101
|
Shirjang S, Alizadeh N, Mansoori B, Mahmoodpoor A, Kafil HS, Hojjat-Farsangi M, Yousefi M. Promising immunotherapy: Highlighting cytokine-induced killer cells. J Cell Biochem 2018; 120:8863-8883. [PMID: 30556298 DOI: 10.1002/jcb.28250] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022]
Abstract
For many years, cancer therapy has appeared to be a challenging issue for researchers and physicians. By the introduction of novel methods in immunotherapy, the prospect of cancer therapy even more explained than before. Cytokine-induced killer (CIK) cell-based immunotherapy demonstrated to have potentiality in improving clinical outcomes and relieving major side effects of standard treatment options. In addition, given the distinctive features such as high safety, low toxicity effects on healthy cells, numerous clinical trials conducted on CIK cells. Due to the shortcomings that observed in CIK cell immunotherapy alone, arising a tendency to make modifications (combined modality therapy or combination therapy) including the addition of various types of cytokines, genetic engineering, combination with immune checkpoints, and so on. In this review, we have tried to bring forth the latest immunotherapy methods and their overview. We have discussed the combination therapies with CIK cells and the conducted clinical trials. This helps the future studies to use integrated therapies with CIK cells as a promising treatment of many types of cancers.
Collapse
Affiliation(s)
- Solmaz Shirjang
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene therapy Lab, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, Stockholm, Sweden
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
102
|
Chen J, Chen Y, Feng F, Chen C, Zeng H, Wen S, Xu X, He J, Li J. Programmed cell death protein-1/programmed death-ligand 1 blockade enhances the antitumor efficacy of adoptive cell therapy against non-small cell lung cancer. J Thorac Dis 2018; 10:6711-6721. [PMID: 30746216 DOI: 10.21037/jtd.2018.10.111] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Cytokine-induced killer (CIK) cells and natural killer (NK) cells are employed by two different approaches to adoptive cell immunotherapy for cancer. It has been reported that adoptive cell immunotherapy could prolong the overall survival (OS) of advanced cancer patients. The introduction of agents that induce immune checkpoint blockades has improved the efficacy of immune-mediated therapy for metastatic cancers. However, the effects of combining a checkpoint inhibitor with CIK cells or NK cells to target non-small cell lung cancer (NSCLC)remain unknown. Methods The present study investigated the effects of combining CIK cells with a programmed cell death protein-1 (PD-1) inhibitor (an anti-PD-1 monoclonal antibody). During the expansion cultivation, the addition of the PD-1 antibody promoted CIK-mediated cytotoxicity in H1975 lung adenocarcinoma cells. Co-cultivation of CIK cells with the PD-1 antibody for 6 days induced CD3+CD56+ T cell expansion, with increases in the levels of CD107a and interferon γ (IFN-γ). Results When NK cells were co-cultured with 5 µg/mL of an anti-programmed death-ligand 1 (PD-L1) mAb for 24 hours at an effector cell: target ratio of 10:1, it led to more potent cytotoxicity compared to other time points and concentrations. However, combining NK cells with the anti-PD-L1 mAb showed no significant advantages over treatment with NK cells alone. Conclusions Our results suggest that combining CIK cells with PD-1 blockade before transfusion might improve the efficiency of CIK therapy for NSCLC patients. This effect does not seem to occur for NK cell therapy.
Collapse
Affiliation(s)
- Jingyi Chen
- State Key Laboratory of Respiratory Disease, the Thoracic Surgery Department, the First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Yusong Chen
- State Key Laboratory of Respiratory Disease, the Thoracic Surgery Department, the First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Fenglan Feng
- State Key Laboratory of Respiratory Disease, the Thoracic Surgery Department, the First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Cheng Chen
- State Key Laboratory of Respiratory Disease, the Thoracic Surgery Department, the First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Haikang Zeng
- State Key Laboratory of Respiratory Disease, the Thoracic Surgery Department, the First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Shuai Wen
- State Key Laboratory of Respiratory Disease, the Thoracic Surgery Department, the First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Xin Xu
- State Key Laboratory of Respiratory Disease, the Thoracic Surgery Department, the First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Jianxing He
- State Key Laboratory of Respiratory Disease, the Thoracic Surgery Department, the First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Jin Li
- State Key Laboratory of Respiratory Disease, the Thoracic Surgery Department, the First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| |
Collapse
|
103
|
Zhou Y, Chen CL, Jiang SW, Feng Y, Yuan L, Chen P, Zhang L, Huang S, Li J, Xia JC, Zheng M. Retrospective analysis of the efficacy of adjuvant CIK cell therapy in epithelial ovarian cancer patients who received postoperative chemotherapy. Oncoimmunology 2018; 8:e1528411. [PMID: 30713783 PMCID: PMC6343777 DOI: 10.1080/2162402x.2018.1528411] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/26/2018] [Accepted: 09/20/2018] [Indexed: 12/24/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are demonstrated to possess potent cytolytic effect against ovarian cancer cells in vitro and in vivo. However, the clinical efficacy of maintenance therapy of CIK cells in patients with epithelial ovarian cancer (EOC) after first-line treatment remains unclear. This retrospective study included 646 cases of postoperative EOC patients, 72 of which received chemotherapy and sequential immunotherapy (CIT group), and 574 of which received only chemotherapy (Control group). Patients in the CIT group received at least four cycles of CIK cell (range 8.0 × 109 – 1.3 × 1010 cells) transfusion, with the interval of each cycle being 2 weeks. Survival analysis showed a significantly higher overall survival (OS) rate in the CIT group compared with the control group, as well as a favorable progression-free survival (PFS). Univariate and multivariate analyses indicated that adjuvant CIT was an independent prognostic factor for the OS of patients with EOC. Furthermore, subgroup analyses showed that adjuvant CIT significantly improved the OS of patients older than 45 years, with CA125 ≤ 1000, or with moderate or poorly differentiated tumors, and prolonged the PFS of patients with residual disease > 1 cm. Additionally, Kaplan-Meier analyses revealed that a higher fraction of CD3+CD8+/CD3+CD56+ phenotypes or lower percentage of CD3+CD4+/CD3−CD56+ phenotypes in the infused CIK cells significantly associated with better survival of patients with EOC. Furthermore, across all processes of CIK cell immunotherapy in the CIT group, 12.5% (9/72) of patients developed self-limiting light fevers and shivering at grade 1 or 2. No immunotherapy-related serious reactions were recorded. These data indicate that adjuvant CIT with CIK cells is an effective therapeutic approach to prolonging the survival of EOC patients.
Collapse
Affiliation(s)
- Yun Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Gynecology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Chang-Long Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Radiation Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Sen-Wei Jiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Gynecology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yanling Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Gynecology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Linjing Yuan
- Department of Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Ping Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of VIP region, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Lan Zhang
- Department of Radiotherapy, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, Kunming, P. R. China
| | - Shuting Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Gynecology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jundong Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Gynecology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jian-Chuan Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Min Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.,Department of Gynecology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| |
Collapse
|
104
|
Fan J, Shang D, Han B, Song J, Chen H, Yang JM. Adoptive Cell Transfer: Is it a Promising Immunotherapy for Colorectal Cancer? Am J Cancer Res 2018; 8:5784-5800. [PMID: 30555581 PMCID: PMC6276301 DOI: 10.7150/thno.29035] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/04/2018] [Indexed: 12/12/2022] Open
Abstract
The last decade has witnessed significant advances in the adoptive cell transfer (ACT) technique, which has been appreciated as one of the most promising treatments for patients with cancer. Utilization of ACT can enhance the function of the immune system or improve the specificity and persistence of transferred cells. Various immune cells including T lymphocytes, natural killer cells, dendritic cells, and even stem cells can be used in the ACT despite their different functional mechanisms. Colorectal cancer (CRC) is among the most common malignancies and causes millions of deaths worldwide every year. In this review, we discuss the status and perspective of the ACT in the treatment of CRC.
Collapse
|
105
|
Recombinant Adenovirus KGHV500 and CIK Cells Codeliver Anti-p21-Ras scFv for the Treatment of Gastric Cancer with Wild-Type Ras Overexpression. MOLECULAR THERAPY-ONCOLYTICS 2018; 11:90-101. [PMID: 30534583 PMCID: PMC6280635 DOI: 10.1016/j.omto.2018.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/16/2018] [Indexed: 12/20/2022]
Abstract
The development of gastric cancer is frequently related to the overexpression of wild-type p21 proteins, but it is rarely related to mutated Ras proteins. We previously constructed a broad-spectrum anti-p21-Ras single-chain variable fragment antibody (scFv), which was carried by the oncolytic adenovirus KGHV500. Here we explored the antitumor effects of this recombinant oncolytic adenovirus carried by cytokine-induced killer (CIK) cells on human gastric SGC7901 cells that overexpress wild-type Ras. The MTT assay, scratch test, Transwell assay, and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay were performed in vitro to investigate the proliferation, migration, invasiveness, and cell apoptosis rate, respectively, of the human gastric cell line SGC7901 treated with KGHV500 adenovirus. Then, the tumor-targeting ability and systemic safety of KGHV500 adenovirus delivered by CIK cells were explored in vivo. We found that KGHV500 adenovirus could significantly inhibit proliferation, migration, and invasiveness and promote cell apoptosis in SGC7901 cells in vitro. In vivo studies showed that CIK cells could successfully deliver KGHV500 adenovirus to the tumor site; the two vectors synergistically killed tumor cells, and the treatment was relatively safe for normal tissues. In conclusion, this therapeutic strategy of recombinant adenovirus KGHV500 delivered by CIK cells offers a positive prospect for the targeted therapy of Ras-related cancers.
Collapse
|
106
|
Zhang W, Cai H, Tan WS. Dynamic suspension culture improves ex vivo expansion of cytokine-induced killer cells by upregulating cell activation and glucose consumption rate. J Biotechnol 2018; 287:8-17. [PMID: 30273619 DOI: 10.1016/j.jbiotec.2018.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/17/2018] [Accepted: 09/26/2018] [Indexed: 12/20/2022]
Abstract
Ex vivo expansion is an effective strategy to acquire cytokine-induced killer (CIK) cells needed for clinical trials. In this work, the effects of dynamic suspension culture, which was carried out by shake flasks on a shaker, on CIK cells were investigated by the analysis of expansion characteristics and physiological functions, with the objective to optimize the culture conditions for ex vivo expansion of CIK cells. The results showed that the expansion folds of total cells in dynamic cultures reached 69.36 ± 30.36 folds on day 14, which were significantly higher than those in static cultures (9.24 ± 1.12 folds, P < 0.05), however, the proportions of CD3+ cells and CD3+CD56+ cells in both cultures were similar, leading to much higher expansion of CD3+ cells and CD3+CD56+ cells in dynamic cultures. Additionally, expanded CIK cells in two cultures possessed comparable physiological functions. Notably, significantly higher percentages of CD25+ cells and CD69+ cells were found in dynamic cultures (P < 0.05). Besides, much higher glucose consumption rate of cells (P < 0.05) but similar YLac/gluc were observed in dynamic cultures. Further, cells in dynamic cultures had better glucose utilization efficiency. Together, these results suggested that dynamic cultures improved cell activation, then accelerated glucose consumption rate, which enhanced cell expansion and promoted glucose utilization efficiency of cells.
Collapse
Affiliation(s)
- Weiwei Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, PR China
| | - Haibo Cai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, PR China.
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, PR China
| |
Collapse
|
107
|
Shi B, Sun A, Zhang X. Influence of different ex vivo cell culture methods on the proliferation and anti-tumor activity of cytokine-induced killer cells from gastric cancer patients. Onco Targets Ther 2018; 11:2657-2672. [PMID: 29780258 PMCID: PMC5951225 DOI: 10.2147/ott.s162281] [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: 12/13/2022] Open
Abstract
Purpose In cytokine-induced killer (CIK) cell therapy, the phenotypes and the numbers of CIK cells have a great influence on the therapeutic effects. This study aimed to investigate the effects of different ex vivo cell culture methods on the proliferation and cytotoxicity of CIK cells that were obtained from gastric cancer patients. Patients and methods CIK precursor (Pre-CIK) cells were collected by either hydroxyethyl starch (HES) sedimentation (HES method, unpurified group) or Ficoll-Hypaque density gradient centrifugation (Ficoll method, purified group). Cell number, collection time, and morphology of Pre-CIK cells in the two groups were determined. The proliferation ability, cytokines, phenotypes, and cytotoxicity of CIK cells in the two groups were evaluated ex vivo and in vivo. Results In this study, the number of Pre-CIK cells in the unpurified group was significantly higher than that in the purified group (P<0.05). Numbers of erythrocytes, platelets, and granulocytes were reduced significantly following the purification step (P<0.05). Compared to CIK cells in the purified group, those in the unpurified group showed more active proliferation, accompanied by higher percentages of CD8+, CD3-CD56+, and CD3+CD56+ cells, which were responsible for cytotoxicity of CIK cells (P<0.05). This research also showed that the levels of interferon-γ, interleukin-2, and tumor necrosis factor-α, which can enhance the proliferation and cytotoxicity of CIK cells, were significantly increased in the unpurified group (P<0.05). Furthermore, CIK cells in the unpurified group also showed stronger anti-tumor effects against gastric cancer cells than those in the purified group, both ex vivo and in vivo (P<0.05). Conclusion The removal of Ficoll-Hypaque purification step reduces the time and cost of the Pre-CIK separation and provides more CIK cells with higher cytotoxicity, which is of great importance in the clinical application of CIK cell therapy.
Collapse
Affiliation(s)
- Bin Shi
- Department of Gastrointestinal Surgery, Liaocheng People's Hospital of Taishan Medical University, Liaocheng, Shandong Province, China
| | - Aixia Sun
- Department of Clinical Laboratory, Liaocheng People's Hospital, Liaocheng Clinical School of Taishan Medical University, Liaocheng, Shandong Province, China
| | - Xiaorui Zhang
- Department of Health, Liaocheng People's Hospital of Taishan Medical University, Liaocheng, Shandong Province, China
| |
Collapse
|
108
|
Shi H, Yu F, Mao Y, Ju Q, Wu Y, Bai W, Wang P, Xu R, Jiang M, Shi J. EphA2 chimeric antigen receptor-modified T cells for the immunotherapy of esophageal squamous cell carcinoma. J Thorac Dis 2018; 10:2779-2788. [PMID: 29997940 DOI: 10.21037/jtd.2018.04.91] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background It is urgent to explore an effective potential therapeutic strategy for ESCC. In recent years, cell-based cancer immunotherapy has become a potentially close for carcinoma therapy. Chimeric antigen receptor (CAR) T cell technology is a kind of adoptive cell therapy technique which has been developed rapidly. We sought to obtain EphA2.CAR-T cell and revealed the ability of EphA2.CAR-T cells to kill esophageal squamous cell carcinoma (ESCC) cells in vitro. Methods Firstly, the expression and location of EphA2 in ESCC tissues and cells was tested by immunohistochemistry staining and Western blot. Secondly, the second generation of EphA2.CAR was constructed via molecular biology technology, and transduced into T cells to obtain the EphA2.CAR-T cell. The transduction efficacies were assessed using flow cytometry (FCM). Thirdly, the effect of cell killing of EphA2.CAR-T cell on ESCC cells in vitro was detected by co-culture experiments. The productions of cytokines (TNF-α and IFN-γ) by EphA2.CAR-T cell after co-culture with ESCC cells were analyzed by ELISA assay. Results The expression of EphA2 was significantly upregulated in ESCC tissues and cells (P<0.05). EphA2 was expressed on the membrane of ESCC cells, so it could be served as tumor-associated surface antigens (TAA) of CAR for ESCC treatment. The EphA2.CAR-T cell was obtained successfully, and its' transduction efficacies was 61.4% by FCM. The ability of cell killing of EphA2.CAR-T cell was better than that of T cells (P<0.01), and demonstrated a dose-dependent cell killing. The results of ELISA assay showed that the levels of TNF-α and IFN-γ in EphA2.CAR-T cells were notably raised compared with T cells (P<0.05). Conclusions We firstly constructed the second generation of EphA2.CAR and established EphA2.CAR-T cells. The EphA2.CAR-T cells showed a dose-dependent cell killing of ESCC cells, and promoted the production of cytokines in vitro. These findings open a new way for treatment of ESCC by immunotherapy in the future.
Collapse
Affiliation(s)
- Hui Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Feng Yu
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Yinting Mao
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Qianqian Ju
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Yingcheng Wu
- Medical College, Nantong University, Nantong 226001, China
| | - Wen Bai
- Medical College, Nantong University, Nantong 226001, China
| | - Peiwen Wang
- Medical College, Nantong University, Nantong 226001, China
| | - Ran Xu
- Medical College, Nantong University, Nantong 226001, China
| | - Maorong Jiang
- Laboratory Animals Center, Nantong University, Nantong 226001, China
| | - Jiahai Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| |
Collapse
|
109
|
Immunotherapy for Hepatocellular Carcinoma: Current Advances and Future Expectations. J Immunol Res 2018; 2018:8740976. [PMID: 29785403 PMCID: PMC5896259 DOI: 10.1155/2018/8740976] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 01/02/2018] [Accepted: 01/21/2018] [Indexed: 12/14/2022] Open
Abstract
Primary liver cancer is a common kind of digestive cancers with high malignancy, causing 745,500 deaths each year. Hepatocellular carcinoma is the major pathological type of primary liver cancer. Traditional treatment methods for patients with hepatocellular carcinoma have shown poor efficacy in killing residual cancer cells for a long time. In recent years, tumor immunotherapy has emerged as a promising method owing to its safety and efficacy with respect to delaying the progression of advanced tumors and protecting postoperative patients against tumor relapse and metastasis. Immune tolerance and suppression in tumor microenvironments are the theoretical basis of immunotherapy. Adoptive cell therapy functions by stimulating and cultivating autologous lymphocytes ex vivo and then reinfusing them into the patient to kill cancer cells. Cancer vaccination is performed using antigenic substances to activate tumor-specific immune responses. Immune checkpoint inhibitors can reactivate tumor-specific T cells and develop an antitumor effect by suppressing checkpoint-mediated signaling. Oncolytic viruses may selectively replicate in tumor cells and cause lysis without harming normal tissues. Here, we briefly introduce the mechanism of immunosuppression in hepatocellular carcinoma and summarize the rationale of the four major immunotherapeutic approaches with their current advances.
Collapse
|
110
|
Saudemont A, Jespers L, Clay T. Current Status of Gene Engineering Cell Therapeutics. Front Immunol 2018; 9:153. [PMID: 29459866 PMCID: PMC5807372 DOI: 10.3389/fimmu.2018.00153] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/17/2018] [Indexed: 12/27/2022] Open
Abstract
Ex vivo manipulations of autologous patient’s cells or gene-engineered cell therapeutics have allowed the development of cell and gene therapy approaches to treat otherwise incurable diseases. These modalities of personalized medicine have already shown great promises including product commercialization for some rare diseases. The transfer of a chimeric antigen receptor or T cell receptor genes into autologous T cells has led to very promising outcomes for some cancers, and particularly for hematological malignancies. In addition, gene-engineered cell therapeutics are also being explored to induce tolerance and regulate inflammation. Here, we review the latest gene-engineered cell therapeutic approaches being currently explored to induce an efficient immune response against cancer cells or viruses by engineering T cells, natural killer cells, gamma delta T cells, or cytokine-induced killer cells and to modulate inflammation using regulatory T cells.
Collapse
Affiliation(s)
| | | | - Timothy Clay
- GlaxoSmithKline, Collegeville, PA, United States
| |
Collapse
|
111
|
Katzman D, Wu S, Sterman DH. Immunological Aspects of Cryoablation of Non-Small Cell Lung Cancer: A Comprehensive Review. J Thorac Oncol 2018; 13:624-635. [PMID: 29391289 DOI: 10.1016/j.jtho.2018.01.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 01/18/2018] [Accepted: 01/19/2018] [Indexed: 01/18/2023]
Abstract
In cryoimmunotherapy, target tumors are treated with cryoablation to generate antitumor immune responses. Because immune checkpoint inhibitors have demonstrated that lung cancer can be an immunotherapy-responsive disease, there has been renewed interest in the immunological aspects of cryoablation of lung cancer. Herein, we review preclinical and clinical trials of cryoablation of primary lung tumors. We examine the magnitude of cryoablation-induced antitumor immune responses and the synergy between cryoablation and either other immunotherapies or molecular targeted therapies to improve treatment responses in advanced lung cancer. We further discuss a rationale for the addition of cryoablation to immune checkpoint inhibitors for the treatment of advanced lung cancer, which is currently under clinical investigation.
Collapse
Affiliation(s)
- Daniel Katzman
- Division of Pulmonary, Critical Care and Sleep Medicine, New York University Langone Health, New York, New York.
| | - Shirley Wu
- New York University School of Medicine, New York, New York
| | - Daniel H Sterman
- Division of Pulmonary, Critical Care and Sleep Medicine, New York University Langone Health, New York, New York
| |
Collapse
|
112
|
Li Z, Yu XF, Chu PK. Recent advances in cell-mediated nanomaterial delivery systems for photothermal therapy. J Mater Chem B 2018; 6:1296-1311. [DOI: 10.1039/c7tb03166a] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cell-mediated “Trojan Horse” delivery vehicles overcome the drug delivery barriers to transport nano-agents enhancing the efficiency of photothermal therapy.
Collapse
Affiliation(s)
- Zhibin Li
- Department of Physics and Department of Materials Science and Engineering
- City University of Hong Kong
- Kowloon
- China
- Center for Biomedical Materials and Interfaces
| | - Xue-Feng Yu
- Center for Biomedical Materials and Interfaces
- Shenzhen Institutes of Advanced Technology
- Chinese Academy of Sciences
- Shenzhen 518055
- P. R. China
| | - Paul K. Chu
- Department of Physics and Department of Materials Science and Engineering
- City University of Hong Kong
- Kowloon
- China
| |
Collapse
|
113
|
Wang Y, Lv B, Li K, Zhang A, Liu H. Adjuvant immunotherapy of dendritic cells and cytokine-induced killer cells is safe and enhances chemotherapy efficacy for multiple myeloma in China: a meta-analysis of clinical trials. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:3245-3256. [PMID: 29180849 PMCID: PMC5695269 DOI: 10.2147/dddt.s146959] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective The aim of this study was to systematically evaluate the efficacy and safety of the combination of dendritic cells and cytokine-induced killer cells (DC–CIK) adjuvant immunotherapy and chemotherapy in the treatment of multiple myeloma (MM). Methods Clinical trials were gathered by searching Web of Science, PubMed, Embase, Cochrane Library, Wanfang, and CNKI database. Outcome measurements including therapeutic efficacy, prognosis, immune function, and adverse events were extracted and evaluated. Results A total of 12 trials including 594 MM patients were involved in this study for statistical analysis. Results indicated that compared to chemotherapy alone, the combination of DC–CIK immunotherapy with chemotherapy significantly improved patients’ overall response rate (ORR, odds ratio [OR] =2.77, 95% confidence interval [CI] =1.88–4.10, P<0.00001), disease control rate (DCR, OR =2.90, CI =1.72–4.90, P<0.0001), and life quality (P<0.00001). The combined therapy showed advantages over chemotherapy alone in prognostic indicators including percentage of tumor cells (P=0.04), serum levels of β2-microglobin (P<0.0001), M protein (P<0.00001), and creatinine (P<0.0001), and 24 h urine light chains (P<0.00001). After combined treatment, CD4+ lymphocyte subsets’ percentages, CD4+/CD8+ ratio, and cytokines levels of AgNOR, IFN-γ, IL-2, and IL-12 were significantly increased (P<0.05), whereas CD8+ and CD4+CD25+ percentages and IL-4, IL-6, IL-10, and TGF-β levels were obviously decreased (P<0.01), indicating a recovered immune condition. Conclusion Adjuvant DC–CIK immunotherapy enhances the efficacy of chemotherapy for MM and improves prognosis probably by reconstructing immune function.
Collapse
Affiliation(s)
- Yan Wang
- Department of Clinical Laboratory
| | | | - Ke Li
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng Clinical School of Taishan Medical University, Liaocheng, Shandong Province, China
| | - Anqi Zhang
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng Clinical School of Taishan Medical University, Liaocheng, Shandong Province, China
| | - Hong Liu
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng Clinical School of Taishan Medical University, Liaocheng, Shandong Province, China
| |
Collapse
|
114
|
Martín-Antonio B, Suñe G, Perez-Amill L, Castella M, Urbano-Ispizua A. Natural Killer Cells: Angels and Devils for Immunotherapy. Int J Mol Sci 2017; 18:ijms18091868. [PMID: 28850071 PMCID: PMC5618517 DOI: 10.3390/ijms18091868] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 08/16/2017] [Accepted: 08/19/2017] [Indexed: 02/06/2023] Open
Abstract
In recent years, the relevance of the immune system to fight cancer has led to the development of immunotherapy, including the adoptive cell transfer of immune cells, such as natural killer (NK) cells and chimeric antigen receptors (CAR)-modified T cells. The discovery of donor NK cells’ anti-tumor activity in acute myeloid leukemia patients receiving allogeneic stem cell transplantation (allo-SCT) was the trigger to conduct many clinical trials infusing NK cells. Surprisingly, many of these studies did not obtain optimal results, suggesting that many different NK cell parameters combined with the best clinical protocol need to be optimized. Various parameters including the high array of activating receptors that NK cells have, the source of NK cells selected to treat patients, different cytotoxic mechanisms that NK cells activate depending on the target cell and tumor cell survival mechanisms need to be considered before choosing the best immunotherapeutic strategy using NK cells. In this review, we will discuss these parameters to help improve current strategies using NK cells in cancer therapy. Moreover, the chimeric antigen receptor (CAR) modification, which has revolutionized the concept of immunotherapy, will be discussed in the context of NK cells. Lastly, the dark side of NK cells and their involvement in inflammation will also be discussed.
Collapse
Affiliation(s)
- Beatriz Martín-Antonio
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.
- Josep Carreras Leukaemia Research Institute, 08036 Barcelona, Spain.
| | - Guillermo Suñe
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.
- Josep Carreras Leukaemia Research Institute, 08036 Barcelona, Spain.
| | - Lorena Perez-Amill
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.
| | - Maria Castella
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.
- Josep Carreras Leukaemia Research Institute, 08036 Barcelona, Spain.
| | - Alvaro Urbano-Ispizua
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.
- Josep Carreras Leukaemia Research Institute, 08036 Barcelona, Spain.
| |
Collapse
|