1
|
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: 0] [Impact Index Per Article: 0] [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
|
2
|
Cappuzzello E, Vigolo E, D’Accardio G, Astori G, Rosato A, Sommaggio R. How can Cytokine-induced killer cells overcome CAR-T cell limits. Front Immunol 2023; 14:1229540. [PMID: 37675107 PMCID: PMC10477668 DOI: 10.3389/fimmu.2023.1229540] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/02/2023] [Indexed: 09/08/2023] Open
Abstract
The successful treatment of patients affected by B-cell malignancies with Chimeric Antigen Receptor (CAR)-T cells represented a breakthrough in the field of adoptive cell therapy (ACT). However, CAR-T therapy is not an option for every patient, and several needs remain unmet. In particular, the production of CAR-T cells is expensive, labor-intensive and logistically challenging; additionally, the toxicities deriving from CAR-T cells infusion, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), have been documented extensively. Alternative cellular therapy products such as Cytokine-induced killer (CIK) cells have the potential to overcome some of these obstacles. CIK cells are a heterogeneous population of polyclonal CD3+CD56+ T cells with phenotypic and functional properties of NK cells. CIK cell cytotoxicity is exerted in a major histocompatibility complex (MHC)-unrestricted manner through the engagement of natural killer group 2 member D (NKG2D) molecules, against a wide range of hematological and solid tumors without the need for prior antigen exposure or priming. The foremost potential of CIK cells lies in the very limited ability to induce graft-versus-host disease (GvHD) reactions in the allogeneic setting. CIK cells are produced with a simple and extremely efficient expansion protocol, which leads to a massive expansion of effector cells and requires a lower financial commitment compared to CAR-T cells. Indeed, CAR-T manufacturing involves the engineering with expensive GMP-grade viral vectors in centralized manufacturing facilities, whereas CIK cell production is successfully performed in local academic GMP facilities, and CIK cell treatment is now licensed in many countries. Moreover, the toxicities observed for CAR-T cells are not present in CIK cell-treated patients, thus further reducing the costs associated with hospitalization and post-infusion monitoring of patients, and ultimately encouraging the delivery of cell therapies in the outpatient setting. This review aims to give an overview of the limitations of CAR-T cell therapy and outline how the use of CIK cells could overcome such drawbacks thanks to their unique features. We highlight the undeniable advantages of using CIK cells as a therapeutic product, underlying the opportunity for further research on the topic.
Collapse
Affiliation(s)
- Elisa Cappuzzello
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Emilia Vigolo
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Giulia D’Accardio
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Department of Hematology, San Bortolo Hospital of Vicenza, Vicenza, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Roberta Sommaggio
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| |
Collapse
|
3
|
Huang H, Gong Z, Zhu X, Tan W, Cai H. Xanthan gum enhances peripheral blood CIK cells cytotoxicity in serum‐free medium. Biotechnol Prog 2022; 38:e3279. [DOI: 10.1002/btpr.3279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/22/2022] [Accepted: 05/29/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Huimin Huang
- State Key Laboratory of Bioreactor Engineering East China University of Science and Technology Shanghai P. R. China
| | - Zizhen Gong
- State Key Laboratory of Bioreactor Engineering East China University of Science and Technology Shanghai P. R. China
| | - Xuejun Zhu
- State Key Laboratory of Bioreactor Engineering East China University of Science and Technology Shanghai P. R. China
| | - Wen‐song Tan
- State Key Laboratory of Bioreactor Engineering East China University of Science and Technology Shanghai P. R. China
| | - Haibo Cai
- State Key Laboratory of Bioreactor Engineering East China University of Science and Technology Shanghai P. R. China
| |
Collapse
|
4
|
Autologous Cytokine-Induced Killer Cell Immunotherapy Enhances Chemotherapy Efficacy against Multidrug-Resistant Tuberculosis. J Immunol Res 2022; 2022:2943113. [PMID: 35340584 PMCID: PMC8947923 DOI: 10.1155/2022/2943113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 11/17/2022] Open
Abstract
Objective Multidrug-resistant tuberculosis (MDR-TB) causes persistent infection and challenges tuberculosis control worldwide. T cell-mediated immunity plays a critical role in controlling Mycobacterium tuberculosis (Mtb) infection, and therefore, enhancing Mtb-specific T cell immune responses represents a promising therapeutic strategy against TB. Cytokine-induced killer (CIK) immunotherapy is based on autologous infusion of in vitro expanded bulk T cells, which include both pathogen-specific and nonspecific T cells from patient peripheral blood mononuclear cells (PBMC) into TB patients. Preclinical mouse studies have shown that the adoptive T cell therapy inhibited Mtb infection. However, the efficacy of CIK immunotherapy in the treatment of MDR-TB infection has not been evaluated in clinical trials. Methods We performed a retrospective study of MDR-TB patients who received CIK immunotherapy in combination with anti-TB chemotherapy and those who had standard chemotherapy. Results Our results showed that CIK immunotherapy in combination with anti-TB chemotherapy treatment increased the conversion rate of sputum smear and Mtb culture, alleviated symptoms, improved lesion absorption, and increased recovery. The kinetics of serology and immunology index monitoring data showed good safety profiles for the CIK treatment. Conclusion Our study has provided strong evidence that CIK immunotherapy in combination with anti-TB chemotherapy is beneficial for MDR-TB patients. A multicenter clinical trial is warranted to evaluate CIK as a new immune therapy for MDR-TB.
Collapse
|
5
|
Abbasi B, Shamsasenjan K, Ahmadi M, Beheshti SA, Saleh M. Mesenchymal stem cells and natural killer cells interaction mechanisms and potential clinical applications. Stem Cell Res Ther 2022; 13:97. [PMID: 35255980 PMCID: PMC8900412 DOI: 10.1186/s13287-022-02777-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/09/2021] [Indexed: 12/29/2022] Open
Abstract
Natural killer cells (NK cells) are innate immune cells that are activated to fight tumor cells and virus-infected cells. NK cells also play an important role in the graft versus leukemia response. However, they can over-develop inflammatory reactions by secreting inflammatory cytokines and increasing Th1 differentiation, eventually leading to tissue damage. Today, researchers have attributed some autoimmune diseases and GVHD to NK cells. On the other hand, it has been shown that mesenchymal stem cells (MSCs) can modulate the activity of NK cells, while some researchers have shown that NK cells can cause MSCs to lysis. Therefore, we considered it is necessary to investigate the effect of these two cells and their signaling pathway in contact with each other, also their clinical applications.
Collapse
Affiliation(s)
- Batol Abbasi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyedeh Ameneh Beheshti
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahshid Saleh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
The Hematology of Tomorrow Is Here-Preclinical Models Are Not: Cell Therapy for Hematological Malignancies. Cancers (Basel) 2022; 14:cancers14030580. [PMID: 35158848 PMCID: PMC8833715 DOI: 10.3390/cancers14030580] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/13/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cell therapy is revolutionizing the prospect of deadly hematological malignancies such as high-risk acute myeloid leukemia. Stem cell therapy of allogeneic source from compatible human leukocyte antigen donor has exceptional success promoting durable remissions, but the rate of relapse is currently still high and there is transplant-related mortality. This review presents the current knowledge on the clinical use of mesenchymal stromal cells to improve outcomes in hematopoietic stem cell transplants. As an alternative or adjuvant approach to prevent relapse, we summarize the status of the promising forms of cellular immunotherapy aimed at targeting not only the bulk but also the cells of origin of leukemia. Finally, we discuss the available in vivo models for disease modelling and treatment efficacy prediction in these contexts. Abstract The purpose of this review is to present the current knowledge on the clinical use of several forms of cell therapy in hematological malignancies and the preclinical models available for their study. In the context of allogeneic hematopoietic stem cell transplants, mesenchymal stromal cells are pursued to help stem cell engraftment and expansion, and control graft versus host disease. We further summarize the status of promising forms of cellular immunotherapy including CAR T cell and CAR NK cell therapy aimed at eradicating the cells of origin of leukemia, i.e., leukemia stem cells. Updates on other forms of cellular immunotherapy, such as NK cells, CIK cells and CAR CIK cells, show encouraging results in AML. The considerations in available in vivo models for disease modelling and treatment efficacy prediction are discussed, with a particular focus on their strengths and weaknesses for the study of healthy and diseased hematopoietic stem cell reconstitution, graft versus host disease and immunotherapy. Despite current limitations, cell therapy is a rapidly evolving field that holds the promise of improved cure rates, soon. As a result, we may be witnessing the birth of the hematology of tomorrow. To further support its development, improved preclinical models including humanized microenvironments in mice are urgently needed.
Collapse
|
7
|
Fan Y, He S. The Characteristics of Tumor Microenvironment in Triple Negative Breast Cancer. Cancer Manag Res 2022; 14:1-17. [PMID: 35018117 PMCID: PMC8740624 DOI: 10.2147/cmar.s316700] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a special subtype of breast cancer, accounting for 10-20% of breast cancers with high intrinsic heterogeneity. Its unique immune microenvironment, including high expression of vascular endothelial growth factors, tumor infiltrating lymphocytes (TILs), tumor-associated macrophages (TAMs), and other molecules that promote the growth and migration of tumor cells, has been shown to play a dual role in the occurrence, growth, and metastasis of TNBC. Understanding the TNBC microenvironment is of great significance for the prognosis and treatment of TNBC. In this article, we describe the composition and function of immune cells in the TNBC microenvironment and summarize the major cytokine growth factors and chemokines in the TNBC microenvironment. Finally, we discuss the progress of TNBC, cytokine-induced killer cell therapy, and immune checkpoint therapy.
Collapse
Affiliation(s)
- Yiqi Fan
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People’s Republic of China
| | - Shuai He
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People’s Republic of China
| |
Collapse
|
8
|
Clinical Studies on Cytokine-Induced Killer Cells: Lessons from Lymphoma Trials. Cancers (Basel) 2021; 13:cancers13236007. [PMID: 34885117 PMCID: PMC8656601 DOI: 10.3390/cancers13236007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 01/03/2023] Open
Abstract
Simple Summary Lymphoma is a heterogeneous group of neoplasms including over 70 different subtypes. Its biological characteristic of deriving from lymphoid tissues makes it ideal for immunotherapy. In this paper, we provide insights into lymphoma-specific clinical trials based on cytokine-induced killer (CIK) cell therapy. We also reviewed pre-clinical lymphoma models where CIK cells have been used along with other synergetic tumor-targeting immune modules to improve their therapeutic potential. From a broader perspective, we will highlight that CIK cell therapy has potential, and in this rapidly evolving landscape of cancer therapies its optimization (as a personalized therapeutic approach) will be beneficial in lymphomas. Abstract Cancer is a complex disease where resistance to therapies and relapses often pose a serious clinical challenge. The scenario is even more complicated when the cancer type itself is heterogeneous in nature, e.g., lymphoma, a cancer of the lymphocytes which constitutes more than 70 different subtypes. Indeed, the treatment options continue to expand in lymphomas. Herein, we provide insights into lymphoma-specific clinical trials based on cytokine-induced killer (CIK) cell therapy and other pre-clinical lymphoma models where CIK cells have been used along with other synergetic tumor-targeting immune modules to improve their therapeutic potential. From a broader perspective, we will highlight that CIK cell therapy has potential, and in this rapidly evolving landscape of cancer therapies its optimization (as a personalized therapeutic approach) will be beneficial in lymphomas.
Collapse
|
9
|
Issabekova A, Zhumabekova M, Zhunussova M, Ogay V. The Crosstalk Between Dendritic Cells, Cytokine-Induced Killer Cells And Cancer Cells From The Perspective Of Combination Therapy. RUSSIAN OPEN MEDICAL JOURNAL 2021. [DOI: 10.15275/rusomj.2021.0209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Dendritic cells (DCs) are considered the most potent professional antigen-presenting cells (APCs) that elicit adaptive antitumour immunity. DCs integrate multiple environmental signals by efficiently processing tumour-associated antigens (TAAs) and migrating to draining lymph nodes (dLNs), where they present foreign antigens to T cells for priming. DCs thus serve as a major link between innate and adaptive immunity. Although DCs (mostly monocyte-derived DCs [mo-DCs]) have already been used in cancer therapies, such approaches have shown limited efficacy. Mo-DCs have the unique ability to present antigens to T cells in peripheral tissues. CD3+CD56+ cytokine-induced killer (CIK) cells are characterized by both MHC-restricted and MHC-unrestricted antitumour cytotoxicity against a broad range of cancer cells. This review presents an overview of the mechanisms by which mo-DCs and CIK cells’ interact with each other and with tumours. The maturation of DCs was identified as a crucial step in the development of effective DC-based vaccines against cancer. A further improved adoptive immunotherapy strategy involves a combination of mature mo-DCs and CIK cells. Combination therapy presents many opportunities for cancer treatment, as reported by a number of clinical trials. However, there is a lack of fundamental studies on the interaction of in vitro-generated mo-DCs with CIK cells. We discuss several methods of boosting DC-based vaccines and review the current knowledge of contact-dependent and cytokine-induced interactions of mo-DCs with CIK cells. We highlight that the combination of mo-DCs with CIK cells activates MHC-restricted and MHC-unrestricted immune responses.
Collapse
|
10
|
The effects of stereotactic body radiotherapy on peripheral natural killer and CD3 +CD56 + NKT-like cells in patients with hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int 2021; 20:240-250. [PMID: 33454220 DOI: 10.1016/j.hbpd.2020.12.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 12/15/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Both natural killer (NK) and CD3+CD56+natural killer T (NKT)-like cells play critical roles in the antitumor response. This study aimed to explore the effects of stereotactic body radiotherapy (SBRT) on peripheral NK and NKT-like cells in patients with hepatocellular carcinoma (HCC), and to identify possible surface markers on these cells that correlate with the prognosis. METHODS Twenty-five HCC patients were prospectively enrolled in our study, and 10 healthy individuals were served as healthy controls. Flow cytometry was used to determine the counts and the percentages of peripheral NK and NKT-like cells, cells with certain receptors, and cells with intracellular interferon-γ and TNF-α secretion at different time points, including time points of prior to SBRT, at post-SBRT, and 3-month and 6-month after treatment. The Kaplan-Meier method with the log-rank test was applied for survival analysis. RESULTS The peripheral NKT-like cells was increased at post-SBRT. Meanwhile, elevated levels of inhibitory receptors and reduced levels of activating receptors of NK cells were also observed in NK cells at post-SBRT, but the levels was not significantly different at 3-month and 6-month as compared with the baseline levels. Lower percentage of NKp30+NK cells before SBRT and higher percentage of CD158b+NK cells after SBRT were associated with poor progression-free survival. In addition, higher percentage of CD3+CD56+ NKT-like cells was associated with a higher overall survival rate in HCC patients. CONCLUSIONS SBRT has an apparent effect on both peripheral NK and CD3+CD56+NKT-like cells. Lower percentage of NKp30+NK cells before SBRT and higher percentage of CD158b+NK cells after SBRT are correlated with poor patients' PFS. Higher percentage of CD3+CD56+ NKT-like cells is associated with higher OS in HCC patients.
Collapse
|
11
|
Zhang X, Huang H, Han L, Li T, Wang Z, Gao Q. Advanced Renal-Cell Carcinoma Pseudoprogression After Combined Immunotherapy: Case Report and Literature Review. Front Oncol 2021; 11:640447. [PMID: 34123792 PMCID: PMC8194346 DOI: 10.3389/fonc.2021.640447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/26/2021] [Indexed: 11/30/2022] Open
Abstract
Treatment with a combination of programmed cell death-1 (PD-1) blocker and cytokine-induced killer (CIK) cells has improved outcome in cancer patients but is also associated with various patterns of responses. Pseudoprogression is a unique and uncommon phenomenon with no clear criteria for rapid diagnosis. Although some reports of pseudoprogression during immunotherapy exist, there are few reports of pseudoprogression occurring twice in the same patient. Here, we report the case of 51-year-old female patient with advanced renal cell carcinoma, who received a combination treatment of PD-1 blocker and CIK cells, and where pseudoprogression of lung and brain tumors occurred successively during treatment.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Hao Huang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Lu Han
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Tiepeng Li
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Zibing Wang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Quanli Gao
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
12
|
Recruitment, Infiltration, and Cytotoxicity of HLA-Independent Killer Lymphocytes in Three-Dimensional Melanoma Models. Cancers (Basel) 2021; 13:cancers13102302. [PMID: 34065007 PMCID: PMC8151151 DOI: 10.3390/cancers13102302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 04/28/2021] [Accepted: 05/05/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Limited therapeutic results of immune checkpoint inhibitors in definite tumor settings, such as melanoma, call for alternative or complementary approaches. Among these, adoptive cell therapy (ACT) by means of HLA-independent tumor killer lymphocytes is a promising approach. We aimed at developing a pre-clinical 3D model to investigate and visualize the interaction between tumor and immune effectors in melanoma. To this aim, we employed Cytokine-Induced Killer cells (CIK) and NK-92 on patient-derived melanoma samples. By means of imaging-based methods, we measured the effector recruitment on the 3D targets, their infiltration, and cytotoxic activity. Our results and methodologies can be easily generalized to other effectors and other classes of tumors and help elucidate fundamental questions on the basic biology and kinetics of immune effector recruitment in a realistic 3D setting mimicking a solid tumor. Abstract Cancer adoptive cell therapy (ACT) with HLA-independent tumor killer lymphocytes is a promising approach, with intrinsic features potentially addressing crucial tumor-escape mechanisms of checkpoint inhibitors. Cytokine-induced Killer (CIK) and Natural Killer (NK) lymphocytes share similar tumor-killing mechanisms, with preclinical evidence of intense activity against multiple solid tumors and currently testing in clinical studies. To improve the effective clinical translation of such ACT approaches, several fundamental questions still need to be addressed within appropriate preclinical contexts, capable of overcoming limitations imposed by most traditional two-dimensional assays. Here, we developed a novel experimental approach to explore, dissect, and visualize the interactions of CIK and NK lymphocytes with melanoma tumors in vitro in 3D. Primary melanoma cells were assembled into small tumors that were dispersed in a 3D matrix and challenged with patient-derived CIK or the NK-92 cell line. By means of imaging-based methods, we reported, visualized, and quantitatively measured the recruitment of CIK and NK on the 3D targets, their infiltration, and cytotoxic activity. Our results support the effective tumor recruitment and tumor infiltration by CIK and NK. Such features appeared dependent on the specific geometric aspects of the environment but can be explained in terms of directional migration toward the tumor, without invoking major feedback components. Overall, our 3D platform allows us to monitor the processes of tumor recruitment, infiltration, and killing by means of live measurements, revealing important kinetic aspects of ACT with CIK and NK against melanoma.
Collapse
|
13
|
Yuan X, Zhang AZ, Ren YL, Wang XL, Jiang CH, Yang L, Liu CX, Liang WH, Pang LJ, Gu WY, Li F, Hu JM. Cytokine-induced killer cells/dendritic cells and cytokine-induced killer cells immunotherapy for the treatment of esophageal cancer: A meta-analysis. Medicine (Baltimore) 2021; 100:e24519. [PMID: 33787569 PMCID: PMC8021386 DOI: 10.1097/md.0000000000024519] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES This meta-analysis was designed to systematically evaluate whether autologous cytokine-induced killer cells (CIK) or dendritic cells and cytokine-induced killer cells (DC-CIK) immunotherapy combined with chemotherapy can improve the therapeutic effect and safety of chemotherapy in esophageal cancer (EC). MATERIALS AND METHODS Randomized controlled trials (RCTs) were electronically searched databases including CNKI, WanFang, WeiPu, CBMDisc, PubMed, Web of Science, EMbase, the Cochrane Library, and Clinical Trials. The databases were searched for articles published until June 2019. Two researchers independently screened the literature, extracted data, and evaluated the quality of the included literature. Meta-analysis was performed using RevMan5.3. RESULTS Seventeen studies (1416 participants) were included. The differences between CIK/DC-CIK combination chemotherapy and chemotherapy alone were significant. The results displayed that the number of CD3+, CD4+, CD4+/CD8+, and NK cells was significantly increased after 1 to 2 weeks of treatment with CIK/DC-CIK cells in the treatment group (all P < .05). In addition, the results shown that 1-year overall survival was significantly prolonged (P < .0001) and quality of life was improved (P = .001) in EC chemotherapy combined with immunotherapy groups compared with conventional treatment. Furthermore, cytokine expression levels of interleukin 2 (IL-2), tumor necrosis factor α (TNF-α), and interleukin 12 (IL-12) were significantly increased (P = .0003) as well as the levels of immunoglobulins were elevated (P < .00001). Serum levels of tumor marker molecules, carcinoembryonic antigen (CEA), carbohydrate antigen (CA)-199, and CA-125 were lower in treatment groups than that of control groups (P < .00001). No fatal adverse reactions were noted (P = .04). CONCLUSIONS It is safe and effective for patients to use chemotherapy combined with CIK/DC-CIK immunotherapy. Immunotherapy can simultaneously improve the antitumor immune response. Specifically, DC-CIK cells can increase T lymphocyte subsets, CIK cells, NK cells, and immunoglobulins in peripheral blood to enhance antitumor immunity. Therefore, combination therapy enhances the immune function and improves the therapeutic efficacy of patients with EC.
Collapse
Affiliation(s)
- Xin Yuan
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang, China
| | - An Zhi Zhang
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang, China
| | - Yi Lin Ren
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang, China
| | - Xue Li Wang
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang, China
| | - Chen Hao Jiang
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang, China
| | - Lan Yang
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang, China
| | - Chun Xia Liu
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang, China
- Australian Institute of Bioengineering and Nanotechnology, University of Queensland, QLD, Australia
| | - Wei Hua Liang
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang, China
| | - Li Juan Pang
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang, China
| | - Wen Yi Gu
- Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Feng Li
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang, China
- Australian Institute of Bioengineering and Nanotechnology, University of Queensland, QLD, Australia
| | - Jian Ming Hu
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang, China
| |
Collapse
|
14
|
Dai F, Zhang PB, Feng Q, Pan XY, Song SL, Cui J, Yang JL. Cytokine-induced killer cells carrying recombinant oncolytic adenovirus expressing p21Ras scFv inhibited liver cancer. J Cancer 2021; 12:2768-2776. [PMID: 33854636 PMCID: PMC8040716 DOI: 10.7150/jca.51434] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 02/18/2021] [Indexed: 02/02/2023] Open
Abstract
Background: Oncolytic adenovirus-mediated gene therapy is an emerging strategy for cancer treatment. However, oncolytic adenoviruses are mainly administered locally at tumor site. Intravenous administration of oncolytic adenovirus for cancer gene therapy is a problem that needs to be solved urgently. Methods: We constructed recombinant oncolytic adenovirus KGHV500 carrying anti-p21Ras scFv and employed CIK cells to deliver KGHV500. TUNEL, wound healing, MTT, and Transwell invasion assays were used to determine the anti-tumor efficacy of KGHV500 on liver cancer cells in vitro. Nude mouse xenograft model was used to examine the anti-tumor efficacy of CIK cells combined with KGHV500 in vivo. Furthermore, KGHV500 accumulation in different organs was detected to assess the safety. Results: KGHV500 inhibited the migration, proliferation, invasion, and induced the apoptosis of liver cancer cells. CIK cells carrying KGHV500 reached tumor site and exerted much better anti-tumor efficacy than CIK cells or KGHV500 alone in nude mouse xenograft model. Moreover, we detected KGHV500 and anti-p21Ras scFv in different organs of nude mice, with little effects on the organs. Conclusions: We develop a novel strategy for the treatment of Ras-driven liver cancer by combining CIK cells with oncolytic adenovirus expressing anti-p21Ras scFv. Intravenous injection of CIK cells carrying KGHV500 in vivo significantly inhibits tumor growth, has little effect on normal organs, and is relatively safe.
Collapse
Affiliation(s)
- Fang Dai
- Graduate School, Kunming Medical University, Kunming, Yunnan, China.,920 th Hospital of the Joint Logistics Support Force of PLA, Kunming, Yunnan, China
| | - Peng-Bo Zhang
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qiang Feng
- 920 th Hospital of the Joint Logistics Support Force of PLA, Kunming, Yunnan, China
| | - Xin-Yan Pan
- 920 th Hospital of the Joint Logistics Support Force of PLA, Kunming, Yunnan, China
| | - Shu-Ling Song
- 920 th Hospital of the Joint Logistics Support Force of PLA, Kunming, Yunnan, China
| | - Jing Cui
- 920 th Hospital of the Joint Logistics Support Force of PLA, Kunming, Yunnan, China
| | - Ju-Lun Yang
- Graduate School, Kunming Medical University, Kunming, Yunnan, China.,920 th Hospital of the Joint Logistics Support Force of PLA, Kunming, Yunnan, China
| |
Collapse
|
15
|
Merker M, Wagner J, Kreyenberg H, Heim C, Moser LM, Wels WS, Bonig H, Ivics Z, Ullrich E, Klingebiel T, Bader P, Rettinger E. ERBB2-CAR-Engineered Cytokine-Induced Killer Cells Exhibit Both CAR-Mediated and Innate Immunity Against High-Risk Rhabdomyosarcoma. Front Immunol 2020; 11:581468. [PMID: 33193388 PMCID: PMC7641627 DOI: 10.3389/fimmu.2020.581468] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
High-risk rhabdomyosarcoma (RMS) occurring in childhood to young adulthood is associated with a poor prognosis; especially children above the age of 10 with advanced stage alveolar RMS still succumb to the disease within a median of 2 years. The advent of chimeric antigen receptor (CAR)-engineered T cells marked significant progress in the treatment of refractory B cell malignancies, but experience for solid tumors has proven challenging. We speculate that this is at least in part due to the poor quality of the patient's own T cells and therefore propose using CAR-modified cytokine-induced killer (CIK) cells as effector cells. CIK cells are a heterogeneous population of polyclonal T cells that acquire phenotypic and cytotoxic properties of natural killer (NK) cells through the cultivation process, becoming so-called T-NK cells. CIK cells can be genetically modified to express CARs. They are minimally alloreactive and can therefore be acquired from haploidentical first-degree relatives. Here, we explored the potential of ERBB2-CAR-modified random-donor CIK cells as a treatment for RMS in xenotolerant mice bearing disseminated high-risk RMS tumors. In otherwise untreated mice, RMS tumors engrafted 13-35 days after intravenous tumor cell injection, as shown by in vivo bioluminescence imaging, immunohistochemistry, and polymerase chain reaction for human gDNA, and mice died shortly thereafter (median/range: 62/56-66 days, n = 5). Wild-type (WT) CIK cells given at an early stage delayed and eliminated RMS engraftment in 4 of 6 (67%) mice, while ERBB2-CAR CIK cells inhibited initial tumor load in 8 of 8 (100%) mice. WT CIK cells were detectable but not as active as CAR CIK cells at distant tumor sites. CIK cell therapies during advanced RMS delayed but did not inhibit tumor progression compared to untreated controls. ERBB2-CAR CIK cell therapy also supported innate immunity as evidenced by selective accumulation of NK and T-NK cell subpopulations in disseminated RMS tumors, which was not observed for WT CIK cells. Our data underscore the power of heterogenous immune cell populations (T, NK, and T-NK cells) to control solid tumors, which can be further enhanced with CARs, suggesting ERBB2-CAR CIK cells as a potential treatment for high-risk RMS.
Collapse
MESH Headings
- Adolescent
- Animals
- Cell Line, Tumor
- Cytokine-Induced Killer Cells/immunology
- Humans
- Immunity, Innate/immunology
- Immunotherapy, Adoptive/methods
- Killer Cells, Natural/immunology
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Natural Killer T-Cells/immunology
- Receptor, ErbB-2/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Chimeric Antigen/immunology
- Rhabdomyosarcoma/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Michael Merker
- Division for Stem Cell Transplantation, Immunology, and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - Juliane Wagner
- Division of Medical Biotechnology, Paul-Ehrlich-Institute, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Hermann Kreyenberg
- Division for Stem Cell Transplantation, Immunology, and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Catrin Heim
- Division for Stem Cell Transplantation, Immunology, and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Laura M. Moser
- Division for Stem Cell Transplantation, Immunology, and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Winfried S. Wels
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Halvard Bonig
- Department of Cellular Therapeutics/Cell Processing (Good Manufacturing Practice, GMP), Institute for Transfusion Medicine and Immunotherapy, Goethe University, Frankfurt, Germany
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul-Ehrlich-Institute, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Evelyn Ullrich
- Division for Stem Cell Transplantation, Immunology, and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany
- Experimental Immunology, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Thomas Klingebiel
- Division for Stem Cell Transplantation, Immunology, and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - Peter Bader
- Division for Stem Cell Transplantation, Immunology, and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - Eva Rettinger
- Division for Stem Cell Transplantation, Immunology, and Intensive Care Medicine, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| |
Collapse
|
16
|
Moloudizargari M, Govahi A, Fallah M, Rezvanfar MA, Asghari MH, Abdollahi M. The mechanisms of cellular crosstalk between mesenchymal stem cells and natural killer cells: Therapeutic implications. J Cell Physiol 2020; 236:2413-2429. [PMID: 32892356 DOI: 10.1002/jcp.30038] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/11/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) are mesenchymal precursors of various origins, with well-known immunomodulatory effects. Natural killer (NK) cells, the major cells of the innate immune system, are critical for the antitumor and antiviral defenses; however, in certain cases, they may be the main culprits in the pathogenesis of some NK-related conditions such as autoimmunities and hematological malignancies. On the other hand, these cells seem to be the major responders in beneficial phenomena like graft versus leukemia. Substantial data suggest that MSCs can variably affect NK cells and can be affected by these cells. Accordingly, acquiring a profound understanding of the crosstalk between MSCs and NK cells and the involved mechanisms seems to be a necessity to develop therapeutic approaches based on such interactions. Therefore, in this study, we made a thorough review of the existing literature on the interactions between MSCs and NK cells with a focus on the underlying mechanisms. The current knowledge herein suggests that MSCs possess a great potential to be used as tools for therapeutic targeting of NK cells in disease context and that preconditioning of MSCs, as well as their genetic manipulation before administration, may provide a wider variety of options in terms of eliciting more specific and desirable therapeutic outcomes. Nevertheless, our knowledge regarding the effects of MSCs on NK cells is still in its infancy, and further studies with well-defined conditions are warranted herein.
Collapse
Affiliation(s)
- Milad Moloudizargari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Govahi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Fallah
- Department of Pharmacology and Toxicology, Medicinal Plant Research Centre, Faculty of Pharmacy, Islamic Azad University, Amol, Iran
| | - Mohammad A Rezvanfar
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), and Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad H Asghari
- Department of Pharmacology and Toxicology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), and Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
17
|
Zhang Y, Ellinger J, Ritter M, Schmidt-Wolf IGH. Clinical Studies Applying Cytokine-Induced Killer Cells for the Treatment of Renal Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12092471. [PMID: 32882824 PMCID: PMC7564072 DOI: 10.3390/cancers12092471] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/28/2020] [Accepted: 08/28/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Cytokine-induced killer (CIK) cells are a heterogeneous population of polyclonal T effector cells expanded ex vivo. Here, we updated our last review published in 2012 and provided a synopsis of current 15 clinical studies, including 382 patients with renal cell carcinoma (RCC) enrolled in CIK cell immunotherapy. CIK cells exhibited promising synergistic anti-tumor effects when combined with conventional therapies and showed mild adverse effects in patients with RCC. Preclinical researches also identified potential molecular targets that augmented CIK cell cytotoxicity against renal carcinoma cells. In future, large randomized clinical trials should be organized to further evaluate the clinical efficacy and optimize the treatment modality of CIK cells in RCC. Abstract There is growing interest in cytokine-induced killer (CIK) cells on the integrated therapy of patients with RCC, especially those in the late stage or refractory to conventional chemotherapy and radiotherapy. In this review, a total of 15 clinical studies including 681 patients enrolled in CIK cell immunotherapy were outlined. Three-hundred-and-eighty-two patients with RCC were treated with CIK cells alone or in combination with DC vaccination, targeted agents sunitinib or sorafenib, and the PD-1 inhibitor pembrolizumab. Significantly improved 3-year overall survival rate was reported in four trials, whereas remarkably longer median progression-free survival was observed in three studies. Adverse reactions were mild and usually controllable fever and fatigue. Besides, preclinical research progresses were reviewed to increase our understanding about the underlying mechanisms of CIK cell cytotoxicity and identify potential targets to enhance their anti-tumor activity. These studies suggest that CIK cell-based immunotherapy has potential clinical benefits with a good safety profile and could become a promising approach in the combined therapies of RCC patients. However, further large-scale studies are required to evaluate the clinical efficacy of CIK cells and more efforts should be performed to identify the optimal CIK cell-based therapeutic regimen for RCC patients.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Integrated Oncology, CIO Bonn, University Hospital Bonn, Venusberg-Campus 1, D 53127 Bonn, Germany;
| | - Jörg Ellinger
- Department of Urology, University Hospital Bonn, Venusberg-Campus 1, D 53127 Bonn, Germany; (J.E.); (M.R.)
| | - Manuel Ritter
- Department of Urology, University Hospital Bonn, Venusberg-Campus 1, D 53127 Bonn, Germany; (J.E.); (M.R.)
| | - Ingo G. H. Schmidt-Wolf
- Department of Integrated Oncology, CIO Bonn, University Hospital Bonn, Venusberg-Campus 1, D 53127 Bonn, Germany;
- Correspondence: ; Tel.: +492-2828-717-048
| |
Collapse
|
18
|
Dendritic cell therapy in cancer treatment; the state-of-the-art. Life Sci 2020; 254:117580. [DOI: 10.1016/j.lfs.2020.117580] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/14/2020] [Accepted: 03/19/2020] [Indexed: 12/29/2022]
|
19
|
Xu H, Qin W, Feng H, Song D, Yang X, Zhang J. Analysis of the Clinical Efficacy of Dendritic Cell -cytokine Induced Killer Cell-based Adoptive Immunotherapy for Colorectal Cancer. Immunol Invest 2020; 50:622-633. [PMID: 32718264 DOI: 10.1080/08820139.2020.1781881] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Background: To analyze the efficacy and safety of dendritic cell - cytokine - induced killer (DC-CIK) immunotherapy combined with chemotherapy for colorectal cancer. Method: A retrospective analysis was conducted in 116 patients from February 2012 to December 2017, who were divided into postoperative adjuvant chemotherapy group alone, combined DC-CIK immunotherapy group, advanced cancer palliative care group, and palliative care + DC-CIK immunotherapy group, to evaluate cellular immune function, disease-free survival(DFS) and overall survival(OS). Results: In the adjuvant therapy and palliative care group, the percentages of CD3+, CD8+ and NK cells after treatment were significantly lower than before, whereas in the other two groups given DC-CIK immunotherapy, the percentages of CD3+, CD8+, NK and NKT cells after treatment were all higher than before, with a significant increase compared with the chemotherapy group (P < .05). DFS (42.4 ± 5.26 m) in the group receiving postoperative adjuvant chemotherapy + DC-CIK immunotherapy was significantly longer than that (23.5 ± 2.79 m) in the group only given postoperative adjuvant chemotherapy (P < .05). OS in the group receiving palliative care + DC-CIK immunotherapy was slightly longer than that in the group only given palliative care for advanced cancer (29 m vs 26 m, P > .05).Conclusion: Combination with DC-CIK immunotherapy could effectively improve cellular immune function. Postoperative adjuvant chemotherapy in combination with DC-CIK immunotherapy could significantly prolong DFS, but palliative care in combination with DC-CIK immunotherapy did not significantly prolong OS in patients with advanced cancer.
Collapse
Affiliation(s)
- Huiru Xu
- Department of Thoracic Oncology, Cancer Center, Shanxi Academy of Medical Sciences (Shanxi Bethune Hospital), Taiyuan, China
| | - Weishan Qin
- Department of Ophthalmology, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Huijing Feng
- Department of Thoracic Oncology, Cancer Center, Shanxi Academy of Medical Sciences (Shanxi Bethune Hospital), Taiyuan, China
| | - Dong Song
- Department of Thoracic Oncology, Cancer Center, Shanxi Academy of Medical Sciences (Shanxi Bethune Hospital), Taiyuan, China
| | - Xiaoling Yang
- Department of Thoracic Oncology, Cancer Center, Shanxi Academy of Medical Sciences (Shanxi Bethune Hospital), Taiyuan, China
| | - Junping Zhang
- Department of Thoracic Oncology, Cancer Center, Shanxi Academy of Medical Sciences (Shanxi Bethune Hospital), Taiyuan, China
| |
Collapse
|
20
|
Zou Y, Liang J, Li D, Fang J, Wang L, Wang J, Zhang J, Guo Q, Yan X, Tang H. Application of the chemokine-chemokine receptor axis increases the tumor-targeted migration ability of cytokine-induced killer cells in patients with colorectal cancer. Oncol Lett 2020; 20:123-134. [PMID: 32565940 PMCID: PMC7286113 DOI: 10.3892/ol.2020.11539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 02/14/2020] [Indexed: 12/24/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are a group of heterogeneous immune cells which can be isolated from human peripheral blood mononuclear cells and have demonstrated therapeutic benefit both in hematologic malignancies and solid tumors, including colorectal cancer. However, poor tumor-targeted migration has limited the clinical efficacy of CIK cell treatment. The chemokine-chemokine receptor (CK-CKR) axis serves a role in the tumor-directed trafficking capacity of immune cells. Investigating the relationship between CKR profiles on the surface of CIK cells and chemokine expression levels in the tumor microenvironment may improve CIK cell therapy. In the present study, the spectrum of chemokine expression levels in tumor tissues from patients with colorectal cancer (CRC) and CKR expression profiles in CIK cells obtained from the same individuals with CRC were investigated. The results showed that chemokine expression levels in tumor tissues exhibited variability and cell line heterogeneity. However, the expression levels of a number of chemokines were similar in different CRC donors and cell lines. Expression levels of CXCLL10, CXCL11 and CCL3 were significantly higher in most tumor tissues compared with adjacent normal tissues and highly expressed in most CRC cell lines. In accordance with chemokine expression levels, CKR profiles on the surface of CIK cells also showed donor-to-donor variability. However, concordant expression profiles of CKRs were identified in different patients with CRC. CXCR3 and CXCR4 were highly expressed on the surface of CIK cells through the culture process. Importantly, the expression levels of all CKRs, especially CCR4, CXCR4 and CXCR3, were notably decreased during the course of CIK cell expansion. The changing trend of CKR profiles were not correlated with the chemokine expression profiles in CRC tissues (CCL3, CXCL12 and CXCL10/CXCL11 were highly expressed in CRC tissue). Re-stimulating CIK cells using chemokines (CCL21 and CXCL11) at the proper time point increased corresponding CKR expression levels on the surface of CIK cells and enhance tumor-targeted trafficking in vitro. These results demonstrated that modification of the CK-CKR axis using exogenous recombinant chemokines at the proper time point enhanced CIK cell trafficking ability and improved CIK antitumor effects.
Collapse
Affiliation(s)
- Yunlian Zou
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650504, P.R. China
- Institute of Medical Sciences, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Jianhua Liang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650504, P.R. China
- Institute of Medical Sciences, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Danyang Li
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650504, P.R. China
- Institute of Medical Sciences, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Jingjing Fang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650504, P.R. China
- Institute of Medical Sciences, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Linping Wang
- Institute of Medical Sciences, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Jinli Wang
- Institute of Medical Sciences, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Jinping Zhang
- Institute of Medical Sciences, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Qiang Guo
- Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Xinmin Yan
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650504, P.R. China
- Institute of Medical Sciences, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Hui Tang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650504, P.R. China
- Institute of Medical Sciences, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
21
|
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: 58] [Impact Index Per Article: 14.5] [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
|
22
|
Cytokine-Induced Killer (CIK) Cells, In Vitro Expanded under Good Manufacturing Process (GMP) Conditions, Remain Stable over Time after Cryopreservation. Pharmaceuticals (Basel) 2020; 13:ph13050093. [PMID: 32408620 PMCID: PMC7281026 DOI: 10.3390/ph13050093] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/09/2020] [Accepted: 05/10/2020] [Indexed: 12/28/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are advanced therapy medicinal products, so their production and freezing process has to be validated before their clinical use, to verify their stability as a drug formulation according to the good manufacturing practice (GMP) guidelines. We designed a stability program for our GMP-manufactured CIK cells, evaluating the viability, identity and potency of cryopreserved CIK cells at varying time periods from freezing, and compared them with fresh CIK cells. We evaluated the effects of the cryopreservation method, transportation, and the length of time of different process phases (pre-freezing, freezing and post-thawing) on the stability of CIK cells. This included a worst case for each stage. The expanded CIK cells were viable for up to 30 min from the addition of the freezing solution, when transported on dry ice within 48 h once frozen, within 60 min from thawing and from 12 months of freezing while preserving their cytotoxic effects. The reference samples, cryopreserved simultaneously in tubes and following the same method, were considered representative of the batch and useful in the case of further analysis. Data obtained from this drug stability program can inform the accurate use of CIK cells in clinical settings.
Collapse
|
23
|
Increase in Efficacy of Checkpoint Inhibition by Cytokine-Induced-Killer Cells as a Combination Immunotherapy for Renal Cancer. Int J Mol Sci 2020; 21:ijms21093078. [PMID: 32349280 PMCID: PMC7246811 DOI: 10.3390/ijms21093078] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 01/05/2023] Open
Abstract
Cytokine-induced killer (CIK) cells are heterogeneous, major histocompatibility complex (MHC)-unrestricted T lymphocytes that have acquired the expression of several natural killer (NK) cell surface markers following the addition of interferon gamma (IFN-γ), OKT3 and interleukin-2 (IL-2). Treatment with CIK cells demonstrates a practical approach in cancer immunotherapy with limited, if any, graft versus host disease (GvHD) toxicity. CIK cells have been proposed and tested in many clinical trials in cancer patients by autologous, allogeneic or haploidentical administration. The possibility of combining them with specific monoclonal antibodies nivolumab and ipilimumab will further expand the possibility of their clinical utilization. Initially, phenotypic analysis was performed to explore CD3, CD4, CD56, PD-1 and CTLA-4 expression on CIK cells and PD-L1/PD-L2 expression on tumor cells. We further treated CIK cells with nivolumab and ipilimumab and measured the cytotoxicity of CIK cells cocultured to renal carcinoma cell lines, A-498 and Caki-2. We observed a significant decrease in viability of renal cell lines after treating with CIK cells (p < 0.0001) in comparison to untreated renal cell lines and anti-PD-1 or anti-CTLA-4 treatment had no remarkable effect on the viability of tumor cells. Using CCK-8, Precision Count Beads™ and Cell Trace™ violet proliferation assays, we proved significant increased proliferation of CIK cells in the presence of a combination of anti-PD-1 and anti-CTLA-4 antibodies compared to untreated CIK cells. The IFN-γ secretion increased significantly in the presence of A-498 and combinatorial blockade of PD-1 and CTLA-4 compared to nivolumab or ipilimumab monotreatment (p < 0.001). In conclusion, a combination of immune checkpoint inhibition with CIK cells augments cytotoxicity of CIK cells against renal cancer cells.
Collapse
|
24
|
Guo Q, Zhang Z, Zhao P, Zou S, Li L, Li N, Sun W, Wei X, Hou L, Yang Z, Gao D. Bispecific antibody activated T cells: A newly developed T cells with enhanced proliferation ability and cytotoxicity. Immunol Lett 2020; 220:79-87. [DOI: 10.1016/j.imlet.2019.12.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 12/12/2019] [Accepted: 12/30/2019] [Indexed: 01/07/2023]
|
25
|
Wang Q, Peng W, Jiang M, Wu L. [Research Progress of Immunotherapy and Prognostic Markers in Small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:182-188. [PMID: 32102135 PMCID: PMC7118334 DOI: 10.3779/j.issn.1009-3419.2020.03.08] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
近年来,虽然非小细胞肺癌患者的治疗效果得到了明显的改善,但小细胞肺癌(small cell lung cancer, SCLC)患者仍因治疗选择有限而预后很差。SCLC发病率占肺癌总体发病率的15%,具有恶性程度高、生长迅速、易发生远处转移及复发的特点,治疗十分棘手。随着免疫治疗获批于非小细胞肺癌等多种实体瘤,作为免疫源性相对较强的癌种,SCLC的相关临床研究也在开展中,并已取得一定的进展。另一方面,由于肿瘤异质性的存在,探索能够预测SCLC疗效的相关标志物对患者精准治疗至关重要。本综述阐述了目前SCLC免疫治疗及与SCLC免疫治疗相关生物标志物研究的最新进展。
Collapse
Affiliation(s)
- Qingyi Wang
- Second Department of Thoracic Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China
| | - Wenying Peng
- Second Department of Thoracic Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China
| | - Meilin Jiang
- Second Department of Thoracic Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China
| | - Lin Wu
- Second Department of Thoracic Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China
| |
Collapse
|
26
|
Li H, Zhang Z, Duan X, Maimela NR, Yang S, Zhao X, Huang J, Zhang Y. Efficacy of cascade-primed cell infusion as an adjuvant immunotherapy with concurrent chemotherapy for patients with non-small-cell lung cancer: A retrospective observational study with a 5-year follow-up. Cytotherapy 2020; 22:35-43. [PMID: 31902660 DOI: 10.1016/j.jcyt.2019.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 11/19/2019] [Accepted: 12/03/2019] [Indexed: 01/23/2023]
Abstract
BACKGROUND Clinical studies have shown the efficacy of combination therapy for various malignancies. In this study, the characteristics, safety and feasibility of use of cascade-primed (CAPRI) cells for the combination treatment of non-small-cell lung cancer (NSCLC) were evaluated both in vitro and in vivo. METHODS Sixty-five patients with stage II-IV NSCLC were recruited. Of these patients, 31 patients received CAPRI cell therapy combined with chemotherapy (CAPRI group), and the other 34 patients constituted the control group and received chemotherapy alone. This study primarily aimed to evaluate the overall survival (OS), progression-free survival (PFS), short-term responses and treatment efficacy. RESULTS CD83, CD1a, CD80 and CD86 marker levels were significantly upregulated in CAPRI cells. Interferon-γ expression levels were highest in CD3+CD8+ cells (33.77% ± 4.40%). Furthermore, interleukin-2 levels were highest in CD3+CD56+ cells (26.73% ± 6.63%), whereas perforin expression levels were similar in CD3+CD8+ and CD3+CD56+ cells. Furthermore, CAPRI cells had a better anti-tumor potential in CD3+CD56+ cells and displayed the highest expression levels of CD107a to H460 and A549 cell lines. The 5-year OS was significantly greater in the CAPRI group than in the control group (P = 0.008), and the PFS of two groups exhibited a significant difference (P = 0.007). Median OS (48 versus 31.6 months; P = 0.004) and PFS (48 versus 36.4 months; P = 0.016) differed between these two groups. Moreover, treatment-associated toxicities were mild and well-tolerated by patients with NSCLC. CONCLUSION CAPRI cell therapy potentially prolongs the survival of patients with NSCLC when combined with chemotherapy.
Collapse
Affiliation(s)
- Hong Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoran Duan
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | | | - Shuangning Yang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuan Zhao
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianmin Huang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China; School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
27
|
Zhang X, Yang J, Zhang G, Song L, Su Y, Shi Y, Zhang M, He J, Song D, Lv F, Wu P, Wang H, Wang T, Zhang Y, Liu H, Lu P. 5 years of clinical DC-CIK/NK cells immunotherapy for acute myeloid leukemia – a summary. Immunotherapy 2020; 12:63-74. [PMID: 31914839 DOI: 10.2217/imt-2019-0108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: To assess the efficacy of dendritic cells-cytokine induced killer (DC-CIK) and natural killer (NK) cell-based immunotherapy in treating the low- and intermediate-risk acute myeloid leukemia. Patients & methods: DC-CIK or NK cells were infused once every 3 months for 2–4 cycles to 85 patients. Results & conclusion: The 5-year overall survival (OS) and relapse-free survival (RFS) rates were 90.5 and 65.2%, respectively. The OS of the very favorable, the favorable and the intermediate-risk groups was 94.4, 86.3 and 93.3% (p = 0.88), and the RFS 83.3, 81.8 and 62.2% (p = 0.14), respectively. The OS and RFS of the 60 patients treated with DC-CIK alternating with NK cells were better than the 25 patients treated with DC-CIK or NK alone (96.5 vs 71.2%; p = 0.003. 79.5 vs 28.9%; p < 0.001).
Collapse
Affiliation(s)
- Xian Zhang
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
- Lu Daopei Institute of Hematology, Beijing 100176, China
| | - Junfang Yang
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
- Lu Daopei Institute of Hematology, Beijing 100176, China
| | - Gailing Zhang
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
| | - Lisong Song
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
| | - Yunchao Su
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
| | - Yanze Shi
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
| | - Min Zhang
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
| | - Jiujiang He
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
| | - Dan Song
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
| | - Fanyong Lv
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
- Lu Daopei Institute of Hematology, Beijing 100176, China
| | - Ping Wu
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
- Lu Daopei Institute of Hematology, Beijing 100176, China
| | - Hui Wang
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
- Lu Daopei Institute of Hematology, Beijing 100176, China
| | - Tong Wang
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
- Lu Daopei Institute of Hematology, Beijing 100176, China
| | - Yang Zhang
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
| | - Hongxing Liu
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
- Lu Daopei Institute of Hematology, Beijing 100176, China
| | - Peihua Lu
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
- Lu Daopei Institute of Hematology, Beijing 100176, China
| |
Collapse
|
28
|
Chen X, Feng R, Xiong D, Yang S, Lin T. Effect of lentiviral vector-packaged interleukin-18 gene on the malignant behavior of lung cancer. Exp Ther Med 2019; 19:319-326. [PMID: 31853306 DOI: 10.3892/etm.2019.8204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 06/27/2019] [Indexed: 11/06/2022] Open
Abstract
Interleukin-18 (IL-18) is a multifunctional cytokine that exhibits antitumor, anti-infection and immunoregulatory functions. This study aimed to investigate the effects of lentiviral vector-packaged interleukin (IL)-18 gene on the malignant behavior of lung cancer and the potential underlying molecular mechanism of IL-18 anticancer activity. Human lung adenocarcinoma A549 cells transfected with human IL-18 gene-containing lentiviral expression vector were the IL-18 intervention group (group A), cells transfected with the empty lentiviral expression vector were empty vector group (group B), and cells without any intervention were the blank control group (group C). Reverse transcription-quantitative PCR and western blotting were used to determine IL-18 mRNA and protein expression levels. Cell Counting Kit-8, colony-formation, flow cytometry, invasion and wound-healing assays were used to evaluate the malignant behavior of A549 cells transfected with the IL-18 lentiviral vector. The expression levels of the T helper (Th)1 cell cytokine interferon-γ (IFN-γ) and the Th2 cell cytokine IL-4 were tested by ELISA, and western blotting was used to test the expressing of nuclear factor κB (NF-κB). The results demonstrated that IL-18 mRNA and protein expression levels in group A were significantly increased compared with groups B and C; the expression levels of IFN-γ in group A were higher and the expression levels of IL-4 in group A were lower compared with those in groups B and C; and the expression of NF-κB was increased in the cytoplasm and decreased in the nucleus in group A compared with groups B and C. The data indicated that, compared with the control groups, the IL-18 gene lentiviral expression vector increased the expression of IL-18, diminished A549 cell proliferative ability, enhanced apoptosis, decreased the invasive and metastatic capacities of the cells, promoted the secretion of IFN-γ, decreased the production of IL-4, reversed the imbalance of Th1/Th2 cell subsets and inhibited the nuclear activation of NF-κB, which collectively present an anti-lung cancer mechanism and deserve further study.
Collapse
Affiliation(s)
- Xiangqi Chen
- Teaching and Research Department of Respiratory Medicine, Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China.,Department of Respiratory Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Rui Feng
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Donglan Xiong
- Teaching and Research Department of Respiratory Medicine, Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Sheng Yang
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China.,Teaching and Research Department of Oncology Medicine, Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China.,Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350001, P.R. China.,Fujian Medical University Stem Cell Research Institute, Fuzhou, Fujian 350001, P.R. China
| | - Tingyan Lin
- Teaching and Research Department of Respiratory Medicine, Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China.,Department of Respiratory Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
29
|
Aldehyde dehydrogenase-positive melanoma stem cells in tumorigenesis, drug resistance and anti-neoplastic immunotherapy. Mol Biol Rep 2019; 47:1435-1443. [PMID: 31838656 DOI: 10.1007/s11033-019-05227-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/07/2019] [Indexed: 02/07/2023]
Abstract
Cancer stem cells (CSCs), a rare subset of cancer cells, are well known for their self-renewing capacity. CSCs play a critical role in therapeutic failure and are responsible for poor prognosis in leukemia and various solid tumors. However, it is still unclear how CSCs initiate carcinogenesis and evade the immune response. In humans, the melanoma initiating cells (MICs) are recognized as the CSCs in melanomas, and were verified to possess CSC potentials. The enzymatic system, aldehyde dehydrogenase (ALDH) is considered to be a specific marker for CSCs in several tumors. The expression of ALDH in MICs may be closely correlated with phenotypic heterogeneity, melanoma-genesis, metastasis, and drug resistance. The ALDH+ CSCs/MICs not only serve as an indicator for therapeutic efficacy, but have also become a target for the treat of melanoma. In this review, we initially introduce the multiple capacities of MICs in melanoma. Then, we summarize in vivo and in vitro studies that illustrate the relationship between ALDH and MICs. Furthermore, understanding of chemotherapy resistance in melanoma relies on ALDH+ MICs. Finally, we review studies that focus on melanoma immunotherapies, rendering ALDH a potential marker to evaluate the efficacy of anti-neoplastic therapies or an adjuvant anti-melanoma target.
Collapse
|
30
|
Huang L, Qiao G, Morse MA, Wang X, Zhou X, Wu J, Hobeika A, Ren J, Lyerly HK. Predictive significance of T cell subset changes during ex vivo generation of adoptive cellular therapy products for the treatment of advanced non-small cell lung cancer. Oncol Lett 2019; 18:5717-5724. [PMID: 31788044 PMCID: PMC6865835 DOI: 10.3892/ol.2019.10964] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 06/06/2019] [Indexed: 12/24/2022] Open
Abstract
Adoptive T cell immunotherapy with cytokine-induced killer cells (CIKs) has been demonstrated to prolong the survival of patients with advanced non-small cell lung cancer (NSCLC). The aim of the present study was to evaluate whether the expansion of effector T cells and the decrease of regulatory T cells (Tregs) that occurred during the ex vivo generation of DC-CIKs were associated with improved clinical outcome in patients who received treatment. CIKs were generated ex vivo over a 28-day period from the peripheral blood apheresis product of 163 patients with advanced cancer (including 30 with NSCLC). CIKs were also generated from an additional cohort of 65 patients with NSCLC over a 15-day period. The progression-free survival (PFS) and overall survival (OS) time of patients treated with CIKs was determined by reviewing the patients' medical records. The number of CIKs gradually increased during the culture period and peaked at day 15, followed by a slight decline until day 28. Similarly, the percentages of T cell subtypes associated with anti-tumor activity (CD3+, CD3+CD4+, CD3+CD8+ and CD8+CD28+) peaked at day 15. Although the percentage of CD4+CD25+CD127+ Tregs increased by day 7, a decrease was subsequently observed. Among the 95 patients with NSCLC, those with a post/pre-culture ratio of CD8+CD28+ T lymphocytes >2.2 had significantly better PFS and OS compared with those with ratios ≤2.2. Those with a post/pre-culture CD4+CD25+CD127+ Treg ratio ≤0.6 had significantly better OS and PFS compared with those with ratios >0.6. The peak expansion of CIKs from peripheral blood mononuclear cells occurred at day 15 of ex vivo culture. PFS and OS were associated with post/pre-culture CD8+CD28+ T lymphocyte ratio >2.2 and post/pre-culture CD4+CD25+CD127+ Treg ratio <0.6 in the CIKs of patients with advanced NSCLC treated with adoptive T cell immunotherapy. Further efforts are underway to optimize the DC-CIK infusion for cancer immunotherapy.
Collapse
Affiliation(s)
- Lefu Huang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Beijing 100038, P.R. China
| | - Guoliang Qiao
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Beijing 100038, P.R. China
| | - Michael A Morse
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA.,Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Xiaoli Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Beijing 100038, P.R. China
| | - Xinna Zhou
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Beijing 100038, P.R. China
| | - Jiangping Wu
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Beijing 100038, P.R. China
| | - Amy Hobeika
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Jun Ren
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Beijing 100038, P.R. China.,Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Herbert K Lyerly
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
31
|
Wang J, Shen T, Wang Q, Zhang T, Li L, Wang Y, Fang Y. The long-term efficacy of cytokine-induced killer cellular therapy for hepatocellular carcinoma: a meta-analysis. Immunotherapy 2019; 11:1325-1335. [PMID: 31578914 DOI: 10.2217/imt-2019-0079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Aim: The long-term efficacy of cytokine-induced killer cellular therapy for hepatocellular carcinoma patients after curative treatments remains controversial. Methods: A meta-analysis was conducted, and the outcomes were the recurrence rate and overall survival. Results: Eight randomized clinical trials with 1038 participants were included. Compared with the control group, cytokine-induced killer cellular therapy group could reduce 1-year, 3-year recurrence rates, as well as improve 1-5 years overall survival for hepatocellular carcinoma patients (p < 0.05). However, it failed to affect the 5-year recurrence rate and 6-year overall survival (p > 0.05). Conclusion: Cytokine-induced killer cellular adjuvant therapy exerted a favorable role in improving early and long-term efficacy for hepatocellular carcinoma patients.
Collapse
Affiliation(s)
- Jiaxue Wang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,Department of Pharmacy Administration & Clinical Pharmacy, School of Pharmaceutical, Peking University, Beijing, China
| | - Tiantian Shen
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Qi Wang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,Department of Pharmacy Administration & Clinical Pharmacy, School of Pharmaceutical, Peking University, Beijing, China
| | - Tan Zhang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,Department of Pharmacy Administration & Clinical Pharmacy, School of Pharmaceutical, Peking University, Beijing, China
| | - Lujin Li
- The Center for Drug Clinical Research of Shanghai University of TCM, Shanghai, China
| | - Yu Wang
- Immunotech Applied Science Limited, Beijing, China
| | - Yi Fang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| |
Collapse
|
32
|
Wu D, Yu Y, Zhao C, Shou X, Piao Y, Zhao X, Zhao Y, Wang S. NK-Cell-Encapsulated Porous Microspheres via Microfluidic Electrospray for Tumor Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:33716-33724. [PMID: 31454227 DOI: 10.1021/acsami.9b12816] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Immunotherapy has recently garnered significant research interest in the field of clinical cancer management. The potential of tumor immunotherapy has been demonstrated for targeting a variety of tumors, both in vivo and in vitro, yielding some remarkable therapeutic effects. Herein, inspired by the stem cell niche, we developed a scale-up approach to generating porous microspheres with encapsulated natural killer (NK) cells via microfluidic electrospray for in situ tumor immunotherapy. The generated microspheres contained porous microstructures with tunable morphologies because of versatile but precise fluid control in the microfluidic electrospray system. NK-92MI cells encapsulated in porous microspheres were protected from the outer complex environment, allowing for improved proliferation and functionality. As observed, perforin and granzymes were sustainably secreted from the encapsulated NK-92MI cells, which exhibited robust killing effects on tumors both in vitro and in vivo. With continual proliferation, NK-92MI cells budded from the surface of porous microspheres and migrated into the surrounding residual tumor tissues, further destroying tumor cells. More importantly, no side effects owing to the native host immune system were observed by injecting the NK-92MI cell-encapsulated microspheres into tumors in vivo. Therefore, the NK-cell-encapsulated porous microspheres show great potential for tumor immunotherapy, offering a robust and attractive treatment option for cancer patient management.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cells, Immobilized/immunology
- Cells, Immobilized/pathology
- Cells, Immobilized/transplantation
- Humans
- Immunity, Cellular
- Immunotherapy
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Killer Cells, Natural/transplantation
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Microspheres
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Porosity
Collapse
Affiliation(s)
- Dan Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases , National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University , Hangzhou , Zhejiang Province 310003 , China
- Institute for Translational Medicine , Zhejiang University , Hangzhou , Zhejiang Province 310029 , China
| | - Yunru Yu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering , Southeast University , Nanjing 210096 , China
| | - Cheng Zhao
- Department of General Surgery, Jinling Hospital , Medical School of Nanjing University , Nanjing 210002 , China
| | - Xin Shou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases , National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University , Hangzhou , Zhejiang Province 310003 , China
- Institute for Translational Medicine , Zhejiang University , Hangzhou , Zhejiang Province 310029 , China
| | - Yun Piao
- Department of Biomedical Engineering , The Hong Kong Polytechnic University , Hong Kong 999077 , China
| | - Xin Zhao
- Department of Biomedical Engineering , The Hong Kong Polytechnic University , Hong Kong 999077 , China
| | - Yuanjin Zhao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering , Southeast University , Nanjing 210096 , China
| | - Shuqi Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases , National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University , Hangzhou , Zhejiang Province 310003 , China
- Institute for Translational Medicine , Zhejiang University , Hangzhou , Zhejiang Province 310029 , China
| |
Collapse
|
33
|
Zou B, Xia S, Du X, Xu Y, Ning N, Li S, Teng D, Li H, Hu Z, Hu S, Wang Y. Treatment Effect of Tuftsin and Antigen Peptide Combined with Immune Cells on Colorectal Cancer. Med Sci Monit 2019; 25:5465-5472. [PMID: 31333222 PMCID: PMC6668490 DOI: 10.12659/msm.915037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background The aim of this study was to investigate the effect of antigenic peptides on dendritic cell maturation and activation as well as the role of dendritic cell induced cell function. The tumor-specific cytotoxic T lymphocytes induced by activation of the dendritic cells were also evaluated. Material/Methods SW-480 cell lysate and peptide antigens were selected as adjuvants in dendritic cell sensitization, and tuftsin was used to induce the phagocytosis of dendritic cells. Immature dendritic cells were stimulated with the antigen and adjuvant as follows: group A was negative control; group B was SW-480 (20 μg/mL); group C was SW-480 (20 μg/mL)+tumor necrosis factor (TNF)-α (10 μg/mL); group D was SW-480 (20 μg/mL)+tuftsin (20 μg/mL); group E was antigen peptide (2 μg/mL); group F was antigen peptide (2 μg/mL)+TNF-α (10 μg/mL); group G was antigen peptide (2 μg/mL)+tuftsin (20 μg/mL). Cytotoxic T lymphocytes activation and in vitro anti-tumor effects were examined by detecting the maturation marks of dendritic cells as well as interleukin (IL)-10 and IL-12 levels secreted by dendritic cells. Cells with the strongest immunizing effects were injected into nude mice and tumor suppression status was evaluated. Results Group D (SW-480+tuftsin), group E (antigen peptides), group F (antigen peptide+TNF-α), and group G (antigen peptides+tuftsin) displayed significant differences compared to the control group (P<0.05). Group G (antigen peptides+tuftsin) could also promote the secretion of cytokines IL-12, as well as inhibit cytokine IL-10 secretion, compared to the other experimental groups (P<0.05). In the in vivo experiments of tumor inhibitions, antigenic polypeptide+tuftsin was the most effective (P<0.05). Conclusions Combination of cytotoxic T lymphocytes and T peptide therapy in treating human colorectal cancer might be used as a new treatment strategy based on adoptive cellular immunotherapy.
Collapse
Affiliation(s)
- Boyuan Zou
- Department of Retroperitoneal Tumor Surgery, Peking University International Hospital, Beijing, China (mainland).,Department of General Surgery, The General Hospital of People's Liberation Army, Beijing, China (mainland)
| | - Shaoyou Xia
- Department of General Surgery, The General Hospital of People's Liberation Army, Beijing, China (mainland)
| | - Xiaohui Du
- Department of General Surgery, The General Hospital of People's Liberation Army, Beijing, China (mainland)
| | - Yingxin Xu
- Department of General Surgery, The General Hospital of People's Liberation Army, Beijing, China (mainland)
| | - Ning Ning
- Department of General Surgery, The General Hospital of People's Liberation Army, Beijing, China (mainland).,Department of Gastrointestinal Surgery, Peking University International Hospital, Beijing, China (mainland)
| | - Songyan Li
- Department of General Surgery, The General Hospital of People's Liberation Army, Beijing, China (mainland)
| | - Da Teng
- Department of General Surgery, The General Hospital of People's Liberation Army, Beijing, China (mainland)
| | - Hao Li
- Department of General Surgery, The General Hospital of People's Liberation Army, Beijing, China (mainland)
| | - Zilong Hu
- Department of General Surgery, The General Hospital of People's Liberation Army, Beijing, China (mainland)
| | - Shidong Hu
- Department of General Surgery, The General Hospital of People's Liberation Army, Beijing, China (mainland)
| | - Yufeng Wang
- Department of General Surgery, The General Hospital of People's Liberation Army, Beijing, China (mainland)
| |
Collapse
|
34
|
Xue W, Yan D, Kan Q. Interleukin-35 as an Emerging Player in Tumor Microenvironment. J Cancer 2019; 10:2074-2082. [PMID: 31205568 PMCID: PMC6548173 DOI: 10.7150/jca.29170] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 03/05/2019] [Indexed: 12/15/2022] Open
Abstract
IL-35 is the newest member of IL-12 family. A dimeric protein consisting of two separate subunits has manifested suppressive actions on immune system, which is counterproductive in the context of cancers. Various reports have confirmed its inhibitory role on immune system which is carried out via formation of IL-35-producing regulatory T cells (iTr35), increased Treg development and suppressive Th17 cells growth. Although last decade has seen a great deal of scientific interest on this subject, the exact role, precise signal transduction and elaborative functions of IL-35 in tumor microenvironment (TME) remained elusive. Search for anti-IL-35 therapies have exhibited limited success in animal models. Contrarily, few studies have denied the idea that IL-35 plays a role in cancer. The purpose of this review is to analyze the reported scientific data on continuous symphony of IL-35 in cancers since the inception of former.
Collapse
Affiliation(s)
- Wenhua Xue
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Dan Yan
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Quancheng Kan
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
35
|
Liu C, Cui X, Zhou D, Li C, Zhao M, Jin Y, Ding C, Zhu Y. Cytokine-induced killer cells co-cultured with non-cell derived targeting peptide-loaded dendritic cells induce a specific antitumor response. Cancer Biol Ther 2019; 20:720-728. [PMID: 30777479 DOI: 10.1080/15384047.2018.1564561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cancer is a severe lethal disease. Currently, immunotherapy has become an effective alternative therapeutic approach for cancers. Cytokine-induced killer (CIK) cells have a higher proliferation rate, increased efficacy with few side-effects, and non-MHC-restricted killing after co-culturing with dendritic cells (DCs). Therefore, it has been widely studied and applied in the treatment of cancers. In our study, we explored the antitumor effects of CIK cells co-culturing with DCs pulsed with non-cell derived targeting peptides, which could specifically bind to certain tumor cells. Our results indicated that targeting peptide-loaded DCs could enhance the differentiation and cytotoxicity of CIK cells. Moreover, CIK cells, which were treated with specific targeting peptide-loaded DCs, could effectively and specifically kill tumor cells in vitro and in vivo, as long as tumor cells were pre-coated with the specific binding peptides. In conclusion, targeting peptides could guide DC-CIK to effectively and specifically kill tumor cells which were pre-coated with these targeting peptides and non-cell derived targeting peptide-loaded-DC-CIK may work as a novel means for cancer therapy.
Collapse
Affiliation(s)
- Cuijuan Liu
- a School of Nano Technology and Nano Bionics , University of Science and Technology of China , Hefei , China.,b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China
| | - Xueyuan Cui
- b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China.,c College of Life Sciences , Shanghai University , Shanghai , China
| | - Dayong Zhou
- d Department of Vascular Surgery , Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital , Suzhou , China
| | - Chunlin Li
- b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China
| | - Mengya Zhao
- b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China.,c College of Life Sciences , Shanghai University , Shanghai , China
| | - Yaqing Jin
- b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China
| | - Chen Ding
- b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China.,e China Pharmaceutical University , Nanjing , China
| | - Yimin Zhu
- b CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics , Chinese Academy of Sciences , Suzhou , China
| |
Collapse
|
36
|
Zhang Y, Wang S, Yang B, Lu S, Du Y, Liu H. Adjuvant treatment for triple-negative breast cancer: a retrospective study of immunotherapy with autologous cytokine-induced killer cells in 294 patients. Cancer Biol Med 2019; 16:350-360. [PMID: 31516755 PMCID: PMC6713632 DOI: 10.20892/j.issn.2095-3941.2018.0378] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objective To examine the efficacy and safety of a sequential combination of chemotherapy and autologous cytokine-induced killer (CIK) cell treatment in triple-negative breast cancer (TNBC) patients. Methods A total of 294 post-surgery TNBC patients participated in the research from January 1, 2009 to January 1, 2015. After adjuvant chemotherapy, autologous CIK cells were introduced in 147 cases (CIK group), while adjuvant chemotherapy alone was used to treat the remaining 147 cases (control group). The major endpoints of the investigation were the disease-free survival (DFS) and overall survival (OS). Additionally, the side effects of the treatment were evaluated. Results In the CIK group, the DFS and OS intervals of the patients were significantly longer than those of the control group (DFS: P = 0.047; OS: P = 0.007). The multivariate analysis demonstrated that the TNM (tumor-node-metastasis) stage and adjuvant CIK treatment were independent prognostic factors for both DFS [hazard ratio (HR) = 0.520, 95% confidence interval (CI):0.271-0.998, P = 0.049; HR = 1.449, 95% CI:1.118-1.877, P = 0.005, respectively] and OS (HR=0.414, 95% CI:0.190-0.903, P = 0.027; HR = 1.581, 95% CI:1.204-2.077, P = 0.001, respectively) in patients with TNBC. Additionally, longer DFS and OS intervals were associated with increased number of CIK treatment cycles (DFS: P = 0.020; OS: P = 0.040). The majority of the patients who benefitted from CIK cell therapy were relatively early-stage TNBC patients.
Conclusion Chemotherapy in combination with adjuvant CIK could be used to lower the relapse and metastasis rate, thus effectively extending the survival time of TNBC patients, especially those at early stages.
Collapse
Affiliation(s)
- Yuhan Zhang
- The Second Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Shuaibing Wang
- Oncology Department, China National Petroleum Corporation Central Hospital, Langfang 065000, China
| | - Beibei Yang
- The Second Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Su Lu
- The Second Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Yiyi Du
- The Second Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Hong Liu
- The Second Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| |
Collapse
|
37
|
Zhou X, Mo X, Qiu J, Zhao J, Wang S, Zhou C, Su Y, Lin Z, Ma H. Chemotherapy combined with dendritic cell vaccine and cytokine-induced killer cells in the treatment of colorectal carcinoma: a meta-analysis. Cancer Manag Res 2018; 10:5363-5372. [PMID: 30464632 PMCID: PMC6225919 DOI: 10.2147/cmar.s173201] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aim To investigate the efficacy and safety of dendritic cell (DC) vaccine combined with cytokine-induced killer (CIK) cell therapy in colorectal carcinoma (CRC). Patients and methods PubMed, Embase, and Cochrane Library databases were searched systematically for clinical trials of DC vaccine and CIK cell therapy combined with chemotherapy for CRC. The primary and secondary endpoints were overall survival (OS) and disease-free survival (DFS), respectively. Pooled risk ratios were used to assess the treatment efficacy. Both random and fixed effects models were used for statistical analysis. The study population consisted of 871 CRC patients enrolled in four trials. Results OS and DFS were significantly improved in patients who received chemotherapy combined with DC vaccine and CIK cells, and no severe adverse events were shown. Conclusions The study demonstrated that the addition of DC vaccine and CIK cell therapy to chemotherapy is feasible and effective in patients with CRC.
Collapse
Affiliation(s)
- Xiuling Zhou
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China, ;
| | - Xiangqiong Mo
- Department of General Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Junlan Qiu
- Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science & Technology Town Hospital, Jiangsu 215153, China
| | - Jingjing Zhao
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shuncong Wang
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China, ;
| | - Cuiling Zhou
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China, ;
| | - Yonghui Su
- Department of General Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Zhong Lin
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China, ;
| | - Haiqing Ma
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China, ;
| |
Collapse
|
38
|
Combination of DC/CIK adoptive T cell immunotherapy with chemotherapy in advanced non-small-cell lung cancer (NSCLC) patients: a prospective patients’ preference-based study (PPPS). Clin Transl Oncol 2018; 21:721-728. [DOI: 10.1007/s12094-018-1968-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/16/2018] [Indexed: 01/06/2023]
|
39
|
Zhao JJ, Zhou S, Chen CL, Zhang HX, Zhou ZQ, Wu ZR, Liu Y, Pan QZ, Zhu Q, Tang Y, Xia JC, Weng DS. Clinical Effect of Adjuvant Cytokine-Induced Killer Cells Immunotherapy in Patients with Stage II-IVB Nasopharyngeal Carcinoma after Chemoradiotherapy: A propensity score analysis. J Cancer 2018; 9:4204-4214. [PMID: 30519321 PMCID: PMC6277608 DOI: 10.7150/jca.25790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/27/2018] [Indexed: 12/24/2022] Open
Abstract
As an adjuvant immunotherapy, cytokine-induced killer cells (CIKs) infusion has been demonstrated to exert potent effectiveness in several types of cancer patients who received curative treatment. However, controversy exists regarding whether nasopharyngeal carcinoma (NPC) patients can benefit from additional treatment after radical radiotherapy or chemoradiotherapy to improve their distant control and survival. In this retrospective study, we aimed to evaluate the efficacy of adjuvant CIK cells therapy in NPC patients with stage II-IVB after curative treatment. From January 1, 2005 to December 31, 2012, 85 pairs of NPC patients matching by propensity score matching (PSM) method to balance prognostic factors were included in this study: 85 cases underwent radical treatment, 85 cases received radical treatment and sequential CIKs infusion. We found that disease-free survival (DFS) and overall survival (OS) were significantly better in the CIK group than that in the control group (P = 0.009, P < 0.001, respectively). Adjuvant CIK cells immunotherapy was showed to be an independent prognostic factor for survival of the patients in further multivariate analysis. In subgroup analyses, the DFS and OS of patients with T3/4, III and IV A-B TNM (tumor-node-metastasis) stages were significantly enhanced in CIK group compared to control group. Nevertheless, both NPC patients with high and low EBV DNA benefited from adjuvant CIK cells immunotherapy. In conclusion, CIKs infusion is an effective adjuvant immunotherapy for enhancing the prognosis of NPC patients who have received the standard treatment, particularly for those with more aggressive tumor (T3/4) or advanced TNM stage.
Collapse
Affiliation(s)
- Jing-Jing Zhao
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shu Zhou
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chang-Long Chen
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hong-Xia Zhang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zi-Qi Zhou
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zheng-Rong Wu
- Department of Pathology, School of Basic Medicine, Southern Medical University, Guangzhou, China
| | - Yuan Liu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiu-Zhong Pan
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qian Zhu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan Tang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jian-Chuan Xia
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - De-Sheng Weng
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
40
|
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: 0.7] [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
|
41
|
Castiglia S, Adamini A, Rustichelli D, Castello L, Mareschi K, Pinnetta G, Leone M, Mandese A, Ferrero I, Mesiano G, Fagioli F. Cytokines induced killer cells produced in good manufacturing practices conditions: identification of the most advantageous and safest expansion method in terms of viability, cellular growth and identity. J Transl Med 2018; 16:237. [PMID: 30157948 PMCID: PMC6116438 DOI: 10.1186/s12967-018-1613-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 08/21/2018] [Indexed: 12/14/2022] Open
Abstract
Background Cytokine-induced killer (CIK) cells are a very promising cell population raising growing interest in the field of cellular antitumor therapy. The aim of our study was to validate the most advantageous expansion method for this advanced therapy medicinal product (ATMP) and to translate it from preclinical field to good manufacturing practices (GMP). GMP ensures that ATMP are consistently produced and controlled to the quality standards required to their intended use. For this reason, the use of the xenogenic sera tended to be minimized by GMP for their high variability and the associated risk of transmitting infectious agents. Results We decided to replace Fetal Bovine Serum (FBS), largely used as medium supplement for CIKs expansion, with other culture media. Firstly, Human Serum (HS) and Human Pool Plasma (HPP) were tested as medium supplements giving not compliant results to acceptance criteria, established for CIKs, probably for the great batch to batch variability. Consequently, we decided to test three different serum free expansion media: X-VIVO 15, (largely used by other groups) and Tex Macs and Cell Genix GMP SCGM: two GMP manufactured media. We performed a validation consisting in three run-sand even if the small number of experiments didn’t permit us to obtained statistical results we demonstrated that both X-VIVO 15 and Tex Macs fulfilled the quality standards in terms of cellular growth, viability and identity while Cell Genix GMP SCGM resulted not compliant as it caused some technical problems such as high mortality. Conclusion In conclusion, these preclinical validation data lay the bases for a GMP-compliant process to improve the CIKs expansion method.
Collapse
Affiliation(s)
- Sara Castiglia
- City of Health and Science Hospital of Turin, Pediatric Oncoematology, Regina Margherita Children's Hospital, Piazza Polonia 94, 10126, Turin, Italy.
| | - Aloe Adamini
- City of Health and Science Hospital of Turin, Pediatric Oncoematology, Regina Margherita Children's Hospital, Piazza Polonia 94, 10126, Turin, Italy
| | - Deborah Rustichelli
- City of Health and Science Hospital of Turin, Pediatric Oncoematology, Regina Margherita Children's Hospital, Piazza Polonia 94, 10126, Turin, Italy
| | - Laura Castello
- City of Health and Science Hospital of Turin, Pediatric Oncoematology, Regina Margherita Children's Hospital, Piazza Polonia 94, 10126, Turin, Italy
| | - Katia Mareschi
- City of Health and Science Hospital of Turin, Pediatric Oncoematology, Regina Margherita Children's Hospital, Piazza Polonia 94, 10126, Turin, Italy.,Department of Public Health and Pediatrics, University of Turin, 10126, Turin, Italy
| | - Giuseppe Pinnetta
- City of Health and Science Hospital of Turin, Pediatric Oncoematology, Regina Margherita Children's Hospital, Piazza Polonia 94, 10126, Turin, Italy
| | - Marco Leone
- City of Health and Science Hospital of Turin, Pediatric Oncoematology, Regina Margherita Children's Hospital, Piazza Polonia 94, 10126, Turin, Italy
| | - Alessandra Mandese
- City of Health and Science Hospital of Turin, Pediatric Oncoematology, Regina Margherita Children's Hospital, Piazza Polonia 94, 10126, Turin, Italy
| | - Ivana Ferrero
- City of Health and Science Hospital of Turin, Pediatric Oncoematology, Regina Margherita Children's Hospital, Piazza Polonia 94, 10126, Turin, Italy.,Department of Public Health and Pediatrics, University of Turin, 10126, Turin, Italy
| | | | - Franca Fagioli
- City of Health and Science Hospital of Turin, Pediatric Oncoematology, Regina Margherita Children's Hospital, Piazza Polonia 94, 10126, Turin, Italy.,Department of Public Health and Pediatrics, University of Turin, 10126, Turin, Italy
| |
Collapse
|
42
|
Xia F, Hou W, Liu Y, Wang W, Han Y, Yang M, Zhi X, Li C, Qi D, Li T, Martinez de la Fuente J, Zhang C, Song J, Cui D. Cytokine induced killer cells-assisted delivery of chlorin e6 mediated self-assembled gold nanoclusters to tumors for imaging and immuno-photodynamic therapy. Biomaterials 2018; 170:1-11. [DOI: 10.1016/j.biomaterials.2018.03.048] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/26/2018] [Accepted: 03/28/2018] [Indexed: 01/02/2023]
|
43
|
Li M, Wang Y, Wei F, An X, Zhang N, Cao S, Ren B, Zhang X, Ren X. Efficiency of Cytokine-Induced Killer Cells in Combination with Chemotherapy for Triple-Negative Breast Cancer. J Breast Cancer 2018; 21:150-157. [PMID: 29963110 PMCID: PMC6015982 DOI: 10.4048/jbc.2018.21.2.150] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 04/18/2018] [Indexed: 12/31/2022] Open
Abstract
Purpose The treatment of triple-negative breast cancer (TNBC) remains challenging, due to the absence of estrogen, progesterone, and human epidermal growth factor receptors. This study was designed to evaluate the efficiency and safety of cytokine-induced killer (CIK) cell immunotherapy, following regular chemotherapy, for patients with TNBC. Methods A total of 340 patients with postmastectomy TNBC, from January 1, 2010 to June 30, 2014, were included in this retrospective study. Seventy-seven patients received CIK cell immunotherapy, following regular chemotherapy (arm 1), and 263 patients received regular chemotherapy alone (arm 2). The primary aim was overall survival (OS) and disease-free survival (DFS), and the treatment responses and adverse events were also evaluated. Results The 5-year DFS and OS rates in arm 1 were 77.9% and 94.3%, compared with 69.8% and 85.6% in arm 2, respectively (p=0.159 and p=0.035, respectively). This clearly shows that there was no statistical difference in the 5-year DFS between the two groups. Multivariate analyses of arm 1 indicated that a Karnofsky performance score (KPS) ≥90 and stage I/IIA disease were significantly associated with a prolonged DFS period (hazard ratio [HR], 0.25; 95% confidence interval [CI], 0.09–0.74; p=0.012; and HR 0.21; 95% CI, 0.06–0.82; p=0.024, respectively), but a KPS ≥90 and stage I/IIA disease were not independent prognostic factors for OS. Toxicity was mild in patients who received the CIK therapy. Conclusion The data suggested that CIK cell immunotherapy improved the efficiency of regular chemotherapy in patients with TNBC, and the side effects of CIK cell immunotherapy were mild.
Collapse
Affiliation(s)
- Man Li
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Yang Wang
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Feng Wei
- National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiumei An
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Naining Zhang
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Shui Cao
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Baozhu Ren
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xinwei Zhang
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiubao Ren
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
44
|
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: 0.5] [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
|
45
|
Mukaida N, Nakamoto Y. Emergence of immunotherapy as a novel way to treat hepatocellular carcinoma. World J Gastroenterol 2018; 24:1839-1858. [PMID: 29740200 PMCID: PMC5937202 DOI: 10.3748/wjg.v24.i17.1839] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/15/2018] [Accepted: 04/23/2018] [Indexed: 02/06/2023] Open
Abstract
Tumor immunity proceeds through multiple processes, which consist of antigen presentation by antigen presenting cells (APCs) to educate effector cells and destruction by the effector cytotoxic cells. However, tumor immunity is frequently repressed at tumor sites. Malignantly transformed cells rarely survive the attack by the immune system, but cells that do survive change their phenotypes to reduce their immunogenicity. The resultant cells evade the attack by the immune system and form clinically discernible tumors. Tumor microenvironments simultaneously contain a wide variety of immune suppressive molecules and cells to dampen tumor immunity. Moreover, the liver microenvironment exhibits immune tolerance to reduce aberrant immune responses to massively-exposed antigens via the portal vein, and immune dysfunction is frequently associated with liver cirrhosis, which is widespread in hepatocellular carcinoma (HCC) patients. Immune therapy aims to reduce tumor burden, but it is also expected to prevent non-cancerous liver lesions from progressing to HCC, because HCC develops or recurs from non-cancerous liver lesions with chronic inflammatory states and/or cirrhosis and these lesions cannot be cured and/or eradicated by local and/or systemic therapies. Nevertheless, cancer immune therapy should augment specific tumor immunity by using two distinct measures: enhancing the effector cell functions such as antigen presentation capacity of APCs and tumor cell killing capacity of cytotoxic cells, and reactivating the immune system in immune-suppressive tumor microenvironments. Here, we will summarize the current status and discuss the future perspective on immune therapy for HCC.
Collapse
MESH Headings
- Antigen Presentation/genetics
- Antigens, Neoplasm/immunology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/immunology
- Humans
- Immune Tolerance/genetics
- Immunotherapy/methods
- Immunotherapy/trends
- Liver/immunology
- Liver/pathology
- Liver Neoplasms/genetics
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Lymphocyte Activation/genetics
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/therapy
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Ishikawa, Kanazawa 920-1192, Japan
| | - Yasunari Nakamoto
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Eiheiji-cho, Fukui 910-1193, Japan
| |
Collapse
|
46
|
Peng H, Yao M, Fan H, Song L, Sun J, Zhou Z, Du Y, Lu K, Li T, Yin A, Xu J, Wei S. Effects of Autologous Cytokine-Induced Killer Cells Infusion in Colorectal Cancer Patients: A Prospective Study. Cancer Biother Radiopharm 2018; 32:221-226. [PMID: 28820635 DOI: 10.1089/cbr.2017.2246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIM To evaluate the efficacy and safety of postoperative adjuvant immunotherapy with cytokine-induced killer (CIK) cells in combination with chemotherapy (CT) in colorectal cancer (CRC) patients. MATERIALS AND METHODS A total of 46 patients were randomly assigned to either group 1 (control group) or group 2 (CIK group) using blocked randomization. Both groups received the FOLFOX4 (5-fluorouridine, leucovorin, and oxaliplatin) CT. In the CIK group, patients were given CIK cell infusion after FOLFOX4 CT. Treatment efficacy, adverse effects, and quality of life (QOL) were assessed. RESULTS During the first 2 years of follow-up, the recurrence rate in the CIK group (26.1%, 6 in 23 cases) was significantly lower than the control group (43.5%, 10 in 23). The survival time was significantly longer in the CIK group (41.9 months, 95% confidence interval [CI]: 38.2-45.7) than in the control group (33.8 months, 95% CI: 28.4-39.2). Although QOL was reduced in both treatment groups, adjuvant CIK cell transfusion significantly improved the QOL in patients with CRC. Toxicity was mild in patients with CIK treatment. CONCLUSIONS Immunotherapy with CIK cells may serve as an adjuvant treatment in patients with CRC after CT with prolonged survival of patients, limited side-effects, and improved QOL.
Collapse
Affiliation(s)
- Hao Peng
- Department of General Surgery, Beijing Meitan General Hospital , Beijing, China
| | - Meng Yao
- Department of General Surgery, Beijing Meitan General Hospital , Beijing, China
| | - Hongwei Fan
- Department of General Surgery, Beijing Meitan General Hospital , Beijing, China
| | - Liwei Song
- Department of General Surgery, Beijing Meitan General Hospital , Beijing, China
| | - Jinwen Sun
- Department of General Surgery, Beijing Meitan General Hospital , Beijing, China
| | - Zheng Zhou
- Department of General Surgery, Beijing Meitan General Hospital , Beijing, China
| | - Yunfeng Du
- Department of General Surgery, Beijing Meitan General Hospital , Beijing, China
| | - Keyu Lu
- Department of General Surgery, Beijing Meitan General Hospital , Beijing, China
| | - Tao Li
- Department of General Surgery, Beijing Meitan General Hospital , Beijing, China
| | - Aiguo Yin
- Department of General Surgery, Beijing Meitan General Hospital , Beijing, China
| | - Jianhua Xu
- Department of General Surgery, Beijing Meitan General Hospital , Beijing, China
| | - Shidong Wei
- Department of General Surgery, Beijing Meitan General Hospital , Beijing, China
| |
Collapse
|
47
|
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
|
48
|
Kong DS, Nam DH, Kang SH, Lee JW, Chang JH, Kim JH, Lim YJ, Koh YC, Chung YG, Kim JM, Kim CH. Phase III randomized trial of autologous cytokine-induced killer cell immunotherapy for newly diagnosed glioblastoma in Korea. Oncotarget 2018; 8:7003-7013. [PMID: 27690294 PMCID: PMC5351686 DOI: 10.18632/oncotarget.12273] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/23/2016] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Adoptive cell immunotherapy involves an ex vivo expansion of autologous cytokine-induced killer (CIK) cells before their reinfusion into the host. We evaluated the efficacy and safety of CIK cell immunotherapy with radiotherapy-temozolomide (TMZ) for the treatment of newly diagnosed glioblastomas. EXPERIMENTAL DESIGN In this multi-center, open-label, phase 3 study, we randomly assigned patients with newly diagnosed glioblastoma to receive CIK cell immunotherapy combined with standard TMZ chemoradiotherapy (CIK immunotherapy group) or standard TMZ chemoradiotherapy alone (control group). The efficacy endpoints were analyzed in the intention-to-treat set and in the per protocol set. RESULTS Between December 2008 and October 2012, a total of 180 patients were randomly assigned to the CIK immunotherapy (n = 91) or control group (n = 89). In the intention-to-treat analysis set, median PFS was 8.1 months (95% confidence interval (CI), 5.8 to 8.5 months) in the CIK immunotherapy group, as compared to 5.4 months (95% CI, 3.3 to 7.9 months) in the control group (one-sided log-rank, p = 0.0401). Overall survival did not differ significantly between two groups. Grade 3 or higher adverse events, health-related quality of life and performance status between two groups did not show a significant difference. CONCLUSIONS The addition of CIK cells immunotherapy to standard chemoradiotherapy with TMZ improved PFS. However, the CIK immunotherapy group did not show evidence of a beneficial effect on overall survival.
Collapse
Affiliation(s)
- Doo-Sik Kong
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Do-Hyun Nam
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Shin-Hyuk Kang
- Department of Neurosurgery, College of Medicine, Korea University, Seoul, Korea
| | - Jae Won Lee
- Department of Statistics, Korea University, Seoul, Korea
| | - Jong-Hee Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong-Hoon Kim
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Young-Jin Lim
- Department of Neurosurgery, Kyunghee University Hospital, Seoul, Korea
| | - Young-Cho Koh
- Department of Neurosurgery, Konkuk University Medical Center, Seoul, Korea
| | - Yong-Gu Chung
- Department of Neurosurgery, College of Medicine, Korea University, Seoul, Korea
| | - Jae-Min Kim
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Choong-Hyun Kim
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| |
Collapse
|
49
|
Chemotherapy plus dendritic cells co-cultured with cytokine-induced killer cells versus chemotherapy alone to treat advanced non-small-cell lung cancer: A meta-analysis. Oncotarget 2018; 7:86500-86510. [PMID: 27863436 PMCID: PMC5349930 DOI: 10.18632/oncotarget.13394] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 11/07/2016] [Indexed: 12/21/2022] Open
Abstract
This study was aimed to investigate the efficacy and safety of the combination treatment of dendritic cells co-cultured with cytokine-induced killer cells and chemotherapy for patients with advanced non-small-cell lung cancer (NSCLC). Literatures were searched from the Cochrane Library Central, PubMed, Web of Science and EMBASE. The primary endpoint of interest was overall survival (OS), and secondary endpoints were disease control rate (DCR) and progression free survival (PFS). Finally 7 trials published between January 2005 and March 2016 met inclusion criteria and totally 610 patients were enrolled. The combination group showed advance in DCR (RR = 1.31, 95% CI = 1.13-1.52, p = 0.0004), 1-year OS (RR = 1.18, 95% CI = 1.05-1.33, p = 0.007), and 2-year OS (RR = 1.37, 95% CI = 1.10-1.70, p = 0.005), with statistical significance. The proportions of CD3+ T cells (p = 0.002), NK cells (p = 0.02) and NKT cells (p = 0.001) were significantly higher in the peripheral blood of combination group, compared with those of the control group. Moreover, adverse reactions were obviously decreased in the combination group. However, no significant difference was identified in ORR and PFS between two groups (p > 0.05). In conclusion, the combination therapy was safe and applicable for patients with advanced NSCLC.
Collapse
|
50
|
Introna M, Correnti F. Innovative Clinical Perspectives for CIK Cells in Cancer Patients. Int J Mol Sci 2018; 19:ijms19020358. [PMID: 29370095 PMCID: PMC5855580 DOI: 10.3390/ijms19020358] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 01/19/2018] [Accepted: 01/22/2018] [Indexed: 12/18/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are T lymphocytes that have acquired, in vitro, following extensive manipulation by Interferon gamma (IFN-γ), OKT3 and Interleukin 2 (IL-2) addition, the expression of several Natural Killer (NK) cell-surface markers. CIK cells have a dual "nature", due to the presence of functional TCR as well as NK molecules, even if the antitumoral activity can be traced back only to the NK-like structures (DNAM-1, NKG2D, NKp30 and CD56). In addition to antineoplastic activity in vitro and in several in-vivo models, CIK cells show very limited, if any, GvHD toxicity as well as a strong intratumoral homing. For all such reasons, CIK cells have been proposed and tested in many clinical trials in cancer patients both in autologous and allogeneic combinations, up to haploidentical mismatching. Indeed, genetic modification of CIK cells as well as the possibility of combining them with specific monoclonal antibodies will further expand the possibility of their clinical utilization.
Collapse
Affiliation(s)
- Martino Introna
- USS Center of Cell Therapy "G. Lanzani", USC Ematologia, ASST Papa Giovanni XXIII Bergamo, 24124 Bergamo, Italy.
| | - Fabio Correnti
- USS Center of Cell Therapy "G. Lanzani", USC Ematologia, ASST Papa Giovanni XXIII Bergamo, 24124 Bergamo, Italy.
| |
Collapse
|