1
|
Li C, Xu D, Lu L, Peng S, Zhao H, Zeng C, Hu L, Guo X, Liu L, Huo F, Rong X, Geng Z, Lin P, Zhou X, Wang X, Hobeika A, Morse MA, Lyerly HK, Ren J. Clinical impact of concurrent autologous adoptive T cells immunotherapy in active COVID-19 infected cancer patients for chemotherapy. Infect Agent Cancer 2025; 20:23. [PMID: 40205403 PMCID: PMC11983847 DOI: 10.1186/s13027-025-00654-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND The concurrent presence of COVID-19 infection in advanced cancer patients has increased the mortality since the compromised immunity was inevitably worsen. The role and clinical impact of autologous adoptive T cell immunotherapy (ACT) designed for anti-cancer treatment were not known in such circumstances. The safety and potential immune reconstitution of concurrent ACT in advanced cancer patients with active COVID-19 infection have yet unknown as well. The effect of infused ACT on the symptom severity manifestation should be summarized. METHODS In this respectively clinical observation study, patients were non-randomized enrolled from the two centers according to the regular therapeutic plans including stage IV cancer patients for scheduled ACT, chemotherapy, cancer patients with symptomatic COVID-19 but without ACT, neither cancer or non-ACT but symptomatic cases of COVID-19 infection. We have incorporated the age-adjusted Charlson comorbidity index (aCCI) for each patient to compare the prognosis of the three groups. All patients were planned for the scheduled standard anti-cancer therapeutic considerations, chemotherapy plus ACT as planned as well as the supportive care.The clinical efficacy and impact of ACT on cancer patients within the 3 months from the peripheral blood apheresis, dendritic cell (DC) and cytokine induced killer T cell (CIK-T ) infusion and subsequent co-existence of COVID-19 infection were recorded as the primary objective. During the same period, the cancer cases without ACT and others were collected to compare the occurrence of both severe and death rate respectively. RESULTS There were 123 patients (35 of ACT, 23 of non-ACT, 65 of non-cancer) with similar aCCI. There were similar cohort-level COVID-19 in-hospital case fatality rates consistent with previously reported data for non-cancer (26.2%, 17/65) and non-ACT cancer (52.2%, 12/23) among those admitted severe cases after the adjustment.There were little overlapped adverse reactions during the ACT therapeutic period even in the presence of active COVID-19 infection. No death case was occurred (0/35) when those exposed to ACT regimen. Cancer patients receiving ACT had a shorter mean time to alleviation of symptoms compared with non-ACT and non-cancer (4.46 versus 16.88 and 17.90 days respectively) as well as the lowered severity incidence of symptoms (P = 0.0010). The infused ACT has not significant impact on peripheral blood count whereas the amount of CD3-CD16+CD56+ NK cells increased (P = 0.0017). The quantity of infused ACT was favorable for augmentation of possibility of severe to mild symptom shift. CONCLUSIONS These data demonstrate the clinical safety profiles while ACT infusions with active COVID-19 infection.The intervention of ACT for cancer patients could generate the benefit for symptom alleviation with improved recovery time. The concurrent ACT for advanced cancer patients during such infectious pandemic might simultaneously leverage and reduce the risk of immune compromised situation for subsequent chemotherapy complications.
Collapse
Affiliation(s)
- Congcong Li
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China
| | - Dazhao Xu
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China
| | - Linyao Lu
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China
| | - Shu Peng
- Department of Medical Oncology, Fudan University Huadong Medical Center, Shanghai, China
| | - Haiyang Zhao
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China
| | - Chuxiong Zeng
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China
| | - Lina Hu
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China
| | - Xianzhi Guo
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China
| | - Li Liu
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China
| | - Feifei Huo
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China
| | - Xiumei Rong
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China
| | - Zhenying Geng
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China
| | - Ping Lin
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China
| | - Xinna Zhou
- Center of Clinical Trials & Management, Beijing Shijitan Hospital, Capital Medical University Cancer Center, Beijing, 10038, China
| | - Xiaoli Wang
- Department of Medical Oncology, Beijing Shijitan Hospital, Capital Medical University Cancer Center, Beijing, 10038, China
| | - Amy Hobeika
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Michael A Morse
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | | | - Jun Ren
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China.
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China.
- Department of Medical Oncology, Fudan University Huadong Medical Center, Shanghai, China.
- Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
2
|
Jiang W, Wang Z, Luo Q, Dai Z, Zhu J, Tao X, Xie Y, Du Y, Jiang L, Chu X, Fu G, Lei Z. Combined immunotherapy with dendritic cells and cytokine-induced killer cells for solid tumors: a systematic review and meta-analysis of randomized controlled trials. J Transl Med 2024; 22:1122. [PMID: 39707416 DOI: 10.1186/s12967-024-05940-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: 10/06/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Immunotherapy utilizing dendritic cells (DCs) and cytokine-induced killer (CIK) cells is a promising treatment approach for solid tumors. This systematic review and meta-analysis aimed to evaluate the efficacy and safety of DC-CIK immunotherapy by assessing overall survival, progression-free survival, overall response rate, disease control rate, and adverse events in relevant randomized controlled trials. The results of this analysis will contribute to optimizing treatment strategies and improving cancer immunotherapy outcomes. METHOD This systematic review and meta-analysis adhered to PRISMA guidelines. A comprehensive search was conducted on multiple databases for RCTs studying the combination of DC-CIK immunotherapy for solid tumors. Inclusion criteria were RCTs comparing DC-CIK immunotherapy with control therapy and reporting OS, PFS, ORR, or DCR. Two authors independently performed study selection and data extraction, with disagreements resolved through consensus or consultation with a third reviewer. Extracted data included study characteristics, participant information, interventions, outcomes, and quality assessment. Statistical analysis was performed using Review Manager and Stata software. Heterogeneity was assessed using chi-square and I-squared statistics. Sensitivity analysis and assessment of publication bias were planned. RESULTS A total of 1013 records were initially retrieved, and after a thorough screening process, 13 randomized controlled trials (RCTs) were included in the meta-analysis. These studies involved a total of 1443 patients, with 730 receiving DC-CIK immunotherapy and 713 in the control groups. The included studies covered various cancer types, with the majority conducted in mainland China. The meta-analysis showed that DC-CIK immunotherapy was associated with improved overall survival (OS) and progression-free survival (PFS) compared to control therapy. Furthermore, DC-CIK immunotherapy demonstrated higher overall response rate (ORR) and disease control rate (DCR) compared to non-DC-CIK therapy. Adverse events were reported in both groups, with fever being more common in the DC-CIK immunotherapy group and bone marrow suppression and gastrointestinal reactions more common in the control group. Sensitivity analyses confirmed the stability of the results, while publication bias was observed for PFS and fever. CONCLUSIONS DC-CIK immunotherapy shows promising efficacy and safety for solid tumors, improving survival rates and response rates. Further research is needed to optimize treatment regimens and identify predictive factors.
Collapse
Affiliation(s)
- Wendi Jiang
- Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhongda Wang
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qinghuizi Luo
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhe Dai
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jialong Zhu
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiaoyue Tao
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yiyang Xie
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuanyang Du
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Longwei Jiang
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Nanjing Advanced Institute for Life Sciences, School of Life Sciences, Nanjing University, Nanjing, China.
| | - Xiaoyuan Chu
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| | - Gongbo Fu
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| | - Zengjie Lei
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| |
Collapse
|
3
|
Mao W, Yoo HS. Inorganic Nanoparticle Functionalization Strategies in Immunotherapeutic Applications. Biomater Res 2024; 28:0086. [PMID: 39323561 PMCID: PMC11423863 DOI: 10.34133/bmr.0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/20/2024] [Accepted: 09/05/2024] [Indexed: 09/27/2024] Open
Abstract
Nanotechnology has been increasingly utilized in anticancer treatment owing to its ability of engineering functional nanocarriers that enhance therapeutic effectiveness while minimizing adverse effects. Inorganic nanoparticles (INPs) are prevalent nanocarriers to be customized for a wide range of anticancer applications, including theranostics, imaging, targeted drug delivery, and therapeutics, because they are advantageous for their superior biocompatibility, unique optical properties, and capacity of being modified via versatile surface functionalization strategies. In the past decades, the high adaptation of INPs in this emerging immunotherapeutic field makes them good carrier options for tumor immunotherapy and combination immunotherapy. Tumor immunotherapy requires targeted delivery of immunomodulating therapeutics to tumor locations or immunological organs to provoke immune cells and induce tumor-specific immune response while regulating immune homeostasis, particularly switching the tumor immunosuppressive microenvironment. This review explores various INP designs and formulations, and their employment in tumor immunotherapy and combination immunotherapy. We also introduce detailed demonstrations of utilizing surface engineering tactics to create multifunctional INPs. The generated INPs demonstrate the abilities of stimulating and enhancing the immune response, specific targeting, and regulating cancer cells, immune cells, and their resident microenvironment, sometimes along with imaging and tracking capabilities, implying their potential in multitasking immunotherapy. Furthermore, we discuss the promises of INP-based combination immunotherapy in tumor treatments.
Collapse
Affiliation(s)
- Wei Mao
- Department of Biomedical Materials Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hyuk Sang Yoo
- Department of Biomedical Materials Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea
- Kangwon Radiation Convergence Research Center, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
4
|
Zhao Y, Wang S, Lv S, Liu X, Li W, Song Y, Rong D, Zheng P, Huang H, Zheng H. Combined oral low-dose cyclophosphamide endocrine therapy may improve clinical response among patients with metastatic breast cancer via Tregs in TLSs. Sci Rep 2024; 14:13432. [PMID: 38862586 PMCID: PMC11166640 DOI: 10.1038/s41598-024-64042-3] [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: 11/27/2023] [Accepted: 06/04/2024] [Indexed: 06/13/2024] Open
Abstract
Despite limited research on refractory and/or endocrine therapy failure in elderly metastatic breast cancer (MBC) patients, a prior study showed that low-dose oral cyclophosphamide (CY) can improve the overall survival rate of MBC patients, possibly through the immunoregulation of regulatory T cells (Tregs). We preliminarily investigated the combination of endocrine therapy (ET) with oral low-dose CY as salvage therapy in elderly patients via peripheral blood regulatory T-cell analyses. In addition, we evaluated the associations of tumor tertiary lymphoid structures (TLSs) with therapeutic outcomes. HR+/HER2- advanced breast cancer patients who received low-dose CY combined with ET or ET only from April 2015 to August 2021 were enrolled in this retrospective study. The primary outcome was the clinical control rate (CCR), and the secondary outcome was progression-free survival (PFS). Circulating T lymphocyte subpopulations represented by Tregs were monitored during treatment by flow cytometry methods. TLSs wereconfirmed by hematoxylin-eosin staining of pretreatment specimens, and CD3, CD4, and Foxp3 were detected using Opal multicolor immunofluorescence. A total of 85 patients who received CY + ET and 50 patients who received ET only were enrolled, the percentage of patients who received CCR was 73% (62/85) vs. 70% (45/50), and the objective response rate (ORR) was 28% (24/85) vs. 24% (12/50). No deaths occurred during the study period. The mean PFS time was 13 vs. 11 months (P = 0.03). In the CY + ET group, decreases in CD4+/CD25+/Foxp3+ T cells (P < 0.001) were favorable for both clinical control and prolonged PFS (P < 0.001). Compared with patients without TLSs, those with TLSs were more likely to have better clinical control and PFS (mean time = 6 months), and a greater number of Treg cells during TLS pretreatment correlated with longer PFS (P = 0.043). Oral low-dose CY combined with standard ET exerts immunological effects by decreasing Treg levels to achieve improved clinical responses. Moreover, patients with TLSs might benefit more from such therapy than those without TLSs, and a high Treg cell count in TLSs before treatment predicts better therapeutic efficacy.
Collapse
Affiliation(s)
- Yuze Zhao
- Department of Medical Oncology, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, 10 Tieyi Rd, Beijing, 100038, China
| | - Shuo Wang
- Department of Medical Oncology, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, 10 Tieyi Rd, Beijing, 100038, China
| | - Shuzhen Lv
- Breast Department, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xiaojun Liu
- Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Weiping Li
- Department of Pharmacology, Shanxi Medical University Fenyang College, Fenyang, 032200, China
| | - Yuguang Song
- Department of Medical Oncology, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, 10 Tieyi Rd, Beijing, 100038, China
| | - Dongwen Rong
- Department of Medical Oncology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Peiming Zheng
- Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Hongyan Huang
- Department of Medical Oncology, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, 10 Tieyi Rd, Beijing, 100038, China.
| | - Huixia Zheng
- Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
5
|
Kumari K, Singh A, Chaudhary A, Singh RK, Shanker A, Kumar V, Haque R. Neoantigen Identification and Dendritic Cell-Based Vaccines for Lung Cancer Immunotherapy. Vaccines (Basel) 2024; 12:498. [PMID: 38793749 PMCID: PMC11125796 DOI: 10.3390/vaccines12050498] [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: 03/31/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Immunotherapies can treat many cancers, including difficult-to-treat cases such as lung cancer. Due to its tolerability, long-lasting therapeutic responses, and efficacy in a wide spectrum of patients, immunotherapy can also help to treat lung cancer, which has few treatment choices. Tumor-specific antigens (TSAs) for cancer vaccinations and T-cell therapies are difficult to discover. Neoantigens (NeoAgs) from genetic mutations, irregular RNA splicing, protein changes, or viral genetic sequences in tumor cells provide a solution. NeoAgs, unlike TSAs, are non-self and can cause an immunological response. Next-generation sequencing (NGS) and bioinformatics can swiftly detect and forecast tumor-specific NeoAgs. Highly immunogenic NeoAgs provide personalized or generalized cancer immunotherapies. Dendritic cells (DCs), which originate and regulate T-cell responses, are widely studied potential immunotherapeutic therapies for lung cancer and other cancers. DC vaccines are stable, reliable, and safe in clinical trials. The purpose of this article is to evaluate the current status, limitations, and prospective clinical applications of DC vaccines, as well as the identification and selection of major histocompatibility complex (MHC) class I and II genes for NeoAgs. Our goal is to explain DC biology and activate DC manipulation to help researchers create extremely potent cancer vaccines for patients.
Collapse
Affiliation(s)
- Komal Kumari
- Department of Biotechnology, Central University of South Bihar, Gaya 824236, Bihar, India; (K.K.); (A.C.)
| | - Amarnath Singh
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA;
| | - Archana Chaudhary
- Department of Biotechnology, Central University of South Bihar, Gaya 824236, Bihar, India; (K.K.); (A.C.)
| | - Rakesh Kumar Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India;
| | - Asheesh Shanker
- Department of Bioinformatics, Central University of South Bihar, Gaya 824236, Bihar, India
| | - Vinay Kumar
- Heart and Vascular Institute, Pennsylvania State University, Hershey Medical Center, Hershey, PA 17033, USA;
| | - Rizwanul Haque
- Department of Biotechnology, Central University of South Bihar, Gaya 824236, Bihar, India; (K.K.); (A.C.)
| |
Collapse
|
6
|
Zhong S, Zhang Y, Lu X, Meftahpour V. The Therapeutic Potential of Cytokine-Induced Killer in Patients with Cancer. J Interferon Cytokine Res 2024; 44:99-110. [PMID: 38488758 DOI: 10.1089/jir.2023.0180] [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] [Indexed: 03/19/2024] Open
Abstract
Despite the promising results of immunotherapy, further experiments need to be considered because of several factors ranging from physical barriers to off-tumor adverse effects. It is surprising that adoptive cellular immunotherapy, particularly dendritic cell and cytokine-induced killer (DC-CIK) therapy, is far less emphasized in the treatment of cancer diseases. DC-CIK therapy in cancer patients presents auspicious results with low or no side effects, which should not be overlooked. More interestingly, almost all DC-CIK clinical trials are ongoing in China that highlight the limitations of therapeutic strategies and require large-scale research. To date, it is advisable to consider combination therapy with chemotherapy since it has shown promising outcomes with higher efficacy. In this article, the efficacy of DC-CIK therapy in patients with cancer is summarized by underscoring the lack of experiments on soft cancers on an unprecedented scale. In brief, DC-CIK therapy is a safe and effective therapeutic agent for malignant and nonmalignant diseases that enhances short-term and long-term effects.
Collapse
Affiliation(s)
- Sixun Zhong
- Hai'an People's Hospital, Department of Oncology, Nantong City, Jiangsu Province, China
| | - Yan Zhang
- Hai'an People's Hospital, Department of Oncology, Nantong City, Jiangsu Province, China
| | - Xiaomin Lu
- Hai'an People's Hospital, Department of Oncology, Nantong City, Jiangsu Province, China
| | - Vafa Meftahpour
- Medical Immunology, Cellular and Molecular Research Center, Medical Sciences Department, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
7
|
Wang S, Mu X, Wang X, Chen L, Lu C, Song L. Peripheral Blood CD8 + CD28 + T Cells as an Independent Predictor of Treatment Response and Survival After Concurrent Chemoradiotherapy in Pediatric High-Grade Glioma Patients. Clin Med Insights Oncol 2024; 18:11795549241227421. [PMID: 38322666 PMCID: PMC10845990 DOI: 10.1177/11795549241227421] [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: 05/11/2023] [Accepted: 01/01/2024] [Indexed: 02/08/2024] Open
Abstract
Backgroud The tumor immune microenvironment influences the efficiency of concurrent chemoradiotherapy (CCRT) in high-grade glioma (HGG). This study investigated peripheral blood T lymphocyte subsets as clinical indicators of therapeutic response and prognosis in pediatric high-grade glioma (pHGG). Methods This retrospective study included 77 patients with postoperative pHGG who were treated concurrently with temozolomide and external beam radiotherapy between January 1, 2012, and December 31, 2018. The median follow-up was 26 (range: 5-106) months. Peripheral venous blood samples were collected before and after CCRT. The proportions of peripheral blood T lymphocytes and their association with treatment outcome and survival were determined. Results Sixty-four (83.1%) patients achieved complete remission, partial remission, and stable disease, and 13 (16.9%) patients had progressive disease. Higher CD3+ T cell, CD4+ T cell, and CD8+ CD28+ T cell ratios were predictive of better response, while a higher CD8+ CD28- T cell ratio was predictive of poorer response. Binary logistic regression analysis showed that the CD8+ CD28+ T cell ratio was a significant independent predictor of CCRT response (odds ratio [OR] = 53.521, 95% confidence interval [CI] = 4.294-667.119, P = .002). Univariate and multivariate analysis of prognostic factors associated with survival showed that the CD8+ CD28+ T lymphocyte ratio was a significant independent predictor of progression-free survival (hazard ratio [HR] = 1.80, 95% CI = 1.06-3.08, P = .03), but none of the subsets were significantly associated with overall survival. Conclusion Peripheral blood T lymphocytes have potential as predictors of CCRT response and prognosis in pHGG.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Medical Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaofeng Mu
- Department of Radiotherapy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Wang
- Department of Medical Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Li Chen
- Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Changyu Lu
- Department of Neurosurgery, Peking University International Hospital, Beijing, China
| | - Linan Song
- Department of Radiotherapy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Xu J, Liu C, Wu X, Ma J. Current immune therapeutic strategies in advanced or metastatic non-small cell lung cancer. Chin Med J (Engl) 2023; 136:1765-1782. [PMID: 37257112 PMCID: PMC10405997 DOI: 10.1097/cm9.0000000000002536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Indexed: 06/02/2023] Open
Abstract
ABSTRACT Immune escape mechanisms in non-small cell lung cancer (NSCLC) can disrupt every step of the anti-cancer immune response. In recent years, an increased understanding of the specific mechanisms fueling immune escape has allowed for the development of numerous immunotherapeutic treatments that have been introduced into the clinical practice. The advent of immunotherapy has dramatically changed the current treatment landscape of advanced or metastatic NSCLC because of its durable efficacy and manageable toxicity. In this review, we will first present a brief overview of recent evidence on immune escape mechanisms in NSCLC. We will then discuss the current promising immunotherapeutic strategies in advanced or metastatic NSCLC tumors.
Collapse
Affiliation(s)
- Jing Xu
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Caixia Liu
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiaonan Wu
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jie Ma
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
9
|
Ye W, Li M, Luo K. Therapies Targeting Immune Cells in Tumor Microenvironment for Non-Small Cell Lung Cancer. Pharmaceutics 2023; 15:1788. [PMID: 37513975 PMCID: PMC10384189 DOI: 10.3390/pharmaceutics15071788] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/02/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
The tumor microenvironment (TME) plays critical roles in immune modulation and tumor malignancies in the process of cancer development. Immune cells constitute a significant component of the TME and influence the migration and metastasis of tumor cells. Recently, a number of therapeutic approaches targeting immune cells have proven promising and have already been used to treat different types of cancer. In particular, PD-1 and PD-L1 inhibitors have been used in the first-line setting in non-small cell lung cancer (NSCLC) with PD-L1 expression ≥1%, as approved by the FDA. In this review, we provide an introduction to the immune cells in the TME and their efficacies, and then we discuss current immunotherapies in NSCLC and scientific research progress in this field.
Collapse
Affiliation(s)
- Wei Ye
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510091, China
| | - Meiye Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510091, China
| | - Kewang Luo
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510091, China
- People's Hospital of Longhua, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen 518109, China
| |
Collapse
|
10
|
Huang J, Zhao X, Zhang Z, Yang S, Chen X, Shen C, Wang L, Qi Y, Zhang Y. Adjuvant cytokine-induced killer cell immunotherapy improves long-term survival in patients with stage I-II non-small cell lung cancer after curative surgery. Cytotherapy 2023; 25:202-209. [PMID: 36379882 DOI: 10.1016/j.jcyt.2022.10.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Non-small cell lung cancer (NSCLC) remains the most common cancer worldwide, with an annual incidence of around 1.3 million. Surgery represents the standard treatment in early-stage NSCLC when feasible. However, because of cancer recurrence, only approximately 53% of patients with stage I and II NSCLC survive 5 years after radical surgery. The authors performed a retrospective study to investigate the impact of cytokine-induced killer (CIK) cell immunotherapy on the long-term survival of patients with stage I-II NSCLC after curative resection. METHODS Fifty-seven patients with NSCLC were included in the study, with 41 and 16 in the control and CIK groups, respectively. Clinical characteristics were compared using a t-test and χ2 test. Survival analysis of patients with NSCLC was performed using the Kaplan-Meier method. The phenotypes and anti-tumor functions of CIK cells were evaluated by flow cytometry. RESULTS Patients in the CIK group exhibited significantly longer overall survival (OS) and better disease-free survival (DFS) than those in the control group. Subgroup analysis indicated that patients with a higher risk of recurrence benefited more from CIK treatment and attained longer OS and DFS compared with those in the control group. No severe adverse events related to CIK treatment occurred. CIK cells contained a higher proportion of CD3+CD56+ natural killer (NK) T cells and CD3+ and CD8+ T cells and a lower proportion of CD3-CD56+ NK cells compared with peripheral blood mononuclear cells. CIK cells exhibited potent tumor-killing ability, with longer contact times with tumor cells and a greater number of cells exposed to tumor cells. CONCLUSIONS The authors' data suggest that adjuvant CIK cell therapy is a safe and effective therapeutic strategy for improving OS and DFS in patients with stage I-II NSCLC after curative resection.
Collapse
Affiliation(s)
- Jianmin Huang
- Biotherapy Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuan Zhao
- Biotherapy Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhen Zhang
- Biotherapy Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuangning Yang
- Biotherapy Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinfeng Chen
- Biotherapy Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunyi Shen
- Biotherapy Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liping Wang
- Cancer Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Qi
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Cancer Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; School of Life Sciences, Zhengzhou University, Zhengzhou, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, China.
| |
Collapse
|
11
|
Tan WQ, Yuan L, Cao X, Wu XY, Xing YQ, Ye M. Overexpression of lncRNA TUG1 enhances the efficacy of DC-CIK immunotherapy in neuroblastoma in vitro and in vivo. Cancer Biomark 2023; 36:53-61. [PMID: 36373305 DOI: 10.3233/cbm-210436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Long non-coding RNA (LncRNA) TUG1 plays a critical role in the development of human cancers. This study explored whether TUG1 is involved in the cytotoxicity of dendritic cells and cytokine-induced killer cells (DCs-CIK), an immunotherapy approach, in neuroblastoma. METHODS A TUG1 expression plasmid was transfected into DCs. Neuroblastoma SK-N-SH cells were incubated with CIK cells, DCs-CIK cells, and TUG1-overexpressing DCs-CIK cells, with or without irradiation. SK-N-SH cell viability, colony formation, migration, and apoptosis were analyzed using CCK-8, colony formation assay, transwell assay, and flow cytometry, respectively. Production of IL-12, IL-2 and IFN-γ in the supernatants was determined using ELISA. A dual luciferase activity assay was performed to confirm the molecular interactions between TUG1 and miR-204. Tumor-bearing mice were established by injection of SK-N-SH cells followed by stimulation with CIK cells, DC-CIK cells, and TUG1-overexpressing DCs-CIK cells. RESULTS Compared to CIK alone or DC-CIK therapy, overexpression of TUG1 significantly suppressed tumor cell proliferation, colony formation, and migration of neuroblastoma cells. Moreover, upregulation of TUG1 robustly induced apoptosis and altered key molecules associated with apoptosis and epithelial-mesenchymal transition. Contents of IL-12, IL-2 and IFN-γ were dramatically elevated in the supernatants in the coculturing system upon transfection with TUG1. In addition, TUG1 was found to be act as miR-204 sponge. Furthermore, in vivo experiments demonstrated that upregulation of TUG1 potentiated the antitumor activity of DC-CIK immunotherapy. CONCLUSION Overexpression of TUG1 promotes DC maturation and enhances CIK cytotoxicity, suggesting that TUG1 may be a novel target for enhancing DC-CIK based immunotherapy for neuroblastoma.
Collapse
Affiliation(s)
- Wei-Qiang Tan
- Department of Surgery, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China.,Department of Surgery, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China
| | - Li Yuan
- Department of Surgery, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China.,Department of Surgery, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China
| | - Xu Cao
- Department of Surgery, Children's Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China.,Department of Surgery, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China
| | - Xiao-Yuan Wu
- Department of Surgery, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China
| | - Yi-Qun Xing
- Department of Surgery, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China
| | - Ming Ye
- Department of Surgery, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
12
|
NK and cells with NK-like activities in cancer immunotherapy-clinical perspectives. Med Oncol 2022; 39:131. [PMID: 35716327 DOI: 10.1007/s12032-022-01735-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/13/2022] [Indexed: 01/10/2023]
Abstract
Natural killer (NK) cells are lymphoid cells of innate immunity that take important roles in immune surveillance. NK cells are considered as a bridge between innate and adaptive immunity, and their infiltration into tumor area is related positively with prolonged patient survival. They are defined as CD16+ CD56+ CD3- cells in clinic. NK cells promote cytolytic effects on target cells and induce their apoptosis. Loss of NK cell cytotoxic activity and reduction in the number of activating receptors are the current issues for application of such cells in cellular immunotherapy, which resulted in the diminished long-term effects. The focus of this review is to discuss about the activity of NK cells and cells with NK-like activity including natural killer T (NKT), cytokine-induced killer (CIK) and lymphokine-activated killer (LAK) cells in immunotherapy of human solid cancers.
Collapse
|
13
|
Liu Y, Zhang Z, Tian Y, Wang D, Liu S, Li L, Hao N, Qin G, Zhao X, Yang S, Huang J, Shen C, Lei Q, Wang L, Zhang Y. Long-term clinical efficacy of cytokine-induced killer cell-based immunotherapy in early-stage esophageal squamous cell carcinoma. Cytotherapy 2022; 24:526-533. [PMID: 35219583 DOI: 10.1016/j.jcyt.2021.12.008] [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: 08/04/2021] [Revised: 12/25/2021] [Accepted: 12/27/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND AIMS In this retrospective clinical study, the authors investigated the impact of cytokine-induced killer (CIK) cell-based immunotherapies on the long-term survival of patients with esophageal squamous cell carcinoma (ESCC). METHODS A total of 87 patients with ESCC who received comprehensive treatment were enrolled in the study. Of these patients, 43 were in the control group and 44 were in the CIK treatment group. Flow cytometry analysis was performed to detect the phenotype and anti-tumor function of CIK cells. Clinical characteristics were compared between these two groups, and the survival estimates of ESCC patients were determined using Kaplan-Meier analysis. RESULTS CIK cells contained a high proportion of the main functional fraction (CD3+CD56+ group) and exhibited a strong killing ability for esophageal cancer cells in vitro. Importantly, overall survival (OS) and progression-free survival (PFS) were significantly higher in the CIK group than in the control group in early-stage ESCC. However, patients with advanced-stage ESCC did not benefit from CIK cell-based therapy in terms of OS and PFS compared with the control group. CONCLUSIONS These results demonstrate that CIK cells combined with conventional treatments potentially prolong long-term survival of patients and may serve as a combined therapeutic approach for the treatment of early-stage ESCC.
Collapse
Affiliation(s)
- Yanfen Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Yonggui Tian
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Dan Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Saisai Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Lin Li
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Na Hao
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guohui Qin
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Xuan Zhao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Shuangning Yang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianmin Huang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Chunyi Shen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Qingyang Lei
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Liping Wang
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China; School of Life Sciences, Zhengzhou University, Zhengzhou, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, China.
| |
Collapse
|
14
|
DAP10 Predicted the Outcome of Pediatric B-Cell Acute Lymphoblastic Leukemia and Was Associated with the T-Cell Exhaustion. JOURNAL OF ONCOLOGY 2021; 2021:4824868. [PMID: 34868314 PMCID: PMC8639274 DOI: 10.1155/2021/4824868] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/27/2021] [Indexed: 01/30/2023]
Abstract
B-cell acute lymphoblastic leukemia is the most common malignant tumor in children. About 10–15% of patients will relapse with a 5-year OS of 57.5% for the past 20 years. As tumor microenvironment plays an important role in the disease process, many types of immunotherapy are approached. New immunotherapies including CAR-T cells have been developed for refractory B-ALL treatment. However, CAR-T treatment faces several problems, including loss of the target antigen and in vivo T-cell persistence. Here, we analyzed the tumor microenvironment of pediatric B-ALL patients in TARGET database. Using Cox analysis and PPI network, we finally sorted out the DAP10 gene. We found that DAP10 was hardly expressed in leukemic B cells. DAP10 was downregulated in B-ALL compared with normal individuals, and low expression level of DAP10 predicted poor survival. Furthermore, we found the tumor microenvironment was different in DAP10 high and low expression children. The CD8+ T cells might be hard to activate and more likely to suffer from exhaustion in DAP10 lowly expressed children. In conclusion, our results showed that DAP10 was a well biomarker to indicate the prognosis and tumor microenvironment in pediatric B-ALL. The treatment strategy of immunotherapy for the leukemic children with DAP10 lowly expressed should be adjusted if needed.
Collapse
|
15
|
Chen Z, Chen C, Li L, Zhang T, Wang X. The spliceosome pathway activity correlates with reduced anti-tumor immunity and immunotherapy response, and unfavorable clinical outcomes in pan-cancer. Comput Struct Biotechnol J 2021; 19:5428-5442. [PMID: 34667536 PMCID: PMC8501672 DOI: 10.1016/j.csbj.2021.09.029] [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: 07/07/2021] [Revised: 09/20/2021] [Accepted: 09/26/2021] [Indexed: 12/12/2022] Open
Abstract
Alterations in the spliceosome pathway (SP) have been associated with diverse human cancers. In this study, we explored associations of the SP activity with various clinical features, anti-tumor immune signatures, tumor immunity-related genomic and molecular features, and the response to immunotherapies and targeted therapies in 29 cancer types from The Cancer Genome Atlas (TCGA) database. We showed that the SP activity was an oncogenic signature, as evidenced by its hyperactivation in cancer and invasive cancer subtypes and correlations with unfavorable clinical outcomes and anti-tumor immunosuppression in various cancers. The SP activity showed positive correlations with tumor mutation burden (TMB) and aneuploidy in diverse cancers, suggesting its association with genomic instability. However, the negative association between the SP activity and anti-tumor immune response was independent of its associations with aneuploidy and TMB. Furthermore, we supported that the SP activity had a negative correlation with immunotherapy response in four cancer cohorts treated by immune checkpoint inhibitors. Moreover, elevated SP activity is correlated with increased drug sensitivity for a broad spectrum of anti-tumor targeted therapies. In conclusion, the SP activity is a negative biomarker for anti-tumor immune response, prognosis, and the response to immunotherapeutic and targeted drugs in pan-cancer.
Collapse
Affiliation(s)
- Zuobing Chen
- Department of Rehabilitation Medicine, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Canping Chen
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| | - Lin Li
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| | - Tianfang Zhang
- Department of Rehabilitation Medicine, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
16
|
Feng Q, Song D, Wang X. Pan-cancer analysis reveals that neurotrophin signaling correlates positively with anti-tumor immunity, clinical outcomes, and response to targeted therapies and immunotherapies in cancer. Life Sci 2021; 282:119848. [PMID: 34293398 DOI: 10.1016/j.lfs.2021.119848] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 12/23/2022]
Abstract
AIMS The crosstalk between cancer cells and nerves plays an important role in tumor biology. However, the correlation between the neurotrophin signaling (NS) and anti-tumor immunity and immunotherapy response in cancer remains unexplored. MATERIALS AND METHODS We analyzed associations of NS with anti-tumor immune signatures, tumor immunity-related molecular and genomic features, and clinical features in 33 TCGA cancer types. We also explored the association between NS and the response to immune checkpoint inhibitors (ICIs) in four cancer cohorts. KEY FINDINGS NS scores had significant positive correlations with the enrichment scores of anti-tumor immune signatures, including CD8+ T cells, interferon response, natural killer cells, Toll-like receptor and NOD-like receptor signaling pathways in most cancer types. NS scores were inversely correlated with the scores of DNA damage repair pathways, tumor mutation burden, copy number alterations, intra-tumor heterogeneity, and tumor stemness in diverse cancers. In contrast, NS scores were significantly and positively correlated with the apoptosis pathway's scores in 32 of the 33 cancer types. NS scores were significantly lower in early-stage versus late-stage and in primary versus metastatic tumors in diverse cancers. Higher NS scores were correlated with better survival in pan-cancer and in eight individual cancer types. Moreover, the response rate to ICIs was higher in higher-NS-score than in lower-NS-score tumors in four cancer cohorts. Elevated NS was correlated with increased drug sensitivity for numerous anti-tumor targeted drugs. SIGNIFICANCE NS is a positive biomarker for anti-tumor immune response, prognosis, and the response to targeted and immunotherapeutic drugs in cancer.
Collapse
Affiliation(s)
- Qiushi Feng
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| | - Dandan Song
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
17
|
Interleukin-2 therapy of cancer-clinical perspectives. Int Immunopharmacol 2021; 98:107836. [PMID: 34218217 DOI: 10.1016/j.intimp.2021.107836] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/18/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022]
Abstract
Interleukin (IL)-2 is a pleiotropic cytokine that displays opposing activities on immune system acting either in favor of or against cancer progression. Advanced/metastatic melanoma and renal cell carcinoma (RCC) are the two types of cancers that included most studies implemented for assessing the role of high-dose IL-2 therapy. The use of high-dose IL-2 therapy can, however, increase the rate of toxicities and interferes with the activity of endothelial cells (ECs) and effector T cells in tumor microenvironment (TME). This implies the need for adjusting strategies related to the cytokine therapy, such as suppressing signals that are interfering with the activity of this cytokine or the use of engineered IL-2 variants. The focus of this review is to discuss about pros and cons related to the IL-2 therapy and propose strategies to increase the efficacy of therapy. The outcomes of this literature will call for application of variants of IL-2 engineered to represent higher half-life and efficacy, and are more safe in the area of cancer immunotherapy.
Collapse
|
18
|
van der Hoorn IAE, Flórez-Grau G, van den Heuvel MM, de Vries IJM, Piet B. Recent Advances and Future Perspective of DC-Based Therapy in NSCLC. Front Immunol 2021; 12:704776. [PMID: 34262573 PMCID: PMC8273436 DOI: 10.3389/fimmu.2021.704776] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/15/2021] [Indexed: 12/11/2022] Open
Abstract
Current treatment for patients with non-small-cell lung cancer (NSCLC) is suboptimal since therapy is only effective in a minority of patients and does not always induce a long-lasting response. This highlights the importance of exploring new treatment options. The clinical success of immunotherapy relies on the ability of the immune system to mount an adequate anti-tumor response. The activation of cytotoxic T cells, the effector immune cells responsible for tumor cell killing, is of paramount importance for the immunotherapy success. These cytotoxic T cells are primarily instructed by dendritic cells (DCs). DCs are the most potent antigen-presenting cells (APCs) and are capable of orchestrating a strong anti-cancer immune response. DC function is often suppressed in NSCLC. Therefore, resurrection of DC function is an interesting approach to enhance anti-cancer immune response. Recent data from DC-based treatment studies has given rise to the impression that DC-based treatment cannot induce clinical benefit in NSCLC by itself. However, these are all early-phase studies that were mainly designed to study safety and were not powered to study clinical benefit. The fact that these studies do show that DC-based therapies were well-tolerated and could induce the desired immune responses, indicates that DC-based therapy is still a promising option. Especially combination with other treatment modalities might enhance immunological response and clinical outcome. In this review, we will identify the possibilities from current DC-based treatment trials that could open up new venues to improve future treatment.
Collapse
Affiliation(s)
- Iris A E van der Hoorn
- Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Georgina Flórez-Grau
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Berber Piet
- Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
19
|
Song L, Wang S, Fang T, Qiu X, Wang X, Zhou X, Morse MA, Hobeika A, Wu W, Yang H, Ren J, Lyerly HK. Changes in Peripheral Blood Regulatory T Cells and IL-6 and IL-10 Levels Predict Response of Pediatric Medulloblastoma and Germ Cell Tumors With Residual or Disseminated Disease to Craniospinal Irradiation. Int J Radiat Oncol Biol Phys 2021; 111:479-490. [PMID: 33974888 DOI: 10.1016/j.ijrobp.2021.04.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 03/26/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE Radiation therapy (RT) modulates immune cells and cytokines, resulting in both clinically beneficial and detrimental effects. The changes in peripheral blood T lymphocyte subsets and cytokines during RT for pediatric brain tumors and the association of these changes with therapeutic outcomes have not been well described. METHODS AND MATERIALS The study population consisted of children (n = 83, aged 3~18) with primary brain tumors (medulloblastoma, glioma, germ cell tumors (GCT), and central nervous system embryonal tumor-not otherwise specified), with or without residual or disseminated (R/D) diseases who were starting standard postoperative focal or craniospinal irradiation (CSI). Peripheral blood T lymphocyte subsets collected before and 4 weeks after RT were enumerated by flow cytometry. Plasma levels of interleukin (IL)-2, IL-4, IL-6, IL-10, tumor necrosis factor-α, interferon-γ, and IL-17A were measured by cytometric bead array. RESULTS Patients with R/D lesions receiving CSI (n = 32) had a post-RT increase in the frequency of CD3+T and CD8+T cells, a decrease in CD4+T cells, and an increase in regulatory T cells (Tregs) and CD8+CD28- suppressor cells, which was more predominantly seen in these patients than in other groups. In the CSI group with such R/D lesions, consisting of patients with medulloblastoma and germ cell tumors, 19 experienced a complete response (CR) and 13 experienced a partial response (PR) on imaging at 4 weeks after RT. The post/pre-RT ratio of Tregs (P = .0493), IL-6 (P = .0111), and IL-10 (P = .0070) was lower in the CR group than in the PR group. Multivariate analysis revealed that the post/pre-RT ratios of Treg, IL-6, and IL-10 were independent predictors of CR (P < .0001, P = .018, P < .0001, respectively). The areas under the receiver operating curves and confidence intervals were 0.7652 (0.5831-0.8964), 0.7794 (0.5980-0.9067), and 0.7085 (0.5223-0.8552) for IL-6, IL-10, and Treg, respectively. The sensitivities of IL-6, IL-10, and Treg to predict radiotherapeutic responses were 100%, 92.3%, and 61.5%, and specificity was 52.6%, 57.9%, and 84.2%, respectively. CONCLUSIONS CSI treatment to those with R/D lesions predominantly exerted an effect on antitumor immune response compared with both R/D lesion-free but exposed to focal or CSI RT and with R/D lesions and exposed to focal RT. Such CSI with R/D lesions group experiencing CR is more likely to have a decrease in immunoinhibitory molecules and cells than patients who only achieve PR. Measuring peripheral blood Treg, IL-6, and IL-10 levels could be valuable for predicting radiotherapeutic responses of pediatric brain tumors with R/D lesions to CSI for medulloblastoma and intracranial germ cell tumors.
Collapse
Affiliation(s)
- Linan Song
- Departments of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China; Departments of Radio-Oncology, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Shuo Wang
- Departments of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Tong Fang
- Departments of Radio-Oncology, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xiaoguang Qiu
- Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Xiaoli Wang
- Departments of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xinna Zhou
- Departments of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Michael A Morse
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Amy Hobeika
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Wanshui Wu
- Department of Pediatrics, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Huabing Yang
- Departments of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Jun Ren
- Departments of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China; Department of Surgery, Duke University Medical Center, Durham, North Carolina.
| | - Herbert Kim Lyerly
- Department of Surgery, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
20
|
Liu Y, Tang L, Gao N, Diao Y, Zhong J, Deng Y, Wang Z, Jin G, Wang X. Synthetic MUC1 breast cancer vaccine containing a Toll-like receptor 7 agonist exerts antitumor effects. Oncol Lett 2020; 20:2369-2377. [PMID: 32782554 PMCID: PMC7400475 DOI: 10.3892/ol.2020.11762] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022] Open
Abstract
Adjuvant immunotherapy has recently emerged as a potential treatment strategy for breast cancer. The tumor-associated protein mucin 1 (MUC1) has received increasing attention due to its high expression in numerous types of common tumors, in which MUC1 acts as a cancer antigen. However, the simple mixed composition of an adjuvant and a peptide is not a sufficient rationale for a MUC1 peptide-based vaccine. The present study developed a novel Toll-like receptor 7 (TLR7) agonist-conjugated MUC1 peptide vaccine (T7-MUC1), which elicited an effective immune response and a robust antitumor effect in a mouse breast cancer model. In vitro, T7-MUC1 significantly increased the release of cytokines in mouse bone marrow dendritic cells and spleen lymphocytes, and induced the dendritic cell-cytokine-induced killer response against tumor cells with high MUC1 expression. In vivo, it was observed that the 4T1 tumor weights in mice immunized with the T7-MUC1 conjugate were reduced by ≥70% compared with those in the control group. Furthermore, the therapeutic responses in vivo were attributed to the increase in specific humoral and cellular immunity, including high antibody titers, antibody-dependent cell-mediated cytotoxicity and cytotoxic T-lymphocyte activity. The percentages of CD3+/CD8+ T-cells were significantly higher in the T7-MUC1 treatment group compared with those in the control group. Therefore, the results of the present study suggested that the T7-MUC1 vaccine inhibited tumor growth in mice and thus may have potential as a therapeutic candidate in clinical trials for breast cancer immunotherapy.
Collapse
Affiliation(s)
- Yu Liu
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China.,Department of Research and Education, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518001, P.R. China
| | - Li Tang
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China.,College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of The Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Ningning Gao
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China
| | - Yuwen Diao
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China
| | - Jingjing Zhong
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China
| | - Yongqiang Deng
- Department of Oral and Maxillofacial Surgery, Shenzhen University General Hospital, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China
| | - Zhulin Wang
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China
| | - Guangyi Jin
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, P.R. China
| | - Xiaodong Wang
- International Cancer Center, National-Regional Engineering Lab for Synthetic Biology of Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong 518055, 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: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022]
Abstract
Cytokine-induced killer (CIK) cells represent an exceptional T-cell population uniting a T cell and natural killer cell-like phenotype in their terminally differentiated CD3+ CD56+ subset, which features non-MHC-restricted tumor-killing activity. CIK cells have provided encouraging results in initial clinical studies and revealed synergistic antitumor effects when combined with standard therapeutic procedures. We established the international registry on CIK cells (IRCC) to collect and evaluate clinical trials for the treatment of cancer patients in 2010. Moreover, our registry set new standards on the reporting of results from clinical trials using CIK cells. In the present update, a total of 106 clinical trials including 10,225 patients were enrolled in IRCC, of which 4,889 patients in over 30 distinct tumor entities were treated with CIK cells alone or in combination with conventional or novel therapies. Significantly improved median progression-free survival and overall survival were shown in 27 trials, and 9 trials reported a significantly increased 5-year survival rate. Mild adverse effects and graft-versus-host diseases were also observed in the studies. Recently, more efforts have been put into the improvement of antitumoral efficacy by CIK cells including the administration of immune checkpoint inhibitors and modification with chimeric antigen receptorc. The minimal toxicity and multiple improvements on their tumor-killing activity both make CIK cells a favorable therapeutic tool in the clinical practice of cancer immunotherapy.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| |
Collapse
|
22
|
Sievers NM, Dörrie J, Schaft N. CARs: Beyond T Cells and T Cell-Derived Signaling Domains. Int J Mol Sci 2020; 21:E3525. [PMID: 32429316 PMCID: PMC7279007 DOI: 10.3390/ijms21103525] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/08/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
When optimizing chimeric antigen receptor (CAR) therapy in terms of efficacy, safety, and broadening its application to new malignancies, there are two main clusters of topics to be addressed: the CAR design and the choice of transfected cells. The former focuses on the CAR construct itself. The utilized transmembrane and intracellular domains determine the signaling pathways induced by antigen binding and thereby the cell-specific effector functions triggered. The main part of this review summarizes our understanding of common signaling domains employed in CARs, their interactions among another, and their effects on different cell types. It will, moreover, highlight several less common extracellular and intracellular domains that might permit unique new opportunities. Different antibody-based extracellular antigen-binding domains have been pursued and optimized to strike a balance between specificity, affinity, and toxicity, but these have been reviewed elsewhere. The second cluster of topics is about the cellular vessels expressing the CAR. It is essential to understand the specific attributes of each cell type influencing anti-tumor efficacy, persistence, and safety, and how CAR cells crosstalk with each other and bystander cells. The first part of this review focuses on the progress achieved in adopting different leukocytes for CAR therapy.
Collapse
Affiliation(s)
- Nico M. Sievers
- Department of Dermatology, Universtitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany; (N.M.S.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Östliche Stadtmauerstraße 30, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Ulmenweg 18, 91054 Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, Universtitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany; (N.M.S.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Östliche Stadtmauerstraße 30, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Ulmenweg 18, 91054 Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, Universtitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany; (N.M.S.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Östliche Stadtmauerstraße 30, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Ulmenweg 18, 91054 Erlangen, Germany
| |
Collapse
|
23
|
Pan C, Liu H, Robins E, Song W, Liu D, Li Z, Zheng L. Next-generation immuno-oncology agents: current momentum shifts in cancer immunotherapy. J Hematol Oncol 2020; 13:29. [PMID: 32245497 PMCID: PMC7119170 DOI: 10.1186/s13045-020-00862-w] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/23/2020] [Indexed: 12/30/2022] Open
Abstract
Cancer immunotherapy has reached a critical point, now that immune checkpoint inhibitors and two CAR-T products have received market approval in treating 16 types of cancers and 1 tissue-agnostic cancer indication. Accompanying these advances, the 2018 Nobel Prize was awarded for the discovery of immune checkpoint pathways, which has led to the revolution of anti-cancer treatments. However, expanding the indications of immuno-oncology agents and overcoming treatment resistance face mounting challenges. Although combination immunotherapy is an obvious strategy to pursue, the fact that there have been more failures than successes in this effort has served as a wake-up call, placing emphasis on the importance of building a solid scientific foundation for the development of next-generation immuno-oncology (IO) agents. The 2019 China Cancer Immunotherapy Workshop was held to discuss the current challenges and opportunities in IO. At this conference, emerging concepts and strategies for IO development were proposed, focusing squarely on correcting the immunological defects in the tumor microenvironment. New targets such as Siglec-15 and new directions including neoantigens, cancer vaccines, oncolytic viruses, and cytokines were reviewed. Emerging immunotherapies were discussed in the areas of overcoming primary and secondary resistance to existing immune checkpoint inhibitors, activating effector cells, and targeting immunosuppressive mechanisms in the tumor microenvironment. In this article, we highlight old and new waves of IO therapy development, and provide perspectives on the latest momentum shifts in cancer immunotherapy.
Collapse
Affiliation(s)
- Chongxian Pan
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- University of California, Davis, CA, USA
| | - Hongtao Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- University of Chicago, Chicago, IL, USA
| | - Elizabeth Robins
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, USA
| | - Wenru Song
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- Kira Pharmaceuticals, Cambridge, MA, USA
| | - Delong Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- New York Medical College, Valhalla, NY, USA
| | - Zihai Li
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, USA
| | - Lei Zheng
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.
- Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
24
|
Wang S, Wang X, Zhou X, Lyerly HK, Morse MA, Ren J. DC-CIK as a widely applicable cancer immunotherapy. Expert Opin Biol Ther 2020; 20:601-607. [PMID: 32033522 DOI: 10.1080/14712598.2020.1728250] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Introduction: Immunotherapy is now a standard treatment for many malignancies. Although immune checkpoint inhibition has demonstrated substantial efficacy by enhancing T cell activation and function in the tumor microenvironment, adoptive transfer of T and NK cell products promises to provide activated cells capable of immediate and direct tumor destruction. A widely applicable, non-MHC dependent, cellular therapy, consisting of in vitro generated dendritic cells (DC) combined with cytokine-induced killer cells (CIK), is highly efficient to produce from individual patients and has demonstrated safety and efficacy alone or with chemotherapy.Areas covered: We summarize the clinical data from studies of DC-CIK and discuss future research directions.Expert opinion: Patients with a wide variety of tumor types who have received DC-CIK therapy may experience clinical responses. This versatile therapy synergizes with other anti-cancer therapies including chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinna Zhou
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | | | - Michael A Morse
- Department of Surgery, Duke University Medical Center, Durham, NC U.S.A.,Department of Medicine, Duke University Medical Center, Durham, NC U.S.A
| | - Jun Ren
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Department of Surgery, Duke University Medical Center, Durham, NC U.S.A
| |
Collapse
|
25
|
Therapeutic Cancer Vaccination with Ex Vivo RNA-Transfected Dendritic Cells-An Update. Pharmaceutics 2020; 12:pharmaceutics12020092. [PMID: 31979205 PMCID: PMC7076681 DOI: 10.3390/pharmaceutics12020092] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/09/2020] [Accepted: 01/18/2020] [Indexed: 12/19/2022] Open
Abstract
Over the last two decades, dendritic cell (DC) vaccination has been studied extensively as active immunotherapy in cancer treatment and has been proven safe in all clinical trials both with respect to short and long-term side effects. For antigen-loading of dendritic cells (DCs) one method is to introduce mRNA coding for the desired antigens. To target the whole antigenic repertoire of a tumor, even the total tumor mRNA of a macrodissected biopsy sample can be used. To date, reports have been published on a total of 781 patients suffering from different tumor entities and HIV-infection, who have been treated with DCs loaded with mRNA. The majority of those were melanoma patients, followed by HIV-infected patients, but leukemias, brain tumors, prostate cancer, renal cell carcinomas, pancreatic cancers and several others have also been treated. Next to antigen-loading, mRNA-electroporation allows a purposeful manipulation of the DCs’ phenotype and function to enhance their immunogenicity. In this review, we intend to give a comprehensive summary of what has been published regarding clinical testing of ex vivo generated mRNA-transfected DCs, with respect to safety and risk/benefit evaluations, choice of tumor antigens and RNA-source, and the design of better DCs for vaccination by transfection of mRNA-encoded functional proteins.
Collapse
|
26
|
Clinical Trials with Cytokine-Induced Killer Cells and CAR-T Cell Transplantation for Non-small Cell Lung Cancer Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1292:113-130. [DOI: 10.1007/5584_2020_522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
27
|
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: 0.5] [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
|
28
|
Adoptive immunotherapy with autologous T-cell infusions reduces opioid requirements in advanced cancer patients. Pain 2019; 161:127-134. [DOI: 10.1097/j.pain.0000000000001702] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
29
|
Sprooten J, Ceusters J, Coosemans A, Agostinis P, De Vleeschouwer S, Zitvogel L, Kroemer G, Galluzzi L, Garg AD. Trial watch: dendritic cell vaccination for cancer immunotherapy. Oncoimmunology 2019; 8:e1638212. [PMID: 31646087 PMCID: PMC6791419 DOI: 10.1080/2162402x.2019.1638212] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
Dendritic- cells (DCs) have received considerable attention as potential targets for the development of anticancer vaccines. DC-based anticancer vaccination relies on patient-derived DCs pulsed with a source of tumor-associated antigens (TAAs) in the context of standardized maturation-cocktails, followed by their reinfusion. Extensive evidence has confirmed that DC-based vaccines can generate TAA-specific, cytotoxic T cells. Nonetheless, clinical efficacy of DC-based vaccines remains suboptimal, reflecting the widespread immunosuppression within tumors. Thus, clinical interest is being refocused on DC-based vaccines as combinatorial partners for T cell-targeting immunotherapies. Here, we summarize the most recent preclinical/clinical development of anticancer DC vaccination and discuss future perspectives for DC-based vaccines in immuno-oncology.
Collapse
Affiliation(s)
- Jenny Sprooten
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jolien Ceusters
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
- Department of Gynecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
- Center for Cancer Biology (CCB), VIB, Leuven, Belgium
| | - Steven De Vleeschouwer
- Research Group Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China
- Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
- Université de Paris Descartes, Paris, France
| | - Abhishek D. Garg
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
30
|
Increased glycolysis correlates with elevated immune activity in tumor immune microenvironment. EBioMedicine 2019; 42:431-442. [PMID: 30935888 PMCID: PMC6491961 DOI: 10.1016/j.ebiom.2019.03.068] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/14/2019] [Accepted: 03/25/2019] [Indexed: 01/01/2023] Open
Abstract
Background Prior studies showed that tumor glycolysis and tumor immune evasion are interdependent. However, a systematic investigation of the association between tumor glycolysis and tumor immunity in various cancers remains lacking. Methods Using the Cancer Genome Atlas (TCGA) datasets, we explored the association between glycolytic activity and immune signatures in 14 cancer types. We also explored the associations between glycolytic activity and tumor immunity associated genetic features, including PD-L1 expression, tumor mutation burden (TMB), and tumor aneuploidy. Moreover, we performed in vitro experiments to verify some findings from bioinformatics analysis. Furthermore, we explored the association between tumor glycolytic activity and immunotherapy response. Findings Glycolytic activity was likely correlated with active immune signatures in various cancers and highly glycolytic tumors presented an immune-stimulatory tumor microenvironment. Compared to TMB and aneuploidy, glycolytic activity was a stronger and more consistent predictor for immune signatures in diverse cancers. Both computational and experimental analyses showed that glycolysis could increase PD-L1 expression in tumor. Glycolytic activity had a strong correlation with apoptosis which was a strong positive predictor for immune signatures, suggesting that apoptosis could be an important medium connecting glycolytic activity with immune activity in cancer. Finally, highly glycolytic tumors exhibited a better immunotherapy response and a favorable survival in the immunotherapy setting. Interpretation Tumor glycolysis may increase tumor immunity in diverse cancers. Glycolytic activity enhances PD-L1 expression on tumor cells and thus promotes anti-PD-1/PD-L1 immunotherapy response. Thus, the tumor glycolytic activity could be a predictive biomarker for immunotherapy response in diverse cancers. Fund This work was supported by the China Pharmaceutical University (grant numbers 3150120001, 2632018YX01 to XW).
Collapse
|