1
|
Fe 3O 4 Nanoparticles in Combination with 5-FU Exert Antitumor Effects Superior to Those of the Active Drug in a Colon Cancer Cell Model. Pharmaceutics 2023; 15:pharmaceutics15010245. [PMID: 36678874 PMCID: PMC9865889 DOI: 10.3390/pharmaceutics15010245] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/13/2023] Open
Abstract
(1) Background: Colon cancer is one of the most common cancer types, and treatment options, unfortunately, do not continually improve the survival rate of patients. With the unprecedented development of nanotechnologies, nanomedicine has become a significant direction in cancer research. Indeed, chemotherapeutics with nanoparticles (NPs) in cancer treatment is an outstanding new treatment principle. (2) Methods: Fe3O4 NPs were synthesized and characterized. Caco-2 colon cancer cells were treated during two different periods (24 and 72 h) with Fe3O4 NPs (6 μg/mL), various concentrations of 5-FU (4−16 μg/mL), and Fe3O4 NPs in combination with 5-FU (4−16 μg/mL) (Fe3O4 NPs + 5-FU). (3) Results: The MTT assay showed that treating the cells with Fe3O4 NPs + 5-FU at 16 µg/mL for 24 or 72 h decreased cell viability and increased their LDH release (p < 0.05 and p < 0.01, respectively). Furthermore, at the same treatment concentrations, total antioxidant capacity (TAC) was decreased (p < 0.05 and p < 0.01, respectively), and total oxidant status (TOS) increased (p < 0.05 and p < 0.01, respectively). Moreover, after treatment with Fe3O4-NPs + 5-FU, the IL-10 gene was downregulated and PTEN gene expression was upregulated (p < 0.05 and p < 0.01, respectively) compared with those of the control. (4) Conclusions: Fe3O4 NPs exert a synergistic cytotoxic effect with 5-FU on Caco-2 cells at concentrations below the active drug threshold levels.
Collapse
|
2
|
Chen H, Zhang Y, Li L, Guo R, Shi X, Cao X. Effective CpG Delivery Using Zwitterion-Functionalized Dendrimer-Entrapped Gold Nanoparticles to Promote T Cell-Mediated Immunotherapy of Cancer Cells. BIOSENSORS 2022; 12:71. [PMID: 35200332 PMCID: PMC8869692 DOI: 10.3390/bios12020071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 11/24/2022]
Abstract
Recently, cell-based immunotherapy has become one of the most promising ways to completely eliminate cancer. The major challenge is to effectively promote a proper immune response to kill the cancer cells by activated T cells. This study investigated the effect of T cell-mediated immunotherapy trigged by Au DENPs-MPC (zwitterion 2-methacryloyloxyethyl phosphorylcholine (MPC)-functionalized dendrimer-entrapped gold nanoparticles) loading oli-godeoxynucleotides (ODN) of unmethylated cytosine guanine dinucleotide (CPG). Here, we first synthesized Au DENPs-MPC, evaluated their capability to compress and transfect CpG-ODN to bone marrow dendritic cells (BMDCs), and investigated the potential to use T cells stimulated by matured BMDCs to inhibit the growth of tumor cells. The developed Au DENPs-MPC could apparently reduce the toxicity of Au DENPs, and enhanced transfer CpG-ODN to the BMDCs for the maturation as demonstrated by the 44.41-48.53% increase in different surface maturation markers. The transwell experiments certificated that ex vivo activated T cells display excellent anti-tumor ability, which could effectively inhibit the growth of tumor cells. These results suggest that Au DENPs-MPC can deliver CpG-ODN efficiently to enhance the antigen presentation ability of BMDCs to activate T cells, indicating that T cells-based immunotherapy mediated by Au DENPs-MPC loaded with CpG-ODN may become the most promising treatment of cancer.
Collapse
Affiliation(s)
| | | | | | | | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; (H.C.); (Y.Z.); (L.L.); (R.G.)
| | - Xueyan Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; (H.C.); (Y.Z.); (L.L.); (R.G.)
| |
Collapse
|
3
|
Zhai Z, Wang Z, Jin M, Zhang K. Peripheral blood CD45RO+T cells is a predictor of the effectiveness of neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Medicine (Baltimore) 2021; 100:e26214. [PMID: 34160385 PMCID: PMC8238272 DOI: 10.1097/md.0000000000026214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 05/17/2021] [Indexed: 01/04/2023] Open
Abstract
To investigate the relationship between the changes in circulating CD45RO+T lymphocyte subsets following neoadjuvant therapy for rectal cancer in patients with locally advanced rectal cancer.The clinicopathological data of 185 patients with rectal cancer who received neoadjuvant therapy in the General Surgery Department of Beijing Chaoyang Hospital affiliated to Capital Medical University from June 2015 to June 2017 were analyzed. Venous blood samples were collected 1 week before neoadjuvant therapy and 1 week before surgery, and the expression of CD45RO+T was detected by flow cytometry. The receiver operating characteristic curve analysis was used to determine the optimal cut-off point of CD45RO+ratio. Log-rank test and multivariate Cox regression were used to analyze the overall survival rate (OS) and disease-free survival rate (DFS) associated with CD45RO+ratio.Circulating CD45RO+ratio of 1.07 was determined as the optimal cut-off point and CD45RO+ratio-high was associated with lower tumor regression grade grading (P = .031), T stage (P = .001), and tumor node metastasis (TNM) stage (P = .012). The 3-year DFS and OS rate in the CD45RO+ratio-high group was significantly higher than that in the CD45RO+ratio-low group (89.2% vs 60.1%, P<.001; 94.4% vs 73.2%, P<.001). The multivariate Cox analysis revealed that elevated CD45RO+ratio was an independent factor for better DFS (OR, 0.339; 95% CI, 0.153-0.752; P = .008) and OS (OR, 0.244; 95% CI,0.082-0.726; P = .011).Circulating CD45RO+ratio could predict the tumor regression grade of neoadjuvant therapy for rectal cancer, as well as long-term prognosis. These findings could be used to stratify patients and develop alternative strategies for adjuvant therapy.
Collapse
Affiliation(s)
| | | | - Mulan Jin
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Kunning Zhang
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Béraud E, Collignon A, Franceschi C, Olive D, Lombardo D, Mas E. Investigation of a new tumor-associated glycosylated antigen as target for dendritic cell vaccination in pancreatic cancer. Oncoimmunology 2021; 1:56-61. [PMID: 22720212 PMCID: PMC3376954 DOI: 10.4161/onci.1.1.18459] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Glycoproteins, as valuable targets for dendritic cell (DC)-vaccination in cancers, remain an open question. Glycosylated structures, which are aberrantly modified during cancerisation, impact positively or negatively on glycoprotein immunogenicity. Here is presented an oncofetal glycovariant of bile-salt-dependent-lipase, expressed on human tumoral pancreas and efficiently processed by DC's, inducing T-lymphocyte activation.
Collapse
Affiliation(s)
- Evelyne Béraud
- INSERM; Marseille, France; Aix-Marseille Univ ; Centre de Recherche en Oncologie biologique et Oncopharmacologie; Marseille, France
| | | | | | | | | | | |
Collapse
|
5
|
Gupta A, Sharma A, von Boehmer L, Surace L, Knuth A, van den Broek M. Radiotherapy supports protective tumor-specific immunity. Oncoimmunology 2021; 1:1610-1611. [PMID: 23264910 PMCID: PMC3525619 DOI: 10.4161/onci.21478] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Radiotherapy is an important therapeutic option for the treatment of cancer. Growing evidence indicates that, besides inducing an irreversible DNA damage, radiotherapy promotes tumor-specific immune response, which significantly contribute to therapeutic efficacy. We postulate that radiotherapy activates tumor-associated dendritic cells, thus changing the tolerogenic tumor environment into an immunogenic one.
Collapse
Affiliation(s)
- Anurag Gupta
- Department of Oncology; University Hospital Zurich; Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
6
|
Badr CE, Silver DJ, Siebzehnrubl FA, Deleyrolle LP. Metabolic heterogeneity and adaptability in brain tumors. Cell Mol Life Sci 2020; 77:5101-5119. [PMID: 32506168 PMCID: PMC8272080 DOI: 10.1007/s00018-020-03569-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/18/2020] [Accepted: 05/28/2020] [Indexed: 12/19/2022]
Abstract
The metabolic complexity and flexibility commonly observed in brain tumors, especially glioblastoma, is fundamental for their development and progression. The ability of tumor cells to modify their genetic landscape and adapt metabolically, subverts therapeutic efficacy, and inevitably instigates therapeutic resistance. To overcome these challenges and develop effective therapeutic strategies targeting essential metabolic processes, it is necessary to identify the mechanisms underlying heterogeneity and define metabolic preferences and liabilities of malignant cells. In this review, we will discuss metabolic diversity in brain cancer and highlight the role of cancer stem cells in regulating metabolic heterogeneity. We will also highlight potential therapeutic modalities targeting metabolic vulnerabilities and examine how intercellular metabolic signaling can shape the tumor microenvironment.
Collapse
Affiliation(s)
- Christian E Badr
- Neuro-Oncology Division, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Neuroscience Program, Harvard Medical School, Boston, MA, USA
| | - Daniel J Silver
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Florian A Siebzehnrubl
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, CF24 4HQ, UK
| | - Loic P Deleyrolle
- Lillian S. Wells Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
7
|
Yin X, Zeng W, Wu B, Wang L, Wang Z, Tian H, Wang L, Jiang Y, Clay R, Wei X, Qin Y, Zhang F, Zhang C, Jin L, Liang W. PPARα Inhibition Overcomes Tumor-Derived Exosomal Lipid-Induced Dendritic Cell Dysfunction. Cell Rep 2020; 33:108278. [PMID: 33086073 PMCID: PMC7771208 DOI: 10.1016/j.celrep.2020.108278] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 06/14/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) orchestrate the initiation, programming, and regulation of anti-tumor immune responses. Emerging evidence indicates that the tumor microenvironment (TME) induces immune dysfunctional tumor-infiltrating DCs (TIDCs), characterized with both increased intracellular lipid content and mitochondrial respiration. The underlying mechanism, however, remains largely unclear. Here, we report that fatty acid-carrying tumor-derived exosomes (TDEs) induce immune dysfunctional DCs to promote immune evasion. Mechanistically, peroxisome proliferator activated receptor (PPAR) α responds to the fatty acids delivered by TDEs, resulting in excess lipid droplet biogenesis and enhanced fatty acid oxidation (FAO), culminating in a metabolic shift toward mitochondrial oxidative phosphorylation, which drives DC immune dysfunction. Genetic depletion or pharmacologic inhibition of PPARα effectively attenuates TDE-induced DC-based immune dysfunction and enhances the efficacy of immunotherapy. This work uncovers a role for TDE-mediated immune modulation in DCs and reveals that PPARα lies at the center of metabolic-immune regulation of DCs, suggesting a potential immunotherapeutic target. Yin et al. reveal that tumor-derived exosomes (TDEs), as fatty acid carriers, induce a metabolic shift toward oxidative phosphorylation, driving DC immune dysfunction. Transcriptomic analysis identifies PPARα as the fatty acid sensor mediating the immunosuppressive effects of TDEs on DCs. PPARα blockade effectively restores DC function and enhances the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Xiaozhe Yin
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100864, China
| | - Wenfeng Zeng
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100864, China.
| | - Bowen Wu
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100864, China; Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Luoyang Wang
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Zihao Wang
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100864, China
| | - Hongjian Tian
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100864, China
| | - Luyao Wang
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100864, China
| | - Yunhan Jiang
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL 32610-3033, USA
| | - Ryan Clay
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL 32610-3033, USA
| | - Xiuli Wei
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Qin
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fayun Zhang
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Chunling Zhang
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Lingtao Jin
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL 32610-3033, USA.
| | - Wei Liang
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100864, China.
| |
Collapse
|
8
|
Chen H, Fan Y, Hao X, Yang C, Peng Y, Guo R, Shi X, Cao X. Adoptive cellular immunotherapy of tumors via effective CpG delivery to dendritic cells using dendrimer-entrapped gold nanoparticles as a gene vector. J Mater Chem B 2020; 8:5052-5063. [DOI: 10.1039/d0tb00678e] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PEGylated Au DENPs ({(Au0)25-G5·NH2-mPEG20}) are synthesized and used as a novel nonviral vector to deliver CpG to mature BMDCs for the subsequent activation of T cells for adoptive tumor immunotherapy.
Collapse
Affiliation(s)
- Huan Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
- Shanghai 201620
| | - Yu Fan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
- Shanghai 201620
| | - Xinxin Hao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
- Shanghai 201620
| | - Chao Yang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
- Shanghai 201620
| | - Yucheng Peng
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
- Shanghai 201620
| | - Rui Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
- Shanghai 201620
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
- Shanghai 201620
| | - Xueyan Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
- Shanghai 201620
| |
Collapse
|
9
|
Wang Y, Zhang Q, Chen Y, Liang CL, Liu H, Qiu F, Dai Z. Antitumor effects of immunity-enhancing traditional Chinese medicine. Biomed Pharmacother 2019; 121:109570. [PMID: 31710893 DOI: 10.1016/j.biopha.2019.109570] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/07/2019] [Accepted: 10/20/2019] [Indexed: 02/06/2023] Open
Abstract
Traditional Chinese Medicine (TCM) has been traditionally used to treat patients with cancers in China. It not only alleviates the symptoms of tumor patients and improves their quality of life, but also controls the size of tumors and prolongs the survival of tumor patients. While some herbs of TCM may exert therapeutic effects by directly targeting cancer cells or reducing side effects caused by antitumor drugs, others can control tumor growth and metastasis via enhancing antitumor immunity. In particular, TCM can exert antitumor effects by upregulating immune responses even in immunosuppressive tumor microenvironment. For instance, it reduces the number of M2-type macrophages and Treg cells in the tumor tissue. Although extensive reviews on directly killing cancer cells by TCM have been conducted, a review of anticancer activity of TCM solely based on its immunity-enhancing capacity is unusual. This review will summarize research progress of antitumor TCM that regulates the immune system, including both innate immunity, such as macrophages, dendritic cells, natural killer cells and MDSCs, and adaptive immunity, including CD4+/CD8+ T lymphocytes, regulatory T cells (Tregs) and B cells. As cancer immunotherapy has recently achieved certain success, it is expected that the clinical applications of immunity-enhancing TCM or traditional medicine for treating various cancer patients will be expanded. Further studies on the mechanisms by which TCM regulates immunity will provide new insights into how TCM controls tumor growth and metastasis, and may help improve its therapeutic effects on various cancers in clinic.
Collapse
Affiliation(s)
- Yeshu Wang
- Section of Immunology & Joint Immunology Program, the Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Qunfang Zhang
- Section of Immunology & Joint Immunology Program, the Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Yuchao Chen
- Section of Immunology & Joint Immunology Program, the Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Chun-Ling Liang
- Section of Immunology & Joint Immunology Program, the Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Huazhen Liu
- Section of Immunology & Joint Immunology Program, the Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Feifei Qiu
- Section of Immunology & Joint Immunology Program, the Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Zhenhua Dai
- Section of Immunology & Joint Immunology Program, the Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
10
|
Li Y, Wan YY, Zhu B. Immune Cell Metabolism in Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1011:163-196. [PMID: 28875490 DOI: 10.1007/978-94-024-1170-6_5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tumor microenvironment (TME) is composed of tumor cells, immune cells, cytokines, extracellular matrix, etc. The immune system and the metabolisms of glucose, lipids, amino acids, and nucleotides are integrated in the tumorigenesis and development. Cancer cells and immune cells show metabolic reprogramming in the TME, which intimately links immune cell functions and edits tumor immunology. Recent findings in immune cell metabolism hold the promising possibilities toward clinical therapeutics for treating cancer. This chapter introduces the updated understandings of metabolic reprogramming of immune cells in the TME and suggests new directions in manipulation of immune responses for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Yongsheng Li
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yisong Y Wan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
11
|
Li J, Huang S, Zhou Z, Lin W, Chen S, Chen M, Ye Y. Exosomes derived from rAAV/AFP-transfected dendritic cells elicit specific T cell-mediated immune responses against hepatocellular carcinoma. Cancer Manag Res 2018; 10:4945-4957. [PMID: 30464595 PMCID: PMC6214341 DOI: 10.2147/cmar.s178326] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Dendritic cell (DC)-derived exosomes (Dexs) have been proved to induce and enhance antigen-specific T cell responses in vivo, and previous clinical trials have shown the feasibility and safety of Dexs in multiple human cancers. However, there is little knowledge on the efficacy of Dexs against hepatocellular carcinoma (HCC) until now. Methods In this study, human peripheral blood-derived DCs were loaded with recombinant adeno-associated viral vector (rAAV)-carrying alpha-fetoprotein (AFP) gene (rAAV/AFP), and high-purity Dexs were generated. Then naive T cells were stimulated with Dexs to investigate the specific T cell-mediated immune responses against HCC. Results Our findings showed that Dexs were effective to stimulate naive T cell proliferation and induce T cell activation to become antigen-specific cytotoxic T lymphocytes (CTLs), thereby exhibiting antitumor immune responses against HCC. In addition, Dex-sensitized DC precursors seemed more effective to trigger major histocompatibility complex class I (MHC I)-restricted CTL response and allow DCs to make full use of the minor antigen peptides, thereby maximally activating specific immune responses against HCC. Conclusion It is concluded that Dexs, which combine the advantages of DCs and cell-free vectors, are promising to completely, or at least in part, replace mature DCs (mDCs) to function as cancer vaccines or natural antitumor adjuvant.
Collapse
Affiliation(s)
- Jieyu Li
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China, .,Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350014, China, .,Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China,
| | - Shenglan Huang
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China,
| | - Zhifeng Zhou
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350014, China, .,Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China,
| | - Wansong Lin
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350014, China, .,Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China,
| | - Shuping Chen
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350014, China, .,Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China,
| | - Mingshui Chen
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350014, China, .,Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China,
| | - Yunbin Ye
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China, .,Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350014, China, .,Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China,
| |
Collapse
|
12
|
Kim JW, Kane JR, Panek WK, Young JS, Rashidi A, Yu D, Kanojia D, Hasan T, Miska J, Gómez-Lim MA, Ulasov IV, Balyasnikova IV, Ahmed AU, Wainwright DA, Lesniak MS. A Dendritic Cell-Targeted Adenoviral Vector Facilitates Adaptive Immune Response Against Human Glioma Antigen (CMV-IE) and Prolongs Survival in a Human Glioma Tumor Model. Neurotherapeutics 2018; 15:1127-1138. [PMID: 30027430 PMCID: PMC6277295 DOI: 10.1007/s13311-018-0650-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Antitumor immunotherapeutic strategies represent an especially promising set of approaches with rapid translational potential considering the dismal clinical context of high-grade gliomas. Dendritic cells (DCs) are the body's most professional antigen-presenting cells, able to recruit and activate T cells to stimulate an adaptive immune response. In this regard, specific loading of tumor-specific antigen onto dendritic cells potentially represents one of the most advanced strategies to achieve effective antitumor immunization. In this study, we developed a DC-specific adenoviral (Ad) vector, named Ad5scFvDEC205FF, targeting the DC surface receptor, DEC205. In vitro analysis shows that 60% of DCs was infected by this vector while the infectivity of other control adenoviral vectors was less than 10%, demonstrating superior infectivity on DCs. Moreover, an average of 14% of DCs were infected by Ad5scFvDEC205FF-GFP, while less than 3% of non-DCs were infected following in vivo administration, demonstrating highly selective in vivo DC infection. Importantly, vaccination with this vehicle expressing human glioma-specific antigen, Ad5scFvDEC205FF-CMV-IE, shows a prolonged survival benefit in GL261CMV-IE-implanted murine glioma models (p < 0.0007). Furthermore, when rechallenged, cancerous cells were completely rejected. In conclusion, our novel, viral-mediated, DC-based immunization approach has the significant therapeutic potential for patients with high-grade gliomas.
Collapse
Affiliation(s)
- Julius W Kim
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - J Robert Kane
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Wojciech K Panek
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Jacob S Young
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Aida Rashidi
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Dou Yu
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Deepak Kanojia
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Tanwir Hasan
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Jason Miska
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Miguel A Gómez-Lim
- Departamento de Ingeniería Genética, CINVESTAV Irapuato, Km 9.6 Libramiento Norte Carretera Irapuato-León, 36821, Irapuato, Guanajuato, Mexico
| | - Ilya V Ulasov
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Irina V Balyasnikova
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Atique U Ahmed
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA.
| |
Collapse
|
13
|
Wu Z, Zhang J, Cai Y, Deng R, Yang L, Li J, Deng Y. Reduction of circulating lymphocyte count is a predictor of good tumor response after neoadjuvant treatment for rectal cancer. Medicine (Baltimore) 2018; 97:e11435. [PMID: 30235653 PMCID: PMC6160071 DOI: 10.1097/md.0000000000011435] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Systemic inflammatory indices are correlated with poor prognosis in cancer patients. The presence of lymphocytes in and around the tumor tissue is a predictor in rectal cancer. We aimed to explore the mechanism underlying the changes in circulating lymphocyte during neoadjuvant therapy and the way in which the count correlates with tumor response.Around 307 patients from FOWARC trial and 64 patients from FORTUNE trial were included in the training and validation group. Circulating lymphocyte count was recorded before neoadjuvant therapy and before rectal surgery. Receiver operating characteristic (ROC) curve analysis was performed to determine the optimal cut-off value of the reduction of lymphocytes. A logistic regression model was obtained in multivariate analysis.The blood absolute number of lymphocyte before and after therapy had no correlation with tumor response. However, total lymphocyte count (TLC) reduction was significantly higher in good response group (39.81% vs 33.31% P = .032) in the FOWARC cohort. The optimal cut-off value for TLC was 24.96%. Age, tumor length, and TLC reduction (P = .005, OR = 2.009, 95%CI 1.240-3.254) were significant factors for tumor regression in multivariate analysis. In the FORTUNE cohort, TLC reduction was the only significant factor for tumor regression in both univariate (P = .032, OR = 3.434, 95%CI 1.111-10.614) and multivariate analysis (P = .046, OR = 3.361, 95%CI 1.024-11.035).Circulating lymphocyte count decreases during neoadjuvant therapy for locally advanced rectal cancer, and it is associated with better tumor regression. It may be involved in the immune response provoked by radiotherapy and chemotherapy.
Collapse
|
14
|
Dillinger B, Ahmadi-Erber S, Lau M, Hoelzl MA, Erhart F, Juergens B, Fuchs D, Heitger A, Ladisch S, Dohnal AM. IFN-γ and tumor gangliosides: Implications for the tumor microenvironment. Cell Immunol 2018; 325:33-40. [PMID: 29402391 DOI: 10.1016/j.cellimm.2018.01.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/16/2018] [Accepted: 01/22/2018] [Indexed: 10/18/2022]
Abstract
Gangliosides shed by tumors into their microenvironment (TME) are immunoinhibitory. Interferon-γ (IFN-γ) may boost antitumor immune responses. Thus we wondered whether IFN-γ would counteract tumor ganglioside-mediated immune suppression. To test this hypothesis, we exposed human monocyte-derived LPS-activated dendritic cells (DC) to IFN-γ and to a highly purified ganglioside, GD1a. DC ganglioside exposure decreased TLR-dependent p38 signaling, explaining the previously observed ganglioside-induced down-modulation of pro-inflammatory surface markers and cytokines. Strikingly, while increasing LPS-dependent DC responses, IFN-γ unexpectedly did not counteract the inhibitory effects of GD1a. Rather, induction of indoleamine 2,3-dioxygenase (IDO1), and expression of STAT1/IRF-1 and programmed cell death ligand (PD-L1), indicated that the immunoinhibitory, not an immune stimulatory, IFN-γ-signaling axis, was active. The combination, IFN-γ and DC ganglioside enrichment, markedly impaired DC stimulatory potential of CD8+ T-cells. We suggest that gangliosides and IFN-γ may act in concert as immunosuppressive mediators in the TME, possibly promoting tumor progression.
Collapse
Affiliation(s)
- Barbara Dillinger
- Tumor Immunology, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung e.V., Zimmermannplatz 10, Vienna, Austria
| | - Sarah Ahmadi-Erber
- Tumor Immunology, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung e.V., Zimmermannplatz 10, Vienna, Austria
| | - Manuel Lau
- Tumor Immunology, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung e.V., Zimmermannplatz 10, Vienna, Austria
| | - Markus A Hoelzl
- Tumor Immunology, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung e.V., Zimmermannplatz 10, Vienna, Austria
| | - Friedrich Erhart
- Tumor Immunology, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung e.V., Zimmermannplatz 10, Vienna, Austria
| | - Birgit Juergens
- Tumor Immunology, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung e.V., Zimmermannplatz 10, Vienna, Austria
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innrain 80, Innsbruck, Austria
| | - Andreas Heitger
- Tumor Immunology, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung e.V., Zimmermannplatz 10, Vienna, Austria
| | - Stephan Ladisch
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Medical Center 111 Michigan Avenue, N.W., Washington, DC, USA.
| | - Alexander M Dohnal
- Tumor Immunology, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung e.V., Zimmermannplatz 10, Vienna, Austria.
| |
Collapse
|
15
|
Lee JH, Tak WY, Lee Y, Heo MK, Song JS, Kim HY, Park SY, Bae SH, Lee JH, Heo J, Kim KH, Bae YS, Kim YJ. Adjuvant immunotherapy with autologous dendritic cells for hepatocellular carcinoma, randomized phase II study. Oncoimmunology 2017; 6:e1328335. [PMID: 28811965 DOI: 10.1080/2162402x.2017.1328335] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/04/2017] [Accepted: 05/05/2017] [Indexed: 02/06/2023] Open
Abstract
Our previous phase I/IIA study showed that autologous dendritic cells (DCs) pulsed with tumor-associated antigens are well tolerated in patients with hepatocellular carcinoma (HCC). In this randomized, multicenter, open-label, phase II trial, we investigated the efficacy and safety of this DC-based adjuvant immunotherapy with 156 patients, who treated for HCC with no evidence of residual tumor after standard treatment modalities. Patients were randomly assigned to immunotherapy (n = 77; injection of 3 × 107 DC cells, six times over 14 weeks) or control (n = 79; no treatment). The primary end point was recurrence-free survival (RFS), and the secondary endpoints were immune response and safety. The RFS between the immunotherapy and control groups was not significantly different (hazard ratio [HR], 0.97; 95% confidence interval [CI], 0.60-1.56; p = 0.90). However, post-hoc subgroup analyses revealed that DC immunotherapy significantly reduced the risk of tumor recurrence of non-radiofrequency ablation (non-RFA) group patients (n = 83, HR, 0.49; 95% CI, 0.26-0.94; p = 0.03), whereas unexpectedly increased the risk of recurrence in RFA group (n = 61, p = 0.01). Tumor-specific immune responses were significantly enhanced (both p < 0.01) in the immunotherapy group. Baseline serum interleukin (IL)-15 was statistically correlated with RFS prolongation (HR, 0.16; 95% CI, 0.03-1.58; p = 0.001) within the immunotherapy groups. Overall adverse events were more frequent in the immunotherapy group (p < 0.001) but were mainly mild to moderate in severity. In conclusion, adjuvant immunotherapy with DC vaccine reduces the risk of tumor recurrence in HCC patients who underwent standard treatment modalities other than RFA. Baseline IL-15 might be a candidate biomarker for DC-based HCC immunotherapy.
Collapse
Affiliation(s)
- Jeong-Hoon Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Won Young Tak
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Yoon Lee
- Department of Biological Science, Sungkyunkwan University, Suwon, Korea.,JW CreaGene Research Institute, JW CreaGene Inc., Seongnam-si, Gyeonggi-do, Korea
| | - Min-Kyu Heo
- JW CreaGene Research Institute, JW CreaGene Inc., Seongnam-si, Gyeonggi-do, Korea
| | - Jae-Sung Song
- JW CreaGene Research Institute, JW CreaGene Inc., Seongnam-si, Gyeonggi-do, Korea
| | - Hak-Yeop Kim
- JW CreaGene Research Institute, JW CreaGene Inc., Seongnam-si, Gyeonggi-do, Korea
| | - Soo Young Park
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Si Hyun Bae
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
| | - Joon Hyeok Lee
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeong Heo
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Ki-Hwan Kim
- JW CreaGene Research Institute, JW CreaGene Inc., Seongnam-si, Gyeonggi-do, Korea
| | - Yong-Soo Bae
- Department of Biological Science, Sungkyunkwan University, Suwon, Korea.,JW CreaGene Research Institute, JW CreaGene Inc., Seongnam-si, Gyeonggi-do, Korea
| | - Yoon Jun Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
16
|
Dickinson BL. Unraveling the immunopathogenesis of glomerular disease. Clin Immunol 2016; 169:89-97. [PMID: 27373970 DOI: 10.1016/j.clim.2016.06.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 06/26/2016] [Accepted: 06/28/2016] [Indexed: 02/08/2023]
Abstract
Immune-mediated damage to glomerular structures is largely responsible for the pathology associated with the majority of glomerular diseases. Therefore, a detailed understanding of the basic immune mechanisms responsible for glomerular damage is needed to inform the design of novel intervention strategies. Glomerular injury of immune origin is complex and involves both inflammatory and non-inflammatory processes driven by elements of the innate and adaptive immune system. This review summarizes the basic immune mechanisms that cause glomerular injury leading to the nephritic and nephrotic syndromes. A major focus of the review is to highlight the mechanisms by which antibodies cause glomerular injury through their interactions with glomerular cells, complement proteins, phagocytes bearing complement and Fcγ receptors, and dendritic cells expressing the neonatal receptor for IgG, FcRn.
Collapse
Affiliation(s)
- Bonny L Dickinson
- Department of Biomedical Science, Western Michigan University Homer Stryker MD School of Medicine, 1000 Oakland Drive, Kalamazoo, MI 49008, United States.
| |
Collapse
|
17
|
Takahashi H, Sakakura K, Mito I, Ida S, Chikamatsu K. Dynamic changes in immune cell profile in head and neck squamous cell carcinoma: Immunomodulatory effects of chemotherapy. Cancer Sci 2016; 107:1065-71. [PMID: 27228557 PMCID: PMC4982580 DOI: 10.1111/cas.12976] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/19/2016] [Accepted: 05/24/2016] [Indexed: 12/21/2022] Open
Abstract
Tumor cells have evolved sophisticated means of escape from the host immune system. To date, several important immunological phenomena have been revealed in peripheral blood as well as within tumors. In the present study, we first investigated the proportion and activation status of peripheral immune regulatory cells and CD8(+) T-cell subsets in patients with head and neck squamous cell carcinoma (HNSCC) using a multicolor flow cytometer, and then evaluated how therapy with docetaxel, cisplatin, and 5-fluorouracil modulated the immune cell profile in peripheral blood. The proportion of naïve T cells was lower and that of effector memory T cells (TEM ) was higher in HNSCC patients than in healthy donors. Moreover, the proportions of activated TEM cells and effector T cells (TEFF ) were dramatically increased in patients with advanced stage disease. The proportion of regulatory T cells and CD14(+) HLA-DR(-) myeloid-derived suppressor cells was elevated in HNSCC patients. Of note, after therapy, in addition to the transient reduction in immune regulatory cells, decreases in central memory T cells and increases in TEFF cells were observed among CD8(+) T-cell subsets, suggesting differentiation from central memory T cells into TEFF cells. Our results suggested that, despite the immunosuppressive status in HNSCC patients, tumor-specific immune responses mediated by CD8(+) T cells might be induced and maintained. Moreover, chemotherapy can trigger not only a transient reduction in immune regulatory cells but also further activation of CD8(+) T cells.
Collapse
Affiliation(s)
- Hideyuki Takahashi
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Koichi Sakakura
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ikko Mito
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Shota Ida
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kazuaki Chikamatsu
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
18
|
Biswas SK. Metabolic Reprogramming of Immune Cells in Cancer Progression. Immunity 2016; 43:435-49. [PMID: 26377897 DOI: 10.1016/j.immuni.2015.09.001] [Citation(s) in RCA: 459] [Impact Index Per Article: 57.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 08/25/2015] [Accepted: 08/31/2015] [Indexed: 11/25/2022]
Abstract
Immune cells play a key role in host defense against infection and cancer. Upon encountering danger signals, these cells undergo activation leading to a modulation in their immune functions. However, recent studies reveal that immune cells upon activation also show distinct metabolic changes that impact their immune functions. Such metabolic reprogramming and its functional effects are well known for cancer cells. Given that immune cells have emerged as crucial players in cancer progression, it is important to understand whether immune cells also undergo metabolic reprogramming in tumors and how this might affect their contribution in cancer progression. This emerging aspect of tumor-associated immune cells is reviewed here, discussing metabolic reprogramming of different immune cell types, the key pathways involved, and its impact on tumor progression.
Collapse
Affiliation(s)
- Subhra K Biswas
- Singapore Immunology Network (SIgN), Agency for Science, Technology & Research (A(∗)STAR), #04-06 Immunos, 8A Biomedical Grove, Singapore 138648, Singapore.
| |
Collapse
|
19
|
Joncker NT, Bettini S, Boulet D, Guiraud M, Guerder S. The site of tumor development determines immunogenicity via temporal mobilization of antigen-laden dendritic cells in draining lymph nodes. Eur J Immunol 2015; 46:609-18. [PMID: 26626316 DOI: 10.1002/eji.201545797] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 10/27/2015] [Accepted: 11/27/2015] [Indexed: 01/06/2023]
Abstract
The elimination of solid tumors largely depends on effective T-cell priming by dendritic cells (DCs). For decades, studies focusing on antitumoral immune responses have been performed with tumors transplanted subcutaneously (s.c.). These studies however do not take into account the heterogeneous tissue distribution and functionality of the different DC subsets. Given the crucial role of DCs in inducing protective immune response, we postulated that the anatomic location of tumor development may greatly impact tumor immunogenicity. We therefore implanted tumor cells either in the DC-rich dermis environment or in the s.c. tissue that mainly contains macrophages and monocytes. We showed that intradermal (i.d.), but not s.c. tumors are rapidly rejected in a T-cell-dependent manner and induce protective T-cell responses. The rejection of i.d. tumors correlates with rapid recruitment of dermal DCs presenting the tumor antigen to both CD4 and CD8 T cells in the draining lymph nodes (dLNs). The same DC subsets were mobilized upon s.c. tumor transplantation but with delayed kinetics. Altogether, our results show that the anatomical site of tumor development influences tumor immunogenicity, notably by controlling the kinetics of DC mobilization in the draining LNs.
Collapse
Affiliation(s)
- Nathalie T Joncker
- Centre de Physiopathologie de Toulouse Purpan, Toulouse, France.,INSERM, U1043, Toulouse, France.,CNRS, UMR5282, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Sarah Bettini
- Centre de Physiopathologie de Toulouse Purpan, Toulouse, France.,INSERM, U1043, Toulouse, France.,CNRS, UMR5282, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Delphine Boulet
- Centre de Physiopathologie de Toulouse Purpan, Toulouse, France.,INSERM, U1043, Toulouse, France.,CNRS, UMR5282, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Martine Guiraud
- Centre de Physiopathologie de Toulouse Purpan, Toulouse, France.,INSERM, U1043, Toulouse, France.,CNRS, UMR5282, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Sylvie Guerder
- Centre de Physiopathologie de Toulouse Purpan, Toulouse, France.,INSERM, U1043, Toulouse, France.,CNRS, UMR5282, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| |
Collapse
|
20
|
Vermi W, Micheletti A, Lonardi S, Costantini C, Calzetti F, Nascimbeni R, Bugatti M, Codazzi M, Pinter PC, Schäkel K, Tamassia N, Cassatella MA. slanDCs selectively accumulate in carcinoma-draining lymph nodes and marginate metastatic cells. Nat Commun 2015; 5:3029. [PMID: 24398631 DOI: 10.1038/ncomms4029] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 11/27/2013] [Indexed: 01/01/2023] Open
Abstract
Dendritic cells (DCs) initiate adaptive immune responses to cancer cells by activating naive T lymphocytes. 6-sulfo LacNAc(+) DCs (slanDCs) represent a distinct population of circulating and tissue proinflammatory DCs, whose role in cancer immune surveillance is unknown. Herein, by screening a large set of clinical samples, we demonstrate accumulation of slanDCs in metastatic tumour-draining lymph nodes (M-TDLN) from carcinoma patients. Remarkably, slanDCs are absent at the primary carcinoma site, while their selective nodal recruitment follows the arrival of cancer cells to M-TDLN. slanDCs surround metastatic carcinoma deposits in close proximity to dead cells and efficiently phagocytose tumour cells. In colon carcinoma patients, the contingent of circulating slanDCs remains intact and competent in terms of IL-12p70 and tumour necrosis factor alpha production, induction of T-cell proliferation and migratory capacity to a set of chemokines produced in M-TDLN. We conclude that activated slanDCs represent previously unrecognized players of nodal immune responses to cancer cells.
Collapse
Affiliation(s)
- William Vermi
- 1] Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy [2] Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Alessandra Micheletti
- 1] Section of General Pathology, Department of Pathology and Diagnostics, University of Verona, Verona 37134, Italy [2]
| | - Silvia Lonardi
- 1] Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy [2]
| | - Claudio Costantini
- Section of General Pathology, Department of Pathology and Diagnostics, University of Verona, Verona 37134, Italy
| | - Federica Calzetti
- Section of General Pathology, Department of Pathology and Diagnostics, University of Verona, Verona 37134, Italy
| | - Riccardo Nascimbeni
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Mattia Bugatti
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Manuela Codazzi
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Patrick C Pinter
- Section of Otorhinolaryngology, Department of Surgery, University of Verona, Verona 37134, Italy
| | - Knut Schäkel
- Department of Dermatology, University Hospital Heidelberg, 69115 Heidelberg, Germany
| | - Nicola Tamassia
- Section of General Pathology, Department of Pathology and Diagnostics, University of Verona, Verona 37134, Italy
| | - Marco A Cassatella
- Section of General Pathology, Department of Pathology and Diagnostics, University of Verona, Verona 37134, Italy
| |
Collapse
|
21
|
Matsuhashi T, Shimizu M, Negishi Y, Takeshita T, Takahashi H. A low, non-toxic dose of paclitaxel can prevent dendritic cell-precursors from becoming tolerogenic dendritic cells with impaired functions. Biomed Res 2015; 35:369-80. [PMID: 25743343 DOI: 10.2220/biomedres.35.369] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Tumor infiltrating dendritic cells (TIDCs) are thought to be potent antigen-presenting cells able to activate tumor-specific cytotoxic T lymphocytes (CTLs) or tolerogenic DCs that suppress immune reaction against tumors to escape. We have recently reported that majority of these TIDCs were DEC-205(+) DCs having a cross-presenting ability of captured tumor antigens to CD8(+) T cells via class I MHC (MHC-I) molecules, nevertheless, when the TIDCs expressed down-modulated costimulatory molecules, such as CD80 and CD86, they will inhibit the priming and activation of immune effectors (Immunol. Cell Biol., 91: 545-555, 2013). Here, we show that DC-precursors (preDCs) but not the established DCs become tolerogenic DCs expressing down-regulated costimulatory molecules having low responsiveness to LPS or tumor cells, when exposed to soluble factors released from the encountered ovarian tumors in the early phase of their development. However, we found that we could reduce the secretion of those soluble factors with a low, nontoxic concentration of paclitaxel (PTX) and we could stop the preDCs to be tolerogenic DCs and maintain DC functions. These findings indicate that we could prevent the induction of tolerogenic DCs from preDCs by using low, non-toxic doses of anti-cancer drugs to establish DCs that effectively elicit tumor-specific CTLs.
Collapse
Affiliation(s)
- Tomohiko Matsuhashi
- Department of Microbiology and Immunology, Department of Obstetrics and Gynecology, Nippon Medical School, Tokyo 113-8602, Japan
| | | | | | | | | |
Collapse
|
22
|
Breitbach CJ, Parato K, Burke J, Hwang TH, Bell JC, Kirn DH. Pexa-Vec double agent engineered vaccinia: oncolytic and active immunotherapeutic. Curr Opin Virol 2015; 13:49-54. [DOI: 10.1016/j.coviro.2015.03.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 03/16/2015] [Accepted: 03/19/2015] [Indexed: 01/21/2023]
|
23
|
Effects of Traditional Chinese Medicine on DCs Under Tumor Microenvironment. DENDRITIC CELLS: BIOPHYSICS, TUMOR MICROENVIRONMENT AND CHINESE TRADITIONAL MEDICINE 2015. [DOI: 10.1007/978-94-017-7405-5_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
24
|
Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2014; 1:1111-1134. [PMID: 23170259 PMCID: PMC3494625 DOI: 10.4161/onci.21494] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) occupy a central position in the immune system, orchestrating a wide repertoire of responses that span from the development of self-tolerance to the elicitation of potent cellular and humoral immunity. Accordingly, DCs are involved in the etiology of conditions as diverse as infectious diseases, allergic and autoimmune disorders, graft rejection and cancer. During the last decade, several methods have been developed to load DCs with tumor-associated antigens, ex vivo or in vivo, in the attempt to use them as therapeutic anticancer vaccines that would elicit clinically relevant immune responses. While this has not always been the case, several clinical studies have demonstrated that DC-based anticancer vaccines are capable of activating tumor-specific immune responses that increase overall survival, at least in a subset of patients. In 2010, this branch of clinical research has culminated with the approval by FDA of a DC-based therapeutic vaccine (sipuleucel-T, Provenge®) for use in patients with asymptomatic or minimally symptomatic metastatic hormone-refractory prostate cancer. Intense research efforts are currently dedicated to the identification of the immunological features of patients that best respond to DC-based anticancer vaccines. This knowledge may indeed lead to personalized combination strategies that would extend the benefit of DC-based immunotherapy to a larger patient population. In addition, widespread enthusiasm has been generated by the results of the first clinical trials based on in vivo DC targeting, an approach that holds great promises for the future of DC-based immunotherapy. In this Trial Watch, we will summarize the results of recently completed clinical trials and discuss the progress of ongoing studies that have evaluated/are evaluating DC-based interventions for cancer therapy.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Institut Gustave Roussy; Villejuif, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Song S, Yuan P, Li P, Wu H, Lu J, Wei W. Dynamic analysis of tumor-associated immune cells in DEN-induced rat hepatocellular carcinoma. Int Immunopharmacol 2014; 22:392-9. [PMID: 25066760 DOI: 10.1016/j.intimp.2014.07.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 06/16/2014] [Accepted: 07/08/2014] [Indexed: 12/14/2022]
Abstract
Hepatocellular carcinoma (HCC) is an aggressive disease with poor prognosis and limited methods to predict patient survival. Immune cells infiltrating tumors are known to impact tumor progression. Here, we analyzed the phenotype and function of dendritic cells (DCs), and the frequency of IL-10-producing regulatory B cells (Breg) and Foxp3(+) regulatory T cells (Treg) in different stages of N-nitrosodiethylamine (DEN)-induced rat HCC in order to understand their roles in this disease. 4weeks following DEN treatment, no significant differences in CD80 and CD86 expression were found on DCs from HCC rats and normal rats but 12 and 16weeks following DEN treatment, the expression of CD80, CD86 and MHCII on DCs of HCC rats was significantly decreased. We also found that the frequency of IL-10-producing Breg and CD4(+)CD25(+)Foxp3(+) Treg in HCC rats was obviously increased during all of these three stages. In addition, the bone-marrow derived DCs (BMDCs) from HCC rats displayed lower ability in activating T cells and an increase in IL-10 secretion. No differences in IL-12 level and endocytosis ability were found on BMDCs from HCC rats and normal rats. Our results suggest that the dysfunction of DCs and the increase of IL-10-producing Breg and Foxp3(+) Treg might play important roles in HCC progression.
Collapse
Affiliation(s)
- Shasha Song
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China
| | - Pingfan Yuan
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China
| | - Peipei Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China
| | - Huaxun Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China
| | - Jingtao Lu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China.
| |
Collapse
|
26
|
Platzer B, Stout M, Fiebiger E. Antigen cross-presentation of immune complexes. Front Immunol 2014; 5:140. [PMID: 24744762 PMCID: PMC3978348 DOI: 10.3389/fimmu.2014.00140] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 03/19/2014] [Indexed: 12/23/2022] Open
Abstract
The ability of dendritic cells (DCs) to cross-present tumor antigens has long been a focus of interest to physicians, as well as basic scientists, that aim to establish efficient cell-based cancer immune therapy. A prerequisite for exploiting this pathway for therapeutic purposes is a better understanding of the mechanisms that underlie the induction of tumor-specific cytotoxic T-lymphocyte (CTL) responses when initiated by DCs via cross-presentation. The ability of humans DC to perform cross-presentation is of utmost interest, as this cell type is a main target for cell-based immunotherapy in humans. The outcome of a cross-presentation event is guided by the nature of the antigen, the form of antigen uptake, and the subpopulation of DCs that performs presentation. Generally, CD8α+ DCs are considered to be the most potent cross-presenting DCs. This paradigm, however, only applies to soluble antigens. During adaptive immune responses, immune complexes form when antibodies interact with their specific epitopes on soluble antigens. Immunoglobulin G (IgG) immune complexes target Fc-gamma receptors on DCs to shuttle exogenous antigens efficiently into the cross-presentation pathway. This receptor-mediated cross-presentation pathway is a well-described route for the induction of strong CD8+ T cell responses. IgG-mediated cross-presentation is intriguing because it permits the CD8− DCs, which are commonly considered to be weak cross-presenters, to efficiently cross-present. Engaging multiple DC subtypes for cross-presentation might be a superior strategy to boost CTL responses in vivo. We here summarize our current understanding of how DCs use IgG-complexed antigens for the efficient induction of CTL responses. Because of its importance for human cell therapy, we also review the recent advances in the characterization of cross-presentation properties of human DC subsets.
Collapse
Affiliation(s)
- Barbara Platzer
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Boston Children's Hospital, Harvard Medical School , Boston, MA , USA
| | - Madeleine Stout
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Boston Children's Hospital, Harvard Medical School , Boston, MA , USA
| | - Edda Fiebiger
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Boston Children's Hospital, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
27
|
Song S, Yuan P, Wu H, Chen J, Fu J, Li P, Lu J, Wei W. Dendritic cells with an increased PD-L1 by TGF-β induce T cell anergy for the cytotoxicity of hepatocellular carcinoma cells. Int Immunopharmacol 2014; 20:117-23. [PMID: 24606770 DOI: 10.1016/j.intimp.2014.02.027] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 01/21/2014] [Accepted: 02/19/2014] [Indexed: 11/28/2022]
Abstract
The effects of TGF-β on dendritic cells (DCs) in the tumor microenvironment are not well-understood. In this study, we investigated the effect of TGF-β on the induction of programmed death ligand-1 (PD-L1) expression in DCs and the underlying mechanism, and we further investigated the influence of the DCs with PD-L1 expression altered by TGF-β on T-cell immunity. We determined that TGF-β increased the expression of PD-L1 and signal transducers and activators of transcription 3 (STAT3) in DCs in both a time- and dose-dependent manner, and the expression of PD-L1 was decreased significantly after STAT3 blockade. In addition, TGF-β-treated DCs induced the apoptosis of T cells and increased the percentage of CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs). Furthermore, the cytotoxicity of T cells against mice hepatocellular carcinoma cells (Hepa) was obviously suppressed. These results suggest that PD-L1 may play an important role in TGF-β-induced immune dysfunction, which finally results in a failure in the anti-tumor responses, and the TGF-β-STAT3-PD-L1 signaling pathway may contribute to novel therapeutic targets for the tumor based on DCs.
Collapse
Affiliation(s)
- Shasha Song
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China
| | - Pingfan Yuan
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China
| | - Huaxun Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China
| | - Jingyu Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China
| | - Jingjing Fu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China
| | - Peipei Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China
| | - Jingtao Lu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Antiinflammatory and Immune Medicine, Ministry of Education, Hefei 230032, PR China.
| |
Collapse
|
28
|
Kulzer L, Rubner Y, Deloch L, Allgäuer A, Frey B, Fietkau R, Dörrie J, Schaft N, Gaipl US. Norm- and hypo-fractionated radiotherapy is capable of activating human dendritic cells. J Immunotoxicol 2014; 11:328-36. [PMID: 24512329 DOI: 10.3109/1547691x.2014.880533] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Despite the transient immunosuppressive properties of local radiotherapy (RT), this classical treatment modality of solid tumors is capable of inducing immunostimulatory forms of tumor-cell death. The resulting 'immunotoxicity' in the tumor, but not in healthy tissues, may finally lead to immune-mediated destruction of the tumor. However, little is known about the best irradiation scheme in this setting. This study examines the immunological effects of differently irradiated human colorectal tumor cells on human monocyte-derived dendritic cells (DC). Human SW480 tumor cells were irradiated with a norm-fractionation scheme (5 × 2 Gy), a hypo-fractionated protocol (3 × 5 Gy), and with a high single irradiation dose (radiosurgery; 1 × 15 Gy). Subsequently, human immature DC (iDC) were co-incubated with supernatants (SN) of these differently treated tumor cells. Afterwards, DC were analyzed regarding the expression of maturation markers, the release of cytokines, and the potential to stimulate CD4(+) T-cells. The co-incubation of iDC with SN of tumor cells exposed to norm- or hypo-fractionated RT resulted in a significantly increased secretion of the immune activating cytokines IL-12p70, IL-8, IL-6, and TNFα, compared to iDC co-incubated with SN of tumor cells that received a high single irradiation dose or were not irradiated. In addition, DC-maturation markers CD80, CD83, and CD25 were also exclusively elevated after co-incubation with the SN of fractionated irradiated tumor cells. Furthermore, the SN of tumor cells that were irradiated with norm- or hypo-fractionated RT triggered iDC to stimulate CD4(+) T-cells not only in an allogenic, but also in an antigen-specific manner like mature DC. Collectively, these results demonstrate that norm- and hypo-fractionated RT induces a fast human colorectal tumor-cell death with immunogenic potential that can trigger DC maturation and activation in vitro. Such findings may contribute to the improvement of irradiation protocols for the most beneficial induction of anti-tumor immunity.
Collapse
|
29
|
Kokolus KM, Spangler HM, Povinelli BJ, Farren MR, Lee KP, Repasky EA. Stressful presentations: mild cold stress in laboratory mice influences phenotype of dendritic cells in naïve and tumor-bearing mice. Front Immunol 2014; 5:23. [PMID: 24575090 PMCID: PMC3918933 DOI: 10.3389/fimmu.2014.00023] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/15/2014] [Indexed: 01/07/2023] Open
Abstract
The ability of dendritic cells (DCs) to stimulate and regulate T cells is critical to effective anti-tumor immunity. Therefore, it is important to fully recognize any inherent factors which may influence DC function under experimental conditions, especially in laboratory mice since they are used so heavily to model immune responses. The goals of this report are to 1) briefly summarize previous work revealing how DCs respond to various forms of physiological stress and 2) to present new data highlighting the potential for chronic mild cold stress inherent to mice housed at the required standard ambient temperatures to influence baseline DCs properties in naïve and tumor-bearing mice. As recent data from our group shows that CD8+ T cell function is significantly altered by chronic mild cold stress and since DC function is crucial for CD8+ T cell activation, we wondered whether housing temperature may also be influencing DC function. Here we report that there are several significant phenotypical and functional differences among DC subsets in naïve and tumor-bearing mice housed at either standard housing temperature or at a thermoneutral ambient temperature, which significantly reduces the extent of cold stress. The new data presented here strongly suggests that, by itself, the housing temperature of mice can affect fundamental properties and functions of DCs. Therefore differences in basal levels of stress due to housing should be taken into consideration when interpreting experiments designed to evaluate the impact of additional variables, including other stressors on DC function.
Collapse
Affiliation(s)
- Kathleen M Kokolus
- Department of Immunology, Roswell Park Cancer Institute , Buffalo, NY , USA
| | - Haley M Spangler
- Department of Immunology, Roswell Park Cancer Institute , Buffalo, NY , USA
| | | | - Matthew R Farren
- Department of Immunology, Roswell Park Cancer Institute , Buffalo, NY , USA
| | - Kelvin P Lee
- Department of Immunology, Roswell Park Cancer Institute , Buffalo, NY , USA
| | | |
Collapse
|
30
|
Kim SJ, Ha GH, Kim SH, Kang CD. Combination of cancer immunotherapy with clinically available drugs that can block immunosuppressive cells. Immunol Invest 2013; 43:517-34. [PMID: 24295450 DOI: 10.3109/08820139.2013.857352] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Although cancer immunotherapy, which is able to target specifically cancer cells without detrimental effects to normal cell functions, would serve as an ideal therapeutic modality, most of the randomized clinical trials of cancer immunotherapy have not demonstrated a meaningful survival benefit to cancer patients over preexisting therapeutic modalities. Due to the discrepancy between the impressive preclinical results and the limited clinical results, the cancer immunotherapy is not accepted generally as a standard therapy for cancers. A variety of immune escape mechanisms are thought to be involved in this ineffectiveness of cancer immunotherapy. Therefore, elimination of immunosuppressive activities in tumor microenvironment will enhance the effectiveness of cancer immunotherapy, which is currently focused on activation of tumor-specific immune responses. Since there are now increasing evidences showing that many cytotoxic anticancer drugs including targeted agents given in lower-than-therapeutic doses have not only the ability to eliminate tumor cells but can also block the immunosuppressive activities in tumor microenvironments and consequently favor the development of anticancer immune responses, clinically available drugs can be considered for their rapid application to cancer immunotherapies to enhance the efficacy of cancer immunotherapies with marginal effects on cancer treatment.
Collapse
Affiliation(s)
- So-Jung Kim
- MD-PhD Program, Pusan National University School of Medicine , Yangsan , South Korea 626-870
| | | | | | | |
Collapse
|
31
|
Chen L, Fabian KL, Taylor JL, Storkus WJ. Therapeutic use of dendritic cells to promote the extranodal priming of anti-tumor immunity. Front Immunol 2013; 4:388. [PMID: 24348473 PMCID: PMC3843121 DOI: 10.3389/fimmu.2013.00388] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 11/05/2013] [Indexed: 12/17/2022] Open
Abstract
Ectopic lymphoid tissue, also known as tertiary lymphoid organs (TLO) develop adaptively within sites of chronic tissue inflammation, thereby allowing the host to efficiently crossprime specific immune effector cells within sites of disease. Recent evidence suggests that the presence of TLO in the tumor microenvironment (TME) predicts better overall survival. We will discuss the relevance of extranodal T cell priming within the TME as a means to effectively promote anti-tumor immunity and the strategic use of dendritic cell (DC)-based therapies to reinforce this clinically preferred process in the cancer-bearing host.
Collapse
Affiliation(s)
- Lu Chen
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Jennifer L Taylor
- Department of Dermatology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA ; Department of Dermatology, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA ; University of Pittsburgh Cancer Institute , Pittsburgh, PA , USA
| |
Collapse
|
32
|
Molino NM, Anderson AKL, Nelson EL, Wang SW. Biomimetic protein nanoparticles facilitate enhanced dendritic cell activation and cross-presentation. ACS NANO 2013; 7:9743-52. [PMID: 24090491 PMCID: PMC3893022 DOI: 10.1021/nn403085w] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Many current cancer vaccine strategies suffer from the inability to mount a CD8 T cell response that is strong enough to overcome the low immunogenicity of tumors. Viruses naturally possess the sizes, geometries, and physical properties for which the immune system has evolved to recognize, and mimicking those properties with nanoparticles can produce robust platforms for vaccine design. Using the nonviral E2 core of pyruvate dehydrogenase, we have engineered a viral-mimicking vaccine platform capable of encapsulating dendritic cell (DC)-activating CpG molecules in an acid-releasable manner and displaying MHC I-restricted SIINFEKL peptide epitopes. Encapsulated CpG activated bone marrow-derived DCs at a 25-fold lower concentration in vitro when delivered with the E2 nanoparticle than with unbound CpG alone. Combining CpG and SIINFEKL within a single multifunctional particle induced ∼3-fold greater SIINFEKL display on MHC I by DCs over unbound peptide. Importantly, combining CpG and SIINFEKL to the E2 nanoparticle for simultaneous temporal and spatial delivery to DCs showed increased and prolonged CD8 T cell activation, relative to free peptide or peptide-bound E2. By codelivering peptide epitopes and CpG activator in a particle of optimal DC-uptake size, we demonstrate the ability of a noninfectious protein nanoparticle to mimic viral properties and facilitate enhanced DC activation and cross-presentation.
Collapse
Affiliation(s)
- Nicholas M. Molino
- Department of Chemical Engineering and Materials Science, University of California, 916 Engineering Tower, Irvine, CA 92697-2575
| | | | | | - Szu-Wen Wang
- Department of Chemical Engineering and Materials Science, University of California, 916 Engineering Tower, Irvine, CA 92697-2575
| |
Collapse
|
33
|
Zhou Z, Li W, Song Y, Wang L, Zhang K, Yang J, Zhang W, Su H, Zhang Y. Growth differentiation factor-15 suppresses maturation and function of dendritic cells and inhibits tumor-specific immune response. PLoS One 2013; 8:e78618. [PMID: 24236027 PMCID: PMC3827235 DOI: 10.1371/journal.pone.0078618] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 09/14/2013] [Indexed: 01/24/2023] Open
Abstract
Dendritic cells (DCs) play a key role in the initiation stage of an antigen-specific immune response. A variety of tumor-derived factors (TDFs) can suppress DC maturation and function, resulting in defects in the tumor-specific immune response. To identify unknown TDFs that may suppress DCs maturation and function, we established a high-throughput screening technology based on a human liver tumor T7 phage cDNA library and screened all of the proteins derived from hepatoma cells that potentially interact with immature DCs. Growth/differentiation factor-15 (GDF-15) was detected and chosen for further study. By incubation of DCs cultures with GDF-15, we demonstrate that GDF-15 can inhibit surface protrusion formation during DC maturation; suppress the membrane expression of CD83, CD86 and HLA-DR on DCs; enhance phagocytosis by DCs; reduce IL-12 and elevate TGF-β1 secretion by DCs; inhibit T cell stimulation and cytotoxic T lymphocyte (CTL) activation by DCs. By building tumor-bearing mouse models, we demonstrate that GDF-15 can inhibit the ability of DCs to stimulate a tumor-specific immune response in vivo. These results indicate that GDF-15 may be one of the critical molecules that inhibit DC maturation and function and are involved in tumor immune escape. Thus, GDF-15 may be a novel target in tumor immunotherapy.
Collapse
Affiliation(s)
- Zhizhong Zhou
- The State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weina Li
- The State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yang Song
- Department of Oncology, Tangdu Hospital, Xi'an, Shaanxi, China
| | - Lili Wang
- The State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Kuo Zhang
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Jing Yang
- Department of Oncology, Tangdu Hospital, Xi'an, Shaanxi, China
| | - Wei Zhang
- The State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, Shaanxi, China
- * E-mail: (YQZ); (HS); (WZ)
| | - Haichuan Su
- Department of Oncology, Tangdu Hospital, Xi'an, Shaanxi, China
- * E-mail: (YQZ); (HS); (WZ)
| | - Yingqi Zhang
- The State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, Shaanxi, China
- * E-mail: (YQZ); (HS); (WZ)
| |
Collapse
|
34
|
Fu C, Jiang A. β-catenin-mediated inhibition of cross-priming: A new mechanism for tumors to evade immunosurveillance. Oncoimmunology 2013; 2:e26920. [PMID: 24567866 PMCID: PMC3925153 DOI: 10.4161/onci.26920] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 10/22/2013] [Indexed: 01/30/2023] Open
Abstract
Cross-priming plays a major role in generating CD8+ T cell-dependent antitumor immunity through cross-presentation. However, the cross-presentation of tumor-associated antigens by dendritic cells often promotes tolerance rather than CD8+ T-cell immunity. We have now identified a β-catenin-dependent pathway of cross-priming inhibition as a novel and potentially broad mechanism whereby neoplastic cells promote immunosuppression.
Collapse
Affiliation(s)
- Chunmei Fu
- Department of Immunology; Roswell Park Cancer Institute; Buffalo, NY USA
| | - Aimin Jiang
- Department of Immunology; Roswell Park Cancer Institute; Buffalo, NY USA
| |
Collapse
|
35
|
Nasi A, Fekete T, Krishnamurthy A, Snowden S, Rajnavölgyi E, Catrina AI, Wheelock CE, Vivar N, Rethi B. Dendritic Cell Reprogramming by Endogenously Produced Lactic Acid. THE JOURNAL OF IMMUNOLOGY 2013; 191:3090-9. [DOI: 10.4049/jimmunol.1300772] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
36
|
Bugaut H, Bruchard M, Berger H, Derangère V, Odoul L, Euvrard R, Ladoire S, Chalmin F, Végran F, Rébé C, Apetoh L, Ghiringhelli F, Mignot G. Bleomycin exerts ambivalent antitumor immune effect by triggering both immunogenic cell death and proliferation of regulatory T cells. PLoS One 2013; 8:e65181. [PMID: 23762310 PMCID: PMC3676388 DOI: 10.1371/journal.pone.0065181] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 04/23/2013] [Indexed: 12/22/2022] Open
Abstract
Bleomycin (BLM) is an anticancer drug currently used for the treatment of testis cancer and Hodgkin lymphoma. This drug triggers cancer cell death via its capacity to generate radical oxygen species (ROS). However, the putative contribution of anticancer immune responses to the efficacy of BLM has not been evaluated. We make here the observation that BLM induces immunogenic cell death. In particular, BLM is able to induce ROS-mediated reticulum stress and autophagy, which result in the surface exposure of chaperones, including calreticulin and ERp57, and liberation of HMBG1 and ATP. BLM induces anti-tumor immunity which relies on calreticulin, CD8+ T cells and interferon-γ. We also find that, in addition to its capacity to trigger immunogenic cell death, BLM induces expansion of Foxp3+ regulatory T (Treg) cells via its capacity to induce transforming growth factor beta (TGFβ) secretion by tumor cells. Accordingly, Treg cells or TGFβ depletion dramatically potentiates the antitumor effect of BLM. We conclude that BLM induces both anti-tumor CD8+ T cell response and a counteracting Treg proliferation. In the future, TGFβ or Treg inhibition during BLM treatment could greatly enhance BLM anti-tumor efficacy.
Collapse
Affiliation(s)
- Hélène Bugaut
- INSERM, U866, Dijon, France
- INSERM AVENIR Team, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
| | - Mélanie Bruchard
- INSERM, U866, Dijon, France
- INSERM AVENIR Team, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
| | - Hélène Berger
- INSERM, U866, Dijon, France
- INSERM AVENIR Team, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
| | - Valentin Derangère
- INSERM, U866, Dijon, France
- INSERM AVENIR Team, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
| | - Ludivine Odoul
- University of Bern, Institute of Pathology, Bern, Switzerland
| | - Romain Euvrard
- INSERM, U866, Dijon, France
- INSERM AVENIR Team, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
| | - Sylvain Ladoire
- INSERM, U866, Dijon, France
- INSERM AVENIR Team, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
- Centre Georges François Leclerc, Dijon, France
| | - Fanny Chalmin
- INSERM, U866, Dijon, France
- INSERM AVENIR Team, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
| | - Frédérique Végran
- INSERM, U866, Dijon, France
- INSERM AVENIR Team, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
| | - Cédric Rébé
- INSERM, U866, Dijon, France
- INSERM AVENIR Team, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
- Centre Georges François Leclerc, Dijon, France
| | - Lionel Apetoh
- INSERM, U866, Dijon, France
- INSERM AVENIR Team, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
| | - François Ghiringhelli
- INSERM, U866, Dijon, France
- INSERM AVENIR Team, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
- Centre Georges François Leclerc, Dijon, France
- * E-mail: (FG); (GM)
| | - Grégoire Mignot
- INSERM, U866, Dijon, France
- INSERM AVENIR Team, Dijon, France
- Faculté de Médecine, Université de Bourgogne, Dijon, France
- * E-mail: (FG); (GM)
| |
Collapse
|
37
|
Gatti G, Nuñez NG, Nocera DA, Dejager L, Libert C, Giraudo C, Maccioni M. Direct effect of dsRNA mimetics on cancer cells induces endogenous IFN-β production capable of improving dendritic cell function. Eur J Immunol 2013; 43:1849-61. [PMID: 23636788 DOI: 10.1002/eji.201242902] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 03/13/2013] [Accepted: 04/25/2013] [Indexed: 12/27/2022]
Abstract
Viral double-stranded RNA (dsRNA) mimetics have been explored in cancer immunotherapy to promote antitumoral immune response. Polyinosine-polycytidylic acid (poly I:C) and polyadenylic-polyuridylic acid (poly A:U) are synthetic analogs of viral dsRNA and strong inducers of type I interferon (IFN). We describe here a novel effect of dsRNA analogs on cancer cells: besides their potential to induce cancer cell apoptosis through an IFN-β autocrine loop, dsRNA-elicited IFN-β production improves dendritic cell (DC) functionality. Human A549 lung and DU145 prostate carcinoma cells significantly responded to poly I:C stimulation, producing IFN-β at levels that were capable of activating STAT1 and enhancing CXCL10, CD40, and CD86 expression on human monocyte-derived DCs. IFN-β produced by poly I:C-activated human cancer cells increased the capacity of monocyte-derived DCs to stimulate IFN-γ production in an allogeneic stimulatory culture in vitro. When melanoma murine B16 cells were stimulated in vitro with poly A:U and then inoculated into TLR3(-/-) mice, smaller tumors were elicited. This tumor growth inhibition was abrogated in IFNAR1(-/-) mice. Thus, dsRNA compounds are effective adjuvants not only because they activate DCs and promote strong adaptive immunity, but also because they can directly act on cancer cells to induce endogenous IFN-β production and contribute to the antitumoral response.
Collapse
Affiliation(s)
- Gerardo Gatti
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | | | | | | | | | | |
Collapse
|
38
|
van Spriel AB, de Jong EC. Dendritic cell science: more than 40 years of history. J Leukoc Biol 2012; 93:33-8. [PMID: 23024285 DOI: 10.1189/jlb.0512263] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Over 40 years of research into the field of DCs has revolutionized our understanding into the activation and regulation of the immune system. This minireview discusses the major breakthroughs in DC science that have paved the way to the 2011 Nobel Prize in Physiology-Medicine awarded to Bruce A. Beutler and Jules A. Hoffmann (for their discoveries in innate immune recognition) and Ralph M. Steinman (for his discovery of the DC).
Collapse
Affiliation(s)
- Annemiek B van Spriel
- Nijmegen Centre for Molecular Life Sciences, Geert Grooteplein 28, Nijmegen, The Netherlands.
| | | |
Collapse
|
39
|
CD86 and IL-12p70 are key players for T helper 1 polarization and natural killer cell activation by Toll-like receptor-induced dendritic cells. PLoS One 2012; 7:e44266. [PMID: 22962607 PMCID: PMC3433478 DOI: 10.1371/journal.pone.0044266] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 07/31/2012] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Dendritic cells (DCs) determine the activation and polarization of T cells via expression of costimulatory molecules and secretion of cytokines. The function of DCs derived from monocytes ex vivo strongly depends on the composition of the maturation cocktail used. METHODOLOGY/PRINCIPAL FINDINGS We analyzed the effect of costimulatory molecule expression and cytokine secretion by DCs on T and natural killer (NK) cell activation by conducting a head-to-head comparison of a Toll-like receptor (TLR) agonist-based cocktail with the standard combination of proinflammatory cytokines or IL-10 alone. We could show that TLR-induced DCs are characterized by a predominance of costimulatory over coinhibitory molecules and by high secretion of IL-12p70, but not IL-10. Functionally, these signals translated into an increase in IFN-γ secreting Th1 cells and a decrease in regulatory T cells. T cell activation and polarization were dependent on IL-12p70 and CD86, but remarkably not on CD80 signaling. By means of IL-12p70 secretion, only TLR-induced DCs activated NK cells. CONCLUSIONS/SIGNIFICANCE TLR-matured DCs are highly suitable for application in immunotherapeutic strategies that rely on strong type 1 polarization and NK cell activation. Their effects particularly depend on high CD86 expression and IL-12p70 secretion.
Collapse
|
40
|
Schmidt MA, Förtsch C, Schmidt M, Rau TT, Fietkau R, Distel LV. Circulating regulatory T cells of cancer patients receiving radiochemotherapy may be useful to individualize cancer treatment. Radiother Oncol 2012; 104:131-8. [PMID: 22682747 DOI: 10.1016/j.radonc.2012.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 05/02/2012] [Accepted: 05/14/2012] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND PURPOSE Dendritic cells (DCs) and regulatory T cells (Treg) play a major role in anti-tumor immune response of cancer patients. We investigated the effect of radiochemotherapy on patients' blood immune cells and their predictive value for tumor response. MATERIALS AND METHODS DCs and Treg of colorectal cancer (CRC) or breast cancer (BC) patients were examined through multicolor flow cytometry before the beginning and after the first week of radiochemotherapy (RCT). DCs were stained for BDCA1 and BDCA2, Treg were stained for CD4, CD25, CD127 and FoxP3. IL-2, IL-10 and TNF-α plasma levels of CRC patients were also determined. We examined the interrelationship between immune cell count alterations, applied dose values, cytokine plasma levels as well as histopathological parameters. RESULTS DCs were increased in BC and CRC patients compared to healthy control individuals (HC). CRC patients had higher levels of Treg (59.0%) compared to BC patients (31.3%) and HC (27.0%). Treg of CRC (58.7% vs. 41.3% p<0.001) but not BC patients (31.3% vs. 38.8%, p=0.164) decreased distinctly after the first week of radiation therapy. Applied dose values and decrease of Treg correlated positively (r=0.216, p=0.054). We also found a positive correlation of IL-10 plasma levels and Treg levels (r=0.748, p=0.021). CRC patients with favorable tumor stage (<ypT3a) have higher levels of Treg after 5 days of RCT (49.4% vs. 34.0%, p=0.043). CONCLUSION Higher Treg levels are associated with favorable tumor stage. We hypothesize that a dramatic decrease of Treg after in vivo irradiation may be a good indicator for necessary dose adjustments in radiation therapy of CRC patients.
Collapse
Affiliation(s)
- Manuel A Schmidt
- Department of Radiation Oncology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsstrasse 27, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Zheng J, Liu Q, Yang J, Ren Q, Cao W, Yang J, Yu Z, Yu F, Wu Y, Shi H, Liu W. Co-culture of apoptotic breast cancer cells with immature dendritic cells: a novel approach for DC-based vaccination in breast cancer. Braz J Med Biol Res 2012; 45:510-5. [PMID: 22527124 PMCID: PMC3854298 DOI: 10.1590/s0100-879x2012007500061] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 03/13/2012] [Indexed: 12/22/2022] Open
Abstract
A dendritic cell (DC)-based vaccine strategy could reduce the risk of recurrence and improve the survival of breast cancer patients. However, while therapy-induced apoptosis of hepatocellular and colorectal carcinoma cells can enhance maturation and antigen presentation of DCs, whether this effect occurs in breast cancer is currently unknown. In the present study, we investigated the effect of doxorubicin (ADM)-induced apoptotic MCF-7 breast cancer cells on the activation of DCs. ADM-induced apoptotic MCF-7 cells could effectively induce immature DC (iDC) maturation. The mean fluorescence intensity (MFI) of DC maturity marker CD83 was 23.3 in the ADM-induced apoptotic MCF-7 cell group compared with 8.5 in the MCF-7 cell group. The MFI of DC co-stimulatory marker CD86 and HLA-DR were also increased after iDCs were treated with ADM-induced apoptotic MCF-7 cells. Furthermore, the proliferating autologous T-lymphocytes increased from 14.2 to 40.3% after incubated with DCs induced by apoptotic MCF-7 cells. The secretion of interferon-γ by these T-lymphocytes was also increased. In addition, cell-cell interaction between apoptotic MCF-7 cells and iDCs, but not soluble factors released by apoptotic MCF-7 cells, was crucial for the maturation of iDCs. These findings constitute a novel in vitro DC-based vaccine strategy for the treatment of breast cancer by ADM-induced apoptotic MCF-7 cells.
Collapse
Affiliation(s)
- Jin Zheng
- Department of Oncology, State Key Discipline of Cell Biology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Curiel TJ. Immunotherapy: a useful strategy to help combat multidrug resistance. Drug Resist Updat 2012; 15:106-13. [PMID: 22483359 DOI: 10.1016/j.drup.2012.03.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2011] [Revised: 01/24/2012] [Accepted: 03/13/2012] [Indexed: 12/22/2022]
Abstract
Multidrug resistance (MDR) renders cancer cells relatively invulnerable to treatment with many standard cytotoxic anti-cancer agents. Cancer immunotherapy could be an important adjunct for other strategies to treat MDR positive cancers, as resistance to immunotherapy generally is unrelated to mechanisms of resistance to cytotoxic agents. Immunotherapy to combat MDR positive tumors could use any of the following strategies: direct immune attack against MDR positive cells, using MDR as an immune target to deliver cytotoxic agents, capitalization on other immune properties of MDR positive cells, or conditional immunotoxins expressed under MDR control. Additional insights into the immunogenic potential of some cytotoxic agents can also be brought to bear on these strategies. This review will highlight key concepts in cancer immunotherapy and illustrate immune principles and strategies that have been or could be used to help destroy MDR positive tumor cells, either alone or in rational combinations.
Collapse
Affiliation(s)
- Tyler J Curiel
- University of Texas Health Science Center, Cancer Therapy and Research Center, STRF Adult Cancer Program MC 8252, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904, United States.
| |
Collapse
|
43
|
Wang HY, Wang RF. Enhancing cancer immunotherapy by intracellular delivery of cell-penetrating peptides and stimulation of pattern-recognition receptor signaling. Adv Immunol 2012; 114:151-76. [PMID: 22449781 DOI: 10.1016/b978-0-12-396548-6.00006-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The importance of T-cell-mediated antitumor immunity has been demonstrated in both animal models and human cancer immunotherapy. In the past 30 years, T-cell-based immunotherapy has been improved with an objective clinical response rate of up to 72%. Identification of MHC class I- and II-restricted tumor antigens recognized by tumor-reactive T cells has generated a resurgence of interest in cancer vaccines. Although clinical trials with cancer peptide/protein vaccines have only met a limited success, several phase II/III clinical trials are either completed or ongoing with encouraging results. Recent advances in immunotherapy have led to the approval of two anticancer drugs (sipuleucel-T vaccine and anti-CTLA-4 antibody) by the US FDA for the treatment of metastatic castration-resistant prostate cancer and melanoma, respectively. Intracellular delivery of antigenic peptides into dendritic cells (DCs) prolongs antigen presentation of antigen-presenting cells to T cells, thus further improving clinical efficacy of peptide/protein cancer vaccines. Because innate immune responses are critically important to provide sensing and initiating of adaptive immunity, combined use of cell-penetrating peptide vaccines with stimulation of innate immune signaling may produce potent antitumor immune responses. We will discuss the recent progress and novel strategies in cancer immunotherapy.
Collapse
Affiliation(s)
- Helen Y Wang
- Department of Pathology and Immunology, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
44
|
Naiditch H, Shurin MR, Shurin GV. Targeting myeloid regulatory cells in cancer by chemotherapeutic agents. Immunol Res 2011; 50:276-85. [PMID: 21717082 DOI: 10.1007/s12026-011-8213-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Recent findings in humans and numerous experimental models provide evidence of the important role of immune regulatory cells in cancer and various diseases. "Myeloid regulatory cells" (MRC) include myeloid-derived suppressor cells, regulatory dendritic cells, regulatory macrophages, and subsets of granulocytes that expand during pathologic conditions and that have the ability to suppress cellular immunity. A decrease in MRC population and/or activity has been shown to have positive immune-potentiating effects. Several clinical trials have thus been initiated with the goal of manipulating the expansion or activation of these cells and thereby improving patient immune responses. New data from our own and other laboratories recently revealed that ultralow noncytotoxic doses of certain chemotherapeutic drugs could up-regulate antitumor immunity by modulating the formation, differentiation, and/or function of MRC. This new phenomenon, termed "chemomodulation," allows for the regulation of the tumor microenvironment without the undesirable toxic effects associated with conventional chemotherapy. However, further studies are required before this new targeted therapy can find its way to patients with cancer.
Collapse
Affiliation(s)
- Hiam Naiditch
- Departments of Pathology and Immunology, Experimental Pathology, University of Pittsburgh Medical Center, S735 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
45
|
Alshamsan A. Induction of tolerogenic dendritic cells by IL-6-secreting CT26 colon carcinoma. Immunopharmacol Immunotoxicol 2011; 34:465-9. [PMID: 21999714 DOI: 10.3109/08923973.2011.625034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
One scenario by which tumors escape immune recognition is the constitutive activation of signal transducer and activator of transcription 3 (STAT3). This transcription factor mediates the production of tumor-derived factors that negatively influence target immune cells, such as dendritic cells, and polarize them toward immune-tolerance also through the induction of STAT3 activation. In the current study, the effect of p-STAT3-positive murine colon carcinoma cell line (CT26) on bone marrow-derived DCs was examined. The results showed a remarkable increase in p-STAT3 in dendritic cells (DCs) only after CT26-CM incubation. The induction of p-STAT3 in CT26-CM exposed DCs was attributed at least in part to the high levels of interleukin-6 secreted by CT26 in culture. This was also accompanied by a significant reduction in the response to the immunostimulatory adjuvant lipopolysaccharide by lowering the expression of co-stimulatory molecules CD86 and CD40. Taken together, the results suggest an inhibitory effect of CT26 colon carcinoma on DC maturation through induction of STAT3 phosphorylation. Therefore, tumor-induced p-STAT3 in DCs can be seen as a promising target for colon cancer immunotherapy.
Collapse
Affiliation(s)
- Aws Alshamsan
- Nanomedicine Research Unit, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|