1
|
Jiang Y, Qiu J, Ye N, Xu Y. Current status of cytokine-induced killer cells and combination regimens in breast cancer. Front Immunol 2025; 16:1476644. [PMID: 39981243 PMCID: PMC11839775 DOI: 10.3389/fimmu.2025.1476644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/23/2025] [Indexed: 02/22/2025] Open
Abstract
Breast cancer remains a significant health challenge worldwide, with substantial efforts aimed at understanding its pathogenesis, biological characteristics, and clinical triggers. Recently, immunotherapy such as the cytokine-induced killer cells combined with other drug therapies has offered new hope for patients with advanced breast cancer. However, the specific pathogenesis of combination regimens involving cytokine-induced killer cells remains elusive. Besides, the combination of immunotherapy with cytokine-induced killer cells might represent a novel breakthrough. This review outlines the current status of cytokine-induced killer cell therapies and their combination strategies, especially the combination of chemotherapy with molecularly targeted treatments, for the management of breast cancer.
Collapse
Affiliation(s)
- Yuancong Jiang
- Department of Breast and Thyroid Surgery, Shaoxing People’s Hospital, Shaoxing, China
| | - Jie Qiu
- Department of Breast and Thyroid Surgery, Shaoxing People’s Hospital, Shaoxing, China
| | - Nanwei Ye
- Department of Medical Research Center, Shaoxing People’s Hospital, Shaoxing, China
| | - Yingchun Xu
- Department of Breast and Thyroid Surgery, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
2
|
Sun W, Hu S, Wang X. Advances and clinical applications of immune checkpoint inhibitors in hematological malignancies. Cancer Commun (Lond) 2024; 44:1071-1097. [PMID: 39073258 PMCID: PMC11492363 DOI: 10.1002/cac2.12587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 06/09/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
Immune checkpoints are differentially expressed on various immune cells to regulate immune responses in tumor microenvironment. Tumor cells can activate the immune checkpoint pathway to establish an immunosuppressive tumor microenvironment and inhibit the anti-tumor immune response, which may lead to tumor progression by evading immune surveillance. Interrupting co-inhibitory signaling pathways with immune checkpoint inhibitors (ICIs) could reinvigorate the anti-tumor immune response and promote immune-mediated eradication of tumor cells. As a milestone in tumor treatment, ICIs have been firstly used in solid tumors and subsequently expanded to hematological malignancies, which are in their infancy. Currently, immune checkpoints have been investigated as promising biomarkers and therapeutic targets in hematological malignancies, and novel immune checkpoints, such as signal regulatory protein α (SIRPα) and tumor necrosis factor-alpha-inducible protein 8-like 2 (TIPE2), are constantly being discovered. Numerous ICIs have received clinical approval for clinical application in the treatment of hematological malignancies, especially when used in combination with other strategies, including oncolytic viruses (OVs), neoantigen vaccines, bispecific antibodies (bsAb), bio-nanomaterials, tumor vaccines, and cytokine-induced killer (CIK) cells. Moreover, the proportion of individuals with hematological malignancies benefiting from ICIs remains lower than expected due to multiple mechanisms of drug resistance and immune-related adverse events (irAEs). Close monitoring and appropriate intervention are needed to mitigate irAEs while using ICIs. This review provided a comprehensive overview of immune checkpoints on different immune cells, the latest advances of ICIs and highlighted the clinical applications of immune checkpoints in hematological malignancies, including biomarkers, targets, combination of ICIs with other therapies, mechanisms of resistance to ICIs, and irAEs, which can provide novel insight into the future exploration of ICIs in tumor treatment.
Collapse
Affiliation(s)
- Wenyue Sun
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
| | - Shunfeng Hu
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
| | - Xin Wang
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
- Taishan Scholars Program of Shandong ProvinceJinanShandongP. R. China
- Branch of National Clinical Research Center for Hematologic DiseasesJinanShandongP. R. China
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuP. R. China
| |
Collapse
|
3
|
Ghanbari Sevari F, Mehdizadeh A, Abbasi K, Hejazian SS, Raisii M. Cytokine-induced killer cells: new insights for therapy of hematologic malignancies. Stem Cell Res Ther 2024; 15:254. [PMID: 39135188 PMCID: PMC11321231 DOI: 10.1186/s13287-024-03869-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Cytokine-induced killer (CIK) cells are a novel subgroup of immune effectors, classified as one of the modified T cell-mediated arms for immunotherapy. These cells exert MHC-unrestricted cytotoxicity against both hematological and solid malignancies with low incidence of treatment-related severe complications. This study reviews the application of CIK cells in treating cases with hematologic malignancies. MAIN BODY CIK cells consist of CD3+/CD56+ natural killer (NK) T cells, CD3-/CD56+ NK cells, and CD3+/CD56- cytotoxic T cells. In this regard, the CD3+/CD56+ NK T cells are the primary effectors. Compared with the previously reported antitumor immune cells, CIK cells are characterized by improved in vitro proliferation and amplification, enhanced migration and invasive capacity to tumor region, more significant antitumor activity, and a broader antitumor spectrum. CIK cells can also induce death in tumor cells via numerous pathways and mechanisms. Hence, CIKs-based therapy has been used in various clinical trials and has shown efficacy with a very low graft versus host disease (GVHD) against several cancers, such as hematologic malignancies, even in relapsing cases, or cases not responding to other therapies. Despite the high content of T cells, CIK cells induce low alloreactivity and, thus, pose a restricted threat of GVHD induction even in MHC-mismatched transplantation cases. Phase 1 and 2 clinical trials of CIK cell therapy have also highlighted satisfactory therapeutic advantages against hematologic cancers, indicating the safety of CIK cells even in haploidentical transplantation settings. CONCLUSION CIK cells have shown promising results in the treatment of hematologic malignancies, especially in combination with other antitumor strategies. However, the existing controversies in achieving desired clinical responses underscore the importance of future studies.
Collapse
Affiliation(s)
- Faezeh Ghanbari Sevari
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khadijeh Abbasi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyed Sina Hejazian
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mortaza Raisii
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Huang Z, Zou S, Liu Q, Qi W, Sharma A, Wang Y, Jin A, Schmidt-Wolf IGH, Lu P, Ai W, Liao F. Inferring the diagnostic potential of 18F-FDG-PET/CT in post-renal transplantation from a unique case harboring multiple rare complications. Front Med (Lausanne) 2024; 11:1353466. [PMID: 38371509 PMCID: PMC10869483 DOI: 10.3389/fmed.2024.1353466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 01/08/2024] [Indexed: 02/20/2024] Open
Abstract
Renal transplantation is undoubtedly an effective treatment for patients with end-stage renal disease, but it is certainly not a cure. Patients require lifelong immunosuppression to maintain optimal allograft function, and post-operative risk complications such as cancer in the transplant recipient cannot be ignored. Besides, infection is a silent complication that follows transplantation. Relatedly, herein, we present a report of a 40-year-old patient who underwent renal transplantation and promptly developed a diffuse large B-cell tumor in the liver and Aspergillus infection in the trachea. In addition, an inflammatory necrotizing granuloma was also observed in the muscles. Of importance, we also described the potential of 18F-FDG-PET/CT, which was instrumental in monitoring and evaluating these relevant post-operative complications in this rare case.
Collapse
Affiliation(s)
- Zizhen Huang
- Sterilization and Supply Center, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Shiwei Zou
- Department of Ultrasound Medicine, Dongxiang District Hospital of Traditional Chinese Medicine, Fuzhou, China
| | - Qian Liu
- Department of Pathology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Wanling Qi
- Department of Nuclear Medicine, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital of Bonn, Bonn, Germany
- Department of Stereotactic and Functional Neurosurgery, University Hospital of Bonn, Bonn, Germany
| | - Yulu Wang
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Aifang Jin
- Department of Nuclear Medicine, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Ingo G. H. Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital of Bonn, Bonn, Germany
| | - Ping Lu
- Department of Nuclear Medicine, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Wuping Ai
- Department of Orthopaedics, Dongxiang District Hospital of Traditional Chinese Medicine, Fuzhou, China
| | - Fengxiang Liao
- Department of Nuclear Medicine, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
5
|
Wang Q, Huang H, Liang P, Wang L, Zheng J, Zhang Y, Wang H. Development of PD-1 blockade peptide-cell conjugates to enhance cellular therapies for T-cell acute lymphoblastic leukemia. Med Oncol 2023; 41:14. [PMID: 38078948 DOI: 10.1007/s12032-023-02235-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023]
Abstract
Blockade of the interaction of the immune checkpoint receptor programmed cell death protein (PD)-1 and its ligand PD-L1 has been found to be a promising cancer treatment. Our previous studies identified that nABPD1 competed with PD-L1 to bind PD-1. The aim of this study was to evaluate the efficacy and safety of anti-tumor immunotherapy of ICIK cells conjugated with peptides in vivo and in vitro. Here, we synthesized the nABPD1 derivatives SBP1 and SBP2 and showed that their binding efficiency to PD-1-positive improving cytokine-induced killer (ICIK) cells was 98 and 82%, respectively. The cytotoxicity of ICIK cells to T-cell acute lymphoblastic leukemia (T-ALL) cells was increased by conjugating with SBP1 or SBP2, which was 2 times higher than that of ICIK cells alone. Furthermore, mice experiments showed that the fluorescence intensity of leukemia cells in T-ALL xenograft models was reduced by more than 95%, indicating that the peptides enhanced the therapeutic effect in vivo, while morphological evaluations showed that the peptides had no toxicity to important organs. Therefore, peptide-cell conjugates (PCCs) may be a novel method to improve the efficacy of cancer immunotherapy by blocking PD-1 in T-ALL patients.
Collapse
Affiliation(s)
- Quanxiao Wang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan West Road, Guangzhou, 510055, Guangdong, China
| | - Hongxing Huang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan West Road, Guangzhou, 510055, Guangdong, China
| | - Peisheng Liang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan West Road, Guangzhou, 510055, Guangdong, China
| | - Lili Wang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan West Road, Guangzhou, 510055, Guangdong, China
| | - Junheng Zheng
- Guangzhou Yidai Pharmaceutical Co., Ltd, Guangzhou, Guangdong, China
- Zhuhai Taisujian Biotechnology Co., Ltd, Zhuhai, Guangdong, China
- Cheerland Taisujian BioPharm. Co., Ltd, Shenzhen, Guangdong, China
| | - Yan Zhang
- Laboratory of Cancer and Stem Cell Biology, Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou Higher Education Mega Center, Guangzhou, China
| | - Hua Wang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan West Road, Guangzhou, 510055, Guangdong, China.
- Guangzhou Yidai Pharmaceutical Co., Ltd, Guangzhou, Guangdong, China.
- Zhuhai Taisujian Biotechnology Co., Ltd, Zhuhai, Guangdong, China.
- Cheerland Taisujian BioPharm. Co., Ltd, Shenzhen, Guangdong, China.
| |
Collapse
|
6
|
Wang Y, Sharma A, Ge F, Chen P, Yang Y, Liu H, Liu H, Zhao C, Mittal L, Asthana S, Schmidt-Wolf IGH. Non-oncology drug (meticrane) shows anti-cancer ability in synergy with epigenetic inhibitors and appears to be involved passively in targeting cancer cells. Front Oncol 2023; 13:1157366. [PMID: 37274234 PMCID: PMC10235775 DOI: 10.3389/fonc.2023.1157366] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/05/2023] [Indexed: 06/06/2023] Open
Abstract
Emerging evidence suggests that chemotherapeutic agents and targeted anticancer drugs have serious side effects on the healthy cells/tissues of the patient. To overcome this, the use of non-oncology drugs as potential cancer therapies has been gaining momentum. Herein, we investigated one non-oncology drug named meticrane (a thiazide diuretic used to treat essential hypertension), which has been reported to indescribably improve the therapeutic efficacy of anti-CTLA4 in mice with AB1 HA tumors. In our hypothesis-driven study, we tested anti-cancer potential meticrane in hematological malignance (leukemia and multiple myeloma) and liver cancer cell lines. Our analysis showed that: 1) Meticrane induced alteration in the cell viability and proliferation in leukemia cells (Jurkat and K562 cells) and liver cancer (SK-hep-1), however, no evidence of apoptosis was detectable. 2) Meticrane showed additive/synergistic effects with epigenetic inhibitors (DNMT1/5AC, HDACs/CUDC-101 and HDAC6/ACY1215). 3) A genome-wide transcriptional analysis showed that meticrane treatment induces changes in the expression of genes associated with non-cancer associated pathways. Of importance, differentially expressed genes showed favorable correlation with the survival-related genes in the cancer genome. 4) We also performed molecular docking analysis and found considerable binding affinity scores of meticrane against PD-L1, TIM-3, CD73, and HDACs. Additionally, we tested its suitability for immunotherapy against cancers, but meticrane showed no response to the cytotoxicity of cytokine-induced killer (CIK) cells. To our knowledge, our study is the first attempt to identify and experimentally confirm the anti-cancer potential of meticrane, being also the first to test the suitability of any non-oncology drug in CIK cell therapy. Beyond that, we have expressed some concerns confronted during testing meticrane that also apply to other non-oncology drugs when considered for future clinical or preclinical purposes. Taken together, meticrane is involved in some anticancer pathways that are passively targeting cancer cells and may be considered as compatible with epigenetic inhibitors.
Collapse
Affiliation(s)
- Yulu Wang
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Fangfang Ge
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| | - Peng Chen
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| | - Yu Yang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Hongjia Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Hongde Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Chunxia Zhao
- School of Nursing, Nanchang University, Nanchang, China
| | - Lovika Mittal
- Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Shailendra Asthana
- Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Ingo G. H. Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| |
Collapse
|
7
|
Li Y, Sharma A, Wu X, Weiher H, Skowasch D, Essler M, Schmidt-Wolf IGH. A Combination of Cytokine-Induced Killer Cells With PD-1 Blockade and ALK Inhibitor Showed Substantial Intrinsic Variability Across Non-Small Cell Lung Cancer Cell Lines. Front Oncol 2022; 12:713476. [PMID: 35646685 PMCID: PMC9130779 DOI: 10.3389/fonc.2022.713476] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 03/31/2022] [Indexed: 11/23/2022] Open
Abstract
Background Cancer heterogeneity poses a serious challenge concerning the toxicity and adverse effects of therapeutic inhibitors, especially when it comes to combinatorial therapies that involve multiple targeted inhibitors. In particular, in non-small cell lung cancer (NSCLC), a number of studies have reported synergistic effects of drug combinations in the preclinical models, while they were only partially successful in the clinical setup, suggesting those alternative clinical strategies (with genetic background and immune response) should be considered. Herein, we investigated the antitumor effect of cytokine-induced killer (CIK) cells in combination with ALK and PD-1 inhibitors in vitro on genetically variable NSCLC cell lines. Methods We co-cultured the three genetically different NSCLC cell lines NCI-H2228 (EML4-ALK), A549 (KRAS mutation), and HCC-78 (ROS1 rearrangement) with and without nivolumab (PD-1 inhibitor) and crizotinib (ALK inhibitor). Additionally, we profiled the variability of surface expression multiple immune checkpoints, the concentration of absolute dead cells, intracellular granzyme B on CIK cells using flow cytometry as well as RT-qPCR. ELISA and Western blot were performed to verify the activation of CIK cells. Results Our analysis showed that (a) nivolumab significantly weakened PD-1 surface expression on CIK cells without impacting other immune checkpoints or PD-1 mRNA expression, (b) this combination strategy showed an effective response on cell viability, IFN-γ production, and intracellular release of granzyme B in CD3+ CD56+ CIK cells, but solely in NCI-H2228, (c) the intrinsic expression of Fas ligand (FasL) as a T-cell activation marker in CIK cells was upregulated by this additive effect, and (d) nivolumab induced Foxp3 expression in CD4+CD25+ subpopulation of CIK cells significantly increased. Taken together, we could show that CIK cells in combination with crizotinib and nivolumab can enhance the anti-tumor immune response through FasL activation, leading to increased IFN-γ and granzyme B, but only in NCI-H2228 cells with EML4-ALK rearrangement. Therefore, we hypothesize that CIK therapy may be a potential alternative in NSCLC patients harboring EML4-ALK rearrangement, in addition, we support the idea that combination therapies offer significant potential when they are optimized on a patient-by-patient basis.
Collapse
Affiliation(s)
- Yutao Li
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, Bonn, Germany
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, Bonn, Germany.,Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Xiaolong Wu
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, Bonn, Germany
| | - Hans Weiher
- Department of Applied Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Dirk Skowasch
- Department of Internal Medicine II, Cardiology, Pneumology and Angiology, University Hospital Bonn, Bonn, Germany
| | - Markus Essler
- Department of Nuclear Medicine, University Hospital Bonn, Bonn, Germany
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
8
|
Liu S, Meng Y, Liu L, Lv Y, Yu W, Liu T, Wang L, Mu D, Zhou Q, Liu M, Ren Y, Zhang D, Li B, Sun Q, Ren X. CD4 + T cells are required to improve the efficacy of CIK therapy in non-small cell lung cancer. Cell Death Dis 2022; 13:441. [PMID: 35523765 PMCID: PMC9076680 DOI: 10.1038/s41419-022-04882-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 12/13/2022]
Abstract
As a widely studied adoptive treatment method, CIK (cytokine-induced killer cells) treatment has shown clinical benefits in many clinical trials on non-small cell lung cancer. As a heterogeneous cell population, however, CIK cells have a strong instability and individual differences in their efficacies, which are collaboratively regulated by the tumor microenvironment and CIK subpopulations. Among them, CD4+ T cells belong to a crucial subgroup of the CIK cell population, and their influence on CIK therapy is still unclear. Herein, we show how CD4+ T cells positively regulate the functions of CD3+CD56+ T and CD3+CD8+ T cells. During this process, we found that Th1/Th17 CD4+ subgroups can induce the phosphorylation of the AKT pathway by secreting IL-17A, and upregulate the expression of T-bet/Eomes transcription factors, thereby restoring the function of CD8+/CD3+CD56+ T cells and reversing the exhaustion of PD-1+Tim-3+ T cells. These findings will provide guidance for the clinical screening of suitable populations for CIK treatment and formulation of strategies for CIK therapy plus immune checkpoint treatment. Based on these findings, we are conducting an open-label phase II study (NCT04836728) is to evaluate the effects of autologous CIKs in combination with PD-1 inhibitor in the first-line treatment of IV NSCLC, and hope to observe patients' benefits in this clinical trial.
Collapse
Affiliation(s)
- Shaochuan Liu
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuan Meng
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Liang Liu
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yingge Lv
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wenwen Yu
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ting Liu
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Limei Wang
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Di Mu
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qiuru Zhou
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Min Liu
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yulin Ren
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Dong Zhang
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Baihui Li
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qian Sun
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiubao Ren
- grid.411918.40000 0004 1798 6427Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin, China ,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
9
|
Garofano F, Sharma A, Abken H, Gonzalez-Carmona MA, Schmidt-Wolf IGH. A Low Dose of Pure Cannabidiol Is Sufficient to Stimulate the Cytotoxic Function of CIK Cells without Exerting the Downstream Mediators in Pancreatic Cancer Cells. Int J Mol Sci 2022; 23:3783. [PMID: 35409142 PMCID: PMC8998663 DOI: 10.3390/ijms23073783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 12/18/2022] Open
Abstract
Despite numerous studies conducted over the past decade, the exact role of the cannabinoid system in cancer development remains unclear. Though research has focused on two cannabinoid receptors (CB1, CB2) activated by most cannabinoids, CB2 holds greater attention due to its expression in cells of the immune system. In particular, cytokine-induced killer cells (CIKs), which are pivotal cytotoxic immunological effector cells, express a high-level of CB2 receptors. Herein, we sought to investigate whether inducing CIK cells with cannabidiol can enhance their cytotoxicity and if there are any possible counter effects in its downstream cascade of phosphorylated p38 and CREB using a pancreatic ductal adenocarcinoma cell line (PANC-1). Our results showed that IL-2 modulates primarily the expression of the CB2 receptor on CIK cells used during ex vivo CIK expansion. The autophagosomal-associated scaffold protein p62 was found to co-localize with CB2 receptors in CIK cells and the PANC-1 cell line. CIK cells showed a low level of intracellular phospho-p38 and, when stimulated with cannabidiol (CBD), a donor specific variability in phospho-CREB. CBD significantly decreases the viability of PANC-1 cells presumably by increasing the cytotoxicity of CIK cells. Taken together, in our preclinical in vitro study, we propose that a low effective dose of CBD is sufficient to stimulate the cytotoxic function of CIK without exerting any associated mediator. Thus, the combinatorial approach of non-psychoactive CBD and CIK cells appears to be safe and can be considered for a clinical perspective in pancreatic cancer.
Collapse
Affiliation(s)
- Francesca Garofano
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, 53127 Bonn, Germany; (F.G.); (A.S.)
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, 53127 Bonn, Germany; (F.G.); (A.S.)
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Hinrich Abken
- RCI Regensburg Center for Interventional Immunology, Department Genetic Immunotherapy, University Hospital Regensburg, 93053 Regensburg, Germany;
| | | | - Ingo G. H. Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, 53127 Bonn, Germany; (F.G.); (A.S.)
| |
Collapse
|
10
|
Zhao L, Li T, Song Y, Yang Y, Ma B, Zhang Y, Shang Y, Xu B, Guo J, Qin P, Han L, Fu X, Lin H, Liu L, Ren X, Wang Z, Gao Q. High Complete Response Rate in Patients With Metastatic Renal Cell Carcinoma Receiving Autologous Cytokine-Induced Killer Cell Therapy Plus Anti-Programmed Death-1 Agent: A Single-Center Study. Front Immunol 2022; 12:779248. [PMID: 35058923 PMCID: PMC8764153 DOI: 10.3389/fimmu.2021.779248] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/08/2021] [Indexed: 12/24/2022] Open
Abstract
Background and Objective The results of the CheckMate 025 trial established the status of nivolumab in the second-line treatment of metastatic renal cell carcinoma (mRCC), with an objective response rate (ORR) of 25% and a complete response (CR) rate of 1%. Thus, the efficacy of anti-programmed death (PD)-1 antibodies in the second-line treatment of mRCC requires improvement. The purpose of this study was to explore the clinical efficacy and safety of anti-PD-1 agents combined with cytokine-induced killer (CIK) cell therapy for refractory mRCC. Patients and Methods Patients with mRCC refractory to previous targeted therapy were included in this study. All patients received anti-PD-1 plus CIK cell therapy. The ORR and CR rate, progression-free survival (PFS), overall survival (OS), and safety were assessed. Results CR was observed in seven of the 29 patients, and partial response was observed in five patients. The ORR was 41.4% and the median PFS was 15.0 months. Up to the last follow-up, 15 patients died with an average survival time of 37 months. Among the patients who achieved a CR, one experienced cerebellar metastasis 18.8 months after discontinuation, but achieved CR again after localized gamma knife and 1-month axitinib treatment. This regimen was tolerated well and there was no treatment-related death. Conclusions Combination therapy with anti-PD-1 and CIK cell therapy is safe and effective in patients with mRCC refractory to previous targeted therapy. The high CR rate and long disease-free survival even after long-term discontinued therapy suggest that this combination treatment may represent a potential curative regimen for this type of malignancy.
Collapse
Affiliation(s)
- Lingdi Zhao
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Tiepeng Li
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yongping Song
- Hematology Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yonghao Yang
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Baozhen Ma
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yong Zhang
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yiman Shang
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Benling Xu
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jindong Guo
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Peng Qin
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Lu Han
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xiaomin Fu
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Hongwei Lin
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Liang Liu
- Biotherapy Department, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiubao Ren
- Biotherapy Department, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zibing Wang
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Quanli Gao
- Immunotherapy Department, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
11
|
Li Y, Sharma A, Maciaczyk J, Schmidt-Wolf IGH. Recent Development in NKT-Based Immunotherapy of Glioblastoma: From Bench to Bedside. Int J Mol Sci 2022; 23:ijms23031311. [PMID: 35163235 PMCID: PMC8835986 DOI: 10.3390/ijms23031311] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/15/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive and dismal disease with a median overall survival of around 15 months and a 5-year survival rate of 7.2%. Owing to genetic mutations, drug resistance, disruption to the blood–brain barrier (BBB)/blood–brain tumor barrier (BBTB), and the complexity of the immunosuppressive environment, the therapeutic approaches to GBM represent still major challenges. Conventional therapies, including surgery, radiotherapy, and standard chemotherapy with temozolomide, have not resulted in satisfactory improvements in the overall survival of GBM patients. Among cancer immunotherapeutic approaches, we propose that adjuvant NKT immunotherapy with invariant NKT (iNKT) and cytokine-induced killer (CIK) cells may improve the clinical scenario of this devastating disease. Considering this, herein, we discuss the current strategies of NKT therapy for GBM based primarily on in vitro/in vivo experiments, clinical trials, and the combinatorial approaches with future therapeutic potential.
Collapse
Affiliation(s)
- Yutao Li
- Center for Integrated Oncology (CIO), Department of Integrated Oncology, University Hospital Bonn, 53127 Bonn, Germany;
| | - Amit Sharma
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany; (A.S.); (J.M.)
| | - Jarek Maciaczyk
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany; (A.S.); (J.M.)
- Department of Surgical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Ingo G. H. Schmidt-Wolf
- Center for Integrated Oncology (CIO), Department of Integrated Oncology, University Hospital Bonn, 53127 Bonn, Germany;
- Correspondence: ; Tel.: +49-228-2871-7050
| |
Collapse
|
12
|
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: 1.0] [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
|