51
|
Li LM, Liu ZX, Cheng QY. Exosome plays an important role in the development of hepatocellular carcinoma. Pathol Res Pract 2019; 215:152468. [PMID: 31171380 DOI: 10.1016/j.prp.2019.152468] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/19/2019] [Accepted: 05/21/2019] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most malignant cancers around the world. However, the early biomarkers for its detection and treatment are limited currently. Exosomes, classified as intercellular messenger shuttling their cargoes between cells, regulate cell differentiation and tissue development. They contain messenger RNA (mRNA), microRNA (miRNA), long non-coding RNA (lncRNA), circular RNA (circRNA), proteins, lipids and transcription factors. Therefore, exosomes play a crucial role in the development of HCC. In this review, we highlight the exosomal cargoes which could serve as biomarkers for the prediction and diagnosis of HCC. Exosomes are involved in metastases of HCC and they show great potential in immunotherapy and drug resistance mechanism. In summary, exosome suggests new clues in clinical application of HCC.
Collapse
Affiliation(s)
- Li-Man Li
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhen-Xian Liu
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qing-Yuan Cheng
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
52
|
Seifi-Alan M, Shamsi R, Ghafouri-Fard S. Application of cancer-testis antigens in immunotherapy of hepatocellular carcinoma. Immunotherapy 2019; 10:411-421. [PMID: 29473472 DOI: 10.2217/imt-2017-0154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a worldwide common malignancy with poor prognosis. Several studies have aimed at identification of appropriate biomarkers for early detection of this cancer. Cancer-testis antigens (CTAs) as a novel group of tumor-associated antigens have been demonstrated to be expressed in HCC samples as well as peripheral blood samples from these patients but not in the corresponding adjacent noncancerous samples. Such pattern of expression has provided them an opportunity to be used as immunotherapeutic targets. The detection of spontaneous immune responses against CTAs in HCC patients has prompted design of CTA-based immunotherapeutic protocols in these patients. The results of some clinical trials have been promising in a subset of patients.
Collapse
Affiliation(s)
- Mahnaz Seifi-Alan
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roshanak Shamsi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
53
|
Huang KW, Jayant K, Lee PH, Yang PC, Hsiao CY, Habib N, Sodergren MH. Positive Immuno-Modulation Following Radiofrequency Assisted Liver Resection in Hepatocellular Carcinoma. J Clin Med 2019; 8:385. [PMID: 30893948 PMCID: PMC6463076 DOI: 10.3390/jcm8030385] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/12/2019] [Accepted: 03/14/2019] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) often develops on a background of chronic inflammation and a complex immunosuppressive network with increased regulatory T cells, impaired CD8⁺ T cells and the secretion of immunosuppressive cytokines. Previous clinical studies have reported a superior disease-free survival (DFS) following a radiofrequency-based ablation or resection in HCC tumours compared to conventional liver resection techniques. The aim of this study was to investigate whether there is any correlation with the use of a radiofrequency-assisted liver resection and clinical outcome. MATERIAL AND METHODS Patients' peripheral blood was collected prior and 7 days following surgery from patients undergoing a liver resection for HCC. There were 5 liver resections performed using CUSA and 6 liver resections with the RF-based device, HabibTM 4X. The primary endpoint of the study was to assess the immunological parameters of circulating immune cell populations as well as serum cytokines. The Student's t-test, chi-square or Fisher's Exact test were applied for statistical comparisons, as appropriate. RESULTS Patients undergoing an RF-assisted liver resection with HabibTM 4X had a significant decrease in the inhibitory Treg cells (p = 0.002) and a significant increase in CD8⁺ T lymphocytes (p = 0.050) and CD4⁺CD45RO⁺/CD4⁺ memory T cells (p = 0.002) compared to those patients undergoing a liver resection with CUSA. It was also noted that the RF-assisted liver resection group had a significant decrease in circulating TGF-ß (p = 0.000), IL10 (p = 0.000) and a significant increase in IFN-gamma (p = 0. 027) and IL-17 compared to the CUSA group. CONCLUSION A liver resection with RF-based device HabibTM 4X was associated with positive immunomodulatory changes in circulating immune cells and circulating cytokines which could explain the significant improvement in DFS.
Collapse
Affiliation(s)
- Kai Wen Huang
- Department of Surgery & Hepatitis Research Center, National Taiwan University Hospital, Taipei 10051, Taiwan.
- Centre of Mini-invasive Interventional Oncology, National Taiwan University Hospital, Taipei 10051, Taiwan.
- Department of Surgery and Cancer, Imperial College London, London, SW7 2AZ, UK.
| | - Kumar Jayant
- Department of Surgery and Cancer, Imperial College London, London, SW7 2AZ, UK.
- Department of Renal Transplant Surgery, Royal Free Hospital, London NW3 2QG, UK.
- Warwick Medical School, University of Warwick, Coventry, CV4 7HL, UK.
| | - Po-Huang Lee
- Department of Surgery & Hepatitis Research Center, National Taiwan University Hospital, Taipei 10051, Taiwan.
- Centre of Mini-invasive Interventional Oncology, National Taiwan University Hospital, Taipei 10051, Taiwan.
| | - Po-Chih Yang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
- Center for Organ Transplantation and Liver Disease Treatment, Fu Jen Catholic University Hospital, New Taipei City 24352, Taiwan.
- School of Medicine, Fu Jen Catholic University, New Taipei City 24352, Taiwan.
| | - Chih-Yang Hsiao
- Department of Surgery & Hepatitis Research Center, National Taiwan University Hospital, Taipei 10051, Taiwan.
- Centre of Mini-invasive Interventional Oncology, National Taiwan University Hospital, Taipei 10051, Taiwan.
| | - Nagy Habib
- Department of Surgery and Cancer, Imperial College London, London, SW7 2AZ, UK.
| | - Mikael H Sodergren
- Department of Surgery and Cancer, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
54
|
Abstract
Exosomes are a class of extracellular vesicles released by multiple cells types including tumor cells, with a size range of 30-100 nm and a lipid bilayer membrane. Recently, the role of exosomes in cell-to-cell communication has been extensively studied, showed that exosomes can deliver their functional RNAs and proteins to recipient cells, impacting transcription and translation of recipient cells. Emerging evidence suggests that hepatocellular carcinoma (HCC) cell-derived exosomes can construct a fertile environment to support HCC cells proliferation, grow, invasion and metastasis, development of drug resistance. Circulating exosomes can be used as noninvasive biomarkers for early diagnosis, moreover as drug delivery vehicles, provide new insights into the treatment of HCC.
Collapse
|
55
|
Yang Y, Qin Z, Du D, Wu Y, Qiu S, Mu F, Xu K, Chen J. Safety and Short-Term Efficacy of Irreversible Electroporation and Allogenic Natural Killer Cell Immunotherapy Combination in the Treatment of Patients with Unresectable Primary Liver Cancer. Cardiovasc Intervent Radiol 2019; 42:48-59. [PMID: 30151798 PMCID: PMC6267679 DOI: 10.1007/s00270-018-2069-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/21/2018] [Indexed: 01/10/2023]
Abstract
PURPOSE This study aimed to investigate the safety and short-term efficacy of irreversible electroporation (IRE) combined with allogenic natural killer (NK) cell immunotherapy in the treatment of patients with unresectable primary liver cancer. MATERIALS AND METHODS Between October 2015 and December 2016, 40 patients were enrolled and randomly allocated to either the IRE group (n = 22) or the IRE-NK group (n = 18). All adverse events experienced by the patients were recorded; the changes in tumor biomarkers [AFP, CA 19-9, circulating tumor cells (CTCs)], lymphocyte number and function, quality of life, clinical response, progression-free survival (PFS) and overall survival (OS) were assessed. RESULTS Patients who received combination therapy exhibited significantly longer median PFS and OS than who just received IRE (PFS 15.1 vs. 10.6 months, P < 0.05, OS 17.9 vs. 23.2 months, P < 0.05). The combination therapy of IRE and NK cell immunotherapy significantly reduced CTCs and increased immune function and Karnofsky performance status. CONCLUSION Our data suggest a novel, promising combination therapy using IRE and allogenic NK cell immunotherapy. Larger clinical trials are required to confirm these conclusions.
Collapse
Affiliation(s)
- Yumei Yang
- Department of Interventional Therapy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, No. 3002 of SunGang West Road, FuTian, Shenzhen, 518035, China
| | - Zilin Qin
- Chongqing Health Service Center, Chongqing, 400020, China
| | - Duanming Du
- Department of Interventional Therapy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, No. 3002 of SunGang West Road, FuTian, Shenzhen, 518035, China.
| | - Yumin Wu
- Department of Interventional Therapy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, No. 3002 of SunGang West Road, FuTian, Shenzhen, 518035, China
| | - Shuibo Qiu
- Department of Interventional Therapy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, No. 3002 of SunGang West Road, FuTian, Shenzhen, 518035, China
| | - Feng Mu
- Department of Oncology, Fuda Cancer Hospital of Jinan University, Guangzhou, China
| | | | - Jibing Chen
- Biotherapy Center, Fuda Cancer Hospital of Jinan University, Guangzhou, 510665, China.
- Fuda Cancer Institute, Guangzhou, China.
| |
Collapse
|
56
|
Azim HA, Omar A, Atef H, Zawahry H, Shaker MK, Abdelmaksoud AK, EzzElarab M, Abdel-Rahman O, Ismail M, Kassem L, Waked I. Sorafenib plus tegafur-uracil (UFT) versus sorafenib as first line systemic treatment for patients with advanced stage HCC: a Phase II trial (ESLC01 study). J Hepatocell Carcinoma 2018; 5:109-119. [PMID: 30510922 PMCID: PMC6250115 DOI: 10.2147/jhc.s169285] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Phase II trials found that tegafur–uracil (UFT) is an effective drug in hepatocellular carcinoma (HCC), while preclinical data suggested that its combination with sorafenib may have a promising activity. Our Phase II randomized trial aimed to evaluate efficacy and tolerability of sorafenib plus UFT vs sorafenib in advanced HCC. Methods Patients with advanced HCC, with no prior systemic therapy, were randomized to receive either UFT at 125 mg/m2 twice daily for 4 out of 5 weeks plus sorafenib at 400 mg twice daily (arm 1) or single agent sorafenib at 400 mg twice daily (arm 2). Primary end point was time to progression (TTP). Results Between March 2012 and March 2014, 76 eligible patients – out of 143 preplanned – were randomized. The study was terminated early because of futility. This is the final analysis of the study, after a median follow-up of 10.2 months and death of 86% of randomized patients (n=64). Median TTP was 7.5 months and 8.2 months in arms 1 and 2 respectively (HR: 1.07; 95% CI, 0.52–2.22; P=0.855), while the median overall survival was 8.2 months and 10.5 months respectively (HR: 1.58; 95% CI: 0.90–2.76, P=0.112). Nine patients (25%) in the combination arm discontinued treatment because of toxicity vs eight patients (21.1%) in the sorafenib monotherapy arm (P=0.899). Conclusion In patients with advanced HCC, adding UFT to sorafenib is feasible, but it did not improve efficacy outcome over sorafenib monotherapy.
Collapse
Affiliation(s)
- Hamdy A Azim
- Department of Clinical Oncology, Faculty of Medicine, Cairo University, Cairo, Egypt, ,
| | - Ashraf Omar
- Department of Gastroenterology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hesham Atef
- Department of Clinical Oncology, Faculty of Medicine, Cairo University, Cairo, Egypt, ,
| | - Heba Zawahry
- Department of Medical Oncology, National Cancer Institute, Cairo, Egypt
| | - Mohamed K Shaker
- Tropical Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Mohamed EzzElarab
- National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed Ismail
- Clinical Oncology Department, Cairo Oncology Center, Cairo, Egypt
| | - Loay Kassem
- Department of Clinical Oncology, Faculty of Medicine, Cairo University, Cairo, Egypt, ,
| | - Imam Waked
- Institute of Liver Disease, Menoufiya University, Menoufiya, Egypt
| |
Collapse
|
57
|
Li Z, Chen G, Cai Z, Dong X, Qiu L, Xu H, Zeng Y, Liu X, Liu J. Genomic and transcriptional Profiling of tumor infiltrated CD8 + T cells revealed functional heterogeneity of antitumor immunity in hepatocellular carcinoma. Oncoimmunology 2018; 8:e1538436. [PMID: 30713796 PMCID: PMC6343808 DOI: 10.1080/2162402x.2018.1538436] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 02/06/2023] Open
Abstract
As key players in HCC antitumor response, the functions of tumor infiltrated CD8+ T cells are significantly affected by surrounding microenvironment. A detailed profiling of their genomic and transcriptional changes could provide valuable insights for both future immunotherapy development and prognosis evaluation. We performed whole exome and transcriptome sequencing on tumor infiltrated CD8+ T cells and CD8+ T cells isolated from other tissue origins (peritumor tissues and corresponding PBMCs) in eight treatment-naive HCC patients. The results demonstrated that transcriptional changes, rather than genomic alterations were the main contributors to the functional alterations of CD8+ T cells in the process of tumor progression. The origins of CD8+ T cells defined their transcriptional landscape, while the tumor infiltrated CD8+ T cells shared more similarity with peritumor-derived CD8+ T cells compared with those CD8+ T cells in blood. In addition, tumor infiltrated CD8+ T cells also showed larger transcriptional heterogeneity among individuals, which was modulated by clinical features such as HBV levels, preoperative anti-viral treatment and the degree of T cell infiltration. We also identified multiple inter-connected pathways involved in the activation and exhaustion of tumor infiltrated CD8+ T cells, among which IL-12 mediated pathway could dynamically reflect the functional status of CD8+ TILs and activation of this pathway indicated a better prognosis. Our results presented an overview picture of CD8+ TILs' genomic and transcriptional landscape and features, as well as how the functional status of CD8+ TILs correlated with patients' clinical course.
Collapse
Affiliation(s)
- Zhenli Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Geng Chen
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Xiuqing Dong
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Liman Qiu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Haipo Xu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Yongyi Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Jingfeng Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| |
Collapse
|
58
|
Vrecko S, Guenat D, Mercier-Letondal P, Faucheu H, Dosset M, Royer B, Galaine J, Boidot R, Kim S, Jary M, Adotévi O, Borg C, Godet Y. Personalized identification of tumor-associated immunogenic neoepitopes in hepatocellular carcinoma in complete remission after sorafenib treatment. Oncotarget 2018; 9:35394-35407. [PMID: 30459932 PMCID: PMC6226040 DOI: 10.18632/oncotarget.26247] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 10/08/2018] [Indexed: 12/11/2022] Open
Abstract
Sorafenib, a multi-targeted kinase inhibitor, is the current standard systemic treatment for advanced hepatocellular carcinoma. Sorafenib has anti-angiogenic and anti-proliferative properties and is also known to favor anti-tumor T cell responses by reducing the population of immunosuppressive cells such as Treg and MDSC. Anti-tumor immune responses, especially mediated by CD4+ T-cells, are critical for tumor cells eradication and therapies modulating those responses are appealing in a growing number of cancers. Here, we report and investigate the case of a patient diagnosed with an advanced HCC treated by sorafenib who experienced a complete histological response. We aimed to identify immunogenic peptides derived from tumor mutated proteins that stimulated CD4+ T cells responses thus favoring the exceptional recovery process of this patient. Tumor neoantigens were identified using whole exome sequencing of normal and tumor tissue and peptide MHC binding prediction algorithms. Among 442 tumor-specific somatic variants, 50 missense mutations and 20 neoepitopes predicted to bind MHC-II were identified. Candidate neoepitopes immunogenicity was assessed by IFN-γ ELISpot after culture of patient's PBMCs in presence of synthetic neopeptides. CD4+ memory T cell responses were detected against a mutated IL-1βS230F peptide and two additional neoepitopes from HELZ2V241M and MLL2A4458V suggesting that efficient anti-tumor immune response occurred in this patient. These results showed that T cells can recognize neoantigens and may lead to the cancer elimination after immunomodulation in the tumor-microenvironment induced by sorafenib. This observation indicates that other immunotherapies in combination with sorafenib could potentially increase the response rate in HCC at advanced stage.
Collapse
Affiliation(s)
- Sindy Vrecko
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
| | - David Guenat
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
- University Bourgogne Franche-Comté, LabEx LipSTIC ANR-11-LABX-0021, Besançon F-25000, France
- University Hospital of Besançon, Department of Molecular and Cell Biology, Besançon F-25000, France
- Stanford Cancer Institute, Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305, USA
| | - Patricia Mercier-Letondal
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
| | - Hugues Faucheu
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
- University Hospital of Besançon, Department of Molecular and Cell Biology, Besançon F-25000, France
| | - Magalie Dosset
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
- University Bourgogne Franche-Comté, LabEx LipSTIC ANR-11-LABX-0021, Besançon F-25000, France
| | - Bernard Royer
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
- University Hospital of Besançon, Department of Pharmacology, Besançon F-25000, France
| | - Jeanne Galaine
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
| | - Romain Boidot
- Centre Georges-François Leclerc, Platform of Transfer in Cancer Biology, Department of Biology and Pathology of Tumours, Centre de Recherche INSERM LNC-UMR123, Dijon F-21000, France
| | - Stefano Kim
- University Hospital of Besançon, Department of Medical Oncology, Besançon F-25000, France
| | - Marine Jary
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
- University Hospital of Besançon, Department of Medical Oncology, Besançon F-25000, France
| | - Olivier Adotévi
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
- University Hospital of Besançon, Department of Medical Oncology, Besançon F-25000, France
| | - Christophe Borg
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
- University Hospital of Besançon, Department of Medical Oncology, Besançon F-25000, France
| | - Yann Godet
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon F-25000, France
| |
Collapse
|
59
|
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
|
60
|
Sheppard S, Ferry A, Guedes J, Guerra N. The Paradoxical Role of NKG2D in Cancer Immunity. Front Immunol 2018; 9:1808. [PMID: 30150983 PMCID: PMC6099450 DOI: 10.3389/fimmu.2018.01808] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
The activating receptor NKG2D and its ligands are recognized as a potent immune axis that controls tumor growth and microbial infections. With regards to cancer surveillance, various studies have demonstrated the antitumor function mediated by NKG2D on natural killer cells and on conventional and unconventional T cells. The use of NKG2D-deficient mice established the importance of NKG2D in delaying tumor development in transgenic mouse models of cancer. However, we recently demonstrated an unexpected, flip side to this coin, the ability for NKG2D to contribute to tumor growth in a model of inflammation-driven liver cancer. With a focus on the liver, here, we review current knowledge of NKG2D-mediated tumor surveillance and discuss evidence supporting a dual role for NKG2D in cancer immunity. We postulate that in certain advanced cancers, expression of ligands for NKG2D can drive cancer progression rather than rejection. We propose that the nature of the microenvironment within and surrounding tumors impacts the outcome of NKG2D activation. In a form of autoimmune attack, NKG2D promotes tissue damage, mostly in the inflamed tissue adjacent to the tumor, facilitating tumor progression while being ineffective at rejecting transformed cells in the tumor bed.
Collapse
Affiliation(s)
- Sam Sheppard
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Memorial Sloan Kettering Cancer Center, Zuckerman Research Center, New York, NY, United States
| | - Amir Ferry
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Joana Guedes
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Nadia Guerra
- Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
61
|
Wu MY, Yiang GT, Cheng PW, Chu PY, Li CJ. Molecular Targets in Hepatocarcinogenesis and Implications for Therapy. J Clin Med 2018; 7:213. [PMID: 30104473 PMCID: PMC6112027 DOI: 10.3390/jcm7080213] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 02/07/2023] Open
Abstract
Hepatocarcinogenesis comprises of multiple, complex steps that occur after liver injury and usually involve several pathways, including telomere dysfunction, cell cycle, WNT/β-catenin signaling, oxidative stress and mitochondria dysfunction, autophagy, apoptosis, and AKT/mTOR signaling. Following liver injury, gene mutations, accumulation of oxidative stress, and local inflammation lead to cell proliferation, differentiation, apoptosis, and necrosis. The persistence of this vicious cycle in turn leads to further gene mutation and dysregulation of pro- and anti-inflammatory cytokines, such as interleukin (IL)-1β, IL-6, IL-10, IL-12, IL-13, IL-18, and transforming growth factor (TGF)-β, resulting in immune escape by means of the NF-κB and inflammasome signaling pathways. In this review, we summarize studies focusing on the roles of hepatocarcinogenesis and the immune system in liver cancer. In addition, we furnish an overview of recent basic and clinical studies to provide a strong foundation to develop novel anti-carcinogenesis targets for further treatment interventions.
Collapse
Affiliation(s)
- Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Giuo-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Pei-Wen Cheng
- Yuh-Ing Junior College of Health Care & Management, Kaohsiung 807, Taiwan.
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
| | - Pei-Yi Chu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 231, Taiwan.
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
- National Institute of Cancer Research, National Health Research Institutes, Miaoli 704, Taiwan.
| | - Chia-Jung Li
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| |
Collapse
|
62
|
The Expanding Role of Systemic Therapy in the Management of Hepatocellular Carcinoma. Can J Gastroenterol Hepatol 2018; 2018:4763832. [PMID: 30159302 PMCID: PMC6106970 DOI: 10.1155/2018/4763832] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 07/25/2018] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) represents a global health problem, with the majority of patients presenting at an advanced or incurable stage. The development of effective systemic therapy options for this disease has been challenging because many HCC patients suffer from underlying liver cirrhosis that precludes the safe delivery of systemic therapy. The current review seeks to provide an overview of the current systemic therapeutic approaches for advanced HCC as well as some of the novel management strategies that are currently being evaluated.
Collapse
|
63
|
Zhu W, Peng Y, Wang L, Hong Y, Jiang X, Li Q, Liu H, Huang L, Wu J, Celis E, Merchen T, Kruse E, He Y. Identification of α-fetoprotein-specific T-cell receptors for hepatocellular carcinoma immunotherapy. Hepatology 2018; 68:574-589. [PMID: 29443377 PMCID: PMC7368991 DOI: 10.1002/hep.29844] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/04/2018] [Accepted: 02/12/2018] [Indexed: 12/17/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) is the major form of liver cancer for which there is no effective therapy. Genetic modification with T-cell receptors (TCRs) specific for HCC-associated antigens, such as α-fetoprotein (AFP), can potentially redirect human T cells to specifically recognize and kill HCC tumor cells to achieve antitumor effects. In this study, using lentivector and peptide immunization, we identified a population of cluster of differentiation 8 (CD8) T cells in human leukocyte antigen (HLA)-A2 transgenic AAD mice that recognized AFP158 epitope on human HCC cells. Adoptive transfer of the AFP158 -specific mouse CD8 T cells eradicated HepG2 tumor xenografts as large as 2 cm in diameter in immunocompromised nonobese diabetic severe combined immunodeficient gamma knockout (NSG) mice. We then established T-cell hybridoma clones from the AFP158 -specific mouse CD8 T cells and identified three sets of paired TCR genes out of five hybridomas. Expression of the murine TCR genes redirected primary human T cells to bind HLA-A2/AFP158 tetramer. TCR gene-engineered human T (TCR-T) cells also specifically recognized HLA-A2+ AFP+ HepG2 HCC tumor cells and produced effector cytokines. Importantly, the TCR-T cells could specifically kill HLA-A2+ AFP+ HepG2 tumor cells without significant toxicity to normal primary hepatocytes in vitro. Adoptive transfer of the AFP-specific TCR-T cells could eradicate HepG2 tumors in NSG mice. CONCLUSION We have identified AFP-specific murine TCR genes that can redirect human T cells to specifically recognize and kill HCC tumor cells, and those AFP158 -specific TCRs have a great potential to engineer a patient's autologous T cells to treat HCC tumors. (Hepatology 2018).
Collapse
Affiliation(s)
- Wei Zhu
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA
| | - Yibing Peng
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA
| | - Lan Wang
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA
| | - Yuan Hong
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA
| | - Xiaotao Jiang
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA
| | - Qi Li
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA
| | - Heping Liu
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA
| | - Lei Huang
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA
| | - Juan Wu
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA
| | - Esteban Celis
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA
| | - Todd Merchen
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA.,Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Edward Kruse
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA.,Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Yukai He
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA.,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| |
Collapse
|
64
|
Stereotactic Ablative Radiation Therapy Induces Systemic Differences in Peripheral Blood Immunophenotype Dependent on Irradiated Site. Int J Radiat Oncol Biol Phys 2018; 101:1259-1270. [PMID: 29891204 DOI: 10.1016/j.ijrobp.2018.04.038] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 04/07/2018] [Accepted: 04/16/2018] [Indexed: 01/13/2023]
Abstract
PURPOSE Despite the strong interest in combining stereotactic ablative radiation therapy (SAR) with immunotherapy, limited data characterizing the systemic immune response after SAR are available. We hypothesized that the systemic immune response to SAR would differ by irradiated site owing to inherent differences in the microenvironment of various organs. METHODS AND MATERIALS Patients receiving SAR to any organ underwent prospective blood banking before and 1 to 2 weeks after SAR. Peripheral blood mononuclear cells (PBMCs) and serum were isolated. PBMCs were stained with fluorophore-conjugated antibodies against T and natural killer (NK) cell markers. Cells were interrogated by flow cytometry, and the results were analyzed using FlowJo software. Serum cytokine and chemokine levels were measured using Luminex. We analyzed the changes from before to after therapy using paired t tests or 1-way analysis of variance (ANOVA) with Bonferroni's post-test. RESULTS A total of 31 patients had evaluable PBMCs for flow cytometry and 37 had evaluable serum samples for Luminex analysis. The total number of NK cells and cytotoxic (CD56dimCD16+) NK cells decreased (P = .02) and T-cell immunoglobulin- and mucin domain-containing molecule-3-positive (TIM3+) NK cells increased (P = .04) after SAR to parenchymal sites (lung and liver) but not to bone or brain. The total memory CD4+ T cells, activated inducible co-stimulator-positive and CD25+CD4+ memory T cells, and activated CD25+CD8+ memory T cells increased after SAR to parenchymal sites but not bone or brain. The circulating levels of tumor necrosis factor-α (P = .04) and multiple chemokines, including RANTES (P = .04), decreased after SAR to parenchymal sites but not bone or brain. CONCLUSIONS Our data suggest SAR to parenchymal sites induces systemic immune changes, including a decrease in total and cytotoxic NK cells, an increase in TIM3+ NK cells, and an increase in activated memory CD4+ and CD8+ T cells. SAR to nonparenchymal sites did not induce these changes. By comparing the immune response after radiation to different organs, our data suggest SAR induces systemic immunologic changes that are dependent on the irradiated site.
Collapse
|
65
|
Kumar N, Khakoo SI. Hepatocellular carcinoma: Prospects for natural killer cell immunotherapy. HLA 2018; 92:3-11. [PMID: 29667374 DOI: 10.1111/tan.13275] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 12/12/2022]
Abstract
Liver disease is a growing cause of death in the United Kingdom and the incidence of hepatocellular carcinoma (HCC) is rising (http://www.cancerresearchuk.org/). The combination of an immunosuppressive environment within the liver and suboptimal host anti-tumour immune responses may account for the poor survival outcome of HCC. Understanding how tumours evade immune recognition coupled with new insights into the unique immunological environment within the liver will be critical to developing liver-specific immunotherapies.
Collapse
Affiliation(s)
- N Kumar
- Section of Hepatology, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - S I Khakoo
- Department of Clinical and Experimental Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, UK
| |
Collapse
|
66
|
Wu X, Roberto JB, Knupp A, Kenerson HL, Truong CD, Yuen SY, Brempelis KJ, Tuefferd M, Chen A, Horton H, Yeung RS, Crispe IN. Precision-cut human liver slice cultures as an immunological platform. J Immunol Methods 2018; 455:71-79. [PMID: 29408707 PMCID: PMC6689534 DOI: 10.1016/j.jim.2018.01.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/17/2018] [Accepted: 01/24/2018] [Indexed: 12/16/2022]
Abstract
The liver is the central metabolic organ in the human body, and also plays an essential role in innate and adaptive immunity. While mouse models offer significant insights into immune-inflammatory liver disease, human immunology differs in important respects. It is not easy to address those differences experimentally. Therefore, to improve the understanding of human liver immunobiology and pathology, we have established precision-cut human liver slices to study innate immunity in human tissue. Human liver slices collected from resected livers could be maintained in ex vivo culture over a two-week period. Although an acute inflammatory response accompanied by signs of tissue repair was observed in liver tissue following slicing, the expression of many immune genes stabilized after day 4 and remained stable until day 15. Remarkably, histological evidence of pre-existing liver diseases was preserved in the slices for up to 7 days. Following 7 days of culture, exposure of liver slices to the toll-like receptor (TLR) ligands, TLR3 ligand Poly-I:C and TLR4 ligand LPS, resulted in a robust activation of acute inflammation and cytokine genes. Moreover, Poly-I:C treatment induced a marked antiviral response including increases of interferons IFNB, IL-28B and a group of interferon-stimulated genes. Therefore, precision-cut liver slices emerge as a valuable tool to study human innate immunity.
Collapse
Affiliation(s)
- Xia Wu
- Department of Pathology, University of Washington, Seattle, WA 98195, USA.
| | - Jessica B Roberto
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Allison Knupp
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Heidi L Kenerson
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Camtu D Truong
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Sebastian Y Yuen
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | | | - Marianne Tuefferd
- Infectious Diseases and Vaccines, Janssen Research and Development, B-2340 Beerse, Belgium
| | - Antony Chen
- Infectious Diseases and Vaccines, Janssen Research and Development, B-2340 Beerse, Belgium
| | - Helen Horton
- Infectious Diseases and Vaccines, Janssen Research and Development, B-2340 Beerse, Belgium
| | - Raymond S Yeung
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Ian N Crispe
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
67
|
Obeid JM, Kunk PR, Zaydfudim VM, Bullock TN, Slingluff CL, Rahma OE. Immunotherapy for hepatocellular carcinoma patients: is it ready for prime time? Cancer Immunol Immunother 2018; 67:161-174. [PMID: 29052780 PMCID: PMC11028155 DOI: 10.1007/s00262-017-2082-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 10/15/2017] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and the second most common cause of cancer death worldwide. Current treatment options for patients with intermediate and advanced HCC are limited, and there is an unmet need for novel therapeutic approaches. HCC is an attractive target for immunomodulation therapy, since it arises in an inflammatory milieu due to hepatitis B and C infections and cirrhosis. However, a major barrier to the development and success of immunotherapy in patients with HCC is the liver's inherent immunosuppressive function. Recent advances in the field of cancer immunology allowed further characterization of immune cell subsets and function, and created new opportunities for therapeutic modulation of the immune system. In this review, we present the different immune cell subsets involved in potential immune modulation of HCC, discuss their function and clinical relevance, review the variety of immune therapeutic agents currently under investigation in clinical trials, and outline future research directions.
Collapse
Affiliation(s)
- Joseph M Obeid
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Paul R Kunk
- Division of Hematology-Oncology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | | | - Timothy N Bullock
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Craig L Slingluff
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Osama E Rahma
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, 450 Brookline Avenue, M1B13, Boston, MA, 02215, USA.
| |
Collapse
|
68
|
Li L, Huang T, Lan C, Jia A, Mao Y. The growth inhibitory effects of garlic polysaccharide combined with cis-dichlorodiamine platinum on human HepG2 cells. Biologia (Bratisl) 2017. [DOI: 10.1515/biolog-2017-0155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
69
|
Moris D, Rahnemai-Azar AA, Zhang X, Ntanasis-Stathopoulos I, Tsilimigras DI, Chakedis J, Argyrou C, Fung JJ, Pawlik TM. Program death-1 immune checkpoint and tumor microenvironment in malignant liver tumors. Surg Oncol 2017; 26:423-430. [PMID: 29113661 DOI: 10.1016/j.suronc.2017.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/29/2017] [Indexed: 12/28/2022]
Abstract
Hepatic malignancies are one of the leading causes of cancer death globally. Considering the limited efficacy of current standard treatments in management of patients with advanced liver cancers, there has been a growing interest in identifying novel therapies. Despite achieving promising results in initial clinical trials, the therapeutic benefit of immunotherapy is limited due to strong immune-tolerogenic characteristics of liver tumors. Therapeutic regimens that impede tumor immunosuppressive mechanisms or elaborate tumor-specific immunity may improve clinical outcomes of patients with liver malignancies. Programmed cell death 1 (PD-1), an inhibitory checkpoint molecule, and its ligands (PD-L1 and -L2) are the main mediators of immunosuppression within the tumor microenvironment. The expression level of PD-1/PD-L1 may act as a biomarker to predict disease progression, as well as long-term survival. Furthermore, early trials have demonstrated the efficacy and safety of targeting PD-1/PD-L1 as an emerging field in the management of patients with advanced hepatocellular carcinoma. We herein review the role of PD-1/PD-L1 in the pathogenesis of liver malignancies, as well as its potential diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Demetrios Moris
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Amir A Rahnemai-Azar
- Department of Surgery, University of Washington Medical Center, Seattle, WA, USA
| | - XuFeng Zhang
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Diamantis I Tsilimigras
- First Department of Pathology, National and Kapodistrian University of Athens, Athens, Greece
| | - Jeffery Chakedis
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Chrysoula Argyrou
- First Department of Pathology, National and Kapodistrian University of Athens, Athens, Greece
| | - John J Fung
- Department of Surgery, University of Chicago Medicine Transplant Institute, Chicago, IL, USA
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
70
|
Shi C, Zhang Y, Yang H, Dong T, Chen Y, Xu Y, Yang X, Liu P. Ultrasound-targeted microbubble destruction-mediated Foxp3 knockdown may suppress the tumor growth of HCC mice by relieving immunosuppressive Tregs function. Exp Ther Med 2017; 15:31-38. [PMID: 29387180 PMCID: PMC5769241 DOI: 10.3892/etm.2017.5421] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 04/10/2017] [Indexed: 12/21/2022] Open
Abstract
The aim of the present study was to investigate the effect of Forkhead family transcription factor P3 (Foxp3) knockdown on the function of cluster of differentiation (CD)4+CD25+ regulatory T cell (Tregs) and the tumor growth of a hepatocellular carcinoma (HCC) mouse model. CD4+CD25+ Tregs and CD4+CD25- T cells were sorted from peripheral blood mononuclear cells (PBMCs) of patients with HCC. Then, ultrasound-targeted microbubble destruction (UTMD)-mediated Foxp3-microRNA (miRNA) was transfected into Tregs. Subsequently, CD4+CD25- T cells were co-cultured with PBMC and Tregs without Foxp3-miRNA (Foxp3+Tregs) or Tregs with Foxp3-miRNA (Foxp3-Tregs) and the proliferation-inhibition ratio of CD4+CD25- T cells was detected using a Cell Counting Kit-8. Additionally, HCC mice were treated with UTMD-mediated Foxp3-shRNA, the tumor volume was calculated and the content of CD4+ and CD25+ T cells in the blood were detected using flow cytometry. The content of interferon-γ (IFN-γ), interleukin (IL)-2, IL-10, transforming growth factor-β (TGF-β) and vascular endothelial growth factor (VEGF) in cultural supernatant and serum were detected by ELISA analysis. Foxp3-Tregs significantly reduced the inhibition effect of Foxp3+Tregs on the proliferation of CD4+CD25- T cells (P<0.01). The content of IFN-γ and IL-2 significantly increased, while IL-10 and TGF-β significantly decreased in the co-cultured system of Foxp3-Tregs compared with the co-cultured system of Foxp3+Tregs (P<0.01). Following treatment with Foxp3-shRNA, the average tumor volume, ratio of Tregs/CD4+ T cells and level of IL-10, TGF-β and VEGF significantly decreased, however, the level of IFN-γ and IL-2 significantly increased compared with un-treated HCC mice (P<0.05). Foxp3 knockdown may suppress the tumor growth of HCC mice through relieving the immunosuppressive function of Tregs.
Collapse
Affiliation(s)
- Chunying Shi
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, Harbin 150001, P.R. China
| | - Yu Zhang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, Harbin 150001, P.R. China
| | - Haichao Yang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, Harbin 150001, P.R. China
| | - Tianxiu Dong
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, Harbin 150001, P.R. China
| | - Yaodong Chen
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, Harbin 150001, P.R. China
| | - Yutong Xu
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, Harbin 150001, P.R. China
| | - Xiuhua Yang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, Harbin 150001, P.R. China
| | - Pengfei Liu
- MRI Department, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, Harbin 150001, P.R. China
| |
Collapse
|
71
|
Lu Z, Zuo B, Jing R, Gao X, Rao Q, Liu Z, Qi H, Guo H, Yin H. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol 2017; 67:739-748. [PMID: 28549917 DOI: 10.1016/j.jhep.2017.05.019] [Citation(s) in RCA: 305] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Dendritic cell (DC)-derived exosomes (DEXs) form a new class of vaccines for cancer immunotherapy. However, their potency in hepatocellular carcinoma (HCC), a life-threatening malignancy with limited treatment options in the clinic that responds poorly to immunotherapy, remains to be investigated. METHODS Exosomes derived from α-fetoprotein (AFP)-expressing DCs (DEXAFP) were investigated in three different HCC mouse models systemically. Tumor growth and microenvironment were monitored. RESULTS DEXAFP elicited strong antigen-specific immune responses and resulted in significant tumor growth retardation and prolonged survival rates in mice with ectopic, orthotopic and carcinogen-induced HCC tumors that displayed antigenic and pathological heterogeneity. The tumor microenvironment was improved in DEXAFP-treated HCC mice, demonstrated by significantly more γ-interferon (IFN-γ)-expressing CD8+ T lymphocytes, elevated levels of IFN-γ and interleukin-2, and fewer CD25+Foxp3+ regulatory T (Treg) cells and decreased levels of interleukin-10 and transforming growth factor-β in tumor sites. Lack of efficacy in athymic nude mice and CD8+ T cell-depleted mice showed that T cells contribute to DEXAFP-mediated antitumor function. Dynamic examination of the antitumor efficacy and the immune microenvironment in DEXAFP-treated orthotopic HCC mice at different time-points revealed a positive correlation between tumor suppression and immune microenvironment. CONCLUSIONS Our findings provide evidence that AFP-enriched DEXs can trigger potent antigen-specific antitumor immune responses and reshape the tumor microenvironment in HCC mice and thus provide a cell-free vaccine option for HCC immunotherapy. Lay summary: Dendritic cell (DC)-derived exosomes (DEXs) form a new class of vaccines for cancer immunotherapy. However, their potency in hepatocellular carcinoma (HCC) remains unknown. Here, we investigated exosomes from HCC antigen-expressing DCs in three different HCC mouse models and proved their feasibility and capability of treating HCC, and thus provide a cell-free vaccine for HCC immunotherapy.
Collapse
Affiliation(s)
- Zhen Lu
- Department of Cell Biology and Research Centre of Basic Medical Science, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Bingfeng Zuo
- Department of Cell Biology and Research Centre of Basic Medical Science, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Renwei Jing
- Department of Cell Biology and Research Centre of Basic Medical Science, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Xianjun Gao
- Department of Cell Biology and Research Centre of Basic Medical Science, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Quan Rao
- Department of Cell Biology and Research Centre of Basic Medical Science, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China; Third Central Clinical College, Tianjin Medical University, Jintang Road, Hedong District, Tianjin 300170, China
| | - Zhili Liu
- Department of Cell Biology and Research Centre of Basic Medical Science, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Han Qi
- Department of Cell Biology and Research Centre of Basic Medical Science, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Hongxing Guo
- Third Central Clinical College, Tianjin Medical University, Jintang Road, Hedong District, Tianjin 300170, China
| | - HaiFang Yin
- Department of Cell Biology and Research Centre of Basic Medical Science, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China.
| |
Collapse
|
72
|
Liu D, Li G, Avella DM, Kimchi ET, Kaifi JT, Rubinstein MP, Camp ER, Rockey DC, Schell TD, Staveley-O'Carroll KF. Sunitinib represses regulatory T cells to overcome immunotolerance in a murine model of hepatocellular cancer. Oncoimmunology 2017; 7:e1372079. [PMID: 29296523 DOI: 10.1080/2162402x.2017.1372079] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/18/2017] [Accepted: 08/23/2017] [Indexed: 02/08/2023] Open
Abstract
Successful development of immunotherapeutic strategies for hepatocellular cancer (HCC) has been impeded by limited understanding of tumor-induced profound tolerance and lack of a clinically faithful HCC model. Recently, we developed a novel model that recapitulates typical features of human HCC. Using this clinically relevant model, we demonstrate that tumor growth impairs host immunity and causes a profound exhaustion of tumor antigen-specific (TAS) CD8+ T cells. Increase in frequency and suppressive function of regulatory T cells (Tregs) is critically involved in this tumor-induced immune dysfunction. We further demonstrate that sunitinib suppresses Tregs and prevents tumor-induced immune tolerance, allowing TAS immunization to activate endogenous CD8+ T cells. As a result, this combinational strategy delays tumor growth. Importantly, the additional integration of exogenous naïve TAS CD8+ T cells by adoptive cell transfer (ACT) leads to the elimination of the established tumors without recurrence and promotes long-term survival of the treated mice. Mechanistically, sunitinib treatment primes the antitumor immune response by significantly decreasing Treg frequency, reducing TGF-β and IL-10 production by Tregs, and also protecting TAS CD8+ T cells from tumor-induced deletion in the setting of HCC. Taken together, sunitinib quantitatively and qualitatively modifies Tregs to overcome tumor-induced immune deficiency, suggesting the potential of sunitinib as a therapeutic immune activator for HCC control.
Collapse
Affiliation(s)
- Dai Liu
- Department of Surgery, University of Missouri-Columbia, Columbia, MO, USA
| | - Guangfu Li
- Department of Surgery, University of Missouri-Columbia, Columbia, MO, USA.,Department of Microbiology and Immunology, University of Missouri-Columbia, Columbia, MO, USA
| | | | - Eric T Kimchi
- Department of Surgery, University of Missouri-Columbia, Columbia, MO, USA.,Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Jussuf T Kaifi
- Department of Surgery, University of Missouri-Columbia, Columbia, MO, USA.,Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Mark P Rubinstein
- Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO, USA
| | - E Ramsay Camp
- Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Don C Rockey
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA; Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Todd D Schell
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Kevin F Staveley-O'Carroll
- Department of Surgery, University of Missouri-Columbia, Columbia, MO, USA.,Ellis Fischel Cancer Center, University of Missouri-Columbia, Columbia, MO, USA
| |
Collapse
|
73
|
Moris D, Beal EW, Chakedis J, Burkhart RA, Schmidt C, Dillhoff M, Zhang X, Theocharis S, Pawlik TM. Role of exosomes in treatment of hepatocellular carcinoma. Surg Oncol 2017; 26:219-228. [PMID: 28807240 DOI: 10.1016/j.suronc.2017.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/24/2017] [Indexed: 02/07/2023]
Abstract
Exosomes are nanovesicles that may play a role in intercellular communication by acting as carriers of functional contents such as proteins, lipids, RNA molecules and circulating DNA from donor to recipient cells. In addition, exosomes may play a potential role in immunosurveillance and tumor pathogenesis and progression. Recently, research has increasingly focused on the role of exosomes in hepatocellular carcinoma (HCC), the most common primary liver malignancy. We herein review data on emerging experimental and clinical studies focused on the role of exosomes in the pathogenesis, diagnosis, progression and chemotherapy response of patients with HCC. Beyond their diagnostic value in HCC, exosomes are involved in different mechanisms of HCC tumor pathogenesis and progression including angiogenesis and immune escape. Moreover, exosomes have been demonstrated to change the tumor microenvironment to a less tolerogenic state, favoring immune response and tumor suppression. These results underline a practical and potentially feasible role of exosomes in the treatment of patients with HCC, both as a target and a vehicle for drug design. Future studies will need to further elucidate the exact role and reliability of exosomes as screening, diagnostic and treatment targets in patients with HCC.
Collapse
Affiliation(s)
- Demetrios Moris
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Eliza W Beal
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Jeffery Chakedis
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Richard A Burkhart
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Carl Schmidt
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Mary Dillhoff
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Xufeng Zhang
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Cancer Hospital and Solove Research Institute, Columbus, OH, USA.
| |
Collapse
|
74
|
Khemlina G, Ikeda S, Kurzrock R. The biology of Hepatocellular carcinoma: implications for genomic and immune therapies. Mol Cancer 2017; 16:149. [PMID: 28854942 PMCID: PMC5577674 DOI: 10.1186/s12943-017-0712-x] [Citation(s) in RCA: 310] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/15/2017] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC), the most common type of primary liver cancer, is a leading cause of cancer-related death worldwide. It is highly refractory to most systemic therapies. Recently, significant progress has been made in uncovering genomic alterations in HCC, including potentially targetable aberrations. The most common molecular anomalies in this malignancy are mutations in the TERT promoter, TP53, CTNNB1, AXIN1, ARID1A, CDKN2A and CCND1 genes. PTEN loss at the protein level is also frequent. Genomic portfolios stratify by risk factors as follows: (i) CTNNB1 with alcoholic cirrhosis; and (ii) TP53 with hepatitis B virus-induced cirrhosis. Activating mutations in CTNNB1 and inactivating mutations in AXIN1 both activate WNT signaling. Alterations in this pathway, as well as in TP53 and the cell cycle machinery, and in the PI3K/Akt/mTor axis (the latter activated in the presence of PTEN loss), as well as aberrant angiogenesis and epigenetic anomalies, appear to be major events in HCC. Many of these abnormalities may be pharmacologically tractable. Immunotherapy with checkpoint inhibitors is also emerging as an important treatment option. Indeed, 82% of patients express PD-L1 (immunohistochemistry) and response rates to anti-PD-1 treatment are about 19%, and include about 5% complete remissions as well as durable benefit in some patients. Biomarker-matched trials are still limited in this disease, and many of the genomic alterations in HCC remain challenging to target. Future studies may require combination regimens that include both immunotherapies and molecularly matched targeted treatments.
Collapse
Affiliation(s)
- Galina Khemlina
- Department of Geriatrics, University of California, UC San Diego, 9500 Gilman Drive, #9111, La Jolla, CA, 92093-9111, USA. .,Kaiser Permanente Southern California, San Diego, USA.
| | - Sadakatsu Ikeda
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, University of California, Moores Cancer Center, San Diego, USA.,Tokyo Medical and Dental University, Tokyo, Japan
| | - Razelle Kurzrock
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, University of California, Moores Cancer Center, San Diego, USA
| |
Collapse
|
75
|
Pei Y, Sun X, Guo X, Yin H, Wang L, Tian F, Jing H, Liang X, Xu J, Shi P. FGF8 promotes cell proliferation and resistance to EGFR inhibitors via upregulation of EGFR in human hepatocellular carcinoma cells. Oncol Rep 2017; 38:2205-2210. [PMID: 28791365 DOI: 10.3892/or.2017.5887] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 06/13/2017] [Indexed: 11/06/2022] Open
Abstract
Fibroblast growth factor 8 (FGF8), a member of the fibroblast growth factor (FGF) family, is upregulated in several human cancers, including HCC (HCC). Previous studies have demonstrated that FGF8 increased cell growth and invasion of tumor cells. In the present study we investigated whether FGF8 is involved in the cell proliferation and resistance to several drugs in human HCC cells. We stably overexpressed FGF8 by lentiviral transfection. In addition, we also added recombinant FGF8 instead of stably overexpressing FGF8 in human HCC cells. Stable overexpression of FGF8 or exogenous recombinant FGF8 resulted in significantly enhanced cell proliferation in human HCC cells. With the use of CellTiter-Glo assay for the determination of cell viability, we found that FGF8 increased the resistance to epidermal growth factor receptor (EGFR) inhibitors in human HCC cells. Additionally, the expression of EGFR was also upregulated by stably overexpressing FGF8 or exogenous recombinant FGF8. Yes-associated protein 1 (YAP1) was reported to upregulate the expression of EGFR. Moreover, we also found that FGF8 increased the expression of YAP1 and knockdown of YAP1 eliminated the upregulation of EGFR and the resistance to EGFR inhibition induced by FGF8. Our study provides evidence that FGF8 plays an important role in the resistance to EGFR inhibition of human HCC cells.
Collapse
Affiliation(s)
- Yuanmin Pei
- Department of General Surgery, Yidu Central Hospital of Weifang, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Xueling Sun
- Department of General Surgery, Yidu Central Hospital of Weifang, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Xiwei Guo
- Department of General Surgery, Yidu Central Hospital of Weifang, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Huashan Yin
- Department of General Surgery, Yidu Central Hospital of Weifang, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Le Wang
- Shanxi Breast Cancer Center, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Fugu Tian
- Shanxi Breast Cancer Center, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Hongxi Jing
- Shanxi Breast Cancer Center, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xiaobo Liang
- Shanxi Breast Cancer Center, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jun Xu
- Shanxi Breast Cancer Center, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Pengcheng Shi
- Shanxi Breast Cancer Center, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
76
|
Zhang Q, Yuan XF, Lu Y, Li ZZ, Bao SQ, Zhang XL, Yang YY, Fan DM, Zhang YZ, Wu CX, Guo HX, Zhang YJ, Ye Z, Xiong DS. Surface expression of anti-CD3scfv stimulates locoregional immunotherapy against hepatocellular carcinoma depending on the E1A-engineered human umbilical cord mesenchymal stem cells. Int J Cancer 2017. [PMID: 28643325 DOI: 10.1002/ijc.30846] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Tumor antigens is at the core of cancer immunotherapy, however, the ideal antigen selection is difficult especially in poorly immunogenic tumors. In this study, we designed a strategy to modify hepatocellular carcinoma (HCC) cells by surface expressing anti-CD3scfv within the tumor site strictly, which depended on the E1A-engineered human umbilical cord mesenchymal stem cells (HUMSC.E1A) delivery system. Subsequently, membrane-bound anti-CD3scfv actived the lymphocytes which lysed HCC cells bypassing the expression of antigens or MHC restriction. First, we constructed the anti-CD3scfv gene driven by human α-fetoprotein (AFP) promoter into an adenoviral vector and the E1A gene into the lentiviral vector. Our results showed that anti-CD3scfv could specifically express on the surface of HCC cells and activate the lymphocytes to kill target cells effectively in vitro. HUMSC infected by AdCD3scfv followed by LentiR.E1A could support the adenoviral replication and packaging in vitro 36 h after LentiR.E1A infection. Using a subcutaneous HepG2 xenograft model, we confirmed that AdCD3scfv and LentiR.E1A co-transfected HUMSC could migrate selectively to the tumor site and produce considerable adenoviruses. The new generated AdCD3scfv infected and modified tumor cells successfully. Mice injected with the MSC.E1A.AdCD3scfv and lymphocytes significantly inhibited the tumor growth compared with control groups. Furthermore, 5-fluorouracil (5-FU) could sensitize adenovirus infection at low MOI resulting in improved lymphocytes cytotoxicity in vitro and in vivo. In summary, this study provides a promising strategy for solid tumor immunotherapy.
Collapse
Affiliation(s)
- Qing Zhang
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Xiang-Fei Yuan
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.,Institute of Integrative Medicine for Acute Abdominal Diseases, Nankai Hospital, Tianjin, 300100, China
| | - Yang Lu
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Zhen-Zhen Li
- National-Local Joint Engineering Research Center of Biodiagnostics & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Shi-Qi Bao
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Xiao-Long Zhang
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Yuan-Yuan Yang
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Dong-Mei Fan
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Yi-Zhi Zhang
- Central Hospital of Karamay, Karamay, Xinjiang, 834000, China
| | - Chen-Xuan Wu
- the Third Central Hospital of Tianjin Medical University, Tianjin, 300170, China
| | - Hong-Xing Guo
- the Third Central Hospital of Tianjin Medical University, Tianjin, 300170, China
| | - Yan-Jun Zhang
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Zhou Ye
- Central Hospital of Karamay, Karamay, Xinjiang, 834000, China
| | - Dong-Sheng Xiong
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| |
Collapse
|
77
|
Giakoustidis AE, Giakoustidis DE. Immunosuppression strategies in liver transplantation patient; patients with hepatocellular carcinoma. Immunotherapy 2017; 9:197-206. [PMID: 28128716 DOI: 10.2217/imt-2016-0110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) consists the main primary malignant tumor of the liver. There is an underlining liver cirrhosis mainly attributed to chronic hepatitis B virus or hepatitis C virus, alcoholic liver disease, nonalcoholic steatohepatitis and other pathologic conditions. Liver transplantation consists a radical management, treating both cancer and cirrhosis. By introducing the Milan Criteria for liver transplantation in HCC patients there was a 5-year survival escalation. Even though there is a careful selection of patients with HCC for transplantation, recurrent disease is still high. The role of immusuppression therapy is of paramount importance, in order to avoid acute and chronic graft rejection while protecting the patient from tumor recurrence. In recent years newer immunosuppressive agents such as the mTOR inhibitors are proposed, having dual properties, as both immunosuppressive and antitumors agents.
Collapse
Affiliation(s)
- Alexander E Giakoustidis
- Hepato-Pancreato-Biliary Surgery Department, The Royal London Hospital, Barts Health, Whitechapel Road, London E1 1BB, UK
| | - Dimitrios E Giakoustidis
- Division of Transplant Surgery, Department of Surgery, School of Health Sciences, Aristotle University of Thessaloniki & Hippokration General Hospital, Thessaloniki, Greece
| |
Collapse
|
78
|
Scarabel L, Perrone F, Garziera M, Farra R, Grassi M, Musiani F, Russo Spena C, Salis B, De Stefano L, Toffoli G, Rizzolio F, Tonon F, Abrami M, Chiarappa G, Pozzato G, Forte G, Grassi G, Dapas B. Strategies to optimize siRNA delivery to hepatocellular carcinoma cells. Expert Opin Drug Deliv 2017; 14:797-810. [PMID: 28266887 DOI: 10.1080/17425247.2017.1292247] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/03/2017] [Indexed: 02/08/2023]
Abstract
hepatocellular carcinoma (hcc) is the predominant form of primary liver cancer and the second leading cause of cancer-associated mortality worldwide. available therapies for hcc have limited efficacy due to often late diagnosis and the general resistance of hcc to anti-cancer agents; therefore, the development of novel therapeutics is urgently required. small-interfering rna (sirna) molecules are short, double-stranded rnas that specifically recognize and bind the mrna of a target gene to inhibit gene expression. despite the great therapeutic potential of sirnas towards many human tumors including hcc, their use is limited by suboptimal delivery. Areas covered: In this review, we outline the current data regarding the therapeutic potential of siRNAs in HCC and describe the development of effective siRNA delivery systems. We detail the key problems associated with siRNA delivery and discuss the possible solutions. Finally, we provide examples of the various siRNA delivery strategies that have been employed in animal models of HCC and in human patients enrolled in clinical trials. Expert opinion: Despite the existing difficulties in siRNA delivery for HCC, the increasing scientific attention and breakthrough studies in this field is facilitating the design of novel and efficient technical solutions that may soon find practical applications.
Collapse
Affiliation(s)
- Lucia Scarabel
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Francesca Perrone
- b Department of Life Sciences, Cattinara University Hospital , University of Trieste , Trieste , Italy
| | - Marica Garziera
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Rossella Farra
- c Department of Engineering and Architecture , University of Trieste , Trieste , Italy
| | - Mario Grassi
- c Department of Engineering and Architecture , University of Trieste , Trieste , Italy
| | - Francesco Musiani
- d Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Concetta Russo Spena
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Barbara Salis
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Lucia De Stefano
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Giuseppe Toffoli
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Flavio Rizzolio
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Federica Tonon
- c Department of Engineering and Architecture , University of Trieste , Trieste , Italy
| | - Michela Abrami
- b Department of Life Sciences, Cattinara University Hospital , University of Trieste , Trieste , Italy
| | - Gianluca Chiarappa
- c Department of Engineering and Architecture , University of Trieste , Trieste , Italy
| | - Gabriele Pozzato
- e Department of 'Scienze Mediche, Chirurgiche e della Salute' , Cattinara University Hospital, University of Trieste , Trieste , Italy
| | - Giancarlo Forte
- f Center for Translational Medicine, International Clinical Research Center , St. Anne's University Hospital , Brno , Czech Republic
| | - Gabriele Grassi
- b Department of Life Sciences, Cattinara University Hospital , University of Trieste , Trieste , Italy
- e Department of 'Scienze Mediche, Chirurgiche e della Salute' , Cattinara University Hospital, University of Trieste , Trieste , Italy
| | - Barbara Dapas
- b Department of Life Sciences, Cattinara University Hospital , University of Trieste , Trieste , Italy
| |
Collapse
|
79
|
Vujanovic L, Stahl EC, Pardee AD, Geller DA, Tsung A, Watkins SC, Gibson GA, Storkus WJ, Butterfield LH. Tumor-Derived α-Fetoprotein Directly Drives Human Natural Killer-Cell Activation and Subsequent Cell Death. Cancer Immunol Res 2017; 5:493-502. [PMID: 28468916 DOI: 10.1158/2326-6066.cir-16-0216] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 03/22/2017] [Accepted: 04/28/2017] [Indexed: 11/16/2022]
Abstract
Hepatocellular carcinoma (HCC) patients with reduced natural killer (NK)-cell numbers and function have been shown to have a poor disease outcome. Mechanisms underlying NK-cell deficiency and dysfunction in HCC patients remain largely unresolved. α-Fetoprotein (AFP) is an oncofetal antigen produced by HCC. Previous studies demonstrated that tumor-derived AFP (tAFP) can indirectly impair NK-cell activity by suppressing dendritic cell function. However, a direct tAFP effect on NK cells remains unexplored. The purpose of this study was to examine the ability of cord blood-derived AFP (nAFP) and that of tAFP to directly modulate human NK-cell activity and longevity in vitro Short-term exposure to tAFP and, especially, nAFP proteins induced a unique proinflammatory, IL2-hyperresponsive phenotype in NK cells as measured by IL1β, IL6, and TNF secretion, CD69 upregulation, and enhanced tumor cell killing. In contrast, extended coculture with tAFP, but not nAFP, negatively affected long-term NK-cell viability. NK-cell activation was directly mediated by the AFP protein itself, whereas their viability was affected by hydrophilic components within the low molecular mass cargo that copurified with tAFP. Identification of the distinct impact of circulating tAFP on NK-cell function and viability may be crucial to developing a strategy to ameliorate HCC patient NK-cell functional deficits. Cancer Immunol Res; 5(6); 493-502. ©2017 AACR.
Collapse
Affiliation(s)
- Lazar Vujanovic
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pennsylvania.,Department of Medicine, University of Pittsburgh, Pennsylvania
| | - Elizabeth C Stahl
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pennsylvania
| | - Angela D Pardee
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pennsylvania.,Department of Medicine, University of Pittsburgh, Pennsylvania
| | - David A Geller
- University of Pittsburgh School of Medicine, Department of Surgery, University of Pittsburgh, Pennsylvania
| | - Allan Tsung
- University of Pittsburgh School of Medicine, Department of Surgery, University of Pittsburgh, Pennsylvania
| | - Simon C Watkins
- Department of Cell Biology and Physiology, University of Pittsburgh, Pennsylvania
| | - Gregory A Gibson
- Department of Cell Biology and Physiology, University of Pittsburgh, Pennsylvania
| | - Walter J Storkus
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pennsylvania.,Department of Dermatology, University of Pittsburgh, Pennsylvania.,Department of Immunology, University of Pittsburgh, Pennsylvania
| | - Lisa H Butterfield
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pennsylvania. .,Department of Medicine, University of Pittsburgh, Pennsylvania.,University of Pittsburgh School of Medicine, Department of Surgery, University of Pittsburgh, Pennsylvania.,Department of Immunology, University of Pittsburgh, Pennsylvania
| |
Collapse
|
80
|
Yoo SY, Badrinath N, Woo HY, Heo J. Oncolytic Virus-Based Immunotherapies for Hepatocellular Carcinoma. Mediators Inflamm 2017; 2017:5198798. [PMID: 28512387 PMCID: PMC5415860 DOI: 10.1155/2017/5198798] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/08/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma is highly refractory cancer which is resistant to conventional chemotherapy and radiotherapy, carrying a dismal prognosis. Although many anticancer drugs have been developed for treating HCC, sorafenib is the only effective treatment, but it only prolongs survival duration for about 3 months. Recently, oncolytic virotherapy has shown promising results in treating HCCs and the effects can be more enhanced by adopting immune modulatory molecules. This review discusses the current status of treating HCC and the effective strategy of oncolytic virus-based immunotherapy for the treatment of HCCs.
Collapse
Affiliation(s)
- So Young Yoo
- BIO-IT Foundry Technology Institute, Pusan National University, Busan 46241, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Department of Internal Medicine, College of Medicine, Pusan National University and Medical Research Institute, Yangsan 50612, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
| | - Narayanasamy Badrinath
- BIO-IT Foundry Technology Institute, Pusan National University, Busan 46241, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Hyun Young Woo
- Department of Internal Medicine, College of Medicine, Pusan National University and Medical Research Institute, Yangsan 50612, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
| | - Jeong Heo
- Department of Internal Medicine, College of Medicine, Pusan National University and Medical Research Institute, Yangsan 50612, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
| |
Collapse
|
81
|
Abstract
Hepatocellular carcinoma (HCC), also called malignant hepatoma, is one of the deadliest cancers due to its complexities, reoccurrence after surgical resection, metastasis and heterogeneity. Incidence and mortality of HCC are increasing in Western countries and are expected to rise as a consequence of the obesity epidemic. Multiple factors trigger the initiation and progression of HCC including chronic alcohol consumption, viral hepatitis B and C infection, metabolic disorders and age. Although Sorafenib is the only FDA approved drug for the treatment of HCC, numerous treatment modalities such as transcatheter arterial chemoembolization/transarterial chemoembolization (TACE), radiotherapy, locoregional therapy and chemotherapy have been tested in the clinics. Polymeric nanoparticles, liposomes, and micelles carrying small molecules, proteins, peptides and nucleic acids have attracted great attention for the treatment of various cancers including HCC. Herein, we discuss the pathogenesis of HCC in relation to its various recent treatment methodologies using nanodelivery of monoclonal antibodies (mAbs), small molecules, miRNAs and peptides. Synopsis of recent clinical trials of mAbs and peptide drugs has been presented with a broad overview of the pathogenesis of the disease and treatment efficacy.
Collapse
Affiliation(s)
- Rinku Dutta
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States.
| |
Collapse
|
82
|
Noveiry BB, Hirbod-Mobarakeh A, Khalili N, Hourshad N, Greten TF, Abou-Alfa GK, Rezaei N. Specific immunotherapy in hepatocellular cancer: A systematic review. J Gastroenterol Hepatol 2017; 32:339-351. [PMID: 27206802 PMCID: PMC6377153 DOI: 10.1111/jgh.13449] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/07/2016] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND AIM In recent years, several novel immunotherapeutic approaches were developed and investigated in patients with hepatocellular carcinoma (HCC). We designed this systematic review, to evaluate clinical efficacy of specific immunotherapy in patients with HCC, according to the guidelines of Border of Immune Tolerance Education and Research Network (BITERN) and Cochrane collaboration. METHODS We searched Medline, Scopus, CENTRAL, TRIP, DART, OpenGrey, and ProQuest through the 9th of December 2015. One author reviewed and retrieved citations from these seven databases for irrelevant and duplicate studies, and two other authors independently extracted data from the studies and rated their quality. We collated study findings and calculated a weighted treatment effect across studies using Review Manager. RESULTS We found 12144 references in seven databases of which 21 controlled studies with 1885 HCC patients in different stages were included in this systematic review after the primary and secondary screenings. Overall, patients undergoing specific immunotherapy had significantly higher overall survival than those in control group (HR = 0.59; 95% CI = 0.47-0.76, P < 0.0001). There was a significant difference in recurrence-free survival between patients undergoing specific immunotherapy and patients in control groups and patients in immunotherapy groups overall had less recurrence than control group (HR = 0.54; 95% CI = 0.46-0.63, P < 0.00001). CONCLUSIONS Results of this systematic review based on the available literature suggest that overall specific immunotherapeutic approaches could be beneficiary for the treatment of patients with HCC. This further supports the current and ongoing evaluations of specific immunotherapies in the field.
Collapse
Affiliation(s)
- Behnoud Baradaran Noveiry
- Border of Immune Tolerance Education and Research Network (BITERN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Armin Hirbod-Mobarakeh
- Border of Immune Tolerance Education and Research Network (BITERN), Universal Scientific Education and Research Network (USERN), Tehran, Iran,Molecular Immunology Research Center, Department of Immunology, School of Medicine, Tehran, Iran,Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nastaran Khalili
- Border of Immune Tolerance Education and Research Network (BITERN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloufar Hourshad
- Border of Immune Tolerance Education and Research Network (BITERN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Ghassan K Abou-Alfa
- Memorial Sloan Kettering Cancer Center, New York, USA,Weill Cornell Medical College, New York, USA
| | - Nima Rezaei
- Border of Immune Tolerance Education and Research Network (BITERN), Universal Scientific Education and Research Network (USERN), Tehran, Iran,Molecular Immunology Research Center, Department of Immunology, School of Medicine, Tehran, Iran,Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran,Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Sheffield, UK
| |
Collapse
|
83
|
Yang N, Li S, Li G, Zhang S, Tang X, Ni S, Jian X, Xu C, Zhu J, Lu M. The role of extracellular vesicles in mediating progression, metastasis and potential treatment of hepatocellular carcinoma. Oncotarget 2017; 8:3683-3695. [PMID: 27713136 PMCID: PMC5356911 DOI: 10.18632/oncotarget.12465] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 09/28/2016] [Indexed: 12/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related death worldwide. As vectors for intercellular information exchange, the potential role of extracellular vesicles (EVs) in HCC formation, progression and therapy has been widely investigated. In this review, we explore the current status of the researches in this field. Altogether there is undeniable evidence that EVs play a crucial role in HCC development, metastasis. Moreover, EVs have shown great potential as drug delivery systems (DDSs) for the treatment of HCC. Exosomal miRNAs derived from HCC cells can enhance transformed cell growth in recipient cells by modulating the expression of transforming growth factor-β activated kinase-1(TAK1) and downstream signaling molecules. Furthermore, vacuolar protein sortin 4 homolog A(VPS4A) and insulin-like growth factor(IGF)-1 regulate exosome-mediated miRNAs transfer. Immune cells- derived EVs containing integrin αMβ2 or CD147 may facilitate HCC metastasis. In addition, EVs-mediated shuttle of long non-coding RNAs (lncRNAs), specifically linc- VLDLR and linc-ROR promote chemoresistance of malignant cells. Heat shock proteins (HSPs)-harboring exosomes derived from HCC tumor cells increase the antitumor effect of natural killer (NK) cells, thus enhancing HCC immunotherapy. Indeed, inhibition of HCC tumor growth has been associated with tumor cell-derived exosomes (TEX)-pulsed dentritic cells (DCs). Exosomes are also essential in liver metastasis during colorectal carcinoma (CRC) and pancreatic ductal adenocarcinomas (PDAC). Therefore, as nucleic acid and drug delivery vehicles, EVs show a tremendous potential for effective treatment against HCC.
Collapse
Affiliation(s)
- Naibin Yang
- Department of Infection and Liver Diseases, Ningbo First Hospital, Ningbo, China
| | - Shanshan Li
- Department of Infection and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Institute of Liver Research, Wenzhou Medical University, Wenzhou, China
| | - Guoxiang Li
- Department of Infection and Liver Diseases, Ningbo First Hospital, Ningbo, China
| | - Shengguo Zhang
- Department of Infection and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Institute of Liver Research, Wenzhou Medical University, Wenzhou, China
| | - Xinyue Tang
- Department of Infection and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Institute of Liver Research, Wenzhou Medical University, Wenzhou, China
| | - Shunlan Ni
- Department of Infection and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Institute of Liver Research, Wenzhou Medical University, Wenzhou, China
| | - Xiaomin Jian
- Department of The First Clinical Medical, Wenzhou Medical University, Wenzhou, China
| | - Cunlai Xu
- Department of Respiration, Lishui People's Hospital of Wenzhou Medical University, Lishui, China
| | - Jiayin Zhu
- Laboratory Animal Center, Wenzhou Medical University, Wenzhou, China
| | - Mingqin Lu
- Department of Infection and Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Institute of Liver Research, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
84
|
Diabetes mellitus increases the risk of intrahepatic recurrence of hepatocellular carcinoma after surgical resection. TUMORI JOURNAL 2017; 103:279-285. [PMID: 28085178 DOI: 10.5301/tj.5000594] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2016] [Indexed: 12/13/2022]
Abstract
AIMS AND BACKGROUND The relationship between cancer and metabolism has recently been receiving attention. We investigated the prognostic influence of type 2 diabetes mellitus in patients with hepatocellular carcinoma (HCC) treated with curative resection. METHODS AND STUDY DESIGN The records of 58 patients who underwent curative resection for HCC pT1-2N0M0 between 2010 and 2014 were reviewed retrospectively. Fourteen patients (24.1%) had diabetes mellitus at diagnosis. Local control (LC) was defined as time to recurrence in the liver. RESULTS The median follow-up was 23.3 months. Relapses occurred in 20 patients (34.5%) during the follow-up period; 17 of them developed intrahepatic recurrence, which was associated with diabetes mellitus (p = 0.013) and alpha fetoprotein (AFP) levels >500 ng/mL (p = 0.019). Overall relapses (n = 20) were related to T stage (p = 0.044), AFP level (p = 0.005), and diabetes (p = 0.044). The 3-year local control (intrahepatic control), disease-free survival, and overall survival rates were 56.7%, 50.5%, and 84.3%, respectively. LC was affected by diabetes mellitus (p = 0.046), Barcelona Clinic Liver Cancer staging (p<0.001), Milan criteria for transplantation (p = 0.041), serosal invasion (p = 0.032), and microvascular invasion (p = 0.043). Diabetes was also associated with reduced LC in the subgroup with hepatitis B-related HCC (n = 44, p = 0.028). CONCLUSIONS Diabetes mellitus is correlated with intrahepatic HCC recurrence after surgery. Greater attention should be paid to managing patients with HCC and diabetes mellitus.
Collapse
|
85
|
Successful chemoimmunotherapy against hepatocellular cancer in a novel murine model. J Hepatol 2017; 66:75-85. [PMID: 27520877 PMCID: PMC5167655 DOI: 10.1016/j.jhep.2016.07.044] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 07/11/2016] [Accepted: 07/26/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS We have established a clinically relevant animal model of hepatocellular cancer (HCC) in immune competent mice to elucidate the complex dialog between host immunity and tumors during HCC initiation and progression. Mechanistic findings have been leveraged to develop a clinically feasible anti-tumor chemoimmunotherapeutic strategy. METHODS Intraperitoneal injection of carbon tetrachloride and intrasplenic inoculation of oncogenic hepatocytes were combined to induce progressive HCCs in fibrotic livers of immunocompetent mice. Immunization and adoptive cell transfer (ACT) were used to dissect the tumor antigen-specific immune response. The ability of the tyrosine kinase inhibitor sunitinib to enhance immunotherapy in the setting of HCC was evaluated. RESULTS This new mouse model mimics human HCC and reflects its typical features. Tumor-antigen-specific CD8+ T cells maintained a naïve phenotype and remained responsive during early-stage tumor progression. Late tumor progression produced circulating tumor cells, tumor migration into draining lymph nodes, and profound exhaustion of tumor-antigen-specific CD8+ T cells associated with accumulation of programmed cell death protein 1 (PD-1)hi CD8+ T cells and regulatory T cells (Tregs). Sunitinib-mediated tumoricidal effect and Treg suppression synergized with antibody-mediated blockade of PD-1 to powerfully suppress tumor growth and activate anti-tumor immunity. CONCLUSION Treg accumulation and upregulation of PD-1 provide two independent mechanisms to induce profound immune tolerance in HCC. Chemoimmunotherapy using Food and Drug Administration-approved sunitinib with anti-PD-1 antibodies achieved significant tumor control, supporting translation of this approach for the treatment of HCC patients. LAY SUMMARY In the current study, we have established a clinically relevant mouse model which mimics human liver cancer. Using this unique model, we studied the response of the immune system to this aggressive cancer. Findings from this trial have led to the development of an innovative and clinically feasible chemoimmunotherapeutic strategy.
Collapse
|
86
|
Abstract
Approximately 12% of all cancers worldwide are associated with viral infections. To date, eight viruses have been shown to contribute to the development of human cancers, including Epstein-Barr virus (EBV), Hepatitis B and C viruses, and Human papilloma virus, among others. These DNA and RNA viruses produce oncogenic effects through distinct mechanisms. First, viruses may induce sustained disorders of host cell growth and survival through the genes they express, or may induce DNA damage response in host cells, which in turn increases host genome instability. Second, they may induce chronic inflammation and secondary tissue damage favoring the development of oncogenic processes in host cells. Viruses like HIV can create a more permissive environment for cancer development through immune inhibition, but we will focus on the previous two mechanisms in this review. Unlike traditional cancer therapies that cannot distinguish infected cells from non-infected cells, immunotherapies are uniquely equipped to target virus-associated malignancies. The targeting and functioning mechanisms associated with the immune response can be exploited to prevent viral infections by vaccination, and can also be used to treat infection before cancer establishment. Successes in using the immune system to eradicate established malignancy by selective recognition of virus-associated tumor cells are currently being reported. For example, numerous clinical trials of adoptive transfer of ex vivo generated virus-specific T cells have shown benefit even for established tumors in patients with EBV-associated malignancies. Additional studies in other virus-associated tumors have also been initiated and in this review we describe the current status of immunotherapy for virus-associated malignancies and discuss future prospects.
Collapse
|
87
|
"Hepatocellular carcinoma: A life-threatening disease". Biomed Pharmacother 2016; 84:1679-1688. [PMID: 27823920 DOI: 10.1016/j.biopha.2016.10.078] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 10/13/2016] [Accepted: 10/26/2016] [Indexed: 02/07/2023] Open
Abstract
An estimated rise in liver cancer incidence will increase to 95374 new cases by 2020. Hepatocellular Carcinoma (HCC), the most common primary malignant tumour of the liver, is considered to be the third leading cause of all cancer-related deaths and fifth common cancer worldwide. The reported data shows that the rate of HCC incidence in male population is three to four times higher compared with the female population. In the United States, HCV-induced liver cancer is increasing very fast because of the lack of proper treatment option. There are various treatment strategies available for HCC like liver transplantation, resection, ablation, embolization and chemotherapy still the prognosis is destitute. If the patient is eligible, liver transplantation is the only therapeutic option that may give around 90% survival rate, but the scarcity of liver donor limits its broad applicability. A sudden address is necessary to develop specific drugs, personalized medicine, for HCC.
Collapse
|
88
|
Zhang Q, Zhang Z, Peng M, Fu S, Xue Z, Zhang R. CAR-T cell therapy in gastrointestinal tumors and hepatic carcinoma: From bench to bedside. Oncoimmunology 2016; 5:e1251539. [PMID: 28123893 PMCID: PMC5214859 DOI: 10.1080/2162402x.2016.1251539] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 10/17/2016] [Indexed: 12/13/2022] Open
Abstract
The chimeric antigen receptor (CAR) is a genetically engineered receptor that combines a scFv domain, which specifically recognizes the tumor-specific antigen, with T cell activation domains. CAR-T cell therapies have demonstrated tremendous efficacy against hematologic malignancies in many clinical trials. Recent studies have extended these efforts to the treatment of solid tumors. However, the outcomes of CAR-T cell therapy for solid tumors are not as remarkable as the outcomes have been for hematologic malignancies. A series of hurdles has arisen with respect to CAR-T cell-based immunotherapy, which needs to be overcome to target solid tumors. The major challenge for CAR-T cell therapy in solid tumors is the selection of the appropriate specific antigen to demarcate the tumor from normal tissue. In this review, we discuss the application of CAR-T cells to gastrointestinal and hepatic carcinomas in preclinical and clinical research. Furthermore, we analyze the usefulness of several specific markers in the study of gastrointestinal tumors and hepatic carcinoma.
Collapse
Affiliation(s)
- Qi Zhang
- Laboratory of Immunology and Inflammation, Department of Immunology and Research Center of Basic Medical Science, Tianjin Medical University, Tianjin, China; Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Molecular and Cellular Immunology, Tianjin Medical University, Tianjin, China
| | - Zimu Zhang
- Laboratory of Immunology and Inflammation, Department of Immunology and Research Center of Basic Medical Science, Tianjin Medical University, Tianjin, China; Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Molecular and Cellular Immunology, Tianjin Medical University, Tianjin, China
| | - Meiyu Peng
- Department of Immunology, Basic Medical College, Weifang Medical University , Weifang, China
| | - Shuyu Fu
- Laboratory of Immunology and Inflammation, Department of Immunology and Research Center of Basic Medical Science, Tianjin Medical University, Tianjin, China; Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Molecular and Cellular Immunology, Tianjin Medical University, Tianjin, China
| | - Zhenyi Xue
- Laboratory of Immunology and Inflammation, Department of Immunology and Research Center of Basic Medical Science, Tianjin Medical University, Tianjin, China; Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Molecular and Cellular Immunology, Tianjin Medical University, Tianjin, China
| | - Rongxin Zhang
- Laboratory of Immunology and Inflammation, Department of Immunology and Research Center of Basic Medical Science, Tianjin Medical University, Tianjin, China; Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Key Laboratory of Molecular and Cellular Immunology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
89
|
Kumar N, Biswas S, Mathew AE, Varghese S, Mathew JE, Nandakumar K, Aranjani JM, Lobo R. Pro-apoptotic and cytotoxic effects of enriched fraction of Elytranthe parasitica (L.) Danser against HepG2 Hepatocellular carcinoma. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:420. [PMID: 27782823 PMCID: PMC5080845 DOI: 10.1186/s12906-016-1395-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 10/13/2016] [Indexed: 02/07/2023]
Abstract
Background Hepatocellular carcinoma (HCC), the most common type of liver cancer accounts for more than one million deaths worldwide. Current treatment modality for HCC is marginally effective. Plants belonging to Mistletoe family (Loranthaceae) have been used in chemotherapy for many years. The present study was aimed at exploring the anti-proliferative, pro-oxidant and pro-apoptotic potential of stem of Elytranthe parasitica (L.) Danser (EP), a parasitic shrub belonging to Loranthaceae. Methods Elytranthe parasitica (L.) Danser, a climbing parasitic shrub was investigated for its cytotoxic activity against HepG2, a hepatocellular carcinoma cell line by Sulforhodamine B (SRB) assay. Further, pro-oxidant activity of EP extract/fractions was studied using copper phenanthroline assay. To understand the mechanism of cell death, the pro-apoptotic effects of Hep-G2 cells treated with EP extract/fractions were visualized by dual staining using acridine orange and ethidium bromide, a morphological marker of apoptosis. Phytochemical profiling of EP was explored by estimating the phenol, flavonoid and tannin content in its various fractions and extract. The occurrence of gallic acid, a principal polyphenol in EP extract and fractions was detected and further quantified using HPTLC (High Performance Thin Layer Chromatography) fingerprinting. Result Active fraction of Elytranthe parasitica, EP.DEE exhibited potent cytotoxic activity in a dose dependent manner against HepG2 hepatocellular carcinoma cell line with an IC50 of 56.7 ± 7.8 μg/mL. Dual staining with acridine orange and ethidium bromide revealed that HepG2 cells treated with EP active fractions underwent cell death chiefly by apoptosis. Highest phenol, flavonoid and tannin content were observed in active fractions, EP.EA (Ethyl acetate fraction) and EP.DEE (Diethyl ether fraction). Gallic acid was identified and quantified in EP extract and active fractions, EP.DEE and EP.EA. Conclusion Our findings indicate EP active fraction could be a promising contender in the treatment of hepatocellular carcinoma.
Collapse
|
90
|
Rao Q, Zuo B, Lu Z, Gao X, You A, Wu C, Du Z, Yin H. Tumor-derived exosomes elicit tumor suppression in murine hepatocellular carcinoma models and humans in vitro. Hepatology 2016; 64:456-472. [PMID: 26990897 DOI: 10.1002/hep.28549] [Citation(s) in RCA: 214] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 02/04/2016] [Accepted: 03/06/2016] [Indexed: 12/13/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) remains a global challenge due to high morbidity and mortality rates and poor response to treatment. Immunotherapy, based on introduction of dendritic cells (DCs) activated by tumor cell lysates as antigens ex vivo, shows limited response rates in HCC patients. Here, we demonstrate that tumor cell-derived exosomes (TEXs), displaying an array of HCC antigens, can elicit a stronger immune response than cell lysates in vitro and in vivo. Significant tumor growth inhibition was achieved in ectopic and orthotopic HCC mice treated with TEX-pulsed DCs. Importantly, the tumor immune microenvironment was significantly improved in orthotopic HCC mice treated by TEX-pulsed DCs, demonstrated by increased numbers of T lymphocytes, elevated levels of interferon-γ, and decreased levels of interleukin-10 and tumor growth factor-β in tumor sites. As expected, T cells played an essential role in the TEX-pulsed DC-mediated immune response. Notably, exosomes from HCC cells not only promoted HCC-specific cytolysis but also provided cross-protective effects against pancreatic cancer cells. Moreover, HCC-specific cytolysis, elicited by DCs pulsed with human HepG2 cell-derived exosomes, was observed across different human HCC cells irrespective of human leukocyte antigen types. CONCLUSION HCC TEXs can potently carry HCC antigens, trigger a strong DC-mediated immune response, and improve the HCC tumor microenvironment. (Hepatology 2016;64:456-472).
Collapse
Affiliation(s)
- Quan Rao
- Department of Cell Biology and Research Centre of Basic Medical Science, Tianjin Medical University, Heping District, Tianjin, China
- Third Central Clinical College, Tianjin Medical University, Hedong District, Tianjin, China
| | - Bingfeng Zuo
- Department of Cell Biology and Research Centre of Basic Medical Science, Tianjin Medical University, Heping District, Tianjin, China
| | - Zhen Lu
- Department of Cell Biology and Research Centre of Basic Medical Science, Tianjin Medical University, Heping District, Tianjin, China
| | - Xianjun Gao
- Department of Cell Biology and Research Centre of Basic Medical Science, Tianjin Medical University, Heping District, Tianjin, China
| | - Abin You
- Department of Cell Biology and Research Centre of Basic Medical Science, Tianjin Medical University, Heping District, Tianjin, China
- Tianjin Cancer Hospital, Tianjin Medical University, Hexi District, Tianjin, China
| | - Chenxuan Wu
- Third Central Clinical College, Tianjin Medical University, Hedong District, Tianjin, China
| | - Zhi Du
- Third Central Clinical College, Tianjin Medical University, Hedong District, Tianjin, China
- Department of Hepatobiliary Surgery, Key Laboratory of Artificial Cell, Institute for Hepatobiliary Diseases, Third Central Hospital, Tianjin Medical University, Hedong District, Tianjin, China
| | - HaiFang Yin
- Department of Cell Biology and Research Centre of Basic Medical Science, Tianjin Medical University, Heping District, Tianjin, China
| |
Collapse
|
91
|
Youness RA, Rahmoon MA, Assal RA, Gomaa AI, Hamza MT, Waked I, El Tayebi HM, Abdelaziz AI. Contradicting interplay between insulin-like growth factor-1 and miR-486-5p in primary NK cells and hepatoma cell lines with a contemporary inhibitory impact on HCC tumor progression. Growth Factors 2016; 34:128-140. [PMID: 27388576 DOI: 10.1080/08977194.2016.1200571] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/31/2016] [Indexed: 12/16/2022]
Abstract
In this study, an impaired natural killer (NK) cell cytolytic activity in 135 hepatocellular carcinoma (HCC) patients parallel to a reduced expression level of insulin-like growth factor (IGF)-1 in NK cells of HCC patients has been revealed. Ectopic expression of miR-486-5p, a direct upstream regulator of IGF-1, restored the endogenous level of IGF-1 in NK cells of HCC patients, thus augmenting its cytolytic activity against Huh7 cells in an opposite manner to the IGF-1 siRNAs. Unorthodoxly, over-expression of miR-486-5p in target hepatocytes resulted in the repression of IGF-1, suppression of Huh7 cells proliferation and viability in a similar pattern to the IGF-1 siRNAs. Therefore, this study highlights a potential role of IGF-1 in modulating cytolytic potential of NK cells of HCC patients. miR-486-5p acts in a cell-specific manner, differentially modulating IGF-1 expression in NK cells and their target hepatocytes with a contemporary inhibitory impact on HCC progression.
Collapse
Affiliation(s)
- Rana Ahmed Youness
- a Department of Pharmaceutical Biology , Faculty of Pharmacy and Biotechnology, German University in Cairo , Cairo , Egypt
| | - Mai Atef Rahmoon
- a Department of Pharmaceutical Biology , Faculty of Pharmacy and Biotechnology, German University in Cairo , Cairo , Egypt
| | - Reem Amr Assal
- b Department of Pharmacology and Toxicology , Faculty of Pharmacy and Biotechnology, German University in Cairo , Cairo , Egypt
| | - Asmaa Ibrahim Gomaa
- c Department of Hepatology , National Liver Institute, Menoufiya University , Shebin El-Kom , Egypt
| | - Mohamed Tarif Hamza
- d Department of Clinical Pathology , Ain Shams University , Cairo , Egypt , and
| | - Imam Waked
- c Department of Hepatology , National Liver Institute, Menoufiya University , Shebin El-Kom , Egypt
| | - Hend Mohamed El Tayebi
- b Department of Pharmacology and Toxicology , Faculty of Pharmacy and Biotechnology, German University in Cairo , Cairo , Egypt
| | | |
Collapse
|
92
|
Abstract
Hepatocellular cancer (HCC) is a leading cause of cancer death worldwide, and most patients who are diagnosed with HCC are ineligible for curative local therapy. The targeted agent sorafenib provides modest survival benefits in the setting of advanced disease. Novel systemic treatment options for HCC are sorely needed. In this review, we identify and categorize the drugs and targets that are in various phases of testing for use against HCC. We also focus on the potential for combining these agents with radiotherapy. This would help identify directions for future study that are likely to yield positive findings and improve outcomes for patients with HCC.
Collapse
Affiliation(s)
- Nitin Ohri
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY
| | - Andreas Kaubisch
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY
| | - Madhur Garg
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY
| | - Chandan Guha
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
93
|
Abstract
Fighting Cancer with Immunotherapy. 21st Century Cancer Warfare: A Glimpse into the Operations of a Modern Radiotherapy Unit. Is Colorectal Carcinomas due to ‘Bad Luck’ or Is It Preventable? Asian Point of View on Cancer. Improving Overall Survival in Hepatocellular Carcinoma through a Multi-Disciplinary Approach: Intra-Tumoral Heterogeneity, Immunology and the Promise of Better Outcomes. Cancer of the Cervix – Can It be Prevented?
Collapse
|
94
|
Tagliamonte M, Petrizzo A, Tornesello ML, Ciliberto G, Buonaguro FM, Buonaguro L. Combinatorial immunotherapy strategies for hepatocellular carcinoma. Curr Opin Immunol 2016; 39:103-113. [PMID: 26851637 DOI: 10.1016/j.coi.2016.01.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/12/2016] [Accepted: 01/12/2016] [Indexed: 12/18/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common liver malignancy. The prognosis for HCC patients greatly varies according to the stage at diagnosis. Overall it is poor, with a 5-year survival rate of approximately 5-6%. Immunotherapeutic interventions represent a novel and effective therapeutic tool. However, only few immunotherapy trials for HCC have been conducted so far with contrasting results, suggesting that significant improvements are needed. Indeed, the liver is characterized by a strong intrinsic immune suppressive microenvironment which needs to be counterbalanced with immune stimulatory approaches. Therefore, the implementation of combinatorial protocols combining immune stimulatory strategies with specific immunotherapy approaches could result in a dramatic improvement of efficacy and clinical outcome in HCC patients. The present review aims at describing the state of the art in immunotherapy strategies for HCC and future perspectives.
Collapse
Affiliation(s)
- Maria Tagliamonte
- Lab of Molecular Biology & Viral Oncology, Dept Experimental Oncology
| | | | | | - Gennaro Ciliberto
- Scientific Direction, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy
| | | | - Luigi Buonaguro
- Lab of Molecular Biology & Viral Oncology, Dept Experimental Oncology.
| |
Collapse
|
95
|
Tabibian JH, Varghese C, LaRusso NF, O'Hara SP. The enteric microbiome in hepatobiliary health and disease. Liver Int 2016; 36:480-487. [PMID: 26561779 PMCID: PMC4825184 DOI: 10.1111/liv.13009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/02/2015] [Indexed: 12/15/2022]
Abstract
Increasing evidence points to the contribution of the intestinal microbiome as a potentially key determinant in the initiation and/or progression of hepatobiliary disease. While current understanding of this dynamic is incomplete, exciting insights are continually being made and more are expected given the developments in molecular and high-throughput omics techniques. In this brief review, we provide a practical and updated synopsis of the interaction of the intestinal microbiome with the liver and its downstream impact on the initiation, progression and complications of hepatobiliary disease.
Collapse
Affiliation(s)
- James H. Tabibian
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA, USA
| | - Cyril Varghese
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Nicholas F. LaRusso
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Steven P. O'Hara
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
96
|
Butterfield LH. Lessons learned from cancer vaccine trials and target antigen choice. Cancer Immunol Immunother 2016; 65:805-12. [PMID: 26842127 DOI: 10.1007/s00262-016-1801-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/21/2016] [Indexed: 01/15/2023]
Abstract
A wide variety of tumor antigens have been targeted in cancer immunotherapy studies. Traditionally, the focus has been on commonly overexpressed antigens shared across many patients and/or tumor types. As the field has progressed, the identity of human tumor rejection antigens has broadened. Immunologic monitoring of clinical trials has slowly elucidated candidate biomarkers of immune response and clinical response, and conversely, of immune dysfunction and suppression. We have utilized MART-1/Melan-A in our melanoma studies and observed a high frequency of immune responses and several significant clinical responses in patients vaccinated with this melanosomal protein. Alpha-fetoprotein is a shared, overexpressed tumor antigen and secreted glycoprotein that we have tested in hepatocellular cancer vaccines. Our recent studies have identified immunosuppressive and immune-skewing activities of this antigen. The choice of target antigen and its form can have unexpected effects.
Collapse
Affiliation(s)
- Lisa H Butterfield
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Departments of Medicine, Surgery and Immunology, Hillman Cancer Center, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
97
|
Nguyen K, Jack K, Sun W. Hepatocellular Carcinoma: Past and Future of Molecular Target Therapy. Diseases 2015; 4:E1. [PMID: 28933381 PMCID: PMC5456309 DOI: 10.3390/diseases4010001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/03/2015] [Accepted: 12/16/2015] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common causes of cancer related mortality worldwide. The incidence of HCC has been increasing annually. Viral infection, alcohol usage, and other causes of cirrhosis have been identified as major risk factors for HCC development. The underlying pathogenesis has not been as well defined. There have been multiple hypotheses to the specific mechanisms of hepatocarcinogenesis and they share the common theme of chronic inflammation, increase oxidative stress, and genomic alteration. Therapeutic options of HCC have been primarily local and/or regional including transplantation, resection, and radial frequency ablation, chemoembolization or radio-embolization. For unresectable or metastatic disease, the options are limited. Conventional chemotherapeutic options have been noted to have limited benefit. Sorafenib has been the one and only systemic therapy which has demonstrated modest overall survival benefit. This has led to more extensive research with focus on targeted therapy. Numerous pre-clinical and early phase clinical studies have been noted but failed to show efficacy in later phase clinical trials. In an effort to identify new potential therapeutic options, new understanding of underlying pathways to hepatocarcinogenesis should be one of the main focuses. This leads to development of more molecularly targeted agents to specific pathways, and immunotherapy. This article provides a review of major studies of molecular targeted agents which attempts to target these specific pathways in HCC.
Collapse
Affiliation(s)
- Khanh Nguyen
- University of Pittsburgh Medical Center, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, 5150 Center Ave. 5th floor, Pittsburgh, PA 15232, USA.
| | - Kerri Jack
- University of Pittsburgh Medical Center, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, 5150 Center Ave. 5th floor, Pittsburgh, PA 15232, USA.
| | - Weijing Sun
- University of Pittsburgh Medical Center, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, 5150 Center Ave. 5th floor, Pittsburgh, PA 15232, USA.
| |
Collapse
|
98
|
New Tools for Molecular Therapy of Hepatocellular Carcinoma. Diseases 2015; 3:325-340. [PMID: 28943628 PMCID: PMC5548255 DOI: 10.3390/diseases3040325] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/15/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver cancer, arising from neoplastic transformation of hepatocytes or liver precursor/stem cells. HCC is often associated with pre-existing chronic liver pathologies of different origin (mainly subsequent to HBV and HCV infections), such as fibrosis or cirrhosis. Current therapies are essentially still ineffective, due both to the tumor heterogeneity and the frequent late diagnosis, making necessary the creation of new therapeutic strategies to inhibit tumor onset and progression and improve the survival of patients. A promising strategy for treatment of HCC is the targeted molecular therapy based on the restoration of tumor suppressor proteins lost during neoplastic transformation. In particular, the delivery of master genes of epithelial/hepatocyte differentiation, able to trigger an extensive reprogramming of gene expression, could allow the induction of an efficient antitumor response through the simultaneous adjustment of multiple genetic/epigenetic alterations contributing to tumor development. Here, we report recent literature data supporting the use of members of the liver enriched transcription factor (LETF) family, in particular HNF4α, as tools for gene therapy of HCC.
Collapse
|
99
|
Zhang L, Zhu W, Li J, Yang X, Ren Y, Niu J, Pang Y. Clinical outcome of immunotherapy with dendritic cell vaccine and cytokine-induced killer cell therapy in hepatobiliary and pancreatic cancer. Mol Clin Oncol 2015; 4:129-133. [PMID: 26870371 DOI: 10.3892/mco.2015.660] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 10/19/2015] [Indexed: 12/17/2022] Open
Abstract
The aim of this study was to determine the therapeutic effects of adoptive immunotherapy following dendritic cell (DC) vaccine and cytokine-induced killer (CIK) cell therapy and evaluate its cytotoxicity, survival benefits and quality of life (QOL) changes in patients with hepatobiliary and pancreatic cancer (HPC). We performed a retrospective analysis of 407 clinical cases, including 77 patients with HPC who received immunotherapy with DC vaccine and CIK cells (I group) and 330 patients with similar characteristics who underwent baseline treatment but did not receive immunotherapy [non-immunotherapy (NI) group)] as the control group. After a follow-up period of 294±207.5 days, the median survival time (MST) of the two groups was compared using the Kaplan-Meier method. In the I group, 61% of the patients developed a positive, delayed-type hypersensitivity response and 65% of the patients exhibited an improvement in QOL. The most notable adverse events included fever (28%), insomnia (25%), anorexia (17%), skin rash (12%) and arthralgia (31%). No severe toxicities were observed in patients in the I group; in addition, the MST was significantly longer in the I group compared with that in the NI group (P=0.014). Thus, the DC vaccine and CIK cell therapy was associated with mild adverse effects, but was able to induce an immune response and effectively eliminate tumor cells, thereby improving the QOL and prolonging the MST of the patients.
Collapse
Affiliation(s)
- Lihong Zhang
- School of Medicine, NanKai University, Tianjin 300071, P.R. China
| | - Wei Zhu
- Graduate School of Tianjin Medical University, Tianjin 300070, P.R. China
| | - Jiali Li
- Department of Oncology, Tianjin Union Medicine Centre, Tianjin 300121, P.R. China
| | - Xuejing Yang
- Department of Oncology, Tianjin Union Medicine Centre, Tianjin 300121, P.R. China
| | - Yanjie Ren
- Department of Oncology, Tianjin Union Medicine Centre, Tianjin 300121, P.R. China
| | - Jingxiu Niu
- Department of Oncology, Tianjin Union Medicine Centre, Tianjin 300121, P.R. China
| | - Yan Pang
- Department of Oncology, Tianjin Union Medicine Centre, Tianjin 300121, P.R. China
| |
Collapse
|
100
|
Bupathi M, Kaseb A, Meric-Bernstam F, Naing A. Hepatocellular carcinoma: Where there is unmet need. Mol Oncol 2015; 9:1501-9. [PMID: 26160430 DOI: 10.1016/j.molonc.2015.06.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 05/05/2015] [Accepted: 06/17/2015] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a complex and heterogeneous tumor most commonly associated with underlying chronic liver disease, especially hepatitis. It is a growing problem in the United States and worldwide. There are two potential ways to prevent HCC. Primary prevention which is based on vaccination or secondary prevention involving agents that slow down carcinogenesis. Several pathways have been thought to play a role in the development of HCC; specifically, those involving vascular endothelial growth factor (VEGF)-mediated angiogenesis, WNT, phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR), AMP-activated protein kinase (AMPK), and c-MET. Currently, there are only a limited number of drugs which have been proven as effective treatment options for HCC and several clinical trials are testing drugs which target aberrations in the pathways mentioned above. In this review, we discuss currently approved therapies, monotherapies and combination therapy for the treatment of HCC.
Collapse
Affiliation(s)
- Manojkumar Bupathi
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ahmed Kaseb
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|