by BFD dose-dependently. In vivo, BFD suppressed A549 and H1975 tumor growth and decreased the expression of IL-10, PD-L1 and CD206. The results showed that TAMs play an important role in tumor progression of NSCLC, which was associated with tumor proliferation, migration, invasion and immunosuppression. Moreover, the antitumor mechanism of BFD is related to interruption of the link between TAMs and cancer cells by inhibiting the expression of IL-10 and PD-L1 in vitro and in vivo. Our results demonstrated BFD's potential as a novel treatment for NSCLC.
Collapse
Affiliation(s)
- Lina Pang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Shuyan Han
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Yanna Jiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Shantong Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Xiran He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Pingping Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| |
Collapse
|
12
|
Osorio F, Fuentes C, López MN, Salazar-Onfray F, González FE. Role of Dendritic Cells in the Induction of Lymphocyte Tolerance. Front Immunol 2015; 6:535. [PMID: 26539197 PMCID: PMC4611163 DOI: 10.3389/fimmu.2015.00535] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/02/2015] [Indexed: 01/07/2023] Open
Abstract
The ability of dendritic cells (DCs) to trigger tolerance or immunity is dictated by the context in which an antigen is encountered. A large body of evidence indicates that antigen presentation by steady-state DCs induces peripheral tolerance through mechanisms such as the secretion of soluble factors, the clonal deletion of autoreactive T cells, and feedback control of regulatory T cells. Moreover, recent understandings on the function of DC lineages and the advent of murine models of DC depletion have highlighted the contribution of DCs to lymphocyte tolerance. Importantly, these findings are now being applied to human research in the contexts of autoimmune diseases, allergies, and transplant rejection. Indeed, DC-based immunotherapy research has made important progress in the area of human health, particularly in regards to cancer. A better understanding of several DC-related aspects including the features of DC lineages, milieu composition, specific expression of surface molecules, the control of signaling responses, and the identification of competent stimuli able to trigger and sustain a tolerogenic outcome will contribute to the success of DC-based immunotherapy in the area of lymphocyte tolerance. This review will discuss the latest advances in the biology of DC subtypes related to the induction of regulatory T cells, in addition to presenting current ex vivo protocols for tolerogenic DC production. Particular attention will be given to the molecules and signals relevant for achieving an adequate tolerogenic response for the treatment of human pathologies.
Collapse
Affiliation(s)
- Fabiola Osorio
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile , Santiago , Chile
| | - Camila Fuentes
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile , Santiago , Chile
| | - Mercedes N López
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile , Santiago , Chile ; Cell Therapy Laboratory, Blood Bank Service, University of Chile Clinical Hospital , Santiago , Chile
| | - Flavio Salazar-Onfray
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile , Santiago , Chile
| | - Fermín E González
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Laboratory of Experimental Immunology and Cancer, Faculty of Dentistry, University of Chile , Santiago , Chile
| |
Collapse
|
13
|
Karimi S, Chattopadhyay S, Chakraborty NG. Manipulation of regulatory T cells and antigen-specific cytotoxic T lymphocyte-based tumour immunotherapy. Immunology 2015; 144:186-96. [PMID: 25243729 DOI: 10.1111/imm.12387] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 09/12/2014] [Accepted: 09/16/2014] [Indexed: 12/16/2022] Open
Abstract
The most potent killing machinery in our immune system is the cytotoxic T lymphocyte (CTL). Since the possibility for self-destruction by these cells is high, many regulatory activities exist to prevent autoimmune destruction by these cells. A tumour (cancer) grows from the cells of the body and is tolerated by the body's immune system. Yet, it has been possible to generate tumour-associated antigen (TAA) -specific CTL that are also self-antigen specific in vivo, to achieve a degree of therapeutic efficacy. Tumour-associated antigen-specific T-cell tolerance through pathways of self-tolerance generation represents a significant challenge to successful immunotherapy. CD4(+) CD25(+) FoxP3(+) T cells, referred to as T regulatory (Treg) cells, are selected in the thymus as controllers of the anti-self repertoire. These cells are referred to as natural T regulatory (nTreg) cells. According to the new consensus (Nature Immunology 2013; 14:307-308) these cells are to be termed as (tTreg). There is another class of CD4(+) Treg cells also involved in regulatory function in the periphery, also phenotypically CD4(+) CD25(±) , classified as induced Treg (iTreg) cells. These cells are to be termed as peripherally induced Treg (pTreg) cells. In vitro-induced Treg cells with suppressor function should be termed as iTreg. These different Treg cells differ in their requirements for activation and in their mode of action. The current challenges are to determine the degree of specificity of these Treg cells in recognizing the same TAA as the CTL population and to circumvent their regulatory constraints so as to achieve robust CTL responses against cancer.
Collapse
Affiliation(s)
- Shirin Karimi
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | | | | |
Collapse
|
14
|
Kazemi T, Younesi V, Jadidi-Niaragh F, Yousefi M. Immunotherapeutic approaches for cancer therapy: An updated review. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:769-79. [PMID: 25801036 DOI: 10.3109/21691401.2015.1019669] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In spite of specific immune effector mechanisms raised against tumor cells, there are mechanisms employed by the tumor cells to keep them away from immune recognition and elimination; some of these mechanisms have been identified, while others are still poorly understood. Manipulation or augmentation of specific antitumor immune responses are now the preferred approaches for treatment of malignancies, and traditional therapeutic approaches are being replaced by the use of agents which potentiate immune effector mechanisms, broadly called "immunotherapy". Cancer immunotherapy is generally classified into two main classes including active and passive methods. Interventions to augment the immune system of the patient, for example, vaccination or adjuvant therapy, actively promote antitumor effector mechanisms to improve cancer elimination. On the other hand, administration of specific monoclonal antibodies (mAbs) against different tumor antigens and adoptive transfer of genetically-modified specific T cells are currently the most rapidly developing approaches for cancer targeted therapy. In this review, we will discuss the different modalities for active and passive immunotherapy for cancer.
Collapse
Affiliation(s)
- Tohid Kazemi
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Vahid Younesi
- c Department of Immunology , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| | - Farhad Jadidi-Niaragh
- c Department of Immunology , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| | - Mehdi Yousefi
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
15
|
Zhan X, Jia L, Niu Y, Qi H, Chen X, Zhang Q, Zhang J, Wang Y, Dong L, Wang C. Targeted depletion of tumour-associated macrophages by an alendronate-glucomannan conjugate for cancer immunotherapy. Biomaterials 2014; 35:10046-57. [PMID: 25245263 DOI: 10.1016/j.biomaterials.2014.09.007] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 09/01/2014] [Indexed: 12/31/2022]
Abstract
Tumour-associated macrophages (TAMs) are a set of macrophages residing in the tumour microenvironment. They play essential roles in mediating tumour angiogenesis, metastasis and immune evasion. Delivery of therapeutic agents to eliminate TAMs can be a promising strategy for cancer immunotherapy but an efficient vehicle to target these cells is still in pressing need. In this study, we developed a bisphosphonate-glucomannan conjugate that could efficiently target and specifically eliminate TAMs in the tumour microenvironment. We employed the polysaccharide from Bletilla striata (BSP), a glucomannan affinitive for macrophages that express abundant mannose receptors, to conjugate alendronate (ALN), a bisphosphonate compound with in vitro macrophage-inhibiting activities. In both in vitro and in vivo tests, the prepared ALN-BSP conjugate could preferentially accumulate in macrophages and induced them into apoptosis. In the subcutaneous S180 tumour-bearing mice model, the treatment using ALN-BSP effectively eliminated TAMs, remarkably inhibited angiogenesis, recovered local immune surveillance, and eventually suppressed tumour progression, without eliciting any unwanted effect such as systematic immune response. Interestingly, ALN alone failed to exhibit any anti-TAM activity in vivo, probably because this compound was susceptible to the mildly acidic tumour microenvironment. Taken together, these results demonstrate the potential of ALN-BSP as a safe and efficient tool targeted at direct depletion of TAMs for cancer immunotherapy.
Collapse
Affiliation(s)
- Xiudan Zhan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Lixin Jia
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yiming Niu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Haixia Qi
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Qingwen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
| |
Collapse
|
16
|
Pinfold TL, Brown GK, Bettiol SS, Woods GM. Mouse Model of Devil Facial Tumour Disease Establishes That an Effective Immune Response Can be Generated Against the Cancer Cells. Front Immunol 2014; 5:251. [PMID: 24904594 PMCID: PMC4034705 DOI: 10.3389/fimmu.2014.00251] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/13/2014] [Indexed: 11/13/2022] Open
Abstract
The largest carnivorous marsupial in Australia, the Tasmanian devil (Sarcophilus harrisii) is facing extinction in the wild due to a transmissible cancer known as Devil Facial Tumour Disease (DFTD). DFTD is a clonal cell line transmitted from host to host with 100% mortality and no known immunity. While it was first considered that low genetic diversity of the population of devils enabled the allograft transmission of DFTD recent evidence reveals that genetically diverse animals succumb to the disease. The lack of an immune response against the DFTD tumor cells may be due to a lack of immunogenicity of the tumor cells. This could facilitate transmission between devils. To test immunogenicity, mice were injected with viable DFTD cells and anti-DFTD immune responses analyzed. A range of antibody isotypes against DFTD cells was detected, indicating that as DFTD cells can induce an immune response they are immunogenic. This was supported by cytokine production, when splenocytes from mice injected with DFTD cells were cultured in vitro with DFTD cells and the supernatant analyzed. There was a significant production of IFN-γ and TNF-α following the first injection with DFTD cells and a significant production of IL-6 and IL-10 following the second injection. Splenocytes from naïve or immunized mice killed DFTD cells in in vitro cytotoxicity assays. Thus, they are also targets for immunological destruction. We conclude that as an immune response can be generated against DFTD cells they would be suitable targets for a vaccine.
Collapse
Affiliation(s)
- Terry L Pinfold
- Menzies Research Institute Tasmania, University of Tasmania , Hobart, TAS , Australia
| | - Gabriella K Brown
- Menzies Research Institute Tasmania, University of Tasmania , Hobart, TAS , Australia
| | - Silvana S Bettiol
- School of Medicine, University of Tasmania , Hobart, TAS , Australia
| | - Gregory M Woods
- Menzies Research Institute Tasmania, University of Tasmania , Hobart, TAS , Australia
| |
Collapse
|
17
|
Gujar SA, Clements D, Dielschneider R, Helson E, Marcato P, Lee PWK. Gemcitabine enhances the efficacy of reovirus-based oncotherapy through anti-tumour immunological mechanisms. Br J Cancer 2013; 110:83-93. [PMID: 24281006 PMCID: PMC3887295 DOI: 10.1038/bjc.2013.695] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 10/08/2013] [Accepted: 10/09/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Reovirus preferentially infects and kills cancer cells and is currently undergoing clinical trials internationally. While oncolysis is the primary mode of tumour elimination, increasing evidence illustrates that reovirus additionally stimulates anti-tumour immunity with a capacity to target existing and possibly relapsing cancer cells. These virus-induced anti-tumour immune activities largely determine the efficacy of oncotherapy. On the other hand, anti-viral immune responses can negatively affect oncotherapy. Hence, the strategic management of anti-tumour and anti-viral immune responses through complementary therapeutics is crucial to achieve the maximum anti-cancer benefits of oncotherapy. METHODS Intra-peritoneal injection of mouse ovarian surface epithelial cells (ID8 cells) into wild-type C57BL/6 mice was treated with a therapeutic regimen of reovirus and/or gemcitabine and then analysed for prolonged survival, disease pathology, and various immunological parameters. Furthermore, in vitro analyses were conducted to assess apoptosis, viral spread, and viral production during reovirus and/or gemcitabine treatment. RESULTS We demonstrate that reovirus and gemcitabine combination treatment postpones peritoneal carcinomatosis development and prolongs the survival of cancer-bearing hosts. Importantly, these anti-cancer benefits are generated through various immunological mechanisms, including: (1) inhibition of myeloid-derived suppressor cells recruitment to the tumour microenvironment, (2) downmodulation of pro-MDSC factors, and (3) accelerated development of anti-tumour T-cell responses. CONCLUSION The complementation of reovirus with gemcitabine further potentiates virus-initiated anti-cancer immunity and enhances the efficacy of oncotherapy. In the context of ongoing clinical trials, our findings represent clinically relevant information capable of enhancing cancer outcomes.
Collapse
Affiliation(s)
- S A Gujar
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - D Clements
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - R Dielschneider
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - E Helson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - P Marcato
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - P W K Lee
- 1] Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada [2] Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
18
|
Raja H, Snyder MR, Johnston PB, O’Neill BP, Caraballo JN, Balsanek JG, Peters BE, Decker PA, Pulido JS. Effect of intravitreal methotrexate and rituximab on interleukin-10 levels in aqueous humor of treated eyes with vitreoretinal lymphoma. PLoS One 2013; 8:e65627. [PMID: 23750271 PMCID: PMC3672178 DOI: 10.1371/journal.pone.0065627] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 05/01/2013] [Indexed: 11/22/2022] Open
Abstract
Intraocular cytokines are promising diagnostic biomarkers of vitreoretinal lymphoma. Here, we evaluate the utility of IL-10, IL-6 and IL-10/IL-6 for discriminating lymphoma from uveitis and report the effects of intraocular methotrexate and rituximab on aqueous cytokine levels in eyes with lymphoma. This is a retrospective case series including 10 patients with lymphoma and 7 patients with uveitis. Non-parametric Mann-Whitney analysis was performed to determine statistical significance of difference in interleukin levels between lymphoma and uveitis. Compared to eyes with uveitis, eyes with lymphoma had higher levels of IL-10 (U = 7.0; two-tailed p = 0.004) and IL-10/IL-6 (U = 6.0; two-tailed p = 0.003), whereas IL-6 levels were more elevated, although insignificant, in those patients with uveitis than in lymphoma (U = 15.0; two-tailed p = ns). Using a receiver operating characteristic analysis to identify threshold values diagnostic for lymphoma, optimal sensitivity and specificity improved to 80.0% and 100%, respectively, for IL-10>7.025 pg/ml and 90.0% and 100.0%, respectively, for IL-10/IL-6>0.02718. In patients in whom serial interleukin levels were available, regular intravitreal treatment with methotrexate and rituximab was associated with reduction in IL-10 levels over time. In conclusion, optimal IL-10 and IL-10/IL-6 threshold values are associated with a diagnostic sensitivity ≥80% and specificity of 100%. Therefore, these cytokines may serve as a useful adjunct in the diagnosis of lymphoma. While negative IL-10 and IL-10/IL-6 values do not exclude a diagnosis of lymphoma, elevated levels do appear to be consistent with lymphoma clinically. Moreover, elevated levels of IL-10 in the setting of a clinically quiet eye may point to impending disease recurrence. Lastly, once lymphoma is diagnosed, IL-10 levels can be monitored over time to assess disease activity and therapeutic response.
Collapse
Affiliation(s)
- Harish Raja
- Mayo School of Graduate Medical Education, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Melissa R. Snyder
- Department of Laboratory Medicine and Pathology, Antibody Immunology Laboratory, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Patrick B. Johnston
- Department of Hematology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Brian P. O’Neill
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Juline N. Caraballo
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Joseph G. Balsanek
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Brian E. Peters
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Paul A. Decker
- Department of Biomedical Statistics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jose S. Pulido
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
19
|
Miyatake J, Ohguro N, Kawauchi M, Kumode T, Yamaguchi T, Morita Y, Tatsumi Y, Maeda Y, Matsumura I. A case of intraocular lymphoma with central nervous system involvement and high interleukin-10 levels in both vitreous humor and cerebrospinal fluids: successful treatment with a combination of intravitreal, intrathecal, and systemic therapy. Int Cancer Conf J 2013. [DOI: 10.1007/s13691-012-0063-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
20
|
Zheng MM, Zhang Z, Bemis K, Belch AR, Pilarski LM, Shively JE, Kirshner J. The systemic cytokine environment is permanently altered in multiple myeloma. PLoS One 2013; 8:e58504. [PMID: 23544044 PMCID: PMC3609759 DOI: 10.1371/journal.pone.0058504] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 02/05/2013] [Indexed: 02/03/2023] Open
Abstract
Multiple myeloma (MM) is an incurable bone marrow malignancy of the B cell lineage. Utilizing multiplex Luminex technology we measured levels of 25 cytokines in the plasma of normal donors (n = 177), those with monoclonal gammopathy of undetermined significance (n = 8), and MM patients (n = 55) with either active disease, on treatment, or in remission. The cytokine levels were compared between normal donors and MM patients as well as between various phases of MM, and discriminant analysis was used to create a predictive classification model based on the differentially expressed cytokines. Evaluating age- and gender-dependence of cytokine expression, we determined that with age there is a shift toward a pro-inflammatory environment. Moreover, we observed a strong gender bias in cytokine expression. However, the profile of differentially expressed cytokines was heavily skewed toward an anti-inflammatory, pro-tumorigenic response in patients with MM. Significantly, our predictive model placed all patients in remission in the same category as those with active disease. Thus, our study demonstrates that the homeostasis of systemic cytokines is not restored when MM patients enter remission, suggesting that once an individual has cancer, the microenvironment is permanently altered and the system is primed for a relapse.
Collapse
Affiliation(s)
- Mary M. Zheng
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Zhifang Zhang
- Division of Immunology, Beckman Research Institute of the City of Hope National Medical Center, Duarte, California, United States of America
| | - Kyle Bemis
- Department of Statistics, Purdue University, West Lafayette, Indiana, United States of America
| | - Andrew R. Belch
- Department of Oncology, University of Alberta and Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Linda M. Pilarski
- Department of Medical Oncology, University of Alberta and Cross Cancer Institute, Edmonton, Alberta, Canada
| | - John E. Shively
- Division of Immunology, Beckman Research Institute of the City of Hope National Medical Center, Duarte, California, United States of America
| | - Julia Kirshner
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail:
| |
Collapse
|
21
|
Ghosh S, Adhikary A, Chakraborty S, Nandi P, Mohanty S, Chakraborty S, Bhattacharjee P, Mukherjee S, Putatunda S, Chakraborty S, Chakraborty A, Sa G, Das T, Sen PC. Nifetepimine, a dihydropyrimidone, ensures CD4+ T cell survival in a tumor microenvironment by maneuvering sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA). J Biol Chem 2012; 287:32881-96. [PMID: 22851172 DOI: 10.1074/jbc.m112.357889] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Multiple mechanisms have been proposed by which tumors induce T cell apoptosis to circumvent tumor immune-surveillance. Although sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA) have long been known to regulate intracellular Ca(2+) homeostasis, few studies have examined the role of SERCA in processes of T lymphocyte survival and activation. In this context it remains largely unexplored as to how tumors jeopardize SERCA function to disable T cell-mediated anti-tumor immunity. Here, we show that human CD4(+) T cells in the presence of tumor conditions manifested an up-regulation of SERCA3 expression that resulted in development of endoplasmic reticulum stress leading to CD4(+) T cell apoptosis. Prostaglandin E(2) produced by the tumor cell plays a critical role in up-regulating SERCA3 by enhancing the binding of its transcription factor Sp1. Gene manipulation and pharmacological approaches further established that an increase in SERCA expression also resulted in subsequent inhibition of PKCα and -θ and retention of NFκB in the cytosol; however, down-modulation of SERCA3 expression by a dihydropyrimidone derivative, ethyl-4-(3-nitro)-phenyl-6-methyl-2-oxo-1,2,3,4-tetrahydropyrimidine-5 carboxylate (nifetepimine), protected the CD4(+) T cells from tumor-induced apoptosis. In fact, nifetepimine-mediated restoration of PKC activity resulted in nuclear translocation of p65NFκB, thereby ensuring its survival. Studies further undertaken in a tumor-bearing mice model revalidated the immunoprotective role of nifetepimine. Our present study thus strongly suggests that imbalance in cellular calcium homeostasis is an important factor leading to CD4(+) T cell death during cancer and holds promise that nifetepimine may have the potential to be used as an immunorestoring agent in cancer bearers.
Collapse
Affiliation(s)
- Swatilekha Ghosh
- Division of Molecular Medicine, Bose Institute, P1/12 Calcutta Improvement Trust Scheme VIIM Kolkata 700054, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Expression of Leukocyte Inhibitory Immunoglobulin-like Transcript 3 Receptors by Ovarian Tumors in Laying Hen Model of Spontaneous Ovarian Cancer. Transl Oncol 2012; 5:85-91. [PMID: 22496924 DOI: 10.1593/tlo.11328] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 12/23/2011] [Accepted: 01/03/2012] [Indexed: 01/08/2023] Open
Abstract
Attempts to enhance a patient's immune response and ameliorate the poor prognosis of ovarian cancer (OVCA) have largely been unsuccessful owing to the suppressive tumor microenvironment. Leukocyte immunoglobulin-like transcript 3 (ILT3) inhibitory receptors have been implicated in immunosuppression in several malignancies. The expression and role of ILT3 in the progression of ovarian tumors are unknown. This study examined the expression and association of ILT3 in ovarian tumors in laying hens, a spontaneous preclinical model of human OVCA. White Leghorn laying hens were selected by transvaginal ultrasound scanning. Serum and normal ovaries or ovarian tumors were collected. The presence of tumors and the expression of ILT3 were examined by routine histology, immunohistochemistry, Western blot analysis, and reverse transcription-polymerase chain reaction. In addition to stromal immune cell-like cells, the epithelium of the ovarian tumors also expressed ILT3 with significantly high intensity than normal ovaries. Among different subtypes of ovarian carcinomas, serous OVCA showed the highest ILT3 staining intensity, whereas endometrioid OVCA had the lowest intensity. Similar to humans, an immunoreactive protein band of approximately 55 kDa for ILT3 was detected in the ovarian tumors in hens. The patterns of ILT3 protein and messenger RNA expression by ovarian tumors in different subtypes and stages were similar to those of immunohistochemical staining. The results of this study suggest that laying hens may be useful to generate information on ILT3-associated immunosuppression in OVCA. This animal model also offers the opportunity to develop and test anti-ILT3 immunotherapy to enhance antitumor immunity against OVCA in humans.
Collapse
|
23
|
Abstract
One of the most fundamental problems in immunology is the seemingly schizophrenic ability of the immune system to launch robust immunity against pathogens, while acquiring and maintaining a state of tolerance to the body's own tissues and the trillions of commensal microorganisms and food antigens that confront it every day. A fundamental role for the innate immune system, particularly dendritic cells (DCs), in orchestrating immunological tolerance has been appreciated, but emerging studies have highlighted the nature of the innate receptors and the signaling pathways that program DCs to a tolerogenic state. Furthermore, several studies have emphasized the major role played by cellular interactions and the microenvironment in programming tolerogenic DCs. Here, we review these studies and suggest that the innate control of tolerogenic responses can be viewed as different hierarchies of organization, in which DCs, their innate receptors and signaling networks, and their interactions with other cells and local microenvironments represent different levels of the hierarchy.
Collapse
Affiliation(s)
- Santhakumar Manicassamy
- Emory Vaccine Center, Yerkes National Primate Research Center, Department of Pathology, Emory University School of Medicine, Atlanta, GA 30329, USA
| | | |
Collapse
|
24
|
Chakraborty NG. Immunoregulation through IL-10 gene expression and the fate of cytotoxic T lymphocyte-mediated tumor immunotherapy. INDIAN JOURNAL OF HUMAN GENETICS 2011; 15:3-8. [PMID: 20407642 PMCID: PMC2846567 DOI: 10.4103/0971-6866.50862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Gene analysis of tumor associated antigens revealed that tumor antigens are all normal gene product. Inducing tumor reactive cytotoxic T lymphocytes (CT) in the patients is same as inducing autoimmunity in the patients. Immunosuppressive cytokine interleukin-10 (IL-10) plays an important role in maintaining homeostasis or tolerance. To break the tumor tolerance, blocking and IL-10 secretion or intervention in the pathways of IL-10 gene activation is indeed important.
Collapse
Affiliation(s)
- Nitya G Chakraborty
- Department of Medicine, University of Connecticut School of Medicine, Farmington, CT 006030-1628, USA
| |
Collapse
|
25
|
Sharma S, Stabila J, Pietras L, Singh AR, McGonnigal B, Ernerudh J, Matthiesen L, Padbury JF. Haplotype-dependent differential activation of the human IL-10 gene promoter in macrophages and trophoblasts: implications for placental IL-10 deficiency and pregnancy complications. Am J Reprod Immunol 2011; 64:179-87. [PMID: 20482524 DOI: 10.1111/j.1600-0897.2010.00854.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PROBLEM polymorphic changes in the IL-10 gene promoter have been identified that lead to altered IL-10 production. We hypothesized that because of these genotypic changes, the IL-10 promoter might be expressed in a cell type-specific manner and may respond differentially to inflammatory triggers. METHOD OF STUDY we created reporter gene promoter constructs containing GCC, ACC, and ATA haplotypes using DNA from patients harboring polymorphic changes at -1082 (G→A), -819 (C→T), and -592 (C→A) sites in the IL-10 promoter. These individual luciferase reporter constructs were transiently transfected into either primary term trophoblasts or THP1 monocytic cells. DNA-binding studies were performed to implicate the role of the Sp1 transcription factor in response to differential promoter activity. RESULTS our results suggest that the GCC promoter construct was activated in trophoblast cells in response to lipopolysaccharide (LPS), as demonstrated by reporter gene expression, but not in monocytic cells. The ACC construct showed weaker activation in both cell types. Importantly, while the ATA promoter was constitutively activated in both cell types, its expression was selectively repressed in response to LPS, but only in trophoblasts. DNA-nuclear protein binding assays with nuclear extracts from LPS treated or untreated cells suggested a functional relevance for Sp1 binding differences at the -592 position. CONCLUSIONS these results demonstrate cell type-specific effects of the genotypic changes in the IL-10 gene promoter. These responses may be further modulated by bacterial infections or other inflammatory conditions to suppress IL-10 production in human trophoblasts.
Collapse
Affiliation(s)
- Surendra Sharma
- Department of Pediatrics, Women and Infants Hospital-Warren Alpert Medical School of Brown University, Providence, Rhode Island 02905, USA.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Wang R, Lu M, Zhang J, Chen S, Luo X, Qin Y, Chen H. Increased IL-10 mRNA expression in tumor-associated macrophage correlated with late stage of lung cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2011; 30:62. [PMID: 21595995 PMCID: PMC3117740 DOI: 10.1186/1756-9966-30-62] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Accepted: 05/20/2011] [Indexed: 12/31/2022]
Abstract
Background Monocyte recruited into the tumor and maturation to tumor-associated macrophage (TAM). Interleukin-10(IL-10) is a potent immunosuppressive cytokine, which can be secreted from both primary tumor and stromal cells. However, there are controversies regarding its role in the progression of cancer. So it is important to isolate TAM from tumor cells to study the role of IL-10 in the progress of cancer. The aim of our study was to determine whether IL-10 expressed by TAM correlated with clinicopathological factors in NSCLC. Methods TAM in NSCLC was isolated by short-term culture in serum free medium with the modification to literature reports. The mRNA expression levels of IL-10, cathepsin B, cathepsin S, which were closely related with TAM according to the literatures, were evaluated by Quantitative real-time RT-PCR in 63 NSCLC. The relationships between their expression levels and clinicopathological features were investigated. Results We successfully achieved up to 95% purity of TAM, derived from 63 primary lung cancer tissues. TAM expressed high levels of IL-10, cathepsin B in NSCLC. High levels of IL-10 in TAM significantly correlated with stage, tumor size, lymph node metastasis, lymphovascular invasion or histologic poor differentiation. Conclusions Our results revealed that TAM with high levels of IL-10 expression may play an important role in the progression of non-small cell lung cancer. The data also suggested that TAMs may involve in tumor immunosuppression through overexpressed IL-10. Additionally, the phenotype of isolated TAM can be potentially used to predict clinicopathological features as well.
Collapse
Affiliation(s)
- Rui Wang
- Department of Thoracic Surgery, Shanghai Cancer Hospital, Fudan University, China
| | | | | | | | | | | | | |
Collapse
|
27
|
López MN, Pesce B, Kurte M, Pérez C, Segal G, Roa J, Aguillón JC, Mendoza-Naranjo A, Gesser B, Larsen C, Villablanca A, Choudhury A, Kiessling R, Salazar-Onfray F. A synthetic peptide homologous to IL-10 functional domain induces monocyte differentiation to TGF-β+ tolerogenic dendritic cells. Immunobiology 2011; 216:1117-26. [PMID: 21601308 DOI: 10.1016/j.imbio.2011.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 04/09/2011] [Accepted: 04/14/2011] [Indexed: 11/27/2022]
Abstract
We have previously demonstrated that IT9302, a nonameric peptide homologous to the C-terminal domain of human IL-10, mimics several effects of the cytokine including down-regulation of the antigen presentation machinery and increased sensitivity of tumor cells to NK-mediated lysis. In the present report, we have explored a potential therapeutic utility for IT9302 related to the ex vivo production of tolerogenic dendritic cells (DCs). Our results indicate that IT9302 impedes human monocyte response to differentiation factors and reduces antigen presentation and co-stimulatory capacity by DCs. Additionally, peptide-treated DCs show impaired capacity to stimulate T-cell proliferation and IFN-γ production. IT9302 exerts its effect through mechanisms, in part, distinct from IL-10, involving STAT3 inactivation and NF-κB intracellular pathway. IT9302-treated DCs display increased expression of membrane-associated TGF-β, linked to a more effective induction of foxp3+ regulatory T cells. These results illustrate for the first time that a short synthetic peptide can promote monocytes differentiation to tolerogenic DCs with therapeutic potential for the treatment of autoimmune and transplantation-related immunopathologic disease.
Collapse
Affiliation(s)
- Mercedes N López
- Millennium Nucleus on Immunology and Immunotherapy, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Yigit R, Massuger LFAG, Figdor CG, Torensma R. Ovarian cancer creates a suppressive microenvironment to escape immune elimination. Gynecol Oncol 2010; 117:366-72. [PMID: 20144842 DOI: 10.1016/j.ygyno.2010.01.019] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 01/08/2010] [Accepted: 01/12/2010] [Indexed: 12/23/2022]
Abstract
BACKGROUND Considering the high mortality rate of ovarian cancer due to the absence of curative treatment in advanced stage or at recurrence, new therapeutic strategies are urgently needed. Immunotherapy is one of these strategies that yielded promising results in fundamental and animal research in the past years. However, implementation in clinical practice remains poor. The aim of this review is to gain insight into the mechanisms of interaction between ovarian cancer and the immune system in order to develop better immunotherapeutic strategies. METHODS We searched the published literature for studies focusing on interactions between ovarian cancer and the immune system, with emphasis on outcome data in order to create a knowledge base that is well grounded in clinical reality. RESULTS The immunological response against cancer is a critical balance between immune-activating and immune-suppressing mechanisms. Besides the immune-activating tumor infiltrating lymphocytes (TILs), immune-suppressive regulatory T-cells (Tregs), tolerance-inducing plasmacytoid dendritic cells (pDCs), B7-H4+ macrophages, immune-suppressive cytokines such as IL10 and TGF-beta are also found in the tumor environment. Myeloid-derived suppressive cells (MDSCs) are recently found to have a significant role in immune suppression in ovarian cancer in murine studies. Furthermore, vascular endothelial growth factor (VEGF) is also known to have an immune-suppressing role besides its angiogenic role. All those concerted mechanisms result in the creation of an environment where the cancer is invincible and can grow unhampered. CONCLUSION Further knowledge of the mechanisms involved is needed to develop better strategies and improve the clinical applicability of immunotherapy. Effective immunotherapy must combine immune-activating strategies with elimination of immune-suppressing mechanisms. We believe that tilting the balance from an immune-suppressive to an immune-active environment may have an enormous impact on the disease.
Collapse
Affiliation(s)
- Refika Yigit
- Department of Obstetrics and Gynecology (791), Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | | | | | | |
Collapse
|
30
|
Baiocchi OCG, Colleoni GWB, Caballero OL, Vettore AL, Bulgarelli A, Dalbone MA, Granato CFH, Franco MF, Pestana JOM. Epstein – Barr viral load, interleukin-6 and interleukin-10 levels in post-transplant lymphoproliferative disease: A nested case – control study in a renal transplant cohort. Leuk Lymphoma 2009; 46:533-9. [PMID: 16019481 DOI: 10.1080/10428190400027837] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The possible correlation among Epstein-Barr virus (EBV) load, interleukin-6 (IL-6) and interleukin-10 (IL-10) levels has become an attractive issue and can provide a useful tool for diagnosis and monitoring of patients at risk for post-transplant lymphoproliferative disease (PTLD) development. At the time of diagnosis of PTLD, 11 patients were prospectively enrolled and 55 nested controls were selected from a 1800 renal transplant cohort. Real-time polymerase chain reaction (PCR) was used to quantify EBV load in peripheral blood mononuclear cells (PBMC). Serum IL-6 and IL-10 levels were determined using an enzyme-linked immunosorbent assay (ELISA). The median EBV load of PTLD cases was 17400 copies/10(6) PBMC, statistically different from controls (P=0.001). The median IL-6 level of PTLD cases was not different from controls (P=0.079). However, median IL-10 levels showed a significant difference in both groups (P < or = 0.001). The receiver-operating characteristic (ROC) curve analysis was applied to estimate the IL-10 cut-off value predictive of PTLD development. We found that 73.5 pg/ml has high sensitivity (1.00) and specificity (0.85). Also, Pearson's analysis showed a strong correlation between EBV load and serum IL-10 concentration (P < or = 0.001). This nested case-control study demonstrates that EBV load at diagnosis of PTLD correlates with IL-10 levels, and that monitoring of IL-10 can provide a less expensive and less time-consuming tool for PTLD diagnosis and close follow-up of patients at risk. Furthermore, we were able to define a cut-off value of IL-10 mostly predictive of PTLD development in this cohort. Our data suggest that serial measurements prior to PTLD development must be carried out to validate our hypothesis.
Collapse
Affiliation(s)
- Otávio C G Baiocchi
- Hematology and Transfusion Medicine Service, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
ProtEx technology for the generation of novel therapeutic cancer vaccines. Exp Mol Pathol 2009; 86:198-207. [PMID: 19454266 DOI: 10.1016/j.yexmp.2009.01.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2009] [Indexed: 01/15/2023]
Abstract
Therapeutic vaccines present an attractive alternative to conventional treatments for cancer. However, tumors have evolved various immune evasion mechanisms to modulate innate, adaptive, and regulatory immunity for survival. Therefore, successful vaccine formulations may require a non-toxic immunomodulator or adjuvant that not only induces/stimulates innate and adaptive tumor-specific immune responses, but also overcomes immune evasion mechanisms. Given the paramount role costimulation plays in modulating innate, adaptive, and regulatory immune responses, costimulatory ligands may serve as effective immunomodulating components of therapeutic cancer vaccines. Our laboratory has developed a novel technology designated as ProtEx that allows for the generation of recombinant costimulatory ligands with potent immunomodulatory activities and the display of these molecules on the cell surface in a rapid and efficient manner as a practical and safe alternative to gene therapy for immunomodulation. Importantly, the costimulatory ligands not only function when displayed on tumor cells, but also as soluble proteins that can be used as immunomodulatory components of conventional vaccine formulations containing tumor-associated antigens (TAAs). We herein discuss the application of the ProtEx technology to the development of effective cell-based as well as cell-free conventional therapeutic cancer vaccines.
Collapse
|
32
|
Levina V, Su Y, Nolen B, Liu X, Gordin Y, Lee M, Lokshin A, Gorelik E. Chemotherapeutic drugs and human tumor cells cytokine network. Int J Cancer 2008; 123:2031-40. [PMID: 18697197 DOI: 10.1002/ijc.23732] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The ability of human tumor cell lines to produce various cytokines, chemokines, angiogenic and growth factors was investigated using Luminex multiplex technology. Media conditioned by tumor cells protected tumor cells from drug-induced apoptosis and stimulated tumor cell proliferation. Antibodies neutralizing IL-6, CXCL8, CCL2 and CCL5 blocked this stimulation. Treatment of tumor cells with doxorubicin and cisplatin resulted in a substantial increase in the production of IL-6, CXCL8, CCL2, CCL5, BFGF, G-CSF and VEGF. This stimulation was associated with drug-induced activation of NF-kappaB, AP-1, AP-2, CREB, HIF-1, STAT-1, STAT-3, STAT-5 and ATF-2 transcription factors and upregulation of IL-6, CXCL8, FGF-2, CSF-3 and CCL5 gene expression. Treatment of tumor cells with doxorubicin and antibodies neutralizing G-CSF, CCL2 or CCL5 had higher inhibitory effects than each modality used alone. These results indicate that chemokines and growth factors produced by tumor by binding to the cognate receptors on tumor and stroma cells could provide proliferative and antiapoptotic signals helping tumor to escape drug-mediated destruction. Clinical studies showed that antibodies neutralizing VEGF (Avastin/Bevacizumab) or blocking HER2/neu signaling (Herceptin/Trastuzumab) could increase the efficacy of chemotherapy, although these beneficial effects have been limited. It is possible that drug-stimulated production of growth and proangiogenic factors could counterbalance the effects of antibody therapy. In addition, numerous growth factors and chemokines share angiogenic and growth-stimulating properties, and thus reduction of a single factor is insufficient to completely block tumor growth. Thus, a broad disruption of tumor cytokine network is needed to further increase the efficacy of cancer therapy.
Collapse
Affiliation(s)
- Vera Levina
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Das T, Sa G, Paszkiewicz-Kozik E, Hilston C, Molto L, Rayman P, Kudo D, Biswas K, Bukowski RM, Finke JH, Tannenbaum CS. Renal cell carcinoma tumors induce T cell apoptosis through receptor-dependent and receptor-independent pathways. THE JOURNAL OF IMMUNOLOGY 2008; 180:4687-96. [PMID: 18354192 DOI: 10.4049/jimmunol.180.7.4687] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tumors can promote their own progressive growth by inducing T cell apoptosis. Though previous studies suggested that tumor-mediated T cell killing is receptor dependent, we recently showed that tumor gangliosides also participate, a notion consistent with reports indicating that, in some cell types, gangliosides can activate the intrinsic apoptotic pathway by stimulating reactive oxygen species production, cytochrome c release, and caspase-9 activation. In this study, we used normal peripheral blood T cells, as well as caspase-8-, caspase-9-, and Fas-associated death domain protein-deficient Jurkat cells, to assess whether the death ligands and gangliosides overexpressed by the renal cell carcinoma (RCC) cell line SK-RC-45 can independently stimulate T cell apoptosis as a mechanism of immune escape. Anti-FasL Abs and the glycosylceramide synthase inhibitor 1-phenyl-2-hexadecanoylamino-3-pyrrolidino-1-propanol (PPPP) each partially inhibited the ability of SK-RC-45 to kill cocultured activated T cells; together, as purified molecules, RCC gangliosides and rFasL induced a more extensive mitochondrial permeability transition and greater levels of apoptosis than either agent alone, equivalent to that induced by the FasL- and ganglioside-expressing RCC line itself. rFasL-mediated apoptosis was completely inhibited in caspase-8- and Fas-associated death domain protein-negative Jurkat cells, though apoptosis induced by purified gangliosides remained intact, findings that correlate with the observed partial inhibition of SK-RC-45-induced apoptosis in the Jurkat lines with defective death receptor signaling. Western blot analysis performed on lysates made from wild-type and mutant Jurkat cells cocultured with SK-RC-45 revealed caspase activation patterns and other biochemical correlates which additionally supported the concept that tumor-associated gangliosides and FasL independently activate the caspase cascade in T cells through the intrinsic and extrinsic pathways, respectively.
Collapse
Affiliation(s)
- Tanya Das
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Nagano T, Kunisada M, Yu X, Masaki T, Nishigori C. Involvement of Interleukin-10 Promoter Polymorphisms in Nonmelanoma Skin Cancers-A Case Study in Non-Caucasian Skin Cancer Patients†. Photochem Photobiol 2007; 84:63-6. [DOI: 10.1111/j.1751-1097.2007.00245.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
35
|
Zhou J, Ye F, Chen H, Lv W, Gan N. The expression of interleukin-10 in patients with primary ovarian epithelial carcinoma and in ovarian carcinoma cell lines. J Int Med Res 2007; 35:290-300. [PMID: 17593856 DOI: 10.1177/147323000703500302] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
This study determined interleukin-10 (IL-10) expression in patients with ovarian carcinoma and in ovarian carcinoma cell lines, and investigated its clinical significance in the development and progression of ovarian carcinoma. Expression of IL-10 in ovarian carcinoma, benign ovarian tumour, normal control tissues and ovarian carcinoma cell lines was detected by immunohistochemistry and Western blot analysis. IL-10 concentrations in sera and ascites from patients with ovarian carcinoma, in sera from patients with benign ovarian tumour and normal controls, and in supernatants of ovarian carcinoma cell line cultures were measured by enzyme-linked immunosorbent assay. The tissue level of IL-10 in ovarian carcinoma was significantly higher than in benign ovarian tumour and normal controls. IL-10 expression was detectable in cell lysate and supernatant from ovarian carcinoma cell lines. In patients with ovarian carcinoma the IL-10 level in ascitic fluid was significantly higher than in sera, and the serum IL-10 level in ovarian carcinoma was significantly higher than in benign ovarian tumour and normal controls. Ascitic IL-10 levels in ovarian carcinoma were significantly correlated with disease stage but not cytological grade. These results suggest that ovarian carcinoma cells are able to synthesize and secrete IL-10, which probably assists in promoting the development and progression of ovarian carcinoma.
Collapse
Affiliation(s)
- J Zhou
- Department of Gynaecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, China
| | | | | | | | | |
Collapse
|
36
|
Mandal D, Bhattacharyya S, Lahiry L, Chattopadhyay S, Sa G, Das T. Black Tea-Induced Decrease in IL-10 and TGF-β of Tumor Cells Promotes Th1/Tc1 Response in Tumor Bearer. Nutr Cancer 2007; 58:213-21. [PMID: 17640168 DOI: 10.1080/01635580701328503] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Several lines of evidence support that impairment of host immune function by tumor may be related to several strategies of tumor escape from immunosurveillance. We found that in Ehrlich's ascites carcinoma (EAC)-bearing mice, the tumor cells secrete immunosuppressive cytokines, transforming growth factor beta (TGF-beta) and interleukin-10 (IL-10) that induce a general T helper cells type 2 (Th2) dominance dampening the T cytotoxic cells type 1 (Tc1) population. Interestingly, black tea at the antitumor dose of 2.5% significantly reduced TGF-beta and IL-10 in tumor cells in vivo, thereby preventing Th2 dominance in the tumor bearers and initiating a Th1/Tc1 response. Thus, apart from its anticancer activity, this popular beverage also rejuvenates cancer immunosurveillance by modulating cytokine profiles and establishing Th1/Tc1 dominance in the tumor-bearing host.
Collapse
|
37
|
Abstract
Tumors express antigens that should induce immune-mediated rejection, but spontaneous rejection of established tumors is rare. Recent work demonstrates that one reason for the lack of tumor rejection is that tumors actively defeat host immunity. This concept forces us to rethink current approaches to harnessing potent, specific host immunity to battle cancer, most of which are based on the paradigm that inducing more antitumor immune cells alone is therapeutic. However, as I discuss in this Personal Perspective, a newer paradigm predicts that reducing tumor-driven immune suppression will be clinically beneficial. CD4+CD25+ Tregs are one mechanism of tumor-driven immune evasion that provide prototypical targets for testing novel anticancer treatment strategies within the newer paradigm.
Collapse
Affiliation(s)
- Tyler J Curiel
- San Antonio Cancer Institute, University of Texas Health Sciences Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
38
|
Salazar-Onfray F, López MN, Mendoza-Naranjo A. Paradoxical effects of cytokines in tumor immune surveillance and tumor immune escape. Cytokine Growth Factor Rev 2007; 18:171-82. [PMID: 17329145 DOI: 10.1016/j.cytogfr.2007.01.015] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The role of cytokines in modulating the formation of new tumors is mediated by their ability to regulate antigen-specific anti-tumor responses and by the activation of non-specific mechanisms, including those involved in the processes of inflammation and innate resistance. Cytokines may influence the growth of tumors by acting directly on tumor cells as growth promoting or growth inhibiting factors or indirectly by attracting inflammatory cell types and affecting angiogenesis. Due to the potency and complexity of cytokine activity against tumor growth, the improvement of cloning techniques and the availability of recombinant forms of different cytokines, a great effort has been made in the recent years to exploit this anti-tumor potential for cancer therapy. This important goal has been difficult to achieve in most cases due to toxicity of most cytokines which could not be dissociated from their anti-tumoral functions. Nevertheless, if well designed, treatment protocols and/or modifications of the cytokine molecules may in some situations augment the anti-tumor effects while limiting the toxicity. One of these molecular approaches could be the design of peptides containing the functional domain of certain cytokines, exemplified by IT9302, a peptide homologous to the functional domain of IL-10, which has demonstrated to increase tumor NK cell sensitivity.
Collapse
Affiliation(s)
- Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| | | | | |
Collapse
|
39
|
Shafer-Weaver K, Anderson M, Malyguine A, Hurwitz AA. T Cell Tolerance to Tumors and Cancer Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 601:357-68. [PMID: 17713024 DOI: 10.1007/978-0-387-72005-0_38] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
It is widely recognized that the immune system plays a role in cancer progression and that some tumors are inherently immunogenic. The identification of tumor-associated antigens (TAAs) has stimulated research focused on immunotherapies to mediate the regression of established tumors. Cancer-specific immunity has traditionally been aimed at activating CD8+ cytotoxic T lymphocytes (CTLs) directed against major histocompatibility complex (MHC) class I-binding peptide epitopes. Other approaches utilize T cell adoptive therapy where autologous, tumor-specific T cells propagated in vitro are transferred back into recipients. However, these strategies have met with limited success in part due to the regulatory mechanisms of T cell tolerance, which poses a considerable challenge to cancer immunotherapy. Our laboratory utilizes the TRansgenic Adenocarcinoma of the Mouse Prostate (TRAMP) model, a murine model of prostate cancer, to study mechanisms of T cell tolerization to tumor antigens. We previously demonstrated that upon encounter with their cognate antigen in the tumor microenvironment, naive T cell become tolerized. Our ongoing studies are testing whether provision of CD4+ T cells can enhance tumor immunity by preventing CD8+ T cell tolerance. A greater understanding of the interaction between various tumor-specific T cell subsets will facilitate the design of novel approaches to stimulate a more potent antitumor immune response.
Collapse
Affiliation(s)
- Kimberly Shafer-Weaver
- Applied and Developmental Research Support Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD, USA
| | | | | | | |
Collapse
|
40
|
López M, Aguilera R, Pérez C, Mendoza-Naranjo A, Pereda C, Ramirez M, Ferrada C, Aguillón JC, Salazar-Onfray F. The role of regulatory T lymphocytes in the induced immune response mediated by biological vaccines. Immunobiology 2006; 211:127-36. [PMID: 16446177 DOI: 10.1016/j.imbio.2005.11.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2005] [Accepted: 11/17/2005] [Indexed: 12/20/2022]
Abstract
Immunotherapy has become a novel therapeutic alternative for various kinds of tumours. Recently, we have finalized the first phase I clinical study in Chile for the treatment of advanced malignant melanoma, using dendritic cells (DCs) loaded with allogeneic melanoma cell lysate. This study included 20 patients and the obtained results, pioneer in Latin America, showed that DC-based immunotherapy is innocuous, even provided in combination with IL-2. In addition, immunological responses were detected in 50% of the treated patients, establishing a positive correlation between the delayed type hypersensitivity (DTH) reaction, which indicates induction of in vivo immunological memory, and patients surviving. Nevertheless, objective clinical responses in vaccinated patients are still insufficient. Only sporadic objective metastasis regressions have been registered and an important proportion of the treated patients did not respond, or their responses were weak. Several strategies have been described to be used by tumours to escape from the immune response. Actually, we have demonstrated that IL-10 inhibits antigen presentation in melanoma, reducing tumour sensitivity to melanoma-specific cytotoxic T lymphocytes (CTLs). Regulation of the immunological response by inhibitory cells could be another possible cause of clinical unresponsiveness. Lately, the existence of subpopulations of regulatory T lymphocytes (RTL) able to limit the immune response in a specific form has been established, specially inhibiting the proliferation and activity of CD4+ and CD8+ effector T lymphocytes. These cellular subpopulations, mostly CD4+/CD25+/Foxp3+ T lymphocytes (Treg) of thymic origin, or TR1 lymphocytes able to release IL-10, and tumour growth factor beta (TGF-beta) producing TH3 lymphocytes, would be accumulated in the body during tumour growth, inhibiting the immune response. In relation to RTL and cancer, evidence indicates that Treg cell numbers are increased in blood and other tissues in different types of cancer. Additionally, it has been demonstrated that in patients with refractory metastatic melanoma, the adoptive transference of anti-tumour CD8+ T lymphocytes after non-myeloablative chemotherapy was able to induce important tumour regressions that would be due to elimination of RTL populations. Additionally, chemotherapeutical drugs like decarbazine, besides their effect on tumour proliferation, also have an immunosuppressive effect on T lymphocyte populations, as well as on accumulated RTL. In this article, a novel strategy for the study of RTL is proposed, including potential therapeutic innovations, which is being pioneered in current clinical trials.
Collapse
Affiliation(s)
- Mercedes López
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Kershaw MH, Teng MWL, Smyth MJ, Darcy PK. Supernatural T cells: genetic modification of T cells for cancer therapy. Nat Rev Immunol 2006; 5:928-40. [PMID: 16322746 DOI: 10.1038/nri1729] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Immunotherapy is receiving much attention as a means of treating cancer, but complete, durable responses remain rare for most malignancies. The natural immune system seems to have limitations and deficiencies that might affect its ability to control malignant disease. An alternative to relying on endogenous components in the immune repertoire is to generate lymphocytes with abilities that are greater than those of natural T cells, through genetic modification to produce 'supernatural' T cells. This Review describes how such T cells can circumvent many of the barriers that are inherent in the tumour microenvironment while optimizing T-cell specificity, activation, homing and antitumour function.
Collapse
Affiliation(s)
- Michael H Kershaw
- Peter MacCallum Cancer Centre, Saint Andrews Place, East Melbourne, Victoria 3002, Australia.
| | | | | | | |
Collapse
|
42
|
Abstract
The results from in vitro immunological experiments, murine tumor models and patients with cancer clearly demonstrate that tumors have multiple mechanisms to evade the immune response. During the early stages of tumor development malignant cells can be poor stimulators, present poor targets or become resistant to the innate immune response, while at later stages, progressively growing tumors impair the adaptive immune response by blocking the maturation and function of antigen presenting cells and causing alterations in T cell signal transduction and function. Preliminary results also suggest a correlation between some of these changes and an increased metastatic potential of the tumor cells, a diminished response to immunotherapy, and poor prognosis. Carefully coordinated basic research studies and clinical immunotherapy trials will be required to fully determine the impact on the outcome of the disease and the response to treatment. However, understanding the mechanisms used by tumor cells to evade the immune system could result in new therapeutic approaches for preventing and/or reversing these immune alterations and have the potential of improving the current results of immunotherapy trials.
Collapse
Affiliation(s)
- Michael Campoli
- Department of Immunology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263, USA
| | | | | | | | | |
Collapse
|
43
|
Röhn TA, Reitz A, Paschen A, Nguyen XD, Schadendorf D, Vogt AB, Kropshofer H. A Novel Strategy for the Discovery of MHC Class II–Restricted Tumor Antigens: Identification of a Melanotransferrin Helper T-Cell Epitope. Cancer Res 2005; 65:10068-78. [PMID: 16267033 DOI: 10.1158/0008-5472.can-05-1973] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD4+ helper T cells play a critical role in orchestrating host immune responses, including antitumor immunity. The limited availability of MHC class II-associated tumor antigens is still viewed as a major obstacle in the use of CD4+ T cells in cancer vaccines. Here, we describe a novel approach for the identification of MHC class II tumor-associated antigens (TAAs). By combining two-dimensional liquid chromatography and nanoelectrospray ionization tandem mass spectrometry, we developed a highly sensitive method for the detection of human leukocyte antigen (HLA)-DR-associated peptides of dendritic cells upon exposure to necrotic tumor cells. This approach led to the identification of a novel MHC class II-restricted TAA epitope derived from melanotransferrin. The epitope stimulated T cells derived from melanoma patients and healthy individuals and displayed promiscuity in HLA-DR restriction. Moreover, the same peptide was also presented by MHC class II-positive melanoma cells. This strategy may contribute to increase the number of tumor epitopes presented by MHC class II molecules and may support the development of more efficacious vaccines against cancer.
Collapse
Affiliation(s)
- Till A Röhn
- Pharmaceutical Research and Roche Center for Medical Genomics, F. Hoffmann La Roche Ltd., Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
44
|
Liu SH, Zhang M, Zhang WG. Strategies of Antigen-Specific T-Cell–Based Immunotherapy for Cancer. Cancer Biother Radiopharm 2005; 20:491-501. [PMID: 16248765 DOI: 10.1089/cbr.2005.20.491] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The critical role of antigen-specific T-cells in the eradication of cancer has been demonstrated in numerous animal models, while significant challenges need to be conquered before antigen-specific T-cell immunotherapy can achieve true success in clinical practice. These challenges include: (1) weak or nonimmunogenicity of spontaneous tumors, (2) negative immune regulation mechanisms of the host immune system, (3) immune inhibition exerted by tumor cells, (4) physical barrier in solid tumor, and (5) escape or resistance to immune attack by tumor cells. Nonetheless, significant success has been achieved in several clinical trials recently, highlighting the possibility of successful manipulation of the immune system for control and elimination of tumor. We focused our study on summarizing the current knowledge and corresponding strategies for improving autologous cytotoxic T-cell (CTL)-based cancer immunotherapy, which include the following aspects: (1) the selection of tumor antigens for stimulation of CTL, (2) strategies of enhancing maturation and antigen presentation activity of dendritic cells (DC), (3) strategies of activation and maintenance of CTL response, and (4) recruitment of suitable immune effector cells to tumor sites. The successful manipulation of the immune system, based on the more and more detailed knowledge of tumor immunology, may finally reach the goal of "immune surveillance of malignancy."
Collapse
Affiliation(s)
- Su-hu Liu
- Department of Clinical Hematology, Xi'an Jiao Tong University, 5th West Avenue, Box 157, Xi'an 710-004, People's Republic of China.
| | | | | |
Collapse
|
45
|
Chattopadhyay S, Chakraborty NG. Continuous Presence of Th1 Conditions Is Necessary for Longer Lasting Tumor-Specific CTL Activity in Stimulation Cultures With PBL. Hum Immunol 2005; 66:884-91. [PMID: 16216672 DOI: 10.1016/j.humimm.2005.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2005] [Revised: 05/31/2005] [Accepted: 06/03/2005] [Indexed: 11/28/2022]
Abstract
The generation of tumor-associated, but self-antigen specific cytotoxic T lymphocytes (CTL) response is possible by vaccination even in patients at the advanced stages of the disease. The in vivo expansion of such CTLs is now the most important objective of the immunotherapy. In human melanoma, we show here that MART-1(27-35)-specific CTLs generated with purified CD8+ cells survive and maintain their activity longer in culture than those CTLs generated by using total peripheral blood lymphocytes (PBL) taken either from patients or from normal donors. When PBL are grown under Th1 conditioning the quantity and quality of CTL with total PBL are comparable with that of the CTLs generated with purified CD8+ cells. For patients either autologous melanoma tumor cells or MART-1(27-35) peptide pulsed autologous DC were used to generate CTL responses. For normal donors MART-1(27-35) peptide pulsed autologous DC were used. For both normal donors or patients, polarization of PBL with Th1 conditioning with interleukin (IL)-12 (250 U/ml) and anti IL-4 antibody 1 mug/ml for 7 days before CTL generation, induced better and longer living CTL response and prevented the expansion of CD4+ T cells that have downregulatory activity. We show that continuous presence of Th1 conditioning in cultures with total PBL generated significantly higher number of antigen-specific CTLs as detected by MART-1 HLA-A2 tetramer staining. The antigen specificity of such CTLs were determined by IFN-gamma secretion in response to target cells bearing the specific antigen. Our observations indicate that Th1 conditioning results in a longer lasting CTL response in vitro and points toward a newer approach for vaccine strategy.
Collapse
Affiliation(s)
- Subhasis Chattopadhyay
- Division of Hematology/Oncology, Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030-1628, USA
| | | |
Collapse
|
46
|
Abstract
Although myeloablative conditioning can cytoreduce or debulk malignancies, the curative antitumor effects of allogeneic hematopoietic stem cell transplantation (HCT) are mostly mediated by transplanted donor immune cells. A heightened awareness and appreciation of the immune-mediated anticancer effects that occur after allogeneic transplantation has led to the increasing use of reduced-intensity stem cell transplantation (RIST) approaches to treat advanced malignancies. The graft-versus-leukemia effects that occur against hematologic cancers after RIST have recently attracted oncologists to explore the therapeutic potential of allogeneic HCT for treatment-refractory solid tumors. Delayed tumor regression after RIST in a subset of patients with metastatic renal cell, breast, ovarian, pancreatic, and colon carcinoma has recently been reported, confirming the existence of a graft-versus-tumor effect in solid tumors. Advanced disease states, rapidly growing tumors, and accrual of patients with extremely short survival are factors that have been identified to limit the efficacy of allogeneic immunotherapy. This review discusses results of allogeneic HCT for solid tumors and the development of newer transplant strategies to optimize the potential of the graft-versus-tumor effect.
Collapse
Affiliation(s)
- Andreas Lundqvist
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-1652, USA
| | | |
Collapse
|
47
|
Im SH, Hueber A, Monticelli S, Kang KH, Rao A. Chromatin-level Regulation of the IL10 Gene in T Cells. J Biol Chem 2004; 279:46818-25. [PMID: 15319439 DOI: 10.1074/jbc.m401722200] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The immunoregulatory cytokine interleukin 10 (IL-10) modulates the function of diverse immune and non-immune cells. Here, we examine the chromatin structural changes associated with IL10 gene transcription by naive and differentiated murine T cells. Naive T cells lack DNase I hypersensitive (HS) sites in the vicinity of the IL10 gene, whereas differentiated T cells display a strong 3' constitutive HS site as well as several inducible sites. The majority of HS sites map to regions that are strongly conserved in sequence between mouse and human genomes. In committed Th1 cells, the mechanism of IL10 gene silencing is associated with the development of repressive histone modifications near the IL10 promoter and also near intronic hypersensitive regions of the IL10 gene. Our results constitute the first report of chromatin structural differences within the IL10 gene in differentiated Th1 and Th2 cells and emphasize the surprising diversity of mechanisms used to regulate cytokine gene expression at the chromatin level.
Collapse
Affiliation(s)
- Sin-Hyeog Im
- Department of Life Science, Gwangju Institute of Science and Technology, 1 Oryong-dong, Puk-ku, Gwangju 500-712, Korea.
| | | | | | | | | |
Collapse
|
48
|
Tejde A, Mathsson L, Ekdahl KN, Nilsson B, Rönnelid J. Immune complex-stimulated production of interleukin-12 in peripheral blood mononuclear cells is regulated by the complement system. Clin Exp Immunol 2004; 137:521-8. [PMID: 15320901 PMCID: PMC1809153 DOI: 10.1111/j.1365-2249.2004.02569.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Immune complexes (IC) can induce cytokine production in vitro. While immune aggregates (IA) consisting of heat-aggregated gamma globulin (HAGG) as model IC increased interleukin (IL)-10 levels in cell cultures with native human serum, IL-12p40/p70 production was inhibited. Three series of experiments suggested that the effects of IA on IL-12 production depended on a functionally intact complement system: (1) heat-inactivation of serum inverted the inhibitory effect of IA on IL-12p40/p70 production; (2) IA-induced IL-12p40 production in a C4 deficient serum was lowered by addition of C4; and (3) addition of the peptide compstatin, which blocks C3 activation, mimicked the effects of heat inactivation on IL-12p40 levels. Neutralization of IL-12 resulted in modestly increased IL-10 levels, while neutralization of IL-10 had no effects on IL-12p40 production. IA-induced production of IL-10 was partially blocked by anti-Fcgamma RII antibodies, whereas Fcgamma R or CR blockade had no effect on IL-12p40 production. IC and local or systemic complement activation characterize rheumatoid arthritis, systemic lupus erythematosus and many malignancies. Different and complement-dependent effects on the production of IL-10 and IL-12 can be of importance in these diseases, where control of the complement system might be a way to direct IC-induced cytokine production in either a type 1 or type 2 direction.
Collapse
Affiliation(s)
- A Tejde
- Unit of Clinical Immunology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | | | | | | | | |
Collapse
|
49
|
Fliss-Jaber L, Houissa-Kastally R, Bouzouita K, Khediri N, Khelifa R. Cytokine and immunoglobulin production by PWM-stimulated peripheral and tumor-infiltrating lymphocytes of undifferentiated nasopharyngeal carcinoma (NPC) patients. BMC Cancer 2004; 4:68. [PMID: 15450122 PMCID: PMC522818 DOI: 10.1186/1471-2407-4-68] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2004] [Accepted: 09/27/2004] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND Undifferentiated Nasopharyngeal Carcinoma (NPC) patients show a characteristic pattern of antibody responses to the Epstein-Barr virus (EBV) which is regularly associated with this tumor. However, no EBV-specific cytotoxic activity is detectable by the standard chromium-release assay at both peripheral and intratumoral levels. The mechanisms underlying this discrepancy between the humoral and cellular immune responses in NPC are still unknown, but might be related to an imbalance in immunoregulatory interleukin production. In this report, we investigated the ability of peripheral (PBL) and tumor- infiltrating (TIL) lymphocytes of undifferentiated NPC patients to produce in vitro three interleukins (IL-2, IL-6, IL-10) and three immunoglobulin isotypes (IgM, IgG, IgA). METHODS Lymphocytes from 17 patients and 17 controls were cultured in the presence of Pokeweed mitogen (PWM) for 12 days and their culture supernatants were tested for interleukins and immunoglobulins by specific enzyme-linked immunosorbent assays (ELISA). Data were analysed using Student's t-test and probability values below 5% were considered significant. RESULTS The data obtained indicated that TIL of NPC patients produced significantly more IL-2 (p = 0,0002), IL-10 (p = 0,020), IgM (p= 0,0003) and IgG (p < 0,0001) than their PBL. On the other hand, patients PBL produced significantly higher levels of IL-2 (p = 0,022), IL-10 (p = 0,016) and IgM (p = 0,004) than those of controls. No significant differences for IL-6 and IgA were observed. CONCLUSION Taken together, our data reinforce the possibility of an imbalance in immunoregulatory interleukin production in NPC patients. An increased ability to produce cytokines such as IL-10 may underlie the discrepancy between humoral and cellular immune responses characteristic of NPC.
Collapse
Affiliation(s)
- Lilia Fliss-Jaber
- Service des Laboratoires, Hôpital Habib Thameur, Tunis, Tunisia
- Laboratoire National de Contrôle des Médicaments, Tunis, Tunisia
| | | | - Kamel Bouzouita
- Laboratoire National de Contrôle des Médicaments, Tunis, Tunisia
| | - Naceur Khediri
- Service des Laboratoires, Hôpital Habib Thameur, Tunis, Tunisia
| | - Ridha Khelifa
- Service des Laboratoires, Hôpital Habib Thameur, Tunis, Tunisia
| |
Collapse
|
50
|
Matera L, Beltramo E, Martinuzzi E, Buttiglieri S. Effect of prolactin on carcinoembryonic antigen-specific cytotoxic T lymphocyte response induced by dendritic cells. Clin Exp Immunol 2004; 137:320-8. [PMID: 15270849 PMCID: PMC1809106 DOI: 10.1111/j.1365-2249.2004.02533.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The cytokine hormone prolactin (PRL) has been shown previously to modulate native cellular responses and maturation of antigen-presenting cells. Here we have addressed its effect on the antigen-specific response of cytotoxic T lymphocytes (CTL). CTL were generated from HLA-A2 lymphocytes after three rounds of stimulation with autologous dendritic cells loaded with HLA-A2-restricted carcinoembrionic antigen (CEA) Cap-1 (YLSGANLNL) peptide. Selected cultures were expanded on cytokine-supplemented feeder-layers, enriched for CD8+ lymphocytes and analysed for PRL-receptor (PRL-R) expression and PRL responsiveness. Resting CD8+ lymphocytes were negative for PRL-R, whereas antigen-activated CD8+ lymphocytes derived from long-term cultures were highly positive. Results of a 51Cr release assay showed CTL killing of CEA-loaded, but not unloaded, T2 cell line and the CEA-positive gastric carcinoma cell line KATO, but not of the CEA-negative T leukaemia cell line Jurkat. Interferon (IFN)-gamma release, evaluated in an ELISPOT assay against CEA-loaded T2, was enhanced (P < 0.05) by concentrations of PRL (12-25 ng/ml) very close to the physiological levels (6-20 ng/ml), but was decreased (P < 0.05) by high concentrations (200 ng/ml). Pre-incubation of the stimulators with the anti-MHC class I MoAb W6.32 induced a 40-60% decrease of the PRL-boosted IFN-gamma release, thus proving the MHC restriction of the lymphocyte response. Cytotoxicity against CEA-loaded T2 and KATO cell lines was also increased by 12-25 ng (P < 0.05) and decreased (P < 0.05) by 200 ng PRL. Pre-incubation of CTL with an antibody specific for the PRL-R almost completely abrogated this effect.
Collapse
Affiliation(s)
- L Matera
- Department of Internal Medicine, University of Turin, Turin, Italy.
| | | | | | | |
Collapse
|