151
|
Dorhoi A, Kotzé LA, Berzofsky JA, Sui Y, Gabrilovich DI, Garg A, Hafner R, Khader SA, Schaible UE, Kaufmann SH, Walzl G, Lutz MB, Mahon RN, Ostrand-Rosenberg S, Bishai W, du Plessis N. Therapies for tuberculosis and AIDS: myeloid-derived suppressor cells in focus. J Clin Invest 2021; 130:2789-2799. [PMID: 32420917 DOI: 10.1172/jci136288] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The critical role of suppressive myeloid cells in immune regulation has come to the forefront in cancer research, with myeloid-derived suppressor cells (MDSCs) as a main oncology immunotherapeutic target. Recent improvement and standardization of criteria classifying tumor-induced MDSCs have led to unified descriptions and also promoted MDSC research in tuberculosis (TB) and AIDS. Despite convincing evidence on the induction of MDSCs by pathogen-derived molecules and inflammatory mediators in TB and AIDS, very little attention has been given to their therapeutic modulation or roles in vaccination in these diseases. Clinical manifestations in TB are consequences of complex host-pathogen interactions and are substantially affected by HIV infection. Here we summarize the current understanding and knowledge gaps regarding the role of MDSCs in HIV and Mycobacterium tuberculosis (co)infections. We discuss key scientific priorities to enable application of this knowledge to the development of novel strategies to improve vaccine efficacy and/or implementation of enhanced treatment approaches. Building on recent findings and potential for cross-fertilization between oncology and infection biology, we highlight current challenges and untapped opportunities for translating new advances in MDSC research into clinical applications for TB and AIDS.
Collapse
Affiliation(s)
- Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institute, Greifswald-Insel Riems, Germany.,Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| | - Leigh A Kotzé
- Centre for Tuberculosis Research, South African Medical Research Council, Cape Town, South Africa.,DST-NRF Centre of Excellence for Biomedical Tuberculosis Research (CBTBR) and.,Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | | | - Ankita Garg
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Richard Hafner
- Division of AIDS, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ulrich E Schaible
- Cellular Microbiology, Priority Program Infections.,Thematic Translation Unit Tuberculosis, German Center for Infection Research, and.,Leibniz Research Alliance INFECTIONS'21, Research Center Borstel, Borstel, Germany
| | - Stefan He Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany.,Hagler Institute for Advanced Study, Texas A&M University, College Station, Texas, USA
| | - Gerhard Walzl
- Centre for Tuberculosis Research, South African Medical Research Council, Cape Town, South Africa.,DST-NRF Centre of Excellence for Biomedical Tuberculosis Research (CBTBR) and.,Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Manfred B Lutz
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Robert N Mahon
- Division of AIDS, Columbus Technologies & Services Inc., Contractor to National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Suzanne Ostrand-Rosenberg
- Department of Pathology and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - William Bishai
- Center for Tuberculosis Research, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Nelita du Plessis
- Centre for Tuberculosis Research, South African Medical Research Council, Cape Town, South Africa.,DST-NRF Centre of Excellence for Biomedical Tuberculosis Research (CBTBR) and.,Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
152
|
Ayuso JM, Rehman S, Virumbrales-Munoz M, McMinn PH, Geiger P, Fitzgerald C, Heaster T, Skala MC, Beebe DJ. Microfluidic tumor-on-a-chip model to evaluate the role of tumor environmental stress on NK cell exhaustion. SCIENCE ADVANCES 2021; 7:7/8/eabc2331. [PMID: 33597234 PMCID: PMC7888951 DOI: 10.1126/sciadv.abc2331] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 12/28/2020] [Indexed: 05/09/2023]
Abstract
Solid tumors generate a suppressive environment that imposes an overwhelming burden on the immune system. Nutrient depletion, waste product accumulation, hypoxia, and pH acidification severely compromise the capacity of effector immune cells such as T and natural killer (NK) cells to destroy cancer cells. However, the specific molecular mechanisms driving immune suppression, as well as the capacity of immune cells to adapt to the suppressive environment, are not completely understood. Thus, here, we used an in vitro microfluidic tumor-on-a-chip platform to evaluate how NK cells respond to the tumor-induced suppressive environment. The results demonstrated that the suppressive environment created by the tumor gradually eroded NK cell cytotoxic capacity, leading to compromised NK cell surveillance and tumor tolerance. Further, NK cell exhaustion persisted for an extended period of time after removing NK cells from the microfluidic platform. Last, the addition of checkpoint inhibitors and immunomodulatory agents alleviated NK cell exhaustion.
Collapse
Affiliation(s)
- Jose M Ayuso
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA.
| | - Shujah Rehman
- Morgridge Institute for Research, 330 N Orchard Street, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | | | - Patrick H McMinn
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Peter Geiger
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Cate Fitzgerald
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Tiffany Heaster
- Morgridge Institute for Research, 330 N Orchard Street, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
| | - Melissa C Skala
- Morgridge Institute for Research, 330 N Orchard Street, Madison, WI, USA.
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - David J Beebe
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA.
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
153
|
Ma C, Zhang Q, Greten TF. MDSCs in liver cancer: A critical tumor-promoting player and a potential therapeutic target. Cell Immunol 2021; 361:104295. [PMID: 33508529 DOI: 10.1016/j.cellimm.2021.104295] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/23/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
Abstract
Liver cancer is a leading cause of cancer deaths worldwide. Hepatocellular carcinoma (~75-85%) and cholangiocarcinoma (~10-15%) account for the majority of primary liver malignancies. Patients with primary liver cancer are often diagnosed with unresectable diseases and do not respond well to current therapies. The liver is also a common site of metastasis. Liver metastasis is difficult to treat, and the prognosis is poor. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells with immunosuppressive activity. MDSCs are an important component of the tumor microenvironment and promote tumor progression through various mechanisms. MDSCs expand in both liver cancer patients and mouse liver cancer models. Importantly, MDSCs correlate with poor clinical outcomes for liver cancer patients. The tumor-promoting functions of MDSCs have also been shown in mouse liver cancer models. All these studies suggest that targeting MDSCs can potentially benefit liver cancer treatment. This review summarizes the current findings of MDSC regulation in liver cancer and related disease conditions.
Collapse
Affiliation(s)
- Chi Ma
- Gastrointestinal Malignancy Section, Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qianfei Zhang
- Gastrointestinal Malignancy Section, Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; NCI-CCR Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
154
|
Bansal D, Reimers MA, Knoche EM, Pachynski RK. Immunotherapy and Immunotherapy Combinations in Metastatic Castration-Resistant Prostate Cancer. Cancers (Basel) 2021; 13:cancers13020334. [PMID: 33477569 PMCID: PMC7831137 DOI: 10.3390/cancers13020334] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/01/2021] [Accepted: 01/14/2021] [Indexed: 12/21/2022] Open
Abstract
Although most prostate cancers are localized, and the majority are curable, recurrences occur in approximately 35% of men. Among patients with prostate-specific antigen (PSA) recurrence and PSA doubling time (PSADT) less than 15 months after radical prostatectomy, prostate cancer accounted for approximately 90% of the deaths by 15 years after recurrence. An immunosuppressive tumor microenvironment (TME) and impaired cellular immunity are likely largely responsible for the limited utility of checkpoint inhibitors (CPIs) in advanced prostate cancer compared with other tumor types. Thus, for immunologically "cold" malignancies such as prostate cancer, clinical trial development has pivoted towards novel approaches to enhance immune responses. Numerous clinical trials are currently evaluating combination immunomodulatory strategies incorporating vaccine-based therapies, checkpoint inhibitors, and chimeric antigen receptor (CAR) T cells. Other trials evaluate the efficacy and safety of these immunomodulatory agents' combinations with standard approaches such as androgen deprivation therapy (ADT), taxane-based chemotherapy, radiotherapy, and targeted therapies such as tyrosine kinase inhibitors (TKI) and poly ADP ribose polymerase (PARP) inhibitors. Here, we will review promising immunotherapies in development and ongoing trials for metastatic castration-resistant prostate cancer (mCRPC). These novel trials will build on past experiences and promise to usher a new era to treat patients with mCRPC.
Collapse
|
155
|
Sanseviero E, Kim R, Gabrilovich DI. Isolation and Phenotyping of Splenic Myeloid-Derived Suppressor Cells in Murine Cancer Models. Methods Mol Biol 2021; 2236:19-28. [PMID: 33237537 DOI: 10.1007/978-1-0716-1060-2_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) are immunosuppressive myeloid cells that accumulate in tumor sites and peripheral lymphoid organs such as the spleen. In murine cancer models, the spleen is a major reservoir for MDSC, representing an easily accessible tissue from which to isolate high numbers of these cell population for downstream applications. Here we describe an efficient method to phenotype as well as to isolate and assess the functionality of murine splenic MDSC.
Collapse
Affiliation(s)
| | - Rina Kim
- The Wistar Institute, Philadelphia, PA, USA
| | | |
Collapse
|
156
|
Xing Z, Zuo Z, Hu D, Zheng X, Wang X, Yuan L, Zhou L, Qi F, Yao Z. Influenza vaccine combined with moderate-dose PD1 blockade reduces amyloid-β accumulation and improves cognition in APP/PS1 mice. Brain Behav Immun 2021; 91:128-141. [PMID: 32956831 DOI: 10.1016/j.bbi.2020.09.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
Immune dysfunction is implicated in Alzheimer's disease (AD), whereas systemic immune modulation may be neuroprotective. Our previous results have indicated immune challenge with Bacillus Calmette-Guerin attenuates AD pathology in animal models by boosting the systemic immune system. Similarly, independent studies have shown that boosting systemic immune system, by blocking PD-1 checkpoint pathway, modifies AD. Here we hypothesized that influenza vaccine would potentiate function of moderate dose anti-PD-1 and therefore combining them might allow reducing the dose of PD-1 antibody needed to modify the disease. We found that moderate-dose PD-1 in combination with influenza vaccine effectively attenuated cognitive deficit and prevented amyloid-β pathology build-up in APP/PS1 mice in a mechanism dependent on recruitment of peripheral monocyte-derived macrophages into the brain. Eliminating peripheral macrophages abrogated the beneficial effect. Moreover, by comparing CD11b+ compartments in the mouse parenchyma, we observed an elevated subset of Ly6C+ microglia-like cells, which are reportedly derived from peripheral monocytes. In addition, myeloid-derived suppressor cells are strongly elevated in the transgenic model used and normalized by combination treatment, indicating restoration of brain immune homeostasis. Overall, our results suggest that revitalizing brain immunity by combining IV with moderate-dose PD-1 inhibition may represent a therapeutic immunotherapy for AD.
Collapse
Affiliation(s)
- Zhiwei Xing
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Zejie Zuo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Dandan Hu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, PR China
| | - Xiaona Zheng
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Xiao Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Lifang Yuan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Lihua Zhou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Department of Anatomy, Sun Yat-sen University, School of Medicine, Guangzhou 510089, PR China.
| | - Fangfang Qi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China.
| | - Zhibin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China.
| |
Collapse
|
157
|
Progress of exosomes in the diagnosis and treatment of lung cancer. Biomed Pharmacother 2020; 134:111111. [PMID: 33352449 DOI: 10.1016/j.biopha.2020.111111] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/28/2020] [Accepted: 12/04/2020] [Indexed: 12/15/2022] Open
Abstract
The incidence and mortality of lung cancer account for first place all over the world. Lung cancer lacks early diagnostic biomarkers; lung cancer patients are usually diagnosed in both middle and advanced stages and have poor treatment outcomes. It is more important to find the first diagnostic tools for lung cancer with high specificity and sensitivity. Besides, exosomes are usually nanometer-sized bi-layered lipid vesicles formed and produced by various types of cells. As one of the main modes of intercellular communication, they can deliver multiple functional biomolecules, such as DNA, microRNAs, messenger RNA (mRNA), long non-coding RNA, and proteins, and the events as mentioned above affects different physiological processes of recipient cells. It has been reported that exosomes are involved in different types of cancer, including lung cancer. Various studies proved that exosomes are involved in multiple cancer processes such as cell proliferation, metastasis, epithelial-mesenchymal transition (EMT), angiogenesis, and the tumor microenvironment in lung cancer. Tumor-derived exosomes (TEX) contain a variety of stimulatory and inhibitory factors involved in regulating immune response, which can affect the tumor microenvironment (TME) and thus participate in the formation and progression of lung cancer. This review's primary purpose to review the latest research progress of exosomes in diagnosing and treating lung cancer.
Collapse
|
158
|
Blood Myeloid-Derived Suppressor Cells Correlate with Neutrophil-to-Lymphocyte Ratio and Overall Survival in Metastatic Urothelial Carcinoma. Target Oncol 2020; 15:211-220. [PMID: 32207064 DOI: 10.1007/s11523-020-00707-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Myeloid-derived suppressor cells (MDSCs) were linked to pathologic stage in bladder urothelial carcinoma (UC). Neutrophil lymphocyte ratio (NLR) is an inflammatory biomarker with a prognostic role in metastatic (m)UC. OBJECTIVE We hypothesized that MDSC levels correlate with NLR and overall survival (OS) in mUC. PATIENTS AND METHODS MDSCs were measured in blood samples from patients with mUC in fresh unfractionated whole blood (WB) and peripheral blood mononuclear cells (PBMC) by flow cytometry and defined as LinloCD33+/HLADR- (Total MDSC). MDSC subsets were defined as polymorphonuclear (PMN-MDSC: CD15+/CD14-), monocytic (M-MDSC: CD15-/CD14+), and uncommitted (UNC-MDSC: CD15-/CD14-). MDSC populations were presented as a percentage of live nucleated blood cells. Spearman's rank correlation assessed correlations between MDSC and NLR. Kaplan-Meier curves and log-rank test estimated OS from the time of MDSC collection to last follow-up or date of death. RESULTS Of the 76 patients, 78% were men and 43% were never smokers with a median age of 69 years (range 31-83); 72% had pure UC and 76% had lower tract UC. Prior therapies included intravesical therapy (22%), neoadjuvant chemotherapy (30%), cystectomy or nephroureterectomy (55%). Median follow-up for all patients was 12 months (0.6-36.5). PMN-MDSC was the predominant subset in WB and PBMC. There was significant correlation between individual MDSC subsets in WB and PBMC (p ≤ 0.001). Both WB UNC-MDSC/PMN-MDSC ratios (rho = - 0.27, p = 0.03) and PBMC UNC-MDSC/PMN-MDSC (rho = - 0.28, p = 0.02) were negatively correlated with NLR. Median OS was 17.7 months (95% CI: 11.0-NE). Overall 1-year and 3-year survival rates were 0.60 (95% CI 0.49-0.73) and 0.15 (95% CI 0.03-0.67), respectively. Higher WB UNC-MDSC levels (HR 3.78, p = 0.0022) and higher NLR (HR 2.6, p = 0.0179) were associated with shorter OS. CONCLUSIONS Specific MDSC subsets correlate with NLR. Higher WB UNC-MDSC levels and higher NLR were negative prognostic factors. Given the feasibility of serial blood draws, dynamic assessment of MDSC over time and further validation with longer follow-up are warranted.
Collapse
|
159
|
Xu WL, Shi BJ, Li SL, Yu FX, Guo LN, Li M, Hu ZG, Li GX, Zhou H. Targeted inhibition of myeloid-derived suppressor cells in the tumor microenvironment by low-dose doxorubicin to improve immune efficacy in murine neuroblastoma. Chin Med J (Engl) 2020; 134:334-343. [PMID: 33278092 PMCID: PMC7846436 DOI: 10.1097/cm9.0000000000001234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND High agglomeration of myeloid-derived suppressor cells (MDSCs) in neuroblastoma (NB) impeded therapeutic effects. This study aimed to investigate the role and mechanism of targeted inhibition of MDSCs by low-dose doxorubicin (DOX) to enhance immune efficacy in NB. METHODS Bagg albino (BALB/c) mice were used as tumor-bearing mouse models by injecting Neuro-2a cells, and MDSCs were eliminated by DOX or dopamine (DA) administration. Tumor-bearing mice were randomly divided into 2.5 mg/kg DOX, 5.0 mg/kg DOX, 50.0 mg/kg DA, and control groups (n = 20). The optimal drug and its concentration for MDSC inhibition were selected according to tumor inhibition. NB antigen-specific cytotoxic T cells (CTLs) were prepared. Tumor-bearing mice were randomly divided into DOX, CTL, anti-ganglioside (GD2), DOX+CTL, DOX+anti-GD2, and control groups. Following low-dose DOX administration, immunotherapy was applied. The levels of human leukocyte antigen (HLA)-I, CD8, interleukin (IL)-2 and interferon (IFN)-γ in peripheral blood, CTLs, T-helper 1 (Thl)/Th2 cytokines, perforin, granzyme and tumor growth were compared among the groups. The Wilcoxon two-sample test and repeated-measures analysis of variance were used to analyze results. RESULTS The slowest tumor growth (F = 6.095, P = 0.018) and strongest MDSC inhibition (F = 14.632, P = 0.001) were observed in 2.5 mg/kg DOX group. Proliferation of T cells was increased (F = 448.721, P < 0.001) and then decreased (F = 2.047, P = 0.186). After low-dose DOX administration, HLA-I (F = 222.489), CD8 (F = 271.686), Thl/Th2 cytokines, CD4+ and CD8+ lymphocytes, granzyme (F = 2376.475) and perforin (F = 488.531) in tumor, IL-2 (F = 62.951) and IFN-γ (F = 240.709) in peripheral blood of each immunotherapy group were all higher compared with the control group (all of P values < 0.05). The most significant increases in the aforementioned indexes and the most notable tumor growth inhibition were observed in DOX+anti-GD2 and DOX+CTL groups. CONCLUSIONS Low-dose DOX can be used as a potent immunomodulatory agent that selectively impairs MDSC-induced immunosuppression, thereby fostering immune efficacy in NB.
Collapse
Affiliation(s)
- Wei-Li Xu
- Department of Pediatric Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Bao-Jun Shi
- Department of Pediatric Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Suo-Lin Li
- Department of Pediatric Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Feng-Xue Yu
- Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Li-Na Guo
- Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Meng Li
- Department of Pediatric Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Zhi-Gang Hu
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Gui-Xin Li
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Hui Zhou
- Department of Pediatric Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
160
|
Wu X, Zhu D, Tian J, Tang X, Guo H, Ma J, Xu H, Wang S. Granulocytic Myeloid-Derived Suppressor Cell Exosomal Prostaglandin E2 Ameliorates Collagen-Induced Arthritis by Enhancing IL-10 + B Cells. Front Immunol 2020; 11:588500. [PMID: 33329572 PMCID: PMC7734343 DOI: 10.3389/fimmu.2020.588500] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/02/2020] [Indexed: 01/22/2023] Open
Abstract
The results of recent studies have shown that granulocytic-myeloid derived suppressor cells (G-MDSCs) can secrete exosomes that transport various biologically active molecules with regulatory effects on immune cells. However, their roles in autoimmune diseases such as rheumatoid arthritis remain to be further elucidated. In the present study, we investigated the influence of exosomes from G-MDSCs on the humoral immune response in murine collagen-induced arthritis (CIA). G-MDSCs exosomes-treated mice showed lower arthritis index values and decreased inflammatory cell infiltration. Treatment with G-MDSCs exosomes promoted splenic B cells to secrete IL-10 both in vivo and in vitro. In addition, a decrease in the proportion of plasma cells and follicular helper T cells was observed in drainage lymph nodes from G-MDSCs exosomes-treated mice. Moreover, lower serum levels of IgG were detected in G-MDSCs exosomes-treated mice, indicating an alteration of the humoral environment. Mechanistic studies showed that exosomal prostaglandin E2 (PGE2) produced by G-MDSCs upregulated the phosphorylation levels of GSK-3β and CREB, which play a key role in the production of IL-10+ B cells. Taken together, our findings demonstrated that G-MDSC exosomal PGE2 attenuates CIA in mice by promoting the generation of IL-10+ Breg cells.
Collapse
Affiliation(s)
- Xinyu Wu
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Dongwei Zhu
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jie Tian
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xinyi Tang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
| | - Hongye Guo
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jie Ma
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Huaxi Xu
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
161
|
Abstract
Initially identified as a T lymphocyte-elicited inhibitor of macrophage motility, macrophage migration inhibitory factor (MIF) has since been found to be expressed by nearly every immune cell type examined and overexpressed in most solid and hematogenous malignant cancers. It is localized to both extracellular and intracellular compartments and physically interacts with more than a dozen different cell surface and intracellular proteins. Although classically associated with and characterized as a mediator of pro-inflammatory innate immune responses, more recent studies demonstrate that, in malignant disease settings, MIF contributes to anti-inflammatory, immune evasive, and immune tolerant phenotypes in both innate and adaptive immune cell types. This review will summarize the studies describing MIF in tumor-specific innate and adaptive immune responses and attempt to reconcile these various pleiotropic functions in normal physiology.
Collapse
Affiliation(s)
- Jordan T. Noe
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY, United States
- J.G. Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Robert A. Mitchell
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY, United States
- J.G. Brown Cancer Center, University of Louisville, Louisville, KY, United States
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, United States
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States
| |
Collapse
|
162
|
Liu BQ, Bao ZY, Zhu JY, Liu H. Fibrinogen-like protein 2 promotes the accumulation of myeloid-derived suppressor cells in the hepatocellular carcinoma tumor microenvironment. Oncol Lett 2020; 21:47. [PMID: 33281958 PMCID: PMC7709556 DOI: 10.3892/ol.2020.12308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
The tumor microenvironment in hepatocellular carcinoma can be classified into cellular and non-cellular components. Myeloid-derived suppressor cells (MDSCs) are cellular components of this microenvironment that serve an important role in the progression of hepatocellular carcinoma. Fibrinogen-like protein 2 (FGL2) has been demonstrated to promote tumor progression by regulating cellular components of the tumor microenvironment in various types of malignant tumor. The present study aimed to determine the expression of FGL2 in hepatocellular carcinoma and its effect on the tumor microenvironment in order to determine novel targets for liver cancer treatment. Immunohistochemistry and reverse transcription quantitative PCR were performed to determine the expression level of FGL2 and the correlation with surface markers of human MDSCs in hepatocellular carcinoma. Furthermore, a mouse hepatocellular carcinoma cell line overexpressing FGL2 was established by stable transfection of a lentivirus expressing FGL2. In addition, fresh bone marrow cells extracted from mouse femurs were in vitro cultured using conditioned medium derived from the cell line overexpressing FGL2. An orthotopic hepatocellular carcinoma mouse model was also established. The results demonstrated that FGL2 expression level in hepatocellular carcinoma tissues was closely associated with tumor size. FGL2 level was positively correlated with the expression level of the MDSC surface markers CD11b and CD33 in hepatocellular carcinoma. The in vitro results demonstrated that FGL2 could maintain the undifferentiated state of bone marrow cells, therefore promoting MDSC accumulation. Furthermore, in the orthotopic hepatocellular carcinoma mouse model, we observed that overexpression of FGL2 could promote tumor growth and significantly increase the number of MDSCs in the tumors and spleen. Taken together, these findings suggested that FGL2 may promote hepatocellular carcinoma tumor growth by promoting the accumulation of MDSCs in the tumor microenvironment.
Collapse
Affiliation(s)
- Bo-Qian Liu
- Department of Transplant and Hepatobilliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China.,Department of Anorectal Surgery, The People's Hospital of Liaoning Province, Shenyang, Liaoning 110000, P.R. China
| | - Zhi-Ye Bao
- Department of Transplant and Hepatobilliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Jia-Yi Zhu
- Department of Transplant and Hepatobilliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Hao Liu
- Department of Transplant and Hepatobilliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| |
Collapse
|
163
|
Drakes ML, Czerlanis CM, Stiff PJ. Immune Checkpoint Blockade in Gynecologic Cancers: State of Affairs. Cancers (Basel) 2020; 12:cancers12113301. [PMID: 33182298 PMCID: PMC7695253 DOI: 10.3390/cancers12113301] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Most endometrial cancer patients are diagnosed at an early stage, receive standard treatment, and survive well. Ovarian cancer has no specific symptoms and usually escapes diagnosis until the patient has advanced disease. This disease results in the highest number of deaths of gynecologic cancers. Current treatments for gynecologic cancers in the advanced stage are not sufficiently effective for good outcome in most patients. This review discusses two novel treatments, which are immune checkpoint inhibitor antibodies that block immune checkpoint molecules cytotoxic T lymphocyte associated protein-4 (CTLA-4) and programmed death-1 (PD-1) in patients. The antibody blocking of CTLA-4 or PD-1 alone is promising treatment for some categories of advanced disease endometrial cancer, but it has little effect against ovarian cancer. Our study primarily discusses the status of clinical trials for these two diseases and the biological parameters governing the different outcomes to these therapies. We also propose mechanisms whereby blocking CTLA-4 and PD-1 may be used in combination with other agents to give much better survival in advanced disease ovarian cancer patients. Abstract This review provides an update on the current use of immune checkpoint inhibitors (ICI) in female gynecologic cancers, and it addresses the potential of these agents to provide therapy options for disease management and long-term remission in advanced disease patients, where surgery, chemotherapy, and/or radiation fail to meet this goal. The topic of immune checkpoint inhibitors (ICI) blocking cytotoxic T lymphocyte associated protein-4 (CTLA-4) and the programmed death-1 (PD-1) axis has come to the forefront of translational medicine over the last decade for several malignancies. The text will focus primarily on a discussion of ovarian cancer, which is the most frequent cause of death of gynecologic cancers; endometrial cancer, which is the most often diagnosed gynecologic cancer; and cervical cancer, which is the third most common female gynecologic malignancy, all of which unfavorably alter the lives of many women. We will address the critical factors that regulate the outcome of these cancer types to ICI therapy, the ongoing clinical trials in this area, as well as the adverse immune responses that impact the outcome of patients given ICI regimens.
Collapse
|
164
|
Long noncoding RNA: a dazzling dancer in tumor immune microenvironment. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:231. [PMID: 33148302 PMCID: PMC7641842 DOI: 10.1186/s13046-020-01727-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Long noncoding RNAs (lncRNAs) are a class of endogenous, non-protein coding RNAs that are highly linked to various cellular functions and pathological process. Emerging evidence indicates that lncRNAs participate in crosstalk between tumor and stroma, and reprogramming of tumor immune microenvironment (TIME). TIME possesses distinct populations of myeloid cells and lymphocytes to influence the immune escape of cancer, the response to immunotherapy, and the survival of patients. However, hitherto, a comprehensive review aiming at relationship between lncRNAs and TIME is missing. In this review, we focus on the functional roles and molecular mechanisms of lncRNAs within the TIME. Furthermore, we discussed the potential immunotherapeutic strategies based on lncRNAs and their limitations.
Collapse
|
165
|
Abstract
The immune system plays an essential and central role in tumor cell differentiation, proliferation, angiogenesis, apoptosis, invasion, and metastasis. Over the past decade, cancer therapy has rapidly evolved from traditional approaches, such as surgery, chemotherapy, and radiotherapy, to revolutionary new treatment options with immunotherapy. This new era of cancer treatment options has now been clinically tested and applied to many forms of human malignancies, often with quite dramatic results. As we develop more effective combinations of cancer treatment, several agents have been recently investigated, putatively identified as anticancer agents, or immunostimulatory molecules. One such agent is metformin, originally developed as a fairly standard first-line therapy for patients with type-2 diabetes mellitus (T2DM). Given the underlying mechanisms of action, researchers began to examine the alternative functions and possible utility of metformin, finding that the cancer risk in patients with T2DM was reduced. It appears that metformin, at least in part, has an antitumor effect through activation of the 5' adenosine monophosphate-activated protein kinase (AMPK) signaling pathway. Moreover, numerous studies have demonstrated that metformin interferes with key immunopathological mechanisms that are involved in the pathological processes or associated with malignant progression. Such insights may shed light on further analyzing whether metformin enhances the effectiveness of the immunotherapy and overcomes the immunotherapy resistance in the patients. Herein, we provide a comprehensive review of the literature examining the impact of metformin upon the host immune system and cancer immunity.
Collapse
Affiliation(s)
- Ruixia Ma
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, China
| | - Bin Yi
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Adam I Riker
- Geaton and JoAnn DeCesaris Cancer Institute, Anne Arundel Medical Center, Luminis Health, Annapolis, MD, USA.
| | - Yaguang Xi
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| |
Collapse
|
166
|
Wang H, Li X, Dong G, Yan F, Zhang J, Shi H, Ning Z, Gao M, Cheng D, Ma Q, Wang C, Zhao M, Dai J, Li C, Li Z, Zhang H, Xiong H. Toll-like Receptor 4 Inhibitor TAK-242 Improves Fulminant Hepatitis by Regulating Accumulation of Myeloid-Derived Suppressor Cell. Inflammation 2020; 44:671-681. [PMID: 33083887 DOI: 10.1007/s10753-020-01366-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/16/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022]
Abstract
Fulminant hepatitis (FH) is an acute clinical disease with a poor prognosis and high mortality rate. The purpose of this study was to determine the protective effect of the Toll-like receptor 4 (TLR4) inhibitor TAK-242 on lipopolysaccharide (LPS)/D-galactosamine (D-GalN)-induced explosive hepatitis and explore in vivo and in vitro mechanisms. Mice were pretreated with TAK-242 for 3 h prior to LPS (10 μg/kg)/D-GalN (250 mg/kg) administration. Compared to the LPS/D-GalN group, the TAK-242 pretreatment group showed significantly prolonged survival, reduced serum alanine aminotransferase and aspartate aminotransferase levels, relieved oxidative stress, and reduced inflammatory interleukin (IL)-6, IL-12, and tumor necrosis factor-α levels. In addition, TAK-242 increased the accumulation of myeloid-derived suppressor cells (MDSCs). Next, mice were treated with an anti-Gr-1 antibody to deplete MDSCs, and adoptive transfer experiments were performed. We found that TAK-242 protected against FH by regulating MDSCs. In the in vitro studies, TAK-242 regulated the accumulation of MDSCs and promoted the release of immunosuppressive inflammatory cytokines. In addition, TAK-242 inhibited protein expression of nuclear factor-κB and mitogen-activated protein kinases. In summary, TAK-242 had a hepatoprotective effect against LPS/D-GalN-induced explosive hepatitis in mice. Its protective effect may be involved in suppressing inflammation, reducing oxidative stress, and increasing the proportion of MDSCs.
Collapse
Affiliation(s)
- Haiyan Wang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuehui Li
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guanjun Dong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Fenglian Yan
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Hui Shi
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Zhaochen Ning
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Min Gao
- Clinical Laboratory, Jining First People's Hospital, Shandong Province, Jining, 272011, China
| | - Dalei Cheng
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qun Ma
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Changying Wang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Mingsheng Zhao
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Jun Dai
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Chunxia Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Zhihua Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Hui Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China.
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China.
| |
Collapse
|
167
|
Connections between Metabolism and Epigenetic Modification in MDSCs. Int J Mol Sci 2020; 21:ijms21197356. [PMID: 33027968 PMCID: PMC7582655 DOI: 10.3390/ijms21197356] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are major immunosuppressive cells in the tumor microenvironment (TME). During the differentiation and development of MDSCs from myeloid progenitor cells, their functions are also affected by a series of regulatory factors in the TME, such as metabolic reprogramming, epigenetic modification, and cell signaling pathways. Additionally, there is a crosstalk between these regulatory factors. This review mainly introduces the metabolism (especially glucose metabolism) and significant epigenetic modification of MDSCs in the TME, and briefly introduces the connections between metabolism and epigenetic modification in MDSCs, in order to determine the further impact on the immunosuppressive effect of MDSCs, so as to serve as a more effective target for tumor therapy.
Collapse
|
168
|
Amin Yavari S, Castenmiller SM, van Strijp JAG, Croes M. Combating Implant Infections: Shifting Focus from Bacteria to Host. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002962. [PMID: 32914481 DOI: 10.1002/adma.202002962] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/28/2020] [Indexed: 05/06/2023]
Abstract
The widespread use of biomaterials to support or replace body parts is increasingly threatened by the risk of implant-associated infections. In the quest for finding novel anti-infective biomaterials, there generally has been a one-sided focus on biomaterials with direct antibacterial properties, which leads to excessive use of antibacterial agents, compromised host responses, and unpredictable effectiveness in vivo. This review sheds light on how host immunomodulation, rather than only targeting bacteria, can endow biomaterials with improved anti-infective properties. How antibacterial surface treatments are at risk to be undermined by biomaterial features that dysregulate the protection normally provided by critical immune cell subsets, namely, neutrophils and macrophages, is discussed. Accordingly, how the precise modification of biomaterial surface biophysical cues, or the incorporation of immunomodulatory drug delivery systems, can render biomaterials with the necessary immune-compatible and immune-protective properties to potentiate the host defense mechanisms is reviewed. Within this context, the protective role of host defense peptides, metallic particles, quorum sensing inhibitors, and therapeutic adjuvants is discussed. The highlighted immunomodulatory strategies may lay a foundation to develop anti-infective biomaterials, while mitigating the increasing threat of antibacterial drug resistance.
Collapse
Affiliation(s)
- Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Suzanne M Castenmiller
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Michiel Croes
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| |
Collapse
|
169
|
Construction of chlorogenic acid-containing liposomes with prolonged antitumor immunity based on T cell regulation. SCIENCE CHINA-LIFE SCIENCES 2020; 64:1097-1115. [PMID: 33009993 DOI: 10.1007/s11427-020-1739-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022]
Abstract
As a potential cancer immunotherapeutic agent, chlorogenic acid (CHA) has entered phase II clinical trials in China as a lyophilized powder formulation for treating glioma. However, the in vivo instability of CHA necessitates daily intramuscular injections, resulting in patient noncompliance. In this study, CHA-phospholipid complex (PC)-containing PEGylated liposomes (CHA-PC PEG-Lipo, named as CPPL), with CHA-PC as the drug intermediate, were prepared to lower the administration frequency. CPPL demonstrated excellent physicochemical properties, enhanced tumor accumulation, and inhibited tumor growth even when the administration interval was prolonged to 4 days when compared to a CHA solution and CHA-PC loaded liposomes (CHA-PC Lipo, labeled as CPL), both of which only demonstrated antitumor efficacy with once-daily administration. Further evaluation of the in vivo antitumor immune mechanism suggested that the extended antitumor immune efficacy of CPPL could be attributed to its distinct immune-stimulating mechanism when compared with CHA solution and CPL, such as stimulating both CD4+ and CD8+ T cell infiltration, inhibiting myeloid-derived suppressor cell expression, reducing the expression of Th2 related factors, and notably, increasing the memory T cells in tumor tissues. This CHA-containing formulation could reduce the frequency of in vivo CHA administration during cancer treatment via T cells, especially memory T cell regulation.
Collapse
|
170
|
Cancer-Associated Fibroblasts: Versatile Players in the Tumor Microenvironment. Cancers (Basel) 2020; 12:cancers12092652. [PMID: 32957515 PMCID: PMC7564346 DOI: 10.3390/cancers12092652] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/11/2020] [Accepted: 09/13/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Cancer-associated fibroblasts (CAFs) are key players in the tumor microenvironment. They are responsible for potentiating growth and metastasis through versatile functions, including maintenance of the extracellular matrix, blood vessel formation, modulation of tumor metabolism, suppression of antitumor immunity, and promotion of chemotherapy resistance. As such, CAFs are associated with poor prognosis and have emerged as a focus of anticancer research. In this review, we discuss the origins of CAFs, their heterogenous subtypes and their properties. We then detail the current state of preclinical and clinical research targeting CAF activities. We believe the limited efficacy of current cancer therapeutic approaches is driven by an incomplete understanding of CAF functions and by a nonstandardized CAF classification system. Therefore, we suggest a unified CAF classification based on specific functions to develop a new class of therapies that will focus on targeting the pro-tumorigenic properties of CAFs during tumor progression. Abstract Cancer-associated fibroblasts (CAFs) are indispensable architects of the tumor microenvironment. They perform the essential functions of extracellular matrix deposition, stromal remodeling, tumor vasculature modulation, modification of tumor metabolism, and participation in crosstalk between cancer and immune cells. In this review, we discuss our current understanding of the principal differences between normal fibroblasts and CAFs, the origin of CAFs, their functions, and ultimately, highlight the intimate connection of CAFs to virtually all of the hallmarks of cancer. We address the remarkable degree of functional diversity and phenotypic plasticity displayed by CAFs and strive to stratify CAF biology among different tumor types into practical functional groups. Finally, we summarize the status of recent and ongoing trials of CAF-directed therapies and contend that the paucity of trials resulting in Food and Drug Administration (FDA) approvals thus far is a consequence of the failure to identify targets exclusive of pro-tumorigenic CAF phenotypes that are mechanistically linked to specific CAF functions. We believe that the development of a unified CAF nomenclature, the standardization of functional assays to assess the loss-of-function of CAF properties, and the establishment of rigorous definitions of CAF subpopulations and their mechanistic functions in cancer progression will be crucial to fully realize the promise of CAF-targeted therapies.
Collapse
|
171
|
Targeting Myeloid-Derived Suppressor Cells in Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12092626. [PMID: 32942545 PMCID: PMC7564060 DOI: 10.3390/cancers12092626] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Myeloid-Derived Suppressor Cells (MDSCs) have been regarded as the main promoters of cancer development in recent years. They can protect tumor cells from being eliminated by neutralizing the anti-tumor response mediated by T cells, macrophages and dendritic cells (DCs). Therefore, different treatment methods targeting MDSCs, including chemotherapy, radiotherapy and immunotherapy, have been developed and proven to effectively inhibit tumor expansion. Herein, we summarize the immunosuppressive role of MDSCs in the tumor microenvironment and some effective treatments targeting MDSCs, and discuss the differences between different therapies. Abstract Myeloid-derived suppressor cells (MDSCs), which are activated under pathological conditions, are a group of heterogeneous immature myeloid cells. MDSCs have potent capacities to support tumor growth via inhibition of the antitumoral immune response and/or the induction of immunosuppressive cells. In addition, multiple studies have demonstrated that MDSCs provide potential therapeutic targets for the elimination of immunosuppressive functions and the inhibition of tumor growth. The combination of targeting MDSCs and other therapeutic approaches has also demonstrated powerful antitumor effects. In this review, we summarize the characteristics of MDSCs in the tumor microenvironment (TME) and current strategies of cancer treatment by targeting MDSCs.
Collapse
|
172
|
Cassetta L, Bruderek K, Skrzeczynska-Moncznik J, Osiecka O, Hu X, Rundgren IM, Lin A, Santegoets K, Horzum U, Godinho-Santos A, Zelinskyy G, Garcia-Tellez T, Bjelica S, Taciak B, Kittang AO, Höing B, Lang S, Dixon M, Müller V, Utikal JS, Karakoç D, Yilmaz KB, Górka E, Bodnar L, Anastasiou OE, Bourgeois C, Badura R, Kapinska-Mrowiecka M, Gotic M, Ter Laan M, Kers-Rebel E, Król M, Santibañez JF, Müller-Trutwin M, Dittmer U, de Sousa AE, Esendağlı G, Adema G, Loré K, Ersvær E, Umansky V, Pollard JW, Cichy J, Brandau S. Differential expansion of circulating human MDSC subsets in patients with cancer, infection and inflammation. J Immunother Cancer 2020; 8:jitc-2020-001223. [PMID: 32907925 PMCID: PMC7481096 DOI: 10.1136/jitc-2020-001223] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2020] [Indexed: 01/25/2023] Open
Abstract
Background Myeloid-derived suppressor cells (MDSC) are a functional myeloid cell subset that includes myeloid cells with immune suppressive properties. The presence of MDSC has been reported in the peripheral blood of patients with several malignant and non-malignant diseases. So far, direct comparison of MDSC across different diseases and Centers is hindered by technical pitfalls and a lack of standardized methodology. To overcome this issue, we formed a network through the COST Action Mye-EUNITER (www.mye-euniter.eu) with the goal to standardize and facilitate the comparative analysis of human circulating MDSC in cancer, inflammation and infection. In this manuscript, we present the results of the multicenter study Mye-EUNITER MDSC Monitoring Initiative, that involved 13 laboratories and compared circulating MDSC subsets across multiple diseases, using a common protocol for the isolation, identification and characterization of these cells. Methods We developed, tested, executed and optimized a standard operating procedure for the isolation and immunophenotyping of MDSC using blood from healthy donors. We applied this procedure to the blood of almost 400 patients and controls with different solid tumors and non-malignant diseases. The latter included viral infections such as HIV and hepatitis B virus, but also psoriasis and cardiovascular disorders. Results We observed that the frequency of MDSC in healthy donors varied substantially between centers and was influenced by technical aspects such as the anticoagulant and separation method used. Expansion of polymorphonuclear (PMN)-MDSC exceeded the expansion of monocytic MDSC (M-MDSC) in five out of six solid tumors. PMN-MDSC expansion was more pronounced in cancer compared with infection and inflammation. Programmed death-ligand 1 was primarily expressed in M-MDSC and e-MDSC and was not upregulated as a consequence of disease. LOX-1 expression was confined to PMN-MDSC. Conclusions This study provides improved technical protocols and workflows for the multi-center analysis of circulating human MDSC subsets. Application of these workflows revealed a predominant expansion of PMN-MDSC in solid tumors that exceeds expansion in chronic infection and inflammation.
Collapse
Affiliation(s)
- Luca Cassetta
- MRC Centre for Reproductive Health, The University of Edinburgh The Queen's Medical Research Institute, Edinburgh, Edinburgh, UK
| | - Kirsten Bruderek
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Joanna Skrzeczynska-Moncznik
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Małopolska, Poland
| | - Oktawia Osiecka
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Małopolska, Poland
| | - Xiaoying Hu
- Clinical Cooperation Unit Dermato-Oncology, DKFZ, Heidelberg, Baden-Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Centre Mannheim, Mannheim, Baden-Württemberg, Germany
| | - Ida Marie Rundgren
- Department of Biomedical Laboratory Scientist Education and Chemical Engineering, Faculty of Engineering and Natural Sciences, Western Norway University of Applied Sciences, Bergen, Hordaland, Norway
| | - Ang Lin
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute, Stockholm, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institute, Stockholm, Stockholm, Sweden
| | - Kim Santegoets
- Medical Center, Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University, Nijmegen, Gelderland, The Netherlands
| | - Utku Horzum
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Ankara, Turkey
| | - Ana Godinho-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, University of Lisbon, Lisboa, Lisboa, Portugal
| | - Gennadiy Zelinskyy
- Institute for Virology, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Thalia Garcia-Tellez
- HIV Inflammation and Persistence, Pasteur Institute, Paris, Île-de-France, France
| | - Sunčica Bjelica
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Beograd, Beograd, Serbia
| | - Bartłomiej Taciak
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, Warszawa, Poland.,Cellis AG, Zurich, Switzerland
| | | | - Benedikt Höing
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Stephan Lang
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Michael Dixon
- Edinburgh Breast Unit and Breast Cancer Now Research Unit, The University of Edinburgh, Edinburgh, Edinburgh, UK
| | - Verena Müller
- Clinical Cooperation Unit Dermato-Oncology, DKFZ, Heidelberg, Baden-Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Centre Mannheim, Mannheim, Baden-Württemberg, Germany
| | - Jochen Sven Utikal
- Department of Dermatology, Venereology and Allergology, University Medical Centre Mannheim, Mannheim, Baden-Württemberg, Germany.,Clinical Cooperation Unit Dermato-Oncology, German Cancer Research Centre, Heidelberg, Baden-Württemberg, Germany
| | - Derya Karakoç
- Department of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, Ankara, Ankara, Turkey.,Department of General Surgery, Faculty of Medicine, Hacettepe University, Ankara, Ankara, Turkey
| | - Kerim Bora Yilmaz
- Department of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, Ankara, Ankara, Turkey.,Department of General Surgery, Gulhane Egitim ve Arastirma Hastanesi, Ankara, Ankara, Turkey
| | - Emilia Górka
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, Warszawa, Poland.,Cellis AG, Zurich, Switzerland
| | - Lubomir Bodnar
- Department of Oncology and Immunooncology, Hospital Ministry of the Interior and Administration & Warmia and Masuria Oncology Centre, Olsztyn, Poland.,Department of Oncology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | | | - Christine Bourgeois
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris-Sud, Saint-Aubin, Île-de-France, France
| | - Robert Badura
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, University of Lisbon, Lisboa, Lisboa, Portugal.,Serviço de Doenças Infecciosas, Northern Lisbon University Hospital Centre, Lisboa, Lisboa, Portugal
| | | | - Mirjana Gotic
- Clinic of Hematology, Clinical Center of Serbia, Beograd, Beograd, Serbia
| | - Mark Ter Laan
- Medical Center, Department of Neurosurgery, Radboud University, Nijmegen, Gelderland, The Netherlands
| | - Esther Kers-Rebel
- Medical Center, Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University, Nijmegen, Gelderland, The Netherlands
| | - Magdalena Król
- Department of Cancer Biology, Institute of Biology, Warsaw University of Life Sciences, Warszawa, Poland.,Cellis AG, Zurich, Switzerland
| | - Juan Francisco Santibañez
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Beograd, Beograd, Serbia.,Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | | | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Ana Espada de Sousa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, University of Lisbon, Lisboa, Lisboa, Portugal
| | - Güneş Esendağlı
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Ankara, Turkey.,Department of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, Ankara, Ankara, Turkey
| | - Gosse Adema
- Department of Radiation Oncology, Radboud University Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute, Stockholm, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institute, Stockholm, Stockholm, Sweden
| | - Elisabeth Ersvær
- Department of Biomedical Laboratory Scientist Education and Chemical Engineering, Faculty of Engineering and Natural Sciences, Western Norway University of Applied Sciences, Bergen, Hordaland, Norway
| | - Viktor Umansky
- Department of Dermatology, Venereology and Allergology, University Medical Centre Mannheim, Mannheim, Baden-Württemberg, Germany.,Clinical Cooperation Unit Dermato-Oncology, German Cancer Research Centre, Heidelberg, Baden-Württemberg, Germany
| | - Jeffrey W Pollard
- MRC Centre for Reproductive Health, The University of Edinburgh The Queen's Medical Research Institute, Edinburgh, Edinburgh, UK
| | - Joanna Cichy
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Małopolska, Poland
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany .,German Cancer Consortium, Partner Site Essen-Düsseldorf, Germany
| |
Collapse
|
173
|
Siolas D, Morrissey C, Oberstein PE. The Achilles' Heel of Pancreatic Cancer: Targeting pancreatic cancer's unique immunologic characteristics and metabolic dependencies in clinical trials. JOURNAL OF PANCREATOLOGY 2020; 3:121-131. [PMID: 33133736 PMCID: PMC7595263 DOI: 10.1097/jp9.0000000000000052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a high mortality rate and is notoriously refractory to multiple cancer treatments. In recent years, cancer therapy has expanded beyond traditional cytotoxic chemotherapy to targeted agents and immunotherapy which have been successfully implemented in many cancers. Despite robust pre-clinical research, these novel therapies have only had a small impact on PDAC. However, there have been successes with emerging clinical data supporting a potential role for checkpoint inhibitor therapy and targeted therapy with poly (ADP-ribose) polymerase inhibitors for select subsets of PDAC patients. In this clinical review, we discuss recent pre-clinical evidence for targeting metabolic pathways as well as prevalent intratumoral immune subsets, and focus on clinical trials designed to test novel agents in PDAC. The challenge of translating pre-clinical findings to patients remains substantial and many clinical trials yield negative results, but collaborative efforts and renewed focus on novel clinical trials have led to optimism that we will identify additional options for PDAC patients and change outcomes for this deadly disease.
Collapse
Affiliation(s)
- Despina Siolas
- Department of Medicine, Pancreatic Cancer Center, Perlmutter Comprehensive Cancer Center, NYU Langone Health, New York, NY
| | - Christy Morrissey
- Department of Medicine, Pancreatic Cancer Center, Perlmutter Comprehensive Cancer Center, NYU Langone Health, New York, NY
| | - Paul Eliezer Oberstein
- Department of Medicine, Pancreatic Cancer Center, Perlmutter Comprehensive Cancer Center, NYU Langone Health, New York, NY
| |
Collapse
|
174
|
Boone CE, Wang C, Lopez-Ramirez MA, Beiss V, Shukla S, Chariou PL, Kupor D, Rueda R, Wang J, Steinmetz NF. Active Microneedle Administration of Plant Virus Nanoparticles for Cancer in situ Vaccination Improves Immunotherapeutic Efficacy. ACS APPLIED NANO MATERIALS 2020; 3:8037-8051. [PMID: 33969278 PMCID: PMC8101548 DOI: 10.1021/acsanm.0c01506] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The solid tumor microenvironment (TME) poses a significant structural and biochemical barrier to immunotherapeutic agents. To address the limitations of tumor penetration and distribution, and to enhance antitumor efficacy of immunotherapeutics, we present here an autonomous active microneedle (MN) system for the direct intratumoral (IT) delivery of a potent immunoadjuvant, cowpea mosaic virus nanoparticles (CPMV) in vivo. In this active delivery system, magnesium (Mg) microparticles embedded into active MNs react with the interstitial fluid in the TME, generating a propulsive force to drive the nanoparticle payload into the tumor. Active delivery of CPMV payload into B16F10 melanomas in vivo demonstrated substantially more pronounced tumor regression and prolonged survival of tumor-bearing mice compared to that of passive MNs and conventional needle injection. Active MN administration of CPMV also enhanced local innate and systemic adaptive antitumor immunity. Our approach represents an elaboration of conventional CPMV in situ vaccination, highlighting substantial immune-mediated antitumor effects and improved therapeutic efficacy that can be achieved through an active and autonomous delivery system-mediated CPMV in situ vaccination.
Collapse
Affiliation(s)
- Christine E. Boone
- Department of Radiology, UC San Diego Health, University of California, San Diego, La Jolla California 92093, United States
| | - Chao Wang
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Miguel Angel Lopez-Ramirez
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Veronique Beiss
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Sourabh Shukla
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Paul L. Chariou
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Daniel Kupor
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Ricardo Rueda
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Joseph Wang
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering (nanoIE), University of California, San Diego, La Jolla, California 92093, United States
| | - Nicole F. Steinmetz
- Department of Radiology, UC San Diego Health, University of California, San Diego, La Jolla California 92093, United States
- Department of Nanoengineering, University of California, San Diego, La Jolla, California 92093, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, UC San Diego Health, University of California, San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering (nanoIE), University of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
175
|
Daveri E, Vergani E, Shahaj E, Bergamaschi L, La Magra S, Dosi M, Castelli C, Rodolfo M, Rivoltini L, Vallacchi V, Huber V. microRNAs Shape Myeloid Cell-Mediated Resistance to Cancer Immunotherapy. Front Immunol 2020; 11:1214. [PMID: 32793185 PMCID: PMC7387687 DOI: 10.3389/fimmu.2020.01214] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 05/15/2020] [Indexed: 12/20/2022] Open
Abstract
Immunotherapy with immune checkpoint inhibitors can achieve long-term tumor control in subsets of patients. However, its effect can be blunted by myeloid-induced resistance mechanisms. Myeloid cells are highly plastic and physiologically devoted to wound healing and to immune homeostasis maintenance. In cancer, their physiological activities can be modulated, leading to an expansion of pro-inflammatory and immunosuppressive cells, the myeloid-derived suppressor cells (MDSCs), with detrimental consequences. The involvement of MDSCs in tumor development and progression has been widely investigated and MDSC-induced immunosuppression is acknowledged as a mechanism hindering effective immune checkpoint blockade. Small non-coding RNA molecules, the microRNAs (miRs), contribute to myeloid cell regulation at different levels, comprising metabolism and function, as well as their skewing to a MDSC phenotype. miR expression can be indirectly induced by cancer-derived factors or through direct miR import via extracellular vesicles. Due to their structural stability and their presence in body fluids miRs represent promising predictive biomarkers of resistance, as we recently found by investigating plasma samples of melanoma patients undergoing immune checkpoint blockade. Dissection of the miR-driven involved mechanisms would pave the way for the identification of new druggable targets. Here, we discuss the role of these miRs in shaping myeloid resistance to immunotherapy with a special focus on immunosuppression and immune escape.
Collapse
Affiliation(s)
- Elena Daveri
- Unit of Immunotherapy of Human Tumors, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elisabetta Vergani
- Unit of Immunotherapy of Human Tumors, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Eriomina Shahaj
- Unit of Immunotherapy of Human Tumors, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Laura Bergamaschi
- Unit of Immunotherapy of Human Tumors, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Stefano La Magra
- Unit of Immunotherapy of Human Tumors, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michela Dosi
- Unit of Immunotherapy of Human Tumors, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara Castelli
- Unit of Immunotherapy of Human Tumors, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Rodolfo
- Unit of Immunotherapy of Human Tumors, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Viviana Vallacchi
- Unit of Immunotherapy of Human Tumors, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Veronica Huber
- Unit of Immunotherapy of Human Tumors, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
176
|
Abstract
Purpose of review This review focuses on the development and progression of glioblastoma through the brain and glioma microenvironment. Specifically we highlight how the tumor microenvironment contributes to the hallmarks of cancer in hopes of offering novel therapeutic options and tools to target this microenvironment. Recent findings The hallmarks of cancer, which represent elements of cancers that contribute to the disease's malignancy, yet elements within the brain tumor microenvironment, such as other cellular types as well as biochemical and biophysical cues that can each uniquely affect tumor cells, have not been well-described in this context and serve as potential targets for modulation. Summary Here, we highlight how the brain tumor microenvironment contributes to the progression and therapeutic response of tumor cells. Specifically, we examine these contributions through the lens of Hanahan & Weinberg's Hallmarks of Cancer in order to identify potential novel targets within the brain that may offer a means to treat brain cancers, including the deadliest brain cancer, glioblastoma.
Collapse
|
177
|
Murch SH. Common determinants of severe Covid-19 infection are explicable by SARS-CoV-2 secreted glycoprotein interaction with the CD33-related Siglecs, Siglec-3 and Siglec-5/14. Med Hypotheses 2020; 144:110168. [PMID: 33254494 PMCID: PMC7413060 DOI: 10.1016/j.mehy.2020.110168] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/23/2020] [Accepted: 08/05/2020] [Indexed: 12/23/2022]
Abstract
SARS-CoV-2 interaction with the ACE-2 receptor cannot alone explain the demography and remarkable variation in clinical progression of Covid-19 infection. Unlike SARS-CoV, the cause of SARS, several SARS-CoV-2 spike glycans contain sialic acid residues. In contrast to the SARS secreted glycoprotein (SGP), SARS-CoV-2 SGP are thus potential ligands for Sialic acid-binding Siglecs on host immune cells, known to regulate immune function. Such SARS-CoV-2 glycoproteins would contribute to immune deviation. CD33-related Siglecs are important immune regulators. Siglec-5 and −14 are paired receptors with opposed actions on the NLRP3 inflammasome, which is critical in early viral clearance. SGP binding in persons of Siglec-14 null genotype (30–70% in Black, Asian and Minority Ethnic (BAME) persons, 10% in North Europeans) would induce unopposed inhibitory signalling, causing viral persistence through inflammasome inhibition. Siglec-3 (CD33) and Siglec-5 are expressed on CD33 myeloid derived suppressor cells (CD33 MDSC). Immunosuppressive CD33 MDSC populations are increased in all groups at risk of severe Covid-19 infection. CD33 expression is increased in persons with the CD33 rs3865444 CC allele, associated with Alzheimer’s disease, who would thus show enhanced susceptibility. Viral SGP ligation of CD33, potentially in conjunction with Siglec-5, would promote expansion of CD33 MDSC cells, as occurs in cancers but at much greater scale. CD33 is expressed on CNS microglia, potentially activated by SGP penetration through the porous cribriform plate to cause anosmia. Genotyping of severe or fatal Covid-19 cases can confirm or refute this pathophysiological mechanism. Early data have confirmed extremely high-level increase of CD33 MDSC numbers in severe Covid-19 infection, consistent with the proposed mechanism.
Collapse
Affiliation(s)
- Simon H Murch
- Emeritus Professor, Warwick Medical School, University of Warwick, Coventry, UK.
| |
Collapse
|
178
|
Jin S, Yang Z, Hao X, Tang W, Ma W, Zong H. Roles of HMGB1 in regulating myeloid-derived suppressor cells in the tumor microenvironment. Biomark Res 2020; 8:21. [PMID: 32551121 PMCID: PMC7298841 DOI: 10.1186/s40364-020-00201-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are notable contributors to the immunosuppressive tumor microenvironment (TME) and are closely associated with tumor progression; in addition, MDSCs are present in most patients with cancer. However, the molecular mechanisms that regulate MDSCs in the etiopathogenesis of human tumor immunity remain unclear. The secreted alarmin high mobility group box 1 (HMGB1) is a proinflammatory factor and inducer of many inflammatory molecules during MDSC development. In this review, we detail the currently reported characteristics of MDSCs in tumor immune escape and the regulatory role of secreted HMGB1 in MDSC differentiation, proliferation, activity and survival. Notably, different posttranslational modifications of HMGB1 may have various effects on MDSCs, and these effects need further identification. Moreover, exosome-derived HMGB1 is speculated to exert a regulatory effect on MDSCs, but no report has confirmed this hypothesis. Therefore, the effects of HMGB1 on MDSCs need more research attention, and additional investigations should be conducted.
Collapse
Affiliation(s)
- Shuiling Jin
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, NO.1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China
| | - Zhenzhen Yang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, NO.1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China.,Academy of medical science, Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Xin Hao
- Henan college of Health Cadres, Zhengzhou, 450008 Henan China
| | - Wenxue Tang
- Departments of Otolaryngology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan China.,Center for Precision Medicine of Zhengzhou University, Zhengzhou, 450052 Henan China.,Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, NO.40 North Daxue Road, Zhengzhou, 450052 Henan China
| | - Wang Ma
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, NO.1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China
| | - Hong Zong
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, NO.1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China
| |
Collapse
|
179
|
Ning G, Huang YL, Zhen LM, Xu WX, Li XJ, Wu LN, Liu Y, Xie C, Peng L. Prognostic Value of Complement Component 2 and Its Correlation with Immune Infiltrates in Hepatocellular Carcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3765937. [PMID: 32626741 PMCID: PMC7312969 DOI: 10.1155/2020/3765937] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/20/2020] [Accepted: 05/25/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Single nucleotide polymorphism (SNP) of complement component 2 (C2) has been found to be significantly associated with hepatocellular carcinoma (HCC). However, little is known about the role and mechanism of C2 in HCC. In the present study, we aimed to explore the prognostic value of C2 and its correlation with tumor-infiltrating immune cells in HCC. MATERIALS AND METHODS mRNA expression was downloaded from TCGA (365 HCC patients and 50 healthy controls), GSE14520 (220 HCC patients and 220 adjacent normal tissues), and ICGC HCC (232 HCC patients) cohorts. Unpaired Student's t-tests or ANOVA tests were used to evaluate differences of C2 expression. Univariate and multivariate analyses were used to analyze the prognostic value of C2. CIBERSORT was used to calculate the proportion of 22 kinds of tumor-infiltrating immune cells. RESULTS Significantly lower C2 expression was found at HCC compared to healthy controls, and C2 was associated with TNM stages. Higher C2 expression was significantly associated with better prognosis, and multivariate analysis showed that C2 was also an independent factor for the prognosis of HCC. Moreover, elevated CD4 T cells were found at HCC patients with higher C2 expression while the higher proportion of macrophage M0 cells was found in HCC patients with lower C2 expression. KEGG analysis showed that "cell cycle," "AMPK signaling pathway," and "PPAR signaling pathway" were enriched in HCC patients with higher C2 expression. CONCLUSION C2 is a prognostic factor for HCC and may be used as a therapeutic target for future treatment of HCC.
Collapse
Affiliation(s)
- Gang Ning
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan-Lin Huang
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Li-Min Zhen
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wen-Xiong Xu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xue-Jun Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Li-Na Wu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ying Liu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chan Xie
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liang Peng
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
180
|
Wang W, Han Y, Jo HA, Lee J, Song YS. Non-coding RNAs shuttled via exosomes reshape the hypoxic tumor microenvironment. J Hematol Oncol 2020; 13:67. [PMID: 32503591 PMCID: PMC7275461 DOI: 10.1186/s13045-020-00893-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023] Open
Abstract
Exosomes are small extracellular vesicles secreted by almost all the cells. Molecular cargos of exosomes can partially reflect the characteristics of originating cells. Exosome-mediated cell-to-cell interactions in the microenvironment are critical in cancer progression. Hypoxia, a key pro-cancerous feature of the tumor microenvironment, alters the releasing and contents of exosomes. A growing body of evidence shows that hypoxia induces more aggressive phenotypes in cancer. Of note, non-coding RNAs shuttled in hypoxic tumor-derived exosomes have been demonstrated as fundamental molecules in regulating cancer biology and remodeling tumor microenvironment. Furthermore, these hypoxic tumor-derived exosomal non-coding RNAs can be detected in the body fluids, serving as promising diagnostic and prognostic biomarkers. The current review discusses changes in cancer behaviors regulated by exosomes-secreted non-coding RNAs under hypoxic conditions.
Collapse
Affiliation(s)
- Wenyu Wang
- Interdisciplinary Program in Cancer Biology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Youngjin Han
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, 03080, Republic of Korea
| | - Hyun A Jo
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, 03080, Republic of Korea
| | - Juwon Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, 03080, Republic of Korea
| | - Yong Sang Song
- Interdisciplinary Program in Cancer Biology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, 03080, Republic of Korea.
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
181
|
Coletti R, Leonardelli L, Parolo S, Marchetti L. A QSP model of prostate cancer immunotherapy to identify effective combination therapies. Sci Rep 2020; 10:9063. [PMID: 32493951 PMCID: PMC7270132 DOI: 10.1038/s41598-020-65590-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 05/06/2020] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy, by enhancing the endogenous anti-tumor immune responses, is showing promising results for the treatment of numerous cancers refractory to conventional therapies. However, its effectiveness for advanced castration-resistant prostate cancer remains unsatisfactory and new therapeutic strategies need to be developed. To this end, systems pharmacology modeling provides a quantitative framework to test in silico the efficacy of new treatments and combination therapies. In this paper we present a new Quantitative Systems Pharmacology (QSP) model of prostate cancer immunotherapy, calibrated using data from pre-clinical experiments in prostate cancer mouse models. We developed the model by using Ordinary Differential Equations (ODEs) describing the tumor, key components of the immune system, and seven treatments. Numerous combination therapies were evaluated considering both the degree of tumor inhibition and the predicted synergistic effects, integrated into a decision tree. Our simulations predicted cancer vaccine combined with immune checkpoint blockade as the most effective dual-drug combination immunotherapy for subjects treated with androgen-deprivation therapy that developed resistance. Overall, the model presented here serves as a computational framework to support drug development, by generating hypotheses that can be tested experimentally in pre-clinical models.
Collapse
Affiliation(s)
- Roberta Coletti
- University of Trento, Department of mathematics, Trento, 38123, Italy
- Fondazione The Microsoft Research - University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, 38068, Italy
| | - Lorena Leonardelli
- Fondazione The Microsoft Research - University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, 38068, Italy
| | - Silvia Parolo
- Fondazione The Microsoft Research - University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, 38068, Italy
| | - Luca Marchetti
- Fondazione The Microsoft Research - University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, 38068, Italy.
| |
Collapse
|
182
|
A novel methodology of the myeloid-derived suppressor cells (MDSCs) generation with splenic stroma feeder cells. Exp Cell Res 2020; 394:112119. [PMID: 32485182 DOI: 10.1016/j.yexcr.2020.112119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/21/2020] [Accepted: 05/28/2020] [Indexed: 01/17/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a significant obstacle for immunotherapy of cancer. It is of great clinical relevance to study the mechanism of MDSCs accumulation in mouse spleens and establish a stable method to obtain high-purity MDSCs in vitro for further research. Here, we established a new method for amplifying a large number of highly pure MDSCs in vitro. To mimic the microenvironment of MDSCs development in vivo, mouse splenic stroma feeder cells and serum-free medium containing granulocyte-macrophage colony stimulating factor (GM-CSF) were used to induce myeloid precursors in mouse bone marrow cells, which differentiate into MDSCs. Development and immunological functions of the cells were monitored both in vivo and in vitro. A total of 4 × 108 MDSCs could be obtained from the bone marrow from one mouse, the ratio of CD11b+Gr-1+ MDSCs could reach 93.8% ± 3.3% after nine days of culture in vitro. Cultured MDSCs maintained a similar immunophenotype with MDSCs found in tumor-bearing mice. Colony forming assay in vitro and in vivo demonstrated that these were myeloid precursor cells. These cells generated high levels of reactive oxygen species and arginase 1 to prevent proliferation of CD8+ T cells in vitro. These also increased regulatory T (Treg) cells in blood while promoting the growth of lymphoma in vivo. In addition, cultured MDSCs effectively inhibited acute graft-versus-host disease (aGVHD). Our findings suggest that mouse splenic stroma plays an important role in the generation of MDSCs and represent a preliminary mechanism for the accumulation of MDSCs in spleens, and thereby lay the foundation for basic research and the clinical application of MDSCs.
Collapse
|
183
|
Liang H, Shen X. LXR activation radiosensitizes non-small cell lung cancer by restricting myeloid-derived suppressor cells. Biochem Biophys Res Commun 2020; 528:330-335. [PMID: 32448508 DOI: 10.1016/j.bbrc.2020.04.137] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 04/24/2020] [Indexed: 01/18/2023]
Abstract
Radiotherapy (RT) is an important radical treatment for locally advanced non-small cell lung cancer (NSCLC). However, radioresistance greatly impairs the efficacy of this therapy in the clinic. Radioresistance can be caused by radiation-induced myeloid-derived suppressor cell (MDSC) infiltration. Liver-X nuclear receptor (LXR) agonists have demonstrated potent antitumor activity in preclinic animal models. Here, we report for the first time that LXR agonists, GW3965 and RGX-104, radiosensitized NSCLC in a subcutaneous homograft murine model. LXR activation significantly reduced MDSC abundance in the tumor microenvironment (TME). Treatment with RGX-104 greatly promoted MDSC apoptosis in vitro. Depleting MDSC activated cytotoxic T lymphocyte (CTL) and T-helper 1 (Th1) responses in the TME. In conclusion, the immunosuppressive effects of radiotherapy can be abrogated partly with an LXR agonist by depleting MDSC, which sensitizes NSCLC to RT.
Collapse
Affiliation(s)
- Huaizhen Liang
- Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaoli Shen
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
184
|
Understanding the Differentiation, Expansion, Recruitment and Suppressive Activities of Myeloid-Derived Suppressor Cells in Cancers. Int J Mol Sci 2020; 21:ijms21103599. [PMID: 32443699 PMCID: PMC7279333 DOI: 10.3390/ijms21103599] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/28/2020] [Accepted: 03/13/2020] [Indexed: 12/15/2022] Open
Abstract
There has been a great interest in myeloid-derived suppressor cells (MDSCs) due to their biological functions in tumor-mediated immune escape by suppressing antitumor immune responses. These cells arise from altered myelopoiesis in response to the tumor-derived factors. The most recognized function of MDSCs is suppressing anti-tumor immune responses by impairing T cell functions, and these cells are the most important players in cancer dissemination and metastasis. Therefore, understanding the factors and the mechanism of MDSC differentiation, expansion, and recruitment into the tumor microenvironment can lead to its control. However, most of the studies only defined MDSCs with no further characterization of granulocytic and monocytic subsets. In this review, we discuss the mechanisms by which specific MDSC subsets contribute to cancers. A better understanding of MDSC subset development and the specific molecular mechanism is needed to identify treatment targets. The understanding of the specific molecular mechanisms responsible for MDSC accumulation would enable more precise therapeutic targeting of these cells.
Collapse
|
185
|
Ishiguro S, Upreti D, Robben N, Burghart R, Loyd M, Ogun D, Le T, Delzeit J, Nakashima A, Thakkar R, Nakashima A, Suzuki K, Comer J, Tamura M. Water extract from Euglena gracilis prevents lung carcinoma growth in mice by attenuation of the myeloid-derived cell population. Biomed Pharmacother 2020; 127:110166. [PMID: 32361165 DOI: 10.1016/j.biopha.2020.110166] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 12/22/2022] Open
Abstract
The partially purified water extract from Euglena gracilis (EWE) was evaluated for its antitumor and immunomodulatory effects in cell cultures and in a mouse orthotopic lung carcinoma allograft model. In two-dimensional cell culture, the EWE treatment inhibited cell growth of both murine Lewis lung carcinoma (LLC) and human lung carcinoma cells (A549 and H1299) in a dose- and time-dependent manner. In contrast, the growth of mouse bone marrow cells (BMCs), but not mouse splenocytes (SPLs), was stimulated by the treatment with EWE. In three-dimensional spheroid culture, spheroid growth of LLC cells was significantly attenuated by EWE treatment. In a mouse LLC orthotopic allograft model, pretreatment with EWE (150-200 mg/kg/day, via drinking water) three weeks prior to the LLC cell inoculation, but not post-treatment after LLC cell inoculation, significantly attenuated the growth of LLC tumors in immunocompetent syngeneic mouse lung. This tumor growth attenuation coincided with a significant decrease in the population of myeloid-derived cells, primarily neutrophils. Flow cytometric analysis revealed that the EWE treatment significantly attenuated growth of granulocytic myeloid-derived suppressor cells (gMDSC) in BMCs and that this decrease was due to induction of gMDSC-specific apoptosis and differentiation of monocytic MDSCs (mMDSC) to macrophages. The present study provides evidence that EWE pretreatment inhibits lung carcinoma growth mainly by stimulating host antitumor immunity through attenuation of growth of gMDSCs and decreasing the number of peripheral granulocytes. This study suggests that the partially purified extract derived from Euglena gracilis contains significant bioactive materials that prevent lung carcinoma growth.
Collapse
Affiliation(s)
- Susumu Ishiguro
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Deepa Upreti
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Nicole Robben
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Riley Burghart
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Mayme Loyd
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Damilola Ogun
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Tran Le
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Jennifer Delzeit
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Arashi Nakashima
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Ravindra Thakkar
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | | | - Kengo Suzuki
- Euglena Co., Ltd., Minato-ku, Tokyo 108-0014, Japan.
| | - Jeffrey Comer
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Masaaki Tamura
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| |
Collapse
|
186
|
Chen SMY, Krinsky AL, Woolaver RA, Wang X, Chen Z, Wang JH. Tumor immune microenvironment in head and neck cancers. Mol Carcinog 2020; 59:766-774. [PMID: 32017286 DOI: 10.1002/mc.23162] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/18/2020] [Accepted: 01/22/2020] [Indexed: 12/24/2022]
Abstract
Head and neck cancers are a heterogeneous group of tumors that are highly aggressive and collectively represent the sixth most common cancer worldwide. Ninety percent of head and neck cancers are squamous cell carcinomas (HNSCCs). The tumor microenvironment (TME) of HNSCCs consists of many different subsets of cells that infiltrate the tumors and interact with the tumor cells or with each other through various networks. Both innate and adaptive immune cells play a crucial role in mediating immune surveillance and controlling tumor growth. Here, we discuss the different subsets of immune cells and how they contribute to an immunosuppressive TME of HNSCCs. We also briefly summarize recent advances in immunotherapeutic approaches for HNSCC treatment. A better understanding of the multiple factors that play pivotal roles in HNSCC tumorigenesis and tumor progression may help define novel targets to develop more effective immunotherapies for patients with HNSCC.
Collapse
Affiliation(s)
- Samantha M Y Chen
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Alexandra L Krinsky
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Rachel A Woolaver
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Xiaoguang Wang
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Zhangguo Chen
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jing H Wang
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
187
|
Lv M, Wang K, Huang XJ. Myeloid-derived suppressor cells in hematological malignancies: friends or foes. J Hematol Oncol 2019; 12:105. [PMID: 31640764 PMCID: PMC6805310 DOI: 10.1186/s13045-019-0797-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 09/25/2019] [Indexed: 12/25/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are newly identified immature myeloid cells that are characterized by the ability to suppress immune responses and expand during cancer, infection, and inflammatory diseases. Although MDSCs have attracted a lot of attention in the field of tumor immunology in recent years, little is known about their multiple roles in hematological malignancies as opposed to their roles in solid tumors. This review will help researchers better understand the various characteristics and functions of MDSCs, as well as the potential therapeutic applications of MDSCs in hematological malignancies, including lymphoma, multiple myeloma, leukemia, and hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Meng Lv
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of HSCT, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Ke Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of HSCT, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of HSCT, No 11 Xizhimen South Street, Beijing, 100044, China. .,Peking-Tsinghua Center for Life Sciences, Beijing, China.
| |
Collapse
|
188
|
Rojas-Sanchez G, Cotzomi-Ortega I, Pazos-Salazar NG, Reyes-Leyva J, Maycotte P. Autophagy and Its Relationship to Epithelial to Mesenchymal Transition: When Autophagy Inhibition for Cancer Therapy Turns Counterproductive. BIOLOGY 2019; 8:biology8040071. [PMID: 31554173 PMCID: PMC6956138 DOI: 10.3390/biology8040071] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/14/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023]
Abstract
The manipulation of autophagy for cancer therapy has gained recent interest in clinical settings. Although inhibition of autophagy is currently being used in clinical trials for the treatment of several malignancies, autophagy has been shown to have diverse implications for normal cell homeostasis, cancer cell survival, and signaling to cells in the tumor microenvironment. Among these implications and of relevance for cancer therapy, the autophagic process is known to be involved in the regulation of protein secretion, in tumor cell immunogenicity, and in the regulation of epithelial-to-mesenchymal transition (EMT), a critical step in the process of cancer cell invasion. In this work, we have reviewed recent evidence linking autophagy to the regulation of EMT in cancer and normal epithelial cells, and have discussed important implications for the manipulation of autophagy during cancer therapy.
Collapse
Affiliation(s)
- Guadalupe Rojas-Sanchez
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla 72570, Mexico.
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Km 4.5 Carretera Atlixco-Metepec HGZ5, Puebla 74360, Mexico.
| | - Israel Cotzomi-Ortega
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla 72570, Mexico.
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Km 4.5 Carretera Atlixco-Metepec HGZ5, Puebla 74360, Mexico.
| | - Nidia G Pazos-Salazar
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla 72570, Mexico.
| | - Julio Reyes-Leyva
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Km 4.5 Carretera Atlixco-Metepec HGZ5, Puebla 74360, Mexico.
| | - Paola Maycotte
- Consejo Nacional de Ciencia y Tecnología (CONACYT)-CIBIOR, IMSS, Puebla 74360, Mexico.
| |
Collapse
|
189
|
Tian X, Shen H, Li Z, Wang T, Wang S. Tumor-derived exosomes, myeloid-derived suppressor cells, and tumor microenvironment. J Hematol Oncol 2019; 12:84. [PMID: 31438991 PMCID: PMC6704713 DOI: 10.1186/s13045-019-0772-z] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/14/2019] [Indexed: 12/13/2022] Open
Abstract
Plenty of immune cells infiltrate into the tumor microenvironment (TME) during tumor progression, in which myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of immature myeloid cells with immunosuppressive activity. Tumor cells and stromal cells facilitate the activation and expansion of MDSCs in TME via intercellular communication, and expanded MDSCs suppress anti-tumor immune responses through direct and indirect mechanisms. Currently, exosomes, which are a kind of extracellular vesicles (EVs) that can convey functional components, are demonstrated to participate in the local and distal intercellular communication between cells. Numerous studies have supposed that tumor-derived exosomes (TEXs), whose assembly and release can be modulated by TME, are capable of modulating the cell biology of MDSCs, including facilitating their activation, promoting the expansion, and enhancing the immunosuppressive function. Therefore, in this review, we mainly focus on the role of TEXs in the cell-cell communication between tumor cells and MDSCs, and discuss their clinical applications.
Collapse
Affiliation(s)
- Xinyu Tian
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Han Shen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Zhiyang Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.
| | - Tingting Wang
- Department of Laboratory Medicine, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, China.
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
190
|
Leija Montoya G, González Ramírez J, Sandoval Basilio J, Serafín Higuera I, Isiordia Espinoza M, González González R, Serafín Higuera N. Long Non-coding RNAs: Regulators of the Activity of Myeloid-Derived Suppressor Cells. Front Immunol 2019; 10:1734. [PMID: 31404149 PMCID: PMC6671873 DOI: 10.3389/fimmu.2019.01734] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 07/09/2019] [Indexed: 12/29/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous cell population with potent immunosuppressive functions. They play major roles in cancer and many of the pathologic conditions associated with inflammation. Long non-coding RNAs (lncRNAs) are untranslated functional RNA molecules. The lncRNAs are involved in the control of a wide variety of cellular processes and are dysregulated in different diseases. They can participate in the modulation of immune function and activity of inflammatory cells, including MDSCs. This mini review focuses on the emerging role of lncRNAs in MDSC activity. We summarize how lncRNAs modulate the generation, recruitment, and immunosuppressive functions of MDSCs and the underlying mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Mario Isiordia Espinoza
- División de Ciencias Biomédicas, Departamento de Clínicas, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, Guadalajara, Mexico
| | | | - Nicolás Serafín Higuera
- Unidad de Ciencias de la Salud, Facultad de Odontología, Universidad Autónoma de Baja California, Mexicali, Mexico
| |
Collapse
|
191
|
Seebach E, Kubatzky KF. Chronic Implant-Related Bone Infections-Can Immune Modulation be a Therapeutic Strategy? Front Immunol 2019; 10:1724. [PMID: 31396229 PMCID: PMC6664079 DOI: 10.3389/fimmu.2019.01724] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/09/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic implant-related bone infections are a major problem in orthopedic and trauma-related surgery with severe consequences for the affected patients. As antibiotic resistance increases in general and because most antibiotics have poor effectiveness against biofilm-embedded bacteria in particular, there is a need for alternative and innovative treatment approaches. Recently, the immune system has moved into focus as the key player in infection defense and bone homeostasis, and the targeted modulation of the host response is becoming an emerging field of interest. The aim of this review was to summarize the current knowledge of impaired endogenous defense mechanisms that are unable to prevent chronicity of bone infections associated with a prosthetic or osteosynthetic device. The presence of foreign material adversely affects the immune system by generating a local immune-compromised environment where spontaneous clearance of planktonic bacteria does not take place. Furthermore, the surface structure of the implant facilitates the transition of bacteria from the planktonic to the biofilm stage. Biofilm formation on the implant surface is closely linked to the development of a chronic infection, and a misled adaption of the immune system makes it impossible to effectively eliminate biofilm infections. The interaction between the immune system and bone cells, especially osteoclasts, is extensively studied in the field of osteoimmunology and this crosstalk further aggravates the course of bone infection by shifting bone homeostasis in favor of bone resorption. T cells play a major role in various chronic diseases and in this review a special focus was therefore set on what is known about an ineffective T cell response. Myeloid-derived suppressor cells (MDSCs), anti-inflammatory macrophages, regulatory T cells (Tregs) as well as osteoclasts all suppress immune defense mechanisms and negatively regulate T cell-mediated immunity. Thus, these cells are considered to be potential targets for immune therapy. The success of immune checkpoint inhibition in cancer treatment encourages the transfer of such immunological approaches into treatment strategies of other chronic diseases. Here, we discuss whether immune modulation can be a therapeutic tool for the treatment of chronic implant-related bone infections.
Collapse
Affiliation(s)
- Elisabeth Seebach
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Katharina F Kubatzky
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
192
|
Zhang R, Liu Q, Li T, Liao Q, Zhao Y. Role of the complement system in the tumor microenvironment. Cancer Cell Int 2019; 19:300. [PMID: 31787848 PMCID: PMC6858723 DOI: 10.1186/s12935-019-1027-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/11/2019] [Indexed: 12/17/2022] Open
Abstract
The complement system has traditionally been considered a component of innate immunity against invading pathogens and "nonself" cells. Recent studies have demonstrated the immunoregulatory functions of complement activation in the tumor microenvironment (TME). The TME plays crucial roles in tumorigenesis, progression, metastasis and recurrence. Imbalanced complement activation and the deposition of complement proteins have been demonstrated in many types of tumors. Plasma proteins, receptors, and regulators of complement activation regulate several biological functions of stromal cells in the TME and promote the malignant biological properties of tumors. Interactions between the complement system and cancer cells contribute to the proliferation, epithelial-mesenchymal transition, migration and invasion of tumor cells. In this review, we summarize recent advances related to the function of the complement system in the TME and discuss the therapeutic potential of targeting complement-mediated immunoregulation in cancer immunotherapy.
Collapse
Affiliation(s)
- Ronghua Zhang
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Qiaofei Liu
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Tong Li
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Quan Liao
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Yupei Zhao
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| |
Collapse
|