1
|
Cai X, Li J, Wu M, Liu Q. Causal relationship between immune cells and the risk of myeloperoxidase antineutrophil cytoplasmic antibody-associated vasculitis: A Mendelian randomization study. FASEB J 2024; 38:e23821. [PMID: 39018091 DOI: 10.1096/fj.202400141r] [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: 01/18/2024] [Revised: 06/13/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024]
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA) is a rare inflammatory disease categorized as antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis. The majority of patients are ANCA-positive, predominantly against myeloperoxidase (MPO). Previous studies have predominantly concentrated on the association between EGPA and neutrophils, but recent research has emphasized the role of lymphocytes in the development of EGPA. The objective of our research was to examine the causal association between immune cells and MPO + ANCA EGPA. A two-sample bidirectional Mendelian randomization (MR) analysis was performed, which included 159 MPO + ANCA EGPA cases and 6688 controls and utilized Genome-Wind Associaton Studies (GWAS) summary statistics of immune traits from approximately 3757 individuals, encompassing around 22 million single nucleotide polymorphisms (SNPs). Our findings revealed that 23 immunophenotypes were associated with MPO + ANCA EGPA. Furthermore, the reverse MR analysis showed that MPO + ANCA EGPA had significant causal effects on three immunophenotypes within the Treg panel. By integrating existing research, our study unveiled the contributions of Tregs, B cells, and monocytes to the development of EGPA. Subgroup analysis specifically examined the roles of lymphocyte subtypes, cytokines, and their surface molecules in the pathogenic mechanisms of the disease. This comprehensive approach provides a novel perspective on the biological mechanisms and early intervention strategies for MPO + ANCA EGPA by focusing on immune cells.
Collapse
Affiliation(s)
- Xiaojing Cai
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junhua Li
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Manyi Wu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingquan Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Wang Y, Zeng Y, Yang W, Wang X, Jiang J. Targeting CD8 + T cells with natural products for tumor therapy: Revealing insights into the mechanisms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155608. [PMID: 38642413 DOI: 10.1016/j.phymed.2024.155608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/27/2024] [Accepted: 04/07/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND Despite significant advances in cancer immunotherapy over the past decades, such as T cell-engaging chimeric antigen receptor (CAR)-T cell therapy and immune checkpoint blockade (ICB), therapeutic failure resulting from various factors remains prevalent. Therefore, developing combinational immunotherapeutic strategies is of great significance for improving the clinical outcome of cancer immunotherapy. Natural products are substances that naturally exist in various living organisms with multiple pharmacological or biological activities, and some of them have been found to have anti-tumor potential. Notably, emerging evidences have suggested that several natural compounds may boost the anti-tumor effects through activating immune response of hosts, in which CD8+ T cells play a pivotal role. METHODS The data of this review come from PubMed, Web of Science, Google Scholar, and ClinicalTrials (https://clinicaltrials.gov/) with the keywords "CD8+ T cell", "anti-tumor", "immunity", "signal 1", "signal 2", "signal 3", "natural products", "T cell receptor (TCR)", "co-stimulation", "co-inhibition", "immune checkpoint", "inflammatory cytokine", "hesperidin", "ginsenoside", "quercetin", "curcumin", "apigenin", "dendrobium officinale polysaccharides (DOPS)", "luteolin", "shikonin", "licochalcone A", "erianin", "resveratrol", "procyanidin", "berberine", "usnic acid", "naringenin", "6-gingerol", "ganoderma lucidum polysaccharide (GL-PS)", "neem leaf glycoprotein (NLGP)", "paclitaxel", "source", "pharmacological activities", and "toxicity". These literatures were published between 1993 and 2023. RESULTS Natural products have considerable advantages as anti-tumor drugs based on the various species, wide distribution, low price, and few side effects. This review summarized the effects and mechanisms of some natural products that exhibit anti-tumor effects via targeting CD8+ T cells, mainly focused on the three signals that activate CD8+ T cells: TCR, co-stimulation, and inflammatory cytokines. CONCLUSION Clarifying the role and underlying mechanism of natural products in cancer immunotherapy may provide more options for combinational treatment strategies and benefit cancer therapy, to shed light on identifying potential natural compounds for improving the clinical outcome in cancer immunotherapy.
Collapse
Affiliation(s)
- Yuke Wang
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China; Department of Neurosurgery, Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Yan Zeng
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenyong Yang
- Department of Neurosurgery, Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Xiuxuan Wang
- Research and Development Department, Beijing DCTY Biotech Co., Ltd., Beijing, China
| | - Jingwen Jiang
- West China School of Public Health and West China Fourth Hospital, West China School of Basic Medical Sciences & Forensic Medicine and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
3
|
Szukiewicz D. CX3CL1 (Fractalkine)-CX3CR1 Axis in Inflammation-Induced Angiogenesis and Tumorigenesis. Int J Mol Sci 2024; 25:4679. [PMID: 38731899 PMCID: PMC11083509 DOI: 10.3390/ijms25094679] [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: 03/28/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
The chemotactic cytokine fractalkine (FKN, chemokine CX3CL1) has unique properties resulting from the combination of chemoattractants and adhesion molecules. The soluble form (sFKN) has chemotactic properties and strongly attracts T cells and monocytes. The membrane-bound form (mFKN) facilitates diapedesis and is responsible for cell-to-cell adhesion, especially by promoting the strong adhesion of leukocytes (monocytes) to activated endothelial cells with the subsequent formation of an extracellular matrix and angiogenesis. FKN signaling occurs via CX3CR1, which is the only known member of the CX3C chemokine receptor subfamily. Signaling within the FKN-CX3CR1 axis plays an important role in many processes related to inflammation and the immune response, which often occur simultaneously and overlap. FKN is strongly upregulated by hypoxia and/or inflammation-induced inflammatory cytokine release, and it may act locally as a key angiogenic factor in the highly hypoxic tumor microenvironment. The importance of the FKN/CX3CR1 signaling pathway in tumorigenesis and cancer metastasis results from its influence on cell adhesion, apoptosis, and cell migration. This review presents the role of the FKN signaling pathway in the context of angiogenesis in inflammation and cancer. The mechanisms determining the pro- or anti-tumor effects are presented, which are the cause of the seemingly contradictory results that create confusion regarding the therapeutic goals.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
4
|
Kang N, Xue H, Lin YY, Dong X, Classen A, Wu R, Jin Y, Lin D, Volik S, Ong C, Gleave M, Collins C, Wang Y. Influence of ADT on B7-H3 expression during CRPC progression from hormone-naïve prostate cancer. Cancer Gene Ther 2023; 30:1382-1389. [PMID: 37452083 PMCID: PMC10581905 DOI: 10.1038/s41417-023-00644-9] [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: 01/05/2023] [Revised: 06/21/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023]
Abstract
Androgen deprivation therapy (ADT) is the standard care for advanced prostate cancer (PCa) patients. Unfortunately, although tumors respond well initially, they enter dormancy and eventually progress to fatal/incurable castration-resistant prostate cancer (CRPC). B7-H3 is a promising new target for PCa immunotherapy. CD276 (B7-H3) gene has a presumptive androgen receptor (AR) binding site, suggesting potential AR regulation. However, the relationship between B7-H3 and AR is controversial. Meanwhile, the expression pattern of B7-H3 following ADT and during CRPC progression is largely unknown, but critically important for identifying patients and determining the optimal timing of B7-H3 targeting immunotherapy. In this study, we performed a longitudinal study using our unique PCa patient-derived xenograft (PDX) models and assessed B7-H3 expression during post-ADT disease progression. We further validated our findings at the clinical level in PCa patient samples. We found that B7-H3 expression was negatively regulated by AR during the early phase of ADT treatment, but positively associated with PCa proliferation during the remainder of disease progression. Our findings suggest its use as a biomarker for diagnosis, prognosis, and ADT treatment response, and the potential of combining ADT and B7-H3 targeting immunotherapy for hormone-naïve PCa treatment to prevent fatal CRPC relapse.
Collapse
Affiliation(s)
- Ning Kang
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
| | - Hui Xue
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
| | - Yen-Yi Lin
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Xin Dong
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
| | - Adam Classen
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Rebecca Wu
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
| | - Yuxuan Jin
- University of Victoria, Victoria, BC, Canada
| | - Dong Lin
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Christopher Ong
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Martin Gleave
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Colin Collins
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada.
- Vancouver Prostate Centre, Vancouver, BC, Canada.
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
5
|
Ding YN, Ding HY, Li H, Yang R, Huang JY, Chen H, Wang LH, Wang YJ, Hu CM, An YL, Zhang ZY, Yu WP, Tang QS, Shao GL. Photosensitive Small Extracellular Vesicles Regulate the Immune Microenvironment of Triple Negative Breast Cancer. Acta Biomater 2023:S1742-7061(23)00329-X. [PMID: 37302734 DOI: 10.1016/j.actbio.2023.06.004] [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: 02/28/2023] [Revised: 05/25/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Currently, the treatment of triple-negative breast cancer (TNBC) is limited by the special pathological characteristics of this disease. In recent years, photodynamic therapy (PDT) has created new hope for the treatment of TNBC. Moreover, PDT can induce immunogenic cell death (ICD) and improve tumor immunogenicity. However, even though PDT can improve the immunogenicity of TNBC, the inhibitory immune microenvironment of TNBC still weakens the antitumor immune response. Therefore, we used the neutral sphingomyelinase inhibitor GW4869 to inhibit the secretion of small extracellular vesicles (sEVs) by TNBC cells to improve the tumor immune microenvironment and enhance antitumor immunity. In addition, bone mesenchymal stem cell (BMSC)-derived sEVs have good biological safety and a strong drug loading capacity, which can effectively improve the efficiency of drug delivery. In this study, we first obtained primary BMSCs and sEVs, and then the photosensitizers Ce6 and GW4869 were loaded into the sEVs by electroporation to produce immunomodulatory photosensitive nanovesicles (Ce6-GW4869/sEVs). When administered to TNBC cells or orthotopic TNBC models, these photosensitive sEVs could specifically target TNBC and improve the tumor immune microenvironment. Moreover, PDT combined with GW4869-based therapy showed a potent synergistic antitumor effect mediated by direct killing of TNBC and activation of antitumor immunity. Here, we designed photosensitive sEVs that could target TNBC and regulate the tumor immune microenvironment, providing a potential approach for improving the effectiveness of TNBC treatment. STATEMENT OF SIGNIFICANCE: We designed an immunomodulatory photosensitive nanovesicle (Ce6-GW4869/sEVs) with the photosensitizer Ce6 to achieve photodynamic therapy and the neutral sphingomyelinase inhibitor GW4869 to inhibit the secretion of small extracellular vesicles (sEVs) by triple-negative breast cancer (TNBC) cells to improve the tumor immune microenvironment and enhance antitumor immunity. In this study, the immunomodulatory photosensitive nanovesicle could target TNBC cells and regulate the tumor immune microenvironment, thus providing a potential approach for improving the treatment effect in TNBC. We found that the reduction in tumor sEVs secretion induced by GW4869 improved the tumor-suppressive immune microenvironment. Moreover, similar therapeutic strategies can also be applied in other kinds of tumors, especially immunosuppressive tumors, which is of great value for the clinical translation of tumor immunotherapy.
Collapse
Affiliation(s)
- Yi-Nan Ding
- Medical School of Southeast University, Nanjing 210009, China
| | - Hui-Yan Ding
- Medical School of Southeast University, Nanjing 210009, China
| | - Han Li
- Department of tuberculosis, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Rui Yang
- Center of Reproductive Medicine, State Key Laboratory of Reproductive Medicine, Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, 214002, Jiangsu, China
| | - Jia-Yan Huang
- Department of tuberculosis, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, China
| | - He Chen
- First people's hospital of Changzhou, Changzhou, Jiangsu, China
| | - Lu-Hong Wang
- Medical School of Southeast University, Nanjing 210009, China
| | - Yun-Juan Wang
- Medical School of Southeast University, Nanjing 210009, China
| | - Chun-Mei Hu
- Department of tuberculosis, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan-Li An
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Zhi-Yuan Zhang
- Department of Neurosurgery, Nanjing Jinling hospital, Nanjing University, Nanjing 210002, China
| | - Wei-Ping Yu
- Medical School of Southeast University, Nanjing 210009, China..
| | - Qiu-Sha Tang
- Medical School of Southeast University, Nanjing 210009, China..
| | - Guo-Liang Shao
- Department of interventional oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China..
| |
Collapse
|
6
|
Tilden SG, Ricco MH, Hemann EA, Anchordoquy TJ. Reducing off-target drug accumulation by exploiting a type-III interferon response. J Control Release 2023; 358:729-738. [PMID: 37230293 PMCID: PMC10389760 DOI: 10.1016/j.jconrel.2023.05.029] [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: 02/06/2023] [Revised: 05/17/2023] [Accepted: 05/20/2023] [Indexed: 05/27/2023]
Abstract
Nanomedicines have been touted as the future of cancer therapy for decades. However, the field of tumor-targeted nanomedicine has failed to significantly advance toward becoming the primary choice for cancer intervention. One of the largest obstacles that has yet to be overcome is off-target accumulation of the nanoparticles. We propose a novel approach to tumor delivery by focusing on decreasing off-target accumulation of nanomedicines rather than directly increasing tumor delivery. Acknowledging a poorly understood "refractory" response to intravenously injected gene therapy vectors observed in ours and other studies, we hypothesize that virus-like particles (lipoplexes) can be utilized to initiate an anti-viral innate immune response that limits off-target accumulation of subsequently administered nanoparticles. Indeed, our results show a significant reduction in the deposition of both dextran and Doxil® in major organs with a concurrent increase in plasma and tumor accumulation when injection occurred 24 h after a lipoplex injection. Furthermore, our data showing that the direct injection of interferon lambda (IFN-λ) is capable of eliciting this response demonstrates a central role for this type III interferon in limiting accumulation in non-tumor tissues.
Collapse
Affiliation(s)
- Scott G Tilden
- University of Colorado, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America.
| | - Madison H Ricco
- University of Colorado, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Emily A Hemann
- Ohio State University, Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Thomas J Anchordoquy
- University of Colorado, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| |
Collapse
|
7
|
Tang X, Yang Y, Zheng M, Yin T, Huang G, Lai Z, Zhang B, Chen Z, Xu T, Ma T, Pan H, Cai L. Magnetic-Acoustic Sequentially Actuated CAR T Cell Microrobots for Precision Navigation and In Situ Antitumor Immunoactivation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2211509. [PMID: 36807373 DOI: 10.1002/adma.202211509] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/10/2023] [Indexed: 05/05/2023]
Abstract
Despite its clinical success, chimeric antigen receptor T (CAR T)-cell immunotherapy remains limited in solid tumors, owing to the harsh physical barriers and immunosuppressive microenvironment. Here a CAR-T-cell-based live microrobot (M-CAR T) is created by decorating CAR T with immunomagnetic beads using click conjugation. M-CAR Ts are capable of magnetic-acoustic actuation for precision targeting and in situ activation of antitumor immune responses. Sequential actuation endows M-CAR Ts with magnetically actuated anti-flow and obstacle avoidance as well as tissue penetration driven by acoustic propulsion, enabling efficient migration and accumulation in artificial tumor models. In vivo, sequentially actuated M-CAR Ts achieves long-distance targeting and accumulate at the peritumoural area under programmable magnetic guidance, and subsequently acoustic tweezers actuate M-CAR Ts to migrate into deep tumor tissues, resulting in a 6.6-fold increase in accumulated exogenous CD8+ CAR T cells compared with that without actuation. Anti-CD3/CD28 immunomagnetic beads stimulate infiltrated CAR T proliferation and activation in situ, significantly enhancing their antitumor efficacy. Thus, this sequential-actuation-guided cell microrobot combines the merits of autonomous targeting and penetration of intelligent robots with in situ T-cell immunoactivation, and holds considerable promise for precision navigation and cancer immunotherapies.
Collapse
Affiliation(s)
- Xiaofan Tang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Ye Yang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Mingbin Zheng
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory for Nanomedicine, Guangdong Medical University, Dongguan, 523808, P. R. China
| | - Ting Yin
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory for Nanomedicine, Guangdong Medical University, Dongguan, 523808, P. R. China
| | - Guojun Huang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Zhengyu Lai
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Baozhen Zhang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Ze Chen
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Tiantian Xu
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Teng Ma
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Hong Pan
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| |
Collapse
|
8
|
Zhang L, Jiang T, Yang Y, Deng W, Lu H, Wang S, Liu R, Chang M, Wu S, Gao Y, Hao H, Shen G, Xu M, Chen X, Hu L, Yang L, Bi X, Lin Y, Lu Y, Jiang Y, Li M, Xie Y. Postpartum hepatitis and host immunity in pregnant women with chronic HBV infection. Front Immunol 2023; 13:1112234. [PMID: 36685527 PMCID: PMC9846060 DOI: 10.3389/fimmu.2022.1112234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Abstract
In order to develop immune tolerant to the fetal, maternal immune system will have some modification comparing to the time before pregnancy. Immune tolerance starts and develops at the maternal placental interface. In innate immunity, decidual natural killer (dNK) cells, macrophages and dendritic cells play a key role in immue tolerance. In adaptive immunity, a moderate increase of number and immune inhibition function of regulatory T cells (Treg) are essential for immune tolerance. The trophoblast cells and immune cells expressing indoleamine 2,3-dioxygenase (IDO), the trophoblast cells expressing HLA-G, and Th1/Th2 shifting to Th2 dominant and Th17/Treg shifting to Treg domiant are in favor of maternal fetal immune tolerance. Steroids (estrogen and progesterone) and human chorionic gonadotropin (HCG) also participate in immune tolerance by inducing Treg cells or upregulating immunosuppressive cytokines. Most of the patients with chronic HBV infection are in the "HBV immune tolerance period" before pregnancy, and the liver disease is relatively stable during pregnancy. In chronic HBV infection women, after delivery, the relative immunosuppression in vivo is reversed, and Th1 is dominant in Th1/Th2 and Th17 is dominant in Th17/Treg balance. After delivery, the number of Treg decrease and NK cells increase in quantity and cytotoxicity in peripheral blood. Liver NK cells may cause liver inflammation through a non-antigen specific mechanism. After delivery, the number of CD8+ T cells will increase and HBV specific T cell response recovers from the disfunction in pregnancy. Under the background of postpartum inflammation, the rapid decrease of cortisol after delivery, and especially the enhancement of HBV specific T cell response induced by HBV DNA and cytokines, are the main reasons for postpartum hepatitis. HBeAg positive, especially HBeAg<700 S/CO, and HBV DNA>3-5Log10IU/ml are risk factors for postpartum hepatitis. Antiviral treatment in late pregnancy can reduce the incidence of mother to child transmission (MTCT) in chronic HBV infection women. Chronic HBV infection women have hepatitis both during pregnancy and more often in 12 weeks postpartum. It is generally agreed that postpartum hepatitis is mild symptoms and self-limited. Delaying drug withdrawal to 48 weeks can increase the seroconversion rate of HBeAg in delivery women with elevated alanine aminotransferase (ALT) in pregnancy.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Tingting Jiang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ying Yang
- Hepatology Department 2, Xingtai Second Hospital, Xingtai, China
| | - Wen Deng
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Huihui Lu
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China,Department of Obstetrics and Gynecology, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiyu Wang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ruyu Liu
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Min Chang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Shuling Wu
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yuanjiao Gao
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Hongxiao Hao
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ge Shen
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Mengjiao Xu
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaoxue Chen
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Leiping Hu
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Liu Yang
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaoyue Bi
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yanjie Lin
- Department of Hepatology Division 2, Peking University Ditan Teaching Hospital, Beijing, China
| | - Yao Lu
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China,*Correspondence: Yao Lu, ; Yuyong Jiang, ; Minghui Li, ; Yao Xie,
| | - Yuyong Jiang
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China,*Correspondence: Yao Lu, ; Yuyong Jiang, ; Minghui Li, ; Yao Xie,
| | - Minghui Li
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China,Department of Hepatology Division 2, Peking University Ditan Teaching Hospital, Beijing, China,*Correspondence: Yao Lu, ; Yuyong Jiang, ; Minghui Li, ; Yao Xie,
| | - Yao Xie
- Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, China,Department of Hepatology Division 2, Peking University Ditan Teaching Hospital, Beijing, China,*Correspondence: Yao Lu, ; Yuyong Jiang, ; Minghui Li, ; Yao Xie,
| |
Collapse
|
9
|
Mayr P, Lutz M, Schmutz M, Hoeppner J, Liesche-Starnecker F, Schlegel J, Gaedcke J, Claus R. Progressive multifocal leukoencephalopathy associated with chemotherapy induced lymphocytopenia in solid tumors – case report of an underestimated complication. Front Oncol 2022; 12:905103. [PMID: 36003787 PMCID: PMC9394442 DOI: 10.3389/fonc.2022.905103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/24/2022] [Indexed: 11/19/2022] Open
Abstract
Background JC virus reactivation causing progressive multifocal leukoencephalopathy (PML) occurs preferentially in human immunodeficiency virus (HIV) positive individuals or patients suffering from hematologic neoplasms due to impaired viral control. Reactivation in patients suffering from solid malignancies is rarely described in published literature. Case Presentation Here we describe a case of PML in a male patient suffering from esophageal cancer who underwent neoadjuvant radiochemotherapy and surgical resection in curative intent resulting in complete tumor remission. The radiochemotherapy regimen contained carboplatin and paclitaxel (CROSS protocol). Since therapy onset, the patient presented with persistent and progredient leukopenia and lymphopenia in absence of otherwise known risk factors for PML. Symptom onset, which comprised aphasia, word finding disorder, and paresis, was apparent 7 months after therapy initiation. There was no relief in symptoms despite standard of care PML directed supportive therapy. The patient died two months after therapy onset. Conclusion PML is a very rare event in solid tumors without obvious states of immununosuppression and thus harbors the risk of unawareness. The reported patient suffered from lymphopenia, associated with systemic therapy, but was an otherwise immunocompetent individual. In case of neurologic impairment in patients suffering from leukopenia, PML must be considered – even in the absence of hematologic neoplasia or HIV infection.
Collapse
|
10
|
Li X, He G, Liu J, Yan M, Shen M, Xu L, An M, Huang J, Gao Z. CCL2-mediated monocytes regulate immune checkpoint blockade resistance in pancreatic cancer. Int Immunopharmacol 2022; 106:108598. [PMID: 35183036 DOI: 10.1016/j.intimp.2022.108598] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 01/16/2022] [Accepted: 01/27/2022] [Indexed: 12/21/2022]
Abstract
The immunosuppressive microenvironment of pancreatic ductal adenocarcinoma (PDAC) contributes to resistance to immune checkpoint blockade. C-C motif chemokine ligand 2 (CCL2) is believed to participate in pancreatic tumorigenesis, but its role in PDAC progression and resistance to immune checkpoint blockade remains unclear. We hypothesized that CCL2 contributes to the pancreatic immunosuppressive microenvironment. In this study, we found that CCL2 recruits monocytes to and decrease CD8+ T cell infiltration in pancreatic tumors. CCL2 inhibition and monocyte neutralization increased the sensitivity of PDAC to immune checkpoint blockade. The findings of our study suggest the potential of CCL2-mediated monocytes as a target for PDAC treatment.
Collapse
Affiliation(s)
- Xiaocui Li
- Department of Gastroenterology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai 201700, China
| | - Guijun He
- Department of Gastroenterology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai 201700, China
| | - Jican Liu
- Department of Pathology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai 201700, China
| | - Meizhu Yan
- Department of Gastroenterology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai 201700, China
| | - Manru Shen
- Department of Gastroenterology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai 201700, China
| | - Linfang Xu
- Department of Gastroenterology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai 201700, China
| | - Min An
- Department of Gastroenterology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai 201700, China
| | - Jiying Huang
- Department of Gastroenterology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai 201700, China.
| | - Zhenjun Gao
- Department of Gastroenterology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai 201700, China.
| |
Collapse
|
11
|
Betker JL, Anchordoquy TJ. The Effect of Repeat Administration of Lipoplexes on Gene Delivery, Biodistribution, and Cytokine Response in Immunocompetent Tumor-bearing Mice. J Pharm Sci 2021; 111:1926-1936. [PMID: 34929156 DOI: 10.1016/j.xphs.2021.12.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 12/19/2022]
Abstract
It is becoming increasingly clear that the intravenous administration of nanoparticles elicits an immune response that compromises delivery efficiency and can be life threatening. This study investigated both the systemic and tissue-level cytokine response to repeat administration of lipoplexes coated with either lactose or PEG. We report that blood cytokine levels differ significantly from that observed in individual tissues. While we consistently observed a reduced cytokine response to lactosylated particles, this did not result in enhanced delivery or expression as compared to PEGylated formulations. We also document that repeat injection did not increase plasmid levels in the liver, lung, or spleen, but delivery to the tumor was enhanced under these conditions. In addition, we show that changes in neither blood nor tissue cytokines correlated strongly with reporter gene expression, and we observed relatively constant expression efficiencies (RLU/ng plasmid) across all tissues despite a considerably reduced cytokine response in the tumor. Together, these results indicate that both biodistribution and cytokine responses are dramatically altered by a repeat intravenous injection of lipoplexes, and that the mechanisms regulating reporter gene expression are not straightforward.
Collapse
Affiliation(s)
- Jamie L Betker
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Thomas J Anchordoquy
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States.
| |
Collapse
|
12
|
Grote S, Ureña-Bailén G, Chan KCH, Baden C, Mezger M, Handgretinger R, Schleicher S. In Vitro Evaluation of CD276-CAR NK-92 Functionality, Migration and Invasion Potential in the Presence of Immune Inhibitory Factors of the Tumor Microenvironment. Cells 2021; 10:1020. [PMID: 33925968 PMCID: PMC8145105 DOI: 10.3390/cells10051020] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/09/2021] [Accepted: 04/22/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Melanoma is the most lethal of all skin-related cancers with incidences continuously rising. Novel therapeutic approaches are urgently needed, especially for the treatment of metastasizing or therapy-resistant melanoma. CAR-modified immune cells have shown excellent results in treating hematological malignancies and might represent a new treatment strategy for refractory melanoma. However, solid tumors pose some obstacles for cellular immunotherapy, including the identification of tumor-specific target antigens, insufficient homing and infiltration of immune cells as well as immune cell dysfunction in the immunosuppressive tumor microenvironment (TME). METHODS In order to investigate whether CAR NK cell-based immunotherapy can overcome the obstacles posed by the TME in melanoma, we generated CAR NK-92 cells targeting CD276 (B7-H3) which is abundantly expressed in solid tumors, including melanoma, and tested their effectivity in vitro in the presence of low pH, hypoxia and other known factors of the TME influencing anti-tumor responses. Moreover, the CRISPR/Cas9-induced disruption of the inhibitory receptor NKG2A was assessed for its potential enhancement of NK-92-mediated anti-tumor activity. RESULTS CD276-CAR NK-92 cells induced specific cytolysis of melanoma cell lines while being able to overcome a variety of the immunosuppressive effects normally exerted by the TME. NKG2A knock-out did not further improve CAR NK-92 cell-mediated cytotoxicity. CONCLUSIONS The strong cytotoxic effect of a CD276-specific CAR in combination with an "off-the-shelf" NK-92 cell line not being impaired by some of the most prominent negative factors of the TME make CD276-CAR NK-92 cells a promising cellular product for the treatment of melanoma and beyond.
Collapse
Affiliation(s)
- Stefan Grote
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany; (G.U.-B.); (K.C.-H.C.); (C.B.); (M.M.); (R.H.); (S.S.)
| | | | | | | | | | | | | |
Collapse
|
13
|
Cancer Immunotherapy Strategies: Basic Principles. Bioanalysis 2021. [DOI: 10.1007/978-3-030-78338-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
14
|
Kang N, Eccleston M, Clermont PL, Latarani M, Male DK, Wang Y, Crea F. EZH2 inhibition: a promising strategy to prevent cancer immune editing. Epigenomics 2020; 12:1457-1476. [PMID: 32938196 PMCID: PMC7607396 DOI: 10.2217/epi-2020-0186] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023] Open
Abstract
Immunotherapies are revolutionizing the clinical management of a wide range of cancers. However, intrinsic or acquired unresponsiveness to immunotherapies does occur due to the dynamic cancer immunoediting which ultimately leads to immune escape. The evolutionarily conserved histone modifier enhancer of zeste 2 (EZH2) is aberrantly overexpressed in a number of human cancers. Accumulating studies indicate that EZH2 is a main driver of cancer cells' immunoediting and mediate immune escape through downregulating immune recognition and activation, upregulating immune checkpoints and creating an immunosuppressive tumor microenvironment. In this review, we overviewed the roles of EZH2 in cancer immunoediting, the preclinical and clinical studies of current pharmacologic EZH2 inhibitors and the prospects for EZH2 inhibitor and immunotherapy combination for cancer treatment.
Collapse
Affiliation(s)
- Ning Kang
- Department of Experimental Therapeutics, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Mark Eccleston
- Belgian Volition SPRL, Parc Scientifique Créalys, Rue Phocas Lejeune 22, BE-5032 Isnes, Belgium
| | - Pier-Luc Clermont
- Faculty of Medicine, Université Laval, 1050, avenue de la Médecine, Québec, QC, G1V 0A6, Canada
| | - Maryam Latarani
- Cancer Research Group, School of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - David Kingsley Male
- Cancer Research Group, School of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
- Department of Urologic Sciences, The Vancouver Prostate Centre, The University of British Columbia, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Francesco Crea
- Cancer Research Group, School of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| |
Collapse
|
15
|
Simon B, Uslu U. Fasten the seat belt: Increasing safety of CAR T-cell therapy. Exp Dermatol 2020; 29:1039-1045. [PMID: 32627228 DOI: 10.1111/exd.14131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/01/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
Abstract
After the recent success and approvals of chimeric antigen receptor (CAR) T cells in haematological malignancies, its efficacy is currently evaluated in a broad spectrum of tumor entities including melanoma. However, severe and potentially life-threatening side effects like cytokine release syndrome, neurologic toxicities, and the competing risk of morbidity and mortality from the treatment itself are still a major limiting factor in the current CAR T-cell landscape. In addition, especially in solid tumors, the lack of ideal target antigens to avoid on-target/off-tumor toxicities also restricts its use. While various groups are working on strategies to boost CAR T-cell efficacy, mechanisms to increase engineered T-cell safety should not move out of focus. Thus, the aim of this article is to summarize and to discuss current and potential future strategies and mechanisms to increase CAR T-cell safety in order to enable the wide use of this promising approach in melanoma and other tumor entities.
Collapse
Affiliation(s)
- Bianca Simon
- Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany.,Comprehensive Cancer Center Erlangen- European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany.,Division of Genetics, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ugur Uslu
- Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany.,Comprehensive Cancer Center Erlangen- European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| |
Collapse
|
16
|
Singh L, Singh MK, Rizvi MA, Pushker N, Bakhshi S, Sen S, Kashyap S. Prognostic significance of immune checkpoints in the tumour-stromal microenvironment of sebaceous gland carcinoma. Br J Ophthalmol 2020; 105:48-56. [PMID: 32277010 DOI: 10.1136/bjophthalmol-2019-315490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/02/2020] [Accepted: 03/11/2020] [Indexed: 11/04/2022]
Abstract
BACKGROUND Immune checkpoint blockade strategies have gained attention in the treatment/prognosis of cancers by targeting the programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) pathway alone or in combination with cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) blockade and are currently in clinical trials. The present study investigated the expression of the PD-1, PD-L1, CTLA-4, CD4 and CD8 proteins and their prognostic value in the tumour microenvironment of sebaceous gland carcinoma (SGC). METHODS The expression levels of PD-1, PD-L1, CTLA-4, CD4 and CD8 proteins were assessed in 52 cases of SGC by immunohistochemistry and validated by western blotting. mRNA expression was measured by quantitative real-time PCR. Kaplan-Meier curves and Cox proportional hazard models were used to analyse the correlation of protein expression with clinicopathological parameters and disease-free survival. RESULTS The expression of PD-L1 was found to be higher in tumour cells than in stromal cells. In univariate analysis, the expression of PD-1 in tumour-infiltrating lymphocytes (tPD-1) and PD-L1 in tumour cells was associated with reduced disease-free survival, whereas PD-L1 expression in stromal lymphocyte infiltration (sPD-L1) was associated with the increased survival of patients (p<0.05). However, by multivariate analysis, the expression of tPD-1 was found to be an independent prognostic factor for poor survival. CONCLUSION Our study highlights the prognostic outcome of PD-1 and PD-L1 protein expression in cells of tumour-stromal compartments. These results indicate that the PD-1/PD-L1 pathway mediates important interactions within the tumour microenvironment in SGC.
Collapse
Affiliation(s)
- Lata Singh
- Ocular Pathology, All India Institute of Medical Sciences, New Delhi, Delhi, India.,Biosciences, Jamia Millia Islamia, New Delhi, Delhi, India
| | | | | | - Neelam Pushker
- Ophthalmology, Dr R P Centre, AIIMS, New Delhi, Delhi, India
| | | | - Seema Sen
- Ocular Pathology, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Seema Kashyap
- Ocular Pathology, All India Institute of Medical Sciences, New Delhi, Delhi, India
| |
Collapse
|
17
|
Sheppard AD, Lysaght J. Immunometabolism and Its Potential to Improve the Current Limitations of Immunotherapy. Methods Mol Biol 2020; 2184:233-263. [PMID: 32808230 DOI: 10.1007/978-1-0716-0802-9_17] [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: 06/11/2023]
Abstract
The last century of research in tumor immunology has culminated in the advent of immunotherapy, most notably immune checkpoint inhibitors. These drugs have shown encouraging results across a multitude of malignancies and have shifted the paradigm of cancer treatment. However, no more than 40% of patients treated with these immune checkpoint blockade inhibitors respond. Thus, resistance is a barrier to therapy that remains poorly understood. All cells require energy and biosynthetic precursors for survival, growth, and functioning, where multiple metabolic pathways allow for flexibility in how nutrients are utilized. A defining hallmark of many cancers is altered cellular metabolism, creating an imbalanced demand for nutrients within the tumor microenvironment. Immunometabolism is increasingly understood to be vital to the functions and phenotypes of a myriad of immune cell subsets. In tumors, the high demand for nutrients by the tumor drives competition between tumor cells and infiltrating immune cells, culminating in dysfunctional immune responses. This chapter discusses the recent successes in cancer immunotherapy and highlights challenges to therapy. We also outline the major metabolic processes involved in the generation of an immune response, how this can become dysregulated in the context of the tumor microenvironment, and how this contributes to resistance to immunotherapy. Finally, we explore the potential for targeting immunometabolic pathways to improve immunotherapy, and examine current trials targeting various aspects of metabolism in an attempt to improve the outcomes from immunotherapy.
Collapse
Affiliation(s)
- Andrew D Sheppard
- Cancer Immunology and Immunotherapy Group, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland.
| |
Collapse
|
18
|
Simon B, Uslu U. CAR-T cell therapy in melanoma: A future success story? Exp Dermatol 2019; 27:1315-1321. [PMID: 30288790 DOI: 10.1111/exd.13792] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/23/2018] [Accepted: 09/26/2018] [Indexed: 12/13/2022]
Abstract
Chimeric antigen receptor (CAR)-T cells are one of the impressive recent success stories of anti-cancer immunotherapy. Especially in haematological malignancies, this treatment strategy has shown promising results leading to the recent approval of two CAR-T cell constructs targeting CD19 in the United States and the European Union. After the huge success in haematological cancers, the next step will be the evaluation of its efficacy in different solid tumors, which is currently investigated in preclinical as well as clinical settings. A commonly examined tumor model in the context of immunotherapy is melanoma, since it is known for its immunogenic features. However, the first results of CAR-T cell therapy in solid tumors did not reveal the same impressive outcomes that were observed in haematological malignancies, as engineered cells need to cope with several challenges. Obstacles include the lack of migration of CAR-T cells from blood vessels to the tumor site as well as the immunosuppressive tumor microenvironment within solid tumors. Another hurdle is posed by the identification of an ideal target antigen to avoid on-target/off-tumor toxicities. Regarding immune escape mechanisms, which can be developed by tumor cells to bypass immune recognition, the observation of antigen loss should also be considered. This article gives an overview of the challenges displayed in CAR-T cell therapy for the use in solid tumors and discusses different new strategies and approaches that deal with these problems in order to improve CAR-T cell therapy, particularly for its use in melanoma.
Collapse
Affiliation(s)
- Bianca Simon
- Department of Dermatology, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany.,Department of Biology, Division of Genetics, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ugur Uslu
- Department of Dermatology, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
19
|
Synergism of PDL/PD1 and IL33/ST2 Axis in Tumor Immunology. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2019. [DOI: 10.2478/sjecr-2018-0033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Abstract
When it comes to tumor immunology, understanding of molecular pathways is rather important. During oncogenesis, many molecules should be taken in consideration altogether in context of a single malignancy. It is of a great significance to determine whether these molecules act synergistically or contrary, whether to understand a malignant disease more thoroughly, or even more important, to reveal new approaches of therapy. In this review, we discuss whether and how IL-33/ST2 and PD-1/PDL axis involve in antitumor immunity.
Collapse
|
20
|
Wang P, Liu J, Song Y, Liu Q, Wang C, Qian C, Zhang S, Zhu W, Yang X, Wan F, Liu Z, Luo D. Screening of immunosuppressive factors for biomarkers of breast cancer malignancy phenotypes and subtype-specific targeted therapy. PeerJ 2019; 7:e7197. [PMID: 31293831 PMCID: PMC6599676 DOI: 10.7717/peerj.7197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/27/2019] [Indexed: 11/20/2022] Open
Abstract
We aimed to screen and validate immunosuppressive factors in luminal- and basal-like breast cancer cell lines and tissue samples associated with malignant phenotypes. The mRNA microarray datasets, GSE40057 and GSE1561, were downloaded and remodeled, and differentially expressed genes were identified. Weighted gene co-expression network analysis (WGCNA) and gene ontology (GO) and KEGG pathway enrichment analysis were performed to explore the immune-related events related to the basal-like breast cancer. The online resources, GOBO, Kaplan–Meier Plotter and UALCAN, were employed to screen for immunosuppressive factors associated with breast cancer malignant phenotypes. Immunohistochemistry was used to evaluate VEGFA and MIF levels in breast tumors and normal breast tissues; qPCRs and western blots were used to validate the expression of clinical immuno-oncology (IO) therapeutic targets CD274 (PD-L1) and IL8 in cell lines. The results showed that various immune-related events contribute to basal-like breast cancer. First, TGFβ1 and IL8 had higher average expression levels in more malignant cell lines; second, MIF and VEGFA had higher average expression levels in more malignant breast cancer tissues, and the high expression levels were associated with poor survival rate. Third, IO targets CD274 and IL8 which were confirmed to be more suitable for the treatment of basal-like breast cancer. In view of the above, during the formation and development of breast cancer, immune-related genes are always activated, and immunosuppressive factors, IL8, TGFβ1, MIF, and VEGFA are up-regulated. Such molecules could be used as biomarkers for breast cancer prognosis. However, because individual immune-related factors can play several biological roles, the mechanistic relationship between immunosuppressive factors and breast cancer malignant phenotypes and the feasibility of their application as drug targets require further investigation.
Collapse
Affiliation(s)
- Ping Wang
- Queen Mary School, Nanchang University, Nanchang, China
| | - Jiaxuan Liu
- Queen Mary School, Nanchang University, Nanchang, China
| | - Yunlei Song
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Qiang Liu
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Wang
- School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Caiyun Qian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Shuhua Zhang
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Weifeng Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Xiaohong Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Fusheng Wan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Zhuoqi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University, Nanchang, China
| |
Collapse
|
21
|
Nahas MR, Stroopinsky D, Rosenblatt J, Cole L, Pyzer AR, Anastasiadou E, Sergeeva A, Ephraim A, Washington A, Orr S, McMasters M, Weinstock M, Jain S, Leaf RK, Ghiasuddin H, Rahimian M, Liegel J, Molldrem JJ, Slack F, Kufe D, Avigan D. Hypomethylating agent alters the immune microenvironment in acute myeloid leukaemia (AML) and enhances the immunogenicity of a dendritic cell/AML vaccine. Br J Haematol 2019; 185:679-690. [PMID: 30828801 PMCID: PMC6590084 DOI: 10.1111/bjh.15818] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023]
Abstract
Acute myeloid leukaemia (AML) is a lethal haematological malignancy characterized by an immunosuppressive milieu in the tumour microenvironment (TME) that fosters disease growth and therapeutic resistance. Hypomethylating agents (HMAs) demonstrate clinical efficacy in AML patients and exert immunomodulatory activities. In the present study, we show that guadecitabine augments both antigen processing and presentation, resulting in increased AML susceptibility to T cell-mediated killing. Exposure to HMA results in the activation of the endogenous retroviral pathway with concomitant downstream amplification of critical mediators of inflammation. In an immunocompetent murine leukaemia model, guadecitabine negatively regulates inhibitory accessory cells in the TME by decreasing PD-1 (also termed PDCD1) expressing T cells and reducing AML-mediated expansion of myeloid-derived suppressor cells. Therapy with guadecitabine results in enhanced leukaemia-specific immunity, as manifested by increased CD4 and CD8 cells targeting syngeneic leukaemia cells. We have previously reported that vaccination with AML/dendritic cell fusions elicits the expansion of leukaemia-specific T cells and protects against disease relapse. In the present study, we demonstrate that vaccination in conjunction with HMA therapy results in enhanced anti-leukaemia immunity and survival. The combination of a novel personalized dendritic cell/AML fusion vaccine and an HMA has therapeutic potential, and a clinical trial investigating this combination is planned.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/immunology
- Antineoplastic Agents, Immunological/pharmacology
- Azacitidine/analogs & derivatives
- Azacitidine/immunology
- Azacitidine/pharmacology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cancer Vaccines/immunology
- Cell Line, Tumor
- DNA Methylation/drug effects
- Dendritic Cells/immunology
- Disease Models, Animal
- Down-Regulation/drug effects
- Down-Regulation/immunology
- Humans
- Immunity, Cellular/drug effects
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/immunology
- Mice, Inbred C57BL
- Neoplasm Transplantation
- Programmed Cell Death 1 Receptor/metabolism
- Retroviridae/immunology
- Tumor Microenvironment/immunology
- Virus Activation/immunology
Collapse
Affiliation(s)
- Myrna R Nahas
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Dina Stroopinsky
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jacalyn Rosenblatt
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Leandra Cole
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Athalia R Pyzer
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Eleni Anastasiadou
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Anna Sergeeva
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adam Ephraim
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Abigail Washington
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Shira Orr
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Matthew Weinstock
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Salvia Jain
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Rebecca K Leaf
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Haider Ghiasuddin
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Maryam Rahimian
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jessica Liegel
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Frank Slack
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Donald Kufe
- Dana-Farber Cancer Institute, Department of Medical Oncology, Boston, MA
| | - David Avigan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
PD-1 and its ligands are important immune checkpoints in cancer. Oncotarget 2018; 8:2171-2186. [PMID: 27974689 PMCID: PMC5356790 DOI: 10.18632/oncotarget.13895] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 11/21/2016] [Indexed: 12/31/2022] Open
Abstract
Checkpoint programmed death-1 (PD-1)/programmed cell death ligands (PD-Ls) have been identified as negative immunoregulatory molecules that promote immune evasion of tumor cells. The interaction of PD-1 and PD-Ls inhibits the function of T cells and tumor-infiltrating lymphocytes (TIL) while increasing the function of immunosuppressive regulatory T cells (Tregs). This condition causes the tumor cells to evade immune response. Thus, the blockade of PD-1/PD-L1 enhances anti-tumor immunity by reducing the number and/or the suppressive activity of Tregs and by restoring the activity of effector T cells. Furthermore, some monoclonal antibodies blockading PD-1/PD-Ls axis have achieved good effect and received Food and Drug Administration approval. The role of PD-1/PD-Ls in tumors has been well studied, but little is known on the mechanism by which PD-1 blocks T-cell activation. In this study, we provide a brief overview on the discovery and regulatory mechanism of PD-1 and PD-L1 dysregulation in tumors, as well as the function and signaling pathway of PD-1 and its ligands; their roles in tumor evasion and clinical treatment were also studied.
Collapse
|
23
|
Composite analysis of immunological and metabolic markers defines novel subtypes of triple negative breast cancer. Mod Pathol 2018; 31:288-298. [PMID: 28984302 PMCID: PMC5963501 DOI: 10.1038/modpathol.2017.126] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/01/2017] [Accepted: 08/03/2017] [Indexed: 12/22/2022]
Abstract
Cancer biology is influenced by the tumor microenvironment, which impacts disease prognosis and therapeutic interventions. The inter-relationship of tumor-infiltrating lymphocytes, immune response regulators, and a glycolytic tumor environment was evaluated in a cohort of 183 largely consecutive patients with triple negative breast cancer diagnosis. High levels of tumor-infiltrating lymphocytes were associated with improved survival of triple negative breast cancer cases. However, elevated levels of PD-L1, CD163, and FOXP3 were individually associated with significantly decreased overall survival. These three determinants were significantly correlated, and could serve to differentiate the prognostic significance of tumor-infiltrating lymphocytes. Interestingly, a glycolytic tumor environment, as determined by the expression of MCT4 in the tumor stroma, was associated with the immune evasive environment and poor prognosis. Clustering of all markers defined four distinct triple negative breast cancer subtypes that harbored prognostic significance in multivariate analysis. Immune and metabolic markers stratified triple negative breast cancer into subtypes that have prognostic significance and implications for therapies targeting immune checkpoints and tumor metabolism.
Collapse
|
24
|
Hahn SA, Neuhoff A, Landsberg J, Schupp J, Eberts D, Leukel P, Bros M, Weilbaecher M, Schuppan D, Grabbe S, Tueting T, Lennerz V, Sommer C, Jonuleit H, Tuettenberg A. A key role of GARP in the immune suppressive tumor microenvironment. Oncotarget 2018; 7:42996-43009. [PMID: 27248166 PMCID: PMC5190003 DOI: 10.18632/oncotarget.9598] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/14/2016] [Indexed: 12/26/2022] Open
Abstract
In melanoma patients, one of the main reasons for tumor immune escape and therapy failure is the immunosuppressive tumor microenvironment. Herein, suppressive immune cells and inhibitory factors secreted by the tumor itself play a central role. In the present study we show that the Treg activation marker GARP (glycoprotein A repetitions predominant), known to induce peripheral tolerance in a TGF-β dependent way, is also expressed on human primary melanoma. Interestingly, membrane bound GARP is shed from the surface of both, activated Treg and melanoma cells, and, in its soluble form (sGARP), not only induces peripheral Treg but also a tumor associated (M2) macrophage phenotype. Notably, proliferation of cytotoxic T cells and their effector function is inhibited in the presence of sGARP. GARP expression on Treg and melanoma cells is significantly decreased in the presence of agents such as IFN-α, thus explaining at least in part a novel mechanism of action of this adjuvant therapy. In conclusion, GARP in its soluble and membrane bound form contributes to peripheral tolerance in a multipronged way, potentiates the immunosuppressive tumor microenvironment and thus acts as a negative regulator in melanoma patients. Therefore, it may qualify as a promising target and a new checkpoint for cancer immunotherapy.
Collapse
Affiliation(s)
- Susanne A Hahn
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Annemarie Neuhoff
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Jenny Landsberg
- Department of Dermatology, University Medical Center, Bonn, Germany
| | - Jonathan Schupp
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Daniela Eberts
- Department of Medicine II, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Petra Leukel
- Institute of Neuropathology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Martin Weilbaecher
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University, Mainz, Germany.,Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Thomas Tueting
- Department of Dermatology, University Medical Center, Bonn, Germany
| | - Volker Lennerz
- Department of Medicine II, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Clemens Sommer
- Institute of Neuropathology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Helmut Jonuleit
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Andrea Tuettenberg
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
25
|
Kroll AV, Fang RH, Jiang Y, Zhou J, Wei X, Yu CL, Gao J, Luk BT, Dehaini D, Gao W, Zhang L. Nanoparticulate Delivery of Cancer Cell Membrane Elicits Multiantigenic Antitumor Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:10.1002/adma.201703969. [PMID: 29239517 PMCID: PMC5794340 DOI: 10.1002/adma.201703969] [Citation(s) in RCA: 335] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 08/29/2017] [Indexed: 05/08/2023]
Abstract
Anticancer vaccines train the body's own immune system to recognize and eliminate malignant cells based on differential antigen expression. While conceptually attractive, clinical efficacy is lacking given several key challenges stemming from the similarities between cancerous and healthy tissue. Ideally, an effective vaccine formulation would deliver multiple tumor antigens in a fashion that potently stimulates endogenous immune responses against those antigens. Here, it is reported on the fabrication of a biomimetic, nanoparticulate anticancer vaccine that is capable of delivering autologously derived tumor antigen material together with a highly immunostimulatory adjuvant. The two major components, tumor antigens and adjuvant, are presented concurrently in a fashion that maximizes their ability to promote effective antigen presentation and activation of downstream immune processes. Ultimately, it is demonstrated that the formulation can elicit potent antitumor immune responses in vivo. When combined with additional immunotherapies such as checkpoint blockades, the nanovaccine demonstrates substantial therapeutic effect. Overall, the work represents the rational application of nanotechnology for immunoengineering and can provide a blueprint for the future development of personalized, autologous anticancer vaccines with broad applicability.
Collapse
|
26
|
Bull JMC. A review of immune therapy in cancer and a question: can thermal therapy increase tumor response? Int J Hyperthermia 2017; 34:840-852. [PMID: 28974121 DOI: 10.1080/02656736.2017.1387938] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Immune therapy is a successful cancer treatment coming into its own. This is because checkpoint molecules, adoptive specific lymphocyte transfer and chimeric antigen T-cell (CAR-T) therapy are able to induce more durable responses in an increasing number of malignancies compared to chemotherapy. In addition, immune therapies are able to treat bulky disease, whereas standard cytotoxic therapies cannot treat large tumour burdens. Checkpoint inhibitor monoclonal antibodies are becoming widely used in the clinic and although more complex, adoptive lymphocyte transfer and CAR-T therapies show promise. We are learning that there are nuances to predicting the successful use of the checkpoint inhibitors as well as to specific-antigen adoptive and CAR-T therapies. We are also newly aware of a here-to-fore unrealised natural force, the status of the microbiome. However, despite better understanding of mechanisms of action of the new immune therapies, the best responses to the new immune therapies remain 20-30%. Likely the best way to improve this somewhat low response rate for patients is to increase the patient's own immune response. Thermal therapy is a way to do this. All forms of thermal therapy, from fever-range systemic thermal therapy, to high-temperature HIFU and even cryotherapy improve the immune response pre-clinically. It is time to test the immune therapies with thermal therapy in vivo to test for optimal timing of the combinations that will best enhance tumour response and then to begin to test the immune therapies with thermal therapy in the clinic as soon as possible.
Collapse
Affiliation(s)
- Joan M C Bull
- a Division of Oncology, Department of Internal Medicine , The University of Texas Medical School at Houston , Houston , TX , USA
| |
Collapse
|
27
|
Chimeric antigen receptor T-cell therapy for glioblastoma. Transl Res 2017; 187:93-102. [PMID: 28755873 DOI: 10.1016/j.trsl.2017.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 06/25/2017] [Accepted: 07/11/2017] [Indexed: 02/06/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has shown great promise in the treatment of hematological disease, and its utility for treatment of solid tumors is beginning to unfold. Glioblastoma continues to portend a grim prognosis and immunotherapeutic approaches are being explored as a potential treatment strategy. Identification of appropriate glioma-associated antigens, barriers to cell delivery, and presence of an immunosuppressive microenvironment are factors that make CAR T-cell therapy for glioblastoma particularly challenging. However, insights gained from preclinical studies and ongoing clinical trials indicate that CAR T-cell therapy will continue to evolve and likely become integrated with current therapeutic strategies for malignant glioma.
Collapse
|
28
|
Linedale R, Schmidt C, King BT, Ganko AG, Simpson F, Panizza BJ, Leggatt GR. Elevated frequencies of CD8 T cells expressing PD-1, CTLA-4 and Tim-3 within tumour from perineural squamous cell carcinoma patients. PLoS One 2017; 12:e0175755. [PMID: 28423034 PMCID: PMC5396892 DOI: 10.1371/journal.pone.0175755] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/30/2017] [Indexed: 12/31/2022] Open
Abstract
Perineural spread of tumour cells along cranial nerves is a severe complication of primary cutaneous squamous cell carcinomas of the head and neck region. While surgical excision of the tumour is the treatment of choice, removal of all the tumour is often complicated by the neural location and recurrence is frequent. Non-invasive immune treatments such as checkpoint inhibitor blockade may be useful in this set of tumours although little is understood about the immune response to perineural spread of squamous cell carcinomas. Immunohistochemistry studies suggest that perineural tumour contains a lymphocyte infiltrate but it is difficult to quantitate the different proportions of immune cell subsets and expression of checkpoint molecules such as PD-1, Tim-3 and CTLA-4. Using flow cytometry of excised perineural tumour tissue, we show that a T cell infiltrate is prominent in addition to less frequent B cell, NK cell and NKT cell infiltrates. CD8 T cells are more frequent than other T cells in the tumour tissue. Amongst CD8 T cells, the frequency of Tim-3, CTLA-4 and PD-1 expressing cells was significantly greater in the tumour relative to the blood, a pattern that was repeated for Tim-3, CTLA-4 and PD-1 amongst non-CD8 T cells. Using immunohistochemistry, PD-1 and PD-L1-expression could be detected in close proximity amongst perineural tumour tissue. The data suggest that perineural SCC contains a mixture of immune cells with a predominant T cell infiltrate containing CD8 T cells. Elevated frequencies of tumour-associated Tim-3+, CTLA-4+ and PD-1+ CD8 T cells suggests that a subset of patients may benefit from local antibody blockade of these checkpoint inhibitors.
Collapse
Affiliation(s)
- Richard Linedale
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Campbell Schmidt
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia.,Department of Otolaryngology-Head and Neck Surgery, Princess Alexandra Hospital, Brisbane, Australia.,The University of Queensland Faculty of Medicine, Brisbane, Australia
| | - Brigid T King
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia.,The University of Queensland Faculty of Medicine, Brisbane, Australia
| | - Annabelle G Ganko
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia.,The University of Queensland Faculty of Medicine, Brisbane, Australia
| | - Fiona Simpson
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Benedict J Panizza
- Department of Otolaryngology-Head and Neck Surgery, Princess Alexandra Hospital, Brisbane, Australia.,The University of Queensland Faculty of Medicine, Brisbane, Australia
| | - Graham R Leggatt
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| |
Collapse
|
29
|
Mongis A, Piller F, Piller V. Coupling of Immunostimulants to Live Cells through Metabolic Glycoengineering and Bioorthogonal Click Chemistry. Bioconjug Chem 2017; 28:1151-1165. [PMID: 28297599 DOI: 10.1021/acs.bioconjchem.7b00042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The present study investigated the potential of metabolic glycoengineering followed by bioorthogonal click chemistry for introducing into cell-surface glycans different immunomodulating molecules. Mouse tumor models EG7 and MC38-OVA were treated with Ac4GalNAz and Ac4ManNAz followed by ligation of immunostimulants to modified cell-surface glycans of the living cells through bioorthogonal click chemistry. The presence of covalently bound oligosaccharide and oligonucleotide immunostimulants could be clearly established. The activation of a reporter macrophage cell line was determined. Depending on the tumor cell line, covalently and noncovalently bound CpG activated the macrophages by between 67 and 100% over controls. EG7 cells with covalently attached immunostimulants and controls were injected subcutaneously into C57BL/6 mice. All tumor cells subjected to the complete treatment with control molecules formed tumors like nontreated cells confirming cell viability. However, when CpG oligonucleotide was linked to cell-surface glycans, tumor growth was slowed significantly (60% reduction, n = 10, by covalently bound CpG compared to noncovalently bound CpG, n = 10). When mice that had not developed large tumors were challenged with unmodified EG7 cells, no new tumors developed, suggesting protection through the immune system.
Collapse
Affiliation(s)
- Aline Mongis
- Centre de Biophysique Moléculaire, CNRS UPR4301 , Rue Charles Sadron, 45071 Orléans, France
| | - Friedrich Piller
- Centre de Biophysique Moléculaire, CNRS UPR4301 , Rue Charles Sadron, 45071 Orléans, France
| | - Véronique Piller
- Centre de Biophysique Moléculaire, CNRS UPR4301 , Rue Charles Sadron, 45071 Orléans, France
| |
Collapse
|
30
|
Seliger B. Molecular mechanisms of HLA class I-mediated immune evasion of human tumors and their role in resistance to immunotherapies. HLA 2016; 88:213-220. [PMID: 27659281 DOI: 10.1111/tan.12898] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022]
Abstract
Although the human immune system can recognize and eradicate tumor cells, tumors have also been shown to develop different strategies to escape immune surveillance, which has been described for the first time in different mouse models. The evasion of immune recognition was often associated with a poor prognosis and reduced survival of patients. During the last years the molecular mechanisms, which protect tumor cells from this immune attack, have been identified and appear to be more complex than initially expected. However, next to the composition of cellular, soluble and physical components of the tumor microenvironment, the tumor cells changes to limit immune responses. Of particular importance are classical and non-classical human leukocyte antigen (HLA) class I antigens, which often showed a deregulated expression in cancers of distinct origin. Furthermore, HLA class I abnormalities were linked to defects in the interferon signaling, which have both been shown to be essential for mounting immune responses and are involved in resistances to T cell-based immunotherapies. Therefore this review summarizes the expression, regulation, function and clinical relevance of HLA class I antigens in association with the interferon signal transduction pathway and its role in adaptive resistances to immunotherapies.
Collapse
Affiliation(s)
- B Seliger
- Institute of Medical Immunology, Martin-Luther-University Halle-Wittenberg, Halle, Germany.
| |
Collapse
|
31
|
Liu X, Ranganathan R, Jiang S, Fang C, Sun J, Kim S, Newick K, Lo A, June CH, Zhao Y, Moon EK. A Chimeric Switch-Receptor Targeting PD1 Augments the Efficacy of Second-Generation CAR T Cells in Advanced Solid Tumors. Cancer Res 2016; 76:1578-90. [PMID: 26979791 PMCID: PMC4800826 DOI: 10.1158/0008-5472.can-15-2524] [Citation(s) in RCA: 398] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chimeric antigen receptor (CAR)-modified adoptive T-cell therapy has been successfully applied to the treatment of hematologic malignancies, but faces many challenges in solid tumors. One major obstacle is the immune-suppressive effects induced in both naturally occurring and genetically modified tumor-infiltrating lymphocytes (TIL) by inhibitory receptors (IR), namely PD1. We hypothesized that interfering with PD1 signaling would augment CAR T-cell activity against solid tumors. To address this possibility, we introduced a genetically engineered switch receptor construct, comprising the truncated extracellular domain of PD1 and the transmembrane and cytoplasmic signaling domains of CD28, into CAR T cells. We tested the effect of this supplement, "PD1CD28," on human CAR T cells targeting aggressive models of human solid tumors expressing relevant tumor antigens. Treatment of mice bearing large, established solid tumors with PD1CD28 CAR T cells led to significant regression in tumor volume due to enhanced CAR TIL infiltrate, decreased susceptibility to tumor-induced hypofunction, and attenuation of IR expression compared with treatments with CAR T cells alone or PD1 antibodies. Taken together, our findings suggest that the application of PD1CD28 to boost CAR T-cell activity is efficacious against solid tumors via a variety of mechanisms, prompting clinical investigation of this potentially promising treatment modality.
Collapse
Affiliation(s)
- Xiaojun Liu
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Raghuveer Ranganathan
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Shuguang Jiang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Chongyun Fang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Jing Sun
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Soyeon Kim
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Kheng Newick
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Albert Lo
- Department of Biomedical Sciences, School of Veterinary Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Carl H. June
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Yangbing Zhao
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Edmund K. Moon
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
32
|
Chawla S, Warren TA, Wockner LF, Lambie DLJ, Brown IS, Martin TPC, Khanna R, Leggatt GR, Panizza BJ. Galectin-1 is associated with poor prognosis in patients with cutaneous head and neck cancer with perineural spread. Cancer Immunol Immunother 2016; 65:213-22. [PMID: 26759008 PMCID: PMC11028814 DOI: 10.1007/s00262-015-1788-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 12/23/2015] [Indexed: 12/18/2022]
Abstract
Spread of head and neck cancer along the cranial nerves is often a lethal complication of this tumour. Current treatment options include surgical resection and/or radiotherapy, but recurrence is a frequent event suggesting that our understanding of this tumour and its microenvironment is incomplete. In this study, we have analysed the nature of the perineural tumour microenvironment by immunohistochemistry with particular focus on immune cells and molecules, which might impair anti-tumour immunity. Moderate to marked lymphocyte infiltrates were present in 58.8% of the patient cohort including T cells, B cells and FoxP3-expressing T cells. While human leukocyte antigen (HLA) class I and more variably HLA class II were expressed on the tumour cells, this did not associate with patient survival or recurrence. In contrast, galectin-1 staining within lymphocyte areas of the tumour was significantly associated with a poorer patient outcome. Given the known role of galectin-1 in immune suppression, the data suggest that galectin inhibitors might improve the prognosis of patients with perineural spread of cancer.
Collapse
Affiliation(s)
- Sharad Chawla
- Department of Otolaryngology - Head and Neck Surgery, Princess Alexandra Hospital, 237 Ipswich Road, Woolloongabba, Brisbane, QLD, 4102, Australia
| | - Timothy A Warren
- Department of Otolaryngology - Head and Neck Surgery, Princess Alexandra Hospital, 237 Ipswich Road, Woolloongabba, Brisbane, QLD, 4102, Australia
- QIMR Berghofer Institute of Medical Research, Brisbane, QLD, Australia
- The University of Queensland School of Medicine, Brisbane, QLD, Australia
| | - Leesa F Wockner
- QIMR Berghofer Institute of Medical Research, Brisbane, QLD, Australia
| | - Duncan L J Lambie
- The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
- IQ Pathology, Brisbane, QLD, Australia
| | | | - Thomas P C Martin
- Department of Otolaryngology - Head and Neck Surgery, Princess Alexandra Hospital, 237 Ipswich Road, Woolloongabba, Brisbane, QLD, 4102, Australia
| | - Rajiv Khanna
- QIMR Berghofer Institute of Medical Research, Brisbane, QLD, Australia
| | - Graham R Leggatt
- The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Benedict J Panizza
- Department of Otolaryngology - Head and Neck Surgery, Princess Alexandra Hospital, 237 Ipswich Road, Woolloongabba, Brisbane, QLD, 4102, Australia.
- The University of Queensland School of Medicine, Brisbane, QLD, Australia.
| |
Collapse
|
33
|
Lin IYC, Van TTH, Smooker PM. Live-Attenuated Bacterial Vectors: Tools for Vaccine and Therapeutic Agent Delivery. Vaccines (Basel) 2015; 3:940-72. [PMID: 26569321 PMCID: PMC4693226 DOI: 10.3390/vaccines3040940] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 10/30/2015] [Accepted: 10/30/2015] [Indexed: 12/14/2022] Open
Abstract
Genetically attenuated microorganisms, including pathogenic and commensal bacteria, can be engineered to carry and deliver heterologous antigens to elicit host immunity against both the vector as well as the pathogen from which the donor gene is derived. These live attenuated bacterial vectors have been given much attention due to their capacity to induce a broad range of immune responses including localized mucosal, as well as systemic humoral and/or cell-mediated immunity. In addition, the unique tumor-homing characteristics of these bacterial vectors has also been exploited for alternative anti-tumor vaccines and therapies. In such approach, tumor-associated antigen, immunostimulatory molecules, anti-tumor drugs, or nucleotides (DNA or RNA) are delivered. Different potential vectors are appropriate for specific applications, depending on their pathogenic routes. In this review, we survey and summarize the main features of the different types of live bacterial vectors and discussed the clinical applications in the field of vaccinology. In addition, different approaches for using live attenuated bacterial vectors for anti-cancer therapy is discussed, and some promising pre-clinical and clinical studies in this field are outlined.
Collapse
Affiliation(s)
- Ivan Y C Lin
- School of Applied Sciences, RMIT University, Plenty Road, Bundoora VIC-3083, Australia.
| | - Thi Thu Hao Van
- School of Applied Sciences, RMIT University, Plenty Road, Bundoora VIC-3083, Australia.
| | - Peter M Smooker
- School of Applied Sciences, RMIT University, Plenty Road, Bundoora VIC-3083, Australia.
| |
Collapse
|
34
|
Schlößer HA, Drebber U, Kloth M, Thelen M, Rothschild SI, Haase S, Garcia-Marquez M, Wennhold K, Berlth F, Urbanski A, Alakus H, Schauss A, Shimabukuro-Vornhagen A, Theurich S, Warnecke-Ebertz U, Stippel DL, Zippelius A, Büttner R, Hallek M, Hölscher AH, Zander T, Mönig SP, von Bergwelt-Baildon M. Immune checkpoints programmed death 1 ligand 1 and cytotoxic T lymphocyte associated molecule 4 in gastric adenocarcinoma. Oncoimmunology 2015; 5:e1100789. [PMID: 27467911 DOI: 10.1080/2162402x.2015.1100789] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/14/2015] [Accepted: 09/22/2015] [Indexed: 12/14/2022] Open
Abstract
Remarkable efficacy of immune checkpoint inhibition has been reported for several types of solid tumors and early studies in gastric adenocarcinoma are promising. A detailed knowledge about the natural biology of immune checkpoints in gastric adenocarcinoma is essential for clinical and translational evaluation of these drugs. This study is a comprehensive analysis of cytotoxic T lymphocyte associated molecule 4 (CTLA-4) and programmed death 1 ligand 1 (PD-L1) expression in gastric adenocarcinoma. PD-L1 and CTLA-4 were stained on tumor sections of 127 Caucasian patients with gastric adenocarcinoma by immunohistochemistry (IHC) and somatic mutation profiling was performed using targeted next-generation sequencing. Expression of PD-L1 and CTLA-4 on lymphocytes in tumor sections, tumor-draining lymph nodes (TDLN) and peripheral blood were studied by flow-cytometry and immune-fluorescence microscopy in an additional cohort. PD-L1 and CTLA-4 were expressed in 44.9% (57/127) and 86.6% (110/127) of the analyzed gastric adenocarcinoma samples, respectively. Positive tumor cell staining for PD-L1 or CTLA-4 was associated with inferior overall survival. Somatic mutational analysis did not reveal a correlation to expression of PD-L1 or CTLA-4 on tumor cells. Expression of PD-1 (52.2%), PD-L1 (42.2%) and CTLA-4 (1.6%) on tumor infiltrating T cells was significantly elevated compared to peripheral blood. Of note, PD-1 and PD-L1 were expressed far higher by tumor-infiltrating lymphocytes than CTLA-4. In conclusion, specific immune checkpoint-inhibitors should be evaluated in this disease and the combination with molecular targeted therapies might be of benefit. An extensive immune monitoring should accompany these studies to better understand their mode of action in the tumor microenvironment.
Collapse
Affiliation(s)
- Hans A Schlößer
- Department of General, Visceral and Cancer Surgery, University of Cologne, Germany; Cologne Interventional Immunology, University of Cologne, Germany; Gastrointestinal Cancer Group Cologne, University of Cologne, Germany
| | - Uta Drebber
- Institute of Pathology, University of Cologne , Germany
| | - Michael Kloth
- Gastrointestinal Cancer Group Cologne, University of Cologne, Germany; Institute of Pathology, University of Cologne, Germany
| | - Martin Thelen
- Cologne Interventional Immunology, University of Cologne , Germany
| | - Sacha I Rothschild
- Cologne Interventional Immunology, University of Cologne, Germany; Department of Internal Medicine I, University of Cologne, Germany; Department of Internal Medicine, Medical Oncology, University Hospital Basel, Switzerland
| | - Simon Haase
- Department of General, Visceral and Cancer Surgery, University of Cologne , Germany
| | - Maria Garcia-Marquez
- Cologne Interventional Immunology, University of Cologne, Germany; Department of Internal Medicine I, University of Cologne, Germany
| | - Kerstin Wennhold
- Cologne Interventional Immunology, University of Cologne, Germany; Department of Internal Medicine I, University of Cologne, Germany
| | - Felix Berlth
- Department of General, Visceral and Cancer Surgery, University of Cologne , Germany
| | - Alexander Urbanski
- Department of General, Visceral and Cancer Surgery, University of Cologne , Germany
| | - Hakan Alakus
- Department of General, Visceral and Cancer Surgery, University of Cologne, Germany; Gastrointestinal Cancer Group Cologne, University of Cologne, Germany
| | - Astrid Schauss
- Cluster of Excellence in Aging-Associated Disease, Core Facility Imaging, University of Cologne , Germany
| | - Alexander Shimabukuro-Vornhagen
- Cologne Interventional Immunology, University of Cologne, Germany; Department of Internal Medicine I, University of Cologne, Germany
| | - Sebastian Theurich
- Cologne Interventional Immunology, University of Cologne, Germany; Department of Internal Medicine I, University of Cologne, Germany; Max-Planck-Institute for Metabolism Research, Cologne, Germany
| | - Ute Warnecke-Ebertz
- Department of General, Visceral and Cancer Surgery, University of Cologne , Germany
| | - Dirk L Stippel
- Department of General, Visceral and Cancer Surgery, University of Cologne , Germany
| | - Alfred Zippelius
- Department of Internal Medicine, Medical Oncology, University Hospital Basel , Switzerland
| | | | - Michael Hallek
- Department of Internal Medicine I, University of Cologne , Germany
| | - Arnulf H Hölscher
- Department of General, Visceral and Cancer Surgery, University of Cologne , Germany
| | - Thomas Zander
- Gastrointestinal Cancer Group Cologne, University of Cologne, Germany; Department of Internal Medicine I, University of Cologne, Germany
| | - Stefan P Mönig
- Department of General, Visceral and Cancer Surgery, University of Cologne, Germany; Gastrointestinal Cancer Group Cologne, University of Cologne, Germany
| | - Michael von Bergwelt-Baildon
- Cologne Interventional Immunology, University of Cologne, Germany; Department of Internal Medicine I, University of Cologne, Germany
| |
Collapse
|
35
|
Cervical cancer 2015 and beyond: a focus on innovative treatments and attention to survivorship. Clin Ther 2015; 37:6-8. [PMID: 25592087 DOI: 10.1016/j.clinthera.2014.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 12/04/2014] [Indexed: 11/20/2022]
|
36
|
Bae J, Keskin DB, Cowens K, Lee AH, Dranoff G, Munshi NC, Anderson KC. Lenalidomide Polarizes Th1-specific Anti-tumor Immune Response and Expands XBP1 Antigen-Specific Central Memory CD3 +CD8 + T cells against Various Solid Tumors. ACTA ACUST UNITED AC 2015; 3. [PMID: 27668268 PMCID: PMC5032910 DOI: 10.4172/2329-6917.1000178] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Introduction Effective combination immunotherapeutic strategies may be required to enhance effector cells’ anti-tumor activities and improve clinical outcomes. Methods XBP1 antigen-specific cytotoxic T lymphocytes (XBP1-CTL) generated using immunogenic heteroclitic XBP1 US184-192 (YISPWILAV) and XBP1 SP367-375 (YLFPQLISV) peptides or various solid tumor cells over-expressing XBP1 target antigen were evaluated, either alone or in combination with lenalidomide, for phenotype and immune functional activity. Results Lenalidomide treatment of XBP1-CTL increased the proportion of CD45RO+ memory CD3+CD8+ T cells, but not the total CD3+CD8+ T cells. Lenalidomide upregulated critical T cell activation markers and costimulatory molecules (CD28, CD38, CD40L, CD69, ICOS), especially within the central memory CTL subset of XBP1-CTL, while decreasing TCRαβ and T cell checkpoint blockade (CTLA-4, PD-1). Lenalidomide increased the anti-tumor activities of XBP1-CTL memory subsets, which were associated with expression of Th1 transcriptional regulators (T-bet, Eomes) and Akt activation, thereby resulting in enhanced IFN-γ production, granzyme B upregulation and specific CD28/CD38-positive and CTLA-4/PD-1-negative cell proliferation. Conclusions These studies suggest the potential benefit of lenalidomide treatment to boost anti-tumor activities of XBP1-specific CTL against a variety of solid tumors and enhance response to an XBP1-directing cancer vaccine regime.
Collapse
Affiliation(s)
- Jooeun Bae
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Derin B Keskin
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Kristen Cowens
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Ann-Hwee Lee
- Weill Cornell Medical College, New York, NY, USA
| | - Glen Dranoff
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Nikhil C Munshi
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Kenneth C Anderson
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|