151
|
Huang B, Huang M, Li Q. Cancer-Associated Fibroblasts Promote Angiogenesis of Hepatocellular Carcinoma by VEGF-Mediated EZH2/VASH1 Pathway. Technol Cancer Res Treat 2020; 18:1533033819879905. [PMID: 31757187 PMCID: PMC6876164 DOI: 10.1177/1533033819879905] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Hepatocellular carcinoma is a highly vascularized tumor, so it is critical to study its angiogenesis. Cancer-associated fibroblasts and enhancer of zeste homolog 2 play an important role in tumor angiogenesis and became significant hallmarks of cancer. But the relationship between enhancer of zeste homolog-2 and cancer-associated fibroblasts in response to angiogenesis and its precise mechanism remains unclear. Methods: The separation of cancer-associated fibroblasts was identified by immunofluorescence. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide analysis was used to reveal the proliferation of human umbilical vein endothelial cells. Vascular endothelial growth factor level was quantified by enzyme-linked immunosorbent assay. The wound healing, transwell, and vascular tube formation assays were used to identify the capability of migration, invasion, and tube formation of human umbilical vein endothelial cells in vitro. The detection of tumor angiogenesis was also performed in vivo. Finally, the level of enhancer of zeste homolog-2 and vasohibin 1 were determined by quantitative real-time polymerase chain reaction and Western blotting. Results: In comparison to control and condition medium noncancerous fibroblasts groups, the condition medium cancer-associated fibroblasts could significantly promote the proliferation, migration, invasion, and angiogenesis of human umbilical vein endothelial cells. We found that cancer-associated fibroblasts promoted angiogenesis of human umbilical vein endothelial cells via vascular endothelial growth factor secretion in vitro and in vivo. The upregulation of enhancer of zeste homolog 2 by vascular endothelial growth factor inhibited the expression of vasohibin 1, thus promoting the proliferation and angiogenesis of human umbilical vein endothelial cells. Taken together, the cancer-associated fibroblasts of hepatocellular carcinoma regulate the enhancer of zeste homolog-2/vasohibin 1 pathway via vascular endothelial growth factor secretion, thereby promoting the proliferation and angiogenesis of human umbilical vein endothelial cells. Conclusion: Our study identified the relationship between cancer-associated fibroblasts and enhancer of zeste homolog-2 and confirmed the pivotal role of cancer-associated fibroblasts in angiogenesis of hepatocellular carcinoma. Cancer-associated fibroblasts promote angiogenesis of hepatocellular carcinoma by vascular endothelial growth factor–mediated enhancer of zeste homolog-2/vasohibin 1 pathway and may be a potentially useful therapeutic target for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Bin Huang
- Department of Intervention, Hunan Provincial Cancer Hospital, Changsha, People's Republic of China
| | - Manping Huang
- Department of Intervention, Hunan Provincial Cancer Hospital, Changsha, People's Republic of China
| | - Qin Li
- Department of Gynecology, Affiliated Hospital of Hunan Institute of Traditional Chinese Medicine, Changsha, People's Republic of China
| |
Collapse
|
152
|
Origin and role of hepatic myofibroblasts in hepatocellular carcinoma. Oncotarget 2020; 11:1186-1201. [PMID: 32284794 PMCID: PMC7138168 DOI: 10.18632/oncotarget.27532] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 03/03/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer and is the second leading cause of cancer-related death worldwide. Fibrosis and cirrhosis are important risk factors for the development of HCC. Hepatic myofibroblasts are the cells responsible for extracellular matrix deposition, which is the hallmark of liver fibrosis. It is believed that myofibroblasts are predominantly derived from hepatic stellate cells (HSCs), also known as Ito cells. Nevertheless, depending on the nature of insult to the liver, it is thought that myofibroblasts may also originate from a variety of other cell types such as the portal fibroblasts (PFs), fibrocytes, hepatocytes, hepatic progenitor cells (HPCs), and mesothelial cells. Liver myofibroblasts are believed to transform into cancer-associated fibroblasts (CAFs) while HCC is developing. There is substantial evidence suggesting that activated HSCs (aHSCs)/cancer-associated fibroblasts (CAFs) may play an important role in HCC initiation and progression. In this paper, we aim to review current literature on cellular origins of myofibroblasts with a focus on hepatitis B virus (HBV)- and hepatitis C virus (HCV)-induced hepatic fibrosis. We also address the role of aHSCs/CAFs in HCC progression through the regulation of immune cells as well as mechanisms of evolvement of drug resistance.
Collapse
|
153
|
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer and is the second leading cause of cancer-related death worldwide. Fibrosis and cirrhosis are important risk factors for the development of HCC. Hepatic myofibroblasts are the cells responsible for extracellular matrix deposition, which is the hallmark of liver fibrosis. It is believed that myofibroblasts are predominantly derived from hepatic stellate cells (HSCs), also known as Ito cells. Nevertheless, depending on the nature of insult to the liver, it is thought that myofibroblasts may also originate from a variety of other cell types such as the portal fibroblasts (PFs), fibrocytes, hepatocytes, hepatic progenitor cells (HPCs), and mesothelial cells. Liver myofibroblasts are believed to transform into cancer-associated fibroblasts (CAFs) while HCC is developing. There is substantial evidence suggesting that activated HSCs (aHSCs)/cancer-associated fibroblasts (CAFs) may play an important role in HCC initiation and progression. In this paper, we aim to review current literature on cellular origins of myofibroblasts with a focus on hepatitis B virus (HBV)- and hepatitis C virus (HCV)-induced hepatic fibrosis. We also address the role of aHSCs/CAFs in HCC progression through the regulation of immune cells as well as mechanisms of evolvement of drug resistance.
Collapse
|
154
|
Damele L, Ottonello S, Mingari MC, Pietra G, Vitale C. Targeted Therapies: Friends or Foes for Patient's NK Cell-Mediated Tumor Immune-Surveillance? Cancers (Basel) 2020; 12:cancers12040774. [PMID: 32218226 PMCID: PMC7226262 DOI: 10.3390/cancers12040774] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/14/2020] [Accepted: 03/23/2020] [Indexed: 12/30/2022] Open
Abstract
In the last 20 years there has been a huge increase in the number of novel drugs for cancer treatment. Most of them exploit their ability to target specific oncogenic mutations in the tumors (targeted therapies–TT), while others target the immune-checkpoint inhibitor molecules (ICI) or the epigenetic DNA modifications. Among them, TT are the longest established drugs exploited against a wide spectrum of both solid and hematological tumors, often with reasonable costs and good efficacy as compared to other innovative therapies (i.e., ICI). Although they have greatly improved the treatment of cancer patients and their survival, patients often relapse or develop drug-resistance, leading to the impossibility to eradicate the disease. The outcome of TT has been often correlated with their ability to affect not only tumor cells, but also the repertoire of immune cells and their ability to interact with cancer cells. Thus, the possibility to create novel synergies among drugs an immunotherapy prompted scientists and physicians to deeply characterize the effects of TT on immune cells both by in-vitro and by ex-vivo analyses. In this context, NK cells may represent a key issue, since they have been shown to exert a potent anti-tumor activity, both against hematological malignancies and solid tumors. In the present review we will discuss most recent ex-vivo analyses that clarify the effect of TT treatment on patient’s NK cells comparing them with clinical outcome and previous in-vitro data.
Collapse
Affiliation(s)
- Laura Damele
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.D.); (S.O.); (M.C.M.); (G.P.)
| | - Selene Ottonello
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.D.); (S.O.); (M.C.M.); (G.P.)
| | - Maria Cristina Mingari
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.D.); (S.O.); (M.C.M.); (G.P.)
- Dipartimento Medicina Sperimentale (DIMES), Università degli Studi di Genova, 16132 Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), Università degli Studi di Genova, 16132 Genoa, Italy
| | - Gabriella Pietra
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.D.); (S.O.); (M.C.M.); (G.P.)
- Dipartimento Medicina Sperimentale (DIMES), Università degli Studi di Genova, 16132 Genoa, Italy
| | - Chiara Vitale
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.D.); (S.O.); (M.C.M.); (G.P.)
- Dipartimento Medicina Sperimentale (DIMES), Università degli Studi di Genova, 16132 Genoa, Italy
- Correspondence:
| |
Collapse
|
155
|
Piersma B, Hayward MK, Weaver VM. Fibrosis and cancer: A strained relationship. Biochim Biophys Acta Rev Cancer 2020; 1873:188356. [PMID: 32147542 DOI: 10.1016/j.bbcan.2020.188356] [Citation(s) in RCA: 394] [Impact Index Per Article: 78.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/03/2020] [Accepted: 03/03/2020] [Indexed: 12/18/2022]
Abstract
Tumors are characterized by extracellular matrix (ECM) deposition, remodeling, and cross-linking that drive fibrosis to stiffen the stroma and promote malignancy. The stiffened stroma enhances tumor cell growth, survival and migration and drives a mesenchymal transition. A stiff ECM also induces angiogenesis, hypoxia and compromises anti-tumor immunity. Not surprisingly, tumor aggression and poor patient prognosis correlate with degree of tissue fibrosis and level of stromal stiffness. In this review, we discuss the reciprocal interplay between tumor cells, cancer associated fibroblasts (CAF), immune cells and ECM stiffness in malignant transformation and cancer aggression. We discuss CAF heterogeneity and describe its impact on tumor development and aggression focusing on the role of CAFs in engineering the fibrotic tumor stroma and tuning tumor cell tension and modulating the immune response. To illustrate the role of mechanoreciprocity in tumor evolution we summarize data from breast cancer and pancreatic ductal carcinoma (PDAC) studies, and finish by discussing emerging anti-fibrotic strategies aimed at treating cancer.
Collapse
Affiliation(s)
- Bram Piersma
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco (UCSF), USA; Matrix research group, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, the Netherlands
| | - M K Hayward
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco (UCSF), USA
| | - Valerie M Weaver
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California, San Francisco (UCSF), USA; Departments of Radiation Oncology, Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research at UCSF, UCSF Helen Diller Comprehensive Cancer Center, 513 Parnassus Avenue, HSE565, San Francisco, CA 94143-0456, USA.
| |
Collapse
|
156
|
Ruan Q, Wang H, Burke LJ, Bridle KR, Li X, Zhao CX, Crawford DHG, Roberts MS, Liang X. Therapeutic modulators of hepatic stellate cells for hepatocellular carcinoma. Int J Cancer 2020; 147:1519-1527. [PMID: 32010970 DOI: 10.1002/ijc.32899] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/14/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary tumor in the liver and is a leading cause of cancer-related death worldwide. Activated hepatic stellate cells (HSCs) are key components of the HCC microenvironment and play an important role in the onset and progression of HCC through the secretion of growth factors and cytokines. Current treatment modalities that include chemotherapy, radiotherapy and ablation are able to activate HSCs and remodel the tumor microenvironment. Growing evidence has demonstrated that the complex interaction between activated HSCs and tumor cells can facilitate cancer chemoresistance and metastasis. Therefore, therapeutic targeting of activated HSCs has emerged as a promising strategy to improve treatment outcomes for HCC. This review summarizes the molecular mechanisms of HSC activation triggered by treatment modalities, the function of activated HSCs in HCC, as well as the crosstalk between tumor cells and activated HSCs. Pathways of activated HSC reduction are discussed, including inhibition, apoptosis, and reversion to the inactivated state. Finally, we outline the progress and challenges of therapeutic approaches targeting activated HSCs in the development of HCC treatment.
Collapse
Affiliation(s)
- Qi Ruan
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Haolu Wang
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia.,Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Leslie J Burke
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia.,Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Kim R Bridle
- Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Xinxing Li
- Department of General Surgery, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Chun-Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - Darrell H G Crawford
- Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Michael S Roberts
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Xiaowen Liang
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia.,Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
157
|
Fujita M, Yamaguchi R, Hasegawa T, Shimada S, Arihiro K, Hayashi S, Maejima K, Nakano K, Fujimoto A, Ono A, Aikata H, Ueno M, Hayami S, Tanaka H, Miyano S, Yamaue H, Chayama K, Kakimi K, Tanaka S, Imoto S, Nakagawa H. Classification of primary liver cancer with immunosuppression mechanisms and correlation with genomic alterations. EBioMedicine 2020; 53:102659. [PMID: 32113157 PMCID: PMC7048625 DOI: 10.1016/j.ebiom.2020.102659] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The tumor microenvironment can be classified into immunologically active "inflamed" tumors and inactive "non-inflamed" tumors based on the infiltration of cytotoxic immune cells. Previous studies on liver cancer have reported a superior prognosis for inflamed tumors compared to non-inflamed tumors. However, liver cancer is highly heterogeneous immunologically and genetically, and a finer classification of the liver cancer microenvironment may improve our understanding of its immunological diversity and response to immune therapy. METHODS We characterized the immune gene signatures of 234 primary liver cancers, mainly virus-related, from a Japanese population using RNA-Seq of tumors and matched non-tumorous hepatitis livers. We then compared them with the somatic alterations detected using the whole-genome sequencing. FINDINGS Liver cancers expressed lower levels of immune marker genes than non-tumorous hepatitis livers, indicating immunosuppression in the tumor microenvironment. Several immunosuppression mechanisms functioned actively and mutually exclusively, resulting in four immune subclasses of liver cancer: tumor-associated macrophage (TAM), CTNNB1, cytolytic activity (CYT), and regulatory T cell (Treg). The CYT and Treg subclasses represented inflamed tumors, while the TAM and CTNNB1 subclasses represented non-inflamed tumors. The TAM subclass, which comprised 31% of liver cancers, showed a poor survival, expressed elevated levels of extracellular matrix genes, and was associated with somatic mutations of chromatin regulator ARID2. The results of cell line experiments suggested a functional link between ARID2 and chemokine production by liver cancer cells. INTERPRETATION Primary liver cancer was classified into four subclasses based on mutually exclusive mechanisms for immunosuppression. This classification indicate the importance of immunosuppression mechanisms, such as TAM and Treg, as therapeutic targets for liver cancer. FUNDING The Japan Agency for Medical Research and Development (AMED).
Collapse
Affiliation(s)
- Masashi Fujita
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| | - Rui Yamaguchi
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Takanori Hasegawa
- Health Intelligence Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Shu Shimada
- Department of Molecular Oncology Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Koji Arihiro
- Department of Anatomical Pathology, Hiroshima University, Hiroshima, Japan.
| | - Shuto Hayashi
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Kazuhiro Maejima
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| | - Kaoru Nakano
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| | - Akihiro Fujimoto
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Atsushi Ono
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Hiroshi Aikata
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Masaki Ueno
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan.
| | - Shinya Hayami
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan.
| | - Hiroko Tanaka
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Satoru Miyano
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Health Intelligence Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Hiroki Yamaue
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan.
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Kazuhiro Kakimi
- Department of Immuno-therapeutics, The University of Tokyo Hospital, Japan; Cancer Immunology Data Multi-level Integration Unit, RIKEN Medical Innovation Hub Program, Tokyo, Japan.
| | - Shinji Tanaka
- Department of Molecular Oncology Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Seiya Imoto
- Health Intelligence Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Hidewaki Nakagawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| |
Collapse
|
158
|
Fujita M, Yamaguchi R, Hasegawa T, Shimada S, Arihiro K, Hayashi S, Maejima K, Nakano K, Fujimoto A, Ono A, Aikata H, Ueno M, Hayami S, Tanaka H, Miyano S, Yamaue H, Chayama K, Kakimi K, Tanaka S, Imoto S, Nakagawa H. Classification of primary liver cancer with immunosuppression mechanisms and correlation with genomic alterations. EBioMedicine 2020. [PMID: 32113157 DOI: 10.1016/j.ebiom.2020.102659.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The tumor microenvironment can be classified into immunologically active "inflamed" tumors and inactive "non-inflamed" tumors based on the infiltration of cytotoxic immune cells. Previous studies on liver cancer have reported a superior prognosis for inflamed tumors compared to non-inflamed tumors. However, liver cancer is highly heterogeneous immunologically and genetically, and a finer classification of the liver cancer microenvironment may improve our understanding of its immunological diversity and response to immune therapy. METHODS We characterized the immune gene signatures of 234 primary liver cancers, mainly virus-related, from a Japanese population using RNA-Seq of tumors and matched non-tumorous hepatitis livers. We then compared them with the somatic alterations detected using the whole-genome sequencing. FINDINGS Liver cancers expressed lower levels of immune marker genes than non-tumorous hepatitis livers, indicating immunosuppression in the tumor microenvironment. Several immunosuppression mechanisms functioned actively and mutually exclusively, resulting in four immune subclasses of liver cancer: tumor-associated macrophage (TAM), CTNNB1, cytolytic activity (CYT), and regulatory T cell (Treg). The CYT and Treg subclasses represented inflamed tumors, while the TAM and CTNNB1 subclasses represented non-inflamed tumors. The TAM subclass, which comprised 31% of liver cancers, showed a poor survival, expressed elevated levels of extracellular matrix genes, and was associated with somatic mutations of chromatin regulator ARID2. The results of cell line experiments suggested a functional link between ARID2 and chemokine production by liver cancer cells. INTERPRETATION Primary liver cancer was classified into four subclasses based on mutually exclusive mechanisms for immunosuppression. This classification indicate the importance of immunosuppression mechanisms, such as TAM and Treg, as therapeutic targets for liver cancer. FUNDING The Japan Agency for Medical Research and Development (AMED).
Collapse
Affiliation(s)
- Masashi Fujita
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| | - Rui Yamaguchi
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Takanori Hasegawa
- Health Intelligence Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Shu Shimada
- Department of Molecular Oncology Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Koji Arihiro
- Department of Anatomical Pathology, Hiroshima University, Hiroshima, Japan.
| | - Shuto Hayashi
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Kazuhiro Maejima
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| | - Kaoru Nakano
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| | - Akihiro Fujimoto
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Atsushi Ono
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Hiroshi Aikata
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Masaki Ueno
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan.
| | - Shinya Hayami
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan.
| | - Hiroko Tanaka
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Satoru Miyano
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Health Intelligence Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Hiroki Yamaue
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan.
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Kazuhiro Kakimi
- Department of Immuno-therapeutics, The University of Tokyo Hospital, Japan; Cancer Immunology Data Multi-level Integration Unit, RIKEN Medical Innovation Hub Program, Tokyo, Japan.
| | - Shinji Tanaka
- Department of Molecular Oncology Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Seiya Imoto
- Health Intelligence Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Hidewaki Nakagawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| |
Collapse
|
159
|
Zhang C, Hu Y, Shi C. Targeting Natural Killer Cells for Tumor Immunotherapy. Front Immunol 2020; 11:60. [PMID: 32140153 PMCID: PMC7042203 DOI: 10.3389/fimmu.2020.00060] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 01/10/2020] [Indexed: 12/20/2022] Open
Abstract
Natural killer (NK) cells are important innate cytotoxic lymphocytes with a rapid and efficient capacity to recognize and kill tumor cells. In recent years, adoptive transfer of autologous- or allogeneic-activated NK cells has become a promising cellular therapy for cancer. However, the therapeutic efficiency is encouraging in hematopoietic malignancies, but disappointing in solid tumors, for which the use of NK-cell-based therapies presents considerable challenges. It is difficult for NK cells to traffic to, and infiltrate into, tumor sites. NK cell function, phenotype, activation, and persistence are impaired by the tumor microenvironment, even leading to NK cell dysfunction or exhaustion. Many strategies focusing on improving NK cells' durable persistence, activation, and cytolytic activity, including activation with cytokines or analogs, have been attempted. Modifying them with chimeric antigen receptors further increases the targeting specificity of NK cells. Checkpoint blockades can relieve the exhausted state of NK cells. In this review, we discuss how the cytolytic and effector functions of NK cells are affected by the tumor microenvironment and summarize the various immunotherapeutic strategies based on NK cells. In particular, we discuss recent advances in overcoming the suppressive effect of the tumor microenvironment with the aim of enhancing the clinical outcome in solid tumors treated with NK-cell-based immunotherapy.
Collapse
Affiliation(s)
- Cai Zhang
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yuan Hu
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Chongdeng Shi
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
160
|
Gonçalves-Maia M, Gache Y, Basante M, Cosson E, Salavagione E, Muller M, Bernerd F, Avril MF, Schaub S, Sarasin A, Braud VM, Magnaldo T. NK Cell and Fibroblast-Mediated Regulation of Skin Squamous Cell Carcinoma Invasion by CLEC2A Is Compromised in Xeroderma Pigmentosum. J Invest Dermatol 2020; 140:1723-1732. [PMID: 32061658 DOI: 10.1016/j.jid.2020.01.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/12/2020] [Accepted: 01/21/2020] [Indexed: 12/24/2022]
Abstract
The ability of cancer cells to invade and disseminate can be affected by components of the surrounding microenvironment. To identify dermal components that regulate the growth of epidermal carcinomas, we studied the genetic disease called xeroderma pigmentosum that bears mutations in genes involved in the nucleotide excision repair of DNA. Patients with xeroderma pigmentosum are more prone to develop cutaneous tumors than the general population and their dermal fibroblasts display the features of dermal cancer-associated fibroblasts, which promote the invasion of keratinocytes. Here, we report that 3-dimensional dermal cultures of fibroblasts from healthy donors but not from patients with xeroderma pigmentosum complementation group C express CLEC2A, which is the ligand of the activating NK cell receptor NKp65. A similar loss of CLEC2A was observed in sporadic dermal cancer-associated fibroblasts and upon the culture of fibroblasts with cutaneous squamous cell carcinoma-conditioned medium. Using an innovative 3-dimensional organotypic skin culture model that contain NK cells in addition to fibroblasts and squamous cell carcinoma cells, we unveiled a key role of CLEC2A that orchestrates a crosstalk between fibroblasts and NK cells, thereby leading to the control of squamous cell carcinoma invasion. These findings indicate that CLEC2A-expressing dermal fibroblasts play a major role in immune surveillance of the skin.
Collapse
Affiliation(s)
- Maria Gonçalves-Maia
- Université Côte d'Azur, INSERM, CNRS, Institute for Research on Cancer and Aging, Nice, U1081, UMR7284, Nice, France
| | - Yannick Gache
- Université Côte d'Azur, INSERM, CNRS, Institute for Research on Cancer and Aging, Nice, U1081, UMR7284, Nice, France
| | - Miguel Basante
- Université Côte d'Azur, INSERM, CNRS, Institute for Research on Cancer and Aging, Nice, U1081, UMR7284, Nice, France; Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, France
| | - Estelle Cosson
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Valbonne, Sophia Antipolis, France
| | - Emie Salavagione
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Valbonne, Sophia Antipolis, France
| | - Margot Muller
- Université Côte d'Azur, INSERM, CNRS, Institute for Research on Cancer and Aging, Nice, U1081, UMR7284, Nice, France
| | | | - Marie Françoise Avril
- Assistance Publique-Hôpitaux de Paris, Department of Dermatology, Hospital Cochin, University Paris Descartes, Paris, France
| | - Sébastien Schaub
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, France
| | - Alain Sarasin
- Université Paris-Sud, Institut Gustave Roussy, UMR8200, CNRS, F-94805, Villejuif, France
| | - Véronique M Braud
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Valbonne, Sophia Antipolis, France
| | - Thierry Magnaldo
- Université Côte d'Azur, INSERM, CNRS, Institute for Research on Cancer and Aging, Nice, U1081, UMR7284, Nice, France.
| |
Collapse
|
161
|
Luo Q, Luo W, Zhu Q, Huang H, Peng H, Liu R, Xie M, Li S, Li M, Hu X, Zou Y. Tumor-Derived Soluble MICA Obstructs the NKG2D Pathway to Restrain NK Cytotoxicity. Aging Dis 2020; 11:118-128. [PMID: 32010486 PMCID: PMC6961768 DOI: 10.14336/ad.2019.1017] [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: 05/15/2019] [Accepted: 06/28/2019] [Indexed: 12/31/2022] Open
Abstract
The natural killer group 2D (NKG2D) receptor and its ligands play important roles in immune surveillance. In this study, we observed that the average serum soluble MICA (sMICA) concentration of 174 hepatocellular carcinoma (HCC) patients was significantly higher than that in 80 healthy subjects (602.17 ± 338.15 vs. 72.26 ± 87.88 pg/ml, t = 3.107, P=0.002). The levels of serum sMICA in 44 HCC patients with initial levels above 400 pg/ml declined significantly after surgical removal of the liver cancer tissue (P<0.001). Moreover, the mean survival time of HCC patients who had sMICA above 400 pg/ml was significantly shorter than that HCC patients with lower sMICA levels (P<0.001). Using the reporter cell line (NKG2D-2B4) in which activation of the NKG2D receptor pathway results in GFP expression based on the stimulation of immobilized rMICA, we showed that the number of GFP-expressing cells decreased sharply in presence of sMICA. Upon adding sMICA, the release of cytokines IFN-γ, TNF-α, and IL-8 by NK cell line (NKL) under stimulation of immobilized rMICA was blocked. Using MICA-expressing cells as the target cells, we observed that about 80% of target cells were killed by NKL at E:T of 10:1, but in presence of sMICAhigh serum of HCC patients, the dead target cells were reduced to 30.8%. Compared in presence of sMICAlow serum from HCC patients, there were 63.7% of target cells dead (p=0.043). Thus, our data suggested that sMICA obstructs the activation of NKG2D pathway to protect tumor cells from NK cell-mediated cytotoxicity.
Collapse
Affiliation(s)
- Qizhi Luo
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Weiguang Luo
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China.,2Department of Physiology, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Quan Zhu
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Hongjun Huang
- 3Cancer Hospital of Hunan, Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Huiyun Peng
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Rongjiao Liu
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Min Xie
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Shili Li
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Ming Li
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Xiaocui Hu
- 3Cancer Hospital of Hunan, Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Yizhou Zou
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| |
Collapse
|
162
|
Lee HW, Cho KJ, Park JY. Current Status and Future Direction of Immunotherapy in Hepatocellular Carcinoma: What Do the Data Suggest? Immune Netw 2020; 20:e11. [PMID: 32158599 PMCID: PMC7049588 DOI: 10.4110/in.2020.20.e11] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/07/2020] [Accepted: 02/08/2020] [Indexed: 02/07/2023] Open
Abstract
Most patients with hepatocellular carcinoma (HCC) are diagnosed at an advanced stage of disease. Until recently, systemic treatment options that showed survival benefits in HCC have been limited to tyrosine kinase inhibitors, antibodies targeting oncogenic signaling pathways or VEGF receptors. The HCC tumor microenvironment is characterized by a dysfunction of the immune system through multiple mechanisms, including accumulation of various immunosuppressive factors, recruitment of regulatory T cells and myeloid-derived suppressor cells, and induction of T cell exhaustion accompanied with the interaction between immune checkpoint ligands and receptors. Immune checkpoint inhibitors (ICIs) have been interfered this interaction and have altered therapeutic landscape of multiple cancer types including HCC. In this review, we discuss the use of anti-PD-1, anti-PD-L1, and anti-CTLA-4 antibodies in the treatment of advanced HCC. However, ICIs as a single agent do not benefit a significant portion of patients. Therefore, various clinical trials are exploring possible synergistic effects of combinations of different ICIs (anti-PD-1/PD-L1 and anti-CTLA-4 antibodies) or ICIs and target agents. Combinations of ICIs with locoregional therapies may also improve therapeutic responses.
Collapse
Affiliation(s)
- Hye Won Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
- Institue of Gastroenterology, Yonsei University College of Medicine, Seoul 03722, Korea
- Yonsei Liver Center, Severance Hospital, Seoul 03722, Korea
| | - Kyung Joo Cho
- Yonsei Liver Center, Severance Hospital, Seoul 03722, Korea
- BK21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jun Yong Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
- Institue of Gastroenterology, Yonsei University College of Medicine, Seoul 03722, Korea
- Yonsei Liver Center, Severance Hospital, Seoul 03722, Korea
- BK21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
163
|
Bald T, Pedde AM, Corvino D, Böttcher JP. The role of NK cell as central communicators in cancer immunity. Adv Immunol 2020; 147:61-88. [DOI: 10.1016/bs.ai.2020.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
164
|
The Gastrointestinal Tumor Microenvironment: An Updated Biological and Clinical Perspective. JOURNAL OF ONCOLOGY 2019; 2019:6240505. [PMID: 31885581 PMCID: PMC6893275 DOI: 10.1155/2019/6240505] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/30/2019] [Indexed: 12/24/2022]
Abstract
Gastrointestinal cancers are still responsible for high numbers of cancer-related deaths despite advances in therapy. Tumor-associated cells play a key role in tumor biology, by supporting or halting tumor development through the production of extracellular matrix, growth factors, cytokines, and extracellular vesicles. Here, we review the roles of these tumor-associated cells in the initiation, angiogenesis, immune modulation, and resistance to therapy of gastrointestinal cancers. We also discuss novel diagnostic and therapeutic strategies directed at tumor-associated cells and their potential benefits for the survival of these patients.
Collapse
|
165
|
Keenan BP, Fong L, Kelley RK. Immunotherapy in hepatocellular carcinoma: the complex interface between inflammation, fibrosis, and the immune response. J Immunother Cancer 2019; 7:267. [PMID: 31627733 PMCID: PMC6798343 DOI: 10.1186/s40425-019-0749-z] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer deaths worldwide and confers a poor prognosis. Beyond standard systemic therapy with multikinase inhibitors, recent studies demonstrate the potential for robust and durable responses from immune checkpoint inhibition in subsets of HCC patients across disease etiologies. The majority of HCC arises in the context of chronic inflammation and from within a fibrotic liver, with many cases associated with hepatitis virus infections, toxins, and fatty liver disease. Many patients also have concomitant cirrhosis which is associated with both local and systemic immune deficiency. Furthermore, the liver is an immunologic organ in itself, which may enhance or suppress the immune response to cancer arising within it. Here, we explore the immunobiology of the liver from its native state to chronic inflammation, fibrosis, cirrhosis and then to cancer, and summarize how this unique microenvironment may affect the response to immunotherapy.
Collapse
Affiliation(s)
- Bridget P Keenan
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, Room M1286, 505 Parnassus Ave., San Francisco, CA, 94143, USA. .,Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, Room M1286, 505 Parnassus Ave., San Francisco, CA, 94143, USA.,Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Robin K Kelley
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, Room M1286, 505 Parnassus Ave., San Francisco, CA, 94143, USA.,Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
166
|
Mortezaee K, Potes Y, Mirtavoos-Mahyari H, Motevaseli E, Shabeeb D, Musa AE, Najafi M, Farhood B. Boosting immune system against cancer by melatonin: A mechanistic viewpoint. Life Sci 2019; 238:116960. [PMID: 31629760 DOI: 10.1016/j.lfs.2019.116960] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/05/2019] [Accepted: 10/11/2019] [Indexed: 12/15/2022]
Abstract
Cancer is a disease of high complexity. Resistance to therapy is a major challenge in cancer targeted therapies. Overcoming this resistance requires a deep knowledge of the cellular interactions within tumor. Natural killer (NK) cells and cytotoxic T lymphocytes (CTLs) are the main anti-cancer immune cells, while T regulatory cells (Tregs) and cancer associated fibroblasts (CAFs) facilitate immune escape of cancer cells. Melatonin is a natural agent with anti-cancer functions that has also been suggested as an adjuvant in combination with cancer therapy modalities such as chemotherapy, radiotherapy, immunotherapy and tumor vaccination. One of the main effects of melatonin is regulation of immune responses against cancer cells. Melatonin has been shown to potentiate the activities of anti-cancer immune cells, as well as attenuating the activities of Tregs and CAFs. It also has a potent effect on the mitochondria, which may change immune responses against cancer. In this review, we explain the mechanisms of immune regulation by melatonin involved in its anti-cancer effects.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Yaiza Potes
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, C/ Julián Clavería 6, 33006, Oviedo, Spain
| | - Hanifeh Mirtavoos-Mahyari
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
167
|
Liu ML, Zang F, Zhang SJ. RBCK1 contributes to chemoresistance and stemness in colorectal cancer (CRC). Biomed Pharmacother 2019; 118:109250. [DOI: 10.1016/j.biopha.2019.109250] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023] Open
|
168
|
Terrén I, Orrantia A, Vitallé J, Zenarruzabeitia O, Borrego F. NK Cell Metabolism and Tumor Microenvironment. Front Immunol 2019; 10:2278. [PMID: 31616440 PMCID: PMC6769035 DOI: 10.3389/fimmu.2019.02278] [Citation(s) in RCA: 325] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/09/2019] [Indexed: 12/28/2022] Open
Abstract
Natural Killer (NK) cells are characterized by their potential to kill tumor cells by different means without previous sensitization and have, therefore, become a valuable tool in cancer immunotherapy. However, their efficacy against solid tumors is still poor and further studies are required to improve it. One of the major restrictions for NK cell activity is the immunosuppressive tumor microenvironment (TME). There, tumor and other immune cells create the appropriate conditions for tumor proliferation while, among others, preventing NK cell activation. Furthermore, NK cell metabolism is impaired in the TME, presumably due to nutrient and oxygen deprivation, and the higher concentration of tumor-derived metabolic end products, such as lactate. This metabolic restriction of NK cells limits their effector functions, and it could represent a potential target to focus on to improve the efficacy of NK cell-based therapies against solid tumors. In this review, we discuss the potential effect of TME into NK cell metabolism and its influence in NK cell effector functions.
Collapse
Affiliation(s)
- Iñigo Terrén
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Ane Orrantia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Joana Vitallé
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Olatz Zenarruzabeitia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Francisco Borrego
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
169
|
Yin Z, Dong C, Jiang K, Xu Z, Li R, Guo K, Shao S, Wang L. Heterogeneity of cancer-associated fibroblasts and roles in the progression, prognosis, and therapy of hepatocellular carcinoma. J Hematol Oncol 2019; 12:101. [PMID: 31547836 PMCID: PMC6757399 DOI: 10.1186/s13045-019-0782-x] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 08/23/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a lethal disease, and recurrence and metastasis are the major causes of death in HCC patients. Cancer-associated fibroblasts (CAFs), a major stromal cell type in the HCC microenvironment, promote HCC progression, and have gradually become a hot research topic in HCC-targeted therapy. This review comprehensively describes and discusses the heterogeneous tissue distribution, cellular origin, phenotype, and biological functions of HCC-associated fibroblasts. Furthermore, the possible use of CAFs for predicting HCC prognosis and in targeted therapeutic strategies is discussed, highlighting the critical roles of CAFs in HCC progression, diagnosis, and therapy.
Collapse
Affiliation(s)
- Zeli Yin
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China
- Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China
| | - Chengyong Dong
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China
- Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China
| | - Keqiu Jiang
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China
- Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China
| | - Zhe Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China
| | - Rui Li
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China
- Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China
| | - Kun Guo
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China.
- Department of Pathology, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China.
| | - Shujuan Shao
- Key Laboratory of Proteomics, Dalian Medical University, 9 West Lvshun South Road, Lvshunkou District, Dalian, 116027, Liaoning, China.
| | - Liming Wang
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China.
- Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China.
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China.
| |
Collapse
|
170
|
Fu Y, Liu S, Zeng S, Shen H. From bench to bed: the tumor immune microenvironment and current immunotherapeutic strategies for hepatocellular carcinoma. J Exp Clin Cancer Res 2019; 38:396. [PMID: 31500650 PMCID: PMC6734524 DOI: 10.1186/s13046-019-1396-4] [Citation(s) in RCA: 298] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/27/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) ranks the most common primary liver malignancy and the third leading cause of tumor-related mortality worldwide. Unfortunately, despite advances in HCC treatment, less than 40% of HCC patients are eligible for potentially curative therapies. Recently, cancer immunotherapy has emerged as one of the most promising approaches for cancer treatment. It has been proven therapeutically effective in many types of solid tumors, such as non-small cell lung cancer and melanoma. As an inflammation-associated tumor, it's well-evidenced that the immunosuppressive microenvironment of HCC can promote immune tolerance and evasion by various mechanisms. Triggering more vigorous HCC-specific immune response represents a novel strategy for its management. Pre-clinical and clinical investigations have revealed that various immunotherapies might extend current options for needed HCC treatment. In this review, we provide the recent progress on HCC immunology from both basic and clinical perspectives, and discuss potential advances and challenges of immunotherapy in HCC.
Collapse
MESH Headings
- Adaptive Immunity
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Biomarkers, Tumor
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Clinical Trials as Topic
- Combined Modality Therapy/methods
- Humans
- Immunity, Innate
- Immunotherapy/adverse effects
- Immunotherapy/methods
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Translational Research, Biomedical
- Treatment Outcome
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Yaojie Fu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Shanshan Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| |
Collapse
|
171
|
Cariani E, Missale G. Immune landscape of hepatocellular carcinoma microenvironment: Implications for prognosis and therapeutic applications. Liver Int 2019; 39:1608-1621. [PMID: 31314948 DOI: 10.1111/liv.14192] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 02/06/2023]
Abstract
The development of immunotherapy for solid tumours has boosted interest in the contexture of tumour immune microenvironment (TIME). Several lines of evidence indicate that the interplay between tumour cells and TIME components is a key factor for the evolution of hepatocellular carcinoma (HCC) and for the likelihood of response to immunotherapeutics. The availability of high-resolution methods will be instrumental for a better definition of the complexity and diversity of TIME with the aim of predicting disease outcome, treatment response and possibly new therapeutic targets. Here, we review current knowledge about the immunological mechanisms involved in shaping the clinical course of HCC. Effector cells, regulatory cells and soluble mediators are discussed for their role defining TIME and as targets for immune modulation, together with possible immune signatures for optimization of immunotherapeutic strategies.
Collapse
Affiliation(s)
- Elisabetta Cariani
- Toxicology and Advanced Diagnostics, Ospedale S. Agostino-Estense, Modena, Italy
| | - Gabriele Missale
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
172
|
Lu C, Rong D, Zhang B, Zheng W, Wang X, Chen Z, Tang W. Current perspectives on the immunosuppressive tumor microenvironment in hepatocellular carcinoma: challenges and opportunities. Mol Cancer 2019; 18:130. [PMID: 31464625 PMCID: PMC6714090 DOI: 10.1186/s12943-019-1047-6] [Citation(s) in RCA: 298] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Incidence of hepatocellular carcinoma (HCC) is on the rise due to the prevalence of chronic hepatitis and cirrhosis. Although there are surgical and chemotherapy treatment avenues the mortality rate of HCC remains high. Immunotherapy is currently the new frontier of cancer treatment and the immunobiology of HCC is emerging as an area for further exploration. The tumor microenvironment coexists and interacts with various immune cells to sustain the growth of HCC. Thus, immunosuppressive cells play an important role in the anti-tumor immune response. This review will discuss the current concepts of immunosuppressive cells, including tumor-associated macrophages, marrow-derived suppressor cells, tumor-associated neutrophils, cancer-associated fibroblasts, and regulatory T cell interactions to actively promote tumorigenesis. It further elaborates on current treatment modalities and future areas of exploration.
Collapse
Affiliation(s)
- Chen Lu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dawei Rong
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
| | - Betty Zhang
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Wubin Zheng
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuehao Wang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China. .,Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.
| | - Ziyi Chen
- Department of General Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, China.
| | - Weiwei Tang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
173
|
Piñeiro Fernández J, Luddy KA, Harmon C, O'Farrelly C. Hepatic Tumor Microenvironments and Effects on NK Cell Phenotype and Function. Int J Mol Sci 2019; 20:E4131. [PMID: 31450598 PMCID: PMC6747260 DOI: 10.3390/ijms20174131] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/16/2019] [Accepted: 08/20/2019] [Indexed: 02/07/2023] Open
Abstract
The liver is a complex organ with critical physiological functions including metabolism, glucose storage, and drug detoxification. Its unique immune profile with large numbers of cytotoxic CD8+ T cells and significant innate lymphoid population, including natural killer cells, γ δ T cells, MAIT cells, and iNKTcells, suggests an important anti-tumor surveillance role. Despite significant immune surveillance in the liver, in particular large NK cell populations, hepatic cell carcinoma (HCC) is a relatively common outcome of chronic liver infection or inflammation. The liver is also the second most common site of metastatic disease. This discordance suggests immune suppression by the environments of primary and secondary liver cancers. Classic tumor microenvironments (TME) are poorly perfused, leading to accumulation of tumor cell metabolites, diminished O2, and decreased nutrient levels, all of which impact immune cell phenotype and function. Here, we focus on changes in the liver microenvironment associated with tumor presence and how they affect NK function and phenotype.
Collapse
Affiliation(s)
| | - Kimberly A Luddy
- School of Biochemistry and Immunology, Trinity College Dublin, D02 PN40 Dublin, Ireland.
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center, Tampa, FL 33626, USA.
| | - Cathal Harmon
- Brigham and Women's Hospital, Harvard Institutes of Medicine, Harvard Medical School, Boston, MA 02138, USA
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity College Dublin, D02 PN40 Dublin, Ireland.
- School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland.
| |
Collapse
|
174
|
Shembrey C, Huntington ND, Hollande F. Impact of Tumor and Immunological Heterogeneity on the Anti-Cancer Immune Response. Cancers (Basel) 2019; 11:E1217. [PMID: 31438563 PMCID: PMC6770225 DOI: 10.3390/cancers11091217] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 12/16/2022] Open
Abstract
Metastatic tumors are the primary cause of cancer-related mortality. In recent years, interest in the immunologic control of malignancy has helped establish escape from immunosurveillance as a critical requirement for incipient metastases. Our improved understanding of the immune system's interactions with cancer cells has led to major therapeutic advances but has also unraveled a previously unsuspected level of complexity. This review will discuss the vast spatial and functional heterogeneity in the tumor-infiltrating immune system, with particular focus on natural killer (NK) cells, as well as the impact of tumor cell-specific factors, such as secretome composition, receptor-ligand repertoire, and neoantigen diversity, which can further drive immunological heterogeneity. We emphasize how tumor and immunological heterogeneity may undermine the efficacy of T-cell directed immunotherapies and explore the potential of NK cells to be harnessed to circumvent these limitations.
Collapse
Affiliation(s)
- Carolyn Shembrey
- Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Melbourne, VIC 3000, Australia
- Centre for Cancer Research, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Nicholas D Huntington
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Frédéric Hollande
- Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Melbourne, VIC 3000, Australia.
- Centre for Cancer Research, The University of Melbourne, Melbourne, VIC 3000, Australia.
| |
Collapse
|
175
|
Monteran L, Erez N. The Dark Side of Fibroblasts: Cancer-Associated Fibroblasts as Mediators of Immunosuppression in the Tumor Microenvironment. Front Immunol 2019; 10:1835. [PMID: 31428105 PMCID: PMC6688105 DOI: 10.3389/fimmu.2019.01835] [Citation(s) in RCA: 475] [Impact Index Per Article: 79.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are prominent components of the microenvironment in most types of solid tumors, and were shown to facilitate cancer progression by supporting tumor cell growth, extracellular matrix remodeling, promoting angiogenesis, and by mediating tumor-promoting inflammation. In addition to an inflammatory microenvironment, tumors are characterized by immune evasion and an immunosuppressive milieu. In recent years, CAFs are emerging as central players in immune regulation that shapes the tumor microenvironment. CAFs contribute to immune escape of tumors via multiple mechanisms, including secretion of multiple cytokines and chemokines and reciprocal interactions that mediate the recruitment and functional differentiation of innate and adaptive immune cells. Moreover, CAFs directly abrogate the function of cytotoxic lymphocytes, thus inhibiting killing of tumor cells. In this review, we focus on recent advancements in our understanding of how CAFs drive the recruitment and functional fate of tumor-infiltrating immune cells toward an immunosuppressive microenvironment, and provide outlook on future therapeutic implications that may lead to integration of preclinical findings into the design of novel combination strategies, aimed at impairing the tumor-supportive function of CAFs.
Collapse
Affiliation(s)
- Lea Monteran
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
176
|
Gieniec KA, Butler LM, Worthley DL, Woods SL. Cancer-associated fibroblasts-heroes or villains? Br J Cancer 2019; 121:293-302. [PMID: 31289350 PMCID: PMC6738083 DOI: 10.1038/s41416-019-0509-3] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 05/19/2019] [Accepted: 05/22/2019] [Indexed: 01/05/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) were originally presumed to represent a homogeneous population uniformly driving tumorigenesis, united by their morphology and peritumoural location. Our understanding of CAFs has since been shaped by sophisticated in vitro and in vivo experiments, pathological association and, more recently, ablation, and it is now widely appreciated that CAFs form a group of highly heterogeneous cells with no single overarching marker. Studies have demonstrated that the CAF population contains different subtypes based on the expression of marker proteins with the capacity to promote or inhibit cancer, with their biological role as accomplices or adversaries dependent on many factors, including the cancer stage. So, while CAFs have been endlessly shown to promote the growth, survival and spread of tumours via improvements in functionality and an altered secretome, they are also capable of retarding tumorigenesis via largely unknown mechanisms. It is important to reconcile these disparate results so that the functions of, or factors produced by, tumour-promoting subtypes can be specifically targeted to improve cancer patient outcomes. This review will dissect out CAF complexity and CAF-directed cancer treatment strategies in order to provide a case for future, rational therapies.
Collapse
Affiliation(s)
- Krystyna A Gieniec
- School of Medicine, University of Adelaide, Adelaide, SA, Australia.,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Lisa M Butler
- School of Medicine, University of Adelaide, Adelaide, SA, Australia.,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Daniel L Worthley
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Susan L Woods
- School of Medicine, University of Adelaide, Adelaide, SA, Australia. .,Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| |
Collapse
|
177
|
Boesch M, Baty F, Rumpold H, Sopper S, Wolf D, Brutsche MH. Fibroblasts in cancer: Defining target structures for therapeutic intervention. Biochim Biophys Acta Rev Cancer 2019; 1872:111-121. [PMID: 31265878 DOI: 10.1016/j.bbcan.2019.06.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/25/2019] [Accepted: 06/27/2019] [Indexed: 12/11/2022]
Abstract
The functional importance of the tumor stroma for cancer growth and progression is increasingly recognized, but has not resulted in notable therapeutic developments yet. Within the mesenchymal tumor microenvironment, cancer-associated fibroblasts take the center stage and fuel tumor progression in various ways including malignant cell potentiation, immune regulation and fibrosis. However, recent studies have demonstrated pronounced heterogeneity of the fibroblastic tumor stroma, which comprises a plethora of individual cell subsets with varying phenotypes and functions, some of which suppress malignant growth through immune engagement or crosstalk with the tumor vasculature. This article summarizes the various levels at which the fibroblastic tumor stroma may impact cancer progression and highlights potential target structures for future therapeutic intervention(s).
Collapse
Affiliation(s)
- Maximilian Boesch
- Lung Center, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, 9007 St. Gallen, Switzerland.
| | - Florent Baty
- Lung Center, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, 9007 St. Gallen, Switzerland
| | - Holger Rumpold
- Internal Medicine II (Medical Oncology, Hematology, Gastroenterology & Rheumatology), Academic Teaching Hospital Feldkirch, Carinagasse 47, 6807 Feldkirch, Austria
| | - Sieghart Sopper
- Internal Medicine V (Hematology & Oncology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020 Innsbruck, Austria
| | - Dominik Wolf
- Internal Medicine V (Hematology & Oncology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; Medical Clinic 3, Oncology, Hematology, Immunoncology and Rheumatology, University Clinic Bonn (UKB), Sigmund-Freud-Strasse 25, 53127 Bonn, Germany
| | - Martin H Brutsche
- Lung Center, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, 9007 St. Gallen, Switzerland
| |
Collapse
|
178
|
Zhao J, Schlößer HA, Wang Z, Qin J, Li J, Popp F, Popp MC, Alakus H, Chon SH, Hansen HP, Neiss WF, Jauch KW, Bruns CJ, Zhao Y. Tumor-Derived Extracellular Vesicles Inhibit Natural Killer Cell Function in Pancreatic Cancer. Cancers (Basel) 2019; 11:874. [PMID: 31234517 PMCID: PMC6628179 DOI: 10.3390/cancers11060874] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies. Tumor-derived extracellular vesicles (EVs) induce pre-metastatic niche formation to promote metastasis. We isolated EVs from a highly-metastatic pancreatic cancer cell line and patient-derived primary cancer cells by ultracentrifugation. The protein content of EVs was analyzed by mass spectrometry. The effects of PDAC-derived EVs on natural kill (NK) cells were investigated by flow cytometry. The serum EVs' TGF-β1 levels were quantified by ELISA. We found that integrins were enriched in PDAC-derived EVs. The expression of NKG2D, CD107a, TNF-α, and INF-γ in NK cells was significantly downregulated after co-culture with EVs. NK cells also exhibited decreased levels of CD71 and CD98, as well as impaired glucose uptake ability. In addition, NK cell cytotoxicity against pancreatic cancer stem cells was attenuated. Moreover, PDAC-derived EVs induced the phosphorylation of Smad2/3 in NK cells. Serum EVs' TGF-β1 was significantly increased in PDAC patients. Our findings emphasize the immunosuppressive role of PDAC-derived EVs and provide new insights into our understanding of NK cell dysfunction regarding pre-metastatic niche formation in PDAC.
Collapse
Affiliation(s)
- Jiangang Zhao
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany.
- Department of General, Visceral und Vascular Surgery, Ludwig-Maximilian-University (LMU), 81377 Munich, Germany.
| | - Hans A Schlößer
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany.
| | - Zhefang Wang
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany.
| | - Jie Qin
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany.
| | - Jiahui Li
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany.
| | - Felix Popp
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany.
| | - Marie Christine Popp
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany.
| | - Hakan Alakus
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany.
| | - Seung-Hun Chon
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany.
| | - Hinrich P Hansen
- Department I of Internal Medicine, University Hospital of Cologne, Center for Integrated Oncology Cologne-Bonn, CECAD Center of Excellence on "Cellular Stress Responses in Aging-Associated Diseases", Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany.
| | - Wolfram F Neiss
- Department of Anatomy I, Medical Faculty, University of Cologne, 50937 Cologne, Germany.
| | - Karl-Walter Jauch
- Department of General, Visceral und Vascular Surgery, Ludwig-Maximilian-University (LMU), 81377 Munich, Germany.
| | - Christiane J Bruns
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany.
| | - Yue Zhao
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany.
- Department of General, Visceral und Vascular Surgery, Otto von Guericke University, 39120 Magdeburg, Germany.
| |
Collapse
|
179
|
Juengpanich S, Shi L, Iranmanesh Y, Chen J, Cheng Z, Khoo AKJ, Pan L, Wang Y, Cai X. The role of natural killer cells in hepatocellular carcinoma development and treatment: A narrative review. Transl Oncol 2019; 12:1092-1107. [PMID: 31176993 PMCID: PMC6558093 DOI: 10.1016/j.tranon.2019.04.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/11/2019] [Accepted: 04/11/2019] [Indexed: 12/12/2022] Open
Abstract
A major obstacle for treatment of HCC is the inadequate efficacy and limitation of the available therapeutic options. Despite the recent advances in developing novel treatment options, HCC still remains one of the major causes of cancer morbidity and mortality around the world. Achieving effective treatment and eradication of HCC is a challenging task, however recent studies have shown that targeting Natural Killer cells, as major regulators of immune system, can help with the complete treatment of HCC, restoration of normal liver function and subsequently higher survival rate of HCC patients. Studies have shown that decrease in the frequency of NK cells, their dysfunction due to several factors such as dysregulation of receptors and their ligands, and imbalance of different types of inhibitory and stimulating microRNA expression is associated with higher rate of HCC progression and development, and poor survival outcome. Here in our review, we mainly focused on the importance of NK cells in HCC development and treatment.
Collapse
Affiliation(s)
- Sarun Juengpanich
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, Province, China; School of Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Liang Shi
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, Province, China.
| | | | - Jiang Chen
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, Province, China; Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Zhenzhe Cheng
- School of Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Aaron Kah-Jin Khoo
- Faculty of Medicine, The University of Queensland, St Lucia, QLD, 4027, Australia.
| | - Long Pan
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, Province, China; School of Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Yifan Wang
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, Province, China.
| | - Xiujun Cai
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, Province, China.
| |
Collapse
|
180
|
Elwakeel E, Brüne B, Weigert A. PGE 2 in fibrosis and cancer: Insights into fibroblast activation. Prostaglandins Other Lipid Mediat 2019; 143:106339. [PMID: 31100473 DOI: 10.1016/j.prostaglandins.2019.106339] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/18/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023]
Abstract
Fibroblasts are the essential cellular architects of connective tissue and as such are crucial cells in contributing to organ homeostasis. While fulfilling important repair functions during tissue regeneration upon wounding, chronic fibroblast activation provokes pathological organ fibrosis and promotes neoplastic disease progression. Identifying targets that may serve to therapeutically terminate fibroblast activation is therefore desirable. Among the mediators that may be relevant in this context is the prostanoid prostaglandin E2 (PGE2) that is produced during inflammatory settings, where pathological fibrosis occurs. Here, we summarize current, in part controversial, concepts on the impact of PGE2 on fibroblast activation in fibrotic diseases including cancer, and discuss these findings in the context of the evolving concept of fibroblast heterogeneity.
Collapse
Affiliation(s)
- Eiman Elwakeel
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany.
| |
Collapse
|
181
|
Gorchs L, Fernández Moro C, Bankhead P, Kern KP, Sadeak I, Meng Q, Rangelova E, Kaipe H. Human Pancreatic Carcinoma-Associated Fibroblasts Promote Expression of Co-inhibitory Markers on CD4 + and CD8 + T-Cells. Front Immunol 2019. [PMID: 31068935 DOI: 10.3389/fimmu.2019.00847.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Carcinoma-associated pancreatic fibroblasts (CAFs) are the major type of cells in the stroma of pancreatic ductal adenocarcinomas and besides their pathological release of extracellular matrix proteins, they are also perceived as key contributors to immune evasion. Despite the known relevance of tumor infiltrating lymphocytes in cancers, the interactions between T-cells and CAFs remain largely unexplored. Here, we found that CAFs isolated from tumors of pancreatic cancer patients undergoing surgical resection (n = 15) expressed higher levels of the PD-1 ligands PD-L1 and PD-L2 compared to primary skin fibroblasts from healthy donors. CAFs strongly inhibited T-cell proliferation in a contact-independent fashion. Blocking the activity of prostaglandin E2 (PGE2) by indomethacin partially restored the proliferative capacity of both CD4+ and CD8+ T-cells. After stimulation, the proportion of proliferating T-cells expressing HLA-DR and the proportion of memory T-cells were decreased when CAFs were present compared to T-cells proliferating in the absence of CAFs. Interestingly, CAFs promoted the expression of TIM-3, PD-1, CTLA-4 and LAG-3 in proliferating T-cells. Immunohistochemistry stainings further showed that T-cells residing within the desmoplastic stromal compartment express PD-1, indicating a role for CAFs on co-inhibitory marker expression also in vivo. We further found that PGE2 promoted the expression of PD-1 and TIM-3 on T-cells. Functional assays showed that proliferating T-cells expressing immune checkpoints produced less IFN-γ, TNF-α, and CD107a after restimulation when CAFs had been present. Thus, this indicates that CAFs induce expression of immune checkpoints on CD4+ and CD8+ T-cells, which contribute to a diminished immune function.
Collapse
Affiliation(s)
- Laia Gorchs
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Carlos Fernández Moro
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Pathology/Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Bankhead
- Division of Pathology, Centre for Genomic & Experimental Medicine, Western General Hospital, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Katharina P Kern
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Imrul Sadeak
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Qingda Meng
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elena Rangelova
- Department of CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Pancreatic Surgery Unit, Center for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Helen Kaipe
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
182
|
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: 16] [Impact Index Per Article: 2.7] [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
|
183
|
Fibroblasts as Modulators of Local and Systemic Cancer Metabolism. Cancers (Basel) 2019; 11:cancers11050619. [PMID: 31058816 PMCID: PMC6562905 DOI: 10.3390/cancers11050619] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 01/05/2023] Open
Abstract
Fibroblast activation is an accompanying feature of solid tumor progression, resembling a conserved host response to tissue damage. Cancer-associated fibroblasts (CAFs) comprise a heterogeneous and plastic population with increasingly appreciated roles in tumor growth, metastatic capacity, and response to therapy. Classical features of fibroblasts in a wound-healing response, including profound extracellular matrix production and cytokine release, are recapitulated in cancer. Emerging evidence suggests that fibroblastic cells in the microenvironments of solid tumors also critically modulate cellular metabolism in the neoplastic compartment through mechanisms including paracrine transfer of metabolites or non-cell-autonomous regulation of metabolic signaling pathways. These metabolic functions may represent common mechanisms by which fibroblasts stimulate growth of the regenerating epithelium during a wound-healing reaction, or may reflect unique co-evolution of cancer cells and surrounding stroma within the tumor microenvironment. Here we review the recent literature supporting an important role for CAFs in regulation of cancer cell metabolism, and relevant pathways that may serve as targets for therapeutic intervention.
Collapse
|
184
|
Guo Q, Huang F, Goncalves C, Del Rincón SV, Miller WH. Translation of cancer immunotherapy from the bench to the bedside. Adv Cancer Res 2019; 143:1-62. [PMID: 31202357 DOI: 10.1016/bs.acr.2019.03.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The tremendous success of immune checkpoint blockades has revolutionized cancer management. Our increased understanding of the cell types that compose the tumor microenvironment (TME), including those of the innate and adaptive immune system, has helped to shape additional immune modulatory strategies in cancer care. Pre-clinical and clinical investigations targeting novel checkpoint interactions and key pathways that regulate cancer immunity continue to increase rapidly. Various combinatorial drug regimens are being tested in attempt to achieve durable response and survival rates of patients with cancer. This review provides an overview of specific components of the TME, an introduction to novel immune checkpoints, followed by a survey of present day and future combination immune modulatory therapies. The idea that the immune system can recognize and destroy tumor cells was first described in the cancer immunosurveillance hypothesis of Burnet and Thomas. However, early experimental evidence failed to support the concept. It was not until the late 1990s when seminal papers clearly showed the existence of cancer immunosurveillance, leading to the cancer immunoediting hypothesis. In this century, progress in the understanding of negative regulators of the immune response led to the discovery that inhibition of these regulators in patients with cancer could lead to dramatic and durable remissions. Drs. Tasuku Honjo and James P. Allison were awarded the Nobel Prize in 2018 for their pioneering work in this field. We now see rapid advances in cancer immunology and emerging effective therapies revolutionizing cancer care across tumor types in the clinic, while pre-clinical research is moving from a focus on the malignant cells themselves to dissect the highly heterogenic and complex multi-cellular tumor microenvironment (TME).
Collapse
Affiliation(s)
- Qianyu Guo
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Fan Huang
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Christophe Goncalves
- Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Sonia V Del Rincón
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Wilson H Miller
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada; Rossy Cancer Network, Montreal, QC, Canada.
| |
Collapse
|
185
|
Mikulak J, Bruni E, Oriolo F, Di Vito C, Mavilio D. Hepatic Natural Killer Cells: Organ-Specific Sentinels of Liver Immune Homeostasis and Physiopathology. Front Immunol 2019; 10:946. [PMID: 31114585 PMCID: PMC6502999 DOI: 10.3389/fimmu.2019.00946] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/12/2019] [Indexed: 12/16/2022] Open
Abstract
The liver is considered a preferential tissue for NK cells residency. In humans, almost 50% of all intrahepatic lymphocytes are NK cells that are strongly imprinted in a liver-specific manner and show a broad spectrum of cellular heterogeneity. Hepatic NK (he-NK) cells play key roles in tuning liver immune response in both physiological and pathological conditions. Therefore, there is a pressing need to comprehensively characterize human he-NK cells to better understand the related mechanisms regulating their effector-functions within the dynamic balance between immune-tolerance and immune-surveillance. This is of particular relevance in the liver that is the only solid organ whose parenchyma is constantly challenged on daily basis by millions of foreign antigens drained from the gut. Therefore, the present review summarizes our current knowledge on he-NK cells in the light of the latest discoveries in the field of NK cell biology and clinical relevance.
Collapse
Affiliation(s)
- Joanna Mikulak
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Elena Bruni
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Ferdinando Oriolo
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Clara Di Vito
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Milan, Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
186
|
Gorchs L, Fernández Moro C, Bankhead P, Kern KP, Sadeak I, Meng Q, Rangelova E, Kaipe H. Human Pancreatic Carcinoma-Associated Fibroblasts Promote Expression of Co-inhibitory Markers on CD4 + and CD8 + T-Cells. Front Immunol 2019; 10:847. [PMID: 31068935 PMCID: PMC6491453 DOI: 10.3389/fimmu.2019.00847] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/01/2019] [Indexed: 12/20/2022] Open
Abstract
Carcinoma-associated pancreatic fibroblasts (CAFs) are the major type of cells in the stroma of pancreatic ductal adenocarcinomas and besides their pathological release of extracellular matrix proteins, they are also perceived as key contributors to immune evasion. Despite the known relevance of tumor infiltrating lymphocytes in cancers, the interactions between T-cells and CAFs remain largely unexplored. Here, we found that CAFs isolated from tumors of pancreatic cancer patients undergoing surgical resection (n = 15) expressed higher levels of the PD-1 ligands PD-L1 and PD-L2 compared to primary skin fibroblasts from healthy donors. CAFs strongly inhibited T-cell proliferation in a contact-independent fashion. Blocking the activity of prostaglandin E2 (PGE2) by indomethacin partially restored the proliferative capacity of both CD4+ and CD8+ T-cells. After stimulation, the proportion of proliferating T-cells expressing HLA-DR and the proportion of memory T-cells were decreased when CAFs were present compared to T-cells proliferating in the absence of CAFs. Interestingly, CAFs promoted the expression of TIM-3, PD-1, CTLA-4 and LAG-3 in proliferating T-cells. Immunohistochemistry stainings further showed that T-cells residing within the desmoplastic stromal compartment express PD-1, indicating a role for CAFs on co-inhibitory marker expression also in vivo. We further found that PGE2 promoted the expression of PD-1 and TIM-3 on T-cells. Functional assays showed that proliferating T-cells expressing immune checkpoints produced less IFN-γ, TNF-α, and CD107a after restimulation when CAFs had been present. Thus, this indicates that CAFs induce expression of immune checkpoints on CD4+ and CD8+ T-cells, which contribute to a diminished immune function.
Collapse
Affiliation(s)
- Laia Gorchs
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Carlos Fernández Moro
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Pathology/Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Bankhead
- Division of Pathology, Centre for Genomic & Experimental Medicine, Western General Hospital, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Katharina P. Kern
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Imrul Sadeak
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Qingda Meng
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elena Rangelova
- Department of CLINTEC, Karolinska Institutet, Stockholm, Sweden
- Pancreatic Surgery Unit, Center for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Helen Kaipe
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
187
|
Targeting natural killer cells in solid tumors. Cell Mol Immunol 2019; 16:415-422. [PMID: 30911118 DOI: 10.1038/s41423-019-0224-2] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 02/27/2019] [Indexed: 02/07/2023] Open
Abstract
Natural killer (NK) cells are innate lymphoid cells endowed with cytolytic activity and a capacity to secrete cytokines and chemokines. Several lines of evidence suggest that NK cells play an important role in anti-tumor immunity. Some therapies against hematological malignacies make use of the immune properties of NK cells, such as their ability to kill residual leukemic blasts efficiently after conditioning during haploidentical hematopoietic stem cell transplantation. However, knowledge on NK cell infiltration and the status of NK cell responsiveness in solid tumors is limited so far. The pro-angiogenic role of the recently described NK cell-like type 1 innate lymphoid cells (ILC1s) and their phenotypic resemblance to NK cells are confounding factors that add a level of complexity, at least in mice. Here, we review the current knowledge on the presence and function of NK cells in solid tumors as well as the immunotherapeutic approaches designed to harness NK cell functions in these conditions, including those that aim to reinforce conventional anti-tumor therapies to increase the chances of successful treatment.
Collapse
|
188
|
Suppression of Natural Killer cell NKG2D and CD226 anti-tumour cascades by platelet cloaked cancer cells: Implications for the metastatic cascade. PLoS One 2019; 14:e0211538. [PMID: 30908480 PMCID: PMC6433214 DOI: 10.1371/journal.pone.0211538] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/16/2019] [Indexed: 11/19/2022] Open
Abstract
Tumour cell immune evasion is a principal hallmark of successful metastasis. Tumour cells in the vasculature adopt a platelet cloak that efficiently suppresses the innate immune system by directly inhibiting Natural Killer (NK) cells, which normally function to neutralise spreading cancers. Here we describe two novel mechanisms of tumour cell evasion of NK cell anti-tumour functions. The first, an ‘immune decoy’ mechanism in which platelets induce the release of soluble NKG2D ligands from the tumour cell to mask detection and actively suppress NK cell degranulation and inflammatory cytokine (IFNγ) production, concomitantly. This represents a double-hit to immune clearance of malignant cells during metastasis. The second mechanism, a platelet-derived TGFβ-mediated suppression of the CD226/CD96-CD112/CD155 axis, is a novel pathway with poorly understood anti-cancer functions. We have demonstrated that platelets robustly suppress surface expression of CD226 and CD96 on the NK cell surface and their associated ligands on the tumour cell to further enhance NK cell suppression. These highly evolved mechanisms promote successful tumour immune evasion during metastasis and provide a unique opportunity for studying the complexity of cellular interactions in the metastatic cascade and thus novel targets for cancer immunotherapy.
Collapse
|
189
|
Wang Y, Yao R, Zhang L, Xie X, Chen R, Ren Z. IDO and intra-tumoral neutrophils were independent prognostic factors for overall survival for hepatocellular carcinoma. J Clin Lab Anal 2019; 33:e22872. [PMID: 30843276 PMCID: PMC6595287 DOI: 10.1002/jcla.22872] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 02/01/2019] [Accepted: 02/10/2019] [Indexed: 12/24/2022] Open
Abstract
Background Both Indole‐amine‐2,3‐dioxygenase (IDO) and neutrophils were proved to have pro‐tumor effect in some kinds of solid tumors by immune suppression. However, little is known about their effect on hepatocellular carcinoma (HCC) and the relationship between these two immune‐suppressive factors. The aim of this study was to evaluate the prognostic significance of IDO and intra‐tumoral neutrophils and their correlations in HCC. Methods Specimens’ tissue microarray (TMA) for 153 HCC patients was used in this study. We examined intra‐tumoral expression of IDO and CD66b in TMA. The Kaplan‐Meier method and Cox regression models were used to evaluate the prognostic value of IDO expression and CD66b. Results Multivariate analysis showed both IDO expression and intra‐tumoral neutrophils infiltration were independent prognostic factors for overall survival (OS). In high IDO expression group, the percentage of intra‐tumoral neutrophils infiltration was higher than that in low IDO expression group. Conclusion Both IDO and intra‐tumoral neutrophils were independent prognostic factors for overall survival for HCC.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, China.,Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rongrong Yao
- Department of Oncology, Huashan Hospital, Fudan University, Shanghai, China
| | - Lan Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, China.,Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoying Xie
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, China.,Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rongxin Chen
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, China.,Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenggang Ren
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, China.,Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
190
|
Sung PS, Jang JW. Natural Killer Cell Dysfunction in Hepatocellular Carcinoma: Pathogenesis and Clinical Implications. Int J Mol Sci 2018; 19:3648. [PMID: 30463262 PMCID: PMC6274919 DOI: 10.3390/ijms19113648] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is currently the third leading cause of malignancy-related mortalities worldwide. Natural killer (NK) cells are involved in the critical role of first line immunological defense against cancer development. Defects in NK cell functions are recognized as important mechanisms for immune evasion of tumor cells. NK cell function appears to be attenuated in HCC, and many previous reports suggested that NK cells play a critical role in controlling HCC, suggesting that boosting the activity of dysfunctional NK cells can enhance tumor cell killing. However, the detailed mechanisms of NK cell dysfunction in tumor microenvironment of HCC remain largely unknown. A better understanding of the mechanisms of NK cell dysfunction in HCC will help in the NK cell-mediated eradication of cancer cells and prolong patient survival. In this review, we describe the various mechanisms underlying human NK cell dysfunction in HCC. Further, we summarize current advances in the approaches to enhance endogenous NK cell function and in adoptive NK cell therapies, to cure this difficult-to-treat cancer.
Collapse
Affiliation(s)
- Pil Soo Sung
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- The Catholic Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Jeong Won Jang
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- The Catholic Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| |
Collapse
|
191
|
Foster DS, Jones RE, Ransom RC, Longaker MT, Norton JA. The evolving relationship of wound healing and tumor stroma. JCI Insight 2018; 3:99911. [PMID: 30232274 DOI: 10.1172/jci.insight.99911] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The stroma in solid tumors contains a variety of cellular phenotypes and signaling pathways associated with wound healing, leading to the concept that a tumor behaves as a wound that does not heal. Similarities between tumors and healing wounds include fibroblast recruitment and activation, extracellular matrix (ECM) component deposition, infiltration of immune cells, neovascularization, and cellular lineage plasticity. However, unlike a wound that heals, the edges of a tumor are constantly expanding. Cell migration occurs both inward and outward as the tumor proliferates and invades adjacent tissues, often disregarding organ boundaries. The focus of our review is cancer associated fibroblast (CAF) cellular heterogeneity and plasticity and the acellular matrix components that accompany these cells. We explore how similarities and differences between healing wounds and tumor stroma continue to evolve as research progresses, shedding light on possible therapeutic targets that can result in innovative stromal-based treatments for cancer.
Collapse
Affiliation(s)
- Deshka S Foster
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, and.,Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - R Ellen Jones
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, and
| | - Ryan C Ransom
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, and
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, and.,Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Jeffrey A Norton
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, and.,Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
192
|
McEachron TA, Triche TJ, Sorenson L, Parham DM, Carpten JD. Profiling targetable immune checkpoints in osteosarcoma. Oncoimmunology 2018; 7:e1475873. [PMID: 30524885 PMCID: PMC6279416 DOI: 10.1080/2162402x.2018.1475873] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/04/2018] [Accepted: 05/07/2018] [Indexed: 12/26/2022] Open
Abstract
Osteosarcomas are aggressive bone tumors for which therapeutic advances have not improved over several decades. Unlike most pediatric tumors, the osteosarcoma genome is remarkably unstable, characterized by numerous copy number alterations and chromosomal structural aberrations. In this study, we asked if the targetable immune checkpoints CD274 (PD-L1), PDCD1LG2 (PD-L2), CD276 (B7-H3) and IDO1 are impacted by copy number alterations in osteosarcoma. Of the 215 osteosarcoma samples investigated, PD-L1/PD-L2, B7-H3 and IDO1 were independently gained at frequencies of approximately 8-9%, with a cumulative frequency of approximately 24%. RNA sequencing data from two independent cohorts revealed that B7-H3 is the most highly expressed immune checkpoint gene among the four investigated. We also show that IDO1 is preferentially expressed in pediatric solid tumors and that increased protein expression of B7-H3 and IDO1 are significantly associated with inferior survival in patient samples. Using human osteosarcoma cell lines, we demonstrate that IDO1 is gained in MG63 and G292 cells and that the IDO1 inhibitor, epacadostat, inhibits the enzymatic activity of IDO1 in a dose-dependent manner in these cells. Together, these data reveal the genomic and transcriptomic profiles of PD-L1, PD-L2, B7-H3 and IDO1 in osteosarcoma and identifies a potential context for targeted immunotherapeutic intervention in a subset of patients.
Collapse
Affiliation(s)
- Troy A McEachron
- Department of Translational Genomics
- Norris Comprehensive Cancer Center
- Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Timothy J Triche
- Norris Comprehensive Cancer Center
- Department of Pathology, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | | | - David M Parham
- Department of Pathology, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - John D Carpten
- Department of Translational Genomics
- Norris Comprehensive Cancer Center
| |
Collapse
|
193
|
Hosseinzadeh F, Verdi J, Ai J, Hajighasemlou S, Seyhoun I, Parvizpour F, Hosseinzadeh F, Iranikhah A, Shirian S. Combinational immune-cell therapy of natural killer cells and sorafenib for advanced hepatocellular carcinoma: a review. Cancer Cell Int 2018; 18:133. [PMID: 30214375 PMCID: PMC6131874 DOI: 10.1186/s12935-018-0624-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 08/24/2018] [Indexed: 02/06/2023] Open
Abstract
Background High prevalence of hepatocellular carcinoma (HCC) and typically poor prognosis of this disease that lead to late stage diagnosis when potentially curative therapies are least effective; therefore, development of an effective and systematic treatment is an urgent requirement. Main body In this review, several current treatments for HCC patients and their advantages or disadvantages were summarized. Moreover, various recent preclinical and clinical studies about the performances of "two efficient agents, sorafenib or natural killer (NK) cells", against HCC cells were investigated. In addition, the focus this review was on the chemo-immunotherapy approach, correlation between sorafenib and NK cells and their effects on the performance of each other for better suppression of HCC. Conclusion It was concluded that combinational therapy with sorafenib and NK cells might improve the outcome of applied therapeutic approaches for HCC patients. Finally, it was also concluded that interaction between sorafenib and NK cells is dose and time dependent, therefore, a careful dose and time optimizing is necessary for development of a combinational immune-cell therapy.
Collapse
Affiliation(s)
- Faezeh Hosseinzadeh
- 1Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Javad Verdi
- 1Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- 1Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saieh Hajighasemlou
- 1Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Iran Food and Drug Administration, Tehran, Iran
| | - Iman Seyhoun
- 1Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Frzad Parvizpour
- 1Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Abolfazl Iranikhah
- 4Department of Gastroenterology, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Sadegh Shirian
- 5Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.,6Shiraz Molecular Pathology Research Center, Dr. Daneshbod Lab, Shiraz, Iran
| |
Collapse
|
194
|
Fabregat I, Caballero-Díaz D. Transforming Growth Factor-β-Induced Cell Plasticity in Liver Fibrosis and Hepatocarcinogenesis. Front Oncol 2018; 8:357. [PMID: 30250825 PMCID: PMC6139328 DOI: 10.3389/fonc.2018.00357] [Citation(s) in RCA: 241] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/13/2018] [Indexed: 12/11/2022] Open
Abstract
The Transforming Growth Factor-beta (TGF-β) family plays relevant roles in the regulation of different cellular processes that are essential for tissue and organ homeostasis. In the case of the liver, TGF-β signaling participates in different stages of disease progression, from initial liver injury toward fibrosis, cirrhosis and cancer. When a chronic injury takes place, mobilization of lymphocytes and other inflammatory cells occur, thus setting the stage for persistence of an inflammatory response. Macrophages produce profibrotic mediators, among them, TGF-β, which is responsible for activation -transdifferentiation- of quiescent hepatic stellate cells (HSC) to a myofibroblast (MFB) phenotype. MFBs are the principal source of extracellular matrix protein (ECM) accumulation and prominent mediators of fibrogenesis. TGF-β also mediates an epithelial-mesenchymal transition (EMT) process in hepatocytes that may contribute, directly or indirectly, to increase the MFB population. In hepatocarcinogenesis, TGF-β plays a dual role, behaving as a suppressor factor at early stages, but contributing to later tumor progression, once cells escape from its cytostatic effects. As part of its potential pro-tumorigenic actions, TGF-β induces EMT in liver tumor cells, which increases its pro-migratory and invasive potential. In parallel, TGF-β also induces changes in tumor cell plasticity, conferring properties of a migratory tumor initiating cell (TIC). The main aim of this review is to shed light about the pleiotropic actions of TGF-β that explain its effects on the different liver cell populations. The cross-talk with other signaling pathways that contribute to TGF-β effects, in particular the Epidermal Growth Factor Receptor (EGFR), will be presented. Finally, we will discuss the rationale for targeting the TGF-β pathway in liver pathologies.
Collapse
Affiliation(s)
- Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute, Barcelona, Spain.,Department of Physiological Sciences, School of Medicine, University of Barcelona, Barcelona, Spain.,Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona, Spain
| | - Daniel Caballero-Díaz
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute, Barcelona, Spain.,Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona, Spain
| |
Collapse
|
195
|
Natural Killer Cells in Liver Disease and Hepatocellular Carcinoma and the NK Cell-Based Immunotherapy. J Immunol Res 2018; 2018:1206737. [PMID: 30255103 PMCID: PMC6142725 DOI: 10.1155/2018/1206737] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/16/2018] [Indexed: 02/07/2023] Open
Abstract
Nature killer (NK) cells play a critical role in host innate and adaptive immune defense against viral infections and tumors. NK cells are enriched in liver hematopoietic cells with unique NK repertories and functions to safeguard liver cells against hepatitis virus infection or malignancy transformation. However, accumulating evidences were found that the NK cells were modulated by liver diseases and liver cancers including hepatocellular carcinoma (HCC) and showed impaired functions failing to activate the elimination of the viral-infected cells or tumor cells and were further involved in the pathogenesis of liver injury and inflammation. The full characterization of circulation and intrahepatic NK cell phenotype and function in liver disease and liver cancer has not only provided new insight into the disease pathogenesis but has also discovered new targets for developing new NK cell-based therapeutic strategies. This review will discuss and summarize the NK cell phenotypic and functional changes in liver disease and HCC, and the NK cell-based immunotherapy approaches and progresses for cancers including HCC will also be reviewed.
Collapse
|
196
|
Nishida N, Kudo M. Immune checkpoint blockade for the treatment of human hepatocellular carcinoma. Hepatol Res 2018; 48:622-634. [PMID: 29734514 DOI: 10.1111/hepr.13191] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/30/2018] [Accepted: 05/01/2018] [Indexed: 12/23/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers with a high recurrence rate. Currently, tyrosine kinase inhibitors (TKIs) are the first-line treatment for cases refractory to conventional therapies. However, the acquisition of somatic mutations can result in TKI resistance. Clinical evidence suggests that acquired immunity contributes to the suppression of tumor recurrence, indicating the potential of induced antitumor immune reaction for the treatment of HCC. Recently, immune checkpoint inhibitors have become available for the treatment of malignancies. They are effective regardless of the response to prior therapies and a durable effect can be expected, which should be attributed to an adaptive immunity to HCC components. The results of phase I/II trials of nivolumab, an anti-programmed cell death-1 antibody, showed that 20% of patients showed objective response and that nivolumab was effective regardless of prior sorafenib treatment and viral status. Nivolumab received expedited Food and Drug Administration approval in 2017 for the treatment of advanced HCC after failure or intolerance to sorafenib. However, the majority of the patients remain refractory, likely due to the solid immune suppressive status, which involves many stromal cells, humoral mediators, and suppressive checkpoint molecules. Therefore, current clinical trials are focusing on how immunosuppressive conditions in HCC might be overcome using immune checkpoint inhibitors in combination with different types of immune checkpoint blockades, TKIs, and other conventional treatments. The development of immune checkpoint inhibitors is rapidly progressing and these inhibitors are likely to be key agents for HCC treatment in the near feature.
Collapse
Affiliation(s)
- Naoshi Nishida
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| |
Collapse
|
197
|
Dai K, Huang Y, Chen Z, Sun X, Yang L, Jiang Y. Kbtbd2 inhibits the cytotoxic activity of immortalized NK cells through down-regulating mTOR signaling in a mouse hepatocellular carcinoma model. Eur J Immunol 2018; 48:683-695. [PMID: 29331106 DOI: 10.1002/eji.201747281] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/05/2017] [Accepted: 01/04/2018] [Indexed: 12/18/2022]
Abstract
Natural killer cell (NK cell)-based immunotherapy is a promising therapeutic strategy for hepatocellular carcinoma (HCC). However, the molecular mechanisms underlying the regulation of NK cell function in the tumor sites are not completely elucidated. In this study, we identified the enhanced expression of kelch repeat and BTB (POZ) domain containing 2 (Kbtbd2) in intratumoral NK cells in a mouse HCC implantation model as a negative regulator of NK cells. To investigate this interaction, we used a Tet-on inducible expression system to control Kbtbd2 expression in an immortalized mouse NK cell line KIL C.2. With this approach, we found that overexpression of Kbtbd2 reduced KIL C.2 cell proliferation, decreased expression certain of Ly49 receptor family members, and substantially impaired cytotoxic activity of KIL C.2 cells in vitro. Moreover, phosphorylation of mTOR and its target 4E-binding protein 1 was reduced in Kbtbd2-expressing KIL C.2 cells, along with down-regulated phosphorylation of Erk1/2. Adoptively transferred Kbtbd2-expressing KIL C.2 cells exhibited weaker tumoricidal effect on hepatocellular carcinoma cells in the HCC implantation model, in comparison with transferred control KIL C.2 cells. Taken together, our investigation indicates that Kbtbd2 is an inhibitory molecule for the tumoricidal activity of KIL C.2 cells and perhaps intratumoral NK cells.
Collapse
Affiliation(s)
- Kai Dai
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yabing Huang
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zubing Chen
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaomei Sun
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lihua Yang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yingan Jiang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
198
|
Petrizzo A, Mauriello A, Tornesello ML, Buonaguro FM, Tagliamonte M, Buonaguro L. Cellular prognostic markers in hepatitis-related hepatocellular carcinoma. Infect Agent Cancer 2018; 13:10. [PMID: 29599818 PMCID: PMC5870199 DOI: 10.1186/s13027-018-0183-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/15/2018] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and accounts for about 6% of all new cancers diagnosed worldwide. Moreover, it is the third and the fifth leading cause of death from cancer in men and women, respectively. HBV and HCV chronic infection is the main risk factor for HCC. A range of therapies are used in the management of HCC according to the extent and severity of liver disease. In this perspective, evaluation of prognosis represents a crucial step for proper management of HCC patients. However, the clinical outcome can be significantly different in HCC patients within the same stage of disease. Therefore, many efforts have been made to define new parameters with more precise prognostic value, and the search for HCC prognostic markers is gaining momentum. The present review aims at providing an update on cellular prognostic markers for HCC.
Collapse
Affiliation(s)
- A. Petrizzo
- Laboratory of Cancer Immunoregulation, Department of Experimental Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” – IRCCS, Via Mariano Semmola, 1, 80131 Naples, Italy
| | - A. Mauriello
- Laboratory of Cancer Immunoregulation, Department of Experimental Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” – IRCCS, Via Mariano Semmola, 1, 80131 Naples, Italy
| | - M. L. Tornesello
- Lab Molecular Biology and Viral Oncology, Department of Experimental Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” – IRCCS, Via Mariano Semmola, 1, 80131 Naples, Italy
| | - F. M. Buonaguro
- Lab Molecular Biology and Viral Oncology, Department of Experimental Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” – IRCCS, Via Mariano Semmola, 1, 80131 Naples, Italy
| | - M. Tagliamonte
- Laboratory of Cancer Immunoregulation, Department of Experimental Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” – IRCCS, Via Mariano Semmola, 1, 80131 Naples, Italy
| | - L. Buonaguro
- Laboratory of Cancer Immunoregulation, Department of Experimental Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” – IRCCS, Via Mariano Semmola, 1, 80131 Naples, Italy
- Lab Molecular Biology and Viral Oncology, Department of Experimental Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” – IRCCS, Via Mariano Semmola, 1, 80131 Naples, Italy
| |
Collapse
|
199
|
Zekri ARN, El Deeb S, Bahnassy AA, Badr AM, Abdellateif MS, Esmat G, Salama H, Mohanad M, El-dien AE, Rabah S, Abd Elkader A. Role of relevant immune-modulators and cytokines in hepatocellular carcinoma and premalignant hepatic lesions. World J Gastroenterol 2018; 24:1228-1238. [PMID: 29568203 PMCID: PMC5859225 DOI: 10.3748/wjg.v24.i11.1228] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 12/24/2017] [Accepted: 01/16/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To assess the levels of different immune modulators in patients with hepatocellular carcinoma (HCC), in relation to other hepatic diseases.
METHODS Eighty-eight patients were included in the current study and represented patients with HCC (20), liver cirrhosis (28) and chronic hepatitis (CH; 25), and normal controls (NC; 15). Peripheral blood was isolated for immunophenotyping of active myeloid dendritic cells (mDCs; CD1c and CD40), mature inactive myeloid cells (CD1c and HLA), active plasmacytoid cells (pDCs; CD303 and CD40), mature inactive pDCs (CD30 and HLA), active natural killer (NK) cells (CD56 and CD161), active NK cells (CD56 and CD314) and inactive NK cells (CD56 and CD158) was done by flow cytometry. Serum levels of interleukin (IL)-2, IL-10, IL-12, IL-1β, interferon (IFN)-α, IFN-γ and tumor necrosis factor (TNF)-αR2 were assessed by ELISA.
RESULTS Active mDCs (CD1C+/CD40+) and inactive mDCs (CD1c+/HLA+) were significantly decreased in HCC patients in relation to NC (P < 0.001). CD40+ expression on active pDCs was decreased in HCC patients (P < 0.001), and its level was not significantly changed among other groups. Inactive pDCs (CD303+/HLA+), inactive NKs (CD56+/CD158+) and active NKs (CD56+/CD161+) were not statistically changed among the four groups studied; however, the latter was increased in CH (P < 0.05). NKG2D was statistically decreased in HCC, CH and cirrhosis (P < 0.001), and it was not expressed in 63% (12/20) of HCC patients. There was significant decrease of IL-2, IFN-α and IFN-γ (P < 0.001), and a significant increase in IL-10, IL-1β, and TNF-αR2 (P <0.01, P < 0.001 and P < 0.001; respectively) in HCC patients. There was inverted correlation between IL-12 and IL-1β in HCC (r = -0.565, P < 0.01), with a strong correlation between pDCs (CD303+/CD40+) and NKs (CD56+/CD161+; r = 0.512, P < 0.05) as well as inactive mDCs (CD1c+/HLA+) and inactive NK cells (CD56+/CD158+; r = 0.945, P < 0.001).
CONCLUSION NKG2D, CD40, IL-2 and IL-10 are important modulators in the development and progression of HCC.
Collapse
Affiliation(s)
- Abdel-Rahman N Zekri
- Molecular Virology and Immunology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo 11976, Egypt
| | - Somaya El Deeb
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Abeer A Bahnassy
- Department of Pathology, National Cancer Institute, Cairo University, Cairo 11976, Egypt
| | - Abeer M Badr
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo 11976, Egypt
| | - Gamal Esmat
- Department of Hepatology and Tropical Medicine, Faculty of Medicine, Cairo University, Cairo 11441, Egypt
| | - Hosny Salama
- Department of Hepatology and Tropical Medicine, Faculty of Medicine, Cairo University, Cairo 11441, Egypt
| | - Marwa Mohanad
- Department of Biochemistry, Misr University for Science and Technology, 6th October 12945, Giza Governorate, Egypt
| | - Ahmed Esam El-dien
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Shimaa Rabah
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Assmaa Abd Elkader
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| |
Collapse
|
200
|
Ziani L, Chouaib S, Thiery J. Alteration of the Antitumor Immune Response by Cancer-Associated Fibroblasts. Front Immunol 2018; 9:414. [PMID: 29545811 PMCID: PMC5837994 DOI: 10.3389/fimmu.2018.00414] [Citation(s) in RCA: 274] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/14/2018] [Indexed: 12/12/2022] Open
Abstract
Among cells present in the tumor microenvironment, activated fibroblasts termed cancer-associated fibroblasts (CAFs), play a critical role in the complex process of tumor-stroma interaction. CAFs, one of the prominent stromal cell populations in most types of human carcinomas, have been involved in tumor growth, angiogenesis, cancer stemness, extracellular matrix remodeling, tissue invasion, metastasis, and even chemoresistance. During the past decade, these activated tumor-associated fibroblasts have also been involved in the modulation of the anti-tumor immune response on various levels. In this review, we describe our current understanding of how CAFs accomplish this task as well as their potential therapeutic implications.
Collapse
Affiliation(s)
- Linda Ziani
- INSERM, UMR 1186, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Faculty of Medicine, University Paris Sud, Le Kremlin Bicêtre, France
| | - Salem Chouaib
- INSERM, UMR 1186, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Faculty of Medicine, University Paris Sud, Le Kremlin Bicêtre, France
| | - Jerome Thiery
- INSERM, UMR 1186, Villejuif, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Faculty of Medicine, University Paris Sud, Le Kremlin Bicêtre, France
| |
Collapse
|