1
|
Kim K, Wang H, Lee J, Yeom C. Long-term tumor suppression in cholangiocarcinoma using cytokine-induced killer cell therapy and high-dose vitamin C: a case report. KOREAN JOURNAL OF CLINICAL ONCOLOGY 2024; 20:84-87. [PMID: 39778511 PMCID: PMC11717578 DOI: 10.14216/kjco.24012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025]
Abstract
This case study explores the effectiveness of autologous cytokine-induced killer (CIK) cell-based immunotherapy in a 49-year-old male patient with inoperable stage IIIb cholangiocarcinoma, characterized by high levels of the sodium-dependent vitamin C transporter-2 (SVCT2) in immune cells. Despite an initial lack of tumor reduction following chemotherapy, the patient showed a significant decrease in tumor markers and stabilization of the tumor after undergoing radiation and proton therapy. Subsequently, CIK cell therapy, combined with high-dose vitamin C, was administered 52 times over 6 years. The patient's tumor size reduced, and no cancer activity was detected for 7 years and 10 months post-diagnosis, indicating a successful long-term outcome without recurrence. This study suggests that CIK cell therapy, particularly in patients with elevated SVCT2 levels, may offer a promising adjuvant treatment for cholangiocarcinoma and potentially other cancers. Further research is needed to validate SVCT2 as a biomarker for the effectiveness of CIK cell therapy.
Collapse
Affiliation(s)
- Kangseok Kim
- Department of Surgery, Bangre Hospital, Incheon,
Korea
| | - Hyunhye Wang
- Department of Radiology, Bangre Hospital, Incheon,
Korea
| | - Jiewon Lee
- Department of Family Medicine, Yeom Chang Hwan Hospital, Seoul,
Korea
| | - Changhwan Yeom
- Department of Family Medicine, Yeom Chang Hwan Hospital, Seoul,
Korea
| |
Collapse
|
2
|
Tseng YH, Ho CL, Chian CF, Chiang CL, Chao HS, Tsai CL, Perng WC, Hsiao CF, Chuang MH, Ko KH, Cheng YC, Chen SJ, Wang CJ, Chen YM. Immune killer cells treatment for previously treated stage IV NSCLC patients. Sci Rep 2024; 14:19374. [PMID: 39169058 PMCID: PMC11339402 DOI: 10.1038/s41598-024-69587-x] [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: 01/10/2023] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
The 5-year survival is poor for stage IV non-small cell lung cancer (NSCLC). Recently, cell immunotherapy has emerged as a new treatment strategy. This study aimed to evaluate the efficacy and safety of Immune killer cells (IKC) in patients with stage IV NSCLC after the failure of prior chemotherapy. This study enrolled 26 patients with stage IV NSCLC who failed at least two lines of chemotherapy with or without targeted therapy. The IKC was given alone weekly for 24 weeks. The primary endpoint was progression-free survival (PFS). Secondary outcomes included overall survival (OS), pain intensity, quality of life (QOL), and safety. The median PFS for the intent-to-treat (ITT) population (i.e., all enrolled patients) was 3.8 month. In the per-protocol (PP) population (i.e., patients receiving > 12 IKC infusions), the median PFS was 5.6 months. Moreover, the ITT population showed a 1-year survival rate of 60.0%, while that for the PP population was 85.7%. Only 7 out of 200 AEs (3.5%) were related to the IKC infusion, and they were all rated as grade 1 in severity. The IKC infusion was well tolerated. This novel immunotherapy prolonged the PFS and improved the survival compared with historical data. It might be a potential treatment strategy for stage IV NSCLC patient who failed prior chemotherapy.ClinicalTrials.gov identifier: NCT03499834.
Collapse
Affiliation(s)
- Yen-Han Tseng
- Department of Chest Medicine, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Rd., Beitou District, Taipei, 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong Street, Taipei, 112, Taiwan
| | - Ching-Liang Ho
- Division of Hematology and Oncology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, No.325, Sec.2, Chenggong Rd., Neihu District, Taipei, 114, Taiwan
| | - Chih-Feng Chian
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, No.325, Sec.2, Chenggong Rd., Neihu District, Taipei, 114, Taiwan
| | - Chi-Lu Chiang
- Department of Chest Medicine, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Rd., Beitou District, Taipei, 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong Street, Taipei, 112, Taiwan
| | - Heng-Sheng Chao
- Department of Chest Medicine, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Rd., Beitou District, Taipei, 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong Street, Taipei, 112, Taiwan
| | - Chen-Liang Tsai
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, No.325, Sec.2, Chenggong Rd., Neihu District, Taipei, 114, Taiwan
| | - Wann-Cherng Perng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, No.325, Sec.2, Chenggong Rd., Neihu District, Taipei, 114, Taiwan
| | - Chin-Fu Hsiao
- Institute of Population Health Sciences, National Health Research Institutes, No. 35, Keyan Rd., Zhunan, Miaoli, 350, Taiwan
| | - Mei-Hsing Chuang
- Institute of Population Health Sciences, National Health Research Institutes, No. 35, Keyan Rd., Zhunan, Miaoli, 350, Taiwan
| | - Kai-Hsiung Ko
- Department of Radiology, Tri-Service General Hospital, National Defense Medical Center, No.325, Sec.2, Chenggong Rd., Neihu District, Taipei, 114, Taiwan
| | - Yun-Ching Cheng
- Ivy Life Sciences Co., Ltd., No. 76, Yuhe St, Taoyuan, 330, Taiwan
| | - Shin-Jung Chen
- Ivy Life Sciences Co., Ltd., No. 76, Yuhe St, Taoyuan, 330, Taiwan
| | - Chia-Jen Wang
- Ivy Life Sciences Co., Ltd., No. 76, Yuhe St, Taoyuan, 330, Taiwan
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Rd., Beitou District, Taipei, 112, Taiwan.
- School of Medicine, National Yang Ming Chiao Tung University, No.155, Sec.2, Linong Street, Taipei, 112, Taiwan.
| |
Collapse
|
3
|
Yang Y, Zhang C, Jiang Y, He Y, Cai J, Liang L, Chen Z, Pan S, Hua C, Wu K, Wang L, Zhang Z. Harnessing cytokine-induced killer cells to accelerate diabetic wound healing: an approach to regulating post-traumatic inflammation. Regen Biomater 2024; 11:rbad116. [PMID: 38333727 PMCID: PMC10850840 DOI: 10.1093/rb/rbad116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 02/10/2024] Open
Abstract
Impaired immunohomeostasis in diabetic wounds prolongs inflammation and cytokine dysfunction, thus, delaying or preventing wound-surface healing. Extensive clinical studies have been conducted on cytokine-induced killer (CIK) cells recently, as they can be easily proliferated using a straightforward, inexpensive protocol. Therefore, the function of CIK cells in regulating inflammatory environments has been drawing attention for clinical management. Throughout the current investigation, we discovered the regenerative capacity of these cells in the challenging environment of wounds that heal poorly due to diabetes. We demonstrated that the intravenous injection of CIK cells can re-establish a proregenerative inflammatory microenvironment, promote vascularization and, ultimately, accelerate skin healing in diabetic mice. The results indicated that CIK cell treatment affects macrophage polarization and restores the function of regenerative cells under hyperglycemic conditions. This novel cellular therapy offers a promising intervention for clinical applications through specific inflammatory regulation functions.
Collapse
Affiliation(s)
- Yixi Yang
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Department of Orthopaedic Surgery, Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong 510150, P. R. China
| | - Cheng Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Department of Orthopaedic Surgery, Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong 510150, P. R. China
| | - Yuan Jiang
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Department of Orthopaedic Surgery, Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong 510150, P. R. China
| | - Yijun He
- Department of Osteoarthropathy and Sports Medicine, Panyu Central Hospital, Guangzhou 511400, P. R. China
| | - Jiawei Cai
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Department of Orthopaedic Surgery, Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong 510150, P. R. China
| | - Lin Liang
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Department of Orthopaedic Surgery, Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong 510150, P. R. China
| | - Zhaohuan Chen
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Department of Orthopaedic Surgery, Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong 510150, P. R. China
| | - Sicheng Pan
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Department of Orthopaedic Surgery, Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong 510150, P. R. China
| | - Chu Hua
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Department of Orthopaedic Surgery, Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong 510150, P. R. China
| | - Keke Wu
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Department of Orthopaedic Surgery, Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong 510150, P. R. China
| | - Le Wang
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Department of Orthopaedic Surgery, Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong 510150, P. R. China
| | - Zhiyong Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Department of Orthopaedic Surgery, Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong 510150, P. R. China
| |
Collapse
|
4
|
Xie J, Chen L, Liu Q, Li XT, Lei XY. Efficacy of Chemoimmunotherapy versus Chemotherapy for Gastric Cancer: A Meta-Analysis of Survival Outcomes. Curr Med Chem 2024; 31:2649-2660. [PMID: 38265394 DOI: 10.2174/0109298673263335231121103807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/25/2023] [Accepted: 10/20/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND Gastric cancer has been traditionally treated with chemotherapy as the primary mode of treatment. However, recent studies have shown that chemoimmunotherapy is also effective and, in some cases, better than chemotherapy treatment. Current study aimed to find the efficacy of chemoimmunotherapy versus chemotherapy in the treatment of gastric cancer. METHODS Using electronic databases, including PubMed, Embase, and EBSCO, a thorough literature search was carried out for the years 2006 to 2023. The search strategy was designed to identify relevant studies based on chemoimmunotherapy and chemotherapy intervention, and the search was conducted using appropriate keywords and MeSH terms. The retrieved studies were screened for relevance based on their titles, abstracts, and full texts. The studies' inclusion criteria were predefined, and the selected studies were then subjected to a quality assessment using GradePro GDT. The data from selected studies were extracted and analyzed using Revman version 5.4. RESULTS The study found that chemoimmunotherapy treatment resulted in a significant improvement in overall survival (OS) with a risk ratio (RR) of 1.54 and a 95% Confidence Interval (CI) of 1.25 to 1.89. The overall effect was also found to be significant, with a p-value of less than 0.001. Furthermore, we also observed an improvement in the 1-year, 3-year, and 5-year survival rates with risk ratio (RR) of 1.09 (95% CI: 1.01, 1.17), 1.43 (95% CI: 1.28, 1.60), and 1.59 (95% CI: 1.10, 2.30), respectively. In addition, it's also found that chemoimmunotherapy treatment also resulted in an improvement in DFS with an RR of 1.94 and a 95% CI of 1.44 to 2.59. Overall, these results suggest that chemoimmunotherapy treatment can be an effective approach in comparison to chemotherapy for improving overall survival and disease-free survival in the studied population. CONCLUSION This study comparing chemoimmunotherapy versus chemotherapy for gastric cancer showed that both treatments were effective, but chemoimmunotherapy had more significant efficacy. To support these results, additional studies with a large sample size and a longer follow-up time are required.
Collapse
Affiliation(s)
- Juan Xie
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Institute of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Lin Chen
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Qing Liu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xi-Tai Li
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiao-Yong Lei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
5
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593'"] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
6
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593'||'] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
7
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593%' and 2*3*8=6*8 and 'eho8'!='eho8%] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
8
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593' and 2*3*8=6*8 and 'x7c8'='x7c8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
9
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593����%2527%2522\'\"] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
10
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593" and 2*3*8=6*8 and "d5lf"="d5lf] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
11
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 DOI: 10.3748/wjg.v29.i40.5593j4dtuwxg] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China.
| |
Collapse
|
12
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 PMCID: PMC10642438 DOI: 10.3748/wjg.v29.i40.5593] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| |
Collapse
|
13
|
Zhang Z, Liu N, Sun M. Research Progress of Immunotherapy for Gastric Cancer. Technol Cancer Res Treat 2023; 22:15330338221150555. [PMID: 37042029 PMCID: PMC10102952 DOI: 10.1177/15330338221150555] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 10/16/2022] [Accepted: 12/22/2022] [Indexed: 04/13/2023] Open
Abstract
Gastric cancer (GC) is one of the most common gastrointestinal tract cancers worldwide, which has high incidence and mortality rates and poor prognosis. Although multidisciplinary comprehensive therapies consisting of surgery, chemotherapy, radiotherapy, and targeted therapy have made great progress in GC treatment, a satisfactory curative effect still cannot be achieved in many circumstances, and the 5-year survival of patients with GC remains to be very low. In China, about 75% of patients with GC are diagnosed in the advanced stage and thus miss the opportunity of surgical resection. Although the conventional treatment of GC has improved the survival time of advanced patients to a certain extent, the clinical efficacy has encountered a bottleneck and cannot bring higher survival benefits to patients. With the development of immunologic and molecular biologic technologies, immunotherapy has gradually become a new essential treatment for GC, which has attracted extensive attention in the field of oncology. The US Food and Drug Administration (USFDA) and China Food and Drug Administration (CFDA) have approved a variety of immune-related drugs for the treatment of GC, and all of which have achieved good efficacy. In this review, we summarize the recent development in nonspecific enhancer therapy, adoptive immunocell therapy, tumor vaccine therapy, oncolytic virus therapy, and immune checkpoint inhibitor therapy, and their roles in the treatment of GC.
Collapse
Affiliation(s)
- Zhipeng Zhang
- Key Laboratory of Liver and Kidney
Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai,
China
| | - Ningning Liu
- Department of Medical Oncology and
Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai,
China
| | - Mingyu Sun
- Key Laboratory of Liver and Kidney
Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai,
China
| |
Collapse
|
14
|
Zhao Y, Bai Y, Shen M, Li Y. Therapeutic strategies for gastric cancer targeting immune cells: Future directions. Front Immunol 2022; 13:992762. [PMID: 36225938 PMCID: PMC9549957 DOI: 10.3389/fimmu.2022.992762] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Gastric cancer (GC) is a malignancy with a high incidence and mortality, and the emergence of immunotherapy has brought survival benefits to GC patients. Compared with traditional therapy, immunotherapy has the advantages of durable response, long-term survival benefits, and lower toxicity. Therefore, targeted immune cells are the most promising therapeutic strategy in the field of oncology. In this review, we introduce the role and significance of each immune cell in the tumor microenvironment of GC and summarize the current landscape of immunotherapy in GC, which includes immune checkpoint inhibitors, adoptive cell therapy (ACT), dendritic cell (DC) vaccines, reduction of M2 tumor-associated macrophages (M2 TAMs), N2 tumor-associated neutrophils (N2 TANs), myeloid-derived suppressor cells (MDSCs), effector regulatory T cells (eTregs), and regulatory B cells (Bregs) in the tumor microenvironment and reprogram TAMs and TANs into tumor killer cells. The most widely used immunotherapy strategies are the immune checkpoint inhibitor programmed cell death 1/programmed death-ligand 1 (PD-1/PD-L1) antibody, cytotoxic T lymphocyte–associated protein 4 (CTLA-4) antibody, and chimeric antigen receptor T (CAR-T) in ACT, and these therapeutic strategies have significant anti-tumor efficacy in solid tumors and hematological tumors. Targeting other immune cells provides a new direction for the immunotherapy of GC despite the relatively weak clinical data, which have been confirmed to restore or enhance anti-tumor immune function in preclinical studies and some treatment strategies have entered the clinical trial stage, and it is expected that more and more effective immune cell–based therapeutic methods will be developed and applied.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuansong Bai
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Meili Shen
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
- *Correspondence: Yapeng Li, ; Meili Shen,
| | - Yapeng Li
- The National and Local Joint Engineering Laboratory for Synthesis Technology of High Performance Polymer, College of Chemistry, Jilin University, Changchun, China
- *Correspondence: Yapeng Li, ; Meili Shen,
| |
Collapse
|
15
|
Jin X, Liu Z, Yang D, Yin K, Chang X. Recent Progress and Future Perspectives of Immunotherapy in Advanced Gastric Cancer. Front Immunol 2022; 13:948647. [PMID: 35844558 DOI: 10.3389/fimmu.2022.948647%' and 2*3*8=6*8 and 'r4tf'!='r4tf%] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/06/2022] [Indexed: 01/29/2024] Open
Abstract
As one of the most common forms of solid tumours, gastric carcinoma has been revealed as the third leading cause of death worldwide. The symptom of gastric cancer is usually not obvious and thus difficult to detect at earlier stages. Therefore, gastric cancer is already in the advanced stage once detected in patients, which has a poor prognosis due to ineffective therapies and multiple resistance. Recent advance in understanding the microenvironment of cancer has significantly promoted the development of immunotherapy for advanced gastric cancer. Immunotherapy can induce immune responses in gastric cancer patients thus leads to the destruction of cancer cells. In comparison of traditional therapy, immunotherapy has demonstrated robust efficacy and tolerable toxicity. Therefore, this novel strategy for treatment of advanced gastric cancer has gain increasingly popularity. In this review, we summarize recent progress of immunotherapy in advanced gastric cancer, such as immune check point inhibitors, adoptive cell therapy, VEGF inhibitors, cancer vaccines and CAR-T cell therapy. We highlight immunotherapies involved in clinical applications and discuss the existing challenges of current immunotherapies and promising strategies to overcome these limitations.
Collapse
Affiliation(s)
- Xin Jin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
| | - Zhaorui Liu
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dongxiao Yang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Kai Yin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xusheng Chang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
16
|
Jin X, Liu Z, Yang D, Yin K, Chang X. Recent Progress and Future Perspectives of Immunotherapy in Advanced Gastric Cancer. Front Immunol 2022; 13:948647. [PMID: 35844558 DOI: 10.3389/fimmu.2022.948647'"] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/06/2022] [Indexed: 01/29/2024] Open
Abstract
As one of the most common forms of solid tumours, gastric carcinoma has been revealed as the third leading cause of death worldwide. The symptom of gastric cancer is usually not obvious and thus difficult to detect at earlier stages. Therefore, gastric cancer is already in the advanced stage once detected in patients, which has a poor prognosis due to ineffective therapies and multiple resistance. Recent advance in understanding the microenvironment of cancer has significantly promoted the development of immunotherapy for advanced gastric cancer. Immunotherapy can induce immune responses in gastric cancer patients thus leads to the destruction of cancer cells. In comparison of traditional therapy, immunotherapy has demonstrated robust efficacy and tolerable toxicity. Therefore, this novel strategy for treatment of advanced gastric cancer has gain increasingly popularity. In this review, we summarize recent progress of immunotherapy in advanced gastric cancer, such as immune check point inhibitors, adoptive cell therapy, VEGF inhibitors, cancer vaccines and CAR-T cell therapy. We highlight immunotherapies involved in clinical applications and discuss the existing challenges of current immunotherapies and promising strategies to overcome these limitations.
Collapse
Affiliation(s)
- Xin Jin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
| | - Zhaorui Liu
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dongxiao Yang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Kai Yin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xusheng Chang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
17
|
Jin X, Liu Z, Yang D, Yin K, Chang X. Recent Progress and Future Perspectives of Immunotherapy in Advanced Gastric Cancer. Front Immunol 2022; 13:948647. [PMID: 35844558 DOI: 10.3389/fimmu.2022.948647" and 2*3*8=6*8 and "hxww"="hxww] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/06/2022] [Indexed: 01/29/2024] Open
Abstract
As one of the most common forms of solid tumours, gastric carcinoma has been revealed as the third leading cause of death worldwide. The symptom of gastric cancer is usually not obvious and thus difficult to detect at earlier stages. Therefore, gastric cancer is already in the advanced stage once detected in patients, which has a poor prognosis due to ineffective therapies and multiple resistance. Recent advance in understanding the microenvironment of cancer has significantly promoted the development of immunotherapy for advanced gastric cancer. Immunotherapy can induce immune responses in gastric cancer patients thus leads to the destruction of cancer cells. In comparison of traditional therapy, immunotherapy has demonstrated robust efficacy and tolerable toxicity. Therefore, this novel strategy for treatment of advanced gastric cancer has gain increasingly popularity. In this review, we summarize recent progress of immunotherapy in advanced gastric cancer, such as immune check point inhibitors, adoptive cell therapy, VEGF inhibitors, cancer vaccines and CAR-T cell therapy. We highlight immunotherapies involved in clinical applications and discuss the existing challenges of current immunotherapies and promising strategies to overcome these limitations.
Collapse
Affiliation(s)
- Xin Jin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
| | - Zhaorui Liu
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dongxiao Yang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Kai Yin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xusheng Chang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
18
|
Jin X, Liu Z, Yang D, Yin K, Chang X. Recent Progress and Future Perspectives of Immunotherapy in Advanced Gastric Cancer. Front Immunol 2022; 13:948647. [PMID: 35844558 DOI: 10.3389/fimmu.2022.948647'||'] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/06/2022] [Indexed: 01/29/2024] Open
Abstract
As one of the most common forms of solid tumours, gastric carcinoma has been revealed as the third leading cause of death worldwide. The symptom of gastric cancer is usually not obvious and thus difficult to detect at earlier stages. Therefore, gastric cancer is already in the advanced stage once detected in patients, which has a poor prognosis due to ineffective therapies and multiple resistance. Recent advance in understanding the microenvironment of cancer has significantly promoted the development of immunotherapy for advanced gastric cancer. Immunotherapy can induce immune responses in gastric cancer patients thus leads to the destruction of cancer cells. In comparison of traditional therapy, immunotherapy has demonstrated robust efficacy and tolerable toxicity. Therefore, this novel strategy for treatment of advanced gastric cancer has gain increasingly popularity. In this review, we summarize recent progress of immunotherapy in advanced gastric cancer, such as immune check point inhibitors, adoptive cell therapy, VEGF inhibitors, cancer vaccines and CAR-T cell therapy. We highlight immunotherapies involved in clinical applications and discuss the existing challenges of current immunotherapies and promising strategies to overcome these limitations.
Collapse
Affiliation(s)
- Xin Jin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
| | - Zhaorui Liu
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dongxiao Yang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Kai Yin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xusheng Chang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
19
|
Jin X, Liu Z, Yang D, Yin K, Chang X. Recent Progress and Future Perspectives of Immunotherapy in Advanced Gastric Cancer. Front Immunol 2022; 13:948647. [PMID: 35844558 PMCID: PMC9284215 DOI: 10.3389/fimmu.2022.948647] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/06/2022] [Indexed: 12/15/2022] Open
Abstract
As one of the most common forms of solid tumours, gastric carcinoma has been revealed as the third leading cause of death worldwide. The symptom of gastric cancer is usually not obvious and thus difficult to detect at earlier stages. Therefore, gastric cancer is already in the advanced stage once detected in patients, which has a poor prognosis due to ineffective therapies and multiple resistance. Recent advance in understanding the microenvironment of cancer has significantly promoted the development of immunotherapy for advanced gastric cancer. Immunotherapy can induce immune responses in gastric cancer patients thus leads to the destruction of cancer cells. In comparison of traditional therapy, immunotherapy has demonstrated robust efficacy and tolerable toxicity. Therefore, this novel strategy for treatment of advanced gastric cancer has gain increasingly popularity. In this review, we summarize recent progress of immunotherapy in advanced gastric cancer, such as immune check point inhibitors, adoptive cell therapy, VEGF inhibitors, cancer vaccines and CAR-T cell therapy. We highlight immunotherapies involved in clinical applications and discuss the existing challenges of current immunotherapies and promising strategies to overcome these limitations.
Collapse
Affiliation(s)
- Xin Jin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
| | - Zhaorui Liu
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dongxiao Yang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Dongxiao Yang, ; Kai Yin, ; Xusheng Chang,
| | - Kai Yin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Dongxiao Yang, ; Kai Yin, ; Xusheng Chang,
| | - Xusheng Chang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Dongxiao Yang, ; Kai Yin, ; Xusheng Chang,
| |
Collapse
|
20
|
Jin X, Liu Z, Yang D, Yin K, Chang X. Recent Progress and Future Perspectives of Immunotherapy in Advanced Gastric Cancer. Front Immunol 2022; 13:948647. [PMID: 35844558 DOI: 10.3389/fimmu.2022.948647bsd3bmst] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/06/2022] [Indexed: 01/29/2024] Open
Abstract
As one of the most common forms of solid tumours, gastric carcinoma has been revealed as the third leading cause of death worldwide. The symptom of gastric cancer is usually not obvious and thus difficult to detect at earlier stages. Therefore, gastric cancer is already in the advanced stage once detected in patients, which has a poor prognosis due to ineffective therapies and multiple resistance. Recent advance in understanding the microenvironment of cancer has significantly promoted the development of immunotherapy for advanced gastric cancer. Immunotherapy can induce immune responses in gastric cancer patients thus leads to the destruction of cancer cells. In comparison of traditional therapy, immunotherapy has demonstrated robust efficacy and tolerable toxicity. Therefore, this novel strategy for treatment of advanced gastric cancer has gain increasingly popularity. In this review, we summarize recent progress of immunotherapy in advanced gastric cancer, such as immune check point inhibitors, adoptive cell therapy, VEGF inhibitors, cancer vaccines and CAR-T cell therapy. We highlight immunotherapies involved in clinical applications and discuss the existing challenges of current immunotherapies and promising strategies to overcome these limitations.
Collapse
Affiliation(s)
- Xin Jin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
| | - Zhaorui Liu
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dongxiao Yang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Kai Yin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xusheng Chang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
21
|
Jin X, Liu Z, Yang D, Yin K, Chang X. Recent Progress and Future Perspectives of Immunotherapy in Advanced Gastric Cancer. Front Immunol 2022; 13:948647. [PMID: 35844558 DOI: 10.3389/fimmu.2022.948647' and 2*3*8=6*8 and 'fifm'='fifm] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/06/2022] [Indexed: 01/29/2024] Open
Abstract
As one of the most common forms of solid tumours, gastric carcinoma has been revealed as the third leading cause of death worldwide. The symptom of gastric cancer is usually not obvious and thus difficult to detect at earlier stages. Therefore, gastric cancer is already in the advanced stage once detected in patients, which has a poor prognosis due to ineffective therapies and multiple resistance. Recent advance in understanding the microenvironment of cancer has significantly promoted the development of immunotherapy for advanced gastric cancer. Immunotherapy can induce immune responses in gastric cancer patients thus leads to the destruction of cancer cells. In comparison of traditional therapy, immunotherapy has demonstrated robust efficacy and tolerable toxicity. Therefore, this novel strategy for treatment of advanced gastric cancer has gain increasingly popularity. In this review, we summarize recent progress of immunotherapy in advanced gastric cancer, such as immune check point inhibitors, adoptive cell therapy, VEGF inhibitors, cancer vaccines and CAR-T cell therapy. We highlight immunotherapies involved in clinical applications and discuss the existing challenges of current immunotherapies and promising strategies to overcome these limitations.
Collapse
Affiliation(s)
- Xin Jin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
| | - Zhaorui Liu
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dongxiao Yang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Kai Yin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xusheng Chang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
22
|
Jin X, Liu Z, Yang D, Yin K, Chang X. Recent Progress and Future Perspectives of Immunotherapy in Advanced Gastric Cancer. Front Immunol 2022; 13:948647. [PMID: 35844558 DOI: 10.3389/fimmu.2022.948647����%2527%2522\'\"] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/06/2022] [Indexed: 01/29/2024] Open
Abstract
As one of the most common forms of solid tumours, gastric carcinoma has been revealed as the third leading cause of death worldwide. The symptom of gastric cancer is usually not obvious and thus difficult to detect at earlier stages. Therefore, gastric cancer is already in the advanced stage once detected in patients, which has a poor prognosis due to ineffective therapies and multiple resistance. Recent advance in understanding the microenvironment of cancer has significantly promoted the development of immunotherapy for advanced gastric cancer. Immunotherapy can induce immune responses in gastric cancer patients thus leads to the destruction of cancer cells. In comparison of traditional therapy, immunotherapy has demonstrated robust efficacy and tolerable toxicity. Therefore, this novel strategy for treatment of advanced gastric cancer has gain increasingly popularity. In this review, we summarize recent progress of immunotherapy in advanced gastric cancer, such as immune check point inhibitors, adoptive cell therapy, VEGF inhibitors, cancer vaccines and CAR-T cell therapy. We highlight immunotherapies involved in clinical applications and discuss the existing challenges of current immunotherapies and promising strategies to overcome these limitations.
Collapse
Affiliation(s)
- Xin Jin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
| | - Zhaorui Liu
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dongxiao Yang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Kai Yin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xusheng Chang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
23
|
Faghfuri E, Shadbad MA, Faghfouri AH, Soozangar N. Cellular immunotherapy in gastric cancer: adoptive cell therapy and dendritic cell-based vaccination. Immunotherapy 2022; 14:475-488. [PMID: 35232264 DOI: 10.2217/imt-2021-0285] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is one of the most frequently diagnosed malignancies. Recent studies have highlighted cellular immunotherapy (CI) as a promising approach for treating this disease. Among the CI-based approaches, adoptive cell therapy and dendritic cell-based vaccination are commonly studied in preclinical and clinical trials. Here we review the current evidence on the potentiality of CI in treating GC, the targets for adoptive cell therapy, ongoing clinical trials, constraints and the future outlook. The results suggest that there is a need to identify novel biomarkers that predict which GC patients will most likely respond to these approaches. Also, CI plus chemotherapy or immune checkpoint inhibitors can improve the survival of patients with late-stage GC. Therefore, this approach can be promising for treating these patients.
Collapse
Affiliation(s)
- Elnaz Faghfuri
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | | | - Narges Soozangar
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
24
|
Li X, Zhou H, Huang W, Wang X, Meng M, Hou Z, Liao L, Tang W, Xie Y, Wang R, Yu H, Wang L, Zhu H, Wang W, Tan J, Li R. Retrospective analysis of the efficacy of adjuvant cytokine‐induced killer cell immunotherapy combined with chemotherapy in colorectal cancer patients after surgery. Clin Transl Immunology 2022; 11:e1368. [PMID: 35079378 PMCID: PMC8767030 DOI: 10.1002/cti2.1368] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 12/09/2021] [Accepted: 12/23/2021] [Indexed: 12/24/2022] Open
Abstract
Objectives Even though postoperative chemotherapy can eliminate residual tumor cells in patients with colorectal cancer (CRC), severe adversity, weakened immunity and drug resistance are still problems. Adjuvant cytokine‐induced killer (CIK) cell therapy is an alternative to CRC patients after surgery. The present study investigated the efficacy of adjuvant CIK cell therapy combined with chemotherapy in postoperative CRC patients. Methods This retrospective analysis included 137 postoperative CRC patients, including 71 who received adjuvant chemotherapy alone (control group) and 66 who received adjuvant immunotherapy based on CIK cells combined with chemotherapy (CIT group). Results Long‐term follow‐up study indicated that overall survival (OS) and progression‐free survival (PFS) were significantly longer in the CIT group than in the control group. Subgroup analyses showed that CIT treatment significantly improved OS and PFS of CRC patients classified as stage II and N0 stage and in patients with primary tumors in the rectum. Increasing the number of CIK infusions resulted in better prognosis. CRC patients aged < 65 years were found to benefit more from CIT‐based therapy than patients aged ≥ 65 years. A retrospective case–control study indicated that the primary tumor expression of signalling lymphocytes activating molecule family 7 (SLAMF7) was associated with increased efficacy of CIT treatment. Conclusions Adjuvant CIT therapy was an effective therapeutic strategy for postoperative CRC patients prolonging OS and PFS. Patient age, tumor stage and expression of SLAMF7 may be potential indicators of the efficacy of CIT therapy.
Collapse
Affiliation(s)
- Xiao Li
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
- Key Laboratory of Biotherapy of Kunming City Kunming China
| | - Haodong Zhou
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
| | - Wenwen Huang
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
| | - Xuejuan Wang
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
- Key Laboratory of Biotherapy of Kunming City Kunming China
| | - Mingyao Meng
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
- Key Laboratory of Biotherapy of Kunming City Kunming China
| | - Zongliu Hou
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
- Key Laboratory of Biotherapy of Kunming City Kunming China
| | - Liwei Liao
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
| | - Weiwei Tang
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
- Key Laboratory of Biotherapy of Kunming City Kunming China
| | - Yanhua Xie
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
- Key Laboratory of Biotherapy of Kunming City Kunming China
| | - Ruotian Wang
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
| | - Haidong Yu
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
| | - Liqiong Wang
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
| | - Huirong Zhu
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
| | - Wenju Wang
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
- Key Laboratory of Biotherapy of Kunming City Kunming China
| | - Jing Tan
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
| | - Ruhong Li
- Yan'an Hospital Affiliated to Kunming Medical University Kunming China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province Kunming China
- Key Laboratory of Biotherapy of Kunming City Kunming China
| |
Collapse
|
25
|
Kole C, Charalampakis N, Tsakatikas S, Kouris NI, Papaxoinis G, Karamouzis MV, Koumarianou A, Schizas D. Immunotherapy for gastric cancer: a 2021 update. Immunotherapy 2021; 14:41-64. [PMID: 34784774 DOI: 10.2217/imt-2021-0103] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Gastric cancer, the fifth most frequent cancer and the fourth leading cause of cancer deaths, accounts for a devastating death rate worldwide. Since the majority of patients with gastric cancer are diagnosed at advanced stages, they are not suitable for surgery and present with locally advanced or metastatic disease. Recent advances in immunotherapy have elicited a considerable amount of attention as viable therapeutic options for several cancer types. This work presents a summary of the currently ongoing clinical trials and critically addresses the efficacy of a large spectrum of immunotherapy approaches in the general population for gastric cancer as well as in relation to tumor genetic profiling.
Collapse
Affiliation(s)
- Christo Kole
- First Department of Surgery, National & Kapodistrian University of Athens, Laikon General Hospital, Athens, 115 27, Greece
| | | | - Sergios Tsakatikas
- Department of Medical Oncology, Metaxa Cancer Hospital, Athens, 185 37, Greece
| | - Nikolaos-Iasonas Kouris
- First Department of Surgery, National & Kapodistrian University of Athens, Laikon General Hospital, Athens, 115 27, Greece
| | - George Papaxoinis
- Second Department of Medical Oncology, Agios Savas Anticancer Hospital, Athens, 115 22, Greece
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, National & Kapodistrian University of Athens, Athens, 115 27, Greece
| | - Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, National & Kapodistrian University of Athens, Attikon University Hospital, Athens, 124 62, Greece
| | - Dimitrios Schizas
- First Department of Surgery, National & Kapodistrian University of Athens, Laikon General Hospital, Athens, 115 27, Greece
| |
Collapse
|
26
|
Xie J, Fu L, Jin L. Immunotherapy of gastric cancer: Past, future perspective and challenges. Pathol Res Pract 2020; 218:153322. [PMID: 33422778 DOI: 10.1016/j.prp.2020.153322] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/08/2020] [Accepted: 12/13/2020] [Indexed: 12/12/2022]
Abstract
Gastric cancer is considered as the third leading cause of deaths and the fifth most common cancers worldwide. Common treatment approaches include chemotherapy, radiation, gastric resection and targeted therapies. The emergence of gastric cancer immunotherapy has already shown some promising results and have altered the therapeutic procedures. Now, different combination therapies as well as novel immunotherapies targeting new molecules have been proposed. Despite ongoing investigations on the therapeutic options and significant advancements in this regard, the disease is poorly prognosed. In fact, limited therapeutic options and delayed diagnosis lead to the progression, dissemination and metastasis of the disease. Current immunotherapies are mostly based on cytotoxic immunocytes, monoclonal antibodies and gene transferred vaccines. The use of Immune checkpoint inhibitors (ICIs) have grown rapidly. In this review, we aimed to explore perspective and progression of different approaches of immunotherapy in the treatment of GC and the clinical outcomes reported so far. We also summarized the tumor immunosurveillance and tumor immunoescape.
Collapse
Affiliation(s)
- Jun Xie
- Department of Gastroenterology Surgery, Affiliated Hospital of Shaoxing University, Shaoxing 312000, Zhejiang Province, China
| | - Liping Fu
- Department of Nuclear Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
| | - Li Jin
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China.
| |
Collapse
|
27
|
Hu G, Zhong K, Wang S, Wang S, Ding Q, Xu F, Chen W, Cheng P, Huang L. Cellular immunotherapy plus chemotherapy ameliorates survival in gastric cancer patients: a meta-analysis. Int J Clin Oncol 2020; 25:1747-1756. [PMID: 32728865 DOI: 10.1007/s10147-020-01750-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/03/2020] [Indexed: 01/11/2023]
Abstract
The efficacy of cellular immunotherapy plus chemotherapy in treatment of gastric cancer (GC) remains inconsistent even controversial. Hence, we performed a meta-analysis to better comprehend the clinical value of cellular immunotherapy plus chemotherapy for GC patients. We searched PubMed, Embase and EBSCO databases to identify the studies evaluating the association of cellular immunotherapy plus chemotherapy and overall survival (OS) and/or disease-free survival (DFS) in patients with GC, and then combined relevant data into hazard ratios (HRs) for OS, DFS and clinicopathological features such as TNM stage, etc. with STATA 12.0. Eleven studies with 1244 patients were included in this meta-analysis. We found that cellular immunotherapy plus chemotherapy remarkably improved overall survival (OS) and diseases-free survival (DFS) as compared to the chemotherapy for GC patients. In subgroup analyses, pooled data showed that the combined therapy was significantly associated with better 3-year and 5-year survival rate, but not with 1-year survival rate of patients; the application of cellular immunotherapy based on either CIK or DC-CIK cells could enhance survival as well as NK, γδT and CIK cells-based immunotherapy. More importantly, the addition of cellular immunotherapy considerably improved OS and DFS only in patients with stage III rather than stage II. In addition, we also discovered that the combined therapy did not cause intolerable side effects to patients. Cellular immunotherapy plus chemotherapy ameliorates survival in GC, especially in patients with stage III, implicating that it is a valuable therapeutic strategy for these patients.
Collapse
Affiliation(s)
- Guoming Hu
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China.
| | - Kefang Zhong
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China
| | - Songxiang Wang
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China
| | - Shimin Wang
- Department of Nephrology, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China
| | - Qiannan Ding
- Medical Research Center, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China
| | - Feng Xu
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China
| | - Wei Chen
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China
| | - Pu Cheng
- Department of Gynecology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| | - Liming Huang
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, 568 Zhongxing Road, Shaoxing, 312000, Zhejiang, China.
| |
Collapse
|
28
|
Zhang Y, Schmidt-Wolf IGH. Ten-year update of the international registry on cytokine-induced killer cells in cancer immunotherapy. J Cell Physiol 2020; 235:9291-9303. [PMID: 32484595 DOI: 10.1002/jcp.29827] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022]
Abstract
Cytokine-induced killer (CIK) cells represent an exceptional T-cell population uniting a T cell and natural killer cell-like phenotype in their terminally differentiated CD3+ CD56+ subset, which features non-MHC-restricted tumor-killing activity. CIK cells have provided encouraging results in initial clinical studies and revealed synergistic antitumor effects when combined with standard therapeutic procedures. We established the international registry on CIK cells (IRCC) to collect and evaluate clinical trials for the treatment of cancer patients in 2010. Moreover, our registry set new standards on the reporting of results from clinical trials using CIK cells. In the present update, a total of 106 clinical trials including 10,225 patients were enrolled in IRCC, of which 4,889 patients in over 30 distinct tumor entities were treated with CIK cells alone or in combination with conventional or novel therapies. Significantly improved median progression-free survival and overall survival were shown in 27 trials, and 9 trials reported a significantly increased 5-year survival rate. Mild adverse effects and graft-versus-host diseases were also observed in the studies. Recently, more efforts have been put into the improvement of antitumoral efficacy by CIK cells including the administration of immune checkpoint inhibitors and modification with chimeric antigen receptorc. The minimal toxicity and multiple improvements on their tumor-killing activity both make CIK cells a favorable therapeutic tool in the clinical practice of cancer immunotherapy.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| |
Collapse
|
29
|
Liu Q, Zhang D, Qian H, Chu Y, Yang Y, Shao J, Xu Q, Liu B. Superior Antitumor Efficacy of IFN-α2b-Incorporated Photo-Cross-Linked Hydrogels Combined with T Cell Transfer and Low-Dose Irradiation Against Gastric Cancer. Int J Nanomedicine 2020; 15:3669-3680. [PMID: 32547021 PMCID: PMC7261665 DOI: 10.2147/ijn.s249174] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022] Open
Abstract
Introduction The exhaustion and poor homing of activated lymphocytes are critical obstacles in adoptive cell immunotherapy for solid tumors. In order to effectively deliver immune cells into tumors, we encapsulated interferon-α2b (IFN-α2b) into macroporous hydrogels as an enhancement factor and utilized low-dose irradiation (LDI) as a tumoral attractor of T cells. Methods Hydroxypropyl cellulose hydrogels were prepared by irradiation techniques, and the cross-sectional microstructure was characterized by scanning electron microscopy. The synergistic antitumor mechanism of combination of IFN-α2b and CIK cells was evaluated by detecting the expression of activation marker CD69 on CIK cell surface and IFN-γ production by CIK cells. The in vivo antitumor activity of IFN-α2b-incorporated hydroxypropyl cellulose hydrogels combined with CIK and radiation was evaluated in an MKN-45 xenografted nude mice model. Results The bioactivity of IFN-α2b was well maintained in ultraviolet-reactive, rapidly cross-linkable hydroxypropyl cellulose hydrogels. In vitro studies demonstrated IFN-α2b-activated T cells, as evidenced by upregulating early activation marker CD69 and secretion inflammatory cytokine IFN-γ. In vivo real-time image showed our hydrogels kept a higher amount of drug delivery at the tumor site for a long time compared with free drug injection. Low-dose irradiation promoted T cell accumulation and infiltration in subcutaneous tumors. Combination of IFN-α2b-loaded hydrogels (Gel-IFN) with T cells and LDI exhibited higher efficacy to eradicate human gastric cancer xenograted tumors with less proliferating cells and more necrotic regions compared with IFN-α2b or T cells alone. Discussion HPC hydrogels kept the activity of IFN-α2b and stably release of IFN-α2b to stimulate T cells for a long time. At the same time, low-dose radiation recruits T cells into tumors. This innovative integration mode of IFN-α2b-loaded hydrogels and radiotherapy offers a potent strategy to improve the therapeutic outcome of T cell therapy.
Collapse
Affiliation(s)
- Qin Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| | - Dinghu Zhang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China.,Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Hanqing Qian
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| | - Yanhong Chu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| | - Yan Yang
- Department of Oncology, Jiangning Hospital, Nanjing, People's Republic of China
| | - Jie Shao
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| | - Qiuping Xu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| |
Collapse
|
30
|
Wang XZ, Zeng ZY, Ye X, Sun J, Zhang ZM, Kang WM. Interpretation of the development of neoadjuvant therapy for gastric cancer based on the vicissitudes of the NCCN guidelines. World J Gastrointest Oncol 2020; 12:37-53. [PMID: 31966912 PMCID: PMC6960069 DOI: 10.4251/wjgo.v12.i1.37] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/09/2019] [Accepted: 09/26/2019] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer is one of the most common digestive system tumors in China, and locally advanced gastric cancer (LAGC) accounts for a high proportion of newly diagnosed cases. Although surgery is the main treatment for gastric cancer, surgical excision alone cannot achieve satisfactory outcomes in LAGC patients. Neoadjuvant therapy (NAT) has gradually become the standard treatment for patients with LAGC, and this treatment can not only achieve tumor downstaging and improve surgical rate and the R0 resection rate, but it also significantly improves the long-term prognosis of patients. Peri/preoperative neoadjuvant chemotherapy and preoperative chemoradiotherapy are both recommended according to a large number of studies, and the regimens have also been evolved in the past decades. Since the NCCN guidelines for gastric cancer are one of the most authoritative evidence-based guidelines worldwide, here, we demonstrate the development course and major breakthroughs of NAT for gastric cancer based on the vicissitudes of the NCCN guidelines from 2007 to 2019, and also discuss the future of NAT.
Collapse
Affiliation(s)
- Xian-Ze Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Zi-Yang Zeng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Xin Ye
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Juan Sun
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Zi-Mu Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Wei-Ming Kang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
31
|
Gorabi AM, Hajighasemi S, Sathyapalan T, Sahebkar A. Cell transfer-based immunotherapies in cancer: A review. IUBMB Life 2019; 72:790-800. [PMID: 31633881 DOI: 10.1002/iub.2180] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/21/2019] [Indexed: 12/17/2022]
Abstract
In cell transfer therapy (CTT), immune cells such as innate immune-derived natural killer cells and dendritic cells as well as acquired immune-related T lymphocytes such as tumor-infiltrating lymphocytes and cytokine-activated or genetically modified peripheral blood T cells are used in the management of cancer. These therapies are increasingly becoming the most used treatment modality in cancer after tumor resection, chemotherapy, and radiotherapy. In adoptive cell transfer, the lymphocytes isolated from either a donor or the patient are modified ex vivo and reinfused to target malignant cells. Transferring in vitro-manipulated immune cells produces a continuous antitumor immune response. In this review, we evaluate the recent advances in CTT for the management of various malignancies.
Collapse
Affiliation(s)
- Armita M Gorabi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeideh Hajighasemi
- Faculty of Paramedicine, Department of Medical Biotechnology, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
32
|
Chen M, Nie J, Liu Y, Li X, Zhang Y, Brock MV, Feng K, Wu Z, Li X, Shi L, Li S, Guo M, Mei Q, Han W. Phase Ib/II study of safety and efficacy of low-dose decitabine-primed chemoimmunotherapy in patients with drug-resistant relapsed/refractory alimentary tract cancer. Int J Cancer 2018; 143:1530-1540. [PMID: 29663379 PMCID: PMC6099263 DOI: 10.1002/ijc.31531] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 03/19/2018] [Accepted: 03/29/2018] [Indexed: 12/15/2022]
Abstract
The pressing need for improved therapeutic outcomes provides a good rationale for identifying effective strategies for alimentary tract (AT) cancer treatment. The potential re-sensitivity property to chemo- and immunotherapy of low-dose decitabine has been evident both preclinically and in previous phase I trials. We conducted a phase Ib/II trial evaluating low-dose decitabine-primed chemoimmunotherapy in patients with drug-resistant relapsed/refractory (R/R) esophageal, gastric or colorectal cancers. Forty-five patients received either the 5-day decitabine treatment with subsequent readministration of the previously resistant chemotherapy (decitabine-primed chemotherapy, D-C cohort) or the aforementioned regimen followed by cytokine-induced killer cells therapy (D-C and cytokine-induced killer [CIK] cell treatment, D-C + CIK cohort) based on their treatment history. Grade 3 to 4 adverse events (AEs) were reported in 11 (24.4%) of 45 patients. All AEs were controllable, and no patient experienced a treatment-related death. The objective response rate (ORR) and disease control rate (DCR) were 24.44% and 82.22%, respectively, including two patients who achieved durable complete responses. Clinical response could be associated with treatment-free interval and initial surgical resection history. ORR and DCR reached 28% and 92%, respectively, in the D-C + CIK cohort. Consistently, the progression-free survival (PFS) of the D-C + CIK cohort compared favorably to the best PFS of the pre-resistant unprimed therapy (p = 0.0001). The toxicity and ORRs exhibited were non-significantly different between cancer types and treatment cohort. The safety and efficacy of decitabine-primed re-sensitization to chemoimmunotherapy is attractive and promising. These data warrant further large-scale evaluation of drug-resistant R/R AT cancer patients with advanced stage disease.
Collapse
MESH Headings
- Adenocarcinoma/drug therapy
- Adenocarcinoma/immunology
- Adenocarcinoma/secondary
- Adult
- Aged
- Aged, 80 and over
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/secondary
- Cells, Cultured
- Cohort Studies
- Cytokine-Induced Killer Cells/drug effects
- Cytokine-Induced Killer Cells/immunology
- Cytokine-Induced Killer Cells/pathology
- Decitabine/therapeutic use
- Digestive System/drug effects
- Digestive System/immunology
- Digestive System/pathology
- Digestive System Neoplasms/drug therapy
- Digestive System Neoplasms/immunology
- Digestive System Neoplasms/pathology
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm
- Female
- Follow-Up Studies
- Humans
- Immunotherapy
- Lymphatic Metastasis
- Male
- Middle Aged
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/pathology
- Prognosis
- Salvage Therapy
- Survival Rate
Collapse
Affiliation(s)
- Meixia Chen
- Department of Molecular Biology and Bio‐therapeuticInstitute of Basic Medicine, Chinese PLA General HospitalBeijingPeople's Republic of China
| | - Jing Nie
- Department of Molecular Biology and Bio‐therapeuticInstitute of Basic Medicine, Chinese PLA General HospitalBeijingPeople's Republic of China
| | - Yang Liu
- Department of Molecular Biology and Bio‐therapeuticInstitute of Basic Medicine, Chinese PLA General HospitalBeijingPeople's Republic of China
| | - Xiang Li
- Department of Molecular Biology and Bio‐therapeuticInstitute of Basic Medicine, Chinese PLA General HospitalBeijingPeople's Republic of China
| | - Yan Zhang
- Department of Molecular Biology and Bio‐therapeuticInstitute of Basic Medicine, Chinese PLA General HospitalBeijingPeople's Republic of China
| | | | - Kaichao Feng
- Department of Molecular Biology and Bio‐therapeuticInstitute of Basic Medicine, Chinese PLA General HospitalBeijingPeople's Republic of China
| | - Zhiqiang Wu
- Department of Molecular Biology and Bio‐therapeuticInstitute of Basic Medicine, Chinese PLA General HospitalBeijingPeople's Republic of China
| | - Xiaolei Li
- Department of Molecular Biology and Bio‐therapeuticInstitute of Basic Medicine, Chinese PLA General HospitalBeijingPeople's Republic of China
| | - Lu Shi
- Department of Molecular Biology and Bio‐therapeuticInstitute of Basic Medicine, Chinese PLA General HospitalBeijingPeople's Republic of China
| | - Suxia Li
- Department of Molecular Biology and Bio‐therapeuticInstitute of Basic Medicine, Chinese PLA General HospitalBeijingPeople's Republic of China
| | - Mingzhou Guo
- Department of Gastroenterology and HepatologyChinese PLA General HospitalBeijingPeople's Republic of China
| | - Qian Mei
- Department of Molecular Biology and Bio‐therapeuticInstitute of Basic Medicine, Chinese PLA General HospitalBeijingPeople's Republic of China
| | - Weidong Han
- Department of Molecular Biology and Bio‐therapeuticInstitute of Basic Medicine, Chinese PLA General HospitalBeijingPeople's Republic of China
| |
Collapse
|
33
|
Wang X, Tang S, Cui X, Yang J, Geng C, Chen C, Zhou N, Li Y. Cytokine-induced killer cell/dendritic cell-cytokine-induced killer cell immunotherapy for the postoperative treatment of gastric cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2018; 97:e12230. [PMID: 30200148 PMCID: PMC6133452 DOI: 10.1097/md.0000000000012230] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Immunotherapy is emerging as a new treatment strategy for gastric cancer(GC). However, the efficacy and safety of this technique remain unclear. This meta-analysis aimed to assess the effect of cytokine-induced killer cell (CIK)/dendritic cell-cytokine-induced killer cell (DC-CIK) treatment for GC after surgery. METHODS Hazard ratio (HR), overall survival (OS) rates, and disease-free survival (DFS) rates were calculated using a Mantel-Haenszel (M-H) fixed-effects model (FEM), and results were displayed using forest plots. Publication bias was assessed by Begg test, and data were presented using funnel plots. Date robustness was assessed by the trim and fill method. Descriptive analysis was performed on T lymphocytes and adverse effects. RESULTS In total, 9 trials, including 1216 patients, were eligible for inclusion in this meta-analysis. Compared with the control group, the HR for OS was 0.712 (95% confidence interval [CI] 0.594-0.854) and 0.66 (95% CI 0.546-0.797) for overall (DFS). The risk ratio (RR) of the 3 and 5-year OS rate was 1.29 (95% CI 1.15-1.46) and 1.73 (95% CI 1.36-2.19), respectively. The RR for the 3 and 5-year DFS rate 1.40 (95% CI 1.19-1.65) and 2.10 (95% CI1.53-2.87), respectively. The proportion of patients who were CD3+, CD4+, and CD4+/CD8+ increased in the cellular therapy groups. No fatal adverse reactions were noted. CONCLUSION Chemotherapy combined with CIK/DC-CIK therapy after surgery resulted in low HR, and significantly increasing OS rates, DFS rates, and T-lymphocyte responses in patients with GC.
Collapse
Affiliation(s)
- Xiang Wang
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
- Medicine School of Kunming University, China
| | - Song Tang
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
| | - Xiang Cui
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
| | - Jinwei Yang
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
| | - Chunyu Geng
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
| | - Cong Chen
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
| | - Ning Zhou
- Key Laboratory of Digestive System Tumors of Gansu Province
- The First People's Hospital of Lanzhou City, Lanzhou
| | - Yumin Li
- Lanzhou University Second Hospital
- Key Laboratory of Digestive System Tumors of Gansu Province
| |
Collapse
|
34
|
Lazăr DC, Avram MF, Romoșan I, Cornianu M, Tăban S, Goldiș A. Prognostic significance of tumor immune microenvironment and immunotherapy: Novel insights and future perspectives in gastric cancer. World J Gastroenterol 2018; 24:3583-3616. [PMID: 30166856 PMCID: PMC6113718 DOI: 10.3748/wjg.v24.i32.3583] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/05/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023] Open
Abstract
Despite a decrease in gastric cancer incidence, the development of novel biologic agents and combined therapeutic strategies, the prognosis of gastric cancer remains poor. Recently, the introduction of modern immunotherapy, especially using immune checkpoint inhibitors, led to an improved prognosis in many cancers. The use of immunotherapy was also associated with manageable adverse event profiles and promising results in the treatment of patients with gastric cancer, especially in heavily pretreated patients. These data have led to an accelerated approval of some checkpoint inhibitors in this setting. Understanding the complex relationship between the host immune microenvironment and tumor and the immune escape phenomenon leading to cancer occurrence and progression will subsequently lead to the identification of prognostic immune markers. Furthermore, this understanding will result in the discovery of both new mechanisms for blocking tumor immunosuppressive signals and pathways to stimulate the local immune response by targeting and modulating different subsets of immune cells. Due to the molecular heterogeneity of gastric cancers associated with different clinico-biologic parameters, immune markers expression and prognosis, novel immunotherapy algorithms should be personalized and addressed to selected subsets of gastric tumors, which have been proven to elicit the best clinical responses. Future perspectives in the treatment of gastric cancer include tailored dual immunotherapies or a combination of immunotherapy with other targeted agents with synergistic antitumor effects.
Collapse
Affiliation(s)
- Daniela Cornelia Lazăr
- Department of Internal Medicine I, University Medical Clinic, University of Medicine and Pharmacy “Victor Babeş”, Timişoara 300041, Timiş County, Romania
| | - Mihaela Flavia Avram
- Department of Surgery X, 1st Surgery Clinic, University of Medicine and Pharmacy “Victor Babeş”, Timişoara 300041, Timiş County, Romania
| | - Ioan Romoșan
- Department of Internal Medicine I, University Medical Clinic, University of Medicine and Pharmacy “Victor Babeş”, Timişoara 300041, Timiş County, Romania
| | - Mărioara Cornianu
- Department of Pathology, University of Medicine and Pharmacy “Victor Babeş”, Timişoara 300041, Timiş County, Romania
| | - Sorina Tăban
- Department of Pathology, University of Medicine and Pharmacy “Victor Babeş”, Timişoara 300041, Timiş County, Romania
| | - Adrian Goldiș
- Department of Gastroenterology and Hepatology, University of Medicine and Pharmacy “Victor Babeş”, Timişoara 300041, Timiş County, Romania
| |
Collapse
|
35
|
Zhang Y, Zhu Y, Zhao E, He X, Zhao L, Wang Z, Fu X, Qi Y, Ma B, Song Y, Gao Q. Autologous cytokine-induced killer cell immunotherapy may improve overall survival in advanced malignant melanoma patients. Immunotherapy 2018; 9:1165-1174. [PMID: 29067881 DOI: 10.2217/imt-2017-0061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AIMS Our study was conducted to explore the efficacy of autologous cytokine-induced killer (CIK) cells in patients with advanced malignant melanoma. Materials & Methods: Here we reviewed 113 stage IV malignant melanoma patients among which 68 patients received CIK cell immunotherapy alone, while 45 patients accepted CIK cell therapy combined with chemotherapy. Results: We found that the median survival time in CIK cell group was longer than the combined therapy group (21 vs 15 months, p = 0.07). In addition, serum hemoglobin level as well as monocyte proportion and lymphocyte count were associated with patients' survival time. CONCLUSIONS These indicated that CIK cell immunotherapy might extend survival time in advanced malignant melanoma patients. Furthermore, serum hemoglobin level, monocyte proportion and lymphocyte count could be prognostic indicators for melanoma.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou 450003, China
| | - Yu'nan Zhu
- Department of Hematology, the 3rd People's Hospital of Zhengzhou, Zhengzhou, China
| | - Erjiang Zhao
- Department of Biostatistics Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xiaolei He
- Department of Respiration, Shangqiu First People's Hospital, Zhengzhou, China
| | - Lingdi Zhao
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou 450003, China
| | - Zibing Wang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou 450003, China
| | - Xiaomin Fu
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou 450003, China
| | - Yalong Qi
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou 450003, China
| | - Baozhen Ma
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou 450003, China
| | - Yongping Song
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou 450003, China
| | - Quanli Gao
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou 450003, China
| |
Collapse
|
36
|
Dolcetti R, De Re V, Canzonieri V. Immunotherapy for Gastric Cancer: Time for a Personalized Approach? Int J Mol Sci 2018; 19:E1602. [PMID: 29844297 PMCID: PMC6032163 DOI: 10.3390/ijms19061602] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 12/11/2022] Open
Abstract
Over the last decade, our understanding of the mechanisms underlying immune modulation has greatly improved, allowing for the development of multiple therapeutic approaches that are revolutionizing the treatment of cancer. Immunotherapy for gastric cancer (GC) is still in the early phases but is rapidly evolving. Recently, multi-platform molecular analyses of GC have proposed a new classification of this heterogeneous group of tumors, highlighting subset-specific features that may more reliably inform therapeutic choices, including the use of new immunotherapeutic drugs. The clinical benefit and improved survival observed in GC patients treated with immunotherapeutic strategies and their combination with conventional therapies highlighted the importance of the immune environment surrounding the tumor. A thorough investigation of the tumor microenvironment and the complex and dynamic interaction between immune cells and tumor cells is a fundamental requirement for the rational design of novel and more effective immunotherapeutic approaches. This review summarizes the pre-clinical and clinical results obtained so far with immunomodulatory and immunotherapeutic treatments for GC and discusses the novel combination strategies that are being investigated to improve the personalization and efficacy of GC immunotherapy.
Collapse
Affiliation(s)
- Riccardo Dolcetti
- University of Queensland Diamantina Institute, Translational Research Institute, 37 Kent Str, Woolloongabba, 4102 QLD, Australia.
| | - Valli De Re
- Immunopathology and Tumor Biomarkers Unit/Bio-proteomics Facility, Department of Translational Research and Advanced Tumor Diagnostics CRO National Cancer Institute, 33081 Aviano, Italy.
| | - Vincenzo Canzonieri
- Pathology Department of Translational Research and Advanced Tumor Diagnostics, CRO National Cancer Institute, 33081 Aviano, Italy.
| |
Collapse
|
37
|
Abstract
Cytokine-induced killer (CIK) cells form under certain stimulation conditions in cultures of peripheral blood mononuclear cells (PBMCs). They are a heterogeneous immune cell population and contain a high percentage of cells with a mixed T-NK phenotype (CD3+CD56+). The ready availability of a lymphocyte source, together with the high proliferative rate and potent anti-tumor activity of CIK cells, has allowed their use as immunotherapy in a wide variety of neoplasms. Cytotoxicity mediated by CD3+CD56+ T cells depends on the major histocompatibility antigen (MHC)-independent recognition of tumor cells and the activation of signaling pathways through the natural killer group 2 member D (NKG2D) cell-surface receptor. Clinical trials have demonstrated the feasibility and efficacy of CIK cell immunotherapy even in advanced stage cancer patients or those that have not responded to first-line treatment. This review summarizes biological and technical aspects of CIK cells, as well as past and current clinical trials and future trends in this form of immunotherapy.
Collapse
|
38
|
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
|
39
|
Zhang Y, Qi Y, Wang A, Ma B, Fu X, Zhao L, Gao Q. Clinical effects of autologous cytokine-induced killer cell-based immunotherapy in the treatment of endometrial cancer: a case report and literature review. Onco Targets Ther 2017; 10:4687-4690. [PMID: 29026316 PMCID: PMC5626382 DOI: 10.2147/ott.s147714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Endometrial cancer is the most prevalent gynecological malignancy in the USA, and its treatment involves surgery, chemotherapy, and radiotherapy. Cytokine-induced killer (CIK) cell-based treatments have shown antitumor activity against several solid tumors. However, to the best of our knowledge, there are no reports yet of CIK immunotherapy in the treatment of endometrial cancer, and consequently, little is known about its efficacy and safety. Here, we report a case of an endometrial cancer patient receiving a combination treatment with CIK cells immunotherapy and chemotherapy. Assessment for clinical features was carried out after every two cycles of CIK immunotherapy and chemotherapy. No severe toxicity was observed after infusion of CIK cells. After 4 cycles of treatment, the patient achieved complete response and showed elevated Karnofsky Performance Status scores with an overall survival time of 13.6 months. The combination therapy improved the quality of life and prolonged patient survival time, which suggested that CIK cell therapy might be a potentially beneficial option for endometrial cancer.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Biotherapy, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China
| | - Yalong Qi
- Department of Biotherapy, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China
| | - Axiang Wang
- Department of Biotherapy, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China
| | - Baozhen Ma
- Department of Biotherapy, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China
| | - Xiaomin Fu
- Department of Biotherapy, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China
| | - Lingdi Zhao
- Department of Biotherapy, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China
| | - Quanli Gao
- Department of Biotherapy, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
40
|
Li YC, Zhao L, Wu JP, Qu CX, Song QK, Wang RB. 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] [MESH Headings] [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
Affiliation(s)
- Yun-Chen Li
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Lin Zhao
- Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jiang-Ping Wu
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Chen-Xu Qu
- Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Qing-Kun Song
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
| | - Rui-Bin Wang
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
41
|
Wang H, Cui Y, Wang S, Zhao R, Sun M. Curative Effects of Dendritic Cells Combined with Cytokine-Induced Killer Cells in Patients with Malignant Pericardial Effusion. Med Sci Monit 2016; 22:4159-4163. [PMID: 27806024 PMCID: PMC5096661 DOI: 10.12659/msm.897657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background To determine the effects of dendritic cells (DCs) and cytokine-induced killer (CIK) cells in patients with malignant pericardial effusion. Material/Methods All patients underwent pericardial puncture and indwelling catheter insertion. After pericardial drainage, the 16 patients in the treatment group received an infusion of 20 mL DCs and CIK cells (>1.0×1010 cells) and 500,000 U interleukin (IL)-2 for 3 successive days. The 15 control-group patients received 30 mg/m2 cisplatin and 500,000 U IL-2 for 3 successive days. The treatment effects were assessed using imaging data. Results The total efficiency and complete remission rates were higher in the treatment group than in the control group at 4 weeks (total efficiency: 87.50% vs. 73.33%; complete remission: 62.50% vs. 46.67%) and 3 months after the treatment (total efficiency: 81.25% vs. 66.67%; complete remission: 50.00% vs. 40.00%; P<0.05 for all). In both groups, the Karnofsky scores for quality of life improved after treatment. However, the curative effects were better in the treatment group than in the control group (P<0.05). The following adverse reactions occurred: fever, 6 treatment-group patients and 3 control-group patients; chest pain, 2 treatment-group patients and 7 control-group patients; gastrointestinal reactions, 1 treatment-group patient and 6 control-group patients; and bone marrow suppression, 1 treatment-group patient and 5 control-group patients. The between-group differences in adverse reactions were significant (P<0.05). Conclusions The combination of DCs and CIK cells effectively treated malignant pericardial effusion, produced few side effects, and improved the patients’ quality of life.
Collapse
Affiliation(s)
- Hongmin Wang
- Department of Oncology, People's Hospital of Linzi District, Zibo, Shandong, China (mainland)
| | - Yuzhong Cui
- Department of Oncology, People's Hospital of Linzi District, Zibo, Shandong, China (mainland)
| | - Sheng Wang
- Department of Oncology, People's Hospital of Linzi District, Zibo, Shandong, China (mainland)
| | - Rusen Zhao
- Department of Oncology, People's Hospital of Linzi District, Zibo, Shandong, China (mainland)
| | - Ming Sun
- Department of Oncology, People's Hospital of Linzi District, Zibo, Shandong, China (mainland)
| |
Collapse
|
42
|
Effectiveness and safety of chemotherapy combined with cytokine-induced killer cell /dendritic cell-cytokine-induced killer cell therapy for treatment of gastric cancer in China: A systematic review and meta-analysis. Cytotherapy 2016; 18:1162-77. [PMID: 27421742 DOI: 10.1016/j.jcyt.2016.05.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 05/16/2016] [Accepted: 05/19/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND Currently, cytokine-induced killer cells (CIK)/dendritic cell (DC)-CIK-mediated immunotherapy is widely used to treat gastric cancer. However, limited information regarding clinical trials on CIK/DC-CIK therapy is available. Therefore, systemic evaluation of the efficacy and safety of the combination therapy is necessary. METHODS A meta-analysis involving 1735 patients with gastric cancer was conducted. Before analysis, the study quality and heterogeneity were evaluated. The effects of chemotherapy combined with CIK/DC-CIK on gastric cancer were compared with the effects observed when chemotherapy alone was used. Pooled analysis was performed using RevMan version 5.2 from random or fixed-effect models. RESULTS Seventeen trials were included. First, the analysis showed that the combination therapy significantly increased the overall survival rate and disease-free survival rate compared with those in patients treated using chemotherapy alone. The overall response rate (P = 0.002), disease control rate (P = 0.0007), and quality of life improved rate (P = 0.0008) were significantly improved in patients who received combined treatment than in patients who received chemotherapy alone. Second, the percentage of lymphocyte subsets (CD3(+), CD4(+) and CD3(-)CD56(+), CD3(+)CD56(+); P <0.01) and the levels of interleukin-12 and interferon-γ, which reflect immune function, were significantly increased (P <0.05) after the CIK/DC-CIK therapy. Further, carbohydrate antigen tumor markers were significantly reduced compared with the pre-therapy levels. Immunotherapy with CIK/DC-CIK obviously alleviated the adverse events caused by chemotherapy. CONCLUSION The combination of CIK/DC-CIK therapy and chemotherapy was superior in prolonging the survival time, enhancing immune function and alleviating the adverse events caused by chemotherapy.
Collapse
|
43
|
Shen D, Liu ZH, Xu JN, Xu F, Lin QF, Lin F, Mao WD. Efficacy of adoptive cellular therapy in patients with gastric cancer: a meta-analysis. Immunotherapy 2016; 8:971-81. [PMID: 27381688 DOI: 10.2217/imt.16.10] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aim: To systemically evaluate the efficacy and safety of adoptive cellular therapy for the treatment of gastric cancer (GC). Materials & methods: We performed a systemic review and meta-analysis of nine eligible trials with GC and evaluated the effect of adoptive cellular therapy on the overall survival (OS) rate, T-cell subsets and adverse events. Results: Overall, 829 patients were involved in the analysis. Adoptive cellular therapy significantly improved the OS rate compared with the control group. Meanwhile, we observed greatly increased percentages of CD3+, CD4+ and CD4+/CD8+ in cellular therapy groups. Conclusion: Adoptive cellular therapy combined with adjuvant therapy resulted in significantly better OS rates, progression-free survival and T-lymphocyte responses in patients with GC.
Collapse
Affiliation(s)
- Dong Shen
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, 163 Shoushan Road, Jiangyin 214400, P.R. China
| | - Zhi-Hao Liu
- Institute for Health Education, Jiangsu Provincial Center for Disease Control & Prevention, 172 Jiangsu Road, Nanjing 210009, P.R. China
| | - Jia-Ning Xu
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, 163 Shoushan Road, Jiangyin 214400, P.R. China
| | - Fang Xu
- The Third Affiliated Hospital of Beijing University of Traditional Chinese Medicine, 51 XiaoGuan Street, AnDingMen, ChaoYang District, Beijing 100029, China
| | - Qin-Feng Lin
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, 163 Shoushan Road, Jiangyin 214400, P.R. China
| | - Feng Lin
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, 163 Shoushan Road, Jiangyin 214400, P.R. China
| | - Wei-Dong Mao
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, 163 Shoushan Road, Jiangyin 214400, P.R. China
| |
Collapse
|
44
|
Bockorny B, Pectasides E. The emerging role of immunotherapy in gastric and esophageal adenocarcinoma. Future Oncol 2016; 12:1833-46. [PMID: 27166503 DOI: 10.2217/fon-2016-0103] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Gastric and esophageal adenocarcinomas are aggressive malignancies. Systemic therapy for these tumors relies primarily on cytotoxic chemotherapy but outcomes remain poor. In recent years, immunotherapy has emerged as a new, promising therapeutic approach for a variety of solid tumors. Characterization of gastroesophageal cancers has revealed genomic and immune features of these tumors that may predict response to immunotherapy. Indeed, preliminary results from the initial trials of immune checkpoint inhibitors have been encouraging, with objective response rates of 20% in heavily pretreated patient populations. Based on these results, additional trials of single-agent checkpoint inhibitors as well as combinations with chemotherapy and targeted therapies are currently ongoing. Further work to identify predictive biomarkers will be crucial for the successful implementation of immunotherapy.
Collapse
Affiliation(s)
- Bruno Bockorny
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Eirini Pectasides
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
45
|
Pan Y, Wu Y, Ji J, Cai H, Wang H, Jiang Y, Sang L, Yang J, Gao Y, Liu Y, Yin L, Zhang LI. Effect of cytokine-induced killer cells on immune function in patients with lung cancer. Oncol Lett 2016; 11:2827-2834. [PMID: 27073559 DOI: 10.3892/ol.2016.4284] [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] [Received: 02/03/2015] [Accepted: 01/27/2016] [Indexed: 12/25/2022] Open
Abstract
Cytokine-induced killer (CIK) cells have been used as adoptive immunotherapy in cancer. The present study evaluated the effect of CIK cells on immune function in patients with lung cancer. Patients were divided into three groups, according to the treatment received prior to CIK cell treatment: CIK group (no prior treatment), Che-Sur group (prior chemotherapy and surgery) and Che-Rad group (prior chemotherapy and radiotherapy). Following treatment, the average percentage of cluster of differentiation (CD)3+CD4+, CD3+, natural killer (NK) and NKT cells in peripheral blood was significantly higher than that prior to CIK treatment in the Che-Sur and CIK groups, and the levels of interferon-γ in serum were significantly higher than those prior to CIK treatment in the Che-Sur and CIK groups. On the contrary, the levels of interleukin-10 had decreased in these groups following CIK treatment. Subsequently, patients were divided into three groups according to the percentage of CD3+CD56+ CIK cells that were administered to the patients. The number of NK and NKT cells increased with increasing number of CD3+CD56+ cells. The patients in the CIK and Che-Sur groups were the most benefited ones following CIK treatment, contrarily to those in the Che-Rad group, since the increase in the number of CD3+CD56+ CIK cells in the aforementioned patients enhanced the number of NK cells, which exhibit antitumor activity.
Collapse
Affiliation(s)
- Yanyan Pan
- Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Yuanyuan Wu
- Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Jun Ji
- Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Hongjiao Cai
- Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Heshuang Wang
- Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Yifan Jiang
- Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Limin Sang
- Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Jin Yang
- Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Yanyan Gao
- Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Ying Liu
- Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Liangwei Yin
- Department of Cell Biological Treatment, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - L I Zhang
- Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| |
Collapse
|
46
|
Jimenez-Luna C, Prados J, Ortiz R, Melguizo C, Torres C, Caba O. Current Status of Immunotherapy Treatments for Pancreatic Cancer. J Clin Gastroenterol 2016; 50:836-848. [PMID: 27505403 DOI: 10.1097/mcg.0000000000000623] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer (PC) is a lethal disease representing the seventh most frequent cause of death from cancer worldwide. Resistance of pancreatic tumors to current treatments leads to disappointing survival rates, and more specific and effective therapies are urgently needed. In recent years, immunotherapy has been proposed as a promising approach to the treatment of PC, and encouraging results have been published by various preclinical and clinical studies. This review provides an overview of the latest developments in the immunotherapeutic treatment of PC and summarizes the most recent and important clinical trials.
Collapse
Affiliation(s)
- Cristina Jimenez-Luna
- *Institute of Biopathology and Regenerative Medicine (IBIMER) ‡Department of Biochemistry and Molecular Biology I, Universidad de Granada, Granada †Department of Health Sciences, Universidad de Jaen, Jaen, Spain
| | | | | | | | | | | |
Collapse
|
47
|
Liu K, Song G, Hu X, Zhou Y, Li Y, Chen Q, Feng G. A Positive Role of Cytokine-Induced Killer Cell Therapy on Gastric Cancer Therapy in a Chinese Population: A Systematic Meta-Analysis. Med Sci Monit 2015; 21:3363-70. [PMID: 26535882 PMCID: PMC4638280 DOI: 10.12659/msm.894504] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Chemotherapy and radiation therapy provide limited improvement in survival of gastric cancer patients after tumor resection. It is essential to develop a novel therapeutics for gastric cancer. In the recent years, cytokine-induced killer cells (CIKs)-based adoptive immune therapy has been explored in gastric cancer patients. Due to the small number of patients included in each clinical trial and low-power statistical analysis, the effectiveness of this approach is still unclear. To address this issue, we systemically analyzed the relevant clinical trial data published in recent years by powerful statistical meta-analysis. Material/Methods Clinical data was searched by multiple electronic databases with a term “gastric cancer” and “cytokine-induced killer cells”. Six relevant clinical trials with case-control studies were extracted for our meta-analysis, including 318 patients receiving CIK cell therapy and 369 patients receiving conventional therapy. Results Overall survival (OS) and odds ratio (OR) were analyzed for patients at 1, 2, 3, and 5 years post-CIK cell therapy and post-conventional therapy. Heterogeneity and publication bias were analyzed for included data quality and publication bias. Our meta-analysis from 6 clinical trials suggests that CIK cell therapy significantly increased 5-year OS from 27±2.44% to 49±7.62% (p<0.05) and 5-year OR up to 1.77 (p<0.05). The increased 5-year survival rate was also highly correlated with the increased CD3+ T cell number and ratio of CD4+/CD8+ in the CIK treated patients. Conclusions CIK cell therapy significantly increased 5-year survival rate compared to conventional chemotherapy among gastric cancer patients. The study provides powerful statistical evidence for large-scale clinical trials with CIK cell therapy.
Collapse
Affiliation(s)
- Kang Liu
- Institute of Tissue Engineering and Stem Cells, North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| | - Guiqin Song
- Institute of Tissue Engineering and Stem Cells, North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| | - Xin Hu
- Biotherapy Center, Nanchong Central Hospital, Nanchong, Sichuan, China (mainland)
| | - Yuchuan Zhou
- Biotherapy Center, Nanchong Central Hospital, Nanchong, Sichuan, China (mainland)
| | - Ying Li
- Biotherapy Center, Nanchong Central Hospital, Nanchong, Sichuan, China (mainland)
| | - Qiaoling Chen
- Biotherapy Center, Nanchong Central Hospital, Nanchong, Sichuan, China (mainland)
| | - Gang Feng
- Institute of Tissue Engineering and Stem Cells, North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| |
Collapse
|
48
|
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
|
49
|
Giraudo L, Gammaitoni L, Cangemi M, Rotolo R, Aglietta M, Sangiolo D. Cytokine-induced killer cells as immunotherapy for solid tumors: current evidence and perspectives. Immunotherapy 2015; 7:999-1010. [PMID: 26310715 DOI: 10.2217/imt.15.61] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are ex vivo expanded T lymphocytes endowed with potent MHC-independent antitumor activity. CIK cells are emerging as promising therapeutic approach in the field of cancer adoptive immunotherapy, with biologic features favoring their transferability into clinical applications. Aim of this review is to present the biologic characteristic of CIK cells, discussing the main preclinical findings and initial clinical applications in the field of solid tumors.
Collapse
Affiliation(s)
- Lidia Giraudo
- Department of Oncology, University of Torino, Turin, Italy.,Laboratory of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Loretta Gammaitoni
- Laboratory of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Michela Cangemi
- Laboratory of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Ramona Rotolo
- Laboratory of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Massimo Aglietta
- Department of Oncology, University of Torino, Turin, Italy.,Division & Laboratory of Medical Oncology, Candiolo Cancer Institute FPO- IRCCS, Candiolo, Turin, Italy
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Turin, Italy.,Laboratory of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| |
Collapse
|
50
|
Wang XD, Gao NN, Diao YW, Liu Y, Gao D, Li W, Wan YY, Zhong JJ, Jin GY. Conjugation of toll-like receptor-7 agonist to gastric cancer antigen MG7-Ag exerts antitumor effects. World J Gastroenterol 2015; 21:8052-8060. [PMID: 26185376 PMCID: PMC4499347 DOI: 10.3748/wjg.v21.i26.8052] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 04/01/2015] [Accepted: 05/21/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of our tumor vaccines on reversing immune tolerance and generating therapeutic response.
METHODS: Vaccines were synthesized by solid phase using an Fmoc strategy, where a small molecule toll-like receptor-7 agonist (T7) was conjugated to a monoclonal gastric cancer 7 antigen mono-epitope (T7-MG1) or tri-epitope (T7-MG3). Cytokines were measured in both mouse bone marrow dendritic cells and mouse spleen lymphocytes after exposed to the vaccines. BALB/c mice were intraperitoneally immunized with the vaccines every 2 wk for a total of three times, and then subcutaneously challenged with Ehrlich ascites carcinoma (EAC) cells. Three weeks later, the mice were killed, and the tumors were surgically removed and weighed. Serum samples were collected from the mice, and antibody titers were determined by ELISA using an alkaline phosphate-conjugated detection antibody for total IgG. Antibody-dependent cell-mediated cytotoxicity was detected by the lactate dehydrogenase method using natural killer cells as effectors and antibody-labeled EAC cells as targets. Cytotoxic T lymphocyte activities were also detected by the lactate dehydrogenase method using lymphocytes as effectors and EAC cells as targets.
RESULTS: Vaccines were successfully synthesized and validated by analytical high performance liquid chromatography and electrospray mass spectrometry, including T7, T7-MG1, and T7-MG3. Rapid inductions of tumor necrosis factor-α and interleukin-12 in bone marrow dendritic cells and interferon γ and interleukin-12 in lymphocytes occurred in vitro after T7, T7-MG1, and T7-MG3 treatment. Immunization with T7-MG3 reduced the EAC tumor burden in BALB/c mice to 62.64% ± 5.55% compared with PBS control (P < 0.01). Six or nine weeks after the first immunization, the monoclonal gastric cancer 7 antigen antibody increased significantly in the T7-MG3 group compared with the PBS control (P < 0.01). As for antibody-dependent cell-mediated cytotoxicity, antisera obtained by immunization with T7-MG3 were able to markedly enhance cell lysis compared to PBS control (31.58% ± 2.94% vs 18.02% ± 2.26%; P < 0.01). As for cytotoxic T lymphocytes, T7-MG3 exhibited obviously greater cytotoxicity compared with PBS control (40.92% ± 4.38% vs 16.29% ± 1.90%; P < 0.01).
CONCLUSION: A successful method is confirmed for the design of gastric cancer vaccines by chemical conjugation of T7 and multi-repeat-epitope of monoclonal gastric cancer 7 antigen.
Collapse
MESH Headings
- Animals
- Antibody-Dependent Cell Cytotoxicity
- Antigens, Neoplasm/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/chemical synthesis
- Cancer Vaccines/immunology
- Cancer Vaccines/pharmacology
- Carcinoma, Ehrlich Tumor/drug therapy
- Carcinoma, Ehrlich Tumor/immunology
- Carcinoma, Ehrlich Tumor/pathology
- Cells, Cultured
- Cytokines/metabolism
- Epitopes
- Female
- Immunization Schedule
- Immunoconjugates/administration & dosage
- Immunoconjugates/pharmacology
- Injections, Intraperitoneal
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Membrane Glycoproteins/agonists
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Mice, Inbred BALB C
- Signal Transduction/drug effects
- Superantigens
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Time Factors
- Toll-Like Receptor 7/agonists
- Toll-Like Receptor 7/immunology
- Toll-Like Receptor 7/metabolism
- Tumor Burden
- Tumor Escape/drug effects
Collapse
|