201
|
Nguyen HV, Faivre V. Targeted drug delivery therapies inspired by natural taxes. J Control Release 2020; 322:439-456. [PMID: 32259545 DOI: 10.1016/j.jconrel.2020.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/18/2022]
Abstract
A taxis is the movement responding to a stimulus of an organism. This behavior helps organisms to migrate, to find food or to avoid dangers. By mimicking and using natural taxes, many bio-inspired and bio-hybrid drug delivery systems have been synthesized. Under the guidance of physical and chemical stimuli, drug-loaded carriers are led to a target, for example tumors, then locally release the drug, inducing a therapeutic effect without influencing other parts of the body. On the other hand, for moving targets, for example metastasis cancer cells or bacteria, taking advantage of their taxes behavior is a solution to capture and to eliminate them. For instance, several traps and ecological niches have been fabricated to attract cancer cells by releasing chemokines. Cancer cells are then eliminated by drug loaded inside the trap, by radiotherapy focusing on the trap location or by simply removing the trap. Further research is needed to deeply understand the taxis behavior of organisms, which is essential to ameliorate the performance of taxes-inspired drug delivery application.
Collapse
Affiliation(s)
- Hung V Nguyen
- Université Paris-Saclay, CNRS, Institut Galien Paris Sud, 5 rue JB Clément, 92296 Châtenay-Malabry, France
| | - Vincent Faivre
- Université Paris-Saclay, CNRS, Institut Galien Paris Sud, 5 rue JB Clément, 92296 Châtenay-Malabry, France.
| |
Collapse
|
202
|
Bittner KR, Jiménez JM, Peyton SR. Vascularized Biomaterials to Study Cancer Metastasis. Adv Healthc Mater 2020; 9:e1901459. [PMID: 31977160 PMCID: PMC7899188 DOI: 10.1002/adhm.201901459] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/07/2019] [Indexed: 12/15/2022]
Abstract
Cancer metastasis, the spread of cancer cells to distant organs, is responsible for 90% of cancer-related deaths. Cancer cells need to enter and exit circulation in order to form metastases, and the vasculature and endothelial cells are key regulators of this process. While vascularized 3D in vitro systems have been developed, few have been used to study cancer, and many lack key features of vessels that are necessary to study metastasis. This review focuses on current methods of vascularizing biomaterials for the study of cancer, and three main factors that regulate intravasation and extravasation: endothelial cell heterogeneity, hemodynamics, and the extracellular matrix of the perivascular niche.
Collapse
Affiliation(s)
- Katharine R Bittner
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA
| | - Juan M Jiménez
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Shelly R Peyton
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
203
|
Ma F, Lei YY, Ding MG, Luo LH, Xie YC, Liu XL. LncRNA NEAT1 Interacted With DNMT1 to Regulate Malignant Phenotype of Cancer Cell and Cytotoxic T Cell Infiltration via Epigenetic Inhibition of p53, cGAS, and STING in Lung Cancer. Front Genet 2020; 11:250. [PMID: 32296457 PMCID: PMC7136539 DOI: 10.3389/fgene.2020.00250] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/02/2020] [Indexed: 12/31/2022] Open
Abstract
Purpose Lung cancer is the main cause of cancer-related mortality worldwide. We report here the biological role of nuclear paraspeckle assembly transcript 1 (NEAT1) in the pathogenesis of lung cancer and the underlying mechanisms. Methods Reverse transcription–quantitative polymerase chain reaction (RT-qPCR) and Western blotting analysis were used to evaluate expression of mRNA and protein. RNA immunoprecipitation (RIP) assay, chromatin immunoprecipitation followed by qPCR analysis, and reporter assay were used to detect DNA/RNA and protein binding. Tumor-infiltrating lymphocytes were assessed with hematoxylin-eosin staining. Cytotoxic T cell infiltration was evaluated with flow cytometric analysis and immunohistochemistry (IHC) staining. The changes of cell viability and cell invasive and migratory ability were analyzed by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide), colony formation, and Transwell assays, respectively. Syngeneic tumor model was set up to evaluate antitumor effect. Results The results showed that NEAT1 was overexpressed in lung cancer tissues and cancer cell lines. This aberrant expression was closely related with tumor stage and lymph node metastasis. Tumor sample with high CD8+ showed lower NEAT1 expression. In vitro studies displayed that inhibition of NEAT1 with shRNA resulted in suppression of survival and migration/invasion of lung cancer cells. On the other side, NEAT1 was found to promote tumor growth via inhibiting cytotoxic T cell immunity in syngeneic models. Finally, NEAT1 was found to interact with DNMT1, which in turn inhibited P53 and cyclic GMP-AMP synthase stimulator of interferon genes (cGAS/STING) expression. Conclusion Our findings demonstrated that NEAT1 interacted with DNMT1 to regulate cytotoxic T cell infiltration in lung cancer via inhibition of cGAS/STING pathway. The results provided the novel mechanistic insight into the pathogenesis of lung cancer.
Collapse
Affiliation(s)
- Fang Ma
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi-Yu Lei
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Meng-Ge Ding
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li-Hua Luo
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang-Chun Xie
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xian-Ling Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
204
|
Shu C, Sun P, Xie H, Huang W, Qi J, Ma Y. Virus-Like Particles Presenting the FGF-2 Protein or Identified Antigenic Peptides Promoted Antitumor Immune Responses in Mice. Int J Nanomedicine 2020; 15:1983-1996. [PMID: 32308382 PMCID: PMC7146011 DOI: 10.2147/ijn.s237182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/25/2020] [Indexed: 01/29/2023] Open
Abstract
Background Fibroblast growth factor (FGF)-2 is overexpressed in various tumor tissues. It affects tumor cell proliferation, invasion and survival, promotes tumor angiogenesis and is tightly involved in the development of systemic and local immunosuppressive tumor mechanisms. Purpose This study aimed to develop an effective vaccine against FGF-2 and to investigate the effects of anti-FGF-2 immunization on tumor growth and antitumor immune responses. Methods A set of thirteen synthesized overlapping peptides covering all possible linear B-cell epitopes of murine FGF-2 and a recombinant FGF-2 protein were conjugated to virus-like particles (VLPs) of recombinant hepatitis B core antigen (HBcAg). The VLPs were immunized through a preventive or therapeutic strategy in a TC-1 or 4T1 grafted tumor model. Results Immunization with FGF-2 peptides or full-length protein-coupled VLPs produced FGF-2-specific antibodies with a high titer. Peptide 12, which is located in the heparin-binding site of FGF-2, or protein-conjugated VLPs presented the most significant effects on the suppression of TC-1 tumor growth. The levels of IFN-γ-expressing splenocytes and serum IFN-γ were significantly elevated; further, the immune effector cells CD8+ IFN-γ+ cytotoxic T lymphocytes (CTLs) and CD4+ IFN-γ+ Th1 cells were significantly increased, whereas the immunosuppressive cells CD4+ CD25+ FOXP3+ Treg cells and Gr-1+ CD11b+ myeloid-derived suppressor cells (MDSCs) were decreased in the immunized mice. In addition, VLP immunization significantly suppressed tumor vascularization and promoted tumor cell apoptosis. In mice bearing 4T1 breast tumor, preventive immunization with FGF-2-conjugated VLPs suppressed tumor growth and lung metastasis, and increased effector cell responses. Conclusion Active immunization against FGF-2 is a new possible strategy for tumor immunotherapy.
Collapse
Affiliation(s)
- Congyan Shu
- Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming 650118, People's Republic of China.,Sichuan Institute for Food and Drug Control, Chengdu 611731, People's Republic of China
| | - Pengyan Sun
- Yunnan Center for Disease Control and Prevention, Kunming 650022, People's Republic of China
| | - Hanghang Xie
- Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Weiwei Huang
- Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Jialong Qi
- Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming 650118, People's Republic of China
| | - Yanbing Ma
- Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming 650118, People's Republic of China
| |
Collapse
|
205
|
The function and clinical application of extracellular vesicles in innate immune regulation. Cell Mol Immunol 2020; 17:323-334. [PMID: 32203193 PMCID: PMC7109106 DOI: 10.1038/s41423-020-0391-1] [Citation(s) in RCA: 179] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/17/2020] [Indexed: 12/21/2022] Open
Abstract
The innate immune system plays a crucial role in the host defense against viral and microbial infection. Exosomes constitute a subset of extracellular vesicles (EVs) that can be released by almost all cell types. Owing to their capacity to shield the payload from degradation and to evade recognition and subsequent removal by the immune system, exosomes efficiently transport functional components to recipient cells. Accumulating evidence has recently shown that exosomes derived from tumor cells, host cells and even bacteria and parasites mediate the communication between the invader and innate immune cells and thus play an irreplaceable function in the dissemination of pathogens and donor cell-derived molecules, modulating the innate immune responses of the host. In this review, we describe the current understanding of EVs (mainly focusing on exosomes) and summarize and discuss their crucial roles in determining innate immune responses. Additionally, we discuss the potential of using exosomes as biomarkers and cancer vaccines in diagnostic and therapeutic applications.
Collapse
|
206
|
Bhargava A, Mishra DK, Tiwari R, Lohiya NK, Goryacheva IY, Mishra PK. Immune cell engineering: opportunities in lung cancer therapeutics. Drug Deliv Transl Res 2020; 10:1203-1227. [PMID: 32172351 DOI: 10.1007/s13346-020-00719-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Engineered immune cells offer a prime therapeutic alternate for some aggressive and frequently occurring malignancies like lung cancer. These therapies were reported to result in tumor regression and overall improvement in patient survival. However, studies also suggest that the presence of cancer cell-induced immune-suppressive microenvironment, off-target toxicity, and difficulty in concurrent imaging are some prime impendent in the success of these approaches. The present article reviews the need and significance of the currently available immune cell-based strategies for lung cancer therapeutics. It also showcases the utility of incorporating nanoengineered strategies and details the available formulations of nanocarriers. In last, it briefly discussed the existing methods for nanoparticle fuctionalization and challenges in translating basic research to the clinics. Graphical Abstract.
Collapse
Affiliation(s)
- Arpit Bhargava
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Kamla Nehru Hospital,, Building (Gandhi Medical College Campus), Bhopal, Madhya Pradesh, 462001, India
| | | | - Rajnarayan Tiwari
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Kamla Nehru Hospital,, Building (Gandhi Medical College Campus), Bhopal, Madhya Pradesh, 462001, India
| | | | - Irina Yu Goryacheva
- Department of General and Inorganic Chemistry, Saratov State University, Saratov, Russian Federation
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Kamla Nehru Hospital,, Building (Gandhi Medical College Campus), Bhopal, Madhya Pradesh, 462001, India.
| |
Collapse
|
207
|
Li T, Zhang X, Lv Z, Gao L, Yan H. Increased Expression of Myeloid-Derived Suppressor Cells in Patients with HBV-Related Hepatocellular Carcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6527192. [PMID: 32258134 PMCID: PMC7097855 DOI: 10.1155/2020/6527192] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/08/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023]
Abstract
METHODS The percentages of MDSCs, IFN-γ-producing CD4 and CD8 T cells in the peripheral blood of HCC patients, chronic hepatitis B (CHB) patients, and healthy controls (HC) were determined by flow cytometry. The serum concentrations of IL-10 and TNF-α were determined by ELISA. The association of the percentages of MDSCs with tumor burden, liver function parameters, systemic inflammation-related indexes, and IFN-γ-producing T cells was assessed. RESULTS The percentages of MDSCs and PMN-MDSCs were significantly higher in HCC patients than those in CHB patients and HC. The level of MDSCs was correlated with indirect bilirubin and prealbumin, as well as systemic inflammation response index, monocyte/lymphocyte ratio, and monocyte counts. The frequency of IFN-γ-producing CD8 T cells of HCC patients was lower than that of HC. However, there was no relationship between MDSCs and IFN-γ-producing CD8 T cells. The level of IL-10 in HCC patients was significantly higher than that in CHB patients. CONCLUSION MDSCs seem to play an important role in the process leading from chronic HBV infection to HCC. Early inhibiting these cells could affect tumor progression.
Collapse
Affiliation(s)
- Tianyu Li
- Graduate College of Hebei Medical University, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Xinyu Zhang
- Graduate College of Hebei Medical University, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Zhuo Lv
- Graduate College of Hebei Medical University, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Li Gao
- College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Huimin Yan
- Clinical Research Center, Shijiazhuang Fifth Hospital, Shijiazhuang, Hebei 050021, China
| |
Collapse
|
208
|
Lin X, Xiao Z, Chen T, Liang SH, Guo H. Glucose Metabolism on Tumor Plasticity, Diagnosis, and Treatment. Front Oncol 2020; 10:317. [PMID: 32211335 PMCID: PMC7069415 DOI: 10.3389/fonc.2020.00317] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/21/2020] [Indexed: 12/22/2022] Open
Abstract
Malignant cells support tumor proliferation and progression by adopting to metabolic changes. Tumor cells altered metabolism by increasing glucose uptake and fermentation of glucose to lactate, even in the aerobic state and the presence of functioning mitochondria. Glucose metabolism in tumor plasticity has attracted great interests by clinicians and scientists in the past decades. This review discusses the previous and emerging researches on the tumor plasticity altered by changing glucose metabolism in different cancer cells, including cancer stem cells (CSCs). In addition, we summarize the rising applications of glucose metabolism in tumor diagnosis and treatment. Our objective is to direct future investigation on this altered metabolic phenotype and its application in patient care.
Collapse
Affiliation(s)
- Xiaoping Lin
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Zizheng Xiao
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tao Chen
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Huiqin Guo
- Department of Thoracic Surgery, Beijing Sijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
209
|
Ou X, Lv W. Metabolic changes and interaction of tumor cell, myeloid-derived suppressor cell and T cell in hypoxic microenvironment. Future Oncol 2020; 16:383-393. [PMID: 32067476 DOI: 10.2217/fon-2019-0692] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It is universally acknowledged that a large number of immune cells, as well as inflammatory factors, regulatory factors and metabolites, accumulate in the tumor microenvironment to jointly promote tumor escape, development and metastasis. Hypoxia is one of the characteristics in tumor microenvironment and is a common phenomenon in all solid tumors. In tumor hypoxia response, there is a key regulator called HIF-1a, which is a key transcriptional regulatory protein that regulates many critical genes. In this paper, the effects of hypoxia on glucose metabolism of tumor cells, myeloid-derived suppressor cells and T cells in tumor microenvironment were reviewed, and the interaction among the three was also described.
Collapse
Affiliation(s)
- Xiantu Ou
- Clinical laboratory of Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, No. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong Province 528308, PR China
| | - Weibiao Lv
- Clinical laboratory of Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, No. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong Province 528308, PR China
| |
Collapse
|
210
|
Vail DM, LeBlanc AK, Jeraj R. Advanced Cancer Imaging Applied in the Comparative Setting. Front Oncol 2020; 10:84. [PMID: 32117739 PMCID: PMC7019008 DOI: 10.3389/fonc.2020.00084] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/16/2020] [Indexed: 11/13/2022] Open
Abstract
The potential for companion (pet) species with spontaneously arising tumors to act as surrogates for preclinical development of advanced cancer imaging technologies has become more apparent in the last decade. The utility of the companion model specifically centers around issues related to body size (including spatial target/normal anatomic characteristics), physical size and spatial distribution of metastasis, tumor heterogeneity, the presence of an intact syngeneic immune system and a syngeneic tumor microenvironment shaped by the natural evolution of the cancer. Companion species size allows the use of similar equipment, hardware setup, software, and scan protocols which provide the opportunity for standardization and harmonization of imaging operating procedures and quality assurance across imaging protocols, imaging hardware, and the imaged species. Murine models generally do not replicate the size and spatial distribution of human metastatic cancer and these factors strongly influence image resolution and dosimetry. The following review will discuss several aspects of comparative cancer imaging in more detail while providing several illustrative examples of investigational approaches performed or currently under exploration at our institutions. Topics addressed include a discussion on interested consortia; image quality assurance and harmonization; image-based biomarker development and validation; contrast agent and radionuclide tracer development; advanced imaging to assess and predict response to cytotoxic and immunomodulatory anticancer agents; imaging of the tumor microenvironment; development of novel theranostic approaches; cell trafficking assessment via non-invasive imaging; and intraoperative imaging to inform surgical oncology decision making. Taken in totality, these comparative opportunities predict that safety, diagnostic and efficacy data generated in companion species with naturally developing and progressing cancers would better recapitulate the human cancer condition than that of artificial models in small rodent systems and ultimately accelerate the integration of novel imaging technologies into clinical practice. It is our hope that the examples presented should serve to provide those involved in cancer investigations who are unfamiliar with available comparative methodologies an understanding of the potential utility of this approach.
Collapse
Affiliation(s)
- David M Vail
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Robert Jeraj
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States.,Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
211
|
Charan M, Dravid P, Cam M, Setty B, Roberts RD, Houghton PJ, Cam H. Tumor secreted ANGPTL2 facilitates recruitment of neutrophils to the lung to promote lung pre-metastatic niche formation and targeting ANGPTL2 signaling affects metastatic disease. Oncotarget 2020; 11:510-522. [PMID: 32082485 PMCID: PMC7007290 DOI: 10.18632/oncotarget.27433] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/28/2019] [Indexed: 12/28/2022] Open
Abstract
The pre-metastatic niche (PMN) represents an abnormal microenvironment devoid of cancer cells, but favoring tumor growth. Little is known about the mechanisms that generate the PMN or their effects on host cells within metastasis-prone organs. Here, we investigated by using spontaneous metastatic models whether lung epithelial cells are essential for primary tumor induced neutrophil recruitment in lung and subsequently initiating PMN formation in osteosarcoma. We found that serum levels of ANGPTL2 in osteosarcoma patients are significantly higher compared to those in healthy controls and that ANGPTL2 secretion by tumor cells plays an essential role in osteosarcoma metastasis. We determined that tumor-derived ANGPTL2 stimulates lung epithelial cells, which is essential for primary tumor-induced neutrophil recruitment in lung and subsequent pre-metastatic niche formation. Lastly, we identified that a p63 isoform, ΔNp63, drives high level of ANGPTL2 secretion and pharmaceutical inhibition of ANGPTL2 signaling by a non–RGD-based integrin binding peptide (ATN-161) diminished metastatic load in lungs likely due to reduction of the lung pre-metastatic niche formation.
Collapse
Affiliation(s)
- Manish Charan
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Piyush Dravid
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Maren Cam
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Bhuvana Setty
- Department of Hematology & Oncology, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Ryan D Roberts
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Peter J Houghton
- Greehey Children's Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Hakan Cam
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
212
|
Wang J, Sun M, Zhao H, Huang Y, Li D, Mao D, Zhang Z, Zhu X, Dong X, Zhao X. IL-9 Exerts Antitumor Effects in Colon Cancer and Transforms the Tumor Microenvironment In Vivo. Technol Cancer Res Treat 2019; 18:1533033819857737. [PMID: 31242804 PMCID: PMC6598323 DOI: 10.1177/1533033819857737] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
As a newly discovered cytokine, interleukin 9 was initially considered a T-lymphocyte growth factor. Interleukin 9 affects target cells by binding to a member of the γc-family of receptors and is involved in inflammation, autoimmune diseases, and other ailments. In recent years, mounting evidence reveals that interleukin 9 exerts antitumor effects, which has attracted considerable attention. Many previous studies were performed in vivo by establishing a mouse model of melanoma. Here, interleukin 9 protein and messenger RNA expression levels were both low in colon carcinoma tissue specimens, as assessed by immunohistochemistry and quantitative real-time polymerase chain reaction. In addition, interleukin 9 expression in these samples was correlated with TNM staging, Dukes staging, lymph node metastasis, and good prognosis, but not with gender, age, tumor size, tumor differentiation, and hepatic metastasis. In vivo, by establishing a mouse subcutaneous allograft model, we found that interleukin 9 overexpression inhibited tumor growth and resulted in longer survival time. Then, antitumor immune responses were increased by interleukin 9 as demonstrated by flow cytometry. Furthermore, interleukin 9 was shown to exert antitumor effects by regulating T-cell function and killing tumor cells in the tumor microenvironment. Overall, this study revealed that interleukin 9 exerts robust antitumor effects in colon cancer and transforms the tumor microenvironment in vivo.
Collapse
Affiliation(s)
- Jin Wang
- 1 Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.,2 Jiangsu Institute of Clinical Immunology, Soochow University, Suzhou, China.,3 Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.,4 Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mingbing Sun
- 1 Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hua Zhao
- 1 Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Huang
- 1 Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dongbao Li
- 1 Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Deli Mao
- 1 Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhe Zhang
- 1 Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinguo Zhu
- 1 Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoqiang Dong
- 1 Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin Zhao
- 1 Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.,2 Jiangsu Institute of Clinical Immunology, Soochow University, Suzhou, China.,3 Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.,4 Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
213
|
Benites BD, Alvarez MC, Saad STO. Small Particles, Big Effects: The Interplay Between Exosomes and Dendritic Cells in Antitumor Immunity and Immunotherapy. Cells 2019; 8:E1648. [PMID: 31888159 PMCID: PMC6952774 DOI: 10.3390/cells8121648] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/07/2019] [Accepted: 12/10/2019] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells play a fundamental role in the antitumor immunity cycle, and the loss of their antigen-presenting function is a recognized mechanism of tumor evasion. We have recently demonstrated the effect of exosomes extracted from serum of patients with acute myeloid leukemia as important inducers of dendritic cell immunotolerance, and several other works have recently demonstrated the effects of these nanoparticles on immunity to other tumor types as well. The aim of this review was to highlight the recent findings on the effects of tumor exosomes on dendritic cell functions, the mechanisms by which they can lead to tumor evasion, and their manipulation as a possible strategy in cancer treatment.
Collapse
Affiliation(s)
- Bruno Deltreggia Benites
- Hematology and Transfusion Medicine Center, University of Campinas, Campinas 13083-970, Brazil; (M.C.A.); (S.T.O.S.)
| | | | | |
Collapse
|
214
|
A mathematical model for the immune-mediated theory of metastasis. J Theor Biol 2019; 482:109999. [PMID: 31493486 DOI: 10.1016/j.jtbi.2019.109999] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 08/13/2019] [Accepted: 09/03/2019] [Indexed: 12/16/2022]
Abstract
Accumulating experimental and clinical evidence suggest that the immune response to cancer is not exclusively anti-tumor. Indeed, the pro-tumor roles of the immune system - as suppliers of growth and pro-angiogenic factors or defenses against cytotoxic immune attacks, for example - have been long appreciated, but relatively few theoretical works have considered their effects. Inspired by the recently proposed "immune-mediated" theory of metastasis, we develop a mathematical model for tumor-immune interactions at two anatomically distant sites, which includes both anti- and pro-tumor immune effects, and the experimentally observed tumor-induced phenotypic plasticity of immune cells (tumor "education" of the immune cells). Upon confrontation of our model to experimental data, we use it to evaluate the implications of the immune-mediated theory of metastasis. We find that tumor education of immune cells may explain the relatively poor performance of immunotherapies, and that many metastatic phenomena, including metastatic blow-up, dormancy, and metastasis to sites of injury, can be explained by the immune-mediated theory of metastasis. Our results suggest that further work is warranted to fully elucidate the pro-tumor effects of the immune system in metastatic cancer.
Collapse
|
215
|
Schramme F, Crosignani S, Frederix K, Hoffmann D, Pilotte L, Stroobant V, Preillon J, Driessens G, Van den Eynde BJ. Inhibition of Tryptophan-Dioxygenase Activity Increases the Antitumor Efficacy of Immune Checkpoint Inhibitors. Cancer Immunol Res 2019; 8:32-45. [PMID: 31806638 DOI: 10.1158/2326-6066.cir-19-0041] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 06/27/2019] [Accepted: 11/15/2019] [Indexed: 11/16/2022]
Abstract
Tryptophan 2,3-dioxygenase (TDO) is an enzyme that degrades tryptophan into kynurenine and thereby induces immunosuppression. Like indoleamine 2,3-dioxygenase (IDO1), TDO is considered as a relevant drug target to improve the efficacy of cancer immunotherapy. However, its role in various immunotherapy settings has not been fully characterized. Here, we described a new small-molecule inhibitor of TDO that can modulate kynurenine and tryptophan in plasma, liver, and tumor tissue upon oral administration. We showed that this compound improved the ability of anti-CTLA4 to induce rejection of CT26 tumors expressing TDO. To better characterize TDO as a therapeutic target, we used TDO-KO mice and found that anti-CTLA4 or anti-PD1 induced rejection of MC38 tumors in TDO-KO, but not in wild-type mice. As MC38 tumors did not express TDO, we related this result to the high systemic tryptophan levels in TDO-KO mice, which lack the hepatic TDO needed to contain blood tryptophan. The antitumor effectiveness of anti-PD1 was abolished in TDO-KO mice fed on a tryptophan-low diet that normalized their blood tryptophan level. MC38 tumors expressed IDO1, which could have limited the efficacy of anti-PD1 in wild-type mice and could have been overcome in TDO-KO mice due to the high levels of tryptophan. Accordingly, treatment of mice with an IDO1 inhibitor improved the efficacy of anti-PD1 in wild-type, but not in TDO-KO, mice. These results support the clinical development of TDO inhibitors to increase the efficacy of immunotherapy of TDO-expressing tumors and suggest their effectiveness even in the absence of tumoral TDO expression.See article by Hoffmann et al., p. 19.
Collapse
Affiliation(s)
- Florence Schramme
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, UCLouvain, Brussels, Belgium
| | | | | | - Delia Hoffmann
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, UCLouvain, Brussels, Belgium
| | - Luc Pilotte
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, UCLouvain, Brussels, Belgium
| | - Vincent Stroobant
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, UCLouvain, Brussels, Belgium
| | | | | | - Benoit J Van den Eynde
- Ludwig Institute for Cancer Research, Brussels, Belgium. .,de Duve Institute, UCLouvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium
| |
Collapse
|
216
|
Xie F, Zhou X, Fang M, Li H, Su P, Tu Y, Zhang L, Zhou F. Extracellular Vesicles in Cancer Immune Microenvironment and Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901779. [PMID: 31871860 PMCID: PMC6918121 DOI: 10.1002/advs.201901779] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/26/2019] [Indexed: 05/10/2023]
Abstract
Extracellular vesicles (EVs) are secreted by almost all cells. They contain proteins, lipids, and nucleic acids which are delivered from the parent cells to the recipient cells. Thereby, they function as mediators of intercellular communication and molecular transfer. Recent evidences suggest that exosomes, a small subset of EVs, are involved in numerous physiological and pathological processes and play essential roles in remodeling the tumor immune microenvironment even before the occurrence and metastasis of cancer. Exosomes derived from tumor cells and host cells mediate their mutual regulation locally or remotely, thereby determining the responsiveness of cancer therapies. As such, tumor-derived circulating exosomes are considered as noninvasive biomarkers for early detection and diagnosis of tumor. Exosome-based therapies are also emerging as cutting-edge and promising strategies that could be applied to suppress tumor progression or enhance anti-tumor immunity. Herein, the current understanding of exosomes and their key roles in modulating immune responses, as well as their potential therapeutic applications are outlined. The limitations of current studies are also presented and directions for future research are described.
Collapse
Affiliation(s)
- Feng Xie
- Institute of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
| | - Xiaoxue Zhou
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
- Key Laboratory of Head & Neck CancerTranslational Research of Zhejiang ProvinceZhejiang Cancer HospitalHangzhou310058P. R. China
| | - Meiyu Fang
- Key Laboratory of Head & Neck CancerTranslational Research of Zhejiang ProvinceZhejiang Cancer HospitalHangzhou310058P. R. China
| | - Heyu Li
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
| | - Peng Su
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
| | - Yifei Tu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
| | - Fangfang Zhou
- Institute of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| |
Collapse
|
217
|
From Tumor Metastasis towards Cerebral Ischemia-Extracellular Vesicles as a General Concept of Intercellular Communication Processes. Int J Mol Sci 2019; 20:ijms20235995. [PMID: 31795140 PMCID: PMC6928831 DOI: 10.3390/ijms20235995] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) have been tremendous carriers in both experimental and translational science. These vesicles—formerly regarded as artifacts of in vitro research—have a heterogeneous population of vesicles derived from virtually all eukaryotic cells. EVs consist of a bilayer lipid structure with a diameter of about 30 to 1000 nm and have a characteristic protein and non-coding RNA content that make up different forms of EVs such as exosomes, microvesicles, and others. Despite recent progress in the EV field, which is known to serve as potential biomarkers and therapeutic tools under various pathological conditions, fundamental questions are yet to be answered. This short review focuses on recently reported data regarding EVs under pathological conditions with a particular emphasis on the role of EVs under such different conditions like tumor formation and cerebral ischemia. The review strives to point out general concepts of EV intercellular communication processes that might be vital to both diagnostic and therapeutic strategies in the long run.
Collapse
|
218
|
Liao P, Wang H, Tang YL, Tang YJ, Liang XH. The Common Costimulatory and Coinhibitory Signaling Molecules in Head and Neck Squamous Cell Carcinoma. Front Immunol 2019; 10:2457. [PMID: 31708918 PMCID: PMC6819372 DOI: 10.3389/fimmu.2019.02457] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/01/2019] [Indexed: 02/05/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are closely linked with immunosuppression, accompanied by complex immune cell functional activities. The abnormal competition between costimulatory and coinhibitory signal molecules plays an important role in the malignant progression of HNSCC. This review will summarize the features of costimulatory molecules (including CD137, OX40 as well as CD40) and coinhibitory molecules (including CTLA-4, PD-1, LAG3, and TIM3), analyze the underlying mechanism behind these molecules' regulation of the progression of HNSCC, and introduce the clinic application. Vaccines, such as those targeting STING while working synergistically with monoclonal antibodies, are also discussed. A deep understanding of the tumor immune landscape will help find new and improved tumor immunotherapy for HNSCC.
Collapse
Affiliation(s)
- Peng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Haofan Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
219
|
de la Cruz-López KG, Castro-Muñoz LJ, Reyes-Hernández DO, García-Carrancá A, Manzo-Merino J. Lactate in the Regulation of Tumor Microenvironment and Therapeutic Approaches. Front Oncol 2019; 9:1143. [PMID: 31737570 PMCID: PMC6839026 DOI: 10.3389/fonc.2019.01143] [Citation(s) in RCA: 525] [Impact Index Per Article: 105.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 10/15/2019] [Indexed: 12/23/2022] Open
Abstract
Tumor cells must generate sufficient ATP and biosynthetic precursors in order to maintain cell proliferation requirements. Otto Warburg showed that tumor cells uptake high amounts of glucose producing large volumes of lactate even in the presence of oxygen, this process is known as “Warburg effect or aerobic glycolysis.” As a consequence of such amounts of lactate there is an acidification of the extracellular pH in tumor microenvironment, ranging between 6.0 and 6.5. This acidosis favors processes such as metastasis, angiogenesis and more importantly, immunosuppression, which has been associated to a worse clinical prognosis. Thus, lactate should be thought as an important oncometabolite in the metabolic reprogramming of cancer. In this review, we summarized the role of lactate in regulating metabolic microenvironment of cancer and discuss its relevance in the up-regulation of the enzymes lactate dehydrogenase (LDH) and monocarboxilate transporters (MCTs) in tumors. The goal of this review is to expose that lactate is not only a secondary product of cellular metabolic waste of tumor cells, but also a key molecule involved in carcinogenesis as well as in tumor immune evasion. Finally, the possible targeting of lactate production in cancer treatment is discussed.
Collapse
Affiliation(s)
- Karen G de la Cruz-López
- Programa de Doctorado en Ciencias Biomédicas, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico.,Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, México/Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Laboratory of Virus and Cancer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Leonardo Josué Castro-Muñoz
- Programa de Doctorado en Ciencias Biomédicas, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico.,Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, México/Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Diego O Reyes-Hernández
- Programa de Maestría y Doctorado en Ciencias Médicas, Odontológicas y de la Salud, Maestría en Investigación Clínica Experimental, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico.,Biological Cancer Causing Agents Group, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Alejandro García-Carrancá
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, México/Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Laboratory of Virus and Cancer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Joaquín Manzo-Merino
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, México/Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Biological Cancer Causing Agents Group, Instituto Nacional de Cancerología, Mexico City, Mexico.,Cátedras CONACyT-Instituto Nacional de Cancerología, Mexico City, Mexico
| |
Collapse
|
220
|
Zheng Y, Fang YC, Li J. PD-L1 expression levels on tumor cells affect their immunosuppressive activity. Oncol Lett 2019; 18:5399-5407. [PMID: 31612048 PMCID: PMC6781757 DOI: 10.3892/ol.2019.10903] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 08/06/2019] [Indexed: 12/14/2022] Open
Abstract
Programmed cell death 1 (PD-1) is an immuno-checkpoint receptor which is primarily expressed on T cells, monocytes, natural killer cells and macrophages. Programmed death-ligand 1 (PD-L1) is the primary ligand of PD-1 and is constitutively expressed on antigen presenting cells, mesenchymal stem cells and bone marrow-derived mast cells. In addition, PD-L1 is also expressed on a wide range of tumor cells, including lung cancer, breast cancer and melanoma. PD-1 and PD-L1 are important members of the immunoglobulin super-family and participate in immune regulation. In the present study, the immune-suppressive effects of a number of tumor cell lines were determined. The breast tumor cell lines MCF-7 and MDA-MB-231 displayed the largest inhibitory effects on T-cell activation and cytokine secretion in a co-culture system. The HepG2, A549 and A375 cells displayed limited inhibitory effects. MCF-7 and MDA-MB-231 cells expressed the highest level of PD-L1 among the cells used, which may explain their higher immuno-suppressive effects. Compound A0-L, a small molecule inhibitor of the PD-1/PD-L1 interaction, restored T cell functions. Additionally, it was demonstrated that the tumor cells with higher levels of PD-L1 expression suppressed signaling pathways involved in T-cell activation, such as the T-cell receptor- zeta chain of T cell receptor associated protein kinase ZAP70-RAS-GTPase-extracellular-signal-regulated kinases and CD28-PI3K-Akt serine/threonine kinases pathways. These findings suggest that tumor cells with higher expression levels of PD-L1 may exhibit higher immuno-suppressive activity, and that drugs targeting the PD-1/PD-L1 interaction may have improved therapeutic effects on tumors expressing higher levels of PD-L1.
Collapse
Affiliation(s)
- Yang Zheng
- Chinese Academy of Sciences Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, P.R. China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - You-Chen Fang
- Chinese Academy of Sciences Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China.,School of Life Sciences, Shanghai University, Shanghai 200444, P.R. China
| | - Jing Li
- Chinese Academy of Sciences Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| |
Collapse
|
221
|
He Q, Liu Z, Liu Z, Lai Y, Zhou X, Weng J. TCR-like antibodies in cancer immunotherapy. J Hematol Oncol 2019; 12:99. [PMID: 31521180 PMCID: PMC6744646 DOI: 10.1186/s13045-019-0788-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy has been regarded as the most significant scientific breakthrough of 2013, and antibody therapy is at the core of this breakthrough. Despite significant success achieved in recent years, it is still difficult to target intracellular antigens of tumor cells with traditional antibodies, and novel therapeutic strategies are needed. T cell receptor (TCR)-like antibodies comprise a novel family of antibodies that can recognize peptide/MHC complexes on tumor cell surfaces. TCR-like antibodies can execute specific and significant anti-tumor immunity through several distinct molecular mechanisms, and the success of this type of antibody therapy in melanoma, leukemia, and breast, colon, and prostate tumor models has excited researchers in the immunotherapy field. Here, we summarize the generation strategy, function, and molecular mechanisms of TCR-like antibodies described in publications, focusing on the most significant discoveries.
Collapse
Affiliation(s)
- Qinghua He
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Zhaoyu Liu
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Zhihua Liu
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Yuxiong Lai
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Xinke Zhou
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Jinsheng Weng
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1414 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
222
|
Ma S, Li X, Wang X, Cheng L, Li Z, Zhang C, Ye Z, Qian Q. Current Progress in CAR-T Cell Therapy for Solid Tumors. Int J Biol Sci 2019; 15:2548-2560. [PMID: 31754328 PMCID: PMC6854376 DOI: 10.7150/ijbs.34213] [Citation(s) in RCA: 273] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 07/16/2019] [Indexed: 12/22/2022] Open
Abstract
Cancer immunotherapy by chimeric antigen receptor-modified T (CAR-T) cells has shown exhilarative clinical efficacy for hematological malignancies. Recently two CAR-T cell based therapeutics, Kymriah (Tisagenlecleucel) and Yescarta (Axicabtagene ciloleucel) approved by US FDA (US Food and Drug Administration) are now used for treatment of B cell acute lymphoblastic leukemia (B-ALL) and diffuse large B-cell lymphoma (DLBCL) respectively in the US. Despite the progresses made in treating hematological malignancies, challenges still remain for use of CAR-T cell therapy to treat solid tumors. In this landscape, most studies have primarily focused on improving CAR-T cells and overcoming the unfavorable effects of tumor microenvironment on solid tumors. To further understand the current status and trend for developing CAR-T cell based therapies for various solid tumors, this review emphasizes on CAR-T techniques, current obstacles, and strategies for application, as well as necessary companion diagnostics for treatment of solid tumors with CAR-T cells.
Collapse
Affiliation(s)
- Shuo Ma
- Shanghai Baize Medical Laboratory, Shanghai, China
| | - Xinchun Li
- Shanghai Baize Medical Laboratory, Shanghai, China
| | - Xinyue Wang
- Shanghai Baize Medical Laboratory, Shanghai, China
| | - Liang Cheng
- Shanghai Baize Medical Laboratory, Shanghai, China.,Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Zhong Li
- Shanghai Baize Medical Laboratory, Shanghai, China
| | | | - Zhenlong Ye
- Shanghai Baize Medical Laboratory, Shanghai, China.,Shanghai Cell Therapy Research Institute, Shanghai, China.,Shanghai Engineering Research Center for Cell Therapy, Shanghai, China
| | - Qijun Qian
- Shanghai Baize Medical Laboratory, Shanghai, China.,Shanghai Cell Therapy Research Institute, Shanghai, China.,Shanghai Engineering Research Center for Cell Therapy, Shanghai, China
| |
Collapse
|
223
|
Zhang X, Zhang Y, Chen J, Wu Y, Zhang J, Wang J. Nanosecond pulsed electric field inhibits malignant melanoma growth by inducing the change of systemic immunity. Med Oral Patol Oral Cir Bucal 2019; 24:e555-e561. [PMID: 31256187 PMCID: PMC6667006 DOI: 10.4317/medoral.22976] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/14/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Nanosecond pulsed electric fields (nsPEFs) showed an inhibitory effect on proliferation of malignant melanoma. In this study, the growth of melanoma were inhibited by changing the systemic immunity . MATERIAL AND METHODS C57BL/6 mice with B16 malignant were exposed to 200 pulses of 100 ns duration, 30kV/cm. The mice were executed four days later. T lymphocyte has been extracted from spleen. Cell viability was evaluated by CCK-8 assay. CD3+CD4+ T cells, CD3+CD8+ T cells, regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSC) were analyzed by flow cytometry . TNF-α, IL-2, IL-10, TGF-β, IFN- γ levels in supernatants were assessed by ELISA. RESULTS C57 malignant melanoma model were established successfully. After the treatment of nsPEFs(30 kV/cm 100 ns 200p), the numbers of T lymphocytes were increased.CD3+ CD4+ T cells changed from 48% to 51.2%;CD3+CD8+T lymphocytes increased from 39.6% to 40.4%.Treg cells reduced from 4.3% to 2.4%,MDSC decreased by 39.0% to 19.7% . In addition, the level of TNF-α, IL-2 were increased (P < 0.05) and the level of IL-10 were decreased (P < 0.05) and the level of TGF-β and IFN-γ remained stable (P > 0.05). CONCLUSIONS Tumor growth can be effectively inhibited by nsPEFs in vivo, which activate targets of immune respones, accumulation of inflammatory cells and immune cytokines.
Collapse
Affiliation(s)
- X Zhang
- Lanzhou University, 199 Donggang Western Road Lanzhou Gansu 730000, China
| | | | | | | | | | | |
Collapse
|
224
|
Kotsakis A, Kallergi G, Aggouraki D, Lyristi Z, Koinis F, Lagoudaki E, Koutsopoulos A, Georgoulias V, Vetsika EK. CD8 + PD-1 + T-cells and PD-L1 + circulating tumor cells in chemotherapy-naïve non-small cell lung cancer: towards their clinical relevance? Ther Adv Med Oncol 2019; 11:1758835919853193. [PMID: 31217824 PMCID: PMC6563396 DOI: 10.1177/1758835919853193] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 02/12/2019] [Indexed: 12/17/2022] Open
Abstract
Background Since tumor cells may escape from immune surveillance through the programmed cell death 1 (PD-1)/programmed death ligand (PD-L)1 axis, this study was designed in order to evaluate whether there is a correlation between the levels of PD-1+ and PD-L1+-expressing immune cells (ICs) and circulating tumor cells (CTCs) in patients with non-small cell lung cancer (NSCLC). Patients and methods Peripheral blood was obtained from 37 chemotherapy-naïve patients with metastatic NSCLC before treatment. PD-1 and PD-L1 expression was evaluated (1) on ICs with anti-tumor function (CD4+ and CD8+ T-cells, B-cells, monocytes/dendritic cells) using flow cytometry, (2) on CTCs by immunofluorescence and (3) on cells from tumor tissues by immunohistochemistry. The levels of PD-1+ and PD-L1+-expressing ICs were correlated with progression-free survival (PFS). Results The presence of PD-1+ CD8+ cells, with reduced interferon (IFN)-γ expression, but not other ICs, were positively correlated with PD-L1+ CTCs (p < 0.04). Increased percentages of PD-1+ CD8+ T-cells, were associated with a worse response to treatment (p = 0.032) and shorter PFS (p = 0.023) which, in multivariate analysis, was revealed as an independent predictor for decreased PFS [hazard ratio (HR): 4.1, p = 0.0007]. Conclusion The results of the current study, for first time, provide evidence for a possible interaction between ICs and CTCs in NSCLC patients via the PD-1/PD-L1 axis and strongly support that the levels of PD-1+ CD8+ in these patients may be of clinical relevance.
Collapse
Affiliation(s)
- Athanasios Kotsakis
- School of Medical Sciences, University of Thessaly, GR-41334, Larissa, Thessaly, Greece
| | - Galatea Kallergi
- Laboratory Tumor Cell Biology, University of Crete, Medical School, Heraklion, Crete, Greece
| | | | - Zaharoula Lyristi
- Laboratory Tumor Cell Biology, University of Crete, Medical School, Heraklion, Crete, Greece
| | - Filippos Koinis
- Department of Medical Oncology, University General Hospital of Larissa, Thessaly, Greece
| | | | | | - Vassilis Georgoulias
- Laboratory Tumor Cell Biology, University of Crete, Medical School, Heraklion, Crete, Greece
| | | |
Collapse
|
225
|
Wang J, Sun M, Zhu X, Zhao H, Mao D, Zhang Z, Zhao X. Lentivirus-mediated RNA interference targeting programmed death receptor ligand 1 increases the immunologic anti-tumor effect of dendritic cell vaccination against pancreatic cancer in SCID-hu mice. Oncol Lett 2019; 18:1539-1547. [PMID: 31423221 PMCID: PMC6607057 DOI: 10.3892/ol.2019.10426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/30/2019] [Indexed: 12/31/2022] Open
Abstract
Programmed death receptor ligand 1 (PD-L1), which belongs to the B7 family, is overexpressed in a variety of human cancer types and serves a crucial role in immune escape by malignant cells. Programmed death receptor 1 (PD-1) is a specific PD-L1 receptor. PD-1/PD-L1 signaling inhibits the antitumor effects of dendritic cell (DC) immunization for tumor treatment. The aim of the present study was to determine whether inhibiting PD-L1 may increase the immunologic anti-tumor effect of dendritic cells against pancreatic cancer. In the present study, PD-L1 levels in non-cancerous and malignant tissue samples were compared, and the impact of PD-L1 downregulation on human pancreatic cancer PaTu8988 cells was determined by lentivirus-based RNA interference and DC immunotherapy. PD-L1 expression in pancreatic specimens was assessed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry. PaTu8988 cells expressing reduced levels of PD-L1 were generated by lentivirus-based knockdown to assess the mechanism by which the inhibition of PD-L1 signaling in DC immunization affects therapeutic outcomes in pancreatic cancer-bearing SCID-hu mice. PD-L1 levels were markedly elevated in pancreatic adenocarcinoma samples compared with in non-cancerous tissue. PD-L1 silencing in pancreatic adenocarcinoma cells resulted in improved treatment outcomes of DC immunization in vitro and in vivo compared with traditional DC immunization. PD-L1 silencing enhances the antitumor response of cytotoxic T cells by increasing interferon γ production in vitro. In vivo, this method prevented tumor growth and lung metastasis, and prolonged survival in the SCID-hu model. In conclusion, the results of the present study suggested that suppressing PD-L1 in malignant cells during DC immunization may be a useful tool for immunotherapy in pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Jin Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Jiangsu Institute of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Mingbing Sun
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Xinguo Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Hua Zhao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Deli Mao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Zhe Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Xin Zhao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Jiangsu Institute of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, P.R. China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
226
|
Lu M, Wang Y, Zhan X. The MAPK Pathway-Based Drug Therapeutic Targets in Pituitary Adenomas. Front Endocrinol (Lausanne) 2019; 10:330. [PMID: 31231308 PMCID: PMC6558377 DOI: 10.3389/fendo.2019.00330] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/07/2019] [Indexed: 12/13/2022] Open
Abstract
Mitogen-activated protein kinases (MAPKs) include ERK, p38, and JNK MAPK subfamilies, which are crucial regulators of cellular physiology, cell pathology, and many diseases including cancers. For the MAPK signaling system in pituitary adenomas (PAs), the activation of ERK signaling is generally thought to promote cell proliferation and growth; whereas the activations of p38 and JNK signaling are generally thought to promote cell apoptosis. The role of MAPK in treatment of PAs is demonstrated through the effects of currently used medications such as somatostatin analogs such as SOM230 and OCT, dopamine agonists such as cabergoline and bromocriptine, and retinoic acid which inhibit the MAPK pathway. Further, there are potential novel therapies based on putative molecular targets of the MAPK pathway, including 18beta-glycyrrhetinic acid (GA), dopamine-somatostatin chimeric compound (BIM-23A760), ursolic acid (UA), fulvestrant, Raf kinase inhibitory protein (RKIP), epidermal growth factor pathway substrate number 8 (Eps8), transmembrane protein with EGF-like and two follistatin-like domains (TMEFF2), cold inducible RNA-binding protein (CIRP), miR-16, and mammaliansterile-20-like kinase (MST4). The combined use of ERK inhibitor (e.g., SOM230, OCT, or dopamine) plus p38 activator (e.g., cabergoline, bromocriptine, and fulvestrant) and/or JNK activator (e.g., UA), or the development of single drug (e.g., BIM-23A760) to target both ERK and p38 or JNK pathways, might produce better anti-tumor effects on PAs. This article reviews the advances in understanding the role of MAPK signaling in pituitary tumorigenesis, and the MAPK pathway-based potential therapeutic drugs for PAs.
Collapse
Affiliation(s)
- Miaolong Lu
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Ya Wang
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Xianquan Zhan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
227
|
Natural killer cells involved in tumour immune escape of hepatocellular carcinomar. Int Immunopharmacol 2019; 73:10-16. [PMID: 31078921 DOI: 10.1016/j.intimp.2019.04.057] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/21/2019] [Accepted: 04/25/2019] [Indexed: 02/08/2023]
Abstract
Natural killer cells are the first line of host immune surveillance and play major roles in the defence against infection and tumours. Hepatic NK cells exhibit unique phenotypic and functional characteristics compared to circulating and spleen NK cells, such as higher levels of cytolytic activity and cytotoxicity mediators against tumour cells. However, the activities of NK cells may be reversed during tumour progression. Recent studies demonstrated that hepatic NK cells were exhausted in hepatocellular carcinoma (HCC) and exhibited impaired cytolytic activity and decreased production of effector cytokines. The present review discusses current knowledge on the role of exhausted NK cells in promoting HCC development and the mechanisms contributing to tumour immune escape, including an imbalance of activating and inhibitory receptors on NK cells, abnormal receptor-ligand interaction, and cross-talk with immune cells and other stromal cells in the tumour environment. We provide a fundamental basis for further study of innate immunity in tumour progression and serve the purpose of exploring new HCC treatment strategies.
Collapse
|
228
|
Medeiros B, Allan AL. Molecular Mechanisms of Breast Cancer Metastasis to the Lung: Clinical and Experimental Perspectives. Int J Mol Sci 2019; 20:E2272. [PMID: 31071959 PMCID: PMC6540248 DOI: 10.3390/ijms20092272] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/01/2019] [Accepted: 05/06/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer in women worldwide, and >90% of breast cancer-related deaths are associated with metastasis. Breast cancer spreads preferentially to the lung, brain, bone and liver; termed organ tropism. Current treatment methods for metastatic breast cancer have been ineffective, compounded by the lack of early prognostic/predictive methods to determine which organs are most susceptible to developing metastases. A better understanding of the mechanisms that drive breast cancer metastasis is crucial for identifying novel biomarkers and therapeutic targets. Lung metastasis is of particular concern as it is associated with significant patient morbidity and a mortality rate of 60-70%. This review highlights the current understanding of breast cancer metastasis to the lung, including discussion of potential new treatment approaches for development.
Collapse
Affiliation(s)
- Braeden Medeiros
- London Regional Cancer Program, London Health Sciences Centre, Department of Anatomy & Cell Biology, Western University, London, ON N6A 5W9, Canada.
| | - Alison L Allan
- London Regional Cancer Program, London Health Sciences Centre, Departments of Anatomy & Cell Biology and Oncology, Western University, London, ON N6A 5W9, Canada.
| |
Collapse
|
229
|
Flores-Martín JF, Perea F, Exposito-Ruiz M, Carretero FJ, Rodriguez T, Villamediana M, Ruiz-Cabello F, Garrido F, Cózar-Olmo JM, Aptsiauri N. A Combination of Positive Tumor HLA-I and Negative PD-L1 Expression Provides an Immune Rejection Mechanism in Bladder Cancer. Ann Surg Oncol 2019; 26:2631-2639. [PMID: 31011905 DOI: 10.1245/s10434-019-07371-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND Tumor human leukocyte antigen class I (HLA-I) expression plays an important role in T cell-mediated tumor rejection. Loss of HLA-I is associated with cancer progression and resistance to immunotherapy, including antibodies blocking programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) signaling. Our objective was to analyze a correlation between HLA-I, tumor immune infiltration, and PD-L1/PD-1 axis in bladder cancer in association with the clinicopathologic features of patients. METHODS We analyzed 85 cryopreserved bladder tumors by immunohistochemistry to investigate the expression of HLA-I, PD-L1, PD-1, CD3, CD8, and CXC chemokine receptor 4 (CXCR4). The results were correlated with tumor stage and other clinicopathologic variables of patients. RESULTS We found a strong positive correlation between tumor HLA-I expression and infiltration with CD3+ and CD8 + T cells. PD-L1 expression was positive in 15.5% of tumors and heterogeneous in 40.5%, and was linked to a more advanced tumor stage. The majority of HLA-I-positive/heterogeneous tumors also expressed PD-L1 and PD-1, which were significantly correlated with each other and with lymphocyte infiltration. Interestingly, the analysis of the simultaneous expression of both markers revealed that 85.2% of tumors with a positive/heterogeneous HLA-I phenotype and negative for PD-L1 were mostly non-invasive, representing a 'tumor rejection' immune phenotype. CONCLUSIONS High tumor HLA-I expression with absence of PD-L1 provides bladder cancer with an immune rejection mechanism. Evaluation of PD-L1 and HLA-I together should be considered in bladder cancer and may provide a new predictive biomarker of tumor invasiveness and of the response to 'immune checkpoint' therapy.
Collapse
Affiliation(s)
- José Francisco Flores-Martín
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Granada, Spain. .,Unidad de Gestión Clínica de Urología, Hospital Universitario Virgen de las Nieves de Granada, Granada, Spain. .,Unidad de Gestión Clínica de Urología, Complejo Hospitalario Universitario de Jaén, Jaén, Spain.
| | - Francisco Perea
- Departamento de Bioquímica, Biología Molecular e Inmunología III, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Manuela Exposito-Ruiz
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Granada, Spain.,Fundación para la Investigación Biosanitaria de Andalucía Oriental (FIBAO), Granada, Spain
| | - Francisco Javier Carretero
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Granada, Spain.,Departamento de Bioquímica, Biología Molecular e Inmunología III, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Teresa Rodriguez
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Granada, Spain.,Departamento de Bioquímica, Biología Molecular e Inmunología III, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Marina Villamediana
- Departamento de Bioquímica, Biología Molecular e Inmunología III, Facultad de Medicina, Universidad de Granada, Granada, Spain.,Unidad de Laboratorio Clínico, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Francisco Ruiz-Cabello
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Granada, Spain.,Departamento de Bioquímica, Biología Molecular e Inmunología III, Facultad de Medicina, Universidad de Granada, Granada, Spain.,Unidad de Laboratorio Clínico, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Federico Garrido
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Granada, Spain.,Departamento de Bioquímica, Biología Molecular e Inmunología III, Facultad de Medicina, Universidad de Granada, Granada, Spain.,Unidad de Laboratorio Clínico, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - José Manuel Cózar-Olmo
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Granada, Spain.,Unidad de Gestión Clínica de Urología, Hospital Universitario Virgen de las Nieves de Granada, Granada, Spain
| | - Natalia Aptsiauri
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Granada, Spain.,Departamento de Bioquímica, Biología Molecular e Inmunología III, Facultad de Medicina, Universidad de Granada, Granada, Spain
| |
Collapse
|
230
|
Würfel FM, Winterhalter C, Trenkwalder P, Wirtz RM, Würfel W. European Patent in Immunoncology: From Immunological Principles of Implantation to Cancer Treatment. Int J Mol Sci 2019; 20:ijms20081830. [PMID: 31013867 PMCID: PMC6514949 DOI: 10.3390/ijms20081830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 12/19/2022] Open
Abstract
The granted European patent EP 2 561 890 describes a procedure for an immunological treatment of cancer. It is based on the principles of the HLA-supported communication of implantation and pregnancy. These principles ensure that the embryo is not rejected by the mother. In pregnancy, the placenta, more specifically the trophoblast, creates an “interface” between the embryo/fetus and the maternal immune system. Trophoblasts do not express the “original” HLA identification of the embryo/fetus (HLA-A to -DQ), but instead show the non-classical HLA groups E, F, and G. During interaction with specific receptors of NK cells (e.g., killer-immunoglobulin-like receptors (KIR)) and lymphocytes (lymphocyte-immunoglobulin-like receptors (LIL-R)), the non-classical HLA groups inhibit these immunocompetent cells outside pregnancy. However, tumors are known to be able to express these non-classical HLA groups and thus make use of an immuno-communication as in pregnancies. If this occurs, the prognosis usually worsens. This patent describes, in a first step, the profiling of the non-classical HLA groups in primary tumor tissue as well as metastases and recurrent tumors. The second step comprises tailored antibody therapies, which is the subject of this patent. In this review, we analyze the underlying mechanisms and describe the currently known differences between HLA-supported communication of implantation and that of tumors.
Collapse
Affiliation(s)
- Franziska M Würfel
- STRATIFYER Molecular Pathology GmbH, D-50935 Cologne, Werthmannstrasse 1c, 50935 Cologne, Germany.
| | | | | | - Ralph M Wirtz
- STRATIFYER Molecular Pathology GmbH, D-50935 Cologne, Werthmannstrasse 1c, 50935 Cologne, Germany.
| | | |
Collapse
|
231
|
Piranlioglu R, Lee E, Ouzounova M, Bollag RJ, Vinyard AH, Arbab AS, Marasco D, Guzel M, Cowell JK, Thangaraju M, Chadli A, Hassan KA, Wicha MS, Celis E, Korkaya H. Primary tumor-induced immunity eradicates disseminated tumor cells in syngeneic mouse model. Nat Commun 2019; 10:1430. [PMID: 30926774 PMCID: PMC6441000 DOI: 10.1038/s41467-019-09015-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 02/14/2019] [Indexed: 02/07/2023] Open
Abstract
Although clinically apparent metastasis is associated with late stages of cancer development, micro-metastatic dissemination may be an early event. However, the fate of these early disseminated tumor cells (DTC) remains elusive. We show that despite their capacity to disseminate into secondary organs, 4T1 tumor models develop overt metastasis while EMT6-tumor bearing mice clear DTCs shed from primary tumors as well as those introduced by intravenous (IV) injection. Following the surgical resection of primary EMT6 tumors, mice do not develop detectable metastasis and reject IV-injected tumor cells. In contrast, these cells readily grow and metastasize in immuno-deficient athymic or Rag2−/− mice, an effect mimicked by CD8+ T-cell depletion in immunocompetent mice. Furthermore, recombinant G-CSF or adoptive transfer of granulocytic-MDSCs isolated from 4T1 tumor-bearing mice, induce metastasis by suppressing CD8+ T-cells in EMT6-primed mice. Our studies support the concept of immune surveillance providing molecular insights into the immune mechanisms during tumor progression. Dissemination of tumor cells from the primary site is an early event. Here, the authors show that the early disseminated tumor cells are actively cleared by the host cytotoxic T lymphocytes induced by the primary tumor and that infiltration of granulocytic myeloid-derived suppressor cells counteracts such immune protection and allow metastasis development.
Collapse
Affiliation(s)
- Raziye Piranlioglu
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - EunMi Lee
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Maria Ouzounova
- Cancer Research Center of Lyon, 28 Rue Laennec, 69008, Lyon, France
| | - Roni J Bollag
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Alicia H Vinyard
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Ali S Arbab
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Daniela Marasco
- Department of Pharmacy, University of Naples "Federico II", 80134, Naples, Italy
| | - Mustafa Guzel
- Regenerative and Restorative Research Center (REMER), Medipol University, Kavacık Mah. Ekinciler Cad. No.19 Kavacık Kavşağı - Beykoz, 34810, İstanbul Istanbul, Turkey
| | - John K Cowell
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Muthushamy Thangaraju
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Ahmed Chadli
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Khaled A Hassan
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Max S Wicha
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Esteban Celis
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA
| | - Hasan Korkaya
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Blvd. CN2136, Augusta, GA, 30912, USA.
| |
Collapse
|
232
|
Guo Y, Ji X, Liu J, Fan D, Zhou Q, Chen C, Wang W, Wang G, Wang H, Yuan W, Ji Z, Sun Z. Effects of exosomes on pre-metastatic niche formation in tumors. Mol Cancer 2019; 18:39. [PMID: 30857545 PMCID: PMC6413442 DOI: 10.1186/s12943-019-0995-1] [Citation(s) in RCA: 276] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 02/28/2019] [Indexed: 12/15/2022] Open
Abstract
A pre-metastatic niche is a microenvironment prepared for the colonization of circulating tumor cells in specific organs. Exosomes are extracellular vesicles with a variety of biological functions. Exosomes play an irreplaceable role in the development of pre-metastatic niches, and mainly function as communication medium. In this review, we analyzed the effects of exosomes on pre-metastatic niches from various perspectives, including inflammation, immune response, angiogenesis, organotropism, matrix remodeling and biomarker expression. In particular, exosomes express programmed death ligand 1 (PD-L1) and cause the immune escape of tumor cells. The immunomodulatory effects of exosomes and their potential in liquid diagnosis have drawn our attention. The potential value of exosomes and pre-metastatic niches will be realized in the field of immunity therapy.
Collapse
Affiliation(s)
- Yaxin Guo
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China.,School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xiang Ji
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jinbo Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Dandan Fan
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Quanbo Zhou
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chen Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Guixian Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Haijiang Wang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, Ürümqi, 830011, Xinjiang, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Zhenyu Ji
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China. .,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China. .,School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China. .,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China. .,Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, Ürümqi, 830011, Xinjiang, China.
| |
Collapse
|
233
|
Ingangi V, Minopoli M, Ragone C, Motti ML, Carriero MV. Role of Microenvironment on the Fate of Disseminating Cancer Stem Cells. Front Oncol 2019; 9:82. [PMID: 30847298 PMCID: PMC6393337 DOI: 10.3389/fonc.2019.00082] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
Disseminating Cancer Stem Cells (CSCs) initiate growth in specific niches of the host tissues, the cellular and molecular components of which sustain signaling pathways that support their survival, self-renewal dormancy and reactivation. In the metastatic niche, tumor cells may enter in a dormant state to survive and, consequently, the metastasis can remain latent for years. Despite the clinical importance of metastatic latency, little is known about what induces CSCs to enter a dormant state and what allows them to remain viable for years in this state. CSCs exhibit genetic, epigenetic and cellular adaptations that confer resistance to classical therapeutic approaches. The identification of potential CSC targets is complicated by the fact that CSCs may arise as a consequence of their relationship with the local microenvironment into the metastatic niches. Indeed, microenvironment modulates the capability of CSCs to evade the innate immune response and survive. Some new therapeutic options that include drugs targeting microenvironment components are achieving encouraging results in reducing the number of CSCs in tumors and/or overcoming their resistance in preclinical studies. This review will focus on specific CSC features with an emphasis on the role of tumor microenvironment in supporting metastatic dissemination of CSCs. In addition, it sheds light on potential microenvironment-targeted therapies aimed to counteract seeding and survival of CSCs in the metastatic niche.
Collapse
Affiliation(s)
- Vincenzo Ingangi
- IRCCS Istituto Nazionale Tumori, Fondazione G. Pascale, Naples, Italy
| | - Michele Minopoli
- IRCCS Istituto Nazionale Tumori, Fondazione G. Pascale, Naples, Italy
| | - Concetta Ragone
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maria Letizia Motti
- IRCCS Istituto Nazionale Tumori, Fondazione G. Pascale, Naples, Italy.,Department of Sport Science and Wellness, University Parthenope, Naples, Italy
| | | |
Collapse
|
234
|
Reale A, Vitiello A, Conciatori V, Parolin C, Calistri A, Palù G. Perspectives on immunotherapy via oncolytic viruses. Infect Agent Cancer 2019; 14:5. [PMID: 30792754 PMCID: PMC6371415 DOI: 10.1186/s13027-018-0218-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/20/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND With few exceptions, current chemotherapy and radiotherapy protocols only obtain a slightly prolonged survival with severe adverse effects in patients with advanced solid tumors. In particular, most solid malignancies not amenable to radical surgery still carry a dismal prognosis, which unfortunately is also the case for relapsing disease after surgery. Even though targeted therapies obtained good results, clinical experience showed that tumors eventually develop resistance. On the other hand, earlier attempts of cancer immunotherapy failed to show consistent efficacy. More recently, a deeper knowledge of immunosuppression in the tumor microenvironment (TME) allowed the development of effective drugs: in particular, monoclonal antibodies targeting the so-called immune checkpoint molecules yielded striking and lasting effects in some tumors. Unfortunately, these monoclonal antibodies are not effective in a majority of patients and are ineffective in several solid malignancies. Furthermore, due to their mechanism of action, checkpoint inhibitors often elicit autoimmune-like disease. MAIN BODY The use of viruses as oncolytic agents (OVs) was considered in the past, while only recently OVs revealed a connection with immunotherapy. However, their antitumoral potential has remained largely unexplored, due to safety concerns and some limitations in the techniques to manipulate viruses. OV research was recently revived by a better knowledge of viral/cancer biology and advances in the methodologies to delete virulence/immune-escape related genes from even complex viral genomes or "to arm" OVs with appropriate transgenes. Recently, the first oncolytic virus, the HSV-1 based Talimogene Laherparepvec (T-VEC), was approved for the treatment of non-resectable melanoma in USA and Europe. CONCLUSION OVs have the potential to become powerful agents of cancer immune and gene therapy. Indeed, in addition to their selective killing activity, they can act as versatile gene expression platforms for the delivery of therapeutic genes. This is particularly true for viruses with a large DNA genome, that can be manipulated to address the multiple immunosuppressive features of the TME. This review will focus on the open issues, on the most promising lines of research in the OV field and, more in general, on how OVs could be improved to achieve real clinical breakthroughs in cancers that are usually difficult to treat by immunotherapy.
Collapse
Affiliation(s)
- Alberto Reale
- Department of Molecular Medicine, University of Padua, Via A. Gabelli, 63, 35121 Padua, Italy
| | - Adriana Vitiello
- Department of Molecular Medicine, University of Padua, Via A. Gabelli, 63, 35121 Padua, Italy
| | - Valeria Conciatori
- Department of Molecular Medicine, University of Padua, Via A. Gabelli, 63, 35121 Padua, Italy
| | - Cristina Parolin
- Department of Molecular Medicine, University of Padua, Via A. Gabelli, 63, 35121 Padua, Italy
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padua, Via A. Gabelli, 63, 35121 Padua, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padua, Via A. Gabelli, 63, 35121 Padua, Italy
| |
Collapse
|
235
|
Sun Q, Barz M, De Geest BG, Diken M, Hennink WE, Kiessling F, Lammers T, Shi Y. Nanomedicine and macroscale materials in immuno-oncology. Chem Soc Rev 2019; 48:351-381. [PMID: 30465669 PMCID: PMC7115880 DOI: 10.1039/c8cs00473k] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immunotherapy is revolutionizing the treatment of cancer. It can achieve unprecedented responses in advanced-stage patients, including complete cures and long-term survival. However, immunotherapy also has limitations, such as its relatively low response rates and the development of severe side effects. These drawbacks are gradually being overcome by improving our understanding of the immune system, as well as by establishing combination regimens in which immunotherapy is combined with other treatment modalities. In addition to this, in recent years, progress made in chemistry, nanotechnology and materials science has started to impact immuno-oncology, resulting in more effective and less toxic immunotherapy interventions. In this context, multiple different nanomedicine formulations and macroscale materials have been shown to be able to boost anti-cancer immunity and the efficacy of immunomodulatory drugs. We here review nanotechnological and materials chemistry efforts related to endogenous and exogenous vaccination, to the engineering of antigen-presenting cells and T cells, and to the modulation of the tumor microenvironment. We also discuss limitations, current trends and future directions. Together, the insights provided and the evidence obtained indicate that there is a bright future ahead for engineering nanomedicines and macroscale materials for immuno-oncology applications.
Collapse
Affiliation(s)
- Qingxue Sun
- Department of Nanomedicines and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany
| | - Matthias Barz
- Institute of Organic Chemistry, Johannes Gutenberg University, 55099 Mainz, Germany
| | - Bruno G. De Geest
- Department of Pharmaceutics, Ghent University, B-9000 Ghent, Belgium
| | - Mustafa Diken
- TRON - Translational Oncology at the University Medical Center of Johannes Gutenberg University Mainz gGmbH, 55131, Mainz, Germany
| | - Wim E. Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Fabian Kiessling
- Department of Nanomedicines and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany
- Fraunhofer MEVIS, Institute for Medical Image Computing, 52074 Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicines and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
- Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7500 AE Enschede, The Netherlands
| | - Yang Shi
- Department of Nanomedicines and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
236
|
Cheng H, Fan GL, Fan JH, Zheng RR, Zhao LP, Yuan P, Zhao XY, Yu XY, Li SY. A Self-Delivery Chimeric Peptide for Photodynamic Therapy Amplified Immunotherapy. Macromol Biosci 2018; 19:e1800410. [PMID: 30576082 DOI: 10.1002/mabi.201800410] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/04/2018] [Indexed: 01/28/2023]
Abstract
In this paper, a self-delivery chimeric peptide PpIX-PEG8 -KVPRNQDWL is designed for photodynamic therapy (PDT) amplified immunotherapy against malignant melanoma. After self-assembly into nanoparticles (designated as PPMA), this self-delivery system shows high drug loading rate, good dispersion, and stability as well as an excellent capability in producing reactive oxygen species (ROS). After cellular uptake, the ROS generated under light irradiation could induce the apoptosis and/or necrosis of tumor cells, which would subsequently stimulate the anti-tumor immune response. On the other hand, the melanoma specific antigen (KVPRNQDWL) peptide could also activate the specific cytotoxic T cells for anti-tumor immunity. Compared to immunotherapy alone, the combined photodynamic immunotherapy exhibits significantly enhanced inhibition of melanoma growth. Both in vitro and in vivo investigations confirm that PDT of PPMA has a positive effect on anti-tumor immune response. This self-delivery system demonstrates a great potential of this PDT amplified immunotherapy strategy for advanced or metastatic tumor treatment.
Collapse
Affiliation(s)
- Hong Cheng
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Gui-Ling Fan
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Jing-Hao Fan
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Rong-Rong Zheng
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Lin-Ping Zhao
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Ping Yuan
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Xiao-Ya Zhao
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Xi-Yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Shi-Ying Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| |
Collapse
|
237
|
Zhao Y, Wang B, Liu J, Sun P, Liu H. An overview on the methods of determining the activity of Indoleamine 2, 3-Dioxygenase 1. J Drug Target 2018; 27:724-731. [DOI: 10.1080/1061186x.2018.1523416] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Yuandi Zhao
- Collaborative Innovation Center of New Drug Research and Safety, Henan Province, PR China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, Henan Province, PR China
- Key Laboratory of Henan Province for Drug Quality and Evaluation Zhengzhou University, Zhengzhou, Henan Province, PR China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Bo Wang
- Collaborative Innovation Center of New Drug Research and Safety, Henan Province, PR China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, Henan Province, PR China
- Key Laboratory of Henan Province for Drug Quality and Evaluation Zhengzhou University, Zhengzhou, Henan Province, PR China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Jinzhi Liu
- Collaborative Innovation Center of New Drug Research and Safety, Henan Province, PR China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, Henan Province, PR China
- Key Laboratory of Henan Province for Drug Quality and Evaluation Zhengzhou University, Zhengzhou, Henan Province, PR China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Pei Sun
- Collaborative Innovation Center of New Drug Research and Safety, Henan Province, PR China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, Henan Province, PR China
- Key Laboratory of Henan Province for Drug Quality and Evaluation Zhengzhou University, Zhengzhou, Henan Province, PR China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Hongmin Liu
- Collaborative Innovation Center of New Drug Research and Safety, Henan Province, PR China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, Henan Province, PR China
- Key Laboratory of Henan Province for Drug Quality and Evaluation Zhengzhou University, Zhengzhou, Henan Province, PR China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
| |
Collapse
|
238
|
Tantyo NA, Karyadi AS, Rasman SZ, Salim MRG, Devina A, Sumarpo A. The prognostic value of S100A10 expression in cancer. Oncol Lett 2018; 17:1417-1424. [PMID: 30675195 PMCID: PMC6341771 DOI: 10.3892/ol.2018.9751] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 11/15/2018] [Indexed: 12/30/2022] Open
Abstract
S100A10, a member of the S100 protein family, commonly forms a heterotetrameric complex with Annexin A2. This is essential for the generation of cellular plasmin from plasminogen, which leads to a cascade of molecular events crucial for tumor progression. S100A10 upregulation has been reported in a number of cancers, suggesting that it may have potential as a prognostic biomarker, as well as predicting sensitivity to anticancer drugs. This review evaluates the direct and indirect relationships between S100A10 and cancer progression by investigating its role in cancer. Research papers published on PubMed and Google Scholar between 2007–2017 were collated and reviewed. We concluded that S100A10 affects the development of the hallmarks of cancer as explained by Hanahan and Weinberg in 2011, most notably by activating the invasion and metastasis of cancer cells. However, further studies are required to explore the underlying biological mechanisms of S100A10.
Collapse
Affiliation(s)
- Normastuti Adhini Tantyo
- Department of Biomedicine, Indonesia International Institute for Life Sciences, Jakarta Timur 13210, Indonesia
| | - Azrina Saraswati Karyadi
- Department of Biomedicine, Indonesia International Institute for Life Sciences, Jakarta Timur 13210, Indonesia
| | - Siti Zulimas Rasman
- Department of Biomedicine, Indonesia International Institute for Life Sciences, Jakarta Timur 13210, Indonesia
| | | | - Astrella Devina
- Department of Biomedicine, Indonesia International Institute for Life Sciences, Jakarta Timur 13210, Indonesia
| | - Anton Sumarpo
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta Utara 14440, Indonesia
| |
Collapse
|
239
|
Li K, Chen Y, Li A, Tan C, Liu X. Exosomes play roles in sequential processes of tumor metastasis. Int J Cancer 2018; 144:1486-1495. [PMID: 30155891 DOI: 10.1002/ijc.31774] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 06/22/2018] [Accepted: 07/17/2018] [Indexed: 02/05/2023]
Abstract
Overwhelming evidence demonstrates that exosomes, a series of biologically functional small vesicles of endocytic origin carrying a variety of active constituents, especially tumor-derived exosomes, contribute to tumor progression and metastasis. This review focuses on the specific multifaceted roles of exosomes in affecting sequenced four crucial processes of metastasis, through which cancer cells spread from primary to secondary organs and finally form macroscopic metastatic lesions. First, exosomes modulate the primary tumor sites to assist cancer growth and dissemination. In this part, five main biological events are reviewed, including the transfer of oncogenic constituents, the recruitment and activation of fibroblasts, the induction of angiogenesis, immunosuppression and epithelial-mesenchymal transition (EMT) promotion. In Step 2, we list two recently disclosed mechanisms during the organ-specific homing process: the exosomal integrin model and exosomal epidermal growth factor receptor (EGFR)/miR-26/hepatocyte growth factor (HGF) model. Subsequently, Step 3 focuses on the interactions between exosomes and pre-metastatic niche, in which we highlight the specific functions of exosomes in angiogenesis, lymphangiogenesis, immune modulation and metabolic, epigenetic and stromal reprogramming of pre-metastatic niche. Finally, we summarize the mechanisms of exosomes in helping the metastatic circulating tumor cells escape from immunologic surveillance, survive in the blood circulation and proliferate in host organs.
Collapse
Affiliation(s)
- Keyu Li
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yonghua Chen
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ang Li
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Chunlu Tan
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xubao Liu
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
240
|
Lybaert L, Vermaelen K, De Geest BG, Nuhn L. Immunoengineering through cancer vaccines – A personalized and multi-step vaccine approach towards precise cancer immunity. J Control Release 2018; 289:125-145. [DOI: 10.1016/j.jconrel.2018.09.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 02/07/2023]
|
241
|
Pawlik A, Anisiewicz A, Filip-Psurska B, Nowak M, Turlej E, Trynda J, Banach J, Gretkierewicz P, Wietrzyk J. Calcitriol and Its Analogs Establish the Immunosuppressive Microenvironment That Drives Metastasis in 4T1 Mouse Mammary Gland Cancer. Int J Mol Sci 2018; 19:ijms19072116. [PMID: 30037009 PMCID: PMC6073894 DOI: 10.3390/ijms19072116] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/15/2018] [Accepted: 07/18/2018] [Indexed: 02/07/2023] Open
Abstract
In our previous study, calcitriol and its analogs PRI-2191 and PRI-2205 stimulated 4T1 mouse mammary gland cancer metastasis. Therefore, we aimed to analyze the inflammatory response in 4T1-bearing mice treated with these compounds. Gene expression analysis of the splenocytes and regional lymph nodes demonstrated prevalence of the T helper lymphocytes (Th2) response with an increased activity of regulatory T (Treg) lymphocytes in mice treated with these compounds. We also observed an increased number of mature granulocytes and B lymphocytes and a decreased number of TCD4+, TCD4+CD25+, and TCD8+, as well as natural killer (NK) CD335+, cells in the blood of mice treated with calcitriol and its analogs. Among the splenocytes, we observed a significant decrease in NK CD335+ cells and an increase in TCD8+ cells. Calcitriol and its analogs decreased the levels of interleukin (IL)-1β and IL-10 and increased the level of interferon gamma (IFN-γ) in the plasma. In the tumor tissue, they caused an increase in the level of IL-10. Gene expression analysis of lung tissue demonstrated an increased level of osteopontin (Spp1) and transforming growth factor β (TGF-β) mRNA. The expression of Spp1 was also elevated in lymph nodes. Calcitriol and its analogs caused prevalence of tumor-conducive changes in the immune system of 4T1 tumor-bearing mice, despite the induction of some tumor-disadvantageous effects.
Collapse
Affiliation(s)
- Agata Pawlik
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland.
| | - Artur Anisiewicz
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland.
| | - Beata Filip-Psurska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland.
| | - Marcin Nowak
- Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 50-3754 Wroclaw, Poland.
| | - Eliza Turlej
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland.
| | - Justyna Trynda
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland.
| | - Joanna Banach
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland.
| | - Paweł Gretkierewicz
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland.
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland.
| |
Collapse
|
242
|
|
243
|
Ye Z, Qian Q, Jin H, Qian Q. Cancer vaccine: learning lessons from immune checkpoint inhibitors. J Cancer 2018; 9:263-268. [PMID: 29344272 PMCID: PMC5771333 DOI: 10.7150/jca.20059] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023] Open
Abstract
Cancer vaccines have been exclusively studied all through the past decades, and have made exceptional achievements in cancer treatment. Few cancer vaccines have been approved by the US Food and Drug Administration (FDA), for instance, Provenge, which was approved for the treatment of prostate carcinoma in 2012. Moreover, more recently, T-VEC got approval for the treatment of melanoma. While, the overall therapeutic effects of cancer vaccines have been taken into consideration as below expectations, low antigenicity of targeting antigen and tumor heterogeneity are the two key limiting barriers encountered by the cancer vaccines. Nonetheless, recent developments in cancer immune-therapies together with associated technologies, for instance the unparalleled achievements bagged by immune checkpoint inhibitor based therapies and neo-antigen identification tools, envisage potential improvements in cancer vaccines in respect to the treatments of malignancies. This review brings forth measures for the purpose of refining therapeutic cancer vaccines by learning lessons from the success of PD-1 inhibitor based immune-therapies.
Collapse
Affiliation(s)
- ZhenLong Ye
- Shanghai Engineering Research Center for Cell Therapy, 75 Qianyang Road, Shanghai 201805, China
| | - Qiming Qian
- Shanghai Engineering Research Center for Cell Therapy, 75 Qianyang Road, Shanghai 201805, China
| | - HuaJun Jin
- Shanghai Engineering Research Center for Cell Therapy, 75 Qianyang Road, Shanghai 201805, China
| | - QiJun Qian
- Shanghai Engineering Research Center for Cell Therapy, 75 Qianyang Road, Shanghai 201805, China
| |
Collapse
|
244
|
Shen CT, Zhang Y, Liu YM, Yin S, Zhang XY, Wei WJ, Sun ZK, Song HJ, Qiu ZL, Wang CR, Luo QY. A distinct serum metabolic signature of distant metastatic papillary thyroid carcinoma. Clin Endocrinol (Oxf) 2017; 87:844-852. [PMID: 28755525 DOI: 10.1111/cen.13437] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/16/2017] [Accepted: 07/21/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Although the incidence rate for thyroid cancer seems to have begun stabilizing in recent years, an increased rate of advanced stage of this disease has been reported. Additionally, distant metastasis is one of the most important prognostic factors of patients with papillary thyroid carcinoma (PTC). Unfortunately, the underlying mechanisms of distant metastasis, as well as cell status like metabolism changes in distant metastatic tumours have not been clearly elucidated. OBJECTIVE To identify serum metabolic signature of distant metastatic PTC. DESIGN, PATIENTS AND MEASUREMENTS In this study, gas chromatography-time-of-flight mass spectrometry (GC-TOF-MS) was used to analyse the serum from 77 patients diagnosed with PTC (37 in distant metastasis group and 40 in ablation group). Principal component analysis (PCA) and orthogonal partial least-squares-discriminant analysis (OPLS-DA) scores plots were used to analyse the data. RESULTS Principal component analysis and OPLS-DA analyses demonstrated an evident trend of separation between 40 serum samples from the ablation group and 37 samples from distant metastasis group. A total of 31 metabolites were identified, which are related to amino acid, lipid, glucose, vitamin metabolism and diet/gut microbiota interaction. Pathway analysis showed "alanine, aspartate and glutamate metabolism" and "inositol phosphate metabolism" were the most relevant pathways. CONCLUSION Serum metabolomics profiling could significantly discriminate papillary thyroid cancer patients according to distant metastasis. Potential metabolic aberration in distant metastatic PTC could be involved in different biological behaviours of tumour cells including proliferation, invasion/migration and immune escape. Diet/gut microbiota-produced metabolites could play an important role in these effects. This work may provide new clues to find the underlying mechanisms regarding the distant metastasis of PTC as well as potential adjuvant therapy targets.
Collapse
Affiliation(s)
- Chen-Tian Shen
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yinan Zhang
- Shanghai Key Laboratory of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- The Metabolic Diseases Biobank, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yu-Min Liu
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, China
| | - Shan Yin
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, China
| | - Xin-Yun Zhang
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wei-Jun Wei
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhen-Kui Sun
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hong-Jun Song
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhong-Ling Qiu
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Cong-Rong Wang
- Shanghai Key Laboratory of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- The Metabolic Diseases Biobank, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
245
|
Mao M, Wei X, Sheng H, Chi P, Liu Y, Huang X, Xiang Y, Zhu Q, Xing S, Liu W. C-reactive protein/albumin and neutrophil/lymphocyte ratios and their combination predict overall survival in patients with gastric cancer. Oncol Lett 2017; 14:7417-7424. [PMID: 29344182 DOI: 10.3892/ol.2017.7179] [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: 09/27/2016] [Accepted: 06/27/2017] [Indexed: 12/13/2022] Open
Abstract
Multiple studies have reported the prognostic association of certain inflammatory factors with various types of cancer. The present study assessed the prognostic value of the C-reactive protein (CRP)/albumin (Alb) ratio and the neutrophil/lymphocyte ratio (NLR), separately and in combination, in gastric cancer (GC). A total of 337 cases pathologically diagnosed with gastric adenocarcinoma were retrospectively evaluated. The clinicopathological and prognostic relevance of the CRP/Alb ratio and NLR and their combination were analyzed. The optimal cut-off values of the CRP/Alb ratio and NLR were 0.38 and 3.14, respectively. High CRP/Alb ratio (≥0.38) and NLR (≥3.14) values were associated with increased tumor invasion, more distant metastasis and a more advanced tumor-node-metastasis stage (all P<0.05). In addition, a high NLR value was also associated with increased tumor size (P=0.02). The CRP/Alb ratio (≥0.38/<0.38) and NLR (≥3.14/<3.14) were independent prognostic factors for overall survival time (OS) in GC by multivariate analysis (P=0.005 and P=0.001). Using the CRP/Alb ratio and NLR classification, patients were stratified into three subgroups with different OS time (P<0.001), which were identified as independent prognostic variables in multivariate analysis (P<0.001). The present study demonstrated that the CRP/Alb ratio and NLR were independent prognostic factors for OS in patients with GC. The combination of these indexes was associated with significant prognostic value and may further stratify prognosis.
Collapse
Affiliation(s)
- Minjie Mao
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Xiaoli Wei
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Hui Sheng
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Peidong Chi
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Yijun Liu
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Xiaoyan Huang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510275, P.R. China
| | - Yifan Xiang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510275, P.R. China
| | - Qianying Zhu
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Shan Xing
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Wanli Liu
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
246
|
Wang M, Chen B, Sun XX, Zhao XD, Zhao YY, Sun L, Xu CG, Shen B, Su ZL, Xu WR, Zhu W. Gastric cancer tissue-derived mesenchymal stem cells impact peripheral blood mononuclear cells via disruption of Treg/Th17 balance to promote gastric cancer progression. Exp Cell Res 2017; 361:19-29. [PMID: 28964780 DOI: 10.1016/j.yexcr.2017.09.036] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 09/08/2017] [Accepted: 09/26/2017] [Indexed: 12/20/2022]
Abstract
Gastric cancer tissue-derived mesenchymal stem cells (GC-MSCs) are important resident stromal cells in the tumor microenvironment (TME) and have been shown to play a key role in gastric cancer progression. Whether GC-MSCs exert a tumor-promoting function by affecting anti-tumor immunity is still unclear. In this study, we used GC-MSC conditioned medium (GC-MSC-CM) to pretreat peripheral blood mononuclear cells (PBMCs) from healthy donors. We found that GC-MSC-CM pretreatment markedly reversed the inhibitory effect of PBMCs on gastric cancer growth in vivo, but did not affect functions of PBMCs on gastric cancer cell proliferation, cell cycle and apoptosis in vitro. PBMCs pretreated with GC-MSC-CM significantly promoted gastric cancer migration and epithelial-mesenchymal transition in vitro and liver metastases in vivo. Flow cytometry analysis showed that GC-MSC-CM pretreatment increased the proportion of Treg cells and reduced that of Th17 cells in PBMCs. CFSE labeling and naïve CD4+ T cells differentiation analysis revealed that GC-MSC-CM disrupted the Treg/Th17 balance in PBMCs by suppressing Th17 cell proliferation and inducing differentiation of Treg cells. Overall, our collective results indicate that GC-MSCs impair the anti-tumor immune response of PBMCs through disruption of Treg/Th17 balance, thus providing new evidence that gastric cancer tissue-derived MSCs contribute to the immunosuppressive TME.
Collapse
Affiliation(s)
- Mei Wang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bin Chen
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiao-Xian Sun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiang-Dong Zhao
- Zhenjiang Provincial Blood Center, Zhenjiang, Jiangsu, China
| | - Yuan-Yuan Zhao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Li Sun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chang-Gen Xu
- Zhenjiang Provincial Blood Center, Zhenjiang, Jiangsu, China
| | - Bo Shen
- Department of Oncology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhao-Liang Su
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wen-Rong Xu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wei Zhu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
247
|
STING signaling in tumorigenesis and cancer therapy: A friend or foe? Cancer Lett 2017; 402:203-212. [PMID: 28602976 DOI: 10.1016/j.canlet.2017.05.026] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/20/2017] [Accepted: 05/31/2017] [Indexed: 12/19/2022]
Abstract
Stimulator of interferon genes (STING) is a DNA sensor and an important cytoplasmic adaptor for other DNA sensors, such as Z-DNA binding protein 1 (DAI), DEAD-box helicase 41 (DDX41), and interferon-γ-inducible protein 16 (IFI16). The activation of STING signaling leads to the production of type I interferons and some other pro-inflammatory cytokines, which are critical for host defense against viral infection. Recent accumulating evidences suggest that STING is also involved in tumor development. However, the role of STING signaling in tumorigenesis is complicated, and a comprehensive review is still lacking. In this paper, we provided an overview of the dual role of STING signaling in tumor development from clinical significance to fundamental mechanisms, as well as its pre-clinical application in cancer therapy.
Collapse
|
248
|
Fang J, Liu C, Wang Q, Lin P, Cheng F. In silico polypharmacology of natural products. Brief Bioinform 2017; 19:1153-1171. [DOI: 10.1093/bib/bbx045] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Indexed: 12/16/2022] Open
|
249
|
Li YC, Zou JM, Luo C, Shu Y, Luo J, Qin J, Wang Y, Li D, Wang SS, Chi G, Guo F, Zhang GM, Feng ZH. Circulating tumor cells promote the metastatic colonization of disseminated carcinoma cells by inducing systemic inflammation. Oncotarget 2017; 8:28418-28430. [PMID: 28415700 PMCID: PMC5438660 DOI: 10.18632/oncotarget.16084] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/28/2017] [Indexed: 01/18/2023] Open
Abstract
Circulating tumor cells (CTCs) have been studied well in the prognosis for malignant diseases as liquid biopsy, but their contribution to tumor metastasis is not clearly defined. Here we report that CTCs could promote the metastatic colonization of disseminated carcinoma cells by inducing systemic inflammation and neutrophil recruitment to pre-metastatic organs. Depletion of neutrophils in vivo could effectively abrogate the promoting effect of CTCs on tumor cell metastasis. In the presence of CTCs, the pro-tumor function of neutrophils was augmented, whereas the antitumor function of neutrophils was suppressed. Mechanically, CTC-derived ligands for TLR2 and TLR4 (TLR2/4) induced the systemic inflammation, thus increasing the production of proinflammatory cytokines such as G-CSF and IL-6 that could induce the conversion of neutrophil function from tumor-suppressing to tumor-promoting. Moreover, CTCs induced the production of endogenous TLR2/4 ligands such as S100A8, S100A9, and SAA3, which may amplify the stimulating effect that induces the expression of proinflammatory cytokines. The promoting effect of CTCs on tumor cell metastasis could be abrogated by suppressing inflammatory response with IL-37, an anti-inflammatory cytokine, or blocking CTC-derived ligands for TLR2/4. Identification of the metastatic axis of CTCs/systemic inflammation/neutrophils may provide potential targets for preventing tumor cell metastasis.
Collapse
Affiliation(s)
- Yong-Chao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, The People's Republic of China
| | - Jiu-Ming Zou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, The People's Republic of China
| | - Chao Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, The People's Republic of China
| | - Yu Shu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, The People's Republic of China
| | - Jing Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, The People's Republic of China
| | - Jian Qin
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, The People's Republic of China
| | - Yu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, The People's Republic of China
| | - Dong Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, The People's Republic of China
| | - Shan-Shan Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, The People's Republic of China
| | - Gang Chi
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, The People's Republic of China
| | - Fang Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, The People's Republic of China
| | - Gui-Mei Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, The People's Republic of China
| | - Zuo-Hua Feng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, The People's Republic of China
| |
Collapse
|
250
|
Liu Q, Zhu H, Zhang C, Chen T, Cao X. Small GTPase RBJ promotes cancer progression by mobilizing MDSCs via IL-6. Oncoimmunology 2016; 6:e1245265. [PMID: 28197363 DOI: 10.1080/2162402x.2016.1245265] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/29/2016] [Accepted: 10/01/2016] [Indexed: 10/20/2022] Open
Abstract
RBJ has been identified to be dysregulated in gastrointestinal cancer and promotes tumorigenesis and progression by mediating nuclear accumulation of active MEK1/2 and sustained activation of ERK1/2. Considering that nuclear accumulation and constitutive activation of MEK/ERK not only promotes tumor progression directly, but also induces chronic inflammation, we wonder whether and how RBJ impairs host immune-surveillance via chronic inflammation and consequently supports tumor progression. Here, we report that higher expression of RBJ in human breast cancer tissue has been significantly correlated with poorer prognosis in breast cancer patients. The forced expression of RBJ promotes tumor growth and metastasis both in vitro and in vivo. In addition, more accumulation of immune suppressive cells but less antitumor immune cell subpopulations were found in spleen and tumor tissue derived from RBJ force-expressed tumor-bearing mice. Furthermore, forced RBJ expression significantly promotes tumor cell production of pro-inflammatory cytokine IL-6 by constitutive activating MEK/ERK signaling pathway. Accordingly, RBJ knockdown significantly decreases tumor growth and metastasis in vitro and in vivo, with markedly reduced production of IL-6. Administration of anti-IL-6 neutralizing antibody could reduce MDSCs accumulation in tumor tissue in vivo. Therefore, our results demonstrate that RBJ-mediated nuclear constitutive activation of ERK1/2 leads to persistent production of IL-6 and increase of MDSCs recruitment, contributing to promotion of tumor growth and metastasis. These results suggest that RBJ contributes to tumor immune escape, maybe serving a potential target for design of antitumor drug.
Collapse
Affiliation(s)
- Qiuyan Liu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University , Shanghai, China
| | - Ha Zhu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University , Shanghai, China
| | - Chaoxiong Zhang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University , Shanghai, China
| | - Taoyong Chen
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University , Shanghai, China
| | - Xuetao Cao
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China; National Key Laboratory of Medical Molecular Biology & Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|