101
|
Nishikawa H, Koyama S. Mechanisms of regulatory T cell infiltration in tumors: implications for innovative immune precision therapies. J Immunother Cancer 2021; 9:jitc-2021-002591. [PMID: 34330764 PMCID: PMC8327843 DOI: 10.1136/jitc-2021-002591] [Citation(s) in RCA: 172] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2021] [Indexed: 11/04/2022] Open
Abstract
With the broad application of cancer immunotherapies such as immune checkpoint inhibitors in multiple cancer types, the immunological landscape in the tumor microenvironment (TME) has become enormously important for determining the optimal cancer treatment. Tumors can be immunologically divided into two categories: inflamed and non-inflamed based on the extent of immune cell infiltration and their activation status. In general, immunotherapies are preferable for the inflamed tumors than for non-inflamed tumors. Regulatory T cells (Tregs), an immunosuppressive subset of CD4+ T cells, play an essential role in maintaining self-tolerance and immunological homeostasis. In tumor immunity, Tregs compromise immune surveillance against cancer in healthy individuals and impair the antitumor immune response in tumor-bearing hosts. Tregs, therefore, accelerate immune evasion by tumor cells, leading to tumor development and progression in various types of cancer. Therefore, Tregs are considered to be a crucial therapeutic target for cancer immunotherapy. Abundant Tregs are observed in the TME in many types of cancer, both in inflamed and non-inflamed tumors. Diverse mechanisms of Treg accumulation, activation, and survival in the TME have been uncovered for different tumor types, indicating the importance of understanding the mechanism of Treg infiltration in each patient when selecting the optimal Treg-targeted therapy. Here, we review recent advances in the understanding of mechanisms leading to Treg abundance in the TME to optimize Treg-targeted therapy. Furthermore, in addition to the conventional strategies targeting cell surface molecules predominantly expressed by Tregs, reagents targeting molecules and signaling pathways specifically employed by Tregs for infiltration, activation, and survival in each tumor type are illustrated as novel Treg-targeted therapies. The effectiveness of immune precision therapy depends on conditions in the TME of each cancer patient.
Collapse
Affiliation(s)
- Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Tokyo/Chiba, Japan .,Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shohei Koyama
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Tokyo/Chiba, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
102
|
Ge Z, Peppelenbosch MP, Sprengers D, Kwekkeboom J. TIGIT, the Next Step Towards Successful Combination Immune Checkpoint Therapy in Cancer. Front Immunol 2021; 12:699895. [PMID: 34367161 PMCID: PMC8339559 DOI: 10.3389/fimmu.2021.699895] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
T cell immunoreceptor with Ig and ITIM domains (TIGIT) is an inhibitory receptor expressed on several types of lymphocytes. Efficacy of antibody blockade of TIGIT in cancer immunotherapy is currently widely being investigated in both pre-clinical and clinical studies. In multiple cancers TIGIT is expressed on tumor-infiltrating cytotoxic T cells, helper T cells, regulatory T cells and NK cells, and its main ligand CD155 is expressed on tumor-infiltrating myeloid cells and upregulated on cancer cells, which contributes to local suppression of immune-surveillance. While single TIGIT blockade has limited anti-tumor efficacy, pre-clinical studies indicate that co-blockade of TIGIT and PD-1/PD-L1 pathway leads to tumor rejection, notably even in anti-PD-1 resistant tumor models. Among inhibitory immune checkpoint molecules, a unique property of TIGIT blockade is that it enhances not only anti-tumor effector T-cell responses, but also NK-cell responses, and reduces the suppressive capacity of regulatory T cells. Numerous clinical trials on TIGIT-blockade in cancer have recently been initiated, predominantly combination treatments. The first interim results show promise for combined TIGIT and PD-L1 co-blockade in solid cancer patients. In this review, we summarize the current knowledge and identify the gaps in our current understanding of TIGIT’s roles in cancer immunity, and provide, based on these insights, recommendations for its positioning in cancer immunotherapy.
Collapse
Affiliation(s)
- Zhouhong Ge
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center (MC), Rotterdam, Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center (MC), Rotterdam, Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center (MC), Rotterdam, Netherlands
| | - Jaap Kwekkeboom
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center (MC), Rotterdam, Netherlands
| |
Collapse
|
103
|
Ikebuchi R, Moriya T, Ueda M, Yasuda I, Kusumoto Y, Chtanova T, Tomura M. Cutting Edge: Recruitment, Retention, and Migration Underpin Functional Phenotypic Heterogeneity of Regulatory T Cells in Tumors. THE JOURNAL OF IMMUNOLOGY 2021; 207:771-776. [PMID: 34290103 DOI: 10.4049/jimmunol.2001083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 05/24/2021] [Indexed: 12/23/2022]
Abstract
Tumor-infiltrating regulatory T cells (Tregs) have been extensively studied as therapeutic targets. However, not all infiltrating T cells exert their functions equally, presumably because of their heterogeneity and substantial turnover in tissues. In this study, we hypothesized that intertissue migration underlies the functional heterogeneity of Tregs. To test this, we applied in vivo photolabeling to examine single-cell diversity of immunosuppressive molecules in mouse Tregs migrating to, remaining in, and emigrating from MC38 tumors. Neuropilin-1 (Nrp1) expression was inversely correlated with that of six other molecules associated with Treg function. Unsupervised clustering analyses revealed that clusters containing Tregs that were retained in tumors expressed high levels of the six functional molecules but not of Nrp1. However, these clusters represented only half of the Tregs migrating to the tumor, suggesting evolving heterogeneity of tumor-infiltrating Tregs. Thus, we propose progressive pathways of Treg activation and migration between tumors and draining lymph nodes.
Collapse
Affiliation(s)
- Ryoyo Ikebuchi
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Tondabayashi, Japan; .,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Taiki Moriya
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Tondabayashi, Japan
| | - Mizuki Ueda
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Tondabayashi, Japan
| | - Ippei Yasuda
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Tondabayashi, Japan.,Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Yutaka Kusumoto
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Tondabayashi, Japan
| | - Tatyana Chtanova
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, Australia; and.,Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Michio Tomura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Tondabayashi, Japan;
| |
Collapse
|
104
|
Collagenase-Expressing Salmonella Targets Major Collagens in Pancreatic Cancer Leading to Reductions in Immunosuppressive Subsets and Tumor Growth. Cancers (Basel) 2021; 13:cancers13143565. [PMID: 34298778 PMCID: PMC8306875 DOI: 10.3390/cancers13143565] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 01/04/2023] Open
Abstract
Therapeutic resistance in pancreatic ductal adenocarcinoma (PDAC) can be attributed, in part, to a dense extracellular matrix containing excessive collagen deposition. Here, we describe a novel Salmonella typhimurium (ST) vector expressing the bacterial collagenase Streptomyces omiyaensis trypsin (SOT), a serine protease known to hydrolyze collagens I and IV, which are predominantly found in PDAC. Utilizing aggressive models of PDAC, we show that ST-SOT selectively degrades intratumoral collagen leading to decreases in immunosuppressive subsets, tumor proliferation and viability. Ultimately, we found that ST-SOT treatment significantly modifies the intratumoral immune landscape to generate a microenvironment that may be more conducive to immunotherapy.
Collapse
|
105
|
Du W, Pasca di Magliano M, Zhang Y. Therapeutic Potential of Targeting Stromal Crosstalk-Mediated Immune Suppression in Pancreatic Cancer. Front Oncol 2021; 11:682217. [PMID: 34290984 PMCID: PMC8287251 DOI: 10.3389/fonc.2021.682217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/21/2021] [Indexed: 12/19/2022] Open
Abstract
The stroma-rich, immunosuppressive microenvironment is a hallmark of pancreatic ductal adenocarcinoma (PDA). Tumor cells and other cellular components of the tumor microenvironment, such as cancer associated fibroblasts, CD4+ T cells and myeloid cells, are linked by a web of interactions. Their crosstalk not only results in immune evasion of PDA, but also contributes to pancreatic cancer cell plasticity, invasiveness, metastasis, chemo-resistance, immunotherapy-resistance and radiotherapy-resistance. In this review, we characterize several prevalent populations of stromal cells in the PDA microenvironment and describe how the crosstalk among them drives and maintains immune suppression. We also summarize therapeutic approaches to target the stroma. With a better understanding of the complex cellular and molecular networks in PDA, strategies aimed at sensitizing PDA to chemotherapy or immunotherapy through re-programing the tumor microenvironment can be designed, and in turn lead to improved clinical treatment for pancreatic cancer patients.
Collapse
Affiliation(s)
- Wenting Du
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
106
|
Gorchs L, Kaipe H. Interactions between Cancer-Associated Fibroblasts and T Cells in the Pancreatic Tumor Microenvironment and the Role of Chemokines. Cancers (Basel) 2021; 13:2995. [PMID: 34203869 PMCID: PMC8232575 DOI: 10.3390/cancers13122995] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 01/18/2023] Open
Abstract
Less than 10% of patients diagnosed with pancreatic ductal adenocarcinoma (PDAC) survive 5 years or more, making it one of the most fatal cancers. Accumulation of T cells in pancreatic tumors is associated with better prognosis, but immunotherapies to enhance the anti-tumor activity of infiltrating T cells are failing in this devastating disease. Pancreatic tumors are characterized by a desmoplastic stroma, which mainly consists of activated cancer-associated fibroblasts (CAFs). Pancreatic CAFs have emerged as important regulators of the tumor microenvironment by contributing to immune evasion through the release of chemokines, cytokines, and growth factors, which alters T-cell migration, differentiation and cytotoxic activity. However, recent discoveries have also revealed that subsets of CAFs with diverse functions can either restrain or promote tumor progression. Here, we discuss our current knowledge about the interactions between CAFs and T cells in PDAC and summarize different therapy strategies targeting the CAF-T cell axis with focus on CAF-derived soluble immunosuppressive factors and chemokines. Identifying the functions of different CAF subsets and understanding their roles in T-cell trafficking within the tumor may be fundamental for the development of an effective combinational treatment for PDAC.
Collapse
Affiliation(s)
- Laia Gorchs
- Department of Laboratory Medicine, Karolinska Institutet, 14152 Stockholm, Sweden
| | - Helen Kaipe
- Department of Laboratory Medicine, Karolinska Institutet, 14152 Stockholm, Sweden
- Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, 14152 Stockholm, Sweden
| |
Collapse
|
107
|
Abstract
INTRODUCTION Chemokines and their cognate receptors play a major role in modulating inflammatory responses. Depending on their ligand binding, chemokine receptors can stimulate both immune activating and inhibitory signaling pathways. The CC chemokine receptor 5 (CCR5) promotes immune responses by recruiting immune cells to the sites of inflammation/tumor, and is involved in stimulating tumor cell proliferation, invasion and migration through various mechanisms. Moreover, CCR5 also contributes to an immune-suppressive tumor microenvironment by recruiting regulatory T cells and myeloid-derived suppressor cells facilitating tumor development and progression. In summary, cells expressing CCR5 modulate immune response and tumor progression. Expression of CCR5 is increased in various malignancies and associated with poor outcome. Experimental data show promising efficacy signals with CCR5 antagonists in preclinical tumor models. Therefore, CCR5 has been recognized as a potential therapeutic target for cancer. AREAS COVERED In this review, we focus on the role of CCR5 in cancer progression and discuss its impact and potential as a therapeutic target for cancer. EXPERT OPINION Beyond immune-checkpoint inhibitors, potentially synergistic immune-modulatory drugs such as CCR5 antagonists are a promising approach to enlarge our treatment armamentarium against cancer.
Collapse
Affiliation(s)
- Hossein Hemmatazad
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Martin D Berger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
108
|
Scott EN, Gocher AM, Workman CJ, Vignali DAA. Regulatory T Cells: Barriers of Immune Infiltration Into the Tumor Microenvironment. Front Immunol 2021; 12:702726. [PMID: 34177968 PMCID: PMC8222776 DOI: 10.3389/fimmu.2021.702726] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
Regulatory T cells (Tregs) are key immunosuppressive cells that promote tumor growth by hindering the effector immune response. Tregs utilize multiple suppressive mechanisms to inhibit pro-inflammatory responses within the tumor microenvironment (TME) by inhibition of effector function and immune cell migration, secretion of inhibitory cytokines, metabolic disruption and promotion of metastasis. In turn, Tregs are being targeted in the clinic either alone or in combination with other immunotherapies, in efforts to overcome the immunosuppressive TME and increase anti-tumor effects. However, it is now appreciated that Tregs not only suppress cells intratumorally via direct engagement, but also serve as key interactors in the peritumor, stroma, vasculature and lymphatics to limit anti-tumor immune responses prior to tumor infiltration. We will review the suppressive mechanisms that Tregs utilize to alter immune and non-immune cells outside and within the TME and discuss how these mechanisms collectively allow Tregs to create and promote a physical and biological barrier, resulting in an immune-excluded or limited tumor microenvironment.
Collapse
Affiliation(s)
- Ellen N. Scott
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Angela M. Gocher
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
| | - Creg J. Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
| | - Dario A. A. Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| |
Collapse
|
109
|
Lim YW, Coles GL, Sandhu SK, Johnson DS, Adler AS, Stone EL. Single-cell transcriptomics reveals the effect of PD-L1/TGF-β blockade on the tumor microenvironment. BMC Biol 2021; 19:107. [PMID: 34030676 PMCID: PMC8147417 DOI: 10.1186/s12915-021-01034-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The anti-tumor activity of anti-PD-1/PD-L1 therapies correlates with T cell infiltration in tumors. Thus, a major goal in oncology is to find strategies that enhance T cell infiltration and efficacy of anti-PD-1/PD-L1 therapy. TGF-β has been shown to contribute to T cell exclusion, and anti-TGF-β improves anti-PD-L1 efficacy in vivo. However, TGF-β inhibition has frequently been shown to induce toxicity in the clinic, and the clinical efficacy of combination PD-L1 and TGF-β blockade has not yet been proven. To identify strategies to overcome resistance to PD-L1 blockade, the transcriptional programs associated with PD-L1 and/or TGF-β blockade in the tumor microenvironment should be further elucidated. RESULTS We used single-cell RNA sequencing in a mouse model to characterize the transcriptomic effects of PD-L1 and/or TGF-β blockade on nearly 30,000 single cells in the tumor and surrounding microenvironment. Combination treatment led to upregulation of immune response genes, including multiple chemokine genes such as CCL5, in macrophages, and downregulation of extracellular matrix genes in fibroblasts. Analysis of publicly available tumor transcriptome profiles showed that the chemokine CCL5 was strongly associated with immune cell infiltration in various human cancers. Further investigation with in vivo models showed that intratumorally administered CCL5 enhanced cytotoxic lymphocytes and the anti-tumor activity of anti-PD-L1. CONCLUSIONS Taken together, our data could be leveraged translationally to complement or find alternatives to anti-PD-L1 plus anti-TGF-β combination therapy, for example through companion biomarkers, and/or to identify novel targets that could be modulated to overcome resistance.
Collapse
Affiliation(s)
- Yoong Wearn Lim
- GigaGen, Inc., One Tower Place, Suite 750, South San Francisco, CA, 94080, USA
| | - Garry L Coles
- GigaGen, Inc., One Tower Place, Suite 750, South San Francisco, CA, 94080, USA
| | - Savreet K Sandhu
- GigaGen, Inc., One Tower Place, Suite 750, South San Francisco, CA, 94080, USA
| | - David S Johnson
- GigaGen, Inc., One Tower Place, Suite 750, South San Francisco, CA, 94080, USA
| | - Adam S Adler
- GigaGen, Inc., One Tower Place, Suite 750, South San Francisco, CA, 94080, USA.
| | - Erica L Stone
- GigaGen, Inc., One Tower Place, Suite 750, South San Francisco, CA, 94080, USA.
| |
Collapse
|
110
|
Ahmad IM, Dafferner AJ, O’Connell KA, Mehla K, Britigan BE, Hollingsworth MA, Abdalla MY. Heme Oxygenase-1 Inhibition Potentiates the Effects of Nab-Paclitaxel-Gemcitabine and Modulates the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2021; 13:2264. [PMID: 34066839 PMCID: PMC8125955 DOI: 10.3390/cancers13092264] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/22/2021] [Accepted: 05/03/2021] [Indexed: 12/27/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a poor prognosis. Tumor hypoxia plays an active role in promoting tumor progression, malignancy, and resistance to therapy in PDAC. We present evidence that nab-paclitaxel-gemcitabine (NPG) and/or a hypoxic tumor microenvironment (TME) up-regulate heme oxygenase-1 (HO-1), providing a survival advantage for tumors. Using PDAC cells in vitro and a PDAC mouse model, we found that NPG chemotherapy up-regulated expression of HO-1 in PDAC cells and increased its nuclear translocation. Inhibition of HO-1 with ZnPP and SnPP sensitized PDAC cells to NPG-induced cytotoxicity (p < 0.05) and increased apoptosis (p < 0.05). Additionally, HO-1 expression was increased in gemcitabine-resistant PDAC cells (p < 0.05), and HO-1 inhibition increased GEM-resistant PDAC sensitivity to NPG (p < 0.05). NPG combined with HO-1 inhibitor inhibited tumor size in an orthotopic model. In parallel, HO-1 inhibition abrogated the influx of macrophages and FoxP3+ cells, while increasing the proportion of CD8+ infiltration in the pancreatic tumors. These effects were mediated primarily by reducing expression of the immunosuppressive cytokine IL-10.
Collapse
Affiliation(s)
- Iman M. Ahmad
- College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Alicia J. Dafferner
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Kelly A. O’Connell
- Fred & Pamela Buffett Cancer Center, The Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.A.O.); (K.M.); (M.A.H.)
| | - Kamiya Mehla
- Fred & Pamela Buffett Cancer Center, The Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.A.O.); (K.M.); (M.A.H.)
| | - Bradley E. Britigan
- Veterans Affairs Medical Center-Nebraska Western Iowa, Department of Internal Medicine and Research Service, Omaha, NE 68105, USA;
| | - Michael A. Hollingsworth
- Fred & Pamela Buffett Cancer Center, The Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.A.O.); (K.M.); (M.A.H.)
| | - Maher Y. Abdalla
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| |
Collapse
|
111
|
Zalfa C, Paust S. Natural Killer Cell Interactions With Myeloid Derived Suppressor Cells in the Tumor Microenvironment and Implications for Cancer Immunotherapy. Front Immunol 2021; 12:633205. [PMID: 34025641 PMCID: PMC8133367 DOI: 10.3389/fimmu.2021.633205] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/12/2021] [Indexed: 12/17/2022] Open
Abstract
The tumor microenvironment (TME) is a complex and heterogeneous environment composed of cancer cells, tumor stroma, a mixture of tissue-resident and infiltrating immune cells, secreted factors, and extracellular matrix proteins. Natural killer (NK) cells play a vital role in fighting tumors, but chronic stimulation and immunosuppression in the TME lead to NK cell exhaustion and limited antitumor functions. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of myeloid cells with potent immunosuppressive activity that gradually accumulate in tumor tissues. MDSCs interact with innate and adaptive immune cells and play a crucial role in negatively regulating the immune response to tumors. This review discusses MDSC-mediated NK cell regulation within the TME, focusing on critical cellular and molecular interactions. We review current strategies that target MDSC-mediated immunosuppression to enhance NK cell cytotoxic antitumor activity. We also speculate on how NK cell-based antitumor immunotherapy could be improved.
Collapse
Affiliation(s)
| | - Silke Paust
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
112
|
Nussbaum YI, Manjunath Y, Suvilesh KN, Warren WC, Shyu CR, Kaifi JT, Ciorba MA, Mitchem JB. Current and Prospective Methods for Assessing Anti-Tumor Immunity in Colorectal Cancer. Int J Mol Sci 2021; 22:4802. [PMID: 33946558 PMCID: PMC8125332 DOI: 10.3390/ijms22094802] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) remains one of the deadliest malignancies worldwide despite recent progress in treatment strategies. Though immune checkpoint inhibition has proven effective for a number of other tumors, it offers benefits in only a small group of CRC patients with high microsatellite instability. In general, heterogenous cell groups in the tumor microenvironment are considered as the major barrier for unveiling the causes of low immune response. Therefore, deconvolution of cellular components in highly heterogeneous microenvironments is crucial for understanding the immune contexture of cancer. In this review, we assimilate current knowledge and recent studies examining anti-tumor immunity in CRC. We also discuss the utilization of novel immune contexture assessment methods that have not been used in CRC research to date.
Collapse
Affiliation(s)
- Yulia I. Nussbaum
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65201, USA; (Y.I.N.); (C.-R.S.); (J.T.K.)
| | - Yariswamy Manjunath
- Department of Surgery, Columbia, MO 65212, USA; (Y.M.); (K.N.S.); (W.C.W.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Kanve N. Suvilesh
- Department of Surgery, Columbia, MO 65212, USA; (Y.M.); (K.N.S.); (W.C.W.)
| | - Wesley C. Warren
- Department of Surgery, Columbia, MO 65212, USA; (Y.M.); (K.N.S.); (W.C.W.)
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Chi-Ren Shyu
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65201, USA; (Y.I.N.); (C.-R.S.); (J.T.K.)
| | - Jussuf T. Kaifi
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65201, USA; (Y.I.N.); (C.-R.S.); (J.T.K.)
- Department of Surgery, Columbia, MO 65212, USA; (Y.M.); (K.N.S.); (W.C.W.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Matthew A. Ciorba
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA;
- Division of Gastroenterology, Department of Medicine, Washington School of Medicine, St. Louis, MO 63110, USA
| | - Jonathan B. Mitchem
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65201, USA; (Y.I.N.); (C.-R.S.); (J.T.K.)
- Department of Surgery, Columbia, MO 65212, USA; (Y.M.); (K.N.S.); (W.C.W.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA;
| |
Collapse
|
113
|
Huang L, Guo Y, Liu S, Wang H, Zhu J, Ou L, Xu X. Targeting regulatory T cells for immunotherapy in melanoma. MOLECULAR BIOMEDICINE 2021; 2:11. [PMID: 34806028 PMCID: PMC8591697 DOI: 10.1186/s43556-021-00038-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 04/01/2021] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells (Tregs) are essential in the maintenance of immunity, and they are also a key to immune suppressive microenvironment in solid tumors. Many studies have revealed the biology of Tregs in various human pathologies. Here we review recent understandings of the immunophenotypes and suppressive functions of Tregs in melanoma, including Treg recruitment and expansion in a tumor. Tregs are frequently accumulated in melanoma and the ratio of CD8+ T cells versus Tregs in the melanoma is predictive for patient survival. Hence, depletion of Tregs is a promising strategy for the enhancement of anti-melanoma immunity. Many recent studies are aimed to target Tregs in melanoma. Distinguishing Tregs from other immune cells and understanding the function of different subsets of Tregs may contribute to better therapeutic efficacy. Depletion of functional Tregs from the tumor microenvironment has been tested to induce clinically relevant immune responses against melanomas. However, the lack of Treg specific therapeutic antibodies or Treg specific depleting strategies is a big hurdle that is yet to be overcome. Additional studies to fine-tune currently available therapies and more agents that specifically and selectively target tumor infiltrating Tregs in melanoma are urgently needed.
Collapse
Affiliation(s)
- Lili Huang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Yeye Guo
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Shujing Liu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Huaishan Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Jinjin Zhu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Lingling Ou
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
114
|
Cendrowicz E, Sas Z, Bremer E, Rygiel TP. The Role of Macrophages in Cancer Development and Therapy. Cancers (Basel) 2021; 13:1946. [PMID: 33919517 PMCID: PMC8073377 DOI: 10.3390/cancers13081946] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023] Open
Abstract
Macrophages are critical mediators of tissue homeostasis and influence various aspects of immunity. Tumor-associated macrophages are one of the main cellular components of the tumor microenvironment. Depending on their activation status, macrophages can exert a dual influence on tumorigenesis by either antagonizing the cytotoxic activity of immune cells or, less frequently, by enhancing antitumor responses. In most situations, TAMs suppress T cell recruitment and function or regulate other aspects of tumor immunity. The importance of TAMs targeting in cancer therapy is derived from the strong association between the high infiltration of TAMs in the tumor tissue with poor patient prognosis. Several macrophage-targeting approaches in anticancer therapy are developed, including TAM depletion, inhibition of new TAM differentiation, or re-education of TAM activation for cancer cell phagocytosis. In this review, we will describe the role of TAMs in tumor development, including such aspects as protumorigenic inflammation, immune suppression, neoangiogenesis, and enhancement of tissue invasion and distant metastasis. Furthermore, we will discuss therapeutic approaches that aim to deplete TAMs or, on the contrary, re-educate TAMs for cancer cell phagocytosis and antitumor immunity.
Collapse
Affiliation(s)
- Ewa Cendrowicz
- Department of Hematology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (E.C.); (E.B.)
| | - Zuzanna Sas
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 Street, Building F, 02-097 Warsaw, Poland;
| | - Edwin Bremer
- Department of Hematology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (E.C.); (E.B.)
| | - Tomasz P. Rygiel
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 Street, Building F, 02-097 Warsaw, Poland;
| |
Collapse
|
115
|
Recruitment and Expansion of Tregs Cells in the Tumor Environment-How to Target Them? Cancers (Basel) 2021; 13:cancers13081850. [PMID: 33924428 PMCID: PMC8069615 DOI: 10.3390/cancers13081850] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/04/2021] [Accepted: 04/08/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary The immune response against cancer is generated by effector T cells, among them cytotoxic CD8+ T cells that destroy cancer cells and helper CD4+ T cells that mediate and support the immune response. This antitumor function of T cells is tightly regulated by a particular subset of CD4+ T cells, named regulatory T cells (Tregs), through different mechanisms. Even if the complete inhibition of Tregs would be extremely harmful due to their tolerogenic role in impeding autoimmune diseases in the periphery, the targeted blockade of their accumulation at tumor sites or their targeted depletion represent a major therapeutic challenge. This review focuses on the mechanisms favoring Treg recruitment, expansion and stabilization in the tumor microenvironment and the therapeutic strategies developed to block these mechanisms. Abstract Regulatory T cells (Tregs) are present in a large majority of solid tumors and are mainly associated with a poor prognosis, as their major function is to inhibit the antitumor immune response contributing to immunosuppression. In this review, we will investigate the mechanisms involved in the recruitment, amplification and stability of Tregs in the tumor microenvironment (TME). We will also review the strategies currently developed to inhibit Tregs’ deleterious impact in the TME by either inhibiting their recruitment, blocking their expansion, favoring their plastic transformation into other CD4+ T-cell subsets, blocking their suppressive function or depleting them specifically in the TME to avoid severe deleterious effects associated with Treg neutralization/depletion in the periphery and normal tissues.
Collapse
|
116
|
Sobhani N, Tardiel-Cyril DR, Davtyan A, Generali D, Roudi R, Li Y. CTLA-4 in Regulatory T Cells for Cancer Immunotherapy. Cancers (Basel) 2021; 13:1440. [PMID: 33809974 PMCID: PMC8005092 DOI: 10.3390/cancers13061440] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have obtained durable responses in many cancers, making it possible to foresee their potential in improving the health of cancer patients. However, immunotherapies are currently limited to a minority of patients and there is a need to develop a better understanding of the basic molecular mechanisms and functions of pivotal immune regulatory molecules. Immune checkpoint cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and regulatory T (Treg) cells play pivotal roles in hindering the anticancer immunity. Treg cells suppress antigen-presenting cells (APCs) by depleting immune stimulating cytokines, producing immunosuppressive cytokines and constitutively expressing CTLA-4. CTLA-4 molecules bind to CD80 and CD86 with a higher affinity than CD28 and act as competitive inhibitors of CD28 in APCs. The purpose of this review is to summarize state-of-the-art understanding of the molecular mechanisms underlining CTLA-4 immune regulation and the correlation of the ICI response with CTLA-4 expression in Treg cells from preclinical and clinical studies for possibly improving CTLA-4-based immunotherapies, while highlighting the knowledge gap.
Collapse
Affiliation(s)
- Navid Sobhani
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Dana Rae Tardiel-Cyril
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Aram Davtyan
- Atomwise, 717 Market St, San Francisco, CA 94103, USA;
| | - Daniele Generali
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34147 Trieste, Italy;
| | - Raheleh Roudi
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Yong Li
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA;
| |
Collapse
|
117
|
Conversion of AML-blasts to leukemia-derived dendritic cells (DCleu) in 'DC-culture-media' shifts correlations of released chemokines with antileukemic T-cell reactions. Immunobiology 2021; 226:152088. [PMID: 33838552 DOI: 10.1016/j.imbio.2021.152088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 02/21/2021] [Accepted: 03/10/2021] [Indexed: 11/22/2022]
Abstract
Dendritic cells (DC) and T-cells are mediators of CTL-responses. Autologous (from patients with acute myeloid leukaemia (AML) or myelodysplasia (MDS)) or allogeneic (donor)-T-cells stimulated by DCleu, gain an efficient lysis of naive blasts, although not in every case. CXCL8, -9, -10, CCL2, -5 and Interleukin (IL-12) were quantified by Cytometric Bead Array (CBA) in supernatants from 5 DC-generating methods and correlated with AML-/MDS-patients' serum-values, DC-/T-cell-interactions/antileukemic T-cell-reactions after mixed lymphocyte culture (MLC) and patients' clinical course. The blast-lytic activity of T-cells stimulated with DC or mononuclear cells (MNC) was quantified in a cytotoxicity assay. Despite great variations of chemokine-levels, correlations with post-stimulation (after stimulating T-cells with DC in MLC) improved antileukemic T-cell activity were seen: higher released chemokine-values correlated with improved T-cells' antileukemic activity (compared to stimulation with blast-containing MNC) - whereas with respect to the corresponding serum values higher CXCL8-, -9-, and -10- but lower CCL5- and -2-release correlated with improved antileukemic activity of DC-stimulated (vs. blast-stimulated) T-cells. In DC-culture supernatants higher chemokine-values correlated with post-stimulation improved antileukemic T-cell reactivity, whereas higher serum-values of CXCL8, -9, and -10 but lower serum-values of CCL5 and -2 correlated with post-stimulation improved antileukemic T-cell-reactivity. In a context of 'DC'-stimulation (vs serum) this might point to a change of (CCL5 and -2-associated) functionality from a more 'inflammatory' or 'tumor-promoting' to a more 'antitumor'-reactive functionality. This knowledge could contribute to develop immune-modifying strategies that promote antileukemic (adaptive) immune-responses.
Collapse
|
118
|
Pere H, Tanchot C, Bayry J, Terme M, Taieb J, Badoual C, Adotevi O, Merillon N, Marcheteau E, Quillien VR, Banissi C, Carpentier A, Sandoval F, Nizard M, Quintin-Colonna F, Kroemer G, Fridman WH, Zitvogel L, Oudard SP, Tartour E. Comprehensive analysis of current approaches to inhibit regulatory T cells in cancer. Oncoimmunology 2021; 1:326-333. [PMID: 22737608 PMCID: PMC3382865 DOI: 10.4161/onci.18852] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CD4+CD25+Foxp3+ regulatory T cells (Treg) have emerged as a dominant T cell population inhibiting anti-tumor effector T cells. Initial strategies used for Treg-depletion (cyclophosphamide, anti-CD25 mAb…) also targeted activated T cells, as they share many phenotypic markers. Current, ameliorated approaches to inhibit Treg aim to either block their function or their migration to lymph nodes and the tumor microenvironment. Various drugs originally developed for other therapeutic indications (anti-angiogenic molecules, tyrosine kinase inhibitors,etc) have recently been discovered to inhibit Treg. These approaches are expected to be rapidly translated to clinical applications for therapeutic use in combination with immunomodulators.
Collapse
Affiliation(s)
- Helene Pere
- INSERM U970 PARCC (Paris Cardiovascular Research Center); Université Paris Descartes; Sorbonne Paris Cité; Paris, France ; Hôpital Européen Georges Pompidou; Service de Microbiologie; Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
119
|
Willenbrock F, Cox CM, Parkes EE, Wilhelm-Benartzi CS, Abraham AG, Owens R, Sabbagh A, Jones CM, Hughes DLI, Maughan T, Hurt CN, O'Neill EE, Mukherjee S. Circulating biomarkers and outcomes from a randomised phase 2 trial of gemcitabine versus capecitabine-based chemoradiotherapy for pancreatic cancer. Br J Cancer 2021; 124:581-586. [PMID: 33100327 PMCID: PMC7851394 DOI: 10.1038/s41416-020-01120-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The Phase 2 SCALOP trial compared gemcitabine with capecitabine-based consolidation chemoradiotherapy (CRT) in locally advanced pancreatic cancer (LAPC). METHODS Thirty-five systematically identified circulating biomarkers were analysed in plasma samples from 60 patients enroled in SCALOP. Each was measured in triplicate at baseline (prior to three cycles of gemcitabine-capecitabine induction chemotherapy) and, for a subset, prior to CRT. Association with overall survival (OS) was determined using univariable Cox regression and optimal thresholds delineating low to high values identified using time-dependent ROC curves. Independence from known prognostic factors was assessed using Spearman correlation and the Wilcoxon rank sum test prior to multivariable Cox regression modelling including independent biomarkers and known prognostic factors. RESULTS Baseline circulating levels of C-C motif chemokine ligand 5 (CCL5) were significantly associated with OS, independent of other clinicopathological characteristics. Patients with low circulating CCL5 (CCL5low) had a median OS of 18.5 (95% CI 11.76-21.32) months compared to 11.3 (95% CI 9.86-15.51) months in CCL5high; hazard ratio 1.95 (95% CI 1.04-8.65; p = 0.037). CONCLUSIONS CCL5 is an independent prognostic biomarker in LAPC. Given the known role of CCL5 in tumour invasion, metastasis and the induction of an immunosuppressive micro-environment, targeting of CCL5-mediated pathways may offer therapeutic potential in pancreatic cancer. CLINICAL TRIAL REGISTRATION The SCALOP trial was registered with ISRCTN, number 96169987 (registered 29 May 2008).
Collapse
Affiliation(s)
| | | | | | | | | | - Robert Owens
- Oxford University Hospital NHS Trust, Oxford, UK
| | - Ahmad Sabbagh
- Weston Park Cancer Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | | | | | - Tim Maughan
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Eric E O'Neill
- Department of Oncology, University of Oxford, Oxford, UK
| | | |
Collapse
|
120
|
TLR expression profiles are a function of disease status in rheumatoid arthritis and experimental arthritis. J Autoimmun 2021; 118:102597. [PMID: 33493980 DOI: 10.1016/j.jaut.2021.102597] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/21/2020] [Accepted: 01/07/2021] [Indexed: 11/23/2022]
Abstract
The role of the innate immune system has been established in the initiation and perpetuation of inflammatory disease, but less attention has been paid to its role in the resolution of inflammation and return to homeostasis. Toll-like receptor (TLR) expression profiles were analysed in tissues with differing disease status in rheumatoid arthritis (RA), ankylosing spondylitis (AS), and in experimental arthritis. TLR gene expression was measured in whole blood and monocytes, before and after TNF blockade. In RA and osteoarthritis synovia, the expression of TLRs was quantified by standard curve qPCR. In addition, four distinct stages of disease were defined and validated in collagen-induced arthritis (CIA), the gold standard animal model for RA - pre-onset, early disease, late disease and immunised mice that were resistant to the development of disease. TLR expression was measured in spleens, lymph nodes, blood cells, liver and the paws (inflamed and unaffected). In RA whole blood, the expression of TLR1, 4 and 6 was significantly reduced by TNF blockade but the differences in TLR expression profiles between responders and non-responders were less pronounced than the differences between RA and AS patients. In RA non-responders, monocytes had greater TLR2 expression prior to therapy compared to responders. The expression of TLR1, 2, 4 and 8 was higher in RA synovium compared to control OA synovium. Circulating cytokine levels in CIA resistant mice were similar to naïve mice, but anti-collagen antibodies were similar to arthritic mice. Distinct profiles of inflammatory gene expression were mapped in paws and organs with differing disease status. TLR expression in arthritic paws tended to be similar in early and late disease, with TLR1 and 2 moderately higher in late disease. TLR expression in unaffected paws varied according to gene and disease status but was generally lower in resistant paws. Disease status-specific profiles of TLR expression were observed in spleens, lymph nodes, blood cells and the liver. Notably, TLR2 expression rose then fell in the transition from naïve to pre-onset to early arthritis. TLR gene expression profiles are strongly associated with disease status. In particular, increased expression in the blood precedes clinical manifestation.
Collapse
|
121
|
Dadey RE, Workman CJ, Vignali DAA. Regulatory T Cells in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1273:105-134. [PMID: 33119878 DOI: 10.1007/978-3-030-49270-0_6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Regulatory T cells (Tregs) are an immunosuppressive subpopulation of CD4+ T cells that are endowed with potent suppressive activity and function to limit immune activation and maintain homeostasis. These cells are identified by the hallmark transcription factor FOXP3 and the high-affinity interleukin-2 (IL-2) receptor chain CD25. Tregs can be recruited to and persist within the tumor microenvironment (TME), acting as a potent barrier to effective antitumor immunity. This chapter will discuss [i] the history and hallmarks of Tregs; [ii] the recruitment, development, and persistence of Tregs within the TME; [iii] Treg function within TME; asnd [iv] the therapeutic targeting of Tregs in the clinic. This chapter will conclude with a discussion of likely trends and future directions.
Collapse
Affiliation(s)
- Rebekah E Dadey
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, USA.,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, USA.,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, USA. .,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
122
|
Wang N, Wang S, Wang X, Zheng Y, Yang B, Zhang J, Pan B, Gao J, Wang Z. Research trends in pharmacological modulation of tumor-associated macrophages. Clin Transl Med 2021; 11:e288. [PMID: 33463063 PMCID: PMC7805405 DOI: 10.1002/ctm2.288] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
As one of the most abundant immune cell populations in the tumor microenvironment (TME), tumor-associated macrophages (TAMs) play important roles in multiple solid malignancies, including breast cancer, prostate cancer, liver cancer, lung cancer, ovarian cancer, gastric cancer, pancreatic cancer, and colorectal cancer. TAMs could contribute to carcinogenesis, neoangiogenesis, immune-suppressive TME remodeling, cancer chemoresistance, recurrence, and metastasis. Therefore, reprogramming of the immune-suppressive TAMs by pharmacological approaches has attracted considerable research attention in recent years. In this review, the promising pharmaceutical targets, as well as the existing modulatory strategies of TAMs were summarized. The chemokine-chemokine receptor signaling, tyrosine kinase receptor signaling, metabolic signaling, and exosomal signaling have been highlighted in determining the biological functions of TAMs. Besides, both preclinical research and clinical trials have suggested the chemokine-chemokine receptor blockers, tyrosine kinase inhibitors, bisphosphonates, as well as the exosomal or nanoparticle-based targeting delivery systems as the promising pharmacological approaches for TAMs deletion or reprogramming. Lastly, the combined therapies of TAMs-targeting strategies with traditional treatments or immunotherapies as well as the exosome-like nanovesicles for cancer therapy are prospected.
Collapse
Affiliation(s)
- Neng Wang
- The Research Center for Integrative MedicineSchool of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Shengqi Wang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Xuan Wang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Yifeng Zheng
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Bowen Yang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Juping Zhang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Bo Pan
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Jianli Gao
- Academy of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Zhiyu Wang
- The Research Center for Integrative MedicineSchool of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| |
Collapse
|
123
|
Sarkar T, Dhar S, Sa G. Tumor-infiltrating T-regulatory cells adapt to altered metabolism to promote tumor-immune escape. CURRENT RESEARCH IN IMMUNOLOGY 2021; 2:132-141. [PMID: 35492399 PMCID: PMC9040151 DOI: 10.1016/j.crimmu.2021.08.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/16/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor mass and its microenvironment alter host immune system in various ways to promote tumor growth. One of the modifications is evasion of immune surveillance by augmenting the number of Tregs in tumor vicinity. Elevated levels of Tregs are seen in peripheral circulation and tumor tissue of cancer patients. Cancer cells release several chemokines to attract Tregs in tumor-site. Infiltration of Tregs has clinical significance because being immunosuppressive infiltrating Tregs suppress other immune cells making the tumor microenvironment favorable for tumor growth. On the other hand, infiltrating Tregs show metabolic alteration in tumor microenvironment which allows their selective survival over the others. Persistence of Tregs in the tumor microenvironment and subsequent immunosuppression makes Tregs a potential therapeutic obstacle and the reason behind the failure of immunotherapy. In this review, we emphasize the recent development in the metabolic adaptation of tumor-infiltrating Tregs and the therapeutic approaches to boost immunity against cancer.
Collapse
|
124
|
Kolb HR, Borcherding N, Zhang W. Understanding and Targeting Human Cancer Regulatory T Cells to Improve Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1278:229-256. [PMID: 33523451 DOI: 10.1007/978-981-15-6407-9_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Regulatory T cells (Tregs) are critical in maintaining immune homeostasis under various pathophysiological conditions. A growing body of evidence demonstrates that Tregs play an important role in cancer progression and that they do so by suppressing cancer-directed immune responses. Tregs have been targeted for destruction by exploiting antibodies against and small-molecule inhibitors of several molecules that are highly expressed in Tregs-including immune checkpoint molecules, chemokine receptors, and metabolites. To date, these strategies have had only limited antitumor efficacy, yet they have also created significant risk of autoimmunity because most of them do not differentiate Tregs in tumors from those in normal tissues. Currently, immune checkpoint inhibitor (ICI)-based cancer immunotherapies have revolutionized cancer treatment, but the resistance to ICI is common and the elevation of Tregs is one of the most important mechanisms. Therapeutic strategies that can selectively eliminate Tregs in the tumor (i.e. therapies that do not run the risk of causing autoimmunity by affecting normal tissue), are urgently needed for the development of cancer immunotherapies. This chapter discusses specific properties of human Tregs under the context of cancer and the various ways to target Treg for cancer immunotherapy.
Collapse
Affiliation(s)
- H Ryan Kolb
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University, St. Louis, MO, USA
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
125
|
González-Navajas JM, Fan DD, Yang S, Yang FM, Lozano-Ruiz B, Shen L, Lee J. The Impact of Tregs on the Anticancer Immunity and the Efficacy of Immune Checkpoint Inhibitor Therapies. Front Immunol 2021; 12:625783. [PMID: 33717139 PMCID: PMC7952426 DOI: 10.3389/fimmu.2021.625783] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/02/2021] [Indexed: 12/21/2022] Open
Abstract
Although cancers arise from genetic mutations enabling cells to proliferate uncontrollably, they cannot thrive without failure of the anticancer immunity due in a large part to the tumor environment's influence on effector and regulatory T cells. The field of immune checkpoint inhibitor (ICI) therapy for cancer was born out of the fact that tumor environments paralyze the immune cells that are supposed to clear them by activating the immune checkpoint molecules such as PD-1. While various subsets of effector T cells work collaboratively to eliminate cancers, Tregs enriched in the tumor environment can suppress not only the native anticancer immunity but also diminish the efficacy of ICI therapies. Because of their essential role in suppressing autoimmunity, various attempts to specifically deplete tumor-associated Tregs are currently underway to boost the efficacy of ICI therapies without causing systemic autoimmune responses. A better understanding the roles of Tregs in the anti-cancer immunity and ICI therapies should provide more specific targets to deplete intratumoral Tregs. Here, we review the current understanding on how Tregs inhibit the anti-cancer immunity and ICI therapies as well as the advances in the targeted depletion of intratumoral Tregs.
Collapse
Affiliation(s)
- Jose M. González-Navajas
- Alicante Institute for Health and Biomedical Research (ISABIAL), Hospital General Universitario de Alicante, Alicante, Spain
- Networked Biomedical Research Center for Hepatic and Digestive Diseases (CIBERehd), Institute of Health Carlos III, Madrid, Spain
- Department of Pharmacology, Pediatrics and Organic Chemistry, University Miguel Hernández, Elche, Spain
- Institute of Research, Development and Innovation in Healthcare Biotechnology in Elche (IDiBE), University Miguel Hernández, Elche, Spain
- Jose M. González-Navajas
| | - Dengxia Denise Fan
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Shuang Yang
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Fengyuan Mandy Yang
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Beatriz Lozano-Ruiz
- Alicante Institute for Health and Biomedical Research (ISABIAL), Hospital General Universitario de Alicante, Alicante, Spain
- Networked Biomedical Research Center for Hepatic and Digestive Diseases (CIBERehd), Institute of Health Carlos III, Madrid, Spain
| | - Liya Shen
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jongdae Lee
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Jongdae Lee ;
| |
Collapse
|
126
|
Hyper-Progressive Disease: The Potential Role and Consequences of T-Regulatory Cells Foiling Anti-PD-1 Cancer Immunotherapy. Cancers (Basel) 2020. [PMID: 33375291 DOI: 10.3390/cancers13010048.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Antibody-mediated disruption of the programmed cell death protein 1 (PD-1) pathway has brought much success to the fight against cancer. Nevertheless, a significant proportion of patients respond poorly to anti-PD-1 treatment. Cases of accelerated and more aggressive forms of cancer following therapy have also been reported. Termed hyper-progressive disease (HPD), this phenomenon often results in fatality, thus requires urgent attention. Among possible causes of HPD, regulatory T-cells (Tregs) are of suspect due to their high expression of PD-1, which modulates Treg activity. Tregs are a subset of CD4+ T-cells that play a non-redundant role in the prevention of autoimmunity and is functionally dependent on the X chromosome-linked transcription factor FoxP3. In cancer, CD4+FoxP3+ Tregs migrate to tumors to suppress anti-tumor immune responses, allowing cancer cells to persist. Hence, Treg accumulation in tumors is associated with poor prognosis. In mice, the anti-tumor efficacy of anti-PD-1 can be enhanced by depleting Tregs. This suggests Tregs pose resistance to anti-PD-1 therapy. In this article, we review the relevant Treg functions that suppress tumor immunity and the potential effects anti-PD-1 could have on Tregs which are counter-productive to the treatment of cancer, occasionally causing HPD.
Collapse
|
127
|
Tay C, Qian Y, Sakaguchi S. Hyper-Progressive Disease: The Potential Role and Consequences of T-Regulatory Cells Foiling Anti-PD-1 Cancer Immunotherapy. Cancers (Basel) 2020; 13:E48. [PMID: 33375291 PMCID: PMC7796137 DOI: 10.3390/cancers13010048] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
Antibody-mediated disruption of the programmed cell death protein 1 (PD-1) pathway has brought much success to the fight against cancer. Nevertheless, a significant proportion of patients respond poorly to anti-PD-1 treatment. Cases of accelerated and more aggressive forms of cancer following therapy have also been reported. Termed hyper-progressive disease (HPD), this phenomenon often results in fatality, thus requires urgent attention. Among possible causes of HPD, regulatory T-cells (Tregs) are of suspect due to their high expression of PD-1, which modulates Treg activity. Tregs are a subset of CD4+ T-cells that play a non-redundant role in the prevention of autoimmunity and is functionally dependent on the X chromosome-linked transcription factor FoxP3. In cancer, CD4+FoxP3+ Tregs migrate to tumors to suppress anti-tumor immune responses, allowing cancer cells to persist. Hence, Treg accumulation in tumors is associated with poor prognosis. In mice, the anti-tumor efficacy of anti-PD-1 can be enhanced by depleting Tregs. This suggests Tregs pose resistance to anti-PD-1 therapy. In this article, we review the relevant Treg functions that suppress tumor immunity and the potential effects anti-PD-1 could have on Tregs which are counter-productive to the treatment of cancer, occasionally causing HPD.
Collapse
Affiliation(s)
- Christopher Tay
- Immunology Frontier Research Center, Department of Experimental Immunology, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan; (C.T.); (Y.Q.)
| | - Yamin Qian
- Immunology Frontier Research Center, Department of Experimental Immunology, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan; (C.T.); (Y.Q.)
| | - Shimon Sakaguchi
- Immunology Frontier Research Center, Department of Experimental Immunology, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan; (C.T.); (Y.Q.)
- Laboratory of Experimental Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
128
|
Zhang Y, Guan XY, Jiang P. Cytokine and Chemokine Signals of T-Cell Exclusion in Tumors. Front Immunol 2020; 11:594609. [PMID: 33381115 PMCID: PMC7768018 DOI: 10.3389/fimmu.2020.594609] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/30/2020] [Indexed: 12/14/2022] Open
Abstract
The success of cancer immunotherapy in solid tumors depends on a sufficient distribution of effector T cells into malignant lesions. However, immune-cold tumors utilize many T-cell exclusion mechanisms to resist immunotherapy. T cells have to go through three steps to fight against tumors: trafficking to the tumor core, surviving and expanding, and maintaining the memory phenotype for long-lasting responses. Cytokines and chemokines play critical roles in modulating the recruitment of T cells and the overall cellular compositions of the tumor microenvironment. Manipulating the cytokine or chemokine environment has brought success in preclinical models and early-stage clinical trials. However, depending on the immune context, the same cytokine or chemokine signals may exhibit either antitumor or protumor activities and induce unwanted side effects. Therefore, a comprehensive understanding of the cytokine and chemokine signals is the premise of overcoming T-cell exclusion for effective and innovative anti-cancer therapies.
Collapse
Affiliation(s)
- Yu Zhang
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- Department of Clinical Oncology, University of Hong Kong, Hong Kong, Hong Kong
| | - Xin-yuan Guan
- Department of Clinical Oncology, University of Hong Kong, Hong Kong, Hong Kong
| | - Peng Jiang
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
129
|
Korbecki J, Grochans S, Gutowska I, Barczak K, Baranowska-Bosiacka I. CC Chemokines in a Tumor: A Review of Pro-Cancer and Anti-Cancer Properties of Receptors CCR5, CCR6, CCR7, CCR8, CCR9, and CCR10 Ligands. Int J Mol Sci 2020; 21:ijms21207619. [PMID: 33076281 PMCID: PMC7590012 DOI: 10.3390/ijms21207619] [Citation(s) in RCA: 238] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/05/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
CC chemokines (or β-chemokines) are 28 chemotactic cytokines with an N-terminal CC domain that play an important role in immune system cells, such as CD4+ and CD8+ lymphocytes, dendritic cells, eosinophils, macrophages, monocytes, and NK cells, as well in neoplasia. In this review, we discuss human CC motif chemokine ligands: CCL1, CCL3, CCL4, CCL5, CCL18, CCL19, CCL20, CCL21, CCL25, CCL27, and CCL28 (CC motif chemokine receptor CCR5, CCR6, CCR7, CCR8, CCR9, and CCR10 ligands). We present their functioning in human physiology and in neoplasia, including their role in the proliferation, apoptosis resistance, drug resistance, migration, and invasion of cancer cells. We discuss the significance of chemokine receptors in organ-specific metastasis, as well as the influence of each chemokine on the recruitment of various cells to the tumor niche, such as cancer-associated fibroblasts (CAF), Kupffer cells, myeloid-derived suppressor cells (MDSC), osteoclasts, tumor-associated macrophages (TAM), tumor-infiltrating lymphocytes (TIL), and regulatory T cells (Treg). Finally, we show how the effect of the chemokines on vascular endothelial cells and lymphatic endothelial cells leads to angiogenesis and lymphangiogenesis.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (S.G.)
| | - Szymon Grochans
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (S.G.)
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (S.G.)
- Correspondence: ; Tel.: +48-914661515
| |
Collapse
|
130
|
Seifert AM, Eymer A, Heiduk M, Wehner R, Tunger A, von Renesse J, Decker R, Aust DE, Welsch T, Reissfelder C, Weitz J, Schmitz M, Seifert L. PD-1 Expression by Lymph Node and Intratumoral Regulatory T Cells Is Associated with Lymph Node Metastasis in Pancreatic Cancer. Cancers (Basel) 2020; 12:cancers12102756. [PMID: 32987956 PMCID: PMC7599971 DOI: 10.3390/cancers12102756] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Pancreatic cancer is a devastating disease and among the most immune-resistant tumor types. Single-agent immunotherapy has not demonstrated clinical benefits in pancreatic cancer patients, and combinational therapies targeting multiple mechanisms of immunosuppression are likely needed. T cell activation in lymph nodes is required for the efficacy of immunotherapy. Here, we phenotypically and functionally analyze T cells from tumor-draining lymph nodes, blood and tumors from patients with pancreatic cancer to decipher unknown immunosuppressive mechanisms and to identify potential immunotherapeutic targets. Abstract Pancreatic ductal adenocarcinoma (PDAC) is characterized by a mostly immunosuppressive microenvironment. Tumor-draining lymph nodes (TDLN) are a major site for priming of tumor-reactive T cells and also tumor metastasis. However, the phenotype and function of T cells in TDLNs from PDAC patients is unknown. In this study, lymph nodes from the pancreatic head (PH), the hepatoduodenal ligament (HDL) and the interaortocaval (IAC) region were obtained from 25 patients with adenocarcinoma of the pancreatic head. Additionally, tumors and matched blood were analyzed from 16 PDAC patients. Using multicolor flow cytometry, we performed a comprehensive analysis of T cells. CD4+ T cells were the predominant T cell subset in PDAC-draining lymph nodes. Overall, lymph node CD4+ and CD8+ T cells had a similar degree of activation, as measured by CD69, inducible T cell co-stimulator (ICOS) and CD137 (4-1BB) expression and interferon-γ (IFNγ) secretion. Expression of the inhibitory receptor programmed death 1 (PD-1) by lymph node and tumor-infiltrating regulatory T cells (Tregs) correlated with lymph node metastasis. Collectively, Treg cells and PD-1 are two relevant components of the immunosuppressive network in PDAC-draining lymph nodes and may be particularly attractive targets for combinatorial immunotherapeutic strategies in selected patients with node-positive PDAC.
Collapse
Affiliation(s)
- Adrian M. Seifert
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- Correspondence:
| | - Annabel Eymer
- Institute of Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (A.E.); (R.W.); (A.T.)
| | - Max Heiduk
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Rebekka Wehner
- Institute of Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (A.E.); (R.W.); (A.T.)
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Antje Tunger
- Institute of Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (A.E.); (R.W.); (A.T.)
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Janusz von Renesse
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
| | - Rahel Decker
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
| | - Daniela E. Aust
- Department of Pathology, University Hospital Carl Gustav Carus, Medical Faculty, University of Dresden, 01307 Dresden, Germany;
- NCT Biobank Dresden, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Thilo Welsch
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Christoph Reissfelder
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Marc Schmitz
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- Institute of Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (A.E.); (R.W.); (A.T.)
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Lena Seifert
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (M.H.); (J.v.R.); (R.D.); (T.W.); (J.W.); (L.S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| |
Collapse
|
131
|
Targeting Myeloid-Derived Suppressor Cells in Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12092626. [PMID: 32942545 PMCID: PMC7564060 DOI: 10.3390/cancers12092626] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Myeloid-Derived Suppressor Cells (MDSCs) have been regarded as the main promoters of cancer development in recent years. They can protect tumor cells from being eliminated by neutralizing the anti-tumor response mediated by T cells, macrophages and dendritic cells (DCs). Therefore, different treatment methods targeting MDSCs, including chemotherapy, radiotherapy and immunotherapy, have been developed and proven to effectively inhibit tumor expansion. Herein, we summarize the immunosuppressive role of MDSCs in the tumor microenvironment and some effective treatments targeting MDSCs, and discuss the differences between different therapies. Abstract Myeloid-derived suppressor cells (MDSCs), which are activated under pathological conditions, are a group of heterogeneous immature myeloid cells. MDSCs have potent capacities to support tumor growth via inhibition of the antitumoral immune response and/or the induction of immunosuppressive cells. In addition, multiple studies have demonstrated that MDSCs provide potential therapeutic targets for the elimination of immunosuppressive functions and the inhibition of tumor growth. The combination of targeting MDSCs and other therapeutic approaches has also demonstrated powerful antitumor effects. In this review, we summarize the characteristics of MDSCs in the tumor microenvironment (TME) and current strategies of cancer treatment by targeting MDSCs.
Collapse
|
132
|
Antineoplastic effects of targeting CCR5 and its therapeutic potential for colorectal cancer liver metastasis. J Cancer Res Clin Oncol 2020; 147:73-91. [PMID: 32902795 PMCID: PMC7810651 DOI: 10.1007/s00432-020-03382-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022]
Abstract
Purpose Liver metastasis is observed in up to 50% of colorectal cancer (CRC) patients. Available treatment options are limited and disease recurrence is often. Chemokine receptor 5 (CCR5) has attracted attention as novel therapeutic target for treating cancers. In this study, we reinforced the importance of CCR5 as therapeutic target in CRC and its liver metastasis by applying in vitro, in vivo and clinical investigations. Methods By targeting CCR5 via siRNAs or an FDA approved antagonist (maraviroc), we investigated the ensuing antineoplastic effects in three CRC cell lines. An animal model for CRC liver metastasis was used to evaluate time-dependent expressional modulation of the CCR5 axis by cDNA microarray. The model was also used to evaluate the in vivo efficacy of targeting CCR5 by maraviroc. Circulatory and tumor associated levels of CCR5 and its cognate ligands (CCL3, CCL4, CCL5) were analyzed by ELISA, qRT-PCR and immunohistochemistry. Results Targeting the CCR5 inhibited proliferative, migratory and clonogenic properties and interfered with cell cycle-related signaling cascades. In vivo findings showed significant induction of the CCR5 axis during the early liver colonization phase. Treatment with maraviroc significantly inhibited CRC liver metastasis in the animal model. Differential expression profiles of circulatory and tumor associated CCR5/ligands were observed in CRC patients and healthy controls. Conclusion The findings indicate that targeting the CCR5 axis can be an effective strategy for treating CRC liver metastasis. Electronic supplementary material The online version of this article (10.1007/s00432-020-03382-9) contains supplementary material, which is available to authorized users.
Collapse
|
133
|
Diao X. CXCR4 polymeric nanocomplex-mediated pulmonary delivery of siPD-L1: A novel procedure to enhance immunotherapy. Thorac Cancer 2020; 11:2753-2754. [PMID: 32893969 DOI: 10.1111/1759-7714.13627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 11/29/2022] Open
Affiliation(s)
- Xiayao Diao
- Department of Urology, Sun Yat-sen Memorial Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant, Tumor Epigenetics and Gene Regulation, Sun Yat-sen, Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
134
|
Cancer Acidity and Hypertonicity Contribute to Dysfunction of Tumor-Associated Dendritic Cells: Potential Impact on Antigen Cross-Presentation Machinery. Cancers (Basel) 2020; 12:cancers12092403. [PMID: 32847079 PMCID: PMC7565485 DOI: 10.3390/cancers12092403] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/13/2020] [Accepted: 08/16/2020] [Indexed: 01/21/2023] Open
Abstract
Macrophages (MΦ) and dendritic cells (DC), major players of the mononuclear phagocyte system (MoPh), are potent antigen presenting cells that steadily sense and respond to signals from the surrounding microenvironment, leading to either immunogenic or tolerogenic outcomes. Next to classical MHC-I/MHC-II antigen-presentation pathways described in the vast majority of cell types, a subset of MoPh (CD8+, XCR1+, CLEC9A+, BDCA3+ conventional DCs in human) is endowed with a high competence to cross-present external (engulfed) antigens on MHC-I molecules to CD8+ T-cells. This exceptional DC function is thought to be a crucial crossroad in cytotoxic antitumor immunity and has been extensively studied in the past decades. Biophysical and biochemical fingerprints of tumor micromilieus show significant spatiotemporal differences in comparison to non-neoplastic tissue. In tumors, low pH (mainly due to extracellular lactate accumulation via the Warburg effect and via glutaminolysis) and high oncotic and osmotic pressure (resulting from tumor debris, increased extracellular matrix components but in part also triggered by nutritive aspects) are—despite fluctuations and difficulties in measurement—likely the most constant general hallmarks of tumor microenvironment. Here, we focus on the influence of acidic and hypertonic micromilieu on the capacity of DCs to cross-present tumor-specific antigens. We discuss complex and in part controversial scientific data on the interference of these factors with to date reported mechanisms of antigen uptake, processing and cross-presentation, and we highlight their potential role in cancer immune escape and poor clinical response to DC vaccines.
Collapse
|
135
|
Saka D, Gökalp M, Piyade B, Cevik NC, Arik Sever E, Unutmaz D, Ceyhan GO, Demir IE, Asimgil H. Mechanisms of T-Cell Exhaustion in Pancreatic Cancer. Cancers (Basel) 2020; 12:cancers12082274. [PMID: 32823814 PMCID: PMC7464444 DOI: 10.3390/cancers12082274] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
T-cell exhaustion is a phenomenon that represents the dysfunctional state of T cells in chronic infections and cancer and is closely associated with poor prognosis in many cancers. The endogenous T-cell immunity and genetically edited cell therapies (CAR-T) failed to prevent tumor immune evasion. The effector T-cell activity is perturbed by an imbalance between inhibitory and stimulatory signals causing a reprogramming in metabolism and the high levels of multiple inhibitory receptors like programmed cell death protein-1 (PD-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), and Lymphocyte-activation gene 3 (Lag-3). Despite the efforts to neutralize inhibitory receptors by a single agent or combinatorial immune checkpoint inhibitors to boost effector function, PDAC remains unresponsive to these therapies, suggesting that multiple molecular mechanisms play a role in stimulating the exhaustion state of tumor-infiltrating T cells. Recent studies utilizing transcriptomics, mass cytometry, and epigenomics revealed a critical role of Thymocyte selection-associated high mobility group box protein (TOX) genes and TOX-associated pathways, driving T-cell exhaustion in chronic infection and cancer. Here, we will review recently defined molecular, genetic, and cellular factors that drive T-cell exhaustion in PDAC. We will also discuss the effects of available immune checkpoint inhibitors and the latest clinical trials targeting various molecular factors mediating T-cell exhaustion in PDAC.
Collapse
Affiliation(s)
- Didem Saka
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
| | - Muazzez Gökalp
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
| | - Betül Piyade
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
| | - Nedim Can Cevik
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
| | - Elif Arik Sever
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
| | - Derya Unutmaz
- Jackson Laboratory of Genomic Medicine, Farmington, CT 06032, USA;
| | - Güralp O. Ceyhan
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
- Correspondence: (G.O.C.); (I.E.D.); Tel.: +90-5320514424 (G.O.C.); +49-8941405868 (I.E.D.)
| | - Ihsan Ekin Demir
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
- Department of Surgery, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- Correspondence: (G.O.C.); (I.E.D.); Tel.: +90-5320514424 (G.O.C.); +49-8941405868 (I.E.D.)
| | - Hande Asimgil
- Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey; (D.S.); (M.G.); (B.P.); (N.C.C.); (E.A.S.); (H.A.)
- Department of Surgery, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
136
|
De Cicco P, Ercolano G, Ianaro A. The New Era of Cancer Immunotherapy: Targeting Myeloid-Derived Suppressor Cells to Overcome Immune Evasion. Front Immunol 2020; 11:1680. [PMID: 32849585 PMCID: PMC7406792 DOI: 10.3389/fimmu.2020.01680] [Citation(s) in RCA: 218] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022] Open
Abstract
Suppression of antitumor immune responses is one of the main mechanisms by which tumor cells escape from destruction by the immune system. Myeloid-derived suppressor cells (MDSCs) represent the main immunosuppressive cells present in the tumor microenvironment (TME) that sustain cancer progression. MDSCs are a heterogeneous group of immature myeloid cells with a potent activity against T-cell. Studies in mice have demonstrated that MDSCs accumulate in several types of cancer where they promote invasion, angiogenesis, and metastasis formation and inhibit antitumor immunity. In addition, different clinical studies have shown that MDSCs levels in the peripheral blood of cancer patients correlates with tumor burden, stage and with poor prognosis in multiple malignancies. Thus, MDSCs are the major obstacle to many cancer immunotherapies and their targeting may be a beneficial strategy for improvement the efficiency of immunotherapeutic interventions. However, the great heterogeneity of these cells makes their identification in human cancer very challenging. Since both the phenotype and mechanisms of action of MDSCs appear to be tumor-dependent, it is important to accurately characterized the precise MDSC subsets that have clinical relevance in each tumor environment to more efficiently target them. In this review we summarize the phenotype and the suppressive mechanisms of MDSCs populations expanded within different tumor contexts. Further, we discuss about their clinical relevance for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Paola De Cicco
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Giuseppe Ercolano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy.,Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland.,Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Angela Ianaro
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| |
Collapse
|
137
|
Yang T, Li J, Li R, Yang C, Zhang W, Qiu Y, Yang C, Rong R. Correlation between MDSC and Immune Tolerance in Transplantation: Cytokines, Pathways and Cell-cell Interaction. Curr Gene Ther 2020; 19:81-92. [PMID: 31237207 DOI: 10.2174/1566523219666190618093707] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/06/2019] [Accepted: 05/24/2019] [Indexed: 11/22/2022]
Abstract
MDSCs play an important role in the induction of immune tolerance. Cytokines and chemokines (GM-CSF, IL-6) contributed to the expansion, accumulation of MDSCs, and MDSCs function through iNOS, arginase and PD-L1. MDSCs are recruited and regulated through JAK/STAT, mTOR and Raf/MEK/ERK signaling pathways. MDSCs' immunosuppressive functions were realized through Tregs-mediated pathways and their direct suppression of immune cells. All of the above contribute to the MDSC-related immune tolerance in transplantation. MDSCs have huge potential in prolonging graft survival and reducing rejection through different ways and many other factors worthy to be further investigated are also introduced.
Collapse
Affiliation(s)
- Tianying Yang
- Department of Urology, ZhongShan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Jiawei Li
- Department of Urology, ZhongShan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ruimin Li
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.,Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunchen Yang
- Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weitao Zhang
- Department of Urology, ZhongShan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yue Qiu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Yang
- Department of Urology, ZhongShan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ruiming Rong
- Department of Urology, ZhongShan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.,Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
138
|
Ellwanger JH, Kulmann-Leal B, Kaminski VDL, Rodrigues AG, Bragatte MADS, Chies JAB. Beyond HIV infection: Neglected and varied impacts of CCR5 and CCR5Δ32 on viral diseases. Virus Res 2020; 286:198040. [PMID: 32479976 PMCID: PMC7260533 DOI: 10.1016/j.virusres.2020.198040] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022]
Abstract
CCR5 regulates multiple cell types (e.g., T regulatory and Natural Killer cells) and immune responses. The effects of CCR5, CCR5Δ32 (variant associated with reduced CCR5 expression) and CCR5 antagonists vary between infections. CCR5 affects the pathogenesis of flaviviruses, especially in the brain. The genetic variant CCR5Δ32 increases the risk of symptomatic West Nile virus infection. The triad “CCR5, extracellular vesicles and infections” is an emerging topic.
The interactions between chemokine receptors and their ligands may affect susceptibility to infectious diseases as well as their clinical manifestations. These interactions mediate both the traffic of inflammatory cells and virus-associated immune responses. In the context of viral infections, the human C-C chemokine receptor type 5 (CCR5) receives great attention from the scientific community due to its role as an HIV-1 co-receptor. The genetic variant CCR5Δ32 (32 base-pair deletion in CCR5 gene) impairs CCR5 expression on the cell surface and is associated with protection against HIV infection in homozygous individuals. Also, the genetic variant CCR5Δ32 modifies the CCR5-mediated inflammatory responses in various conditions, such as inflammatory and infectious diseases. CCR5 antagonists mimic, at least in part, the natural effects of the CCR5Δ32 in humans, which explains the growing interest in the potential benefits of using CCR5 modulators for the treatment of different diseases. Nevertheless, beyond HIV infection, understanding the effects of the CCR5Δ32 variant in multiple viral infections is essential to shed light on the potential effects of the CCR5 modulators from a broader perspective. In this context, this review discusses the involvement of CCR5 and the effects of the CCR5Δ32 in human infections caused by the following pathogens: West Nile virus, Influenza virus, Human papillomavirus, Hepatitis B virus, Hepatitis C virus, Poliovirus, Dengue virus, Human cytomegalovirus, Crimean-Congo hemorrhagic fever virus, Enterovirus, Japanese encephalitis virus, and Hantavirus. Subsequently, this review addresses the impacts of CCR5 gene editing and CCR5 modulation on health and viral diseases. Also, this article connects recent findings regarding extracellular vesicles (e.g., exosomes), viruses, and CCR5. Neglected and emerging topics in “CCR5 research” are briefly described, with focus on Rocio virus, Zika virus, Epstein-Barr virus, and Rhinovirus. Finally, the potential influence of CCR5 on the immune responses to coronaviruses is discussed.
Collapse
Affiliation(s)
- Joel Henrique Ellwanger
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Bruna Kulmann-Leal
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Valéria de Lima Kaminski
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Biotecnologia, Laboratório de Imunologia Aplicada, Instituto de Ciência e Tecnologia - ICT, Universidade Federal de São Paulo - UNIFESP, São José dos Campos, São Paulo, Brazil
| | - Andressa Gonçalves Rodrigues
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Marcelo Alves de Souza Bragatte
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Núcleo de Bioinformática do Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - José Artur Bogo Chies
- Laboratório de Imunobiologia e Imunogenética, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil.
| |
Collapse
|
139
|
Del Prete A, Sozio F, Barbazza I, Salvi V, Tiberio L, Laffranchi M, Gismondi A, Bosisio D, Schioppa T, Sozzani S. Functional Role of Dendritic Cell Subsets in Cancer Progression and Clinical Implications. Int J Mol Sci 2020; 21:ijms21113930. [PMID: 32486257 PMCID: PMC7312661 DOI: 10.3390/ijms21113930] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells (DCs) constitute a complex network of cell subsets with common functions but also with many divergent aspects. All dendritic cell subsets share the ability to prime T cell response and to undergo a complex trafficking program related to their stage of maturation and function. For these reasons, dendritic cells are implicated in a large variety of both protective and detrimental immune responses, including a crucial role in promoting anti-tumor responses. Although cDC1s are the most potent subset in tumor antigen cross-presentation, they are not sufficient to induce full-strength anti-tumor cytotoxic T cell response and need close interaction and cooperativity with the other dendritic cell subsets, namely cDC2s and pDCs. This review will take into consideration different aspects of DC biology, including the functional role of dendritic cell subsets in both fostering and suppressing tumor growth, the mechanisms underlying their recruitment into the tumor microenvironment, as well as the prognostic value and the potentiality of dendritic cell therapeutic targeting. Understanding the specificity of dendritic cell subsets will allow to gain insights on role of these cells in pathological conditions and to design new selective promising therapeutic approaches.
Collapse
Affiliation(s)
- Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
- Humanitas Clinical and Research Center—IRCCS, Via Manzoni 56, 20089 Rozzano (MI), Italy
| | - Francesca Sozio
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
- Humanitas Clinical and Research Center—IRCCS, Via Manzoni 56, 20089 Rozzano (MI), Italy
| | - Ilaria Barbazza
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
| | - Valentina Salvi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
| | - Laura Tiberio
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
| | - Mattia Laffranchi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
| | - Angela Gismondi
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy;
| | - Daniela Bosisio
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
| | - Tiziana Schioppa
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (A.D.P.); (F.S.); (I.B.); (V.S.); (L.T.); (M.L.); (D.B.); (T.S.)
- Humanitas Clinical and Research Center—IRCCS, Via Manzoni 56, 20089 Rozzano (MI), Italy
| | - Silvano Sozzani
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy;
- Correspondence: ; Tel.: +39-06-4434-0632
| |
Collapse
|
140
|
Li Z, Wang Y, Shen Y, Qian C, Oupicky D, Sun M. Targeting pulmonary tumor microenvironment with CXCR4-inhibiting nanocomplex to enhance anti-PD-L1 immunotherapy. SCIENCE ADVANCES 2020; 6:eaaz9240. [PMID: 32440550 PMCID: PMC7228744 DOI: 10.1126/sciadv.aaz9240] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/02/2020] [Indexed: 05/19/2023]
Abstract
Anti-programmed cell death 1 ligand 1 (PD-L1) therapy is extraordinarily effective in select patients with cancer. However, insufficient lymphocytic infiltration, weak T cell-induced inflammation, and immunosuppressive cell accumulation in the tumor microenvironment (TME) may greatly diminish the efficacy. Here, we report development of the FX@HP nanocomplex composed of fluorinated polymerized CXCR4 antagonism (FX) and paclitaxel-loaded human serum albumin (HP) for pulmonary delivery of anti-PD-L1 small interfering RNA (siPD-L1) to treat orthotopic lung tumors. FX@HP induced T cell infiltration, increased expression of calreticulin on tumor cells, and reduced the myeloid-derived suppressor cells/regulatory T cells in the TME, thereby acting synergistically with siPD-L1 for effective immunotherapy. Our work suggests that the CXCR4-inhibiting nanocomplex decreases tumor fibrosis, facilitates T cell infiltration and relieves immunosuppression to modulate the immune process to improve the objective response rate of anti-PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Zhaoting Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yixin Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yuexin Shen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Chenggen Qian
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - David Oupicky
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Minjie Sun
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
141
|
Xie M, Wei J, Xu J. Inducers, Attractors and Modulators of CD4 + Treg Cells in Non-Small-Cell Lung Cancer. Front Immunol 2020; 11:676. [PMID: 32425930 PMCID: PMC7212357 DOI: 10.3389/fimmu.2020.00676] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/26/2020] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the leading cause of cancer-associated deaths worldwide, with non-small cell-lung cancer (NSCLC) accounting for approximately 80% of cases. Immune escape has been demonstrated to play a key role in the initiation and progression of NSCLC, although the underlying mechanisms are diverse and their puzzling nature is far from being understood. As a critical participant in immune escape, the CD4+ T cell subset of regulatory T (Treg) cells, with their immunosuppressive functions, has been implicated in the occurrence of many types of cancers. Additionally, therapies based on Treg blockade have benefited a portion of cancer patients, including those with NSCLC. Accumulating literature has noted high Treg infiltration in NSCLC tumor tissues, bone marrow, lymph nodes and/or blood; moreover, the tumor milieu is involved in regulating the proliferation, differentiation, recruitment and suppressive functions of Treg cells. Multifarious mechanisms by which CD4+ Treg cells are generated, attracted and modulated in the NSCLC milieu will be discussed in this review.
Collapse
Affiliation(s)
- Mengxiao Xie
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Jia Wei
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| | - Jian Xu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, Nanjing, China
| |
Collapse
|
142
|
Seng A, Krausz KL, Pei D, Koestler DC, Fischer RT, Yankee TM, Markiewicz MA. Coexpression of FOXP3 and a Helios isoform enhances the effectiveness of human engineered regulatory T cells. Blood Adv 2020; 4:1325-1339. [PMID: 32259202 PMCID: PMC7160257 DOI: 10.1182/bloodadvances.2019000965] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/04/2020] [Indexed: 12/13/2022] Open
Abstract
Regulatory T cells (Tregs) are a subset of immune cells that suppress the immune response. Treg therapy for inflammatory diseases is being tested in the clinic, with moderate success. However, it is difficult to isolate and expand Tregs to sufficient numbers. Engineered Tregs (eTregs) can be generated in larger quantities by genetically manipulating conventional T cells to express FOXP3. These eTregs can suppress in vitro and in vivo but not as effectively as endogenous Tregs. We hypothesized that ectopic expression of the transcription factor Helios along with FOXP3 is required for optimal eTreg immunosuppression. To test this theory, we generated eTregs by retrovirally transducing total human T cells (CD4+ and CD8+) with FOXP3 alone or with each of the 2 predominant isoforms of Helios. Expression of both FOXP3 and the full-length isoform of Helios was required for eTreg-mediated disease delay in a xenogeneic graft-versus-host disease model. In vitro, this corresponded with superior suppressive function of FOXP3 and full-length Helios-expressing CD4+ and CD8+ eTregs. RNA sequencing showed that the addition of full-length Helios changed gene expression in cellular pathways and the Treg signature compared with FOXP3 alone or the other major Helios isoform. Together, these results show that functional human CD4+ and CD8+ eTregs can be generated from total human T cells by coexpressing FOXP3 and full-length Helios.
Collapse
Affiliation(s)
- Amara Seng
- Department of Microbiology, Molecular Genetics, and Immunology, and
| | - Kelsey L Krausz
- Department of Microbiology, Molecular Genetics, and Immunology, and
| | - Dong Pei
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS; and
| | - Devin C Koestler
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS; and
| | - Ryan T Fischer
- Pediatric Gastroenterology, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO
| | - Thomas M Yankee
- Department of Microbiology, Molecular Genetics, and Immunology, and
| | | |
Collapse
|
143
|
Zhou Q, Tao X, Xia S, Guo F, Pan C, Xiang H, Shang D. T Lymphocytes: A Promising Immunotherapeutic Target for Pancreatitis and Pancreatic Cancer? Front Oncol 2020; 10:382. [PMID: 32266154 PMCID: PMC7105736 DOI: 10.3389/fonc.2020.00382] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/04/2020] [Indexed: 01/18/2023] Open
Abstract
Pancreatic disorders cause a broad spectrum of clinical diseases, mainly including acute and chronic pancreatitis and pancreatic cancer, and are associated with high global rates of morbidity and mortality. Unfortunately, the pathogenesis of pancreatic disease remains obscure, and there is a lack of specific treatments. T lymphocytes (T cells) play a vital role in the adaptive immune systems of multicellular organisms. During pancreatic disease development, local imbalances in T-cell subsets in inflammatory and tumor environments and the circulation have been observed. Furthermore, agents targeting T cells have been shown to reverse the natural course of pancreatic diseases. In this review, we have discussed the clinical relevance of T-cell alterations as a potential outcome predictor and the underlying mechanisms, as well as the present status of immunotherapy targeting T cells in pancreatitis and neoplasms. The breakthrough findings summarized in this review have important implications for innovative drug development and the prospective use of immunotherapy for pancreatitis and pancreatic cancer.
Collapse
Affiliation(s)
- Qi Zhou
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xufeng Tao
- School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Shilin Xia
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fangyue Guo
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Chen Pan
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of General Surgery, Pancreatic-Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hong Xiang
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of General Surgery, Pancreatic-Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
144
|
Xu H, Hu M, Liu M, An S, Guan K, Wang M, Li L, Zhang J, Li J, Huang L. Nano-puerarin regulates tumor microenvironment and facilitates chemo- and immunotherapy in murine triple negative breast cancer model. Biomaterials 2020; 235:119769. [PMID: 31986348 PMCID: PMC7093100 DOI: 10.1016/j.biomaterials.2020.119769] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/22/2019] [Accepted: 01/07/2020] [Indexed: 01/05/2023]
Abstract
Tumor associated fibroblasts (TAFs) are key stromal cells mediating the desmoplastic reaction and being partially responsible for the drug-resistance and immunosuppressive microenvironment formation in solid tumors. Delivery of genotoxic drugs off-targetedly to kill TAFs results in production of Wnt16 which renders the neighboring tumor cells drug resistant as shown in our previous study (PMC4623876). Our current approach looks for means to deactivate, rather than kill, TAFs. Reactive oxygen species (ROS) are the central hub of multiple profibrogenic pathways and indispensable for TAFs activation. Herein, puerarin was identified to effectively downregulate ROS production in the activated myofibroblast. In this study, a novel puerarin nanoemulsion (nanoPue) was developed to improve the solubility and bioavailability of puerarin. NanoPue significantly deactivated the stromal microenvironment (e.g., ~6-fold reduction of TAFs in nanoPue treated mice compared with the PBS control, p < 0.0001) and facilitated chemotherapy effect of nano-paclitaxel in the desmoplastic triple-negative breast cancer (TNBC) model. Moreover, the removal of the physical barrier increased intra-tumoral infiltration of cytotoxic T cell by 2-fold. This activated immune microenvironment allowed nanoPue to synergize PD-L1 blockade therapy in TNBC model.
Collapse
Affiliation(s)
- Huan Xu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States; Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, People's Republic of China
| | - Mengying Hu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Mengrui Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Sai An
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Kaiyun Guan
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Menglin Wang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Lei Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Jing Zhang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Jun Li
- ZY Therapeutics Inc., 400 Park Office Dr. Suite 310, Research Triangle Park, NC, 27709, United States
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States.
| |
Collapse
|
145
|
Fan JQ, Wang MF, Chen HL, Shang D, Das JK, Song J. Current advances and outlooks in immunotherapy for pancreatic ductal adenocarcinoma. Mol Cancer 2020; 19:32. [PMID: 32061257 PMCID: PMC7023714 DOI: 10.1186/s12943-020-01151-3] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/06/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an incurable cancer resistant to traditional treatments, although a limited number of early-stage patients can undergo radical resection. Immunotherapies for the treatment of haematological malignancies as well as solid tumours have been substantially improved over the past decades, and impressive results have been obtained in recent preclinical and clinical trials. However, PDAC is likely the exception because of its unique tumour microenvironment (TME). In this review, we summarize the characteristics of the PDAC TME and focus on the network of various tumour-infiltrating immune cells, outlining the current advances in PDAC immunotherapy and addressing the effect of the PDAC TME on immunotherapy. This review further explores the combinations of different therapies used to enhance antitumour efficacy or reverse immunodeficiencies and describes optimizable immunotherapeutic strategies for PDAC. The concordant combination of various treatments, such as targeting cancer cells and the stroma, reversing suppressive immune reactions and enhancing antitumour reactivity, may be the most promising approach for the treatment of PDAC. Traditional treatments, especially chemotherapy, may also be optimized for individual patients to remodel the immunosuppressive microenvironment for enhanced therapy.
Collapse
Affiliation(s)
- Jia-qiao Fan
- Third General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Meng-Fei Wang
- Third General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hai-Long Chen
- Third General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Third General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jugal K. Das
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, TX USA
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, TX USA
| |
Collapse
|
146
|
Kim JH, Kim BS, Lee SK. Regulatory T Cells in Tumor Microenvironment and Approach for Anticancer Immunotherapy. Immune Netw 2020; 20:e4. [PMID: 32158592 PMCID: PMC7049587 DOI: 10.4110/in.2020.20.e4] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 01/30/2020] [Accepted: 02/02/2020] [Indexed: 12/21/2022] Open
Abstract
Tregs have a role in immunological tolerance and immune homeostasis by suppressing immune reactions, and its therapeutic potential is critical in autoimmune diseases and cancers. There have been multiple studies conducted on Tregs because of their roles in immune suppression and therapeutic potential. In tumor immunity, Tregs can promote the development and progression of tumors by preventing effective anti-tumor immune responses in tumor-bearing hosts. High infiltration of Tregs into tumor tissue results in poor survival in various types of cancer patients. Identifying factors specifically expressed in Tregs that affect the maintenance of stability and function of Tregs is important for understanding cancer pathogenesis and identifying therapeutic targets. Thus, manipulation of Tregs is a promising anticancer strategy, but finding markers for Treg-specific depletion and controlling these cells require fine-tuning and further research. Here, we discuss the role of Tregs in cancer and the development of Treg-targeted therapies to promote cancer immunotherapy.
Collapse
Affiliation(s)
- Jung-Ho Kim
- Research Institute for Precision Immune-Medicine, Good T Cells, Inc., Seoul 03722, Korea
| | - Beom Seok Kim
- Research Institute for Precision Immune-Medicine, Good T Cells, Inc., Seoul 03722, Korea
| | - Sang-Kyou Lee
- Research Institute for Precision Immune-Medicine, Good T Cells, Inc., Seoul 03722, Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
147
|
Do HTT, Lee CH, Cho J. Chemokines and their Receptors: Multifaceted Roles in Cancer Progression and Potential Value as Cancer Prognostic Markers. Cancers (Basel) 2020; 12:E287. [PMID: 31991604 PMCID: PMC7072521 DOI: 10.3390/cancers12020287] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/14/2020] [Accepted: 01/19/2020] [Indexed: 12/19/2022] Open
Abstract
Chemokines are chemotactic cytokines that mediate immune cell chemotaxis and lymphoid tissue development. Recent advances have indicated that chemokines and their cognate receptors play critical roles in cancer-related inflammation and cancer progression. On the basis of these findings, the chemokine system has become a new potential drug target for cancer immunotherapy. In this review, we summarize the essential roles of the complex network of chemokines and their receptors in cancer progression. Furthermore, we discuss the potential value of the chemokine system as a cancer prognostic marker. The chemokine system regulates the infiltration of immune cells into the tumor microenvironment, which induces both pro- and anti-immunity and promotes or suppresses tumor growth and proliferation, angiogenesis, and metastasis. Increasing evidence indicates the promising prognostic value of the chemokine system in cancer patients. While CCL2, CXCL10, and CX3CL1/CX3CR1 can serve as favorable or unfavorable prognostic factors depending on the cancer types, CCL14 and XCL1 possess good prognostic value. Other chemokines such as CXCL1, CXCL8, and CXCL12 are poor prognostic markers. Despite vast advances in our understanding of the complex nature of the chemokine system in tumor biology, knowledge about the multifaceted roles of the chemokine system in different types of cancers is still limited. Further studies are necessary to decipher distinct roles within the chemokine system in terms of cancer progression and to validate their potential value in cancer prognosis.
Collapse
Affiliation(s)
| | | | - Jungsook Cho
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi 10326, Korea; (H.T.T.D.); (C.H.L.)
| |
Collapse
|
148
|
Siret C, Collignon A, Silvy F, Robert S, Cheyrol T, André P, Rigot V, Iovanna J, van de Pavert S, Lombardo D, Mas E, Martirosyan A. Deciphering the Crosstalk Between Myeloid-Derived Suppressor Cells and Regulatory T Cells in Pancreatic Ductal Adenocarcinoma. Front Immunol 2020; 10:3070. [PMID: 32038621 PMCID: PMC6987391 DOI: 10.3389/fimmu.2019.03070] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a fatal disease with rising incidence and a remarkable resistance to current therapies. The reasons for this therapeutic failure include the tumor's extensive infiltration by immunosuppressive cells such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). By using light sheet fluorescent microscopy, we identified here direct interactions between these major immunoregulatory cells in PDAC. The in vivo depletion of MDSCs led to a significant reduction in Tregs in the pancreatic tumors. Through videomicroscopy and ex vivo functional assays we have shown that (i) MDSCs are able to induce Treg cells in a cell-cell dependent manner; (ii) Treg cells affect the survival and/or the proliferation of MDSCs. Furthermore, we have observed contacts between MDSCs and Treg cells at different stages of human cancer. Overall our findings suggest that interactions between MDSCs and Treg cells contribute to PDAC immunosuppressive environment.
Collapse
Affiliation(s)
- Carole Siret
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Aurélie Collignon
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| | - Françoise Silvy
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| | - Stéphane Robert
- Aix Marseille Univ, INSERM, VRCM, Centre de Recherche Vasculaire de Marseille, Marseille, France
| | - Thierry Cheyrol
- Aix Marseille Univ, CEFOS, Centre d'exploration Fonctionnelle Scientifique, Marseille, France
| | - Perrine André
- Aix Marseille Univ, CEFOS, Centre d'exploration Fonctionnelle Scientifique, Marseille, France
| | - Véronique Rigot
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| | - Juan Iovanna
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Centre de Recherche en Cancérologie de Marseille, Marseille, France
| | - Serge van de Pavert
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Dominique Lombardo
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| | - Eric Mas
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| | - Anna Martirosyan
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| |
Collapse
|
149
|
The CCR5 antagonist maraviroc causes remission of pancreatic cancer liver metastasis in nude rats based on cell cycle inhibition and apoptosis induction. Cancer Lett 2020; 474:82-93. [PMID: 31954769 DOI: 10.1016/j.canlet.2020.01.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 12/05/2019] [Accepted: 01/10/2020] [Indexed: 12/18/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease, and novel therapeutic strategies are urgently needed. Recently, expression of the C-C chemokine receptor 5 (CCR5) and its ligands has been found to play an important role in cancer progression and metastasis. In this study, we blocked the CCR5 receptor by the FDA approved antagonist maraviroc (MVC) in Suit2-007 and MIA-PaCa-2 human PDAC cells. The treatment significantly inhibited their proliferation and induced apoptosis of exposed cells as evidenced by caspases activation and increased Bax levels. Moreover, MVC inhibited the cell cycle by down regulating the proteins of the complexes of cyclin dependent kinase (CDK) 4/6 - Cyclin D and CDK2 - Cyclin E, as well as by increasing the protein levels of CDK inhibitors p18, p21 and p27. In line with this, MVC caused significant retardation of Suit2-007 cells growing in a PDAC liver metastasis xenograft model (p < 0.05). These results suggest that maraviroc could be a promising treatment strategy for PDAC patients with liver metastases.
Collapse
|
150
|
Mempel TR, Marangoni F. Guidance factors orchestrating regulatory T cell positioning in tissues during development, homeostasis, and response. Immunol Rev 2020; 289:129-141. [PMID: 30977195 DOI: 10.1111/imr.12761] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 12/29/2022]
Abstract
Over their lifetime, regulatory T cells (Treg) recalibrate their expression of trafficking receptors multiple times as they progress through development, respond to immune challenges, or adapt to the requirements of functioning in various non-lymphoid tissue environments. These trafficking receptors, which include chemokine receptors and other G-protein coupled receptors, integrins, as well as selectins and their ligands, enable Treg not only to enter appropriate tissues from the bloodstream via post-capillary venules, but also to navigate these tissues to locally execute their immune-regulatory functions, and finally to seek out the right antigen-presenting cells and interact with these, in part in order to receive the signals that sustain their survival, proliferation, and functional activity, in part in order to execute their immuno-regulatory function by altering antigen presenting cell function. Here, we will review our current knowledge of when and in what ways Treg alter their trafficking properties. We will focus on the chemokine system and try to identify specialized, non-redundant roles of individual receptors as well as similarities and differences to the conventional T cell compartment.
Collapse
Affiliation(s)
- Thorsten R Mempel
- The Center for Immunology and Inflammatory Diseases at Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Francesco Marangoni
- The Center for Immunology and Inflammatory Diseases at Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|