1
|
Alfar R, Napoleon JV, Shahriar I, Finnell R, Walchle C, Johnson A, Low PS. Selective reprogramming of regulatory T cells in solid tumors can strongly enhance or inhibit tumor growth. Front Immunol 2023; 14:1274199. [PMID: 37928524 PMCID: PMC10623129 DOI: 10.3389/fimmu.2023.1274199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023] Open
Abstract
Folate receptor delta (FRδ) has been used as a biomarker for regulatory T cells (Tregs), because its expression is limited to Tregs and ovum. Although FRδ is unable to bind folate, we have used molecular docking software to identify a folate congener that binds FRδ with high affinity and have exploited this FRδ-specific ligand to target attached drugs (imaging agents, immune activators, and immune suppressors) specifically to Tregs in murine tumor xenografts. Analysis of treated tumors demonstrates that targeting of a Toll-like receptor 7 agonist inhibits Treg expression of FOXP3, PD-1, CTLA4, and HELIOS, resulting in 40-80% reduction in tumor growth and repolarization of other tumor-infiltrating immune cells to more inflammatory phenotypes. Targeting of the immunosuppressive drug dexamethasone, in contrast, promotes enhanced tumor growth and shifts the tumor-infiltrating immune cells to more anti-inflammatory phenotypes. Since Tregs comprise <1% of cells in the tumor masses examined, and since the targeted drugs are not internalized by cancer cells, these data demonstrate that Tregs exert a disproportionately large effect on tumor growth. Because the targeted drug did not bind to Tregs or other immune cells in healthy tissues, the data demonstrate that the immunosuppressive properties of Tregs in tumors can be manipulated without causing systemic toxicities associated with global reprogramming of the immune system.
Collapse
Affiliation(s)
- Rami Alfar
- Department of Chemistry, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - John V. Napoleon
- Department of Chemistry, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Imrul Shahriar
- Department of Chemistry, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Richard Finnell
- Departments of Molecular and Cellular Biology, Molecular and Human Genetics and Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Cole Walchle
- Department of Chemistry, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Austin Johnson
- Department of Chemistry, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| | - Philip S. Low
- Department of Chemistry, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
2
|
Zhang M, Fang Y, Fu X, Liu J, Liu Y, Zhu Z, Ni Y, Yao M, Pan Q, Cao W, Li Z, Dong C. Cancer-associated fibroblasts nurture LGR5 marked liver tumor-initiating cells and promote their tumor formation, growth, and metastasis. Cancer Med 2023; 12:18032-18049. [PMID: 37578396 PMCID: PMC10524013 DOI: 10.1002/cam4.6408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 06/27/2023] [Accepted: 07/25/2023] [Indexed: 08/15/2023] Open
Abstract
BACKGROUND & AIMS In liver cancer, leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) compartment represents an important tumor-initiating cell (TIC) population and served as a potential therapeutic target. Cancer-associated fibroblasts (CAFs) is a critical part of the tumor microenvironment, heavily influenced TIC function and fate. However, deeply investigations have been hindered by the lack of accurate preclinical models to investigate the interaction between CAFs and TIC. Organoids model have achieved major advancements as a precious research model for recapitulating the morphological aspects of organs, and thus also serving as a candidate model to investigate the mutual interaction between different cell types. Consequently, this study aimed to construct a three-dimensional (3D) co-culture organoid model of primary LGR5-expressing tumor stem cells from primary murine liver tumors with CAFs to investigate the impact of CAFs on LGR5 marked TICs in liver cancer. MATERIALS AND METHODS First, both of the transgenic LGR5-diphtheria toxin receptor (DTR)-GFP knock-in mice and transgenic Rosa26-mT mice developed primary liver tumors by diethylnitrosamine (DEN) administration. Tumor organoids and CAFs were generated from those primary liver cancer separately. Second, LGR5-expressing TICs organoid with CAFs were established ex vivo based on cell-cell contact or trans-well co-culture system, and the mutual influence between those two types of cells was further investigated. Subsequently, immunodeficient mouse-based xenograft model was further adopted to evaluate the influence of CAFs to LGR5 tumor stem cell, tumor formation, and metastasis. RESULTS The co-culture organoid model composed of murine liver tumor LGR5+ tumor-initiating cells and CAFs in 3D co-culture was successfully established, with the intention to investigate their mutual interaction. The existence of CAFs upon engrafting tumor organoids resulted in dramatic higher number of LGR5+ cells in the neoplasia when compared with engrafting tumor organoids alone. Furthermore, ex vivo culture of isolated LGR5+ cells from tumors of co-engrafted mice formed significantly larger size of organoids than mono-engrafted. Our results also indicated significantly larger size and number of formed organoids, when LGR5+ cells co-cultured with CAF in both cell-cell contact and paracrine signaling in vitro, comparing to LGR5+ cells alone. Furthermore, we found that specific knockout of LGR5 expressing cells suppressed CAF-mediated promotion of tumor formation, growth, and metastasis in the experimental mice model. CONCLUSIONS Altogether, in a 3D co-culture type of murine liver LGR5+ cells and cancer-associated fibroblasts, we have demonstrated robust effects of CAFs in the promotion of LGR5 marked liver TICs. We also further revealed the influence of tumor microenvironment on stem cell-related therapy, suggesting the possibility of combing CAF-targeted and tumor stem cell targeted therapy in treating liver cancer.
Collapse
Affiliation(s)
- Mingna Zhang
- Department of OncologyPostgraduate Training Base of Jinzhou Medical University, Shanghai East HospitalShanghaiChina
| | - Yiqiao Fang
- Department of Thyroid and Parathyroid Surgery, West China HospitalSichuan UniversityChengduSichuanChina
- Laboratory of Thyroid and Parathyroid Diseases, Frontiers Science Center for Disease‐Related Molecular Network, West China HospitalSichuan UniversityChengduSichuanChina
| | - Xia Fu
- Department of Outpatients, West China HospitalSichuan UniversityChengduSichuanChina
| | - Jiaye Liu
- Department of Thyroid and Parathyroid Surgery, West China HospitalSichuan UniversityChengduSichuanChina
- Laboratory of Thyroid and Parathyroid Diseases, Frontiers Science Center for Disease‐Related Molecular Network, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yang Liu
- Department of Obsterics and Gynecology, Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Zhounan Zhu
- Department of OncologyShanghai East Hospital, Tongji University School of Medicine, Tongji UniversityShanghaiPeople's Republic of China
| | - Yinyun Ni
- Department of Respiratory and Critical Care Medicine, National Clinic al Research Center for Geriatrics, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital, West China School of MedicineSichuan UniversityChengduSichuanChina
| | - Menglin Yao
- Department of Respiratory and Critical Care Medicine, National Clinic al Research Center for Geriatrics, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital, West China School of MedicineSichuan UniversityChengduSichuanChina
| | - Qiuwei Pan
- Department of Gastroenterology and HepatologyErasmus Medical CenterRotterdamthe Netherlands
| | - Wanlu Cao
- Department of OncologyShanghai East Hospital, Tongji University School of Medicine, Tongji UniversityShanghaiPeople's Republic of China
| | - Zhihui Li
- Department of Thyroid and Parathyroid Surgery, West China HospitalSichuan UniversityChengduSichuanChina
- Laboratory of Thyroid and Parathyroid Diseases, Frontiers Science Center for Disease‐Related Molecular Network, West China HospitalSichuan UniversityChengduSichuanChina
| | - Chunyan Dong
- Department of OncologyPostgraduate Training Base of Jinzhou Medical University, Shanghai East HospitalShanghaiChina
- Department of Oncology, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and EngineeringEast Hospital Affiliated to Tongji University, Tongji University School of Medicine, Tongji UniversityShanghaiPeople's Republic of China
| |
Collapse
|
3
|
Ueyama A, Nogami W, Nashiki K, Haruna M, Miwa H, Hagiwara M, Nagira M, Wada H, Nagira Y. Immunotherapy Targeting CCR8+ Regulatory T Cells Induces Antitumor Effects via Dramatic Changes to the Intratumor CD8+ T Cell Profile. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:673-682. [PMID: 37350632 DOI: 10.4049/jimmunol.2300067] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/02/2023] [Indexed: 06/24/2023]
Abstract
Regulatory T cells (Tregs) contribute to the formation of a tumor-immunosuppressive microenvironment. CCR8 is reportedly selectively expressed in tumor Tregs, and an anti-CCR8 Ab can exert potent antitumor effects by eliminating intratumor Tregs in murine tumor models. In this study, we analyzed changes to intratumor immunity after anti-CCR8 Ab administration, especially in CD8+ T cells, which are involved in cancer cell killing, using the CT26 colorectal carcinoma mouse model. Immunophenotyping of tumor-infiltrating cells by mass cytometry after Ab administration on day 5 of tumor inoculation revealed that CD8+ T cell subsets were dramatically altered in the CCR8 Ab-treated group, with an increase in naive cells and nonexhausted effector cells and a decrease in exhausted cells with high expression levels of TOX. These results were corroborated with flow cytometry analysis. Delayed administration of the anti-CCR8 Ab on day 9 or 12, when the amount of CCR8+ Tregs and CD8+ T cell exhaustion were more progressed, also resulted in a decrease in exhausted CD8+ T cells, leading to tumor regression. Finally, we confirmed that high CCR8+ Treg infiltration was associated with high TOX expression in CD8+ T cells in human cancer patients. In conclusion, administration of an anti-CCR8 Ab can dramatically alter the activation and exhaustion state of intratumor CD8+ T cells, resulting in strong antitumor effects. In cancer patients with an advanced tumor-immunosuppressive environment, CD8+ T cell exhaustion has progressed along with CCR8+ Treg induction. Therefore, targeted depletion of CCR8+ Tregs is expected to be effective in these patients.
Collapse
Affiliation(s)
- Azumi Ueyama
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
- Department of Clinical Research in Tumor Immunology, Osaka University, Suita, Japan
| | - Wataru Nogami
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Kunitaka Nashiki
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Miya Haruna
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
- Department of Clinical Research in Tumor Immunology, Osaka University, Suita, Japan
| | - Hiroto Miwa
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
- Department of Clinical Research in Tumor Immunology, Osaka University, Suita, Japan
| | - Masaki Hagiwara
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Morio Nagira
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Hisashi Wada
- Department of Clinical Research in Tumor Immunology, Osaka University, Suita, Japan
| | - Yoji Nagira
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
| |
Collapse
|
4
|
Buhrmann C, Kunnumakkara AB, Kumar A, Samec M, Kubatka P, Aggarwal BB, Shakibaei M. Multitargeting Effects of Calebin A on Malignancy of CRC Cells in Multicellular Tumor Microenvironment. Front Oncol 2021; 11:650603. [PMID: 34660256 PMCID: PMC8511772 DOI: 10.3389/fonc.2021.650603] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 09/09/2021] [Indexed: 12/19/2022] Open
Abstract
Background Tumor microenvironment (TME) provides the essential prerequisite niche for promoting cancer progression and metastasis. Calebin A, a component of Curcuma longa, has long been investigated as a safe multitargeted agent with antitumor and anti-inflammatory properties. However, the multicellular-TME-induced malignancy and the antitumorigenic potential of Calebin A on colorectal cancer (CRC) cells in 3D-alginate cultures are not yet understood, and more in-depth research is needed. Methods 3D-alginate tumor cultures (HCT116 cells) in the multicellular proinflammatory TME (fibroblast cells/T lymphocytes), tumor necrosis factor beta (TNF-β)-TME (fibroblast cells/TNF-β) were treated with/without Calebin A to address the pleiotropic actions of Calebin A in the CRC. Results We found that Calebin A downmodulated proliferation, vitality, and migration of HCT116 cells in 3D-alginate cultures in multicellular proinflammatory TME or TNF-β-TME. In addition, Calebin A suppressed TNF-β-, similar to multicellular-TME-induced phosphorylation of nuclear factor kappa B (NF-κB) in a concentration-dependent manner. NF-κB-promoting proinflammatory mediators, associated with tumor growth and antiapoptotic molecules (i.e.,MMP-9, CXCR4, Ki-67, β1-integrin, and Caspase-3) and its translocation to the nucleus in HCT116 cells, were increased in both TME cultures. The multicellular-TME cultures further induced the survival of cancer stem cells (CSCs) (upregulation of CD133, CD44, and ALDH1). Last but not the least, Calebin A suppressed multicellular-, similar to TNF-β-TME-induced rigorous upregulation of NF-κB phosphorylation, various NF-κB-regulated gene products, CSCs activation, and survival in 3D-alginate tumor cultures. Conclusions The downmodulation of multicellular proinflammatory-, similar to TNF-β-TME-induced CRC proliferation, survival, and migration by the multitargeting agent Calebin A could be a new therapeutic strategy to suppress inflammation and CRC tumorigenesis.
Collapse
Affiliation(s)
- Constanze Buhrmann
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Munich, Germany.,Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory & Department of Biotechnology-National institute of Advanced Industrial Science and Technology (DBT-AIST) International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory & Department of Biotechnology-National institute of Advanced Industrial Science and Technology (DBT-AIST) International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Marek Samec
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | | | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, Munich, Germany
| |
Collapse
|
5
|
Kraus S, Kolman T, Yeung A, Deming D. Chemokine Receptor Antagonists: Role in Oncology. Curr Oncol Rep 2021; 23:131. [PMID: 34480662 DOI: 10.1007/s11912-021-01117-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW To evaluate the clinical potential of chemokine receptor antagonists for the treatment of patients with cancer. RECENT FINDINGS Chemokine receptors and their ligands can have a significant impact on the infiltration of cells into the tumor microenvironment. The receptors are increasingly being investigated as targets for the treatment of cancers. Recent studies are demonstrating the promise of chemokine receptor antagonists in this setting. There are many chemokine receptors, and each can have different functions depending on the cellular context. Targeting chemokine receptors is a promising strategy in both pre-clinical research and clinical trials. Inhibiting chemokine receptors that either recruit suppressive cells or improve cancer mobility and viability while sparing those necessary for proper immune trafficking may prove to dramatically improve treatment responses. Further research in this area is warranted and has the potential to dramatically improve patient outcomes.
Collapse
Affiliation(s)
- Sean Kraus
- Division of Hematology, Oncology and Palliative Care, Department of Medicine, University of WI-Madison, Madison, WI, USA
| | - Thomas Kolman
- Division of Hematology, Oncology and Palliative Care, Department of Medicine, University of WI-Madison, Madison, WI, USA
| | - Austin Yeung
- Division of Hematology, Oncology and Palliative Care, Department of Medicine, University of WI-Madison, Madison, WI, USA
| | - Dustin Deming
- Division of Hematology, Oncology and Palliative Care, Department of Medicine, University of WI-Madison, Madison, WI, USA. .,University of Wisconsin Carbone Cancer Center, Madison, WI, USA. .,McArdle Laboratory for Cancer Research, Department of Oncology, University of WI-Madison, Madison, WI, USA. .,6507 WI Institutes for Medical Research, 1111 Highland Ave, Madison, WI, 53705, USA.
| |
Collapse
|
6
|
Luo Y, Yin S, Lu J, Zhou S, Shao Y, Bao X, Wang T, Qiu Y, Yu H. Tumor microenvironment: a prospective target of natural alkaloids for cancer treatment. Cancer Cell Int 2021; 21:386. [PMID: 34284780 PMCID: PMC8290600 DOI: 10.1186/s12935-021-02085-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 07/08/2021] [Indexed: 12/17/2022] Open
Abstract
Malignant tumor has become one of the major diseases that seriously endangers human health. Numerous studies have demonstrated that tumor microenvironment (TME) is closely associated with patient prognosis. Tumor growth and progression are strongly dependent on its surrounding tumor microenvironment, because the optimal conditions originated from stromal elements are required for cancer cell proliferation, invasion, metastasis and drug resistance. The tumor microenvironment is an environment rich in immune/inflammatory cells and accompanied by a continuous, gradient of hypoxia and pH. Overcoming immunosuppressive environment and boosting anti-tumor immunity may be the key to the prevention and treatment of cancer. Most traditional Chinese medicine have been proved to have good anti-tumor activity, and they have the advantages of better therapeutic effect and few side effects in the treatment of malignant tumors. An increasing number of studies are giving evidence that alkaloids extracted from traditional Chinese medicine possess a significant anticancer efficiency via regulating a variety of tumor-related genes, pathways and other mechanisms. This paper reviews the anti-tumor effect of alkaloids targeting tumor microenvironment, and further reveals its anti-tumor mechanism through the effects of alkaloids on different components in tumor microenvironment.
Collapse
Affiliation(s)
- Yanming Luo
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shuangshuang Yin
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jia Lu
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shiyue Zhou
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yingying Shao
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiaomei Bao
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tao Wang
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| | - Haiyang Yu
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
7
|
Fujikawa M, Koma YI, Hosono M, Urakawa N, Tanigawa K, Shimizu M, Kodama T, Sakamoto H, Nishio M, Shigeoka M, Kakeji Y, Yokozaki H. Chemokine (C-C Motif) Ligand 1 Derived from Tumor-Associated Macrophages Contributes to Esophageal Squamous Cell Carcinoma Progression via CCR8-Mediated Akt/Proline-Rich Akt Substrate of 40 kDa/Mammalian Target of Rapamycin Pathway. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:686-703. [PMID: 33460563 DOI: 10.1016/j.ajpath.2021.01.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/10/2020] [Accepted: 01/06/2021] [Indexed: 12/26/2022]
Abstract
Tumor-associated macrophages (TAMs) promote tumor progression. The number of infiltrating TAMs is associated with poor prognosis in esophageal squamous cell carcinoma (ESCC) patients; however, the mechanism underlying this phenomenon is unclear. cDNA microarray analysis indicates that the expression of chemokine (C-C motif) ligand 1 (CCL1) is up-regulated in peripheral blood monocyte-derived macrophages stimulated using conditioned media from ESCC cells (TAM-like macrophages). Here, we evaluated the role of CCL1 in ESCC progression. CCL1 was overexpressed in TAM-like macrophages, and CCR8, a CCL1 receptor, was expressed on ESCC cell surface. TAM-like macrophages significantly enhanced the motility of ESCC cells, and neutralizing antibodies against CCL1 or CCR8 suppressed this increased motility. Recombinant human CCL1 promoted ESCC cell motility via the Akt/proline-rich Akt substrate of 40 kDa/mammalian target of rapamycin pathway. Phosphatidylinositol 3-kinase or Akt inhibitors, CCR8 silencing, and neutralizing antibody against CCR8 could significantly suppress these effects. The overexpression of CCL1 in stromal cells or CCR8 in ESCC cells was significantly associated with poor overall survival (P = 0.002 or P = 0.009, respectively) and disease-free survival (P = 0.009 or P = 0.047, respectively) in patients with ESCC. These results indicate that the interaction between stromal CCL1 and CCR8 on cancer cells promotes ESCC progression via the Akt/proline-rich Akt substrate of 40 kDa/mammalian target of rapamycin pathway, thereby providing novel therapeutic targets.
Collapse
Affiliation(s)
- Masataka Fujikawa
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yu-Ichiro Koma
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Masayoshi Hosono
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoki Urakawa
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kohei Tanigawa
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masaki Shimizu
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takayuki Kodama
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroki Sakamoto
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Mari Nishio
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Manabu Shigeoka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshihiro Kakeji
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Yokozaki
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
8
|
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
|
9
|
Resveratrol Suppresses Cross-Talk between Colorectal Cancer Cells and Stromal Cells in Multicellular Tumor Microenvironment: A Bridge between In Vitro and In Vivo Tumor Microenvironment Study. Molecules 2020; 25:molecules25184292. [PMID: 32962102 PMCID: PMC7570736 DOI: 10.3390/molecules25184292] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/08/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
The interaction between tumor cells and the tumor microenvironment (TME) is an important process for the development of tumor malignancy. Modulation of paracrine cross-talk could be a promising strategy for tumor control within the TME. The exact mechanisms of multi-targeted compound resveratrol are not yet fully understood. Whether resveratrol can modulate paracrine signal transduction-induced malignancy in the multicellular-TME of colorectal cancer cells (CRC) was investigated. An in vitro model with 3D-alginate HCT116 cells in multicellular-TME cultures (fibroblast cells, T-lymphocytes) was used to elucidate the role of TNF-β, Sirt1-ASO and/or resveratrol in the proliferation, invasion and cancer stem cells (CSC) of CRC cells. We found that multicellular-TME, similar to TNF-β-TME, promoted proliferation, colony formation, invasion of CRC cells and enabled activation of CSCs. However, after co-treatment with resveratrol, the malignancy of multicellular-TME reversed to HCT116. In addition, resveratrol reduced the secretion of T-lymphocyte/fibroblast (TNF-β, TGF-β3) proteins, antagonized the T-lymphocyte/fibroblast-promoting NF-κB activation, NF-κB nuclear translocation and thus the expression of NF-κB-promoting biomarkers, associated with proliferation, invasion and survival of CSCs in 3D-alginate cultures of HCT116 cells induced by TNF-β- or multicellular-TME, but not by Sirt1-ASO, indicating the central role of this enzyme in the anti-tumor function of resveratrol. Our results suggest that in vitro multicellular-TME promotes crosstalk between CRC and stromal cells to increase survival, migration of HCT116 and the resveratrol/Sirt1 axis suppresses this loop by modulating paracrine agent secretion and NF-κB signaling. Fibroblasts and T-lymphocytes are promising targets for resveratrol in the prevention of CRC metastasis.
Collapse
|