101
|
Stiff A, Trikha P, Mundy-Bosse B, McMichael E, Mace TA, Benner B, Kendra K, Campbell A, Gautam S, Abood D, Landi I, Hsu V, Duggan M, Wesolowski R, Old M, Howard JH, Yu L, Stasik N, Olencki T, Muthusamy N, Tridandapani S, Byrd JC, Caligiuri M, Carson WE. Nitric Oxide Production by Myeloid-Derived Suppressor Cells Plays a Role in Impairing Fc Receptor-Mediated Natural Killer Cell Function. Clin Cancer Res 2018; 24:1891-1904. [PMID: 29363526 DOI: 10.1158/1078-0432.ccr-17-0691] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 11/22/2017] [Accepted: 01/19/2018] [Indexed: 12/14/2022]
Abstract
Purpose: mAbs are used to treat solid and hematologic malignancies and work in part through Fc receptors (FcRs) on natural killer cells (NK). However, FcR-mediated functions of NK cells from patients with cancer are significantly impaired. Identifying the mechanisms of this dysfunction and impaired response to mAb therapy could lead to combination therapies and enhance mAb therapy.Experimental Design: Cocultures of autologous NK cells and MDSC from patients with cancer were used to study the effect of myeloid-derived suppressor cells (MDSCs) on NK-cell FcR-mediated functions including antibody-dependent cellular cytotoxicity, cytokine production, and signal transduction in vitro Mouse breast cancer models were utilized to study the effect of MDSCs on antibody therapy in vivo and test the efficacy of combination therapies including a mAb and an MDSC-targeting agent.Results: MDSCs from patients with cancer were found to significantly inhibit NK-cell FcR-mediated functions including antibody-dependent cellular cytotoxicity, cytokine production, and signal transduction in a contact-independent manner. In addition, adoptive transfer of MDSCs abolished the efficacy of mAb therapy in a mouse model of pancreatic cancer. Inhibition of iNOS restored NK-cell functions and signal transduction. Finally, nonspecific elimination of MDSCs or inhibition of iNOS in vivo significantly improved the efficacy of mAb therapy in a mouse model of breast cancer.Conclusions: MDSCs antagonize NK-cell FcR-mediated function and signal transduction leading to impaired response to mAb therapy in part through nitric oxide production. Thus, elimination of MDSCs or inhibition of nitric oxide production offers a strategy to improve mAb therapy. Clin Cancer Res; 24(8); 1891-904. ©2018 AACR.
Collapse
Affiliation(s)
- Andrew Stiff
- Medical Scientist Training Program, Columbus, Ohio.,Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Prashant Trikha
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | | | - Elizabeth McMichael
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Thomas A Mace
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Brooke Benner
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Kari Kendra
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Amanda Campbell
- Medical Scientist Training Program, Columbus, Ohio.,Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Shalini Gautam
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - David Abood
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Ian Landi
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Vincent Hsu
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Megan Duggan
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio
| | - Robert Wesolowski
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Matthew Old
- Department of Otolaryngology, The Ohio State University, Columbus, Ohio
| | - John Harrison Howard
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, Ohio
| | - Lianbo Yu
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Nancy Stasik
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Thomas Olencki
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Natarajan Muthusamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Susheela Tridandapani
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Internal Medicine and Dorothy M. Davis Heart and Lung Research Institute, Columbus, Ohio
| | - John C Byrd
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Michael Caligiuri
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - William E Carson
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio. .,Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, Ohio
| |
Collapse
|
102
|
Otegbeye F, Ojo E, Moreton S, Mackowski N, Lee DA, de Lima M, Wald DN. Inhibiting TGF-beta signaling preserves the function of highly activated, in vitro expanded natural killer cells in AML and colon cancer models. PLoS One 2018; 13:e0191358. [PMID: 29342200 PMCID: PMC5771627 DOI: 10.1371/journal.pone.0191358] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 01/03/2018] [Indexed: 01/08/2023] Open
Abstract
Natural killer cells harnessed from healthy individuals can be expanded ex vivo using various platforms to produce large doses for adoptive transfer into cancer patients. During such expansion, NK cells are increasingly activated and more efficient at killing cancer cells. Adoptive transfer however introduces these activated cells into a highly immunosuppressive tumor microenvironment mediated in part by excessive transforming growth factor beta (TGF-beta) from both cancer cells and their surrounding stroma. This microenvironment ultimately limits the clinical efficacy of NK cell therapy. In this study, we examined the use of a TGF-beta receptor kinase inhibitor, LY2157299, in preserving the cytotoxic function of ex vivo expanded, highly activated NK cells following sustained exposure to pathologic levels of TGF-beta in vitro and in a liver metastases model of colon cancer. Using myeloid leukemia and colon cancer cell lines, we show that the TGF-beta driven impairment of NK cell cytotoxicity is mitigated by LY2157299. We demonstrate this effect using quantitative cytotoxicity assays as well as by showing a preserved activated phenotype with high NKG2D/CD16 expression and enhanced cytokine production. In a mouse liver metastases model of colon cancer, we observed significantly improved eradication of liver metastases in mice treated with adoptive NK cells combined with LY2157299 compared with mice receiving NK cells or TGF beta inhibition alone. We propose that the therapeutic efficacy of adoptive NK cell therapy clinically will be markedly enhanced by complementary approaches targeting TGF-beta signaling in vivo.
Collapse
Affiliation(s)
- Folashade Otegbeye
- Department of Medicine, Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
- Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Evelyn Ojo
- Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Stephen Moreton
- Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Nathan Mackowski
- Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Dean A. Lee
- Nationwide Children’s Hospital Division of Hematology/Oncology, Columbus, Ohio, United States of America
| | - Marcos de Lima
- Department of Medicine, Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
- Case Western Reserve University, Cleveland, Ohio, United States of America
| | - David N. Wald
- Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
| |
Collapse
|
103
|
Bonavida B. Linking Autophagy and the Dysregulated NFκB/ SNAIL/YY1/RKIP/PTEN Loop in Cancer: Therapeutic Implications. Crit Rev Oncog 2018; 23:307-320. [PMID: 30311562 PMCID: PMC6370039 DOI: 10.1615/critrevoncog.2018027212] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The role of autophagy in the pathogenesis of various cancers has been well documented in many reports. Autophagy in cancer cells regulates cell proliferation, viability, invasion, epithelial-to-mesenchymal transition (EMT), metastasis, and responses to chemotherapeutic and immunotherapeutic treatment strategies. These manifestations are the result of various regulatory gene products that govern autophagic, biochemical, and molecular mechanisms. In several human cancer cell models, the presence of a dysregulated circuit-namely, NFκB/SNAIL/YY1/RKIP/PTEN-that plays a major role in the regulation of tumor cell unique characteristics just listed for autophagy-regulated activities. Accordingly, the autophagic mechanism and the dysregulated circuit in cancer cells share many of the same properties and activities. Thus, it has been hypothesized that there must exist a biochemical/molecular link between the two. The present review describes the link and the association of each gene product of the dysregulated circuit with the autophagic mechanism and delineates the presence of crosstalk. Crosstalk between autophagy and the dysregulated circuit is significant and has important implications in the development of targeted therapies aimed at either autophagy or the dysregulated gene products in cancer cells.
Collapse
Affiliation(s)
- Benjamin Bonavida
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90025-1747,
| |
Collapse
|
104
|
Li Q, Huang Q, Huyan T, Wang Y, Huang Q, Shi J. Bifacial effects of engineering tumour cell-derived exosomes on human natural killer cells. Exp Cell Res 2017; 363:141-150. [PMID: 29269076 DOI: 10.1016/j.yexcr.2017.12.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 12/03/2017] [Accepted: 12/07/2017] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EVs) are nano vesicular structures that are secreted by almost all kinds of cells. Exosomes are small EVs derived from endosomes, with a diameter between 30-100nm. Tumour-derived exosomes carry many molecules and factors from tumour cells. These exosomes are recognized and taken up by immunocytes. However, tumour-derived exosomes can not only suppress immune cell functions but also help tumours escape immune surveillance in the tumour microenvironment. The present work investigated the effect of exosomes derived from genetical modified K562 cells (GMK cells), which express IL-15, IL-18 and 4-1BBL (TNFSF9) on their surface. The results showed that these GME exosomes, carrying IL-15, IL-18 and 4-1BBL proteins similar to their host cells, could activate NK cells, increase the cytotoxicity of NK cells on some tumour cells in a short treatment (4h) and promote NK cells proliferation. However, with an extended treatment time (48h), these exosomes could inhibite the cytotoxicity of NK cells by inhibiting activated receptor expression on NK cells. These results indicated the bifacial effects of GMK exosomes on NK cells, which will be helpful to explore the possibility of using transformed exosomes as an anti-tumour immune vaccine or a therapeutic tool in future.
Collapse
Affiliation(s)
- Qi Li
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China.
| | - Qiuping Huang
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China
| | - Ting Huyan
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China
| | - Yilin Wang
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China
| | - Qingsheng Huang
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China
| | - Junling Shi
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China
| |
Collapse
|
105
|
Prognostic value of MICA/B in cancers: a systematic review and meta-analysis. Oncotarget 2017; 8:96384-96395. [PMID: 29221214 PMCID: PMC5707108 DOI: 10.18632/oncotarget.21466] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 09/22/2017] [Indexed: 12/11/2022] Open
Abstract
Purpose MHC class I chain related-proteins A (MICA) and B (MICB) are natural killer group 2D ligands that mediate tumor surveillance. Several studies have suggested that MICA/B levels predict clinical outcomes in patients with cancer; however, this remains contentious. Here, we present a systematic review and meta-analysis of available studies of the prognostic value of MICA/B in cancer. Materials and Methods We searched PubMed, Embase, Clinicaltrials.gov, and Cochrane Library to identify studies published from inception to July 2017 that assessed MICA/B in patients with cancer. The hazard ratio (HR) and 95% confidence interval (CI) of MICA/B were extracted for overall survival (OS) analysis. Results A total of 19 studies comprising 2,588 patients with 10 different types of cancer were included in the study. Low sMICA/B levels were found associated with significantly longer OS (HR = 1.65, 95% CI [1.42–1.92], P < 0.00001). Patients with cancers of digestive system that exhibited high MICA/B expression had significantly longer OS in (HR = 0.56, 95% CI [0.39–0.80], P = 0.002) compared with those with lower MICA/B expression (I2 = 35%, P = 0.18). Conclusions Serum soluble MICA/B represents a potential prognostic marker in various human cancers. High cell-surface MICA/B expression in cancers of the digestive system was found associated with increased survival.
Collapse
|
106
|
Paul S, Lal G. The Molecular Mechanism of Natural Killer Cells Function and Its Importance in Cancer Immunotherapy. Front Immunol 2017; 8:1124. [PMID: 28955340 PMCID: PMC5601256 DOI: 10.3389/fimmu.2017.01124] [Citation(s) in RCA: 467] [Impact Index Per Article: 66.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/28/2017] [Indexed: 12/31/2022] Open
Abstract
Natural killer (NK) cells are innate immune cells that show strong cytolytic function against physiologically stressed cells such as tumor cells and virus-infected cells. NK cells show a broad array of tissue distribution and phenotypic variability. NK cells express several activating and inhibitory receptors that recognize the altered expression of proteins on target cells and control the cytolytic function. NK cells have been used in several clinical trials to control tumor growth. However, the results are encouraging only in hematological malignancies but not very promising in solid tumors. Increasing evidence suggests that tumor microenvironment regulate the phenotype and function of NK cells. In this review, we discussed the NK cell phenotypes and its effector function and impact of the tumor microenvironment on effector and cytolytic function of NK cells. We also summarized various NK cell-based immunotherapeutic strategies used in the past and the possibilities to improve the function of NK cell for the better clinical outcome.
Collapse
Affiliation(s)
- Sourav Paul
- National Centre for Cell Science, Pune, India
| | | |
Collapse
|
107
|
Angka L, Khan ST, Kilgour MK, Xu R, Kennedy MA, Auer RC. Dysfunctional Natural Killer Cells in the Aftermath of Cancer Surgery. Int J Mol Sci 2017; 18:ijms18081787. [PMID: 28817109 PMCID: PMC5578175 DOI: 10.3390/ijms18081787] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 08/13/2017] [Accepted: 08/14/2017] [Indexed: 02/06/2023] Open
Abstract
The physiological changes that occur immediately following cancer surgeries initiate a chain of events that ultimately result in a short pro-, followed by a prolonged anti-, inflammatory period. Natural Killer (NK) cells are severely affected during this period in the recovering cancer patient. NK cells play a crucial role in anti-tumour immunity because of their innate ability to differentiate between malignant versus normal cells. Therefore, an opportunity arises in the aftermath of cancer surgery for residual cancer cells, including distant metastases, to gain a foothold in the absence of NK cell surveillance. Here, we describe the post-operative environment and how the release of sympathetic stress-related factors (e.g., cortisol, prostaglandins, catecholamines), anti-inflammatory cytokines (e.g., IL-6, TGF-β), and myeloid derived suppressor cells, mediate NK cell dysfunction. A snapshot of current and recently completed clinical trials specifically addressing NK cell dysfunction post-surgery is also discussed. In collecting and summarizing results from these different aspects of the surgical stress response, a comprehensive view of the NK cell suppressive effects of surgery is presented. Peri-operative therapies to mitigate NK cell suppression in the post-operative period could improve curative outcomes following cancer surgery.
Collapse
Affiliation(s)
- Leonard Angka
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada.
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| | - Sarwat T Khan
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada.
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| | - Marisa K Kilgour
- Deeley Research Centre, BC Cancer Agency, Victoria, BC V8R 6V5, Canada.
| | - Rebecca Xu
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada.
| | - Michael A Kennedy
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada.
| | - Rebecca C Auer
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada.
- Department of Surgery, University of Ottawa, Ottawa, ON K1H 8L6, Canada.
| |
Collapse
|
108
|
Zhu L, Li XJ, Kalimuthu S, Gangadaran P, Lee HW, Oh JM, Baek SH, Jeong SY, Lee SW, Lee J, Ahn BC. Natural Killer Cell (NK-92MI)-Based Therapy for Pulmonary Metastasis of Anaplastic Thyroid Cancer in a Nude Mouse Model. Front Immunol 2017; 8:816. [PMID: 28785259 PMCID: PMC5519537 DOI: 10.3389/fimmu.2017.00816] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/27/2017] [Indexed: 12/22/2022] Open
Abstract
Objective Natural killer (NK) cells represent the third largest population of lymphocytes, and they play an important role in immune surveillance against tumors. The lungs are a common metastatic site for anaplastic thyroid cancer (ATC), and metastasis is one of the most frequent causes of mortality in this type of cancer. In the current study, we evaluated the effects of NK cell-based immunotherapy for pulmonary metastasis of ATC and determined how it affects the effector molecules of NK cells. Methods Human NK cells (NK-92MI) were retrovirally transduced to express the effluc gene. Human ATC cells (CAL-62) were transduced with the effluc and Rluc genes. The cytotoxicity of NK cells against CAL-62 cells was assessed using the CytoTox 96® Non-Radioactive Cytotoxicity Assay system. Pulmonary metastases of ATC were developed by i.v. injection of CAL-62, and metastasis growth was monitored using bioluminescence imaging (BLI). To treat the metastases, five million NK-92MI cells were injected twice into the caudal vein of nude mice. To assess the targetability of NK cells to ATC tumors, NK-92MI cells expressing the effluc gene (NK/F) were administered through the tail vein of nude mice with a pulmonary metastasis or tumor xenograft. BLI was subsequently performed at 1, 3, 24, and 48 h. Results NK/F and CAL-62 cells expressing the effluc or Rluc gene (CAL-62/F, CAL-62/R) were successfully established. Expression of the effluc and Rluc genes in NK/F, CAL-62/F, and CAL-62/R cells was verified by RT-polymerase chain reaction, western blotting, and luciferase assay. After coculture of NK-92MI and CAL-62/F cells for 24 h, the BLI signal intensity of CAL-62/F cells proportionally decreased with the number of cocultured NK cells. An ATC pulmonary metastasis mouse model was successfully generated, and NK cells significantly inhibited the growth of the metastasis (p < 0.01). The NK/F cells exhibited targetability to the pulmonary metastasis and tumor xenograft in the mouse model. Conclusion The results of present study suggest that NK cells are able to target ATC tumors and that NK cell-based immunotherapy may serve as an effective therapeutic approach for pulmonary metastases of ATC.
Collapse
Affiliation(s)
- Liya Zhu
- Department of Nuclear Medicine, Kyungpook School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Xiu Juan Li
- Department of Nuclear Medicine, Kyungpook School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea.,Department of Radiology, Taian City Central Hospital, Taian, China
| | - Senthilkumar Kalimuthu
- Department of Nuclear Medicine, Kyungpook School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, Kyungpook School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, Kyungpook School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Ji Min Oh
- Department of Nuclear Medicine, Kyungpook School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Se Hwan Baek
- Department of Nuclear Medicine, Kyungpook School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| |
Collapse
|
109
|
Zhu L, Kalimuthu S, Gangadaran P, Oh JM, Lee HW, Baek SH, Jeong SY, Lee SW, Lee J, Ahn BC. Exosomes Derived From Natural Killer Cells Exert Therapeutic Effect in Melanoma. Theranostics 2017; 7:2732-2745. [PMID: 28819459 PMCID: PMC5558565 DOI: 10.7150/thno.18752] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/08/2017] [Indexed: 12/13/2022] Open
Abstract
Objective: Exosomes are nanovesicles that are released from normal and tumor cells and are detectable in cell culture supernatant and human biological fluids. Although previous studies have explored exosomes released from cancer cells, little is understood regarding the functions of exosomes released by normal cells. Natural killer (NK) cells display rapid immunity to metastatic or hematological malignancies, and efforts have been undertaken to clinically exploit the antitumor properties of NK cells. However, the characteristics and functions of exosomes derived from NK cells remain unknown. In this study, we explored NK cell-derived exosome-mediated antitumor effects against aggressive melanoma in vitro and in vivo. Methods: B16F10 cells were transfected with enhanced firefly luciferase (effluc) and thy1.1 genes, and thy1.1-positive cells were immunoselected using microbeads. The resulting B16F10/effluc cells were characterized using reverse transcriptase polymerase chain reaction (RT-PCR), western blotting, and luciferase activity assays. Exosomes derived from NK-92MI cells (NK-92 Exo) were isolated by ultracentrifugation and density gradient ultracentrifugation. NK-92 Exo were characterized by transmission electron microscopy and western blotting. We also performed an enzyme-linked immunosorbent assay to measure cytokines retained in NK-92 Exo cells. The in vitro cytotoxicity of NK-92 Exo against the cancer cells was determined using a bioluminescence imaging system (BLI) and CCK-8 assays. To investigate the possible side effects of NK-92 Exo on healthy cells, we also performed the BLI and CCK-8 assays using the human kidney Phoenix™-Ampho cell line. Flow cytometry and western blotting confirmed that NK-92 Exo induced apoptosis in the B16F10/effluc cells. In vivo, we used a B16F10/effluc cell xenograft model to detect the immunotherapeutic effect of NK-92 Exo. We injected NK-92 Exo into tumors, and tumor growth progression was monitored using the IVIS Lumina imaging system and ultrasound imaging. Tumor mass was monitored after in vivo experiments. Results: RT-PCR and western blotting confirmed effluc gene expression and protein levels in B16F10/effluc cells. B16F10/effluc activity was found to increase with increasing cell numbers, using BLI assay. For NK-92 Exo characterization, western blotting was performed on both ultracentrifuged and density gradient-isolated exosomes. The results confirmed that NK cell-derived exosomes express two typical exosome proteins, namely CD63 and ALIX. We demonstrated by western blot analysis that NK-92 Exo presented two functional NK proteins, namely perforin and FasL. Moreover, we confirmed the membrane expression of FasL. The enzyme-linked immunosorbent assay results indicated that NK-92 Exo can secrete tumor necrosis factor (TNF)-α, which affected the cell proliferation signaling pathway. The antitumor effect of NK-92 Exo against B16F10/effluc cells in vitro was confirmed by BLI (p < 0.001) and CCK-8 assays (p < 0.001). Furthermore, in normal healthy cells, even after 24 h of co-culture, NK-92 Exo did not exhibit significant side effects. In the in vivo experiments, tumors in the vehicle control group were significantly increased, compared with those in the NK-92 Exo-treated group (p < 0.05). Conclusion: The results of the current study suggest that exosomes derived from NK cells exert cytotoxic effects on melanoma cells and thus warrant further development as a potential immunotherapeutic strategy for cancer.
Collapse
|
110
|
Shenouda MM, Gillgrass A, Nham T, Hogg R, Lee AJ, Chew MV, Shafaei M, Aarts C, Lee DA, Hassell J, Bane A, Dhesy-Thind S, Ashkar AA. Ex vivo expanded natural killer cells from breast cancer patients and healthy donors are highly cytotoxic against breast cancer cell lines and patient-derived tumours. Breast Cancer Res 2017; 19:76. [PMID: 28668076 PMCID: PMC5493877 DOI: 10.1186/s13058-017-0867-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 06/14/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Natural killer (NK) cells play a critical role in cancer immunosurveillance. Recent developments in NK cell ex-vivo expansion makes it possible to generate millions of activated NK cells from a small volume of peripheral blood. We tested the functionality of ex vivo expanded NK cells in vitro against breast cancer cell lines and in vivo using a xenograft mouse model. The study aim was to assess functionality and phenotype of expanded NK cells from breast cancer patients against breast cancer cell lines and autologous primary tumours. METHODS We used a well-established NK cell co-culture system to expand NK cells ex vivo from healthy donors and breast cancer patients and examined their surface marker expression. Moreover, we tested the ability of expanded NK cells to lyse the triple negative breast cancer and HER2-positive breast cancer cell lines MDA-MB-231 and MDA-MB-453, respectively. We also tested their ability to prevent tumour growth in vivo using a xenograft mouse model. Finally, we tested the cytotoxicity of expanded NK cells against autologous and allogeneic primary breast cancer tumours in vitro. RESULTS After 3 weeks of culture we observed over 1000-fold expansion of NK cells isolated from either breast cancer patients or healthy donors. We also showed that the phenotype of expanded NK cells is comparable between those from healthy donors and cancer patients. Moreover, our results confirm the ability of ex vivo expanded NK cells to lyse tumour cell lines in vitro. While the cell lines examined had differential sensitivity to NK cell killing we found this was correlated with level of major histocompatibility complex (MHC) class I expression. In our in vivo model, NK cells prevented tumour establishment and growth in immunocompromised mice. Finally, we showed that NK cells expanded from the peripheral blood of breast cancer patients show high cytotoxicity against allogeneic and autologous patient-derived tumour cells in vitro. CONCLUSION NK cells from breast cancer patients can be expanded similarly to those from healthy donors, have a high cytotoxic ability against breast cancer cell lines and patient-derived tumour cells, and can be compatible with current cancer treatments to restore NK cell function in cancer patients.
Collapse
Affiliation(s)
- Mira M. Shenouda
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, 1280 Main Street West, MDCL 4015 Hamilton, ON Canada
| | - Amy Gillgrass
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, 1280 Main Street West, MDCL 4015 Hamilton, ON Canada
| | - Tina Nham
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, 1280 Main Street West, MDCL 4015 Hamilton, ON Canada
| | - Richard Hogg
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, 1280 Main Street West, MDCL 4015 Hamilton, ON Canada
| | - Amanda J. Lee
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, 1280 Main Street West, MDCL 4015 Hamilton, ON Canada
| | - Marianne V. Chew
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, 1280 Main Street West, MDCL 4015 Hamilton, ON Canada
| | - Mahsa Shafaei
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, 1280 Main Street West, MDCL 4015 Hamilton, ON Canada
| | - Craig Aarts
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, 1280 Main Street West, MDCL 4015 Hamilton, ON Canada
| | - Dean A. Lee
- Cellular Therapy and Cancer Immunology Program, Department of Hematology/Oncology and BMT, Nationwide Children’s Hospital, The Ohio State University Comprehensive Cancer Center, Ohio, USA
| | - John Hassell
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, 1280 Main Street West, MDCL 4015 Hamilton, ON Canada
| | - Anita Bane
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, 1280 Main Street West, MDCL 4015 Hamilton, ON Canada
- Department of Oncology, McMaster University, Hamilton, ON Canada
| | | | - Ali A. Ashkar
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, 1280 Main Street West, MDCL 4015 Hamilton, ON Canada
| |
Collapse
|
111
|
Bhattacharya S, Muhammad N, Steele R, Kornbluth J, Ray RB. Bitter Melon Enhances Natural Killer-Mediated Toxicity against Head and Neck Cancer Cells. Cancer Prev Res (Phila) 2017; 10:337-344. [PMID: 28465362 PMCID: PMC5499682 DOI: 10.1158/1940-6207.capr-17-0046] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/10/2017] [Accepted: 04/25/2017] [Indexed: 01/06/2023]
Abstract
Natural killer (NK) cells are one of the major components of innate immunity, with the ability to mediate antitumor activity. Understanding the role of NK-cell-mediated tumor killing in controlling of solid tumor growth is still in the developmental stage. We have shown recently that bitter melon extract (BME) modulates the regulatory T cell (Treg) population in head and neck squamous cell carcinoma (HNSCC). However, the role of BME in NK-cell modulation against HNSCC remains unknown. In this study, we investigated whether BME can enhance the NK-cell killing activity against HNSCC cells. Our results indicated that treatment of human NK-cell line (NK3.3) with BME enhances ability to kill HNSCC cells. BME increases granzyme B accumulation and translocation/accumulation of CD107a/LAMP1 in NK3.3 cells exposed to BME. Furthermore, an increase in cell surface expression of CD16 and NKp30 in BME-treated NK3.3 cells was observed when cocultured with HNSCC cells. Collectively, our results demonstrated for the first time that BME augments NK-cell-mediated HNSCC killing activity, implicating an immunomodulatory role of BME. Cancer Prev Res; 10(6); 337-44. ©2017 AACR.
Collapse
MESH Headings
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/immunology
- Cell Line, Tumor
- Cytotoxicity, Immunologic/drug effects
- GPI-Linked Proteins/metabolism
- Granzymes/metabolism
- Head and Neck Neoplasms/drug therapy
- Head and Neck Neoplasms/immunology
- Humans
- Immunomodulation/drug effects
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lysosomal Membrane Proteins/metabolism
- Medicine, Traditional/methods
- Momordica charantia/chemistry
- Natural Cytotoxicity Triggering Receptor 3/metabolism
- Plant Extracts/pharmacology
- Plant Extracts/therapeutic use
- Receptors, IgG/metabolism
- Squamous Cell Carcinoma of Head and Neck
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
| | - Naoshad Muhammad
- Department of Pathology, Saint Louis University, St. Louis, Missouri
| | - Robert Steele
- Department of Pathology, Saint Louis University, St. Louis, Missouri
| | - Jacki Kornbluth
- Department of Pathology, Saint Louis University, St. Louis, Missouri
- Saint Louis VA Health Care System, St. Louis, Missouri
| | - Ratna B Ray
- Department of Pathology, Saint Louis University, St. Louis, Missouri.
| |
Collapse
|
112
|
Zhang C, Oberoi P, Oelsner S, Waldmann A, Lindner A, Tonn T, Wels WS. Chimeric Antigen Receptor-Engineered NK-92 Cells: An Off-the-Shelf Cellular Therapeutic for Targeted Elimination of Cancer Cells and Induction of Protective Antitumor Immunity. Front Immunol 2017; 8:533. [PMID: 28572802 PMCID: PMC5435757 DOI: 10.3389/fimmu.2017.00533] [Citation(s) in RCA: 221] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 04/21/2017] [Indexed: 12/27/2022] Open
Abstract
Significant progress has been made in recent years toward realizing the potential of natural killer (NK) cells for cancer immunotherapy. NK cells can respond rapidly to transformed and stressed cells and have the intrinsic potential to extravasate and reach their targets in almost all body tissues. In addition to donor-derived primary NK cells, also the established NK cell line NK-92 is being developed for adoptive immunotherapy, and general safety of infusion of irradiated NK-92 cells has been established in phase I clinical trials with clinical responses observed in some of the cancer patients treated. To enhance their therapeutic utility, NK-92 cells have been modified to express chimeric antigen receptors (CARs) composed of a tumor-specific single chain fragment variable antibody fragment fused via hinge and transmembrane regions to intracellular signaling moieties such as CD3ζ or composite signaling domains containing a costimulatory protein together with CD3ζ. CAR-mediated activation of NK cells then bypasses inhibitory signals and overcomes NK resistance of tumor cells. In contrast to primary NK cells, CAR-engineered NK-92 cell lines suitable for clinical development can be established from molecularly and functionally well-characterized single cell clones following good manufacturing practice-compliant procedures. In preclinical in vitro and in vivo models, potent antitumor activity of NK-92 variants targeted to differentiation antigens expressed by hematologic malignancies, and overexpressed or mutated self-antigens associated with solid tumors has been found, encouraging further development of CAR-engineered NK-92 cells. Importantly, in syngeneic mouse tumor models, induction of endogenous antitumor immunity after treatment with CAR-expressing NK-92 cells has been demonstrated, resulting in cures and long-lasting immunological memory protecting against tumor rechallenge at distant sites. Here, we summarize the current status and future prospects of CAR-engineered NK-92 cells as off-the-shelf cellular therapeutics, with special emphasis on ErbB2 (HER2)-specific NK-92 cells that are approaching clinical application.
Collapse
Affiliation(s)
- Congcong Zhang
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pranav Oberoi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Sarah Oelsner
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Anja Waldmann
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Aline Lindner
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Torsten Tonn
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Red Cross Blood Donation Service North-East, Institute for Transfusion Medicine, Dresden, Germany.,Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
| | - Winfried S Wels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
113
|
Linking tumor glycolysis and immune evasion in cancer: Emerging concepts and therapeutic opportunities. Biochim Biophys Acta Rev Cancer 2017; 1868:212-220. [PMID: 28400131 DOI: 10.1016/j.bbcan.2017.04.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 03/30/2017] [Accepted: 04/06/2017] [Indexed: 12/17/2022]
Abstract
Metabolic reprogramming and immune evasion are two hallmarks of cancer. Metabolic reprogramming is exemplified by cancer's propensity to utilize glucose at an exponential rate which in turn is linked with "aerobic glycolysis", popularly known as the "Warburg effect". Tumor glycolysis is pivotal for the efficient management of cellular bioenergetics and uninterrupted cancer growth. Mounting evidence suggests that tumor glycolysis also plays a key role in instigating immunosuppressive networks that are critical for cancer cells to escape immune surveillance ("immune evasion"). Recent data show that induction of cellular stress or metabolic dysregulation sensitize cancer cells to antitumor immune cells implying that metabolic reprogramming and immune evasion harmonize during cancer progression. However, the molecular link between these two hallmarks of cancer remains obscure. In this review the molecular intricacies of tumor glycolysis that facilitate immune evasion has been discussed in the light of recent research to explore immunotherapeutic potential of targeting cancer metabolism.
Collapse
|
114
|
Natural Killer Cells in the Orchestration of Chronic Inflammatory Diseases. J Immunol Res 2017; 2017:4218254. [PMID: 28428965 PMCID: PMC5385901 DOI: 10.1155/2017/4218254] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/04/2017] [Accepted: 01/18/2017] [Indexed: 02/06/2023] Open
Abstract
Inflammation, altered immune cell phenotype, and functions are key features shared by diverse chronic diseases, including cardiovascular, neurodegenerative diseases, diabetes, metabolic syndrome, and cancer. Natural killer cells are innate lymphoid cells primarily involved in the immune system response to non-self-components but their plasticity is largely influenced by the pathological microenvironment. Altered NK phenotype and function have been reported in several pathological conditions, basically related to impaired or enhanced toxicity. Here we reviewed and discussed the role of NKs in selected, different, and “distant” chronic diseases, cancer, diabetes, periodontitis, and atherosclerosis, placing NK cells as crucial orchestrator of these pathologic conditions.
Collapse
|
115
|
Li Y, Lu H, Gu Y, Ning Z, Cao T, Chen C, Hu C, Tao M. Enhancement of NK cells proliferation and function by Shikonin. Immunopharmacol Immunotoxicol 2017; 39:124-130. [PMID: 28303727 DOI: 10.1080/08923973.2017.1299174] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Yan Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
- Department of Oncology, The Affiliated Wujiang Hospital of Nantong University, Suzhou, P.R. China
| | - Hailin Lu
- Department of Oncology, The Affiliated Wujiang Hospital of Nantong University, Suzhou, P.R. China
| | - Yongchun Gu
- Central Laboratory, The Affiliated Wujiang Hospital of Nantong University, Suzhou, P.R. China
| | - Zhiqiang Ning
- Department of Oncology, The Affiliated Wujiang Hospital of Nantong University, Suzhou, P.R. China
| | - Tinghua Cao
- Department of Oncology, The Affiliated Wujiang Hospital of Nantong University, Suzhou, P.R. China
| | - Chao Chen
- Department of Oncology, The Affiliated Wujiang Hospital of Nantong University, Suzhou, P.R. China
| | - Chengru Hu
- Department of Oncology, The Affiliated Wujiang Hospital of Nantong University, Suzhou, P.R. China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| |
Collapse
|
116
|
Miao X, Chen Y, Hao K, Zheng M, Chen B, Li K, Wang Y, Zhang W, Zhang Y, Mou X, Jiang S, Wang Z. CD103 + Cell Growth Factor Flt3L Enhances the Efficacy of Immune Checkpoint Blockades in Murine Glioblastoma Model. Oncol Res 2017; 26:173-182. [PMID: 28109087 PMCID: PMC7844614 DOI: 10.3727/096504017x14841698396865] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Glioblastoma is a lethal disease featuring a high proliferation of tumor cells, excessive angiogenesis, and heavy drug resistance. The overall survival of glioblastoma patients has been dismal, even with an intensive standard of care. Recent advances in immune checkpoint blockades are changing the treatment of cancers. However, the efficacy of immune checkpoint blockades in glioblastoma is still unclear. Here we investigated the roles of CD103+ cells in regulating the effect of immune checkpoint blockades in glioblastoma mouse models. Our findings indicated that the murine glioblastoma model was not sensitive to immune checkpoint blockades. Flt3L, a growth factor for CD103+ cells, could significantly increase the number of CD103+ dendritic cells in the murine glioblastoma model and, thus, sensitize murine glioblastoma to immune checkpoint blockades. Downstream analysis indicated that the Flt3L and immune checkpoint blockade combination increased the number of tumor-infiltrating CD8+ cells, decreased immune checkpoint expression, and therefore enhanced the antitumor immune response in the murine glioblastoma model. These findings suggested that Flt3L could enhance the efficacy of immune checkpoint blockades in glioblastoma via expanding CD103+ dendritic cells and downstream antitumor immune response.
Collapse
Affiliation(s)
- Xiaolin Miao
- Eye Hospital of Wenzhou Medical University, Zhejiang, P.R. China
| | - Yiqi Chen
- Eye Hospital of Wenzhou Medical University, Zhejiang, P.R. China
| | - Ke Hao
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Zhejiang, P.R. China
| | - Meiqin Zheng
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, P.R. China
| | - Bingyu Chen
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Zhejiang, P.R. China
| | - Kaiqiang Li
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Zhejiang, P.R. China
| | - Ying Wang
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Zhejiang, P.R. China
| | - Wei Zhang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Zhejiang, P.R. China
| | - Yu Zhang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Zhejiang, P.R. China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Zhejiang, P.R. China
| | - Shanshan Jiang
- Department of Gynecology, Zhejiang Provincial People's Hospital, Zhejiang, P.R. China
| | - Zhen Wang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, P.R. China
| |
Collapse
|
117
|
Poggi A, Giuliani M. Mesenchymal Stromal Cells Can Regulate the Immune Response in the Tumor Microenvironment. Vaccines (Basel) 2016; 4:E41. [PMID: 27834810 PMCID: PMC5192361 DOI: 10.3390/vaccines4040041] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/01/2016] [Accepted: 10/31/2016] [Indexed: 12/19/2022] Open
Abstract
The tumor microenvironment is a good target for therapy in solid tumors and hematological malignancies. Indeed, solid tumor cells' growth and expansion can influence neighboring cells' behavior, leading to a modulation of mesenchymal stromal cell (MSC) activities and remodeling of extracellular matrix components. This leads to an altered microenvironment, where reparative mechanisms, in the presence of sub-acute inflammation, are not able to reconstitute healthy tissue. Carcinoma cells can undergo epithelial mesenchymal transition (EMT), a key step to generate metastasis; these mesenchymal-like cells display the functional behavior of MSC. Furthermore, MSC can support the survival and growth of leukemic cells within bone marrow participating in the leukemic cell niche. Notably, MSC can inhibit the anti-tumor immune response through either carcinoma-associated fibroblasts or bone marrow stromal cells. Experimental data have indicated their relevance in regulating cytolytic effector lymphocytes of the innate and adaptive arms of the immune system. Herein, we will discuss some of the evidence in hematological malignancies and solid tumors. In particular, we will focus our attention on the means by which it is conceivable to inhibit MSC-mediated immune suppression and trigger anti-tumor innate immunity.
Collapse
Affiliation(s)
- Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS AOU San Martino IST, 16132 Genoa, Italy.
| | - Massimo Giuliani
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg City L-1526, Luxembourg.
| |
Collapse
|
118
|
Abstract
Lipid metabolism, in particular the synthesis of fatty acids (FAs), is an essential cellular process that converts nutrients into metabolic intermediates for membrane biosynthesis, energy storage and the generation of signalling molecules. This Review explores how different aspects of FA synthesis promote tumorigenesis and tumour progression. FA synthesis has received substantial attention as a potential target for cancer therapy, but strategies to target this process have not yet translated into clinical practice. Furthermore, efforts to target this pathway must consider the influence of the tumour microenvironment.
Collapse
Affiliation(s)
- Florian Röhrig
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, Am Hubland, 97074 Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Josef-Schneider-Strasse 6, 97080 Würzburg, Germany
| | - Almut Schulze
- Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, Am Hubland, 97074 Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Josef-Schneider-Strasse 6, 97080 Würzburg, Germany
| |
Collapse
|
119
|
Xu W, Yang Z, Lu N. From pathogenesis to clinical application: insights into exosomes as transfer vectors in cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:156. [PMID: 27686593 PMCID: PMC5043625 DOI: 10.1186/s13046-016-0429-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 09/15/2016] [Indexed: 12/21/2022]
Abstract
Exosomes are nanoscale extracellular membrane vesicles that are created by the fusion of an intracellular multivesicular body with the cell membrane. They are widely distributed in serum, urine, saliva and other biological fluids. As important transfer vectors for intercellular communication and genetic material, exosomes can stimulate target cells directly via receptor-mediated interactions or via the transfer of various bioactive molecules, such as cell membrane receptors, proteins, mRNAs and microRNAs, thus exerting their biological functions. This review focuses on the biological characteristics of exosomes, as well as their role and underlying mechanisms of action in the evolution of tumor formation, metastasis, drug resistance and other malignant behaviors. Additionally, this review emphasizes the potential applications of exosomes in the treatment of tumors. Further research may provide new ideas and methods to establish effective, exosome-based strategies for the early diagnosis and treatment of tumors.
Collapse
Affiliation(s)
- Wenting Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 YongWaizheng Street, Nanchang, Jiangxi, 330006, China
| | - Zhen Yang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 YongWaizheng Street, Nanchang, Jiangxi, 330006, China.
| | - Nonghua Lu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 YongWaizheng Street, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
120
|
Wabnitz GH, Balta E, Schindler S, Kirchgessner H, Jahraus B, Meuer S, Samstag Y. The pro-oxidative drug WF-10 inhibits serial killing by primary human cytotoxic T-cells. Cell Death Discov 2016; 2:16057. [PMID: 27551545 PMCID: PMC4979520 DOI: 10.1038/cddiscovery.2016.57] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/05/2016] [Indexed: 12/22/2022] Open
Abstract
Cytotoxic T-cells (CTLs) play an important role in many immune-mediated inflammatory diseases. Targeting cytotoxicity of CTLs would allow to interfere with immune-mediated tissue destruction. Here we demonstrate that WF-10, a pro-oxidative compound, inhibits CTL-mediated cytotoxicity. WF-10 did not influence early steps of target-cell killing, but impaired the ability of CTLs to detach from the initial target cell and to move to a second target cell. This reduced serial killing was accompanied by stronger enrichment of the adhesion molecule LFA-1 in the cytolytic immune synapse. LFA-1 clustering requires activation of the actin-bundling protein L-plastin and was accordingly diminished in L-plastin knockdown cells. Interestingly, WF-10 likely acts through regulating L-plastin: (I) It induced L-plastin activation through phosphorylation leading to enhanced LFA-1-mediated cell adhesion, and, importantly, (II) WF-10 lost its influence on target-cell killing in L-plastin knockdown cells. Finally, we demonstrate that WF-10 can improve immunosuppression by conventional drugs. Thus, while cyclosporine A alone had no significant effect on cytotoxicity of CTLs, a combination of cyclosporine A and WF-10 blocked target-cell killing synergistically. Together, our findings suggest that WF-10 – either alone or in combination with conventional immunosuppressive drugs – may be efficient to control progression of diseases, in which CTLs are crucially involved.
Collapse
Affiliation(s)
- G H Wabnitz
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| | - E Balta
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| | - S Schindler
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| | - H Kirchgessner
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| | - B Jahraus
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| | - S Meuer
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| | - Y Samstag
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| |
Collapse
|
121
|
Curtis KK, Wong WW, Ross HJ. Past approaches and future directions for targeting tumor hypoxia in squamous cell carcinomas of the head and neck. Crit Rev Oncol Hematol 2016; 103:86-98. [DOI: 10.1016/j.critrevonc.2016.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 04/04/2016] [Accepted: 05/10/2016] [Indexed: 12/27/2022] Open
|
122
|
Mahasa KJ, Ouifki R, Eladdadi A, Pillis LD. Mathematical model of tumor-immune surveillance. J Theor Biol 2016; 404:312-330. [PMID: 27317864 DOI: 10.1016/j.jtbi.2016.06.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 06/09/2016] [Accepted: 06/09/2016] [Indexed: 12/26/2022]
Abstract
We present a novel mathematical model involving various immune cell populations and tumor cell populations. The model describes how tumor cells evolve and survive the brief encounter with the immune system mediated by natural killer (NK) cells and the activated CD8(+) cytotoxic T lymphocytes (CTLs). The model is composed of ordinary differential equations describing the interactions between these important immune lymphocytes and various tumor cell populations. Based on up-to-date knowledge of immune evasion and rational considerations, the model is designed to illustrate how tumors evade both arms of host immunity (i.e. innate and adaptive immunity). The model predicts that (a) an influx of an external source of NK cells might play a crucial role in enhancing NK-cell immune surveillance; (b) the host immune system alone is not fully effective against progression of tumor cells; (c) the development of immunoresistance by tumor cells is inevitable in tumor immune surveillance. Our model also supports the importance of infiltrating NK cells in tumor immune surveillance, which can be enhanced by NK cell-based immunotherapeutic approaches.
Collapse
Affiliation(s)
- Khaphetsi Joseph Mahasa
- DST/NRF Centre of Excellence in Epidemiological Modelling and Analysis (SACEMA), University of Stellenbosch, Stellenbosch, South Africa.
| | - Rachid Ouifki
- DST/NRF Centre of Excellence in Epidemiological Modelling and Analysis (SACEMA), University of Stellenbosch, Stellenbosch, South Africa
| | | | | |
Collapse
|
123
|
Smits NC, Coupet TA, Godbersen C, Sentman CL. Designing multivalent proteins based on natural killer cell receptors and their ligands as immunotherapy for cancer. Expert Opin Biol Ther 2016; 16:1105-12. [PMID: 27248342 DOI: 10.1080/14712598.2016.1195364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Natural killer (NK) cells are an important component of the innate immune system that play a key role in host immunity against cancer. NK cell recognition and activation is based on cell surface receptors recognizing specific ligands that are expressed on many types of tumor cells. Some of these receptors are capable of activating NK cell function while other receptors inhibit NK cell function. Therapeutic approaches to treat cancer have been developed based on preventing NK cell inhibition or using NK cell receptors and their ligands to activate NK cells or T cells to destroy tumor cells. AREAS COVERED This article describes the various strategies for targeting NK cell receptors and NK cell receptor ligands using multivalent proteins to activate immunity against cancer. EXPERT OPINION NK cell receptors work in synergy to activate NK cell effector responses. Effective anti-cancer strategies will need to not only kill tumor cells but must also lead to the destruction of the tumor microenvironment. Immunotherapy based on NK cells and their receptors has the capacity to accomplish this through triggering lymphocyte cytotoxicity and cytokine production.
Collapse
Affiliation(s)
- Nicole C Smits
- a Department of Microbiology and Immunology and the Center for Synthetic Immunity , The Geisel School of Medicine at Dartmouth , Lebanon , NH , USA
| | - Tiffany A Coupet
- a Department of Microbiology and Immunology and the Center for Synthetic Immunity , The Geisel School of Medicine at Dartmouth , Lebanon , NH , USA
| | - Claire Godbersen
- a Department of Microbiology and Immunology and the Center for Synthetic Immunity , The Geisel School of Medicine at Dartmouth , Lebanon , NH , USA
| | - Charles L Sentman
- a Department of Microbiology and Immunology and the Center for Synthetic Immunity , The Geisel School of Medicine at Dartmouth , Lebanon , NH , USA
| |
Collapse
|
124
|
Abstract
Metastasis is the underlying cause of death for the majority of breast cancer patients. Despite significant advances in recent years in basic research and clinical development, therapies that specifically target metastatic breast cancer remain inadequate, and represents the single greatest obstacle to reducing mortality of late-stage breast cancer. Recent efforts have leveraged genomic analysis of breast cancer and molecular dissection of tumor-stromal cross-talk to uncover a number of promising candidates for targeted treatment of metastatic breast cancer. Rational combinations of therapeutic agents targeting tumor-intrinsic properties and microenvironmental components provide a promising strategy to develop precision treatments with higher specificity and less toxicity. In this review, we discuss the emerging therapeutic targets in breast cancer metastasis, from tumor-intrinsic pathways to those that involve the host tissue components, including the immune system.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, United States
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, United States.
| |
Collapse
|
125
|
Velásquez SY, Killian D, Schulte J, Sticht C, Thiel M, Lindner HA. Short Term Hypoxia Synergizes with Interleukin 15 Priming in Driving Glycolytic Gene Transcription and Supports Human Natural Killer Cell Activities. J Biol Chem 2016; 291:12960-77. [PMID: 27129235 DOI: 10.1074/jbc.m116.721753] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Indexed: 01/08/2023] Open
Abstract
Natural killer (NK) cells induce apoptosis in infected and transformed cells and are important producers of immunoregulatory cytokines. Therefore, they operate under low oxygen conditions (hypoxia) in inflammatory and tumor environments. In vitro studies of NK cells are, however, commonly performed in ambient air (normoxia). We used global gene expression profiling to evaluate changes in transcriptional pathways in primary human NK cells following short term culture under hypoxia compared with normoxia and in response to interleukin 15 (IL-15) priming using a 2 × 2 factorial design. The largest contrasts observed were priming dependences for associations between hypoxia and the hypoxia-inducible factor (Hif) 1 signaling and glycolysis pathways. RT-PCR confirmed positive synergistic hypoxia/IL-15 interactions for genes of key regulatory and metabolic enzymes. IL-15 primes NK cells for effector functions, which were recently demonstrated to depend on glycolytic switching. We did not, however, observe important increases in glycolytic flux through hypoxia and priming alone. Chemical Hif-1α inhibition suggested equal importance of this transcription factor for glycolysis and energy production under normoxia and hypoxia. Hypoxia promoted secretion of CC chemokines Ccl3/4/5 and macrophage migration inhibitory factor. Unexpectedly, hypoxia also stimulated migration of NK cells through the extracellular matrix and shifted amounts of susceptible leukemia target cells toward late apoptosis in a cell killing assay. We conclude that short term hypoxia supports these activities by positively interacting with NK cell priming at the level of glycolytic gene transcription. Hypoxic conditioning of NK cells may thus benefit their use in cell-based immunotherapy of cancer.
Collapse
Affiliation(s)
- Sonia Y Velásquez
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| | - Doreen Killian
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| | - Jutta Schulte
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| | - Carsten Sticht
- Medical Research Center, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Manfred Thiel
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| | - Holger A Lindner
- From the Department of Anesthesiology and Surgical Intensive Care Medicine, and
| |
Collapse
|
126
|
Koguchi Y, Hoen HM, Bambina SA, Rynning MD, Fuerstenberg RK, Curti BD, Urba WJ, Milburn C, Bahjat FR, Korman AJ, Bahjat KS. Serum Immunoregulatory Proteins as Predictors of Overall Survival of Metastatic Melanoma Patients Treated with Ipilimumab. Cancer Res 2016; 75:5084-92. [PMID: 26627641 DOI: 10.1158/0008-5472.can-15-2303] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Treatment with ipilimumab improves overall survival (OS) in patients with metastatic melanoma. Because ipilimumab targets T lymphocytes and not the tumor itself, efficacy may be uniquely sensitive to immunomodulatory factors present at the time of treatment. We analyzed serum from patients with metastatic melanoma (247 of 273, 90.4%) randomly assigned to receive ipilimumab or gp100 peptide vaccine. We quantified candidate biomarkers at baseline and assessed the association of each using multivariate analyses. Results were confirmed in an independent cohort of similar patients (48 of 52, 92.3%) treated with ipilimumab. After controlling for baseline covariates, elevated chemokine (C-X-C motif) ligand 11 (CXCL11) and soluble MHC class I polypeptide-related chain A (sMICA) were associated with poor OS in ipilimumab-treated patients [log10 CXCL11: HR, 1.88; 95% confidence interval (CI), 1.14-3.12; P = 0.014; and log10 sMICA quadratic effect P = 0.066; sMICA (≥ 247 vs. 247): HR, 1.75; 95% CI, 1.02-3.01]. Multivariate analysis of an independent ipilimumab-treated cohort confirmed the association between log10 CXCL11 and OS (HR, 3.18; 95% CI, 1.13-8.95; P = 0.029), whereas sMICA was less strongly associated with OS [log10 sMICA quadratic effect P = 0.16; sMICA (≥ 247 vs. 247): HR, 1.48; 95% CI, 0.67-3.27]. High baseline CXCL11 and sMICA were associated with poor OS in patients with metastatic melanoma after ipilimumab treatment but not vaccine treatment. Thus, pretreatment CXCL11 and sMICA may represent predictors of survival benefit after ipilimumab treatment as well as therapeutic targets.
Collapse
Affiliation(s)
- Yoshinobu Koguchi
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, Oregon
| | - Helena M Hoen
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, Oregon
| | - Shelly A Bambina
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, Oregon
| | | | | | - Brendan D Curti
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, Oregon
| | - Walter J Urba
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, Oregon
| | | | | | | | - Keith S Bahjat
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, Oregon.
| |
Collapse
|
127
|
Song X, Hong SH, Kwon WT, Bailey LM, Basse P, Bartlett DL, Kwon YT, Lee YJ. Secretory TRAIL-Armed Natural Killer Cell-Based Therapy: In Vitro and In Vivo Colorectal Peritoneal Carcinomatosis Xenograft. Mol Cancer Ther 2016; 15:1591-601. [PMID: 27196776 DOI: 10.1158/1535-7163.mct-15-0937] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/03/2016] [Indexed: 12/31/2022]
Abstract
Since its discovery in 1995, TNF-related apoptosis-inducing ligand (TRAIL) has sparked growing interest among oncologists due to its remarkable ability to induce apoptosis in malignant human cells, but not in most normal cells. However, one major drawback is its fast clearance rate in vivo Thus, the development of an alternative means of delivery may increase the effectiveness of TRAIL-based therapy. In this study, we developed a secretory TRAIL-armed natural killer (NK) cell-based therapy and assessed its cytotoxic effects on colorectal cancer cells and its tumoricidal efficacy on colorectal peritoneal carcinomatosis xenograft. We generated genetically modified NK cells by transduction with a lentiviral vector consisting of a secretion signal domain, a trimerization domain, and an extracellular domain of the TRAIL gene. These NK cells secreted a glycosylated form of TRAIL fusion protein that induced apoptotic death. Intraperitoneally, but not intravenously, injected NK cells effectively accumulated at tumor sites, infiltrated tumor tissue, induced apoptosis, and delayed tumor growth. These results shed light on the therapeutic potential of genetically engineered NK cells to treat peritoneal carcinomatosis. Mol Cancer Ther; 15(7); 1591-601. ©2016 AACR.
Collapse
Affiliation(s)
- Xinxin Song
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Se-Hoon Hong
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William T Kwon
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lisa M Bailey
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Per Basse
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David L Bartlett
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yong Tae Kwon
- Protein Metabolism Medical Research Center and Department of Biomedical Science, College of Medicine, Seoul National University, Seoul, Korea
| | - Yong J Lee
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania. Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
128
|
Janji B, Viry E, Moussay E, Paggetti J, Arakelian T, Mgrditchian T, Messai Y, Noman MZ, Van Moer K, Hasmim M, Mami-Chouaib F, Berchem G, Chouaib S. The multifaceted role of autophagy in tumor evasion from immune surveillance. Oncotarget 2016; 7:17591-607. [PMID: 26910842 PMCID: PMC4951235 DOI: 10.18632/oncotarget.7540] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/06/2016] [Indexed: 12/18/2022] Open
Abstract
While autophagy is constitutively executed at basal level in all cells, it is activated in cancer cells in response to various microenvironmental stresses including hypoxia. It is now well established that autophagy can act both as tumor suppressor or tumor promoter. In this regard, several reports indicate that the tumor suppressor function of autophagy is associated with its ability to scavenge damaged oxidative organelles, thereby preventing the accumulation of toxic oxygen radicals and limiting the genome instability. Paradoxically, in developed tumors, autophagy can promote the survival of cancer cells and therefore operates as a cell resistance mechanism. The consensus appears to be that autophagy has a dual role in suppressing tumor initiation and in promoting the survival of established tumors. This has inspired significant interest in applying anti-autophagy therapies as an entirely new approach to cancer treatment. While much remains to be learned about the regulation and context-dependent biological role of autophagy, it is now well established that modulation of this process could be an attractive approach for the development of novel anticancer therapeutic strategies. In this review, we will summarize recent reports describing how tumor cells, by activating autophagy, manage to resist the immune cell attack. Data described in this review strongly argue that targeting autophagy may represent a conceptual realm for new immunotherapeutic strategies aiming to block the immune escape and therefore providing rational approach to future tumor immunotherapy design.
Collapse
Affiliation(s)
- Bassam Janji
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Elodie Viry
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Etienne Moussay
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Jérôme Paggetti
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Tsolère Arakelian
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Takouhie Mgrditchian
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Yosra Messai
- UMR 1186, Gustave Roussy Cancer Campus, Villejuif, France
| | - Muhammad Zaeem Noman
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
- UMR 1186, Gustave Roussy Cancer Campus, Villejuif, France
| | - Kris Van Moer
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Meriem Hasmim
- UMR 1186, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Guy Berchem
- Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
- Centre Hospitalier de Luxembourg, Department of Hemato-Oncology, Luxembourg City, Luxembourg
| | - Salem Chouaib
- UMR 1186, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
129
|
NK Cell-Dependent Growth Inhibition of Lewis Lung Cancer by Yu-Ping-Feng, an Ancient Chinese Herbal Formula. Mediators Inflamm 2016; 2016:3541283. [PMID: 27034590 PMCID: PMC4789500 DOI: 10.1155/2016/3541283] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 02/03/2016] [Indexed: 12/15/2022] Open
Abstract
Little is known about Yu-Ping-Feng (YPF), a typical Chinese herbal decoction, for its antitumor efficacy in non-small-cell lung cancer (NSCLC). Here, we found that YPF significantly inhibited the growth of Lewis lung cancer, prolonged the survival of tumor-bearing mice, promoted NK cell tumor infiltration, increased the population of NK cells in spleen, and enhanced NK cell-mediated killing activity. The growth suppression of tumors by YPF was significantly reversed by the depletion of NK cells. Furthermore, we found that YPF significantly downregulated the expression of TGF-β, indoleamine 2,3-dioxygenase, and IL-10 in tumor microenvironment. These results demonstrated that YPF has a NK cell-dependent inhibitory effect on Lewis lung cancer.
Collapse
|
130
|
A cancer treatment based on synergy between anti-angiogenic and immune cell therapies. J Theor Biol 2016; 394:197-211. [PMID: 26826488 DOI: 10.1016/j.jtbi.2016.01.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 07/06/2015] [Accepted: 01/13/2016] [Indexed: 02/06/2023]
Abstract
A mathematical model integrating tumor angiogenesis and tumor-targeted cytotoxicity by immune cells was developed to identify the therapeutic window of two distinct modes to treat cancer: (1) an anti-angiogenesis treatment based on the monoclonal antibody bevacizumab that targets tumor vasculature, and (2) immunotherapy involving the injection of unlicensed dendritic cells to boost the anti-tumor adaptive response. The angiogenic cytokine Vascular Endothelial Growth Factor (VEGF) contributes to the immunosuppressive tumor microenvironment, which is responsible for the short-lived therapeutic effect of cancer-targeted immunotherapy. The effect of immunosuppression on the width of the therapeutic window of each treatment was quantified. Experimental evidence has shown that neutralizing immunosuppressive cytokines results in an enhanced immune response against infections and chronic diseases. The model was used to determine treatment protocols involving the combination of anti-VEGF and unlicensed dendritic cell injections that enhance tumor regression. The model simulations predicted that the most effective method to treat tumors involves administering a series of biweekly anti-VEGF injections to disrupt angiogenic processes and limit tumor growth. The simulations also verified the hypothesis that reducing the concentration of the immunosuppressive factor VEGF prior to an injection of unlicensed dendritic cells enhances the cytotoxicity of CD8+ T cells and results in complete tumor elimination. Feasible treatment protocols for tumors that are diagnosed late and have grown to a relatively large size were identified.
Collapse
|
131
|
Li M, Xing S, Zhang H, Shang S, Li X, Ren B, Li G, Chang X, Li Y, Li W. A matrix metalloproteinase inhibitor enhances anti-cytotoxic T lymphocyte antigen-4 antibody immunotherapy in breast cancer by reprogramming the tumor microenvironment. Oncol Rep 2016; 35:1329-39. [PMID: 26752000 PMCID: PMC4750755 DOI: 10.3892/or.2016.4547] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/23/2015] [Indexed: 02/06/2023] Open
Abstract
Anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) treatment is effective for the treatment of primary tumors, but not sufficient for the treatment of metastatic tumors, likely owing to the effects of the tumor microenvironment. In this study, we aimed to determine the therapeutic effects of combined treatment with a matrix metalloproteinase (MMP) inhibitor (MMPI) and anti-CTLA-4 antibody in a breast cancer model in mice. Interestingly, combined treatment with MMPI and anti-CTLA-4 antibody delayed tumor growth and reduced lung and liver metastases compared with anti-CTLA-4 alone or vehicle treatment. The functions of the liver and kidney in mice in the different groups did not differ significantly compared with that in normal mice. The CD8+/CD4+ ratio in T cells in the spleen and tumor were increased after monotherapy or combined anti-CTLA-4 antibody plus MMPI therapy compared with that in vehicle-treated mice. Anti-CTLA-4 antibody plus MMPI therapy reduced the percentage of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) and decreased the Treg/Th17 cell ratio in the spleen compared with those in the vehicle-treated group. Additionally, anti-CTLA-4 antibody plus MMPI therapy reduced the percentages of regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and Th17 cells in tumors compared with that in the vehicle-treated group. Moreover, combined treatment with MMPI and anti-CTLA-4 antibody reduced the microvessel density (MVD) in tumors compared with that in vehicle or MMPI-treated mice. There was a negative correlation between MVD and the CD8+ T cell percentage, CD4+ T cell percentage, and CD8+/CD4+ T cell ratio, but a positive correlation with Tregs, Th17 cells, Treg/Th17 cell ratio, and MDSCs. Thus, these data demonstrated that addition of MMPI enhanced the effects of anti-CTLA-4 antibody treatment in a mouse model of breast cancer by delaying tumor growth and reducing metastases.
Collapse
Affiliation(s)
- Mingyue Li
- The Key Laboratory of Pathobiology, Ministry of Education, The College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Shugang Xing
- The Key Laboratory of Pathobiology, Ministry of Education, The College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Haiying Zhang
- The Key Laboratory of Pathobiology, Ministry of Education, The College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Siqi Shang
- The Key Laboratory of Pathobiology, Ministry of Education, The College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiangxiang Li
- The Key Laboratory of Pathobiology, Ministry of Education, The College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Bo Ren
- The Key Laboratory of Pathobiology, Ministry of Education, The College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Gaiyun Li
- The Key Laboratory of Pathobiology, Ministry of Education, The College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaona Chang
- The Key Laboratory of Pathobiology, Ministry of Education, The College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yilei Li
- The Key Laboratory of Pathobiology, Ministry of Education, The College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wei Li
- The Key Laboratory of Pathobiology, Ministry of Education, The College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
132
|
Liu LL, Pfefferle A, Yi Sheng VO, Björklund AT, Béziat V, Goodridge JP, Malmberg KJ. Harnessing adaptive natural killer cells in cancer immunotherapy. Mol Oncol 2015; 9:1904-17. [PMID: 26604011 PMCID: PMC5528731 DOI: 10.1016/j.molonc.2015.10.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 09/28/2015] [Accepted: 10/04/2015] [Indexed: 12/17/2022] Open
Abstract
Natural killer (NK) cells are innate lymphocytes with a refined ability to recognize transformed cells through a broad array of activating receptors in combination with stochastically expressed inhibitory receptors that recognize MHC-class I. Recent advances in NK cell biology have revealed a high degree of functional plasticity that can be attributed to dynamic cell-to-cell interactions in concert with transcriptional and epigenetic reprogramming. Here, we discuss how new insights into the adaptive behavior of NK cells pave the way for next generation cell therapy based on guided differentiation and selective expansion of particularly cytotoxic NK cell subsets.
Collapse
Affiliation(s)
- Lisa L Liu
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Aline Pfefferle
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Vincent Oei Yi Sheng
- The KG Jebsen Center for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, Norway; Institute for Cancer Research, Oslo University Hospital, Norway
| | - Andreas T Björklund
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; University Paris Descartes, Imagine Institute, Paris, France
| | - Jodie P Goodridge
- The KG Jebsen Center for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, Norway; Institute for Cancer Research, Oslo University Hospital, Norway
| | - Karl-Johan Malmberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden; The KG Jebsen Center for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, Norway; Institute for Cancer Research, Oslo University Hospital, Norway.
| |
Collapse
|
133
|
Cantoni C, Grauwet K, Pietra G, Parodi M, Mingari MC, Maria AD, Favoreel H, Vitale M. Role of NK cells in immunotherapy and virotherapy of solid tumors. Immunotherapy 2015; 7:861-82. [PMID: 26314197 DOI: 10.2217/imt.15.53] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Although natural killer (NK) cells are endowed with powerful cytolytic activity against cancer cells, their role in different therapies against solid tumors has not yet been fully elucidated. Their interactions with various elements of the tumor microenvironment as well as their possible effects in contributing to and/or limiting oncolytic virotherapy render this potential immunotherapeutic tool still difficult to exploit at the bedside. Here, we will review the current literature with the aim of providing new hints to manage this powerful cell type in future innovative therapies, such as the use of NK cells in combination with new cytokines, specific mAbs (inducing ADCC), Tyr-Kinase inhibitors, immunomodulatory drugs and/or the design of oncolytic viruses aimed at optimizing the effect of NK cells in virotherapy.
Collapse
Affiliation(s)
- Claudia Cantoni
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy.,Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,Istituto Giannina Gaslini, Genova, Italy
| | - Korneel Grauwet
- Laboratory of Immunology, Department of Virology, Parasitology & Immunology, Faculty of Veterinary Medicine, Ghent University, Belgium
| | - Gabriella Pietra
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy.,IRCCS AOU San Martino-IST Genova, Genova, Italy
| | - Monica Parodi
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Maria Cristina Mingari
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy.,Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,IRCCS AOU San Martino-IST Genova, Genova, Italy
| | - Andrea De Maria
- Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,IRCCS AOU San Martino-IST Genova, Genova, Italy.,Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
| | - Herman Favoreel
- Laboratory of Immunology, Department of Virology, Parasitology & Immunology, Faculty of Veterinary Medicine, Ghent University, Belgium
| | | |
Collapse
|
134
|
Yan H, Kamiya T, Suabjakyong P, Tsuji NM. Targeting C-Type Lectin Receptors for Cancer Immunity. Front Immunol 2015; 6:408. [PMID: 26379663 PMCID: PMC4547497 DOI: 10.3389/fimmu.2015.00408] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/26/2015] [Indexed: 12/21/2022] Open
Abstract
C-type lectin receptors (CLRs) are a large family of soluble and trans-membrane pattern recognition receptors that are widely and primarily expressed on myeloid cells. CLRs are important for cell-cell communication and host defense against pathogens through the recognition of specific carbohydrate structures. Similar to a family of Toll-like receptors, CLRs signaling are involved in the various steps for initiation of innate immune responses and promote secretion of soluble factors such as cytokines and interferons. Moreover, CLRs contribute to endocytosis and antigen presentation, thereby fine-tune adaptive immune responses. In addition, there may also be a direct activation of acquired immunity. On the other hand, glycans, such as mannose structures, Lewis-type antigens, or GalNAc are components of tumor antigens and ligate CLRs, leading to immunoregulation. Therefore, agonists or antagonists of CLRs signaling are potential therapeutic reagents for cancer immunotherapy. We aim to overview the current knowledge of CLRs signaling and the application of their ligands on tumor-associating immune response.
Collapse
Affiliation(s)
- Huimin Yan
- Immune Homeostasis Laboratory, Biomedical Research Institute, National Institute for Advanced Industrial Science and Technology (AIST) , Tsukuba , Japan ; Institute for Liver Disease, Fifth Hospital of Shijiazhuang , Shijiazhuang , China
| | - Tomomori Kamiya
- Immune Homeostasis Laboratory, Biomedical Research Institute, National Institute for Advanced Industrial Science and Technology (AIST) , Tsukuba , Japan ; Research Institute for Biomedical Sciences, Tokyo University of Science , Noda-shi , Japan
| | - Papawee Suabjakyong
- Immune Homeostasis Laboratory, Biomedical Research Institute, National Institute for Advanced Industrial Science and Technology (AIST) , Tsukuba , Japan ; Department of Clinical and Analytical Biochemistry, Graduate School of Pharmaceutical Sciences, Chiba University , Chiba-shi , Japan
| | - Noriko M Tsuji
- Immune Homeostasis Laboratory, Biomedical Research Institute, National Institute for Advanced Industrial Science and Technology (AIST) , Tsukuba , Japan
| |
Collapse
|
135
|
Exosomes released by chronic lymphocytic leukemia cells induce the transition of stromal cells into cancer-associated fibroblasts. Blood 2015; 126:1106-17. [PMID: 26100252 DOI: 10.1182/blood-2014-12-618025] [Citation(s) in RCA: 366] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 06/16/2015] [Indexed: 02/08/2023] Open
Abstract
Exosomes derived from solid tumor cells are involved in immune suppression, angiogenesis, and metastasis, but the role of leukemia-derived exosomes has been less investigated. The pathogenesis of chronic lymphocytic leukemia (CLL) is stringently associated with a tumor-supportive microenvironment and a dysfunctional immune system. Here, we explore the role of CLL-derived exosomes in the cellular and molecular mechanisms by which malignant cells create this favorable surrounding. We show that CLL-derived exosomes are actively incorporated by endothelial and mesenchymal stem cells ex vivo and in vivo and that the transfer of exosomal protein and microRNA induces an inflammatory phenotype in the target cells, which resembles the phenotype of cancer-associated fibroblasts (CAFs). As a result, stromal cells show enhanced proliferation, migration, and secretion of inflammatory cytokines, contributing to a tumor-supportive microenvironment. Exosome uptake by endothelial cells increased angiogenesis ex vivo and in vivo, and coinjection of CLL-derived exosomes and CLL cells promoted tumor growth in immunodeficient mice. Finally, we detected α-smooth actin-positive stromal cells in lymph nodes of CLL patients. These findings demonstrate that CLL-derived exosomes actively promote disease progression by modulating several functions of surrounding stromal cells that acquire features of cancer-associated fibroblasts.
Collapse
|
136
|
Li W, Zhou Y, Yang J, Zhang X, Zhang H, Zhang T, Zhao S, Zheng P, Huo J, Wu H. Gastric cancer-derived mesenchymal stem cells prompt gastric cancer progression through secretion of interleukin-8. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:52. [PMID: 25986392 PMCID: PMC4443537 DOI: 10.1186/s13046-015-0172-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/08/2015] [Indexed: 01/26/2023]
Abstract
Background Bone marrow mesenchymal stem cells (BM-MSCs) have been identified to be closely associated with tumor growth and progression. However, the roles of tumor-resident MSCs in cancer have not been thoroughly clarified. This study was to investigate the regulating effect of gastric cancer-derived MSCs (GC-MSCs) on gastric cancer and elucidate the underlying mechanism. Methods GC-MSCs were isolated from primary human gastric cancer tissues and characterized. The effect of GC-MSCs on gastric cancer cell proliferation was analyzed by MTT assay and colony formation assay. Transwell migration assay was performed to evaluate the influence of GC-MSCs in gastric cancer cell migration. The regulating effects of interactions between gastric cancer cells and GC-MSCs on their pro-angiogenic abilities were analyzed in a co-culture system, with the expression, and secretion of pro-angiogenic factors detected by RT-PCR and Luminex assay. Tube formation assay was used to further validate the angiogenic capability of gastric cancer cells or GC-MSCs. Cytokine profiles in the supernatant of GC-MSCs were screened by Luminex assay and neutralizing antibody was used to identify the key effective cytokines. The activations of Akt and Erk1/2 in gastric caner cells were detected by Western blot. Results GC-MSC treatment enhanced the proliferation and migration of BGC-823 and MKN-28 cells, which was more potently than MSCs from adjacent non-cancerous tissues (GCN-MSCs) or bone marrow (BM-MSCs). Higher expression levels of pro-angiogenic factors were detected in GC-MSCs than GCN-MSCs or BM-MSCs. After 10 % GC-MSC-CM treatment, BGC-823, and MKN-28 cells expressed increased levels of pro-angiogenic factors and facilitated tube formation more potently than cancer cells alone. Furthermore, GC-MSCs produced an extremely higher level of interleukin-8 (IL-8) than GCN-MSCs or BM-MSCs. Blockade of IL-8 by neutralizing antibody significantly attenuated the tumor-promoting effect of GC-MSCs. In addition, 10 % CM of IL-8-secreted GC-MSCs induced the activations of Akt or Erk1/2 pathway in BGC-823 and MKN-28 cells. Conclusion Tumor-resident GC-MSCs promote gastric cancer growth and progression more efficiently than GCN-MSCs or BM-MSCs through a considerable secretion of IL-8, which could be a possible target for gastric cancer therapy.
Collapse
Affiliation(s)
- Wei Li
- Center of Research Laboratory, The First People's Hospital of Lianyungang, 182 Tongguan Road, Lianyungang, 222001, China. .,Department of Pathology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, 221004, China.
| | - Ying Zhou
- Center of Research Laboratory, The First People's Hospital of Lianyungang, 182 Tongguan Road, Lianyungang, 222001, China.
| | - Jin Yang
- Center of Research Laboratory, The First People's Hospital of Lianyungang, 182 Tongguan Road, Lianyungang, 222001, China.
| | - Xu Zhang
- School of Medical Science and Laboratory Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, China.
| | - Huanhuan Zhang
- Center of Research Laboratory, The First People's Hospital of Lianyungang, 182 Tongguan Road, Lianyungang, 222001, China.
| | - Ting Zhang
- Center of Research Laboratory, The First People's Hospital of Lianyungang, 182 Tongguan Road, Lianyungang, 222001, China.
| | - Shaolin Zhao
- Center of Research Laboratory, The First People's Hospital of Lianyungang, 182 Tongguan Road, Lianyungang, 222001, China.
| | - Ping Zheng
- Center of Research Laboratory, The First People's Hospital of Lianyungang, 182 Tongguan Road, Lianyungang, 222001, China.
| | - Juan Huo
- Center of Research Laboratory, The First People's Hospital of Lianyungang, 182 Tongguan Road, Lianyungang, 222001, China.
| | - Huiyi Wu
- Center of Research Laboratory, The First People's Hospital of Lianyungang, 182 Tongguan Road, Lianyungang, 222001, China.
| |
Collapse
|
137
|
MicroRNA transcriptomes of distinct human NK cell populations identify miR-362-5p as an essential regulator of NK cell function. Sci Rep 2015; 5:9993. [PMID: 25909817 PMCID: PMC4408982 DOI: 10.1038/srep09993] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 03/25/2015] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells are critical effectors in the immune response against malignancy and infection, and microRNAs (miRNAs) play important roles in NK cell biology. Here we examined miRNA profiles of human NK cells from different cell compartments (peripheral blood, cord blood, and uterine deciduas) and of NKT and T cells from peripheral blood, and we identified a novel miRNA, miR-362-5p, that is highly expressed in human peripheral blood NK (pNK) cells. We also demonstrated that CYLD, a negative regulator of NF-κB signaling, was a target of miR-362-5p in NK cells. Furthermore, we showed that the over-expression of miR-362-5p enhanced the expression of IFN-γ, perforin, granzyme-B, and CD107a in human primary NK cells, and we found that silencing CYLD with a small interfering RNA (siRNA) mirrored the effect of miR-362-5p over-expression. In contrast, the inhibition of miR-362-5p had the opposite effect in NK cells, which was abrogated by CYLD siRNA, suggesting that miR-362-5p promotes NK-cell function, at least in part, by the down-regulation of CYLD. These results provide a resource for studying the roles of miRNAs in human NK cell biology and contribute to a better understanding of the physiologic significance of miRNAs in the regulation of NK cell function.
Collapse
|
138
|
Sinkovics JG. The cell survival pathways of the primordial RNA-DNA complex remain conserved in the extant genomes and may function as proto-oncogenes. Eur J Microbiol Immunol (Bp) 2015; 5:25-43. [PMID: 25883792 PMCID: PMC4397846 DOI: 10.1556/eujmi-d-14-00034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 12/22/2014] [Indexed: 01/09/2023] Open
Abstract
Malignantly transformed (cancer) cells of multicellular hosts, including human cells, operate activated biochemical pathways that recognizably derived from unicellular ancestors. The descendant heat shock proteins of thermophile archaea now chaperon oncoproteins. The ABC cassettes of toxin-producer zooxantella Symbiodinia algae pump out the cytoplasmic toxin molecules; malignantly transformed cells utilize the derivatives of these cassettes to get rid of chemotherapeuticals. High mobility group helix-loop-helix proteins, protein arginine methyltransferases, proliferating cell nuclear antigens, and Ki-67 nuclear proteins, that protect and repair DNA in unicellular life forms, support oncogenes in transformed cells. The cell survival pathways of Wnt-β-catenin, Hedgehog, PI3K, MAPK-ERK, STAT, Ets, JAK, Pak, Myb, achaete scute, circadian rhythms, Bruton kinase and others, which are physiological in uni- and early multicellular eukaryotic life forms, are constitutively encoded in complex oncogenic pathways in selected single cells of advanced multicellular eukaryotic hosts. Oncogenes and oncoproteins in advanced multicellular hosts recreate selected independently living and immortalized unicellular life forms, which are similar to extinct and extant protists. These unicellular life forms are recognized at the clinics as autologous "cancer cells".
Collapse
Affiliation(s)
- J G Sinkovics
- St. Joseph's Hospital Cancer Institute Affiliated with the H. L. Moffitt Comprehensive Cancer Center, Morsani College of Medicine, Department of Molecular Medicine, The University of South Florida Tampa, FL USA
| |
Collapse
|
139
|
Krzewski K, Bryceson YT. Molecular mechanisms regulating cytotoxic lymphocyte development and function, and their associations to human diseases. Front Immunol 2014; 5:279. [PMID: 24966858 PMCID: PMC4052198 DOI: 10.3389/fimmu.2014.00279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 05/28/2014] [Indexed: 12/18/2022] Open
Affiliation(s)
- Konrad Krzewski
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Rockville, MD , USA
| | - Yenan T Bryceson
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge , Stockholm , Sweden
| |
Collapse
|
140
|
Zhao JJ, Pan QZ, Pan K, Weng DS, Wang QJ, Li JJ, Lv L, Wang DD, Zheng HX, Jiang SS, Zhang XF, Xia JC. Interleukin-37 mediates the antitumor activity in hepatocellular carcinoma: role for CD57+ NK cells. Sci Rep 2014; 4:5177. [PMID: 24898887 PMCID: PMC4046124 DOI: 10.1038/srep05177] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 05/02/2014] [Indexed: 12/19/2022] Open
Abstract
The biological role of interleukin-37 (IL-37) in cancer is large unknown. Through immunohistochemical detection using 163 primary hepatocellular carcinoma (HCC) clinical specimens, we found the expression of IL-37 was decreased in tumor tissues, and the expression level was negatively correlated with tumor size. High expression of IL-37 in HCC tumor tissues was associated with better overall survival (OS) and disease-free survival (DFS). IL-37 expression in tumor tissues was positively associated with the density of tumor-infiltrating CD57+ natural killer (NK) cells, but not with the CD3+ and CD8+ T cells. Consistently, in vitro chemotaxis analysis showed that IL-37- overexpressing HCC cells could recruit more NK cells. The in vivo mouse model experiments also revealed that overexpression IL-37 in HCC cells significantly delayed tumor growth and recruited more NK cells into tumors tissues. Our finding suggested that IL-37 might play an important role for the prognosis of HCC patients via regulating innate immune-action.
Collapse
Affiliation(s)
- Jing-Jing Zhao
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2] Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China [3]
| | - Qiu-Zhong Pan
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2]
| | - Ke Pan
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2] Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China [3]
| | - De-Sheng Weng
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2] Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Qi-Jing Wang
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2] Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Jian-Jun Li
- State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Lin Lv
- State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Dan-Dan Wang
- State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Hai-Xia Zheng
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2] Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Shan-Shan Jiang
- State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Xiao-Fei Zhang
- State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Jian-Chuan Xia
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2] Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China
| |
Collapse
|
141
|
Poggi A, Musso A, Dapino I, Zocchi MR. Mechanisms of tumor escape from immune system: role of mesenchymal stromal cells. Immunol Lett 2014; 159:55-72. [PMID: 24657523 DOI: 10.1016/j.imlet.2014.03.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/01/2014] [Accepted: 03/05/2014] [Indexed: 12/12/2022]
Abstract
Tumor microenvironment represents the site where the tumor tries to survive and escape from immune system-mediated recognition. Indeed, to proliferate tumor cells can divert the immune response inducing the generation of myeloid derived suppressor cells and regulatory T cells which can limit the efficiency of effector antitumor lymphocytes in eliminating neoplastic cells. Many components of the tumor microenvironment can serve as a double sword for the tumor and the host. Several types of fibroblast-like cells, which herein we define mesenchymal stromal cells (MSC), secrete extracellular matrix components and surrounding the tumor mass can limit the expansion of the tumor. On the other hand, MSC can interfere with the immune recognition of tumor cells producing immunoregulatory cytokines as transforming growth factor (TGF)ß, releasing soluble ligands of the activating receptors expressed on cytolytic effector cells as decoy molecules, affecting the correct interaction among lymphocytes and tumor cells. MSC can also serve as target for the same anti-tumor effector lymphocytes or simply impede the interaction between these lymphocytes and neoplastic cells. Thus, several evidences point out the role of MSC, both in epithelial solid tumors and hematological malignancies, in regulating tumor cell growth and immune response. Herein, we review these evidences and suggest that MSC can be a suitable target for a more efficient anti-tumor therapy.
Collapse
Affiliation(s)
- Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS AOU San Martino IST, 16132 Genoa, Italy.
| | - Alessandra Musso
- Molecular Oncology and Angiogenesis Unit, IRCCS AOU San Martino IST, 16132 Genoa, Italy
| | - Irene Dapino
- Molecular Oncology and Angiogenesis Unit, IRCCS AOU San Martino IST, 16132 Genoa, Italy
| | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, Istituto Scientifico San Raffaele Milan, 20132 Milan, Italy
| |
Collapse
|