1
|
Wu X, Jin B, Liu X, Mao Y, Wan X, Du S. Research trends of cellular immunotherapy for primary liver cancer: A bibliometric analysis. Hum Vaccin Immunother 2024; 20:2426869. [PMID: 39538378 PMCID: PMC11572085 DOI: 10.1080/21645515.2024.2426869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/21/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Cellular immunotherapy has shown considerable potential for the treatment of primary liver cancer (PLC), particularly hepatocellular carcinoma (HCC), although it is in the early stages of development. This study used bibliometric methods to delineate the evolution of research on cellular immunotherapy for PLC. Data were sourced from the Web of Science Core Collection (WoSCC) on April 22, 2024. Using the "Bibliometrix" R package, we examined primary bibliometric features, collaboration frequency between countries, and article output of the journals. Furthermore, we employed VOSviewer for coauthorship analysis and visualization and CiteSpace to assess keyword co-occurrence, as well as to spotlight keywords and references with the strongest citation bursts. Our analysis encompassed 492 publications focused on PLC and cellular immunotherapy, and we pinpointed China, Japan, and the USA as the foremost contributing nations and identified "Cancer Immunology Immunotherapy" as the journal with the most contributions in this area. Sun Yat-sen University emerged as the institution with the most significant output, and Li Zonghai authored the greatest number of leading articles. Prominent keywords that displayed a notable citation burst in the later years included "chimeric antigen receptor," "combination therapy", "CAR-T cells," "TCR-T cells," and "liver transplantation." This bibliometric study outlined a foundational knowledge framework, surveyed over three decades of research on cellular immunotherapy for PLC, and revealed the key players and trends, thereby offering a thorough understanding of the field, especially in relation to HCC.
Collapse
Affiliation(s)
- Xiang’an Wu
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, China
| | - Bao Jin
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao Liu
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, China
| | - Xueshuai Wan
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, China
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
2
|
Hsu CY, Mustafa MA, Kumar A, Pramanik A, Sharma R, Mohammed F, Jawad IA, Mohammed IJ, Alshahrani MY, Ali Khalil NAM, Shnishil AT, Abosaoda MK. Exploiting the immune system in hepatic tumor targeting: Unleashing the potential of drugs, natural products, and nanoparticles. Pathol Res Pract 2024; 256:155266. [PMID: 38554489 DOI: 10.1016/j.prp.2024.155266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 04/01/2024]
Abstract
Hepatic tumors present a formidable challenge in cancer therapeutics, necessitating the exploration of novel treatment strategies. In recent years, targeting the immune system has attracted interest to augment existing therapeutic efficacy. The immune system in hepatic tumors includes numerous cells with diverse actions. CD8+ T lymphocytes, T helper 1 (Th1) CD4+ T lymphocytes, alternative M1 macrophages, and natural killer (NK) cells provide the antitumor immunity. However, Foxp3+ regulatory CD4+ T cells (Tregs), M2-like tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) are the key immune inhibitor cells. Tumor stroma can also affect these interactions. Targeting these cells and their secreted molecules is intriguing for eliminating malignant cells. The current review provides a synopsis of the immune system components involved in hepatic tumor expansion and highlights the molecular and cellular pathways that can be targeted for therapeutic intervention. It also overviews the diverse range of drugs, natural products, immunotherapy drugs, and nanoparticles that have been investigated to manipulate immune responses and bolster antitumor immunity. The review also addresses the potential advantages and challenges associated with these approaches.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan City 71710, Taiwan
| | | | - Ashwani Kumar
- Department of Life Sciences, School of Sciences, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Atreyi Pramanik
- Institute of Pharma Sciences and Research, Chandigarh University, Mohali, India
| | - Rajiv Sharma
- Institute of Pharma Sciences and Research, Chandigarh University, Mohali, India
| | - Faraj Mohammed
- Department of Pharmacy, Al-Manara College for Medical Sciences, Maysan, Iraq
| | | | - Imad Jasim Mohammed
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia.
| | | | | | - Munther Kadhim Abosaoda
- College of technical engineering, the Islamic University, Najaf, Iraq; College of technical engineering, the Islamic University of Al Diwaniyah, Iraq; College of technical engineering, the Islamic University of Babylon, Iraq
| |
Collapse
|
3
|
Jiang S, Ma W, Ma C, Zhang Z, Zhang W, Zhang J. An emerging strategy: probiotics enhance the effectiveness of tumor immunotherapy via mediating the gut microbiome. Gut Microbes 2024; 16:2341717. [PMID: 38717360 PMCID: PMC11085971 DOI: 10.1080/19490976.2024.2341717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
The occurrence and progression of tumors are often accompanied by disruptions in the gut microbiota. Inversely, the impact of the gut microbiota on the initiation and progression of cancer is becoming increasingly evident, influencing the tumor microenvironment (TME) for both local and distant tumors. Moreover, it is even suggested to play a significant role in the process of tumor immunotherapy, contributing to high specificity in therapeutic outcomes and long-term effectiveness across various cancer types. Probiotics, with their generally positive influence on the gut microbiota, may serve as effective agents in synergizing cancer immunotherapy. They play a crucial role in activating the immune system to inhibit tumor growth. In summary, this comprehensive review aims to provide valuable insights into the dynamic interactions between probiotics, gut microbiota, and cancer. Furthermore, we highlight recent advances and mechanisms in using probiotics to improve the effectiveness of cancer immunotherapy. By understanding these complex relationships, we may unlock innovative approaches for cancer diagnosis and treatment while optimizing the effects of immunotherapy.
Collapse
Affiliation(s)
- Shuaiming Jiang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Wenyao Ma
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Chenchen Ma
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, Shenzhen, PR China
| | - Zeng Zhang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Wanli Zhang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Jiachao Zhang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| |
Collapse
|
4
|
Wu D, Li Y. Application of adoptive cell therapy in hepatocellular carcinoma. Immunology 2023; 170:453-469. [PMID: 37435926 DOI: 10.1111/imm.13677] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/20/2023] [Indexed: 07/13/2023] Open
Abstract
Hepatocellular carcinoma (HCC) remains a global health challenge. Novel treatment modalities are urgently needed to extend the overall survival of patients. The liver plays an immunomodulatory function due to its unique physiological structural characteristics. Therefore, following surgical resection and radiotherapy, immunotherapy regimens have shown great potential in the treatment of hepatocellular carcinoma. Adoptive cell immunotherapy is rapidly developing in the treatment of hepatocellular carcinoma. In this review, we summarize the latest research on adoptive immunotherapy for hepatocellular carcinoma. The focus is on chimeric antigen receptor (CAR)-T cells and T cell receptor (TCR) engineered T cells. Then tumour-infiltrating lymphocytes (TILs), natural killer (NK) cells, cytokine-induced killer (CIK) cells, and macrophages are briefly discussed. The main overview of the application and challenges of adoptive immunotherapy in hepatocellular carcinoma. It aims to provide the reader with a comprehensive understanding of the current status of HCC adoptive immunotherapy and offers some strategies. We hope to provide new ideas for the clinical treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Dengqiang Wu
- Department of Clinical Laboratory, Ningbo No. 6 Hospital, Ningbo, China
| | - Yujie Li
- Clinical Laboratory of Ningbo Medical Centre Lihuili Hospital, Ningbo University, Zhejiang, Ningbo, China
| |
Collapse
|
5
|
Yang F, Zheng X, Koh S, Lu J, Cheng J, Li P, Du C, Chen Y, Chen X, Yang L, Chen W, Wong RW, Wai LE, Wang T, Zhang Q, Chen W. Messenger RNA electroporated hepatitis B virus (HBV) antigen-specific T cell receptor (TCR) redirected T cell therapy is well-tolerated in patients with recurrent HBV-related hepatocellular carcinoma post-liver transplantation: results from a phase I trial. Hepatol Int 2023; 17:850-859. [PMID: 37067675 DOI: 10.1007/s12072-023-10524-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/16/2023] [Indexed: 04/18/2023]
Abstract
BACKGROUND AND AIMS Liver transplantation (LT) is the primary curative option for cirrhotic patients with early-stage hepatocellular carcinoma (HCC). However, tumor recurrence occurs in 15-20% of cases with unfavorable prognosis. We have developed a library of T cell receptors (TCRs) specific for different hepatitis B virus (HBV) antigens, restricted by different molecules of human leucocyte antigen (HLA)-class I, to redirect T cells against HBV antigens (Banu in Sci Rep 4:4166, 2014). We further demonstrated that these transiently functional T cells specific for HBV obtained through messenger RNA (mRNA) electroporation can eliminate HCC cells expressing HBV antigens in vitro and in vivo (Kah in J Clin Invest 127:3177-3188, 2017). A phase I clinical trial for patients with HCC recurrence post-liver transplant was conducted to assess the safety, tolerability, and anti-tumor efficacy of transiently functional HBV-TCR T cells. Here, we report the clinical findings with regard to the safety and anti-tumor efficacy of mRNA electroporated HBV-specific TCR-T cells. (ClinicalTrials.gov identifier: NCT02719782). PATIENTS AND METHODS A total of six patients with HBV-positive recurrent HCC post-liver transplant and HLA-matched to TCR targeting hepatitis B surface antigen (HBsAg) or hepatitis B core antigen (HBcAg) (HLA-A*02:01/HBsAg, HLA-A*11:01/HBcAg, HLA-B*58:01/HBsAg or HLA-C*08:01/HBsAg) were enrolled in this study. The primary objective was to assess the safety of short-lived mRNA electroporated HBV-TCR T cells based on the incidence and severity of the adverse event (AE) graded per National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE), Version 4.0. The secondary objective was to determine the effectiveness of HBV-TCR T cells as per RECIST 1.1 criteria. Patients were followed up for survival for 2 years post-end of treatment. RESULTS The median age of the six patients was 35.5 years (range: 28-47). The median number of HBV-TCR T cell infusions administered was 6.5 (range: 4-12). The treatment-related AE included grade 1 pyrexia. This study reported no cytokine release syndrome nor neurotoxicity. One patient remained alive and five were deceased at the time of the data cutoff (30 April 2020). CONCLUSION This study has demonstrated that multiple infusions of mRNA electroporated HBV-specific TCR T cells were well-tolerated in patients with HBV-positive recurrent HCC post-liver transplant.
Collapse
Affiliation(s)
- Fan Yang
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
- Department of Infectious Diseases, The First People's Hospital of Kashi, The Kashi Affiliated Hospital, Sun Yat-Sen University, Kashi, 844000, China
| | - Xiaofang Zheng
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Sarene Koh
- Lion TCR Pte Ltd, Singapore, Singapore
- Agency for Science and Technology (A*STAR), Singapore Immunology Network, Singapore, Singapore
| | - Jianxi Lu
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Jintao Cheng
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Panlong Li
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Cong Du
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yunhao Chen
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Xiaoyan Chen
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Li Yang
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Wanxin Chen
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | | | - Lu-En Wai
- Lion TCR Pte Ltd, Singapore, Singapore
- Agency for Science and Technology (A*STAR), Singapore Immunology Network, Singapore, Singapore
| | | | - Qi Zhang
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China.
- Department of Infectious Diseases, The First People's Hospital of Kashi, The Kashi Affiliated Hospital, Sun Yat-Sen University, Kashi, 844000, China.
| | - Wenjie Chen
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China.
| |
Collapse
|
6
|
Einloth KR, Gayfield S, McMaster T, Didier A, Dworkin L, Creeden JF. The application, safety, and future of ex vivo immune cell therapies and prognosis in different malignancies. BIOIMPACTS : BI 2023; 13:439-455. [PMID: 38022382 PMCID: PMC10676524 DOI: 10.34172/bi.2023.27521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 04/11/2023] [Accepted: 04/26/2023] [Indexed: 12/01/2023]
Abstract
Introduction Immunotherapy has revolutionized how cancer is treated. Many of these immunotherapies rely on ex vivo expansion of immune cells, classically T cells. Still, several immunological obstacles remain, including tumor impermeability by immune cells and the immunosuppressive nature of the tumor microenvironment (TME). Logistically, high costs of treatment and variable clinical responses have also plagued traditional T cell-based immunotherapies. Methods To review the existing literature on cellular immunotherapy, the PubMed database was searched for publications using variations of the phrases "cancer immunotherapy", "ex vivo expansion", and "adoptive cell therapy". The Clinicaltrials.gov database was searched for clinical trials related to ex vivo cellular therapies using the same phrases. The National Comprehensive Cancer Network guidelines for cancer treatment were also referenced. Results To circumvent the challenges of traditional T cell-based immunotherapies, researchers have developed newer therapies including tumor infiltrating lymphocyte (TIL), chimeric antigen receptor (CAR), T cell receptor (TCR) modified T cell, and antibody-armed T cell therapies. Additionally, newer immunotherapeutic strategies have used other immune cells, including natural killer (NK) and dendritic cells (DC), to modulate the T cell immune response to cancers. From a prognostic perspective, circulating tumor cells (CTC) have been used to predict cancer morbidity and mortality. Conclusion This review highlights the mechanism and clinical utility of various types of ex vivo cellular therapies in the treatment of cancer. Comparing these therapies or using them in combination may lead to more individualized and less toxic chemotherapeutics.
Collapse
Affiliation(s)
- Katelyn R. Einloth
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Scott Gayfield
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Thomas McMaster
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Alexander Didier
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Lance Dworkin
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Justin Fortune Creeden
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
7
|
Villarruel-Melquiades F, Mendoza-Garrido ME, García-Cuellar CM, Sánchez-Pérez Y, Pérez-Carreón JI, Camacho J. Current and novel approaches in the pharmacological treatment of hepatocellular carcinoma. World J Gastroenterol 2023; 29:2571-2599. [PMID: 37213397 PMCID: PMC10198058 DOI: 10.3748/wjg.v29.i17.2571] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/19/2023] [Accepted: 04/11/2023] [Indexed: 05/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignant tumours worldwide. The mortality-to-incidence ratio is up to 91.6% in many countries, representing the third leading cause of cancer-related deaths. Systemic drugs, including the multikinase inhibitors sorafenib and lenvatinib, are first-line drugs used in HCC treatment. Unfortunately, these therapies are ineffective in most cases due to late diagnosis and the development of tumour resistance. Thus, novel pharmacological alternatives are urgently needed. For instance, immune checkpoint inhibitors have provided new approaches targeting cells of the immune system. Furthermore, monoclonal antibodies against programmed cell death-1 have shown benefits in HCC patients. In addition, drug combinations, including first-line treatment and immunotherapy, as well as drug repurposing, are promising novel therapeutic alternatives. Here, we review the current and novel pharmacological approaches to fight HCC. Preclinical studies, as well as approved and ongoing clinical trials for liver cancer treatment, are discussed. The pharmacological opportunities analysed here should lead to significant improvement in HCC therapy.
Collapse
Affiliation(s)
- Fernanda Villarruel-Melquiades
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - María Eugenia Mendoza-Garrido
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Claudia M García-Cuellar
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Yesennia Sánchez-Pérez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Julio Isael Pérez-Carreón
- Instituto Nacional de Medicina Genómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico
| | - Javier Camacho
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico
| |
Collapse
|
8
|
Wang XF, Liu DL, Geng L. The PSCA rs2294008 (C/T) Polymorphism Increases the Risk of Gastric and Bladder Cancer: A Meta-Analysis. Genet Test Mol Biomarkers 2023; 27:44-55. [PMID: 36853840 DOI: 10.1089/gtmb.2022.0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
Background: It has been reported that prostate stem cell antigen (PSCA) is overexpressed in certain cancer types and confers poor prognoses. The rs2294008 (C/T) polymorphism of PSCA is considered to be associated with risk for gastric, bladder, and colorectal cancers; however, these studies have produced inconsistent results, so we performed this meta-analysis to verify the association between the PSCA rs2294008 (C/T) polymorphism and cancer risk. Methods: A systematic literature search was performed using PubMed, EMBASE, and the Chinese National Knowledge Infrastructure, through October 20, 2022 according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. We used odds ratios (ORs) with 95% confidence intervals (CIs) to assess the strength of the association between the PSCA rs2294008 (C/T) polymorphism and cancer risk. In addition, we explored PSCA mRNA expression in cancers through online databases. Results: In total, 45 articles met our inclusion criteria and were analyzed, including 37,586 cancer cases and 51,197 non-cancer controls. Except in the recessive model, the pooled effect indicated the PSCA rs2294008 T allele was associated with an increased overall cancer risk (T vs. C: OR = 1.120, 95% CI = 1.056-1.188, p < 0.01; TT vs. CC: OR = 1.206, 95% CI = 1.066-1.364, p = 0.03; CT vs. CC: OR = 1.249, 95% CI = 1.151-1.356, p < 0.01; [CT+TT] vs. CC: OR = 1.248, 95% CI = 1.147-1.359, p < 0.01; TT vs. [CT+CC]: OR = 1.051, 95% CI = 0.954-1.156, p = 0.314). In the subgroup analysis, there were significant associations between the rs2294008 T allele and increased risk of bladder and gastric cancer. Two different online tools were used to explore the PSCA mRNA levels in cancer and the corresponding normal adjacent tissues. We found that expression of PSCA was significantly lower in gastric cancer patients. Conclusions: The PSCA rs2294008 T polymorphism is related to increased cancer susceptibility, especially for gastric and bladder cancers. This polymorphism results in a decreased PSCA expression level in gastric cancer.
Collapse
Affiliation(s)
- Xiao-Feng Wang
- Department of Medical Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dong-Li Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Ministry of Education, Shanghai, China
| | - Li Geng
- Department of Medical Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Ahmad M, Dhasmana A, Harne PS, Zamir A, Hafeez BB. Chemokine clouding and liver cancer heterogeneity: Does it impact clinical outcomes? Semin Cancer Biol 2022; 86:1175-1185. [PMID: 35189322 DOI: 10.1016/j.semcancer.2022.02.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 02/08/2023]
Abstract
Tumor heterogeneity is a predominant feature of hepatocellular carcinoma (HCC) that plays a crucial role in chemoresistance and limits the efficacy of available chemo/immunotherapy regimens. Thus, a better understanding regarding the molecular determinants of tumor heterogeneity will help in developing newer strategies for effective HCC management. Chemokines, a sub-family of cytokines are one of the key molecular determinants of tumor heterogeneity in HCC and are involved in cell survival, growth, migration, and angiogenesis. Herein, we provide a panoramic insight into the role of chemokines in HCC heterogeneity at genetic, epigenetic, metabolic, immune cell composition, and tumor microenvironment levels and its impact on clinical outcomes. Interestingly, our in-silico analysis data showed that expression of chemokine receptors impacts infiltration of various immune cell populations into the liver tumor and leads to heterogeneity. Thus, it is evident that aberrant chemokines clouding impacts HCC tumor heterogeneity and understanding this phenomenon in depth could be harnessed for the development of personalized medicine strategies in future.
Collapse
Affiliation(s)
- Mudassier Ahmad
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, TX 78504, United States
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, TX 78504, United States; Department of Biosciences and Cancer Research Institute, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, India
| | - Prateek Suresh Harne
- DHR Health Gastroenterology, 5520 Leonardo da Vinci Drive, Suite 100, Edinburg, TX 78539, United States
| | - Asif Zamir
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, TX 78504, United States; DHR Health Gastroenterology, 5520 Leonardo da Vinci Drive, Suite 100, Edinburg, TX 78539, United States
| | - Bilal Bin Hafeez
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, TX 78504, United States; Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, TX 78504, United States.
| |
Collapse
|
10
|
Khan AA, Liu ZK, Xu X. Recent advances in immunotherapy for hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int 2021; 20:511-520. [PMID: 34344612 DOI: 10.1016/j.hbpd.2021.06.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 06/22/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Treatment of hepatocellular carcinoma (HCC) is challenging as most patients are diagnosed at advanced stage with underlying chronic liver conditions. Conventional systemic chemotherapy has failed in HCC, and the clinical efficacy of FDA-approved molecular targeted agents such as sorafenib and lenvatinib remains unsatisfactory. DATA SOURCES Literature search was conducted in PubMed for relevant articles published before January 2021. The search aimed to identify recent developments in immune-based treatment approaches for HCC. Information of clinical trials was obtained from https://clinicaltrials.gov/. RESULTS Two immune checkpoint inhibitors (ICIs), nivolumab and pembrolizumab were approved as monotherapies, which has revolutionized HCC treatment. Besides, combination ICIs have also got accelerated FDA approval recently. Immune-based therapies have challenged targeted drugs owing to their safety, tolerability, and survival benefits. In addition to the significant success in ICIs, other immunotherapeutic strategies such as cancer vaccine, chimeric antigen receptor T-cells, natural killer cells, cytokines, and combination therapy, have also shown promising outcomes in clinical trials. Various diagnostic and prognostic biomarkers have been identified which can help in clinical decision making when starting treatment with ICIs. CONCLUSIONS Immunotherapy has emerged as one of the mainstream treatment modalities for advanced HCC in recent years. However, challenges such as low response rate and acquired resistance in previously respondent patients still exist. Further research is needed to understand the unique resistance mechanism to immunotherapy and to discover more predictive biomarkers to guide clinical decision making.
Collapse
Affiliation(s)
- Abid Ali Khan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Lab of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Ministry of Public Health, Hangzhou 310003, China
| | - Zhi-Kun Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Lab of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Ministry of Public Health, Hangzhou 310003, China
| | - Xiao Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Lab of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Ministry of Public Health, Hangzhou 310003, China; Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
11
|
Bo XW, Sun LP, Yu SY, Xu HX. Thermal ablation and immunotherapy for hepatocellular carcinoma: Recent advances and future directions. World J Gastrointest Oncol 2021; 13:1397-1411. [PMID: 34721773 PMCID: PMC8529921 DOI: 10.4251/wjgo.v13.i10.1397] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/30/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of most common cancers that cause death in the world. Thermal ablation (TA) is an important alternative treatment method for HCC patients who are not appropriate for surgery or liver transplantation. Particularly for small and early HCCs, TA can be considered as the first-line curative treatment. However, local and distant recurrence rates are still high even though the TA equipment and technology develop rapidly. Immunotherapy is a novel systemic treatment method to enhance the anti-tumor immune response of HCC patients, which has the potential to reduce the tumor recurrence and metastasis. The combination of local TA and systemic immunotherapy for HCCs may be an ideal treatment for enhancing the efficacy of TA and controlling the recurrence. Herein we summarize the latest progress in TA, immunotherapy, and their combination for the treatment of patients with HCC and discuss the limitations and future research directions of the combined therapy.
Collapse
Affiliation(s)
- Xiao-Wan Bo
- Center of Minimally Invasive Treatment for Tumor, Department of Medical Ultrasound, Shanghai Tenth People’s Hospital; Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai 200072, China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai 200072, China
| | - Li-Ping Sun
- Center of Minimally Invasive Treatment for Tumor, Department of Medical Ultrasound, Shanghai Tenth People’s Hospital; Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai 200072, China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai 200072, China
| | - Song-Yuan Yu
- Center of Minimally Invasive Treatment for Tumor, Department of Medical Ultrasound, Shanghai Tenth People’s Hospital; Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai 200072, China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai 200072, China
| | - Hui-Xiong Xu
- Center of Minimally Invasive Treatment for Tumor, Department of Medical Ultrasound, Shanghai Tenth People’s Hospital; Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai 200072, China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment; National Clinical Research Center for Interventional Medicine, Shanghai 200072, China
| |
Collapse
|
12
|
Greten TF, Abou-Alfa GK, Cheng AL, Duffy AG, El-Khoueiry AB, Finn RS, Galle PR, Goyal L, He AR, Kaseb AO, Kelley RK, Lencioni R, Lujambio A, Mabry Hrones D, Pinato DJ, Sangro B, Troisi RI, Wilson Woods A, Yau T, Zhu AX, Melero I. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of hepatocellular carcinoma. J Immunother Cancer 2021; 9:e002794. [PMID: 34518290 PMCID: PMC8438858 DOI: 10.1136/jitc-2021-002794] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2021] [Indexed: 12/11/2022] Open
Abstract
Patients with advanced hepatocellular carcinoma (HCC) have historically had few options and faced extremely poor prognoses if their disease progressed after standard-of-care tyrosine kinase inhibitors (TKIs). Recently, the standard of care for HCC has been transformed as a combination of the immune checkpoint inhibitor (ICI) atezolizumab plus the anti-vascular endothelial growth factor (VEGF) antibody bevacizumab was shown to offer improved overall survival in the first-line setting. Immunotherapy has demonstrated safety and efficacy in later lines of therapy as well, and ongoing trials are investigating novel combinations of ICIs and TKIs, in addition to interventions earlier in the course of disease or in combination with liver-directed therapies. Because HCC usually develops against a background of cirrhosis, immunotherapy for liver tumors is complex and oncologists need to account for both immunological and hepatological considerations when developing a treatment plan for their patients. To provide guidance to the oncology community on important concerns for the immunotherapeutic care of HCC, the Society for Immunotherapy of Cancer (SITC) convened a multidisciplinary panel of experts to develop a clinical practice guideline (CPG). The expert panel drew on the published literature as well as their clinical experience to develop recommendations for healthcare professionals on these important aspects of immunotherapeutic treatment for HCC, including diagnosis and staging, treatment planning, immune-related adverse events (irAEs), and patient quality of life (QOL) considerations. The evidence- and consensus-based recommendations in this CPG are intended to give guidance to cancer care providers treating patients with HCC.
Collapse
Affiliation(s)
- Tim F Greten
- Thoracic and GI Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Ghassan K Abou-Alfa
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Medical College at Cornell University, New York, New York, USA
| | - Ann-Lii Cheng
- Department of Medical Oncology, National Taiwan University Cancer Center and National Taiwan University Hospital, Taipei, Taiwan
| | - Austin G Duffy
- The Mater Hospital/University College Dublin, Dublin, Ireland
| | - Anthony B El-Khoueiry
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Richard S Finn
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | | - Lipika Goyal
- Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Aiwu Ruth He
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Ahmed O Kaseb
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Robin Kate Kelley
- Department of Medicine (Hematology/Oncology), UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
| | - Riccardo Lencioni
- Department of Radiology, University of Pisa School of Medicine, Pisa, Italy
- Miami Cancer Institute, Miami, Florida, USA
| | - Amaia Lujambio
- Oncological Sciences Department, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Donna Mabry Hrones
- Thoracic and GI Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - David J Pinato
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Bruno Sangro
- Clinica Universidad de Navarra-Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | | | - Andrea Wilson Woods
- Blue Faery: The Adrienne Wilson Liver Cancer Association, Birmingham, Alabama, USA
| | - Thomas Yau
- Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong
| | - Andrew X Zhu
- Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
- Jiahui Health, Jiahui International Cancer Center, Shanghai, China
| | - Ignacio Melero
- Clinica Universidad de Navarra-Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Foundation for Applied Medical Research (FIMA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
13
|
The State of Immunotherapy in Hepatobiliary Cancers. Cells 2021; 10:cells10082096. [PMID: 34440865 PMCID: PMC8393650 DOI: 10.3390/cells10082096] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/02/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatobiliary cancers, including hepatocellular carcinoma (HCC), cholangiocarcinoma (CCA), and gallbladder carcinoma (GBC), are lethal cancers with limited therapeutic options. Curative-intent treatment typically involves surgery, yet recurrence is common and many patients present with advanced disease not amenable to an operation. Immunotherapy represents a promising approach to improve outcomes, but the immunosuppressive tumor microenvironment of the liver characteristic of hepatobiliary cancers has hampered the development and implementation of this therapeutic approach. Current immunotherapies under investigation include immune checkpoint inhibitors (ICI), the adoptive transfer of immune cells, bispecific antibodies, vaccines, and oncolytic viruses. Programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are two ICIs that have demonstrated utility in HCC, and newer immune checkpoint targets are being tested in clinical trials. In advanced CCA and GBC, PD-1 ICIs have resulted in antitumor responses, but only in a minority of select patients. Other ICIs are being investigated for patients with CCA and GBC. Adoptive transfer may hold promise, with reports of complete durable regression in metastatic CCA, yet this therapeutic approach may not be generalizable. Alternative approaches have been developed and promising results have been observed, but clinical trials are needed to validate their utility. While the treatment of hepatobiliary cancers involves unique challenges that these cancers present, the progress seen with ICIs and adoptive transfer has solidified immunotherapy as an important approach in these challenging patients with few other effective treatment options.
Collapse
|
14
|
Woeste MR, Geller AE, Martin RCG, Polk HC. Optimizing the Combination of Immunotherapy and Trans-Arterial Locoregional Therapy for Stages B and C Hepatocellular Cancer. Ann Surg Oncol 2021; 28:1499-1510. [PMID: 33393028 DOI: 10.1245/s10434-020-09414-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC), the most common primary hepatic malignancy worldwide, is the second leading cause of cancer-related death. Underlying liver dysfunction and advanced stage of disease require treatments to be optimally timed and implemented to minimize hepatic parenchymal damage while maximizing disease response and quality of life. Locoregional therapies (LRTs) such as trans-arterial chemo- and radio-embolization remain effective for intermediate liver-only and advanced HCC disease (i.e., Barcelona-Clinic liver cancer stages B and C) not amendable to primary resection or ablation. Additionally, these minimally invasive interventions have been shown to augment the immune system. This and the recent success of immune-oncologic treatments for HCC have generated interest in applying these therapies in combination with such locoregional interventions to improve patient outcomes and response rates. This report reviews the use of trans-arterial LRTs with immunotherapy for stages B and C HCC, potential biomarkers, and imaging methods for assessing the response and safety of such combinations.
Collapse
Affiliation(s)
- Matthew R Woeste
- Division of Surgical Oncology, Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA.,Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Anne E Geller
- Division of Surgical Oncology, Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA.,Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Robert C G Martin
- Division of Surgical Oncology, Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA.
| | - Hiram C Polk
- Division of Surgical Oncology, Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
15
|
Wang J, Feng L, Zhang L. Combining cellular immunotherapy was an optional choice for unresectable advanced HCC: A systematic review and meta-analysis. Clin Res Hepatol Gastroenterol 2021; 45:101440. [PMID: 32709504 DOI: 10.1016/j.clinre.2020.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The efficacy of cellular immunotherapy in advanced hepatocellular carcinoma (HCC) was controversial. This study was conducted to compare the effectiveness of combining cellular immunotherapy with that of incurable treatment alone. METHODS The Ovid Medline, Embase, Cochrane Library and Pubmed were systematically searched due to January 8th 2020. The keywords include "immunotherapy", "HCC" and study type. Treatment response was evaluated and progression-free survival (PFS) and overall survival (OS) were calculated using hazard ratio (HR) and 95% confidence interval (CI). RESULTS A total of 19 studies with 1275 patients were included in the meta-analysis. The median complete response rate (CR) was 19% in combining cellular immunotherapy comparing to 9% in the control group (RR=0.55, P=0.003). No significant difference was found in partial response and stable disease (RR=1.06 and 0.78, P>0.05, respectively). The progression disease rate was higher in the non-cellular immunotherapy group (31%) compared to the cellular immunotherapy group (RR=2.20, P=0.002). Patients treating with cellular immunotherapy had a better OS and PFS compared to those without cellular immunotherapy (HR=0.52 and 0.63, P<0.001). In the subgroup analysis, the only CIK infusion therapy and combined DC with CIK perfusion therapy patients had a better OS (HR=0.52 and 0.49, P<0.001 and P=0.002, respectively). CONCLUSION Our results suggested that combining use of cellular immunotherapy in advanced HCC could increase the complete response rate, and thereafter extend the progression-free and overall survival rate. Subgroup analysis suggested that combining use of CIK and DC or using CIK alone could provide the benefit in survival outcome.
Collapse
Affiliation(s)
- Jing Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003 Shandong, People's Republic of China.
| | - Lingxin Feng
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003 Shandong, People's Republic of China
| | - Linwei Zhang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266003 Shandong, People's Republic of China
| |
Collapse
|
16
|
Immunotherapy for Hepatocellular Carcinoma: A 2021 Update. Cancers (Basel) 2020; 12:cancers12102859. [PMID: 33020428 PMCID: PMC7600093 DOI: 10.3390/cancers12102859] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/26/2020] [Accepted: 09/30/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of one of the most frequent liver cancers and the fourth leading cause of cancer-related mortality worldwide. Current treatment options such as surgery, neoadjuvant chemoradiotherapy, liver transplantation, and radiofrequency ablation will benefit only a very small percentage of patients. Immunotherapy is a novel treatment approach representing an effective and promising option against several types of cancer. The aim of our study is to present the currently ongoing clinical trials and to evaluate the efficacy of immunotherapy in HCC. In this paper, we demonstrate that combination of different immunotherapies or immunotherapy with other modalities results in better overall survival (OS) and progression-free survival (PFS) compared to single immunotherapy agent. Another objective of this paper is to demonstrate and highlight the importance of tumor microenvironment as a predictive and prognostic marker and its clinical implications in immunotherapy response.
Collapse
|
17
|
Du H, Yang J, Zhang Y. Cytokine-induced killer cell/dendritic cell combined with cytokine-induced killer cell immunotherapy for treating advanced gastrointestinal cancer. BMC Cancer 2020; 20:357. [PMID: 32345239 PMCID: PMC7189715 DOI: 10.1186/s12885-020-06860-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 04/13/2020] [Indexed: 01/10/2023] Open
Abstract
Background This study aimed to investigate the efficacy and safety of cytokine-induced killer (CIK)/dendritic cell combined with CIK (DC–CIK) cell therapy in advanced gastrointestinal cancer (GIC). Methods The PubMed, Cochrane library, and Embase were searched to conduct a meta-analysis of clinical controlled trials to evaluate the efficacy and safety of CIK/DC–CIK cell therapy in advanced GIC. The pooled risk ratios (RRs) or weighted mean difference (WMD) with 95% confidence intervals (95% CIs) were calculated. Results A total of nine studies with 1113 patients were identified. The overall survival (RR = 1.84, 95% CI = 1.41–2.40, Pheterogeneity = 0.654, I2 = 0%), progression-free survival (RR = 1.99, 95% CI = 1.52–2.60, Pheterogeneity = 0.727, I2 = 0%), and quality of life (WMD = 16.09, 95% CI = 1.66–30.52, Pheterogeneity < 0.001, I2 = 98.8%) were significantly improved in patients who received chemotherapy combined with CIK/DC–CIK cells, and no severe adverse events were reported. Conclusion This meta-analysis suggested that the combination of CIK/DC–CIK immunotherapy and chemotherapy was safe and applicable for patients with advanced GIC. It is a feasible choice to prolong survival and improve quality of life.
Collapse
Affiliation(s)
- Hansong Du
- Department of Gastrointestinal Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Yang
- Department of Gastrointestinal Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Zhang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
18
|
Wang J, Shen T, Wang Q, Zhang T, Li L, Wang Y, Fang Y. The long-term efficacy of cytokine-induced killer cellular therapy for hepatocellular carcinoma: a meta-analysis. Immunotherapy 2019; 11:1325-1335. [PMID: 31578914 DOI: 10.2217/imt-2019-0079] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Aim: The long-term efficacy of cytokine-induced killer cellular therapy for hepatocellular carcinoma patients after curative treatments remains controversial. Methods: A meta-analysis was conducted, and the outcomes were the recurrence rate and overall survival. Results: Eight randomized clinical trials with 1038 participants were included. Compared with the control group, cytokine-induced killer cellular therapy group could reduce 1-year, 3-year recurrence rates, as well as improve 1-5 years overall survival for hepatocellular carcinoma patients (p < 0.05). However, it failed to affect the 5-year recurrence rate and 6-year overall survival (p > 0.05). Conclusion: Cytokine-induced killer cellular adjuvant therapy exerted a favorable role in improving early and long-term efficacy for hepatocellular carcinoma patients.
Collapse
Affiliation(s)
- Jiaxue Wang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,Department of Pharmacy Administration & Clinical Pharmacy, School of Pharmaceutical, Peking University, Beijing, China
| | - Tiantian Shen
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Qi Wang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,Department of Pharmacy Administration & Clinical Pharmacy, School of Pharmaceutical, Peking University, Beijing, China
| | - Tan Zhang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,Department of Pharmacy Administration & Clinical Pharmacy, School of Pharmaceutical, Peking University, Beijing, China
| | - Lujin Li
- The Center for Drug Clinical Research of Shanghai University of TCM, Shanghai, China
| | - Yu Wang
- Immunotech Applied Science Limited, Beijing, China
| | - Yi Fang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| |
Collapse
|
19
|
Liu C, Cui X, Zhou D, Li C, Zhao M, Jin Y, Ding C, Zhu Y. Cytokine-induced killer cells co-cultured with non-cell derived targeting peptide-loaded dendritic cells induce a specific antitumor response. Cancer Biol Ther 2019; 20:720-728. [PMID: 30777479 DOI: 10.1080/15384047.2018.1564561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cancer is a severe lethal disease. Currently, immunotherapy has become an effective alternative therapeutic approach for cancers. Cytokine-induced killer (CIK) cells have a higher proliferation rate, increased efficacy with few side-effects, and non-MHC-restricted killing after co-culturing with dendritic cells (DCs). Therefore, it has been widely studied and applied in the treatment of cancers. In our study, we explored the antitumor effects of CIK cells co-culturing with DCs pulsed with non-cell derived targeting peptides, which could specifically bind to certain tumor cells. Our results indicated that targeting peptide-loaded DCs could enhance the differentiation and cytotoxicity of CIK cells. Moreover, CIK cells, which were treated with specific targeting peptide-loaded DCs, could effectively and specifically kill tumor cells in vitro and in vivo, as long as tumor cells were pre-coated with the specific binding peptides. In conclusion, targeting peptides could guide DC-CIK to effectively and specifically kill tumor cells which were pre-coated with these targeting peptides and non-cell derived targeting peptide-loaded-DC-CIK may work as a novel means for cancer therapy.
Collapse
Affiliation(s)
- Cuijuan Liu
- a School of Nano Technology and Nano Bionics , University of Science and Technology of China , Hefei , China.,b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China
| | - Xueyuan Cui
- b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China.,c College of Life Sciences , Shanghai University , Shanghai , China
| | - Dayong Zhou
- d Department of Vascular Surgery , Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital , Suzhou , China
| | - Chunlin Li
- b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China
| | - Mengya Zhao
- b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China.,c College of Life Sciences , Shanghai University , Shanghai , China
| | - Yaqing Jin
- b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China
| | - Chen Ding
- b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China.,e China Pharmaceutical University , Nanjing , China
| | - Yimin Zhu
- b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China
| |
Collapse
|
20
|
Kang S, Li Y, Bao Y, Li Y. High-affinity T cell receptors redirect cytokine-activated T cells (CAT) to kill cancer cells. Front Med 2019; 13:69-82. [PMID: 30725257 DOI: 10.1007/s11684-018-0677-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 11/16/2018] [Indexed: 12/16/2022]
Abstract
Cytokine-activated T cells (CATs) can be easily expanded and are widely applied to cancer immunotherapy. However, the good efficacy of CATs is rarely reported in clinical applications because CATs have no or very low antigen specificity. The low-efficacy problem can be resolved using T cell antigen receptor-engineered CAT (TCR-CAT). Herein, we demonstrate that NY-ESO-1157-165 HLA-A*02:01-specific high-affinity TCR (HAT)-transduced CATs can specifically kill cancer cells with good efficacy. With low micromolar range dissociation equilibrium constants, HAT-transduced CATs showed good specificity with no off-target killing. Furthermore, the high-affinity TCR-CATs delivered significantly better activation and cytotoxicity than the equivalent TCR-engineered T cells (TCR-Ts) in terms of interferon-γ and granzyme B production and in vitro cancer cell killing ability. TCR-CAT may be a very good alternative to the expensive TCR-T, which is considered an effective personalized cyto-immunotherapy.
Collapse
Affiliation(s)
- Synat Kang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanyan Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Yifeng Bao
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Yi Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
21
|
Huang F, Chen J, Lan R, Wang Z, Chen R, Lin J, Zhang L, Fu L. δ-Catenin peptide vaccines repress hepatocellular carcinoma growth via CD8 + T cell activation. Oncoimmunology 2018; 7:e1450713. [PMID: 30221043 DOI: 10.1080/2162402x.2018.1450713] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/01/2018] [Accepted: 03/06/2018] [Indexed: 12/30/2022] Open
Abstract
As classical therapy method of advanced hepatocellular carcinoma (HCC) is not effective enough, HCC immunotherapy is a hot spot for research in recent years. Although in recent years, immune checkpoint inhibitors are focused in cancer therapy, vaccines and adoptive cell therapy (ACT), as traditional immunotherapy methods for HCC are still promising. We found that δ-Catenin might be a new tumor-associated antigen for HCC, for it could be upregulated as a stress associated protein under hypoxia and irradiation treatment. δ-Catenin peptide vaccines could inhibit the growth of subcutaneous hepatocellular tumors in vivo. According to our work, δ-Catenin peptide vaccines could stimulate the activation of cytotoxic T lymphocytes (CTLs) and enhance the infiltration of CD8+ T cells into tumors. Moreover, δ-Catenin peptide vaccines could enhance the secretion of IFN-γ and the killing of tumor cells by T cells. Mechanistically, δ-Catenin peptide vaccines, presented by antigen-presenting cells to T cells, could enhance the activation of T cells via MAPK/ERK signaling and the transcriptional factors Eomes and T-bet. Our research results indicate new potential peptide vaccines, which can be applied in clinical HCC therapy.
Collapse
Affiliation(s)
- Fei Huang
- Central Lab, First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian Platform for Medical Research at First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian key Lab of Individualized Active Immunotherapy, Fuzhou, China.,Key Laboratory of Radiation Biology of Fujian Province Universities, Fuzhou, China
| | - Junying Chen
- Central Lab, First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian Platform for Medical Research at First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian key Lab of Individualized Active Immunotherapy, Fuzhou, China.,Key Laboratory of Radiation Biology of Fujian Province Universities, Fuzhou, China
| | - Ruilong Lan
- Central Lab, First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian Platform for Medical Research at First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian key Lab of Individualized Active Immunotherapy, Fuzhou, China.,Key Laboratory of Radiation Biology of Fujian Province Universities, Fuzhou, China
| | - Zeng Wang
- Central Lab, First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian Platform for Medical Research at First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian key Lab of Individualized Active Immunotherapy, Fuzhou, China.,Key Laboratory of Radiation Biology of Fujian Province Universities, Fuzhou, China
| | - Ruiqing Chen
- Central Lab, First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian Platform for Medical Research at First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian key Lab of Individualized Active Immunotherapy, Fuzhou, China.,Key Laboratory of Radiation Biology of Fujian Province Universities, Fuzhou, China
| | - Jingan Lin
- Central Lab, First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian Platform for Medical Research at First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian key Lab of Individualized Active Immunotherapy, Fuzhou, China.,Key Laboratory of Radiation Biology of Fujian Province Universities, Fuzhou, China
| | - Lurong Zhang
- Central Lab, First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian Platform for Medical Research at First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian key Lab of Individualized Active Immunotherapy, Fuzhou, China.,Key Laboratory of Radiation Biology of Fujian Province Universities, Fuzhou, China
| | - Lengxi Fu
- Central Lab, First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian Platform for Medical Research at First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian key Lab of Individualized Active Immunotherapy, Fuzhou, China.,Key Laboratory of Radiation Biology of Fujian Province Universities, Fuzhou, China
| |
Collapse
|
22
|
Wang M, Zhang L, Liu Z, Zhou J, Pan Q, Fan J, Zang R, Wang L. AGO1 may influence the prognosis of hepatocellular carcinoma through TGF-β pathway. Cell Death Dis 2018; 9:324. [PMID: 29487329 PMCID: PMC5832432 DOI: 10.1038/s41419-018-0338-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 01/09/2018] [Accepted: 01/23/2018] [Indexed: 12/13/2022]
Abstract
AGO1 is a major component of RNA-induced silencing complexes and plays a crucial role in solid tumors. The aim of our study was to investigate AGO1 functions in hepatocellular carcinoma (HCC). Using small interfering RNA, AGO1 functions were investigated in HCCLM3 cell lines. Cell proliferation, immigration, and invasion significantly decreased after AGO1 depletion using MTT, wound-healing, and transwell assay. The associated proteins in the epithelial–mesenchymal transition (EMT) and the activation of its signal pathways were measured using western blot. After AGO1 depleted, increased E-cadherin and decreased N-cadherin, Vimentin, Snail, and Zeb1 were founded. In its upstream pathway, the phosphorylation of ERK1/2(Thr202/Tyr204), Smad2(S425/250/255), and Smad4 were significantly inhibited. Meanwhile, inhibitor of ERK1/2(LY3214996) significantly inhibited the growth and migration of the AGO1 cells. The nuclear importing of Smad4 was blocked and furthermore, the transcription of Snail was also influenced for the decrease of combination between Smad4 and the promotor region of Snail. After Snail was overexpressed, the invasion of HCCLM3 cells was significantly rescued. Immunohistochemistry in tissue microarrays consisting of 200 HCC patients was used to analyze the associations between AGO1 expression and prognosis. Intratumoral AGO1 expression was an independent risk factor for overall survival (P = 0.008) and recurrence-free survival (P < 0.001). In conclusion, AGO1 may promote HCC metastasis through TGF-β pathway, and AGO1 may be a reliable prognostic factor in HCC.
Collapse
Affiliation(s)
- Miao Wang
- Department of Liver Surgery, Shanghai Cancer Center, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, China
| | - Lyu Zhang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zeyang Liu
- Department of Liver Surgery, Shanghai Cancer Center, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, China
| | - Jiamin Zhou
- Department of Liver Surgery, Shanghai Cancer Center, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, China
| | - Qi Pan
- Department of Liver Surgery, Shanghai Cancer Center, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, China
| | - Jia Fan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Rongyu Zang
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China.
| | - Lu Wang
- Department of Liver Surgery, Shanghai Cancer Center, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, China.
| |
Collapse
|
23
|
Kong DS, Nam DH, Kang SH, Lee JW, Chang JH, Kim JH, Lim YJ, Koh YC, Chung YG, Kim JM, Kim CH. Phase III randomized trial of autologous cytokine-induced killer cell immunotherapy for newly diagnosed glioblastoma in Korea. Oncotarget 2018; 8:7003-7013. [PMID: 27690294 PMCID: PMC5351686 DOI: 10.18632/oncotarget.12273] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/23/2016] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Adoptive cell immunotherapy involves an ex vivo expansion of autologous cytokine-induced killer (CIK) cells before their reinfusion into the host. We evaluated the efficacy and safety of CIK cell immunotherapy with radiotherapy-temozolomide (TMZ) for the treatment of newly diagnosed glioblastomas. EXPERIMENTAL DESIGN In this multi-center, open-label, phase 3 study, we randomly assigned patients with newly diagnosed glioblastoma to receive CIK cell immunotherapy combined with standard TMZ chemoradiotherapy (CIK immunotherapy group) or standard TMZ chemoradiotherapy alone (control group). The efficacy endpoints were analyzed in the intention-to-treat set and in the per protocol set. RESULTS Between December 2008 and October 2012, a total of 180 patients were randomly assigned to the CIK immunotherapy (n = 91) or control group (n = 89). In the intention-to-treat analysis set, median PFS was 8.1 months (95% confidence interval (CI), 5.8 to 8.5 months) in the CIK immunotherapy group, as compared to 5.4 months (95% CI, 3.3 to 7.9 months) in the control group (one-sided log-rank, p = 0.0401). Overall survival did not differ significantly between two groups. Grade 3 or higher adverse events, health-related quality of life and performance status between two groups did not show a significant difference. CONCLUSIONS The addition of CIK cells immunotherapy to standard chemoradiotherapy with TMZ improved PFS. However, the CIK immunotherapy group did not show evidence of a beneficial effect on overall survival.
Collapse
Affiliation(s)
- Doo-Sik Kong
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Do-Hyun Nam
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Shin-Hyuk Kang
- Department of Neurosurgery, College of Medicine, Korea University, Seoul, Korea
| | - Jae Won Lee
- Department of Statistics, Korea University, Seoul, Korea
| | - Jong-Hee Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong-Hoon Kim
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Young-Jin Lim
- Department of Neurosurgery, Kyunghee University Hospital, Seoul, Korea
| | - Young-Cho Koh
- Department of Neurosurgery, Konkuk University Medical Center, Seoul, Korea
| | - Yong-Gu Chung
- Department of Neurosurgery, College of Medicine, Korea University, Seoul, Korea
| | - Jae-Min Kim
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Choong-Hyun Kim
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| |
Collapse
|
24
|
Lee S, Loecher M, Iyer R. Immunomodulation in hepatocellular cancer. J Gastrointest Oncol 2018; 9:208-219. [PMID: 29564186 PMCID: PMC5848038 DOI: 10.21037/jgo.2017.06.08] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/22/2017] [Indexed: 12/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fastest growing malignancy in the United States in relation to mortality. HCC relies on a complex immunosuppressive network to modify the host immune system and evade destruction. Intrinsic to the liver's function and anatomy, native hepatic and immune cells produce many inhibitory cytokines that promote tolerogenicity and limit immune response. Since the introduction of sorafenib in 2008, no treatment has been able to demonstrate improved survival in patients with advanced HCC post disease progression treated with sorafenib. More recent studies have shown that sorafenib has an immunomodulatory function in addition to inhibition of multiple tyrosine kinases. Clinical trials have aimed to further enhance this immunomodulatory function with other treatments, most promisingly immune checkpoint inhibitors. Additionally, ongoing studies are using combinatorial approaches with immunomodulatory treatment and liver directed therapies such as transarterial chemoembolization (TACE), radiofrequency ablation (RFA), microwave ablation (MWA), and cryoablation. This article will review recent data describing the immunosuppressive network in HCC, recent results of immunotherapies, and combinatorial approaches to treat advanced HCC.
Collapse
Affiliation(s)
- Sunyoung Lee
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
- University at Buffalo School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Matthew Loecher
- University at Buffalo School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Renuka Iyer
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
- University at Buffalo School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| |
Collapse
|
25
|
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: 21] [Impact Index Per Article: 3.0] [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
|
26
|
Xie Y, Huang L, Chen L, Lin X, Chen L, Zheng Q. Effect of dendritic cell-cytokine-induced killer cells in patients with advanced colorectal cancer combined with first-line treatment. World J Surg Oncol 2017; 15:209. [PMID: 29179719 PMCID: PMC5704402 DOI: 10.1186/s12957-017-1278-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/15/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Surgical resection combined with adjuvant chemotherapy is considered as the gold-standard treatment for advanced colorectal cancer patients. These patients have a poor 5-year survival rate of 5% or less. Furthermore, a large dose of chemotherapy can produce adverse side effects and severe toxicity. Therefore, this retrospective study aimed to evaluate the efficacy of dendritic cell-cytokine-induced killer (DC-CIK) cell infusion as an adjuvant therapy in patients with advanced colorectal cancer combined with first-line treatment. METHODS A total of 142 patients with stage III/IV colorectal carcinoma who had been treated with first-line therapy were included in this study. Among these patients, 71 patients received first-line treatment only (non-DC-CIK group), while the other 71 patients who had similar demographic and clinical characteristics received a DC-CIK cell infusion combined with first-line treatment (DC-CIK group). These patients were followed up until August 2014. Data were analyzed by Kaplan-Meier and Cox regression. RESULTS Our results showed that the 5-year overall survival (OS) rate for the DC-CIK group versus the non-DC-CIK group was 41.3 versus 19.4% (p = 0.001) and the 5-year progression-free survival (PFS) rate for the DC-CIK group versus the non-DC-CIK group was 57.4 versus 33.6% (p = 0.022). CONCLUSIONS Our results showed that patients with advanced colorectal cancer might benefit from DC-CIK immunotherapy combined with first-line therapy by significantly prolonging 5-year OS and PFS.
Collapse
Affiliation(s)
- Yunqing Xie
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Lijie Huang
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Luchuan Chen
- Department of Abdominal Surgery, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Xiaowei Lin
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Li Chen
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Qiuhong Zheng
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China.
| |
Collapse
|
27
|
Abstract
The encouraging results in immunotherapy for melanoma also led the way for translational and clinical research about immune-related mechanisms possibly relevant for gastrointestinal tumours. It is in fact now evident that the immune checkpoint modulation and in particular cell-mediated immune-response through programmed cell death-1 (PD-1) and the cytotoxic T-lymphocyte antigen-4 (CTLA4) receptors along with the regulatory T cells activity all have a relevant role in gastrointestinal cancers as well. This review aims to explore the state of the art of immunotherapy for gastrointestinal tumours, deepening recent scientific evidence regarding anti PD-1/PDL-1 and anti CTLA4 monoclonal antibodies, peptide based vaccine, DNA based vaccine, and pulsed dendritic cells, either alone or in combination with other antineoplastic medical therapy and locoregional treatments. Considering the non-negligible toxicity profile deriving from such a treatment approach, predictive biomarkers of response to immunotherapy in gastrointestinal cancer are also urgently needed in order to better select the patients' group with the highest likelihood of benefit.
Collapse
|
28
|
Zhang J, Wu N, Lian Z, Feng H, Jiang Q, Chen X, Gong J, Qiao Z. The Combined Antitumor Effects of 125I Radioactive Particle Implantation and Cytokine-Induced Killer Cell Therapy on Xenograft Hepatocellular Carcinoma in a Mouse Model. Technol Cancer Res Treat 2017; 16:1083-1091. [PMID: 29332456 PMCID: PMC5762075 DOI: 10.1177/1533034617732204] [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] [Indexed: 12/28/2022] Open
Abstract
The combination of radiotherapy and immunotherapy has shown great promise in eradicating tumors. For example, 125I radioactive particle implantation and cytokine-induced killer cell therapies have demonstrated efficacy in treating hepatocellular carcinoma. However, the mechanism of this combination therapy remains unknown. In this study, we utilized cytokine-induced killer cells obtained from human peripheral blood mononuclear cells along with 125I radioactive particle implantation to treat subcutaneous hepatocellular carcinoma xenograft tumors in BALB/c nude mice. The effects of combination therapy on tumor growth, tumor cell apoptosis and proliferation, animal survival, and immune indexes were then assessed. The results indicated that 125I radioactive particle implantation combined with cytokine-induced killer cells shows a much greater antitumor therapeutic effect than either of the therapies alone when compared to control treatments. Mice treated with a combination of radiotherapy and immunotherapy displayed significantly reduced tumor growth. 125I radioactive particle implantation upregulated the expression of major histocompatibility complex (MHC) class I chain-related gene A in hepatocellular carcinoma cells and enhanced cytokine-induced killer cell–mediated apoptosis through activation of caspase-3. Furthermore, cytokine-induced killer cells supplied immune substrates to induce a strong immune response after 125I radioactive particle implantation therapy. In conclusion, 125I radioactive particle implantation combined with cytokine-induced killer cell therapy significantly inhibits the growth of human hepatocellular carcinoma cells in vivo and improves animal survival times through mutual promotion of antitumor immunity, presenting a promising therapy for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Junyong Zhang
- 1 Chongqing Key Laboratory of Hepatobiliary Surgery, Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,2 Department of Urology Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Nian Wu
- 3 Department of General Surgery, the Fifth People's Hospital of Chongqing City, Chongqing, People's Republic of China
| | - Zhengrong Lian
- 4 Department of Clinical Epidemiology and Biostatistics, Population Health Research Institution, McMaster University, Hamilton, Ontario, Canada
| | - Huyi Feng
- 3 Department of General Surgery, the Fifth People's Hospital of Chongqing City, Chongqing, People's Republic of China
| | - Qing Jiang
- 2 Department of Urology Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xianfeng Chen
- 5 Department of Hepatobiliary Surgery, Fuling Center Hospital, Fuling District, Chongqing, People's Republic of China
| | - Jianping Gong
- 1 Chongqing Key Laboratory of Hepatobiliary Surgery, Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhengrong Qiao
- 6 Department of General Surgery, People's Hospital of Changshou District, Chongqing, People's Republic of China
| |
Collapse
|
29
|
Gao X, Mi Y, Guo N, Xu H, Xu L, Gou X, Jin W. Cytokine-Induced Killer Cells As Pharmacological Tools for Cancer Immunotherapy. Front Immunol 2017; 8:774. [PMID: 28729866 PMCID: PMC5498561 DOI: 10.3389/fimmu.2017.00774] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 06/19/2017] [Indexed: 12/31/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are a heterogeneous population of effector CD3+CD56+ natural killer T cells, which can be easily expanded in vitro from peripheral blood mononuclear cells. CIK cells work as pharmacological tools for cancer immunotherapy as they exhibit MHC-unrestricted, safe, and effective antitumor activity. Much effort has been made to improve CIK cells cytotoxicity and treatments of CIK cells combined with other antitumor therapies are applied. This review summarizes some strategies, including the combination of CIK with additional cytokines, dendritic cells, check point inhibitors, antibodies, chemotherapeutic agents, nanomedicines, and engineering CIK cells with a chimeric antigen receptor. Furthermore, we briefly sum up the clinical trials on CIK cells and compare the effect of clinical CIK therapy with other immunotherapies. Finally, further research is needed to clarify the pharmacological mechanism of CIK and provide evidence to formulate uniform culturing criteria for CIK expansion.
Collapse
Affiliation(s)
- Xingchun Gao
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China.,State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,Department of Instrument Science and Engineering, Institute of Nano Biomedicine and Engineering, Key Lab for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yajing Mi
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China.,State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Na Guo
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Hao Xu
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Lixian Xu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Weilin Jin
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China.,Department of Instrument Science and Engineering, Institute of Nano Biomedicine and Engineering, Key Lab for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, China.,National Centers for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
30
|
Chen JH, Xiang JY, Ding GP, Cao LP. Cholangiocarcinoma-derived exosomes inhibit the antitumor activity of cytokine-induced killer cells by down-regulating the secretion of tumor necrosis factor-α and perforin. J Zhejiang Univ Sci B 2017; 17:537-44. [PMID: 27381730 DOI: 10.1631/jzus.b1500266] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The aim of our study is to observe the impact of cholangiocarcinoma-derived exosomes on the antitumor activities of cytokine-induced killer (CIK) cells and then demonstrate the appropriate mechanism. METHODS Tumor-derived exosomes (TEXs), which are derived from RBE cells (human cholangiocarcinoma line), were collected by ultracentrifugation. CIK cells induced from peripheral blood were stimulated by TEXs. Fluorescence-activated cell sorting (FACS) was performed to determine the phenotypes of TEX-CIK and N-CIK (normal CIK) cells. The concentrations of tumor necrosis factor-α (TNF-α) and perforin in the culture medium supernatant were examined by using an enzyme-linked immunosorbent assay (ELISA) kit. A CCK-8 kit was used to evaluate the cytotoxic activity of the CIK cells to the RBE cell line. RESULTS The concentrations of TNF-α and perforin of the group TEX-CIK were 138.61 pg/ml and 2.41 ng/ml, respectively, lower than those of the group N-CIK 194.08 pg/ml (P<0.01) and 3.39 ng/ml (P<0.05). The killing rate of the group TEX-CIK was 33.35%, lower than that of the group N-CIK (47.35% (P<0.01)). The population of CD3(+), CD8(+), NK (CD56(+)), and CD3(+)CD56(+) cells decreased in the TEX-CIK group ((63.2±6.8)%, (2.5±1.0)%, (0.53±0.49)%, (0.45±0.42)%) compared with the N-CIK group ((90.3±7.3)%, (65.7±3.3)%, (4.2±1.2)%, (15.2±2.7)%), P<0.01. CONCLUSIONS Our results suggest that RBE cells-derived exosomes inhibit the antitumor activity of CIK cells by down-regulating the population of CD3(+), CD8(+), NK (CD56(+)), and CD3(+)CD56(+) cells and the secretion of TNF-α and perforin. TEX may play an important role in cholangiocarcinoma immune escape.
Collapse
Affiliation(s)
- Jiong-Huang Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Jian-Yang Xiang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Guo-Ping Ding
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Li-Ping Cao
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
31
|
Cellular and molecular targets for the immunotherapy of hepatocellular carcinoma. Mol Cell Biochem 2017; 437:13-36. [PMID: 28593566 DOI: 10.1007/s11010-017-3092-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/01/2017] [Indexed: 02/06/2023]
|
32
|
Longo V, Gnoni A, Gardini AC, Pisconti S, Licchetta A, Scartozzi M, Memeo R, Palmieri VO, Aprile G, Santini D, Nardulli P, Silvestris N, Brunetti O. Immunotherapeutic approaches for hepatocellular carcinoma. Oncotarget 2017; 8:33897-33910. [PMID: 28420805 PMCID: PMC5464921 DOI: 10.18632/oncotarget.15406] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/01/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a cancer with a high mortality rate due to the fact that the diagnosis usually occurs at anadvanced stage. Even in case of curative surgical treatment, recurrence is common. Sorafenib and regorafenib are the only therapeutic agents that have been demonstrated to be effective in advanced HCC, thus novel curative approaches are urgently needed. Recent studies focus on the role of immune system in HCC. In fact, the unique immune response in the liver favors tolerance, which can represent a real challenge for conventional immunotherapy in these patients. Spontaneous immune responses against tumor antigens have been detected, and new immune therapies are under investigation: dendritic cell vaccination, immune-modulator strategy, and immune checkpoint inhibition. In recent years different clinical trials examining the use of immunotherapy to treat HCC have been conducted with initial promising results. This review article will summarize the literature data concerning the potential immunotherapeutic approaches in HCC patients.
Collapse
Affiliation(s)
- Vito Longo
- Medical Oncology Unit, Hospital of Taranto, Taranto, Italy
| | - Antonio Gnoni
- Medical Oncology Unit, Hospital of Gallipoli, Gallipoli, Italy
| | - Andrea Casadei Gardini
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, FC, Italy
| | | | | | - Mario Scartozzi
- Department of Medical Oncology, University of Cagliari, Cagliari, Monserrato, CA, Italy
| | - Riccardo Memeo
- Department of Hepatobiliary Surgery, Ospedale Regionale “F.Miulli”, Strada Pr. Acquaviva - Santeramo, Bari, Italy
| | - Vincenzo Ostilio Palmieri
- Department of Biomedical Sciences and Human Oncology, Clinica Medica “A. Murri”, University of Bari Medical School, Bari, Italy
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo Hospital ULSS 6, Vicenza, Italy
| | - Daniele Santini
- Medical Oncology Unit, University Campus Biomedico, Rome, Italy
| | - Patrizia Nardulli
- Pharmacy Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, Viale Orazio Flacco, Bari, Italy
| | - Oronzo Brunetti
- Medical Oncology Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, Viale Orazio Flacco, Bari, Italy
| |
Collapse
|
33
|
Ryu JI, Han MH, Cheong JH, Kim JM, Kim CH. Current update of adoptive immunotherapy using cytokine-induced killer cells to eliminate malignant gliomas. Immunotherapy 2017; 9:411-421. [PMID: 28357913 DOI: 10.2217/imt-2017-0003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The therapeutic outcome for those with malignant glioma is poor, even though diverse therapeutic modalities have been developed. Immunotherapy has emerged as a therapeutic approach for malignant gliomas, making it possible to selectively treat tumors while sparing normal tissue. Here, we review clinical trials of adoptive immunotherapy approaches for malignant gliomas. We also describe a clinical trial that examined the efficacy and safety of autologous cytokine-induced killer (CIK) cells along with concomitant chemoradiotherapy for newly diagnosed glioblastoma. These CIK cells identify and kill autologous tumor cells. This review focuses on the use of adoptive immunotherapy for malignant gliomas and reviews the current literature on the concept of antitumor activity mediated by CIK cells.
Collapse
Affiliation(s)
- Je Il Ryu
- Department of Neurosurgery, Hanyang University Guri Hospital, 153 Gyeongchun-ro, Guri 471-701, Gyonggi-do, Republic of Korea
| | - Myung Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, 153 Gyeongchun-ro, Guri 471-701, Gyonggi-do, Republic of Korea
| | - Jin Hwan Cheong
- Department of Neurosurgery, Hanyang University Guri Hospital, 153 Gyeongchun-ro, Guri 471-701, Gyonggi-do, Republic of Korea
| | - Jae Min Kim
- Department of Neurosurgery, Hanyang University Guri Hospital, 153 Gyeongchun-ro, Guri 471-701, Gyonggi-do, Republic of Korea
| | - Choong Hyun Kim
- Department of Neurosurgery, Hanyang University Guri Hospital, 153 Gyeongchun-ro, Guri 471-701, Gyonggi-do, Republic of Korea
| |
Collapse
|
34
|
Yu R, Yang B, Chi X, Cai L, Liu C, Yang L, Wang X, He P, Lu X. Efficacy of cytokine-induced killer cell infusion as an adjuvant immunotherapy for hepatocellular carcinoma: a systematic review and meta-analysis. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:851-864. [PMID: 28360510 PMCID: PMC5364004 DOI: 10.2147/dddt.s124399] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This study was designed to evaluate the efficacy and safety of cytokine-induced killer (CIK) cell-based immunotherapy as an adjuvant therapy for hepatocellular carcinoma (HCC). Published studies were identified by searching Medline, Cochrane, EMBASE, and Google Scholar databases with the keywords: cytokine-induced killer cell, hepatocellular carcinoma, and immunotherapy. The outcomes of interest were overall survival, progression-free survival, and disease-free survival. Eight randomized controlled trials (RCTs), six prospective studies, and three retrospective studies were included. The overall analysis revealed that patients in the CIK cell-treatment group had a higher survival rate (pooled hazard ratio (HR) =0.594, 95% confidence interval [CI] =0.501–0.703, P<0.001). Patients treated with CIK cells in non-RCTs had a higher progression-free survival rate (pooled HR =0.613, 95% CI =0.510–0.738, P<0.001). However, CIK cell-treated patients in RCTs had progression-free survival rates similar to those of the control group (pooled HR =0.700, 95% CI =0.452–1.084, P=0.110). The comparison between pooled results of RCTs and non-RCTs regarding the progression-free survival rate did not reach statistical significance. Patients in the CIK cell-treatment group had lower rates of relapse in RCTs (pooled HR =0.635, 95% CI =0.514–0.784, P<0.001). Similar results were found when non-RCT and RCTs were pooled (pooled HR =0.623, 95% CI =0.516–0.752, P<0.001). Adjuvant CIK cell-based immunotherapy is a promising therapeutic approach that can improve overall survival and reduce recurrence in patients with HCC.
Collapse
Affiliation(s)
- Ruili Yu
- Department of Allergy, Beijing Shijitan Hospital, Affiliated to Capital Medical University
| | - Bo Yang
- Department of Geriatric Hematology, Chinese PLA General Hospital
| | - Xiaohua Chi
- Department of Pharmacy, Chinese PLA Rocket Force General Hospital
| | - Lili Cai
- Department of Geriatric Laboratory Medicine
| | - Cui Liu
- Department of Geriatric Ultrasound
| | - Lei Yang
- Medical Department, Nanlou Clinic, Chinese PLA General Hospital, Beijing
| | - Xueyan Wang
- Department of Allergy, Beijing Shijitan Hospital, Affiliated to Capital Medical University
| | - Peifeng He
- School of Medical Information Management, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xuechun Lu
- Department of Geriatric Hematology, Chinese PLA General Hospital
| |
Collapse
|
35
|
Aravalli RN, Steer CJ. Immune-Mediated Therapies for Liver Cancer. Genes (Basel) 2017; 8:E76. [PMID: 28218682 PMCID: PMC5333065 DOI: 10.3390/genes8020076] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/06/2017] [Accepted: 02/13/2017] [Indexed: 02/07/2023] Open
Abstract
In recent years, immunotherapy has gained renewed interest as an alternative therapeutic approach for solid tumors. Its premise is based on harnessing the power of the host immune system to destroy tumor cells. Development of immune-mediated therapies, such as vaccines, adoptive transfer of autologous immune cells, and stimulation of host immunity by targeting tumor-evasive mechanisms have advanced cancer immunotherapy. In addition, studies on innate immunity and mechanisms of immune evasion have enhanced our understanding on the immunology of liver cancer. Preclinical and clinical studies with immune-mediated therapies have shown potential benefits in patients with liver cancer. In this review, we summarize current knowledge and recent developments in tumor immunology by focusing on two main primary liver cancers: hepatocellular carcinoma and cholangiocarcinoma.
Collapse
Affiliation(s)
- Rajagopal N Aravalli
- Department of Electrical and Computer Engineering, University of Minnesota, 200 Union Street S.E., Minneapolis, MN 55455, USA.
| | - Clifford J Steer
- Departments of Medicine and Genetics, Cell Biology and Development, University of Minnesota, 420 Delaware Street S.E., Minneapolis, MN 55455, USA.
| |
Collapse
|
36
|
Wang H, Liu A, Bo W, Feng X, Hu Y, Tian L, Zhang H, Tang X. Adjuvant immunotherapy with autologous cytokine-induced killer cells for hepatocellular carcinoma patients after curative resection, a systematic review and meta-analysis. Dig Liver Dis 2016; 48:1275-1282. [PMID: 27481586 DOI: 10.1016/j.dld.2016.07.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 05/26/2016] [Accepted: 07/11/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cytokine-induced killer cells have been used as an adjuvant treatment for hepatocellular carcinoma with curative treatment. However, the outcomes remain controversial. AIM We conducted this meta-analysis to assess the safety and efficacy of cytokine-induced killer cells. METHODS Randomized controlled trials on cytokine-induced killer cells for hepatocellular carcinoma after curative treatments were identified by electronic searches. A meta-analysis was carried out to examine disease-free survival, overall survival rate and adverse effect. RESULTS Six randomized controlled trials with 844 patients (85.9% with hepatitis B or C) were included. Our meta-analysis showed that cytokine-induced killer cells can not only improve the 1-year (RR=1.23, P<0.001), 2-year (RR=1.37, P<0.001) and 3-year (RR=1.35, P=0.004) disease-free survival, but also improve the 1-year (RR=1.08, P=0.001), 2-year (RR=1.14, P<0.001) and 3-year (RR=1.15, P=0.02) overall survival. However, it failed to affect the 4-year and 5-year disease-free survival and overall survival (P>0.05). At the same time, cytokine-induced killer cells treatment was proved to be a safe strategy with the comparable adverse events comparing to the control group (P=0.39). CONCLUSIONS This review provides the best available evidence that adjuvant cytokine-induced killer cells treatment can be safely used to improve the early disease-free survival and survival of hepatitis B or C related hepatocellular carcinoma.
Collapse
Affiliation(s)
- Haiqing Wang
- Department of Hepato-Biliary-Pancreatic Surgery, Sichuan Cancer Hospital, Chengdu, Sichuan Province, China
| | - Aixiang Liu
- Department of Hepato-Biliary-Pancreatic Surgery, Sichuan Cancer Hospital, Chengdu, Sichuan Province, China
| | - Wentao Bo
- Department of Hepato-Biliary-Pancreatic Surgery, Sichuan Cancer Hospital, Chengdu, Sichuan Province, China
| | - Xielin Feng
- Department of Hepato-Biliary-Pancreatic Surgery, Sichuan Cancer Hospital, Chengdu, Sichuan Province, China.
| | - Yong Hu
- Department of Hepato-Biliary-Pancreatic Surgery, Sichuan Cancer Hospital, Chengdu, Sichuan Province, China
| | - Lang Tian
- Department of Hepato-Biliary-Pancreatic Surgery, Sichuan Cancer Hospital, Chengdu, Sichuan Province, China
| | - Hui Zhang
- Department of Hepato-Biliary-Pancreatic Surgery, Sichuan Cancer Hospital, Chengdu, Sichuan Province, China
| | - Xiaoli Tang
- Department of Hepato-Biliary-Pancreatic Surgery, Sichuan Cancer Hospital, Chengdu, Sichuan Province, China
| |
Collapse
|
37
|
Cytokine-induced killer cell infusion combined with conventional treatments produced better prognosis for hepatocellular carcinoma patients with barcelona clinic liver cancer B or earlier stage: A systematic review and meta-analysis. Cytotherapy 2016; 18:1525-1531. [PMID: 27746013 DOI: 10.1016/j.jcyt.2016.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/06/2016] [Indexed: 01/26/2023]
Abstract
BACKGROUND AIMS To investigate the clinical benefits of cytokine-induced killer (CIK) cell infusions on hepatocellular carcinoma (HCC) patients, combined with other conventional treatments. METHODS This was a systematic review and meta-analysis conducted among phase II and III randomized control trials worldwide. Review manager 5.2 version was used to pool the effect size across studies. Sensitivity analyses and risk of bias were estimated among included studies. Egger's test was used to characterize the publication bias. RESULTS Eight randomized controlled trials and 945 patients with HCC were included in the study. CIK infusion reduced cancer recurrence risk to 0.74 (95% confidence interval [CI] 0.5-0.92), I2 75% (P <0.001), and reduced cancer death risk to 0.76 (95% CI 0.65-0.88), I2 50% (P = 0.09). Among studies blinded for outcome assessment and Barcelona Clinic Liver Cancer stages of 0, A and B, CIK infusion reduced recurrence risk by 18% (relative risk [RR] = 0.82, 95% CI 0.70-0.96) and death risk by 37% (RR = 0.63, 95% CI 0.47-0.85); heterogeneity was 0% and 39%, respectively (P > 0.05). The intercepts of linear regressions for recurrence and death were -2.17 and -2.07, respectively, but the P value was 0.17 and 0.38; no significant publication bias was observed with Egger's test. DISCUSSION Among hepatocellular carcinoma patients with Barcelona Clinic Liver Cancer score of B or less, CIK cell infusions combined with conventional treatments significantly prolonged recurrence-free and overall survival. This adoptive immunotherapy could be recommended to HCC patients.
Collapse
|
38
|
Nwangwu CA, Weiher H, Schmidt-Wolf IGH. Increase of CIK cell efficacy by upregulating cell surface MICA and inhibition of NKG2D ligand shedding in multiple myeloma. Hematol Oncol 2016; 35:719-725. [PMID: 27430430 DOI: 10.1002/hon.2326] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 12/12/2022]
Abstract
Multiple myeloma, which is a monoclonal plasma cell malignancy, still remains incurable despite recent progress in our understanding of this disorder. Adoptive immunotherapy of multiple myeloma using cytokine-induced killer cells is yielding promising results in clinical trials; however, some myeloma cells still evade immune surveillance by various unknown molecular mechanisms. This study aims at increasing the efficacy of cytokine-induced killer cells in targeting this tumor, using selective small-molecule inhibitors which increase and stabilize surface expression of the natural killer group 2, member D ligand, major histocompatibility complex class I polypeptide-related sequence A (MICA) on myeloma cells. We treated 2 multiple myeloma cell lines-U266 and KMS-12-PE-with 3 drugs. One of these drugs (sodium butyrate) is a histone deacetylase inhibitor. Another drug which was used (matrix metalloproteinase inhibitor III) blocks ligand shedding while the third drug (phenylarsine oxide) obstructs surface ligand internalization. The effect of these drugs on cell viability was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, surface ligand expression was examined using flow cytometry, and ligand shedding was assessed using enzyme-linked immunosorbent assay. We demonstrated that cytokine-induced killer cells have increased cytotoxicity against multiple myeloma cells after combined drug treatment than without drug pretreatment. We also established that this increased cytotoxicity was due to potent upregulation and stabilization of surface MICA on the surface of these tumor cell lines. Our study thus highlights further therapeutic options which could be used for the treatment of multiple myeloma patients.
Collapse
Affiliation(s)
- Chidimma A Nwangwu
- Center for Integrated Oncology (CIO), Med. Klinik and Poliklinik III, University Hospital Bonn, Bonn, Germany.,Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Hans Weiher
- Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Ingo G H Schmidt-Wolf
- Center for Integrated Oncology (CIO), Med. Klinik and Poliklinik III, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
39
|
Chu H, Du F, Jiang L, Wang Z, Gong Z, Lian P, Li P, Chen J. The Efficacy of CIK-Based Immunotherapies for Advanced Solid Tumors. Technol Cancer Res Treat 2016; 16:577-585. [PMID: 27436839 DOI: 10.1177/1533034616659163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To investigate the efficacy of cytokine-induced killer cell-based immunotherapies in patients with advanced malignant solid tumors and the difference in clinical efficiency among 3 kinds of cytokine-induced killer cell-based immunotherapies. METHODS One hundred forty-six cases with advanced solid tumor, 230 cycles of cytokine-induced killer cell-based immunotherapies, were involved in this study. T-lymphocyte subsets, carcinoembryonic antigen, and adverse reactions were recorded. RESULTS CD3+ T lymphocyte, Th, NKT, and Th/Tc were increased after cytokine-induced killer cell-based treatment, from 55.67 ± 3.64 to 84.12 ± 5.15, 26.56 ± 4.47 to 42.76 ± 3.68, 1.82 ± 0.58 to 7.08 ± 0.92, 0.79 ± 3.64 to 1.35 ± 0.20, respectively ( P < .001). Carcinoembryonic antigen was decreased from 398.39 ± 219.16 to 127.26 ± 153.41 ( P < .001). Difference values were greater than 0 ( P < .001). Difference value of carcinoembryonic antigen was obviously less than 0 ( P < .001). There was no obvious difference in all variations between cytokine-induced killer cell and DC+CIK groups ( P > .05). The highest amount of CD3+ T lymphocyte and Th was recorded after at least 4 cycles of immunotherapy. And CD8+ T/CD4+ T also began to decrease after 4 cycles of immunotherapy. Difference value of T lymphocyte and Tc of patients with surgery is higher than that of patients without surgery. CONCLUSION Cytokine-induced killer cell-based immunotherapy is capable of increasing T-lymphocyte subsets, recovering cellular immunity without severe side effects, and is suitable for different kinds of solid cancer. Clinical efficiency of cytokine-induced killer cell-based immunotherapy is influenced by many factors such as surgery, stage.
Collapse
Affiliation(s)
- Hongjin Chu
- 1 The Central Laboratory, Yantai Yuhuangding Hospital, Affiliated Hospital of Medical College Qingdao University, Yantai, Shandong, People's Republic of China
| | - Fengcai Du
- 2 The First Clinical College of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Lixin Jiang
- 3 Department of Gastrointestinal Surgery, Yantai Yuhuangding Hospital, Affiliated Hospital of Medical College Qingdao University, Yantai, Shandong, People's Republic of China
| | - Zhixin Wang
- 1 The Central Laboratory, Yantai Yuhuangding Hospital, Affiliated Hospital of Medical College Qingdao University, Yantai, Shandong, People's Republic of China
| | - Zhaohua Gong
- 4 Department of Oncology, Yantai Yuhuangding Hospital, Affiliated Hospital of Medical College Qingdao University, Yantai, Shandong, People's Republic of China
| | - Peiwen Lian
- 1 The Central Laboratory, Yantai Yuhuangding Hospital, Affiliated Hospital of Medical College Qingdao University, Yantai, Shandong, People's Republic of China
| | - Peng Li
- 4 Department of Oncology, Yantai Yuhuangding Hospital, Affiliated Hospital of Medical College Qingdao University, Yantai, Shandong, People's Republic of China
| | - Jian Chen
- 1 The Central Laboratory, Yantai Yuhuangding Hospital, Affiliated Hospital of Medical College Qingdao University, Yantai, Shandong, People's Republic of China.,4 Department of Oncology, Yantai Yuhuangding Hospital, Affiliated Hospital of Medical College Qingdao University, Yantai, Shandong, People's Republic of China
| |
Collapse
|
40
|
Qi X, Zhao Y, Li H, Guo X, Han G. Management of hepatocellular carcinoma: an overview of major findings from meta-analyses. Oncotarget 2016; 7:34703-51. [PMID: 27167195 PMCID: PMC5085185 DOI: 10.18632/oncotarget.9157] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/11/2016] [Indexed: 02/07/2023] Open
Abstract
This paper aims to systematically review the major findings from meta-analyses comparing different treatment options for hepatocellular carcinoma (HCC). A total of 153 relevant papers were searched via the PubMed, EMBASE, and Cochrane library databases. They were classified according to the mainstay treatment modalities (i.e., liver transplantation, surgical resection, radiofrequency ablation, transarterial embolization or chemoembolization, sorafenib, and others). The primary outcome data, such as overall survival, diseases-free survival or recurrence-free survival, progression-free survival, and safety, were summarized. The recommendations and uncertainties regarding the treatment of HCC were also proposed.
Collapse
Affiliation(s)
- Xingshun Qi
- 1 Department of Gastroenterology and Meta-analysis Study Interest Group, General Hospital of Shenyang Military Area, Shenyang, 110840 China
- 2 Department of Liver Diseases and Digestive Interventional Radiology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032 China
| | - Yan Zhao
- 2 Department of Liver Diseases and Digestive Interventional Radiology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032 China
- 3 Department of Gastroenterology, First Affiliated Hospital of the Medical College, Xi'an Jiaotong University, Xi'an, 710000 China
| | - Hongyu Li
- 1 Department of Gastroenterology and Meta-analysis Study Interest Group, General Hospital of Shenyang Military Area, Shenyang, 110840 China
| | - Xiaozhong Guo
- 1 Department of Gastroenterology and Meta-analysis Study Interest Group, General Hospital of Shenyang Military Area, Shenyang, 110840 China
| | - Guohong Han
- 2 Department of Liver Diseases and Digestive Interventional Radiology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032 China
| |
Collapse
|
41
|
Yegin EG, Oymaci E, Karatay E, Coker A. Progress in surgical and nonsurgical approaches for hepatocellular carcinoma treatment. Hepatobiliary Pancreat Dis Int 2016; 15:234-56. [PMID: 27298100 DOI: 10.1016/s1499-3872(16)60097-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a complex and heterogeneous malignancy, frequently occurs in the setting of a chronically diseased organ, with multiple confounding factors making its management challenging. HCC represents one of the leading causes of cancer-related mortality globally with a rising trend of incidence in some of the developed countries, which indicates the need for better surgical and nonsurgical management strategies. DATA SOURCES PubMed database was searched for relevant articles in English on the issue of HCC management. RESULTS Surgical resection represents a potentially curative option for appropriate candidates with tumors detected at earlier stages and with well-preserved liver function. The long-term outcome of surgery is impaired by a high rate of recurrence. Surgical approaches are being challenged by local ablative therapies such as radiofrequency ablation and microwave ablation in selected patients. Liver transplantation offers potential cure for HCC and also correction of underlying liver disease, and minimizes the risk of recurrence, but is reserved for patients within a set of criteria proposed for a prudent allocation in the shortage of donor organs. Transcatheter locoregional therapies have become the palliative standard allowing local control for intermediate stage patients with noninvasive multinodular or large HCC who are beyond the potentially curative options. The significant survival benefit with the multikinase inhibitor sorafenib for advanced HCC has shifted the direction of research regarding systemic treatment toward molecular therapies targeting the disregulated pathways of hepatocarcinogenesis. Potential benefit is suggested from simultaneous or sequential multimodal therapies, and optimal combinations are being investigated. Despite the striking progress in preclinical studies of HCC immunotherapy and gene therapy, extensive clinical trials are required to achieve successful clinical applications of these innovative approaches. CONCLUSION Treatment decisions have become increasingly complex for HCC with the availability of multiple surgical and nonsurgical therapeutic options and require a comprehensive, multidisciplinary approach.
Collapse
Affiliation(s)
- Ender Gunes Yegin
- Department of Gastroenterology, Bozyaka Training and Research Hospital, Izmir 35170, Turkey.
| | | | | | | |
Collapse
|
42
|
Xu YC, Xu Q, Li JJ, Gu XF, Lin XL, Sun L, Lu HM, Tang L, Ma Y, Lu Z, Wang HX. Chemotherapy with or without autologous cytokine-induced killer cell transfusion as the first-line treatment for stage IV gastrointestinal cancer: a phase II clinical trial. J Cancer Res Clin Oncol 2016; 142:1315-23. [PMID: 26941189 DOI: 10.1007/s00432-016-2127-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/30/2016] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To investigate the efficacy of autologous cytokine-induced killer (CIK) cell therapy combined with chemotherapy versus chemotherapy alone for the treatment of stage IV gastrointestinal (GI) cancer in the first-line setting. METHODS Thirty-three patients diagnosed with stage IV GI cancer were divided into chemotherapy plus CIK group (chemo-CIK, n = 16) and chemotherapy-alone group (chemo-alone, n = 17). Autologous peripheral blood mononuclear cells were separated by flow cytometry, cultured in vitro to induce CIK cells, and transfused into patients on days 14 and 16 of the first and second chemotherapy cycles. RESULTS The median progression-free survival (PFS) was 5.6 months for patients in the chemo-CIK group and 3.83 months for those in the chemo-alone group. The difference was borderline significant (P = 0.06), indicating a potential advantage for combined CIK cell transfusion with chemotherapy in improving PFS. A favored objective response rate was also observed in the chemo-CIK group than in the chemo-alone group. This study also revealed that CIK cell transfusion restored the cellular immunity in these GI cancer patients. The percentage of natural killer T cells, NK cells, CD3(+) T cells, and T-cell subgroups CD4(+) proportion in the peripheral blood of cancer patients significantly increased after the CIK cell transfusion, while the change in T-cell subgroups CD8(+) and CD4(+)/CD8(+) did not differ significantly. CONCLUSIONS The study showed that the addition of CIK cell transfusion to traditional chemotherapy in the first-line setting was associated with a prolonged PFS and enhanced T-lymphocyte subset activity, supporting a potential treatment choice for advanced GI cancer patients.
Collapse
Affiliation(s)
- Ying-Chun Xu
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200081, China
- Department of Oncology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Qi Xu
- Department of Oncology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jun-Jian Li
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200081, China
| | - Xiao-Feng Gu
- Department of Oncology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiao-Lin Lin
- Department of Oncology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Li Sun
- Department of Oncology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Hong-Min Lu
- Department of Oncology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lei Tang
- Department of Oncology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yue Ma
- Department of Oncology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhimin Lu
- Department of Neuro-Oncology - Research Unit Number: 1002, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Hong-Xia Wang
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200081, China.
| |
Collapse
|
43
|
Dual Effects of Cellular Immunotherapy in Inhibition of Virus Replication and Prolongation of Survival in HCV-Positive Hepatocellular Carcinoma Patients. J Immunol Res 2016; 2016:6837241. [PMID: 27069936 PMCID: PMC4812393 DOI: 10.1155/2016/6837241] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/14/2016] [Accepted: 02/14/2016] [Indexed: 12/11/2022] Open
Abstract
Immune cells play an important role in the development and progression of hepatitis C virus (HCV) and hepatocellular carcinoma (HCC). We conducted a retrospective study to evaluate the influence of adoptive cellular immunotherapy (CIT) on viral load and progression-free survival (PFS) for HCC patients infected with HCV. Patients (n = 104) were divided into a control group (conventional therapy, n = 73) and study group (combination of CIT and conventional therapy, n = 31). Autologous mononuclear cells were induced into natural killer, γδT, and cytokine-induced killer cells and infused intravenously to study group patients. More patients had shown viral load decrease or were stable in study group (100% versus 75%) (p = 0.014). The median PFS of the study group and control group was 16 and 10 months, respectively (p = 0.0041), and only CIT was an independent prognostic factor for PFS (hazard ratio, 0.422; p = 0.005). Three patients developed transient moderate fever after infusion, and there were no significant differences in alanine aminotransferase and aspartate aminotransferase levels before and after treatment in both groups. Our results show that CIT contributes to improvement of prognosis and inhibition of viral replication in HCV-related HCC patients, without impairment of liver function.
Collapse
|
44
|
Tagliamonte M, Petrizzo A, Tornesello ML, Ciliberto G, Buonaguro FM, Buonaguro L. Combinatorial immunotherapy strategies for hepatocellular carcinoma. Curr Opin Immunol 2016; 39:103-13. [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] [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
|
45
|
Wang Y, Xu Z, Zhou F, Sun Y, Chen J, Li L, Jin H, Qian Q. The combination of dendritic cells-cytotoxic T lymphocytes/cytokine-induced killer (DC-CTL/CIK) therapy exerts immune and clinical responses in patients with malignant tumors. Exp Hematol Oncol 2015; 4:32. [PMID: 26561538 PMCID: PMC4641330 DOI: 10.1186/s40164-015-0027-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 10/30/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The clinical trials using immunotherapy have been performed for the treatment of variety of malignant tumors. However, large-scale meta-analysis of combined DC-CTL/CIK therapy on immune and clinical response in patients has not been well studied yet. The purpose of this study is to investigate the role of DC-CTL/CIK therapy and evaluate the changes of immune indicators and tumor serological markers both at an individual level and at a system level, which is an important basis for immunotherapy as well as prognosis estimation. METHODS Three cohorts were designed to estimate therapeutic effects on patients with malignant tumors. Tumor serological markers were detected pre- and post-treatment by immunoradiometric methods using commercially available diagnostic kits. Lymphocyte subsets were identified by flow cytometry. The quality of life was assessed by EORTC QLQ-C30 questionnaire. RESULTS In this study, we found out that Tregs was significantly reduced after transfusion of DC-CTL/CIK cells companied by decreasing serological tumor markers including AFP, CA199 and CA242 in primary liver cancer and CA724 in gastric cancer. A system-level analysis showed that lower percentages of Tregs were detected in patients with long-lasting courses of immunotherapy. Strikingly, a tumor progression indicator, myeloid-derived suppressor cells (MDSC), was dramatically decreased in patients after DC-CTL/CIK treatment. These results suggested that DC-CTL/CIK therapy improves immune functions and the quality of life post-treatment versus pre-therapy, indicating that DC-CTL/CIK therapy might block the deterioration of invasive cancers in these patients. CONCLUSIONS This study demonstrated that DC-CTL/CIK therapy could reduce Tregs, MDSCs, and several crucial serological tumor markers in particular tumors, and improve the function of T cells immune systems and the quality of life in patients with malignant tumor.
Collapse
Affiliation(s)
- Ying Wang
- Laboratory of Gene and Viral Therapy, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University of Chinese PLA, Shanghai, 200438 China
| | - Zenghui Xu
- Laboratory of Gene and Viral Therapy, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University of Chinese PLA, Shanghai, 200438 China
| | - Fuping Zhou
- Department of Biotherapy, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University of Chinese PLA, Shanghai, 200438 China
| | - Yan Sun
- Laboratory of Gene and Viral Therapy, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University of Chinese PLA, Shanghai, 200438 China
| | - Jingbo Chen
- Laboratory of Gene and Viral Therapy, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University of Chinese PLA, Shanghai, 200438 China
| | - Linfang Li
- Laboratory of Gene and Viral Therapy, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University of Chinese PLA, Shanghai, 200438 China
| | - Huajun Jin
- Laboratory of Gene and Viral Therapy, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University of Chinese PLA, Shanghai, 200438 China
| | - Qijun Qian
- Laboratory of Gene and Viral Therapy, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University of Chinese PLA, Shanghai, 200438 China.,Department of Biotherapy, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University of Chinese PLA, Shanghai, 200438 China
| |
Collapse
|
46
|
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
|
47
|
Xu L, Wang J, Kim Y, Shuang ZY, Zhang YJ, Lao XM, Li YQ, Chen MS, Pawlik TM, Xia JC, Li SP, Lau WY. A randomized controlled trial on patients with or without adjuvant autologous cytokine-induced killer cells after curative resection for hepatocellular carcinoma. Oncoimmunology 2015; 5:e1083671. [PMID: 27141337 DOI: 10.1080/2162402x.2015.1083671] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 08/09/2015] [Accepted: 08/11/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND & AIMS There is no generally accepted adjuvant therapy for hepatocellular carcinoma (HCC) after curative resection. Autologous cytokine-induced killer (CIK) cells therapy has been reported to improve outcomes of patients with HCC, but its role as an adjuvant therapy remains unclear. This study aimed to evaluate the efficacy and safety of CIK as an adjuvant therapy for HCC after curative resection. METHODS This is a single center, phase 3, open label, randomized controlled trial (RCT). Two hundred patients who were initially diagnosed with HCC of Barcelona Clinic Liver Cancer (BCLC) stage A or B, and underwent curative hepatectomy were randomly assigned to receive four cycles of CIK treatment (the CIK group, n = 100) or no treatment (the control group, n = 100). The primary outcome was time to recurrence. The secondary outcomes included disease-free survival (DFS), overall survival (OS) and adverse events. RESULTS All patients in the CIK group finished the treatment by protocol. The median time to recurrence (TTR) was 13.6 (IQR 6.5-25.2) mo in the CIK group and 7.8 (IQR 2.7-17.0) mo in the control group (p = 0.01). There were no significant differences between the groups in DFS and OS. All adverse events were grade 1 or 2. There were no significant differences in incidence between the two groups. CONCLUSIONS Four cycles of CIK therapy were safe and effective to prolong the median TTR in patients with HCC after curative resection, but the treatment did not improve the DFS and OS.
Collapse
Affiliation(s)
- Li Xu
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; State Key Laboratory of Oncology in Southern China, Guangzhou, Guangdong, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China; Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jun Wang
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; State Key Laboratory of Oncology in Southern China, Guangzhou, Guangdong, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China; Department of Ultrasound, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yuhree Kim
- Department of Surgery, The Johns Hopkins University School of Medicine , Baltimore, MD, USA
| | - Ze-Yu Shuang
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; State Key Laboratory of Oncology in Southern China, Guangzhou, Guangdong, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Yao-Jun Zhang
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; State Key Laboratory of Oncology in Southern China, Guangzhou, Guangdong, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Xiang-Ming Lao
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; State Key Laboratory of Oncology in Southern China, Guangzhou, Guangdong, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Yong-Qiang Li
- State Key Laboratory of Oncology in Southern China, Guangzhou, Guangdong, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China; Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Min-Shan Chen
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; State Key Laboratory of Oncology in Southern China, Guangzhou, Guangdong, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Timothy M Pawlik
- Department of Surgery, The Johns Hopkins University School of Medicine , Baltimore, MD, USA
| | - Jian-Chuan Xia
- State Key Laboratory of Oncology in Southern China, Guangzhou, Guangdong, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China; Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Sheng-Ping Li
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; State Key Laboratory of Oncology in Southern China, Guangzhou, Guangdong, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Wan-Yee Lau
- Faculty of Medicine, The Chinese University of Hong Kong Prince of Wales Hospital , Hong Kong S.A.R., China
| |
Collapse
|
48
|
Zhang Z, Wang L, Luo Z, Zhao X, Huang J, Li H, Yang S, Zhao X, Zhang L, Li L, Wang F, Huang L, Zhang Y. Efficacy and safety of cord blood-derived cytokine-induced killer cells in treatment of patients with malignancies. Cytotherapy 2015; 17:1130-1138. [PMID: 25963952 DOI: 10.1016/j.jcyt.2015.04.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 04/01/2015] [Accepted: 04/04/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND AIMS Adoptive immunotherapy with the use of cytokine-induced killer (CIK) cells represents an effective therapeutic option for treating malignancies. The characteristics and function of cord blood-derived CIK (CB-CIK) cells have been evaluated both in vitro and in vivo. In this study, we assessed the efficacy and safety of administering CB-CIK cells to patients with cancer. METHODS In this retrospective clinical trial, 15 patients with cancer received CB-CIK therapy with different cycles from April 2012 to August 2014. CB-CIK cells demonstrated a high percentage of main functional fraction CD3(+)CD56(+) and efficient anti-tumor activity in vitro. RESULTS After the infusion of CB-CIK cells, the subsets of CD3(+)CD4(+) T lymphocytes and CD3(-)CD56(+) T cells in the peripheral blood were significantly increased compared with those before the therapy. Of 15 patients, one patient with hepatocellular cancer and one patient with esophageal cancer achieved complete responses, two patients with ovarian cancer obtained partial remissions, 10 patients had stable disease and one patient with hepatocellular cancer had progressive disease. Acute toxicities including fever, slight fever, dizziness and other neurologic toxicities were few and occurred in patients after infusion of CB-CIK cells. CONCLUSIONS These results demonstrated the feasibility and safety of treating malignancies with CB-CIK cells. The study provides a potential therapeutic approach for the patients with poor health or older patients who cannot tolerate repeated collection of blood.
Collapse
Affiliation(s)
- Zhen Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liping Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhenzhen Luo
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuan Zhao
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianmin Huang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hong Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuangning Yang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xianlan Zhao
- Department of Obstetrics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lei Zhang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liuxia Li
- Department of Obstetrics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lan Huang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; School of Life Science, Zhengzhou University, Zhengzhou, Henan, China; Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, Henan, China.
| |
Collapse
|
49
|
Zhang Q, Liu XY, Zhang T, Zhang XF, Zhao L, Long F, Liu ZK, Wang EH. The dual-functional capability of cytokine-induced killer cells and application in tumor immunology. Hum Immunol 2015; 76:385-91. [DOI: 10.1016/j.humimm.2014.09.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 09/27/2014] [Accepted: 09/27/2014] [Indexed: 01/18/2023]
|
50
|
Elia AR, Circosta P, Sangiolo D, Bonini C, Gammaitoni L, Mastaglio S, Genovese P, Geuna M, Avolio F, Inghirami G, Tarella C, Cignetti A. Cytokine-induced killer cells engineered with exogenous T-cell receptors directed against melanoma antigens: enhanced efficacy of effector cells endowed with a double mechanism of tumor recognition. Hum Gene Ther 2015; 26:220-31. [PMID: 25758764 DOI: 10.1089/hum.2014.112] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cytokine-induced killer (CIK) cells consist of a heterogeneous population of polyclonal T lymphocytes displaying NK phenotype and HLA-unrestricted cytotoxic activity against a broad range of tumors. We sought to determine whether transduction of CIK cells with T cell receptor (TCR) genes specific for tumor-associated antigens could generate effector cells endowed with a double mechanism of tumor recognition. HLA-A2-restricted TCR-transduced (TD) CIK directed against the melanoma antigens Mart1 and NY-ESO1 were generated by lentiviral transduction and successfully expanded over a 3-4-week period. TD-CIK cells were both CD3(+)/CD56(-) and CD3(+)/CD56(+) (31±8% and 59±9%, respectively), indicating that both major histocompatibility complex (MHC)-restricted T cells and MHC-unrestricted CIK could be targeted by lentiviral transduction. At the end of the culture, the majority of both unmodified and TD-CIK displayed an effector memory phenotype, without considerable expression of replicative senescence and exhaustion markers. Functionally, TD-CIK specifically recognized tumor cells expressing the relevant antigen as well as maintained their MHC-unrestricted tumor activity. The cytotoxic activity of TD-CIK against HLA-A2(+) melanoma cell lines was significantly higher than the untransduced counterparts at a low effector:target ratio (cytotoxic activity of TD-CIK was from 1.9- to 4.3-fold higher than untransduced counterparts). TD-CIK were highly proficient in releasing high amount of IFN-γ upon antigen-specific stimulation and were able to recognize primary melanoma targets. In conclusion, we showed that (1) the reproducibility and simplicity of CIK transduction and expansion might solve the problem of obtaining adequate numbers of potent antitumor effector cells for adoptive immunotherapy; (2) the presence of both terminal effectors as well as of less differentiated progenitors might confer them long survival in vivo; and (3) the addition of an MHC-restricted antigen recognition allows not only targeting tumor surface antigens but also a wider range of cytoplasmic or nuclear antigens, involved in tumor proliferation and survival. TD-CIK cells with a double mechanism of tumor recognition are an attractive and alternative tool for the development of efficient cell therapeutic strategies.
Collapse
Affiliation(s)
- Angela R Elia
- 1 Molecular Biotechnology Center, University of Torino , 10126 Turin, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|