1
|
Yaseen MM, Abuharfeil NM, Darmani H. MDSC expansion during HIV infection: regulators, ART and immune reconstitution. Genes Immun 2024; 25:242-253. [PMID: 38605259 DOI: 10.1038/s41435-024-00272-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) become expanded in different pathological conditions including human immunodeficiency virus (HIV) infection and this may worsen the disease status and accelerate disease progression. In HIV infection, MDSCs suppress anti-HIV immune responses and hamper immune reconstitution. Understanding the factors and mechanisms of MDSC expansion during HIV infection is central to understanding the pathophysiology of HIV infection. This may pave the way to developing new therapeutic targets or strategies. In this work we addressed (i) the mechanisms that regulate MDSC expansion, (ii) the impact of antiretroviral therapy (ART) on the frequency of MDSCs during HIV infection; (iii) the impact of MDSCs on immune reconstitution during successful ART; and (iv) the potential of MDSCs as a therapeutic target.
Collapse
Affiliation(s)
- Mahmoud Mohammad Yaseen
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan.
| | - Nizar Mohammad Abuharfeil
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Homa Darmani
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| |
Collapse
|
2
|
Zhang H, Li QW, Li YY, Tang X, Gu L, Liu HM. Myeloid-derived suppressor cells and pulmonary hypertension. Front Immunol 2023; 14:1189195. [PMID: 37350962 PMCID: PMC10282836 DOI: 10.3389/fimmu.2023.1189195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/18/2023] [Indexed: 06/24/2023] Open
Abstract
Pulmonary hypertension (PH) is a chronic pulmonary vascular disorder characterized by an increase in pulmonary vascular resistance and pulmonary arterial pressure. The detailed molecular mechanisms remain unclear. In recent decades, increasing evidence shows that altered immune microenvironment, comprised of immune cells, mesenchymal cells, extra-cellular matrix and signaling molecules, might induce the development of PH. Myeloid-derived suppressor cells (MDSCs) have been proposed over 30 years, and the functional importance of MDSCs in the immune system is appreciated recently. MDSCs are a heterogeneous group of cells that expand during cancer, chronic inflammation and infection, which have a remarkable ability to suppress T-cell responses and may exacerbate the development of diseases. Thus, targeting MDSCs has become a novel strategy to overcome immune evasion, especially in tumor immunotherapy. Nowadays, severe PH is accepted as a cancer-like disease, and MDSCs are closely related to the development and prognosis of PH. Here, we review the relationship between MDSCs and PH with respect to immune cells, cytokines, chemokines and metabolism, hoping that the key therapeutic targets of MDSCs can be identified in the treatment of PH, especially in severe PH.
Collapse
Affiliation(s)
- Hui Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- The Fifth People’s Hospital of Chengdu, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (Sichuan University), National Health Commission of China, Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qi-Wei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (Sichuan University), National Health Commission of China, Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yuan-Yuan Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (Sichuan University), National Health Commission of China, Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xue Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (Sichuan University), National Health Commission of China, Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ling Gu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (Sichuan University), National Health Commission of China, Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Han-Min Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (Sichuan University), National Health Commission of China, Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Zhang W, Fang X, Gao C, Song C, He Y, Zhou T, Yang X, Shang Y, Xu J. MDSCs in sepsis-induced immunosuppression and its potential therapeutic targets. Cytokine Growth Factor Rev 2023; 69:90-103. [PMID: 35927154 DOI: 10.1016/j.cytogfr.2022.07.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 02/07/2023]
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. In sepsis, a complicated immune response is initiated, which varies over time with sustained excessive inflammation and immunosuppression. Identifying a promising way to orchestrate sepsis-induced immunosuppression is a challenge. Myeloid-derived suppressor cells (MDSCs) comprise pathologically activated neutrophils and monocytes with potent immunosuppressive activity. They play an important part in inhibiting innate and adaptive immune responses, and have emerged as part of the immune response in sepsis. MDSCs numbers are persistently high in sepsis patients, and associated with nosocomial infections and other adverse clinical outcomes. However, their characteristics and functional mechanisms during sepsis have not been addressed fully. Our review sheds light on the features and suppressive mechanism of MDSCs. We also review the potential applications of MDSCs as biomarkers and targets for clinical treatment of sepsis.
Collapse
Affiliation(s)
- Wanying Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesiology and critical care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiangzhi Fang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenggang Gao
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoying Song
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yajun He
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Zhou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobo Yang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesiology and critical care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Jiqian Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesiology and critical care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
4
|
Chen MF, Chen PT, Hsieh CC, Wang CC. Effect of Proton Therapy on Tumor Cell Killing and Immune Microenvironment for Hepatocellular Carcinoma. Cells 2023; 12:cells12020332. [PMID: 36672266 PMCID: PMC9857172 DOI: 10.3390/cells12020332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/31/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Radiotherapy with proton therapy (PT) has dosimetric advantages over photon therapy, which helps to enlarge the therapeutic window of radiotherapy for hepatocellular carcinoma (HCC). We evaluated the response of HCC to PT and examined the underlying mechanisms. The human liver cancer cell lines HepG2 and HuH7 and the murine liver cancer cell line Hepa1-6 were selected for cell and animal experiments to examine the response induced by PT irradiation. Biological changes and the immunological response following PT irradiation were examined. In vitro experiments showed no significant difference in cell survival following PT compared with photon radiotherapy. In a murine tumor model, the tumors were obviously smaller in size 12 days after PT irradiation. The underlying changes included increased DNA damage, upregulated IL-6 levels, and a regulated immune tumor microenvironment. Protein analysis in vitro and in vivo showed that PT increased the level of programmed cell death ligand 1 (PD-L1) expressed in tumor cells and recruited myeloid-derived suppressor cells (MDSCs). The increase in PD-L1 was positively correlated with the irradiation dose. In Hepa1-6 syngeneic mouse models, the combination of PT with anti-PD-L1 increased tumor growth delay compared with PT alone, which was associated with increased tumor-infiltrating T cells and attenuated MDSC recruitment in the microenvironment. Furthermore, when PT was applied to the primary HCC tumor, anti-PD-L1 antibody-treated mice showed smaller synchronous unirradiated tumors. In conclusion, the response of HCC to PT was determined by tumor cell killing and the immunological response in the tumor microenvironment. The combination with the anti-PD-L1 antibody to enhance antitumor immunity was responsible for the therapeutic synergism for HCC treated with PT. Based on our results, we suggest that PT combined with anti-PD-L1 may be a promising therapeutic policy for HCC.
Collapse
Affiliation(s)
- Miao-Fen Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital at Linko, Taoyuan 333, Taiwan
- College of Medicine, Chang Gung University, Taoyuan333, Taiwan
- Correspondence: (M.-F.C.); (C.-C.W.); Tel.: +886-3-3281000 (ext. 7008) (M.-F.C.)
| | - Ping-Tsung Chen
- Department of Medical Oncology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Ching-Chuan Hsieh
- College of Medicine, Chang Gung University, Taoyuan333, Taiwan
- Department of Surgery, Chang Gung Memorial Hospital at Chiayi, Chiayi 613, Taiwan
| | - Chih-Chi Wang
- Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Correspondence: (M.-F.C.); (C.-C.W.); Tel.: +886-3-3281000 (ext. 7008) (M.-F.C.)
| |
Collapse
|
5
|
MDSCs Aggravate the Asthmatic Progression in Children and OVA-Allergic Mice by Regulating the Th1/Th2/Th17 Responses. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6157385. [PMID: 36045657 PMCID: PMC9423955 DOI: 10.1155/2022/6157385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/12/2022] [Accepted: 07/30/2022] [Indexed: 11/28/2022]
Abstract
Background Asthma is a chronic inflammatory disease of respiratory with serious risks for children. This study explored myeloid-derived suppressor cells (MDSCs) on the pathogenesis of asthmatic children and mice. Methods The clinical study enrolled 30 asthma, 20 pneumonia, and 20 control participants. The MDSCs, Th17 and Th1 cells percentage, and IL-4, IL-12, IL-10, and IFN-γ levels were detected by flow cytometry and ELISA. In experimental asthma, mice were divided into control, ovalbumin (OVA), and OVA + MDSCs groups. The changes in inflammatory cell count and the levels of IL-5, IL-12, and IL-10 in mice BALF and the levels of inflammatory factors, IgE, and IFN-γ in mice were detected by ELISA. The amount of ROS generation and pathological changes and the levels of caspase 1 and caspase 3 were tested by flow cytometry, HE and PAS staining, and immunohistochemistry. The expression of cleaved caspase 1/caspase 1 and cleaved caspase 3/caspase 3 was detected by western blot. Results In clinical trials, the levels of IL-12, IFN-γ, and Th1 percentage decreased in pneumonia and asthma children's peripheral blood, while the levels of IL-4 and IL-10 and the percentages MDSCs and Th17 increased. In asthma mice, pathological staining showed that asthma caused lung inflammation and damage, while the OVA + MDSC group was severer. Moreover, the percentages of eosinophils, neutrophils, lymphocytes, and the levels of inflammatory factors, IgE, ROS production, caspase 1, caspase 3, cleaved caspase 1/caspase 1, and cleaved caspase 3/caspase 3 increased in OVA + MDSC group, while the percentage of macrophages, IL-12, and IFN-γ levels reduced, illustrating that MDSCs exacerbated asthma. Conclusion Our study indicated that MDSCs could aggravate asthma by regulating the Th1/Th2/Th17 response.
Collapse
|
6
|
Festari MF, da Costa V, Rodríguez-Zraquia SA, Costa M, Landeira M, Lores P, Solari-Saquieres P, Kramer MG, Freire T. The tumour-associated Tn antigen fosters lung metastasis and recruitment of regulatory T cells in triple negative breast cancer. Glycobiology 2021; 32:366-379. [PMID: 34939098 DOI: 10.1093/glycob/cwab123] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/01/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
Cancer is a leading cause of death worldwide, accounting for nearly 10 million deaths. Among breast cancers (BC) subtypes, triple-negative (TN) BC, is characterized by metastatic progression and poor patient prognosis. Although, TNBC is initially sensitive to chemotherapy, many TNBC patients rapidly develop resistance, at which point metastatic disease is highly lethal. Cancer cells present phenotypic changes or molecular signatures that distinguish them from healthy cells. The Tn antigen (GalNAc-O-Thr/Ser), that constitutes a powerful tool as tumour marker, was recently reported to contribute to tumour growth. However, its role in BC-derived metastasis has not yet been addressed. In this work we generated a pre-clinical orthotopic Tn+ model of metastatic TNBC, that mimics the patient surgical treatment and is useful to study the role of Tn in metastasis and immunoregulation. We obtained two different cell clones which differed in their Tn antigen expression: a high Tn-expressing and a non-expressing clone. Interestingly, the Tn-positive cell line generated significantly larger tumours and higher degree of lung metastases associated with a lower survival rate than the Tn-negative and parental cell line. Furthermore, we also found that both tumours and draining-lymph nodes from Tn+-tumour bearing mice presented a higher frequency of CD4+ FoxP3+ T cells, while their splenocytes expressed higher levels of IL-10. In conclusion, this work suggests that the Tn antigen participates in breast tumour growth and spreading, favouring metastases to the lungs that are associated to an immunoregulatory state, suggesting that Tn-based immunotherapy could be a strategy of choice to treat these tumours.
Collapse
Affiliation(s)
- María Florencia Festari
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República, Montevideo, Uruguay
| | - Valeria da Costa
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República, Montevideo, Uruguay
| | - Santiago A Rodríguez-Zraquia
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República, Montevideo, Uruguay
| | - Monique Costa
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República, Montevideo, Uruguay
| | - Mercedes Landeira
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República, Montevideo, Uruguay
| | - Pablo Lores
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República, Montevideo, Uruguay
| | - Patricia Solari-Saquieres
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República, Montevideo, Uruguay
| | - M Gabriela Kramer
- Laboratorio de Bioensayos, Campus Interinstitucional, INIA-UdelaR, Tacuarembó; and Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Teresa Freire
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República, Montevideo, Uruguay
| |
Collapse
|
7
|
Chen HM, Sun L, Pan PY, Wang LH, Chen SH. Nutrient supplements from selected botanicals mediated immune modulation of the tumor microenvironment and antitumor mechanism. Cancer Immunol Immunother 2021; 70:3435-3449. [PMID: 33877384 DOI: 10.1007/s00262-021-02927-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 03/23/2021] [Indexed: 11/29/2022]
Abstract
Specific extracts of selected vegetables (SV) have been shown to benefit the survival of stage IIIb/IV non-small cell lung cancer patients in phase I/II studies and is currently in a phase III trial. However, the underlying mechanism of SV-mediated antitumor immune responses has not been elucidated. Our results indicate that SV modulated the NK and adoptive T cell immune responses in antitumor efficacy. Furthermore, antitumor effects of SV were also mediated by innate myeloid cell function, which requires both TLR and β-glucan signaling in a MyD88/TRIF and Dectin-1-dependent manner, respectively. Additionally, SV treatment reduced granulocytic myeloid-derived suppressor cell (MDSC) infiltration into the tumor and limited monocytic MDSC toward the M2-like functional phenotype. Importantly, SV treatment enhanced antigen-specific immune responses by augmenting the activation of antigen-specific TH1/TH17 cells in secondary lymphoid organs and proliferative response, as well as by reducing the Treg population in the tumor microenvironment, which was driven by SV-primed activated M-MDSC. Our results support the idea that SV can subvert immune-tolerance state in the tumor microenvironment and inhibit tumor growth. The present study suggests that features, such as easy accessibility, favorable clinical efficacy, no detectable side effects and satisfactory safety make SV a feasible, appealing and convincing adjuvant therapy for the treatment of cancer patients and prevent tumor recurrence and/or metastases.
Collapse
Affiliation(s)
- Hui-Ming Chen
- Department of Oncological Sciences, Icahn School of Medicine At Mount Sinai, New York, 10029, USA
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Cancer Center, Houston Methodist Research Institute, Houston, 77030, USA
| | - Linus Sun
- Department of Ophthalmology, Columbia University, New York, 10027, USA
| | - Ping-Ying Pan
- Department of Oncological Sciences, Icahn School of Medicine At Mount Sinai, New York, 10029, USA
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Cancer Center, Houston Methodist Research Institute, Houston, 77030, USA
| | - Lu-Hai Wang
- Institute of Integrated Medicine and Chinese Medical Research Center, China Medical University, Taichung, 40402, Taiwan, ROC.
| | - Shu-Hsia Chen
- Department of Oncological Sciences, Icahn School of Medicine At Mount Sinai, New York, 10029, USA.
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Cancer Center, Houston Methodist Research Institute, Houston, 77030, USA.
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, USA.
| |
Collapse
|
8
|
Fleuren EDG, Terry RL, Meyran D, Omer N, Trapani JA, Haber M, Neeson PJ, Ekert PG. Enhancing the Potential of Immunotherapy in Paediatric Sarcomas: Breaking the Immunosuppressive Barrier with Receptor Tyrosine Kinase Inhibitors. Biomedicines 2021; 9:1798. [PMID: 34944614 PMCID: PMC8698536 DOI: 10.3390/biomedicines9121798] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/22/2022] Open
Abstract
Despite aggressive surgery, chemotherapy, and radiotherapy, survival of children and adolescents and young adults (AYAs) with sarcoma has not improved significantly in the past four decades. Immune checkpoint inhibitors (ICIs) are an exciting type of immunotherapy that offer new opportunities for the treatment of paediatric and AYA sarcomas. However, to date, most children do not derive a benefit from this type of treatment as a monotherapy. The immunosuppressive tumour microenvironment is a major barrier limiting their efficacy. Combinations of ICIs, such as anti-PD-1 therapy, with targeted molecular therapies that have immunomodulatory properties may be the key to breaking through immunosuppressive barriers and improving patient outcomes. Preclinical studies have indicated that several receptor tyrosine kinase inhibitors (RTKi) can alter the tumour microenvironment and boost the efficacy of anti-PD-1 therapy. A number of these combinations have entered phase-1/2 clinical trials, mostly in adults, and in most instances have shown efficacy with manageable side-effects. In this review, we discuss the status of ICI therapy in paediatric and AYA sarcomas and the rationale for co-treatment with RTKis. We highlight new opportunities for the integration of ICI therapy with RTK inhibitors, to improve outcomes for children with sarcoma.
Collapse
Affiliation(s)
- Emmy D. G. Fleuren
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick 2031, Australia; (R.L.T.); (M.H.); (P.G.E.)
- School of Women’s and Children’s Health, UNSW Sydney, Randwick 2052, Australia
- Centre for Childhood Cancer Research, UNSW Sydney, Randwick 2031, Australia
| | - Rachael L. Terry
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick 2031, Australia; (R.L.T.); (M.H.); (P.G.E.)
- School of Women’s and Children’s Health, UNSW Sydney, Randwick 2052, Australia
| | - Deborah Meyran
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne 3000, Australia; (D.M.); (J.A.T.); (P.J.N.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3000, Australia
- Inserm, Université de Paris, U976 HIPI Unit, Institut de Recherche Saint-Louis, 75475 Paris, France
| | - Natacha Omer
- Translational Innate Immunotherapy, University of Queensland Diamantina Institute (UQDI), Brisbane 4102, Australia;
- Oncology Services Group, Queensland Children’s Hospital, Brisbane 4101, Australia
| | - Joseph A. Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne 3000, Australia; (D.M.); (J.A.T.); (P.J.N.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3000, Australia
| | - Michelle Haber
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick 2031, Australia; (R.L.T.); (M.H.); (P.G.E.)
- School of Women’s and Children’s Health, UNSW Sydney, Randwick 2052, Australia
| | - Paul J. Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne 3000, Australia; (D.M.); (J.A.T.); (P.J.N.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3000, Australia
| | - Paul G. Ekert
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick 2031, Australia; (R.L.T.); (M.H.); (P.G.E.)
- School of Women’s and Children’s Health, UNSW Sydney, Randwick 2052, Australia
- Centre for Childhood Cancer Research, UNSW Sydney, Randwick 2031, Australia
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne 3000, Australia; (D.M.); (J.A.T.); (P.J.N.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne 3000, Australia
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne 3052, Australia
| |
Collapse
|
9
|
Yaseen MM, Abuharfeil NM, Darmani H. Myeloid-derived suppressor cells and the pathogenesis of human immunodeficiency virus infection. Open Biol 2021; 11:210216. [PMID: 34753323 PMCID: PMC8580465 DOI: 10.1098/rsob.210216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
There are several mechanisms by which human immunodeficiency virus (HIV) can mediate immune dysfunction and exhaustion during the course of infection. Chronic immune activation, after HIV infection, seems to be a key driving force of such unwanted consequences, which in turn worsens the pathological status. In such cases, the immune system is programmed to initiate responses that counteract unwanted immune activation, for example through the expansion of myeloid-derived suppressor cells (MDSCs). Although the expansion of immune suppressor cells in the setting of systemic chronic immune activation, in theory, is expected to contain immune activation, HIV infection is still associated with a remarkably high level of biomarkers of immune activation. Paradoxically, the expansion of immune suppressor cells during HIV infection can suppress potent anti-viral immune responses, which in turn contribute to viral persistence and disease progression. This indicates that HIV hijacks not only immune activation but also the immune regulatory responses to its advantage. In this work, we aim to pave the way to comprehend how such unwanted expansion of MDSCs could participate in the pathology of acute/primary and chronic HIV infection in humans, as well as simian immunodeficiency virus infection in rhesus macaques, according to the available literature.
Collapse
Affiliation(s)
- Mahmoud Mohammad Yaseen
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Nizar Mohammad Abuharfeil
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Homa Darmani
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid 22110, Jordan
| |
Collapse
|
10
|
Cai F, Li S, Huang H, Iqbal J, Wang C, Jiang X. Green synthesis of gold nanoparticles for immune response regulation: Mechanisms, applications, and perspectives. J Biomed Mater Res A 2021; 110:424-442. [PMID: 34331516 DOI: 10.1002/jbm.a.37281] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 01/16/2023]
Abstract
Immune responses are involved in the pathogenesis of many diseases, including cancer, autoimmune diseases, and chronic inflammation. These responses are attributed to immune cells that produce cytokines, mediate cytotoxicity, and synthesize antibodies. Gold nanoparticles (GNPs) are novel agents that intervene with immune responses because of their unique physical-chemical properties. In particular, GNPs enhance anti-tumour activity during immunotherapy and eliminate excessive inflammation in autoimmune diseases. However, GNPs synthesized by conventional methods are toxic to living organisms. Green biosynthesis provides a safe and eco-friendly method to obtain GNPs from microbes or plant extracts. In this review, we describe several patterns for green GNP biosynthesis. The applications of GNPs to target immune cells and modulate the immune response are summarized. In particular, we elaborate on how GNPs regulate innate immunity and adaptive immunity, including inflammatory signaling and immune cell differentiation. Finally, perspectives and challenges in utilizing green biosynthesized GNPs for novel therapeutic approaches are discussed.
Collapse
Affiliation(s)
- Feiyang Cai
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, China.,School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyi Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Huang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Javed Iqbal
- Department of Botany, Bacha Khan University, Charsadda, Khyber Pakhtunkhwa, Pakistan
| | - Canran Wang
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xing Jiang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
11
|
Spiesschaert B, Angerer K, Park J, Wollmann G. Combining Oncolytic Viruses and Small Molecule Therapeutics: Mutual Benefits. Cancers (Basel) 2021; 13:3386. [PMID: 34298601 PMCID: PMC8306439 DOI: 10.3390/cancers13143386] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
The focus of treating cancer with oncolytic viruses (OVs) has increasingly shifted towards achieving efficacy through the induction and augmentation of an antitumor immune response. However, innate antiviral responses can limit the activity of many OVs within the tumor and several immunosuppressive factors can hamper any subsequent antitumor immune responses. In recent decades, numerous small molecule compounds that either inhibit the immunosuppressive features of tumor cells or antagonize antiviral immunity have been developed and tested for. Here we comprehensively review small molecule compounds that can achieve therapeutic synergy with OVs. We also elaborate on the mechanisms by which these treatments elicit anti-tumor effects as monotherapies and how these complement OV treatment.
Collapse
Affiliation(s)
- Bart Spiesschaert
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University Innsbruck, 6020 Innsbruck, Austria; (B.S.); (K.A.)
- Institute of Virology, Medical University Innsbruck, 6020 Innsbruck, Austria
- ViraTherapeutics GmbH, 6063 Rum, Austria
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach a.d. Riss, Germany;
| | - Katharina Angerer
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University Innsbruck, 6020 Innsbruck, Austria; (B.S.); (K.A.)
- Institute of Virology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - John Park
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach a.d. Riss, Germany;
| | - Guido Wollmann
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University Innsbruck, 6020 Innsbruck, Austria; (B.S.); (K.A.)
- Institute of Virology, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
12
|
Khalaf K, Hana D, Chou JTT, Singh C, Mackiewicz A, Kaczmarek M. Aspects of the Tumor Microenvironment Involved in Immune Resistance and Drug Resistance. Front Immunol 2021; 12:656364. [PMID: 34122412 PMCID: PMC8190405 DOI: 10.3389/fimmu.2021.656364] [Citation(s) in RCA: 209] [Impact Index Per Article: 69.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/27/2021] [Indexed: 12/11/2022] Open
Abstract
The tumor microenvironment (TME) is a complex and ever-changing "rogue organ" composed of its own blood supply, lymphatic and nervous systems, stroma, immune cells and extracellular matrix (ECM). These complex components, utilizing both benign and malignant cells, nurture the harsh, immunosuppressive and nutrient-deficient environment necessary for tumor cell growth, proliferation and phenotypic flexibility and variation. An important aspect of the TME is cellular crosstalk and cell-to-ECM communication. This interaction induces the release of soluble factors responsible for immune evasion and ECM remodeling, which further contribute to therapy resistance. Other aspects are the presence of exosomes contributed by both malignant and benign cells, circulating deregulated microRNAs and TME-specific metabolic patterns which further potentiate the progression and/or resistance to therapy. In addition to biochemical signaling, specific TME characteristics such as the hypoxic environment, metabolic derangements, and abnormal mechanical forces have been implicated in the development of treatment resistance. In this review, we will provide an overview of tumor microenvironmental composition, structure, and features that influence immune suppression and contribute to treatment resistance.
Collapse
Affiliation(s)
- Khalil Khalaf
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Doris Hana
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Jadzia Tin-Tsen Chou
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Chandpreet Singh
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Andrzej Mackiewicz
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Mariusz Kaczmarek
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
13
|
Barbosa Bomfim CC, Pinheiro Amaral E, Santiago-Carvalho I, Almeida Santos G, Machado Salles É, Hastreiter AA, Silva do Nascimento R, Almeida FM, Lopes Biá Ventura Simão T, Linhares Rezende A, Hiroyuki Hirata M, Ambrósio Fock R, Álvarez JM, Lasunskaia EB, D'Império Lima MR. Harmful Effects of Granulocytic Myeloid-Derived Suppressor Cells on Tuberculosis Caused by Hypervirulent Mycobacteria. J Infect Dis 2020; 223:494-507. [PMID: 33206171 DOI: 10.1093/infdis/jiaa708] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The role of myeloid-derived suppressor cells (MDSCs) in patients with severe tuberculosis who suffer from uncontrolled pulmonary inflammation caused by hypervirulent mycobacterial infection remains unclear. METHODS This issue was addressed using C57BL/6 mice infected with highly virulent Mycobacterium bovis strain MP287/03. RESULTS CD11b+GR1int population increased in the bone marrow, blood and lungs during advanced disease. Pulmonary CD11b+GR1int (Ly6GintLy6Cint) cells showed granularity similar to neutrophils and expressed immature myeloid cell markers. These immature neutrophils harbored intracellular bacilli and were preferentially located in the alveoli. T-cell suppression occurred concomitantly with CD11b+GR1int cell accumulation in the lungs. Furthermore, lung and bone marrow GR1+ cells suppressed both T-cell proliferation and interferon γ production in vitro. Anti-GR1 therapy given when MDSCs infiltrated the lungs prevented expansion and fusion of primary pulmonary lesions and the development of intragranulomatous caseous necrosis, along with increased mouse survival and partial recovery of T-cell function. Lung bacterial load was reduced by anti-GR1 treatment, but mycobacteria released from the depleted cells proliferated extracellularly in the alveoli, forming cords and clumps. CONCLUSIONS Granulocytic MDSCs massively infiltrate the lungs during infection with hypervirulent mycobacteria, promoting bacterial growth and the development of inflammatory and necrotic lesions, and are promising targets for host-directed therapies.
Collapse
Affiliation(s)
- Caio César Barbosa Bomfim
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Eduardo Pinheiro Amaral
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Igor Santiago-Carvalho
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Gislane Almeida Santos
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Érika Machado Salles
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Araceli Aparecida Hastreiter
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | | | - Fabrício M Almeida
- Laboratório de Biologia do Reconhecer, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Thatiana Lopes Biá Ventura Simão
- Laboratório de Biologia do Reconhecer, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Andreza Linhares Rezende
- Laboratório de Biologia do Reconhecer, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Mario Hiroyuki Hirata
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Ricardo Ambrósio Fock
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - José Maria Álvarez
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Elena B Lasunskaia
- Laboratório de Biologia do Reconhecer, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | | |
Collapse
|
14
|
Kalathil SG, Wang K, Hutson A, Iyer R, Thanavala Y. Tivozanib mediated inhibition of c-Kit/SCF signaling on Tregs and MDSCs and reversal of tumor induced immune suppression correlates with survival of HCC patients. Oncoimmunology 2020; 9:1824863. [PMID: 33101775 PMCID: PMC7553535 DOI: 10.1080/2162402x.2020.1824863] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The immune modulatory effect of tivozanib, a tyrosine kinase inhibitor, and the underlying immune mechanisms impacting survival of HCC patients have not been investigated. Pre-clinical studies have shown that tivozanib reduces Tregs and MDSCs accumulation through inhibition of c-Kit/SCF axis. We rationalized that c-Kit/SCF axis antagonism by tivozanib may reverse tumor-induced immune suppression in HCC patients. The frequency of circulating Tregs, MDSCs, CTLA-4+Tregs, PD-1+T cells, c-Kit+pERK-2+Tregs, and c-Kit+pERK-2+MDSCs were quantified in HCC patients at baseline and two time points during tivozanib treatment. We report for the first time that reduction in Tregs after tivozanib treatment and increased levels of baseline CD4+PD-1+T cells correlated with significant improvement in overall survival (OS) of the patients and these signatures may be potential biomarkers of prognostic significance. This immune modulation resulted from tivozanib-mediated blockade of c-Kit/SCF signaling, impacting ERK2 phosphorylation on Tregs and MDSCs. Low pre-treatment CD4+T cells: Treg ratio and reduction in the frequencies of Foxp3+c-Kit+pERK+Tregs after tivozanib treatment correlated significantly with progression free survival. In a comparative analysis of tivozanib vs sorafenib treatment in HCC patients, we demonstrate that decrease in the baseline numbers or frequencies of Foxp3+Tregs, MDSCs and exhausted T cells was significantly greater following tivozanib treatment. Additionally, greater increase in CD4+T cell: Treg ratio after tivozanib treatment was associated with significant improvement in OS compared to sorafenib treatment, highlighting the greater efficacy of tivozanib. These insights may help identify patients who likely would benefit from c-Kit/SCF antagonism and inform efforts to improve the efficacy of tivozanib in combination with immunotherapy.
Collapse
Affiliation(s)
- Suresh Gopi Kalathil
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, USA
| | - Katy Wang
- Department of Biostatistics & Bioinformatics Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, USA
| | - Alan Hutson
- Department of Biostatistics & Bioinformatics Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, USA
| | - Renuka Iyer
- Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, USA
| | - Yasmin Thanavala
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, USA
| |
Collapse
|
15
|
Hager S, Fittler FJ, Wagner E, Bros M. Nucleic Acid-Based Approaches for Tumor Therapy. Cells 2020; 9:E2061. [PMID: 32917034 PMCID: PMC7564019 DOI: 10.3390/cells9092061] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/24/2022] Open
Abstract
Within the last decade, the introduction of checkpoint inhibitors proposed to boost the patients' anti-tumor immune response has proven the efficacy of immunotherapeutic approaches for tumor therapy. Furthermore, especially in the context of the development of biocompatible, cell type targeting nano-carriers, nucleic acid-based drugs aimed to initiate and to enhance anti-tumor responses have come of age. This review intends to provide a comprehensive overview of the current state of the therapeutic use of nucleic acids for cancer treatment on various levels, comprising (i) mRNA and DNA-based vaccines to be expressed by antigen presenting cells evoking sustained anti-tumor T cell responses, (ii) molecular adjuvants, (iii) strategies to inhibit/reprogram tumor-induced regulatory immune cells e.g., by RNA interference (RNAi), (iv) genetically tailored T cells and natural killer cells to directly recognize tumor antigens, and (v) killing of tumor cells, and reprograming of constituents of the tumor microenvironment by gene transfer and RNAi. Aside from further improvements of individual nucleic acid-based drugs, the major perspective for successful cancer therapy will be combination treatments employing conventional regimens as well as immunotherapeutics like checkpoint inhibitors and nucleic acid-based drugs, each acting on several levels to adequately counter-act tumor immune evasion.
Collapse
Affiliation(s)
- Simone Hager
- Department of Chemistry and Pharmacy, Ludwig-Maximilians-University (LMU), 81377 Munich, Germany;
| | | | - Ernst Wagner
- Department of Chemistry and Pharmacy, Ludwig-Maximilians-University (LMU), 81377 Munich, Germany;
| | - Matthias Bros
- Department of Dermatology, University Medical Center, 55131 Mainz, Germany;
| |
Collapse
|
16
|
Stem cell factor produced by tumor cells expands myeloid-derived suppressor cells in mice. Sci Rep 2020; 10:11257. [PMID: 32647215 PMCID: PMC7347545 DOI: 10.1038/s41598-020-68061-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/16/2020] [Indexed: 12/30/2022] Open
Abstract
Immunotherapy is a novel treatment approach for cancers; however, its therapeutic effects are impeded by myeloid-derived suppressor cells (MDSCs). This study aimed to determine how MDSCs are expanded in cancer hosts. MDSCs were positive for Gr-1 and CD11b. Hepa1-6 hepatoma cells, EL4 lymphoma cells, and mice bearing Hepa1-6 hepatoma or lymphoma were examined. Following the inoculation of Hepa1-6 cells into the flanks of mice, a linear correlation was evident between the frequency of MDSCs in the spleen and tumor sizes. MDSC numbers diminished gradually and returned to the normal level within 3 weeks if the tumors were excised. To identify the cytokines produced by tumor cells that allowed expansion of MDSCs, cytokines in Hepa1-6 cell culture medium and murine serum were examined using a cytokine array. Stem cell factor (SCF) was implicated as the relevant cytokine. When recombinant SCF was added to the spleen cell culture medium, MDSC expansion could occur. In the presence of c-kit blockade, this effect of SCF was partially reversed. In conclusion, MDSCs can be expanded in tumor cells in a process that involves SCF released by tumor cells.
Collapse
|
17
|
Repurposing Food and Drug Administration-Approved Drugs to Promote Antitumor Immunity. ACTA ACUST UNITED AC 2020; 25:88-99. [PMID: 30896530 DOI: 10.1097/ppo.0000000000000368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
There has been a major resurgence of interest in immune-based approaches to treat cancer, based largely on the success of checkpoint inhibitors (anti-cytotoxic T-lymphocyte-associated antigen 4, anti-programmed cell death 1, and anti-programmed cell death ligand 1 antibodies) in several malignancies. However, not all tumors respond to checkpoint therapy, and there is clearly a need for additional approaches for enhancing tumor immunity. We summarize the critical elements necessary for mounting an efficacious T-cell response to a tumor. We cite drugs approved by the Food and Drug Administration for no-cancer indications that could be repurposed and used as part of an antitumor immune cocktail. We also list cancer drugs not initially intended to impact tumor immunity (soft repurposing) but that have been found to modulate the immune system. We highlight those drugs that might be used in combination with checkpoint inhibitors to increase response rates and survival of cancer patients. Our focus will be on drugs for which there are limited but existing human data. We cite supporting mechanistic mouse data as well. Repurposing drugs to modulate antitumor immunity is an opportunity to rapidly bring new, effective, and affordable treatments to cancer patients.
Collapse
|
18
|
|
19
|
Wang Y, Jia A, Bi Y, Wang Y, Liu G. Metabolic Regulation of Myeloid-Derived Suppressor Cell Function in Cancer. Cells 2020; 9:cells9041011. [PMID: 32325683 PMCID: PMC7226088 DOI: 10.3390/cells9041011] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a group of immunosuppressive cells that play crucial roles in promoting tumor growth and protecting tumors from immune recognition in tumor-bearing mice and cancer patients. Recently, it has been shown that the metabolic activity of MDSCs plays an important role in the regulation of their inhibitory function, especially in the processes of tumor occurrence and development. The MDSC metabolism, such as glycolysis, fatty acid oxidation and amino acid metabolism, is rewired in the tumor microenvironment (TME), which enhances the immunosuppressive activity, resulting in effector T cell apoptosis and suppressive cell proliferation. Herein, we summarized the recent progress in the metabolic reprogramming and immunosuppressive function of MDSCs during tumorigenesis.
Collapse
Affiliation(s)
- Yufei Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China; (Y.W.); (A.J.); (Y.W.)
| | - Anna Jia
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China; (Y.W.); (A.J.); (Y.W.)
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China;
| | - Yuexin Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China; (Y.W.); (A.J.); (Y.W.)
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China; (Y.W.); (A.J.); (Y.W.)
- Correspondence: ; Tel./Fax: +86-10-58800026
| |
Collapse
|
20
|
C-kit-derived CD11b + cells are critical for cardiac allograft prolongation by autologous C-kit + progenitor cells. Cell Immunol 2019; 347:104023. [PMID: 31836133 DOI: 10.1016/j.cellimm.2019.104023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 11/06/2019] [Accepted: 11/25/2019] [Indexed: 10/25/2022]
Abstract
Autologous C-kit+ cells robustly prolong cardiac allografts. As C-kit+ cells can transdifferentiate to hematopoietic cells as well as non-hematopoietic cells, we aimed to clarify the class(es) of C-kit-derived cell(s) required for cardiac allograft prolongation. Autologous C-kit+ cells were administered post-cardiac transplantation and allografts were evaluated for C-kit+ inoculum-derived cells. Results suggested that alloimmunity was a major signal for trafficking of C-kit-derived cells to the allograft and demonstrated that C-kit+ inoculum-derived cells expressed CD11b early after transfer. Allograft survival studies with CD11b-DTR C-kit+ cells demonstrated a requirement for C-kit+-derived CD11b+ cells. Co-therapy studies demonstrated near complete abrogation of acute rejection with concomitant CTLA4-Ig therapy and no loss of prolongation in combination with Cyclosporine A. These results strongly implicate a C-kit-derived myeloid population as critical for allograft preservation and demonstrate the potential therapeutic application of autologous C-kit+ progenitor cells as calcineurin inhibitor-sparing agents and possibly as co-therapeutics for durable graft survival.
Collapse
|
21
|
Han X, Wang R, Xu J, Chen Q, Liang C, Chen J, Zhao J, Chu J, Fan Q, Archibong E, Jiang L, Wang C, Liu Z. In situ thermal ablation of tumors in combination with nano-adjuvant and immune checkpoint blockade to inhibit cancer metastasis and recurrence. Biomaterials 2019; 224:119490. [DOI: 10.1016/j.biomaterials.2019.119490] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 08/11/2019] [Accepted: 09/11/2019] [Indexed: 12/27/2022]
|
22
|
Haas L, Obenauf AC. Allies or Enemies-The Multifaceted Role of Myeloid Cells in the Tumor Microenvironment. Front Immunol 2019; 10:2746. [PMID: 31849950 PMCID: PMC6892746 DOI: 10.3389/fimmu.2019.02746] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 11/08/2019] [Indexed: 12/21/2022] Open
Abstract
For decades, cancer was considered a disease driven by genetic mutations in tumor cells, therefore afflicting a single cell type. This simplified view was slowly replaced by the understanding that interactions between malignant cells and neighboring stromal and immune cells-the tumor microenvironment (TME)-profoundly shape cancer progression. This understanding paved the way for an entirely new form of therapy that targets the immune cell compartment, which has revolutionized the treatment of cancer. In particular, agents activating T lymphocytes have become a key focus of these therapies, as they can induce durable responses in several cancer types. However, T cell targeting agents only benefit a fraction of patients. Thus, it is crucial to identify the roles of other immune cell types in the TME and understand how they influence T cell function and/or whether they present valuable therapeutic targets themselves. In this review, we focus on the myeloid compartment of the TME, a heterogeneous mix of cell types with diverse effector functions. We describe how distinct myeloid cell types can act as enemies of cancer cells by inducing or enhancing an existing immune response, while others act as strong allies, supporting tumor cells in their malignant growth and establishing an immune evasive TME. Specifically, we focus on the role of myeloid cells in the response and resistance to immunotherapy, and how modulating their numbers and/or state could provide alternative therapeutic entry-points.
Collapse
Affiliation(s)
| | - Anna C. Obenauf
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| |
Collapse
|
23
|
Expansion of PMN-myeloid derived suppressor cells and their clinical relevance in patients with oral squamous cell carcinoma. Oral Oncol 2019; 95:157-163. [DOI: 10.1016/j.oraloncology.2019.06.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/28/2019] [Accepted: 06/02/2019] [Indexed: 02/08/2023]
|
24
|
Zhang Y, Xu B, Luan B, Zhang Y, Wang X, Xiong X, Shi H. Tumor-derived MDSCs inhibit airway remodeling in asthmatic mice through regulating IL-10 and IL-12. Am J Transl Res 2019; 11:4192-4202. [PMID: 31396328 PMCID: PMC6684897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 06/29/2019] [Indexed: 06/10/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs), a group of newly discovered and heterogeneous myeloid-derived immunosuppressive cells, play an important role in the progress of asthma, however, the specific mechanism is still largely unclear. Our previous study has indicated that during the onset of asthma, the accumulation of MDSCs and the level of serum interleukin (IL)-10 increased, while the level of IL-12 decreased. The present study aimed to investigate whether tumor-derived MDSCs could inhibit airway remodeling in asthmatic mice through regulating IL-10 and IL-12 secretion. To perform our investigation, we established a mouse model of breast cancer, and the extracted MDSCs from breast caner mouse model were injected into a mouse model of asthma induced by ovalbumin (OVA). Then, asthmatic airway remodeling of mice was analyzed and the levels of IL-10 and IL-12 in the serum and bronchoalveolar lavage fluid (BALF) of mice were detected. In addition, the correlation of MDSCs with the levels of IL-10 and IL-12 in the transplantation group was analyzed. The transplantation of tumor-derived MDSCs into asthmatic mice significantly improved airway remodeling, decreased MDSCs and the expression of IL-10, and significantly increased the expression of IL-12. Besides, we confirmed that IL-10 was positively correlated with MDSCs, while IL-12 was negatively correlated with MDSCs. The results indicated that tumor-derived MDSCs could reduce IL-10 level, increase the level of IL-12, and thus correct the Th1/Th2 imbalance in asthmatic mice. In summary, our results revealed that tumor-derived MDSCs could serve as a potential novel target for asthma therapy.
Collapse
Affiliation(s)
- Yanli Zhang
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou UniversityZhengzhou 450052, China
| | - Boyi Xu
- Institute of Psychology, Chinese Academy of SciencesBeijing 100101, China
| | - Bin Luan
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou UniversityZhengzhou 450052, China
| | - Yan Zhang
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou UniversityZhengzhou 450052, China
| | - Xiufang Wang
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou UniversityZhengzhou 450052, China
| | - Xiaorong Xiong
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou UniversityZhengzhou 450052, China
| | - Hongke Shi
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou UniversityZhengzhou 450052, China
| |
Collapse
|
25
|
Safa AR, Kamocki K, Saadatzadeh MR, Bijangi-Vishehsaraei K. c-FLIP, a Novel Biomarker for Cancer Prognosis, Immunosuppression, Alzheimer's Disease, Chronic Obstructive Pulmonary Disease (COPD), and a Rationale Therapeutic Target. BIOMARKERS JOURNAL 2019; 5:4. [PMID: 32352084 PMCID: PMC7189798 DOI: 10.36648/2472-1646.5.1.59] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dysregulation of c-FLIP (cellular FADD-like IL-1β-converting enzyme inhibitory protein) has been shown in several diseases including cancer, Alzheimer's disease, and chronic obstructive pulmonary disease (COPD). c-FLIP is a critical anti-cell death protein often overexpressed in tumors and hematological malignancies and its increased expression is often associated with a poor prognosis. c-FLIP frequently exists as long (c-FLIPL) and short (c-FLIPS) isoforms, regulates its anti-cell death functions through binding to FADD (FAS associated death domain protein), an adaptor protein known to activate caspases-8 and -10 and links c-FLIP to several cell death regulating complexes including the death-inducing signaling complex (DISC) formed by various death receptors. c-FLIP also plays a critical role in necroptosis and autophagy. Furthermore, c-FLIP is able to activate several pathways involved in cytoprotection, proliferation, and survival of cancer cells through various critical signaling proteins. Additionally, c-FLIP can inhibit cell death induced by several chemotherapeutics, anti-cancer small molecule inhibitors, and ionizing radiation. Moreover, c-FLIP plays major roles in aiding the survival of immunosuppressive tumor-promoting immune cells and functions in inflammation, Alzheimer's disease (AD), and chronic obstructive pulmonary disease (COPD). Therefore, c-FLIP can serve as a versatile biomarker for cancer prognosis, a diagnostic marker for several diseases, and an effective therapeutic target. In this article, we review the functions of c-FLIP as an anti-apoptotic protein and negative prognostic factor in human cancers, and its roles in resistance to anticancer drugs, necroptosis and autophagy, immunosuppression, Alzheimer's disease, and COPD.
Collapse
Affiliation(s)
- Ahmad R Safa
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, USA
| | - Krzysztof Kamocki
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, USA
| | - M Reza Saadatzadeh
- Department of Neurosurgery, Indiana University School of Medicine, Indianapolis, USA
| | | |
Collapse
|
26
|
Bryant J, Shields L, Hynes C, Howe O, McCleanc B, Lynga F. DNA Damage and Cytokine Production in Non-Target Irradiated Lymphocytes. Radiat Res 2019; 191:545-555. [DOI: 10.1667/rr15165.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Jane Bryant
- Radiation and Environmental Science Centre, FOCAS Institute
| | - Laura Shields
- Medical Physics Department, St. Luke's Radiation Oncology Centre, Rathgar, Dublin, Ireland
| | | | - Orla Howe
- School of Biological Sciences, Technological University Dublin, Dublin 8, Ireland
| | - Brendan McCleanc
- Medical Physics Department, St. Luke's Radiation Oncology Centre, Rathgar, Dublin, Ireland
| | - Fiona Lynga
- Radiation and Environmental Science Centre, FOCAS Institute
| |
Collapse
|
27
|
Umansky V, Adema GJ, Baran J, Brandau S, Van Ginderachter JA, Hu X, Jablonska J, Mojsilovic S, Papadaki HA, Pico de Coaña Y, Santegoets KCM, Santibanez JF, Serre K, Si Y, Sieminska I, Velegraki M, Fridlender ZG. Interactions among myeloid regulatory cells in cancer. Cancer Immunol Immunother 2019; 68:645-660. [PMID: 30003321 PMCID: PMC11028297 DOI: 10.1007/s00262-018-2200-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/04/2018] [Indexed: 12/14/2022]
Abstract
Mounting evidence has accumulated on the critical role of the different myeloid cells in the regulation of the cancerous process, and in particular in the modulation of the immune reaction to cancer. Myeloid cells are a major component of host cells infiltrating tumors, interacting with each other, with tumor cells and other stromal cells, and demonstrating a prominent plasticity. We describe here various myeloid regulatory cells (MRCs) in mice and human as well as their relevant therapeutic targets. We first address the role of the monocytes and macrophages that can contribute to angiogenesis, immunosuppression and metastatic dissemination. Next, we discuss the differential role of neutrophil subsets in tumor development, enhancing the dual and sometimes contradicting role of these cells. A heterogeneous population of immature myeloid cells, MDSCs, was shown to be generated and accumulated during tumor progression as well as to be an important player in cancer-related immune suppression. Lastly, we discuss the role of myeloid DCs, which can either contribute to effective anti-tumor responses or play a more regulatory role. We believe that MRCs play a critical role in cancer-related immune regulation and suggest that future anti-cancer therapies will focus on these abundant cells.
Collapse
Affiliation(s)
- Viktor Umansky
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht Karl University of Heidelberg, Mannheim, Germany.
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Jaroslaw Baran
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Kraków, Poland
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Jo A Van Ginderachter
- Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Xiaoying Hu
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht Karl University of Heidelberg, Mannheim, Germany
| | - Jadwiga Jablonska
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Slavko Mojsilovic
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Helen A Papadaki
- Department of Hematology, School of Medicine, University of Crete, Heraklion, Greece
| | - Yago Pico de Coaña
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kim C M Santegoets
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Juan F Santibanez
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | - Karine Serre
- Faculty of Medicine, Institute of Molecular Medicine (IMM)-João Lobo Antunes, University of Lisbon, Lisbon, Portugal
| | - Yu Si
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Isabela Sieminska
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Kraków, Poland
| | - Maria Velegraki
- Department of Hematology, School of Medicine, University of Crete, Heraklion, Greece
| | - Zvi G Fridlender
- Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
28
|
The prognosis of head and neck squamous cell carcinoma related to immunosuppressive tumor microenvironment regulated by IL-6 signaling. Oral Oncol 2019; 91:47-55. [PMID: 30926062 DOI: 10.1016/j.oraloncology.2019.02.027] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 01/26/2019] [Accepted: 02/23/2019] [Indexed: 12/17/2022]
Abstract
Evasion of immune surveillance is a significant factor in head and neck squamous cell carcinoma (HNSCC) carcinogenesis. IL-6 signaling is a critical mechanism for the induction of dysfunctional immune responses. In the present study, we examined the role of IL-6 in the prognosis of HNSCC regarding the immunosuppressive tumor microenvironment. We retrospectively analyzed the clinical outcomes of HNSCC patients and examined its correlation with the levels of IL-6 in tumors and circulating myeloid-derived suppressor cells (MDSCs) in peripheral blood. Furthermore, the relationships between IL and 6, programmed death ligand (PD-L1) expression, and immune response were examined in vitro and in vivo. Our data revealed that IL-6 overexpression was associated with the increased risk of developing disease failure and poor prognosis for HNSCC. The immunoreactivity of IL-6 in HNSCC specimens was positively linked to the staining of PD-L1 and the level of circulating MDSCs. By cellular and animal experiments, there were augmented radiation-induced increases in the expression of PD-L1 and the activation of MDSCs noted in IL-6-positive tumors. When IL-6 signaling was inhibited, the levels of PD-L1 and MDSC recruitment were significantly down-regulated. Furthermore, the neutrophil-to-lymphocyte ratio (NLR) was positively correlated with the levels of IL-6 and PD-L1 in tumor, and circulating MDSCs. In conclusion, IL-6 is a significant predictor of treatment outcome in HNSCC patients, and plays an important role in the induction of immunosuppressive tumor microenvironment mediated by increased MDSCs and PD-L1 expression. Furthermore, IL-6 combined with NLR can assist the clinician to make an informed decision regarding treatment options.
Collapse
|
29
|
Rossowska J, Anger N, Wegierek K, Szczygieł A, Mierzejewska J, Milczarek M, Szermer-Olearnik B, Pajtasz-Piasecka E. Antitumor Potential of Extracellular Vesicles Released by Genetically Modified Murine Colon Carcinoma Cells With Overexpression of Interleukin-12 and shRNA for TGF-β1. Front Immunol 2019; 10:211. [PMID: 30814999 PMCID: PMC6381037 DOI: 10.3389/fimmu.2019.00211] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/24/2019] [Indexed: 12/20/2022] Open
Abstract
Recent developments demonstrate that tumor-derived extracellular vesicles (EVs) could become a highly effective tool for delivery of antitumor factors. The main objective of the study was to determine whether EVs secreted by MC38 colon carcinoma cells genetically engineered for overproduction of interleukin (IL-)12 and/or shRNA targeting TGF-β1 are effectively loaded with these molecules and whether the obtained EVs could be an efficient tool for antitumor therapy. Fractions of EVs released by genetically modified MC38 cells [both modified tumor-derived exosomes (mTEx) and modified microvesicles (mTMv)] and those released by unmodified, wild-type MC38 cells were characterized in terms of loading efficacy, using real-time PCR and ELISA, as well as their antitumor potential. In order to examine the therapeutic potential of mTEx, they were applied in the form of sole treatment as well as in combination with dendritic cell (DC)-based vaccines stimulated with mTMv in the therapy of mice with subcutaneously growing MC38 tumors. The results demonstrated that genetic modification of wild-type MC38 tumor cells is an effective method of loading the molecules of interest into extracellular vesicles secreted by the cells (both TEx and TMv). The results also showed that mTEx secreted by cells engineered for overproduction of IL-12 and/or shRNA for TGF-β1 are able to induce tumor growth inhibition as opposed to TEx from unmodified MC38 cells. Additionally, antitumor therapy composed of mTEx (especially those deprived of TGF-β1) and DC-based vaccines allowed for regeneration of antitumor immunity and induction of the systemic Th1 response responsible for the sustained effect of the therapy. In conclusion, tumor-derived exosomes loaded with IL-12 and/or deprived of TGF-β1 could become an efficient adjuvant supporting induction of a specific antitumor response in both immuno- and chemotherapeutic schemes of treatment.
Collapse
Affiliation(s)
- Joanna Rossowska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Natalia Anger
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Katarzyna Wegierek
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Agnieszka Szczygieł
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Jagoda Mierzejewska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Magdalena Milczarek
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Bożena Szermer-Olearnik
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Elżbieta Pajtasz-Piasecka
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
30
|
Lee SE, Lim JY, Kim TW, Ryu DB, Park SS, Jeon YW, Yoon JH, Cho BS, Eom KS, Kim YJ, Kim HJ, Lee S, Cho SG, Kim DW, Lee JW, Min CK. Different role of circulating myeloid-derived suppressor cells in patients with multiple myeloma undergoing autologous stem cell transplantation. J Immunother Cancer 2019; 7:35. [PMID: 30732646 PMCID: PMC6367772 DOI: 10.1186/s40425-018-0491-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/20/2018] [Indexed: 12/16/2022] Open
Abstract
Background The aim of this study is to evaluate the prognostic impact of myeloid-derived suppressor cells (MDSCs) in multiple myeloma (MM) in the context of autologous stem cell transplantation (ASCT). Methods Peripheral blood samples were collected for measuring monocytic (M-) MDSCs (CD14posHLA-DRlow/neg) and early-stage (E-) MDSCs (LinnegHLA-DRnegCD33posCD11bpos) before and after ASCT. Clinical outcomes following ASCT differed according to the frequency of each MDSC phenotype. Results In the pre-ASCT analyses, lower M-MDSCs (<median) but not E-MDSCs were associated with a longer time to progression (TTP), whereas both MDSC phenotypes post-ASCT did not have a role in TTP. Both MDSC phenotypes pre-ASCT but not post-ASCT similarly suppressed in vitro autologous T and natural killer T cell proliferation. Importantly, pre-ASCT M-MDSCs more strongly inhibited the in vitro cytotoxic effect of melphalan compared with pre-ASCT E-MDSCs. Transcriptome analysis of each isolated MDSC subtype showed that expression of osteoclastic differentiation factors, particularly colony-stimulating factor 1 receptor (CSF1R), was significantly increased in M-MDSCs pre-ASCT. Finally, blockade of CSF1R substantially recovered the melphalan-induced cytotoxicity reduced by pre-ASCT M-MDSCs. Conclusions Our data demonstrate that pre-ASCT M-MDSCs are correlated with poor clinical outcomes after ASCT through reduced cytotoxicity of melphalan. We propose that targeting CSF1R on these cells may improve the results of ASCT in MM. Electronic supplementary material The online version of this article (10.1186/s40425-018-0491-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sung-Eun Lee
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, Korea
| | - Ji-Young Lim
- Leukemia Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Tae Woo Kim
- Leukemia Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Da-Bin Ryu
- Leukemia Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Sung Soo Park
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, Korea
| | - Young-Woo Jeon
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, Korea
| | - Jae-Ho Yoon
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, Korea
| | - Byung-Sik Cho
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, Korea.,Leukemia Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Ki-Seong Eom
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, Korea.,Leukemia Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Yoo-Jin Kim
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, Korea.,Leukemia Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Hee-Je Kim
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, Korea.,Leukemia Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Seok Lee
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, Korea.,Leukemia Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Seok-Goo Cho
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, Korea
| | - Dong-Wook Kim
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, Korea.,Leukemia Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Jong Wook Lee
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, Korea
| | - Chang-Ki Min
- Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodae-ro, Seocho-gu, Seoul, 06591, Korea. .,Leukemia Research Institute, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
31
|
Specific inhibition of PI3Kδ/γ enhances the efficacy of anti-PD1 against osteosarcoma cancer. J Bone Oncol 2018; 16:100206. [PMID: 31334002 PMCID: PMC6617297 DOI: 10.1016/j.jbo.2018.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/03/2018] [Accepted: 11/05/2018] [Indexed: 02/06/2023] Open
Abstract
Impressive responses have been observed in patients with cancer treated with checkpoint inhibitory anti-programmed cell death-1 (PD-1) or anti-cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) antibodies through disinhibiting the immune system. However, tumors possess complex immunosuppressive tumor microenvironment to present therapeutic obstacles and the response rates to immune checkpoint inhibition remain low. One significant barrier to the efficacy of anti-PD1 treatment is the recruitment of myeloid-derived suppressor cells (MDSCs) into the tumor. MDSCs dramatically increased in peripheral blood of patients with osteosarcoma and prohibited both T-cell activation and infiltration. Here we demonstrated functional inhibition of G-MDSCs with (S)-(-)-N-[2-(3-Hydroxy-1H-indol-3-yl)-methyl]-acetamide (SNA), a specific inhibitor of PI3Kδ/γ, could prime tumor microenvironment, resultantly enhancing the therapeutic efficacy of anti-PD1 treatment in a syngeneic osteosarcoma tumor model. Combining SNA with anti-PD1 dramatically slowed osteosarcoma tumor growth and prolonged survival time of tumor-bearing mice, at least in part mediated through CD8+ T cells. Our results demonstrated that addition of SNA to anti-PD1 significantly altered infiltration and function of innate immune cells, providing the rationale for combination therapy in patients with osteosarcoma through inhibiting the function of MDSCs with a selective PI3Kδ/γ inhibitor to enhance responses to immune checkpoint blockade.
Collapse
|
32
|
Chen Y, Sun J, Huang Y, Lu B, Li S. Improved Cancer Immunochemotherapy via Optimal Co-delivery of Chemotherapeutic and Immunomodulatory Agents. Mol Pharm 2018; 15:5162-5173. [PMID: 30222360 DOI: 10.1021/acs.molpharmaceut.8b00717] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
It is highly demanded and still a big challenge to develop an effective formulation for immunochemotherapy against advanced tumors. We have previously reported a PEG-NLG-based immunostimulatory nanocarrier (PEG2k-Fmoc-NLG919) for co-delivery of an IDO1 inhibitor (NLG919) and a chemotherapeutic agent (paclitaxel, PTX). Although antitumor immune responses were enhanced with a PTX-loaded nanocarrier, the accumulation of myeloid-derived suppressor cells (MDSCs) was also significantly increased, which may limit the overall efficacy of therapy. In the present work, we developed an improved dual-functional nanocarrier (PEG5k-Fmoc-NLG2) to co-load PTX and sunitinib (SUN, a multitarget receptor tyrosine kinase inhibitor) for improved cancer immunochemotherapy. We found that the recruited MDSCs negatively impacted the overall antitumor activity of the PTX-loaded PEG-NLG nanocarrier. Mechanistic study suggests that this is likely attributed to the PTX-mediated induction of a number of chemokines that are involved in the recruitment of MDSCs. We have further shown that the induction of these chemokines was drastically blocked by SUN. Co-delivery of PTX and SUN via the PEG5k-Fmoc-NLG9192 nanocarrier led to a further improvement in the therapeutic efficacy with a concomitant reduction in MDSCs.
Collapse
Affiliation(s)
- Yichao Chen
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy , University of Pittsburgh , Pittsburgh , Pennsylvania 15261 , United States
| | - Jingjing Sun
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy , University of Pittsburgh , Pittsburgh , Pennsylvania 15261 , United States
| | - Yixian Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy , University of Pittsburgh , Pittsburgh , Pennsylvania 15261 , United States
| | - Binfeng Lu
- Department of Immunology , University of Pittsburgh School of Medicine , 200 Lothrop Street , Pittsburgh , Pennsylvania 15261 , United States
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy , University of Pittsburgh , Pittsburgh , Pennsylvania 15261 , United States
| |
Collapse
|
33
|
Lin H, Wu Y, Chen J, Huang S, Wang Y. (−)-4-O-(4-O-β-D-glucopyranosylcaffeoyl) Quinic Acid Inhibits the Function of Myeloid-Derived Suppressor Cells to Enhance the Efficacy of Anti-PD1 against Colon Cancer. Pharm Res 2018; 35:183. [DOI: 10.1007/s11095-018-2459-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 07/09/2018] [Indexed: 01/04/2023]
|
34
|
Humphreys L, Espona-Fiedler M, Longley DB. FLIP as a therapeutic target in cancer. FEBS J 2018; 285:4104-4123. [PMID: 29806737 DOI: 10.1111/febs.14523] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/11/2018] [Accepted: 05/24/2018] [Indexed: 12/13/2022]
Abstract
One of the classic hallmarks of cancer is disruption of cell death signalling. Inhibition of cell death promotes tumour growth and metastasis, causes resistance to chemo- and radiotherapies as well as targeted agents, and is frequently due to overexpression of antiapoptotic proteins rather than loss of pro-apoptotic effectors. FLIP is a major apoptosis-regulatory protein frequently overexpressed in solid and haematological cancers, in which its high expression is often correlated with poor prognosis. FLIP, which is expressed as long (FLIP(L)) and short (FLIP(S)) splice forms, achieves its cell death regulatory functions by binding to FADD, a critical adaptor protein which links FLIP to the apical caspase in the extrinsic apoptotic pathway, caspase-8, in a number of cell death regulating complexes, such as the death-inducing signalling complexes (DISCs) formed by death receptors. FLIP also plays a key role (together with caspase-8) in regulating another form of cell death termed programmed necrosis or 'necroptosis', as well as in other key cellular processes that impact cell survival, including autophagy. In addition, FLIP impacts activation of the intrinsic mitochondrial-mediated apoptotic pathway by regulating caspase-8-mediated activation of the pro-apoptotic Bcl-2 family member Bid. It has been demonstrated that FLIP can not only inhibit death receptor-mediated apoptosis, but also cell death induced by a range of clinically relevant chemotherapeutic and targeted agents as well as ionizing radiation. More recently, key roles for FLIP in promoting the survival of immunosuppressive tumour-promoting immune cells have been discovered. Thus, FLIP is of significant interest as an anticancer therapeutic target. In this article, we review FLIP's biology and potential ways of targeting this important tumour and immune cell death regulator.
Collapse
Affiliation(s)
- Luke Humphreys
- Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| | - Margarita Espona-Fiedler
- Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| | - Daniel B Longley
- Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| |
Collapse
|
35
|
Yang G, Shen W, Zhang Y, Liu M, Zhang L, Liu Q, Lu HH, Bo J. Accumulation of myeloid-derived suppressor cells (MDSCs) induced by low levels of IL-6 correlates with poor prognosis in bladder cancer. Oncotarget 2018; 8:38378-38388. [PMID: 28418913 PMCID: PMC5503539 DOI: 10.18632/oncotarget.16386] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/27/2017] [Indexed: 01/22/2023] Open
Abstract
Bladder cancer (BC) is one of the most commonly occurring cancers, with a high recurrence rate and poor outcomes in cases of relapsed metastatic disease. Here, we analyzed the markers and significance of myeloid-derived suppressor cells (MDSCs) for BC development and progression. MDSC markers were examined in peripheral blood from 113 BC patients and 20 healthy volunteers. We identified CD11b+CD33lowHLA-DR- CD3- cells as markers of MDSCs in peripheral blood from BC patients. We also demonstrated that MDSC numbers are higher in BC patients than healthy donors, and that MDSC numbers correlate with the clinical grade, stage, and poor prognosis. In addition, serum IL-6 levels are decreased in BC patients with higher MDSC counts. IL-6 blockade increases induction of MDSCs in vitro. Low IL-6 levels inhibit activation of Stat3, resulting in the increased formation of MDSCs in BC. These results indicate that the MDSCs numbers may serve as a novel prognostic marker in BC patients, and that targeting IL-6 signaling may be a promising strategy for BC treatment.
Collapse
Affiliation(s)
- Guoliang Yang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenyan Shen
- Department of laboratory medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Mengyao Liu
- Clinical Stem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lianhua Zhang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Liu
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Hui Lu
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Juanjie Bo
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
36
|
Schafer CC, Wang Y, Hough KP, Sawant A, Grant SC, Thannickal VJ, Zmijewski J, Ponnazhagan S, Deshane JS. Indoleamine 2,3-dioxygenase regulates anti-tumor immunity in lung cancer by metabolic reprogramming of immune cells in the tumor microenvironment. Oncotarget 2018; 7:75407-75424. [PMID: 27705910 PMCID: PMC5340181 DOI: 10.18632/oncotarget.12249] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 09/13/2016] [Indexed: 12/22/2022] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) has been implicated in immune evasion by tumors. Upregulation of this tryptophan (Trp)-catabolizing enzyme, in tumor cells and myeloid-derived suppressor cells (MDSCs) within the tumor microenvironment (TME), leads to Trp depletion that impairs cytotoxic T cell responses and survival; however, exact mechanisms remain incompletely understood. We previously reported that a combination therapy of gemcitabine and a superoxide dismutase mimetic promotes anti-tumor immunity in a mouse model of lung cancer by inhibiting MDSCs, enhancing polyfunctional response of CD8+ memory T cells, and extending survival. Here, we show that combination therapy targets IDO signaling, specifically in MDSCs, tumor cells, and CD8+ T cells infiltrating the TME. Deficiency of IDO caused significant reduction in tumor burden, tumor-infiltrating MDSCs, GM-CSF, MDSC survival and infiltration of programmed death receptor-1 (PD-1)-expressing CD8+ T cells compared to controls. IDO−/− MDSCs downregulated nutrient-sensing AMP-activated protein kinase (AMPK) activity, but IDO−/− CD8+ T cells showed AMPK activation associated with enhanced effector function. Our studies provide proof-of-concept for the efficacy of this combination therapy in inhibiting IDO and T cell exhaustion in a syngeneic model of lung cancer and provide mechanistic insights for IDO-dependent metabolic reprogramming of MDSCs that reduces T cell exhaustion and regulates anti-tumor immunity.
Collapse
Affiliation(s)
- Cara C Schafer
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yong Wang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kenneth P Hough
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anandi Sawant
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stefan C Grant
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victor J Thannickal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jaroslaw Zmijewski
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Jessy S Deshane
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
37
|
Belyaev NN, Abdolla N, Perfilyeva YV, Ostapchuk YO, Krasnoshtanov VK, Kali A, Tleulieva R. Daunorubicin conjugated with alpha-fetoprotein selectively eliminates myeloid-derived suppressor cells (MDSCs) and inhibits experimental tumor growth. Cancer Immunol Immunother 2018; 67:101-111. [PMID: 28956104 PMCID: PMC11028061 DOI: 10.1007/s00262-017-2067-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 09/18/2017] [Indexed: 12/17/2022]
Abstract
Failure of antitumor immunity in cancer was shown to be mediated by myeloid-derived suppressor cells (MDSCs), which are considered to be one of the key factors contributing to the development of malignant diseases. Therefore, the development of pharmacological approaches to effectively eliminate MDSCs in organisms carrying growing tumors is a promising pathway for potential treatment. For this purpose we propose alpha-fetoprotein (AFP) conjugated with a cytotoxic agent as a vector molecule, specifically recognizing MDSCs. The present study was aimed at examination of this suggestion using both in vitro and in vivo approaches. MDSCs, obtained from the spleen of Ehrlich carcinoma bearing mice, selectively bound AFP labeled with fluorescein isothiocyanate. AFP conjugated to daunorubicin (AFP-DR) and DR alone showed similar in vitro cytotoxicity against the granulocytic MDSC subpopulation. The monocytic MDSC subpopulation was resistant to treatment with DR, whereas it was completely depleted in the presence of AFP-DR. Treatment of mice bearing Ehrlich carcinoma with AFP-DR resulted in reduced numbers of splenic MDSCs, normalization of NK cell levels, and inhibition of tumor growth. The obtained results demonstrate that cytotoxic conjugates based on AFP are promising anticancer drugs, which, in addition to the direct effect on tumor cells expressing receptors to AFP, may contribute to elimination of MDSCs.
Collapse
Affiliation(s)
- Nikolai N Belyaev
- Laboratory of Molecular Immunology and Immunobiotechnology, M. A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, 86 Dosmukhamedov St., Almaty, 050012, Kazakhstan.
| | - Nurshat Abdolla
- Laboratory of Molecular Immunology and Immunobiotechnology, M. A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, 86 Dosmukhamedov St., Almaty, 050012, Kazakhstan
- Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Yuliya V Perfilyeva
- Laboratory of Molecular Immunology and Immunobiotechnology, M. A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, 86 Dosmukhamedov St., Almaty, 050012, Kazakhstan
| | - Yekaterina O Ostapchuk
- Laboratory of Molecular Immunology and Immunobiotechnology, M. A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, 86 Dosmukhamedov St., Almaty, 050012, Kazakhstan
| | | | - Aikyn Kali
- Laboratory of Molecular Immunology and Immunobiotechnology, M. A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, 86 Dosmukhamedov St., Almaty, 050012, Kazakhstan
| | - Raikhan Tleulieva
- Laboratory of Molecular Immunology and Immunobiotechnology, M. A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, 86 Dosmukhamedov St., Almaty, 050012, Kazakhstan
| |
Collapse
|
38
|
Schupp J, Krebs FK, Zimmer N, Trzeciak E, Schuppan D, Tuettenberg A. Targeting myeloid cells in the tumor sustaining microenvironment. Cell Immunol 2017; 343:103713. [PMID: 29129292 DOI: 10.1016/j.cellimm.2017.10.013] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/26/2017] [Accepted: 10/26/2017] [Indexed: 12/24/2022]
Abstract
Myeloid cells are the most abundant cells in the tumor microenvironment (TME). The tumor recruits and modulates endogenous myeloid cells to tumor-associated macrophages (TAM), dendritic cells (DC), myeloid-derived suppressor cells (MDSC) and neutrophils (TAN), to sustain an immunosuppressive environment. Pathologically overexpressed mediators produced by cancer cells like granulocyte-macrophage colony-stimulating- and vascular endothelial growth factor induce myelopoiesis in the bone marrow. Excess of myeloid cells in the blood, periphery and tumor has been associated with tumor burden. In cancer, myeloid cells are kept at an immature state of differentiation to be diverted to an immunosuppressive phenotype. Here, we review human myeloid cells in the TME and the mechanisms for sustaining the hallmarks of cancer. Simultaneously, we provide an introduction into current and novel therapeutic approaches to redirect myeloid cells from a cancer promoting to a rather inflammatory, cancer inhibiting phenotype. In addition, the role of platelets for tumor promotion is discussed.
Collapse
Affiliation(s)
- Jonathan Schupp
- Department of Dermatology, University Medical Center, Mainz, Germany
| | - Franziska K Krebs
- Department of Dermatology, University Medical Center, Mainz, Germany; German Cancer Consortium (DKTK), partner site Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Niklas Zimmer
- Department of Dermatology, University Medical Center, Mainz, Germany
| | - Emily Trzeciak
- The Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
39
|
Safarzadeh E, Orangi M, Mohammadi H, Babaie F, Baradaran B. Myeloid-derived suppressor cells: Important contributors to tumor progression and metastasis. J Cell Physiol 2017; 233:3024-3036. [DOI: 10.1002/jcp.26075] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 06/28/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Elham Safarzadeh
- Immunology Research Center; Tabriz University of Medical Sciences; Tabriz Iran
- Faculty of Medicine, Department of Immunology; Tabriz University of Medical Sciences; Tabriz Iran
| | - Mona Orangi
- Immunology Research Center; Tabriz University of Medical Sciences; Tabriz Iran
- Faculty of Medicine, Department of Immunology; Tabriz University of Medical Sciences; Tabriz Iran
| | - Hamed Mohammadi
- Immunology Research Center; Tabriz University of Medical Sciences; Tabriz Iran
- Faculty of Medicine, Department of Immunology; Tabriz University of Medical Sciences; Tabriz Iran
| | - Farhad Babaie
- Immunology Research Center; Tabriz University of Medical Sciences; Tabriz Iran
- Faculty of Medicine, Department of Immunology; Tabriz University of Medical Sciences; Tabriz Iran
| | - Behzad Baradaran
- Immunology Research Center; Tabriz University of Medical Sciences; Tabriz Iran
- Faculty of Medicine, Department of Immunology; Tabriz University of Medical Sciences; Tabriz Iran
| |
Collapse
|
40
|
Stahl M, Gedrich R, Peck R, LaVallee T, Eder JP. Targeting KIT on innate immune cells to enhance the antitumor activity of checkpoint inhibitors. Immunotherapy 2017; 8:767-74. [PMID: 27349976 DOI: 10.2217/imt-2016-0040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Innate immune cells such as mast cells and myeloid-derived suppressor cells are key components of the tumor microenvironment. Recent evidence indicates that levels of myeloid-derived suppressor cells in melanoma patients are associated with poor survival to checkpoint inhibitors. This suggests that targeting both the innate and adaptive suppressive components of the immune system will maximize clinical benefit and elicit more durable responses in cancer patients. Preclinical data suggest that targeting signaling by the receptor tyrosine kinase KIT, particularly on mast cells, may modulate innate immune cell numbers and activity in tumors. Here, we review data highlighting the importance of the KIT signaling in regulating antitumor immune responses and the potential benefit of combining selective KIT inhibitors with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Maximilian Stahl
- Department of Internal Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, USA
| | - Richard Gedrich
- Kolltan Pharmaceuticals, 300 George Street Suite 530, New Haven, CT, USA
| | - Ronald Peck
- Kolltan Pharmaceuticals, 300 George Street Suite 530, New Haven, CT, USA
| | - Theresa LaVallee
- Kolltan Pharmaceuticals, 300 George Street Suite 530, New Haven, CT, USA
| | - Joseph Paul Eder
- Department of Medical Oncology & Yale Cancer Center, Yale School of Medicine, 333 Cedar Street, WWW211, New Haven, CT 06520, USA
| |
Collapse
|
41
|
Chesney JA, Mitchell RA, Yaddanapudi K. Myeloid-derived suppressor cells-a new therapeutic target to overcome resistance to cancer immunotherapy. J Leukoc Biol 2017; 102:727-740. [PMID: 28546500 DOI: 10.1189/jlb.5vmr1116-458rrr] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 12/12/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that accumulate during pathologic conditions, such as cancer. Patients diagnosed with advanced metastatic cancers have an average survival of 12-24 mo, a survival time that hasn't changed significantly in the past 30 yr. Despite some encouraging improvements in response rates and overall survival in patients receiving immunotherapies, such as immune checkpoint inhibitors, most patients will ultimately progress. MDSCs contribute to immunotherapeutic resistance by actively inhibiting antitumor T cell proliferation and cytotoxic activity as well as by promoting expansion of protumorigenic T regulatory cells, thereby, dampening the host immune responses against the tumor. In addition, MDSCs promote angiogenesis, tumor invasion, and metastasis. Thus, MDSCs are potential therapeutic targets in cases of multiple cancers. This review focuses on the phenotypic and functional characteristics of MDSCs and provides an overview of the mono- and combinatorial-therapeutic strategies that target MDSCs with an objective of enhancing the efficacy of cancer immunotherapies.
Collapse
Affiliation(s)
- Jason A Chesney
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA; .,Department of Medicine, University of Louisville, Louisville, Kentucky, USA; and
| | - Robert A Mitchell
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA; .,Department of Medicine, University of Louisville, Louisville, Kentucky, USA; and.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Kavitha Yaddanapudi
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA; .,Department of Medicine, University of Louisville, Louisville, Kentucky, USA; and.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
42
|
Hassel JC, Jiang H, Bender C, Winkler J, Sevko A, Shevchenko I, Halama N, Dimitrakopoulou-Strauss A, Haefeli WE, Jäger D, Enk A, Utikal J, Umansky V. Tadalafil has biologic activity in human melanoma. Results of a pilot trial with Tadalafil in patients with metastatic Melanoma (TaMe). Oncoimmunology 2017; 6:e1326440. [PMID: 28932631 PMCID: PMC5599085 DOI: 10.1080/2162402x.2017.1326440] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/27/2017] [Accepted: 04/29/2017] [Indexed: 01/01/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are known to play a critical role in the suppression of T cell antitumor responses. Our preclinical data showed that the phosphodiesterase (PDE)-5 inhibitor sildenafil impaired MDSC functions, enhanced intratumoral T cell activity and prolonged survival of melanoma-bearing mice. In this study, we evaluated biologic effects, safety and efficacy of palliative treatment with the PDE-5 inhibitor tadalafil in metastatic melanoma patients. We conducted an open-label, dose de-escalation trial with tadalafil in pretreated metastatic melanoma patients. Tumor and peripheral blood samples were taken before and 4 weeks after the start of treatment. Samples were investigated by immunohistochemistry and FACS analysis, for different immune subsets with numbers of CD8+ tumor-infiltrating lymphocytes (TIL) as primary end point. Stable disease was achieved in 3/12 patients (25%). Median progression-free survival was 4.6 mo (range 0.7–7.1), median overall survival (OS) 8.5 mo (range 2.7–23.7). The treatment was well tolerated. Stable patients displayed significantly higher numbers of CD8+ TIL in the center of metastases before treatment as compared with progressive patients. Upon the therapy, they showed increased expression of ζ-chain (used as a marker of T cell activation) in CD8+ and CD4+TILs and CD8+T cells in the peripheral blood as compared with baseline. Our study suggests that the PDE-5 inhibitor tadalafil can improve clinical outcome of advanced melanoma patients by enhancing antitumor immunity and highlights its potential application in combined melanoma immunotherapy.
Collapse
Affiliation(s)
- Jessica C Hassel
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Huanhuan Jiang
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Heidelberg, Germany.,Department of Obstetrics and Gynecology, Center for Reproductive Medicine, The First Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Carolin Bender
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Julia Winkler
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexandra Sevko
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Heidelberg, Germany
| | - Ivan Shevchenko
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Heidelberg, Germany
| | - Niels Halama
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Antonia Dimitrakopoulou-Strauss
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ) and University Hospital Heidelberg, Heidelberg, Germany
| | - Walter E Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexander Enk
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Heidelberg, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
43
|
Rico MJ, Perroud HA, Herrera C, Alasino CM, Roggero EA, Pezzotto SM, Nocito AL, Rozados VR, Scharovsky OG. Putative Biomarkers of Response to Treatment in Breast Cancer Patients: A Pilot Assay. Cancer Invest 2017; 35:377-385. [DOI: 10.1080/07357907.2017.1309545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- María J. Rico
- Institute of Experimental Genetics, School of Medicine, National University of Rosario, Rosario, Argentina
- National Scientific and Technological Research Council (CONICET), Rosario, Argentina
| | - Herman A. Perroud
- Institute of Experimental Genetics, School of Medicine, National University of Rosario, Rosario, Argentina
- National Scientific and Technological Research Council (CONICET), Rosario, Argentina
| | - Cintia Herrera
- Institute of Experimental Genetics, School of Medicine, National University of Rosario, Rosario, Argentina
| | | | - Eduardo A. Roggero
- Institute of Experimental Genetics, School of Medicine, National University of Rosario, Rosario, Argentina
| | - Stella M. Pezzotto
- Institute of Immunology, School of Medical Sciences, National University of Rosario, Rosario, Argentina
- National University of Rosario Research Council (CIUNR), Rosario, Argentina
| | - Ana Lía Nocito
- Department of Pathological Anatomy and Physiology, School of Medical Sciences, National University of Rosario, Rosario, Argentina
| | - Viviana R. Rozados
- Institute of Experimental Genetics, School of Medicine, National University of Rosario, Rosario, Argentina
| | - O. Graciela Scharovsky
- Institute of Experimental Genetics, School of Medicine, National University of Rosario, Rosario, Argentina
- National Scientific and Technological Research Council (CONICET), Rosario, Argentina
- National University of Rosario Research Council (CIUNR), Rosario, Argentina
| |
Collapse
|
44
|
Whiteside TL, Demaria S, Rodriguez-Ruiz ME, Zarour HM, Melero I. Emerging Opportunities and Challenges in Cancer Immunotherapy. Clin Cancer Res 2016; 22:1845-55. [PMID: 27084738 DOI: 10.1158/1078-0432.ccr-16-0049] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 02/25/2016] [Indexed: 12/20/2022]
Abstract
Immunotherapy strategies against cancer are emerging as powerful weapons for treatment of this disease. The success of checkpoint inhibitors against metastatic melanoma and adoptive T-cell therapy with chimeric antigen receptor T cells against B-cell-derived leukemias and lymphomas are only two examples of developments that are changing the paradigms of clinical cancer management. These changes are a result of many years of intense research into complex and interrelated cellular and molecular mechanisms controling immune responses. Promising advances come from the discovery of cancer mutation-encoded neoantigens, improvements in vaccine development, progress in delivery of cellular therapies, and impressive achievements in biotechnology. As a result, radical transformation of cancer treatment is taking place in which conventional cancer treatments are being integrated with immunotherapeutic agents. Many clinical trials are in progress testing potential synergistic effects of treatments combining immunotherapy with other therapies. Much remains to be learned about the selection, delivery, and off-target effects of immunotherapy used alone or in combination. The existence of numerous escape mechanisms from the host immune system that human tumors have evolved still is a barrier to success. Efforts to understand the rules of immune cell dysfunction and of cancer-associated local and systemic immune suppression are providing new insights and fuel the enthusiasm for new therapeutic strategies. In the future, it might be possible to tailor immune therapy for each cancer patient. The use of new immune biomarkers and the ability to assess responses to therapy by noninvasive monitoring promise to improve early cancer diagnosis and prognosis. Personalized immunotherapy based on individual genetic, molecular, and immune profiling is a potentially achievable future goal. The current excitement for immunotherapy is justified in view of many existing opportunities for harnessing the immune system to treat cancer.
Collapse
Affiliation(s)
- Theresa L Whiteside
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sandra Demaria
- Department of Radiation Oncology, University of Cornell, New York, New York
| | - Maria E Rodriguez-Ruiz
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain. Clinica Universidad de Navarra, Pamplona, Spain
| | - Hassane M Zarour
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ignacio Melero
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain. Clinica Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
45
|
Jayashankar L, Hafner R. Adjunct Strategies for Tuberculosis Vaccines: Modulating Key Immune Cell Regulatory Mechanisms to Potentiate Vaccination. Front Immunol 2016; 7:577. [PMID: 28018344 PMCID: PMC5159487 DOI: 10.3389/fimmu.2016.00577] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/23/2016] [Indexed: 12/22/2022] Open
Abstract
Tuberculosis (TB) remains a global health threat of alarming proportions, resulting in 1.5 million deaths worldwide. The only available licensed vaccine, Bacillus Calmette–Guérin, does not confer lifelong protection against active TB. To date, development of an effective vaccine against TB has proven to be elusive, and devising newer approaches for improved vaccination outcomes is an essential goal. Insights gained over the last several years have revealed multiple mechanisms of immune manipulation by Mycobacterium tuberculosis (Mtb) in infected macrophages and dendritic cells that support disease progression and block development of protective immunity. This review provides an assessment of the known immunoregulatory mechanisms altered by Mtb, and how new interventions may reverse these effects. Examples include blocking of inhibitory immune cell coreceptor checkpoints (e.g., programed death-1). Conversely, immune mechanisms that strengthen immune cell effector functions may be enhanced by interventions, including stimulatory immune cell coreceptors (e.g., OX40). Modification of the activity of key cell “immunometabolism” signaling pathway molecules, including mechanistic target of rapamycin, glycogen synthase kinase-3β, wnt/β-catenin, adenosine monophosophate-activated protein kinase, and sirtuins, related epigenetic changes, and preventing induction of immune regulatory cells (e.g., regulatory T cells, myeloid-derived suppressor cells) are powerful new approaches to improve vaccine responses. Interventions to favorably modulate these components have been studied primarily in oncology to induce efficient antitumor immune responses, often by potentiation of cancer vaccines. These agents include antibodies and a rapidly increasing number of small molecule drug classes that have contributed to the dramatic immune-based advances in treatment of cancer and other diseases. Because immune responses to malignancies and to Mtb share many similar mechanisms, studies to improve TB vaccine responses using interventions based on “immuno-oncology” are needed to guide possible repurposing. Understanding the regulation of immune cell functions appropriated by Mtb to promote the imbalance between protective and pathogenic immune responses may guide the development of innovative drug-based adjunct approaches to substantially enhance the clinical efficacy of TB vaccines.
Collapse
Affiliation(s)
- Lakshmi Jayashankar
- Columbus Technologies, Inc., Contractor to the National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD , USA
| | - Richard Hafner
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
46
|
Schröder M, Loos S, Naumann SK, Bachran C, Krötschel M, Umansky V, Helming L, Swee LK. Identification of inhibitors of myeloid-derived suppressor cells activity through phenotypic chemical screening. Oncoimmunology 2016; 6:e1258503. [PMID: 28197378 DOI: 10.1080/2162402x.2016.1258503] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/26/2016] [Accepted: 11/03/2016] [Indexed: 01/04/2023] Open
Abstract
Tumors are infiltrated by cells of the immune system that interact through complex regulatory networks. Although tumor-specific CD8+ T cells can be found in peripheral blood and tumor samples from cancer patients, their function is inhibited by immunosuppressive cells such as regulatory T cells, tumor-associated macrophages, and myeloid-derived suppressor cells (MDSC). Recent clinical successes have demonstrated that alleviating immunosuppression and T cell exhaustion translates into long-term clinical benefits. Although tremendous progress has been achieved, tools that afford unbiased approaches and screenings to uncover new potential inhibitors or gene targets are lacking. In this study, we describe a system based on immortalized progenitors that allows straightforward investigation of myeloid cells. We show that bone marrow progenitors immortalized through the transduction of NUP98-HOXB4 transgene can be differentiated into CD11b+Gr-1+ MDSC that express Arginase-1 and PD-L1, produce reactive oxygen and nitrogen species, and suppress T cell function in vitro. To uncover chemical probes that interfere with MDSC biology, we performed a chemical phenotypic screening and identified 3-deazaneplanocin A as a novel modulator of MDSC functions. We characterized and compared the effect of 3-deazaneplanocin-A and all-trans retinoic acid, a well-known modulator of MDSC activity, on the expression of effector molecules and immunosuppressive functions of MDSC. Altogether, this proof-of-principle opens new possibilities for the identification of drugs targeting myeloid cells with immunosuppressive activities.
Collapse
Affiliation(s)
| | - Simone Loos
- BioMed X Innovation Center , Heidelberg, Germany
| | | | | | | | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Heidelberg, Germany
| | | | - Lee Kim Swee
- BioMed X Innovation Center , Heidelberg, Germany
| |
Collapse
|
47
|
The Role of Myeloid-Derived Suppressor Cells (MDSC) in Cancer Progression. Vaccines (Basel) 2016; 4:vaccines4040036. [PMID: 27827871 PMCID: PMC5192356 DOI: 10.3390/vaccines4040036] [Citation(s) in RCA: 272] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/04/2016] [Accepted: 10/31/2016] [Indexed: 12/27/2022] Open
Abstract
The immunosuppressive tumor microenvironment represents not only one of the key factors stimulating tumor progression but also a strong obstacle for efficient tumor immunotherapy. Immunosuppression was found to be associated with chronic inflammatory mediators including cytokines, chemokines and growth factors produced by cancer and stroma cells. Long-term intensive production of these factors induces the formation of myeloid-derived suppressor cells (MDSCs) representing one of the most important players mediating immunosuppression. Moreover, MDSCs could not only inhibit anti-tumor immune reactions but also directly stimulate tumor growth and metastasis. Therefore, understanding the mechanisms of their generation, expansion, recruitment and activation is required for the development of novel strategies for tumor immunotherapy.
Collapse
|
48
|
Abstract
As a multitargeted kinase inhibitor, sunitinib has carved its way into demonstrating itself as a most effective tyrosine kinase inhibitor in the treatment of metastatic renal cell carcinoma. Mechanistically, sunitinib inhibits multiple receptor tyrosine kinases, especially those involved in angiogenesis, that is, vascular endothelial growth factor receptor, platelet-derived growth factor receptor, and proto-oncogene cKIT. Sunitinib has also been implicated in enhancing cancer invasiveness and metastasis. Mechanisms of resistance are poorly understood, but both intrinsic and acquired mechanisms are thought to be involved. While the side effects are manageable, sunitinib, like many other tyrosine kinase inhibitors, can be associated with serious toxicities that require careful management including frequent dose reductions. Although still in the early stage, emerging evidence points to an immunomodulatory role for sunitinib. It is also likely to contribute to the overall outcomes, especially those seen in metastatic renal cell carcinoma, and such effects are thought to be mediated by the proto-oncogene cKIT receptor. Combination with other modalities such as stereotactic body radiation therapy, therapeutic vaccines, and checkpoint inhibitors is being pursued for improved efficacy.
Collapse
Affiliation(s)
- Zhonglin Hao
- Department of Medicine, Section of Hematology and Oncology, Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ibrahim Sadek
- Department of Medicine, Section of Hematology and Oncology, Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
49
|
Gaither KA, Little AA, McBride AA, Garcia SR, Brar KK, Zhu Z, Platt A, Zhang F, Meadows GG, Zhang H. The immunomodulatory, antitumor and antimetastatic responses of melanoma-bearing normal and alcoholic mice to sunitinib and ALT-803: a combinatorial treatment approach. Cancer Immunol Immunother 2016; 65:1123-34. [PMID: 27481107 PMCID: PMC11029158 DOI: 10.1007/s00262-016-1876-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 07/21/2016] [Indexed: 01/13/2023]
Abstract
ALT-803, a novel IL-15/IL-15 receptor alpha complex, and the tyrosine kinase inhibitor, sunitinib, were examined for their single and combined effects on the growth of subcutaneous B16BL6 melanoma and on lymph node and lung metastasis. The study was conducted in immunocompetent C57BL/6 mice drinking water (Water mice) and in mice that chronically consumed alcohol (Alcohol mice), which are deficient in CD8(+) T cells. Sunitinib inhibited melanoma growth and was more effective in Alcohol mice. ALT-803 did not alter tumor growth or survival in Water or Alcohol mice. Combined ALT-803 and sunitinib inhibited melanoma growth and increased survival, and these effects were greater than sunitinib alone in Water mice. ALT-803 and alcohol independently suppressed lymph node and lung metastasis, whereas sunitinib alone or in combination with ALT-803 increased lymph node and lung metastasis in Water and Alcohol mice. Initially, ALT-803 increased IFN-γ-producing CD8(+)CD44(hi) memory T cells and CD8(+)CD44(hi)CD62L(lo) effector memory T cells and sunitinib decreased immunosuppressive MDSC and T regulatory cells (Treg). However, the impact of these treatments diminished with time. Subcutaneous tumors from Water mice showed increased numbers of CD8(+) T cells, CD8(+)CD44(hi) T cells, NK cells, and MDSC cells and decreased Treg cells after ALT-803 treatment.
Collapse
Affiliation(s)
- Kari A Gaither
- Department of Pharmaceutical Sciences and the Pharmaceutical Sciences Graduate Program, College of Pharmacy, Washington State University Spokane, PBS 323, P. O. Box 1495, Spokane, WA, 99210-1495, USA
| | - Alexander A Little
- Department of Pharmaceutical Sciences and the Pharmaceutical Sciences Graduate Program, College of Pharmacy, Washington State University Spokane, PBS 323, P. O. Box 1495, Spokane, WA, 99210-1495, USA
| | - Alisha A McBride
- Department of Pharmaceutical Sciences and the Pharmaceutical Sciences Graduate Program, College of Pharmacy, Washington State University Spokane, PBS 323, P. O. Box 1495, Spokane, WA, 99210-1495, USA
| | - Savanna R Garcia
- Department of Pharmaceutical Sciences and the Pharmaceutical Sciences Graduate Program, College of Pharmacy, Washington State University Spokane, PBS 323, P. O. Box 1495, Spokane, WA, 99210-1495, USA
| | - Kiranjot K Brar
- Department of Pharmaceutical Sciences and the Pharmaceutical Sciences Graduate Program, College of Pharmacy, Washington State University Spokane, PBS 323, P. O. Box 1495, Spokane, WA, 99210-1495, USA
| | - Zhaohui Zhu
- Department of Pharmaceutical Sciences and the Pharmaceutical Sciences Graduate Program, College of Pharmacy, Washington State University Spokane, PBS 323, P. O. Box 1495, Spokane, WA, 99210-1495, USA
| | - Amity Platt
- Department of Pharmaceutical Sciences and the Pharmaceutical Sciences Graduate Program, College of Pharmacy, Washington State University Spokane, PBS 323, P. O. Box 1495, Spokane, WA, 99210-1495, USA
| | - Faya Zhang
- Department of Pharmaceutical Sciences and the Pharmaceutical Sciences Graduate Program, College of Pharmacy, Washington State University Spokane, PBS 323, P. O. Box 1495, Spokane, WA, 99210-1495, USA
| | - Gary G Meadows
- Department of Pharmaceutical Sciences and the Pharmaceutical Sciences Graduate Program, College of Pharmacy, Washington State University Spokane, PBS 323, P. O. Box 1495, Spokane, WA, 99210-1495, USA.
| | - Hui Zhang
- Department of Pharmaceutical Sciences and the Pharmaceutical Sciences Graduate Program, College of Pharmacy, Washington State University Spokane, PBS 323, P. O. Box 1495, Spokane, WA, 99210-1495, USA.
| |
Collapse
|
50
|
Reinmuth N, Reck M. Immunotherapy for Lung Cancer. Oncol Res Treat 2016; 39:360-8. [PMID: 27259861 DOI: 10.1159/000446726] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/11/2016] [Indexed: 11/19/2022]
Abstract
Immune evasion is recognized as a key strategy for cancer survival and progression. Hence, various approaches to restore antitumor immune responses are currently being investigated. In particular, agents targeting immune checkpoints, such as the cytotoxic T-lymphocyte-associated antigen-4 receptor and programmed death-1 receptor, have shown potential for improving tumor responses and survival in lung cancer patients. With the first immunomodulating agents having been approved for treatment of selected lung cancer patients, there are high expectations that treatment outcomes may be improved with the incorporation of immunotherapies into the various treatment cascades.
Collapse
Affiliation(s)
- Niels Reinmuth
- Department of Thoracic Oncology, Fachklinik Gauting, Member of the German Center for Lung Research (DZL), Gauting, Germany
| | | |
Collapse
|