1
|
Deo A, Sleeman JP, Shaked Y. The role of host response to chemotherapy: resistance, metastasis and clinical implications. Clin Exp Metastasis 2024; 41:495-507. [PMID: 37999904 DOI: 10.1007/s10585-023-10243-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023]
Abstract
Chemotherapy remains the primary treatment for most metastatic cancers. However, the response to chemotherapy and targeted agents is often transient, and concurrent development of resistance is the primary impediment to effective cancer therapy. Strategies to overcome resistance to treatment have focused on cancer cell intrinsic factors and the tumor microenvironment (TME). Recent evidence indicates that systemic chemotherapy has a significant impact on the host that either facilitates tumor growth, allowing metastatic spread, or renders treatment ineffective. These host responses include the release of bone marrow-derived cells, activation of stromal cells in the TME, and induction of different molecular effectors. Here, we provide an overview of chemotherapy-induced systemic host responses that support tumor aggressiveness and metastasis, and which contribute to therapy resistance. Studying host responses to chemotherapy provides a solid basis for the development of adjuvant strategies to improve treatment outcomes and delay resistance to chemotherapy. This review discusses the emerging field of host response to cancer therapy, and its preclinical and potential clinical implications, explaining how under certain circumstances, these host effects contribute to metastasis and resistance to chemotherapy.
Collapse
Affiliation(s)
- Abhilash Deo
- Department of Cell Biology and Cancer Science, Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa, Israel
| | - Jonathan P Sleeman
- European Centre for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Karlsruhe Institute for Technology (KIT), IBCS-BIP, Campus Nord, 76344, Eggenstein- Leopoldshafen, Germany
| | - Yuval Shaked
- Department of Cell Biology and Cancer Science, Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
2
|
Verdura S, Encinar JA, Gratchev A, Llop-Hernández À, López J, Serrano-Hervás E, Teixidor E, López-Bonet E, Martin-Castillo B, Micol V, Bosch-Barrera J, Cuyàs E, Menendez JA. Silibinin is a suppressor of the metastasis-promoting transcription factor ID3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155493. [PMID: 38484626 DOI: 10.1016/j.phymed.2024.155493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/31/2024] [Accepted: 02/26/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND ID3 (inhibitor of DNA binding/differentiation-3) is a transcription factor that enables metastasis by promoting stem cell-like properties in endothelial and tumor cells. The milk thistle flavonolignan silibinin is a phytochemical with anti-metastatic potential through largely unknown mechanisms. HYPOTHESIS/PURPOSE We have mechanistically investigated the ability of silibinin to inhibit the aberrant activation of ID3 in brain endothelium and non-small cell lung cancer (NSCLC) models. METHODS Bioinformatic analyses were performed to investigate the co-expression correlation between ID3 and bone morphogenic protein (BMP) ligands/BMP receptors (BMPRs) genes in NSCLC patient datasets. ID3 expression was assessed by immunoblotting and qRT-PCR. Luciferase reporter assays were used to evaluate the gene sequences targeted by silibinin to regulate ID3 transcription. In silico computational modeling and LanthaScreen TR-FRET kinase assays were used to characterize and validate the BMPR inhibitory activity of silibinin. Tumor tissues from NSCLC xenograft models treated with oral silibinin were used to evaluate the in vivo anti-ID3 effects of silibinin. RESULTS Analysis of lung cancer patient datasets revealed a top-ranked positive association of ID3 with the BMP9 endothelial receptor ACVRL1/ALK1 and the BMP ligand BMP6. Silibinin treatment blocked the BMP9-induced activation of the ALK1-phospho-SMAD1/5-ID3 axis in brain endothelial cells. Constitutive, acquired, and adaptive expression of ID3 in NSCLC cells were all significantly downregulated in response to silibinin. Silibinin blocked ID3 transcription via BMP-responsive elements in ID3 gene enhancers. Silibinin inhibited the kinase activities of BMPRs in the micromolar range, with the lower IC50 values occurring against ACVRL1/ALK1 and BMPR2. In an in vivo NSCLC xenograft model, tumoral overexpression of ID3 was completely suppressed by systematically achievable oral doses of silibinin. CONCLUSIONS ID3 is a largely undruggable metastasis-promoting transcription factor. Silibinin is a novel suppressor of ID3 that may be explored as a novel therapeutic approach to interfere with the metastatic dissemination capacity of NSCLC.
Collapse
Affiliation(s)
- Sara Verdura
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - José Antonio Encinar
- Institute of Research, Development and Innovation in Health Biotechnology of Elche (IDiBE), Universitas Miguel Hernández (UMH), Elche 03202, Spain
| | - Alexei Gratchev
- Laboratory for Tumor Stromal Cell Biology, Institute of Carcinogenesis, Nikolaj Nikolajevich (N.N.) Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia
| | - Àngela Llop-Hernández
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - Júlia López
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - Eila Serrano-Hervás
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - Eduard Teixidor
- Precision Oncology Group (OncoGir-Pro), Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain; Medical Oncology, Catalan Institute of Oncology, Girona, 17007, Spain
| | - Eugeni López-Bonet
- Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain; Department of Anatomical Pathology, Dr. Josep Trueta Hospital of Girona, Girona 17007, Spain
| | - Begoña Martin-Castillo
- Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain; Unit of Clinical Research, Catalan Institute of Oncology, Girona, 17007, Spain
| | - Vicente Micol
- Institute of Research, Development and Innovation in Health Biotechnology of Elche (IDiBE), Universitas Miguel Hernández (UMH), Elche 03202, Spain; CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, 28029, Spain
| | - Joaquim Bosch-Barrera
- Precision Oncology Group (OncoGir-Pro), Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain; Medical Oncology, Catalan Institute of Oncology, Girona, 17007, Spain; Department of Medical Sciences, Medical School, University of Girona, Girona, Spain
| | - Elisabet Cuyàs
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - Javier A Menendez
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain.
| |
Collapse
|
3
|
Yao J, Chen Y, Huang Y, Sun X, Shi X. The role of cardiac microenvironment in cardiovascular diseases: implications for therapy. Hum Cell 2024; 37:607-624. [PMID: 38498133 DOI: 10.1007/s13577-024-01052-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/25/2024] [Indexed: 03/20/2024]
Abstract
Due to aging populations and changes in lifestyle, cardiovascular diseases including cardiomyopathy, hypertension, and atherosclerosis, are the leading causes of death worldwide. The heart is a complicated organ composed of multicellular types, including cardiomyocytes, fibroblasts, endothelial cells, vascular smooth muscle cells, and immune cells. Cellular specialization and complex interplay between different cell types are crucial for the cardiac tissue homeostasis and coordinated function of the heart. Mounting studies have demonstrated that dysfunctional cells and disordered cardiac microenvironment are closely associated with the pathogenesis of various cardiovascular diseases. In this paper, we discuss the composition and the homeostasis of cardiac tissues, and focus on the role of cardiac environment and underlying molecular mechanisms in various cardiovascular diseases. Besides, we elucidate the novel treatment for cardiovascular diseases, including stem cell therapy and targeted therapy. Clarification of these issues may provide novel insights into the prevention and potential targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Jiayu Yao
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuejun Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuqing Huang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| |
Collapse
|
4
|
Deng T, Zhao J, Tong Y, Chen Z, He B, Li J, Chen B, Li R, Deng L, Yu H, Zhang B, Zhang T, Shi Z, Gao B, Jiang J, Shan Y, Yu Z, Jin Y, Wang Y, Xia J, Chen G. Crosstalk between endothelial progenitor cells and HCC through periostin/CCL2/CD36 supports formation of the pro-metastatic microenvironment in HCC. Oncogene 2024; 43:944-961. [PMID: 38351345 DOI: 10.1038/s41388-024-02960-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/19/2024]
Abstract
Metastasis causes most cancer-related deaths, and the role and mechanism of periostin (POSTN) in the metastasis of hepatocellular carcinoma (HCC) remain undiscovered. In this study, DEN and HTVi HCC models were performed in hepatic-specific Postn ablation and Postn knock-in mouse to reveal the role of POSTN in HCC metastasis. Furthermore, POSTN was positively correlated with circulating EPCs level and promoted EPC mobilization and tumour infiltration. POSTN also mediated the crosstalk between HCC and EPCs, which promoted metastasis ability and upregulated CD36 expression in HCC through indirect crosstalk. Chemokine arrays further revealed that hepatic-derived POSTN induced elevated CCL2 expression and secretion in EPCs, and CCL2 promoted prometastatic traits in HCC. Mechanistic studies showed that POSTN upregulated CCL2 expression in EPCs via the αvβ3/ILK/NF-κB pathway. CCL2 further induced CD36 expression via the CCR2/STAT3 pathway by directly binding to the promoter region of CD36. Finally, CD36 was verified to have a prometastatic role in vitro and to be correlated with POSTN expression, metastasis and recurrence in HCC in clinical samples. Our findings revealed that crosstalk between HCC and EPCs is mediated by periostin/CCL2/CD36 signalling which promotes HCC metastasis and emphasizes a potential therapeutic strategy for preventing HCC metastasis.
Collapse
Affiliation(s)
- Tuo Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jungang Zhao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yifan Tong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Ziyan Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Bangjie He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jiacheng Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Bo Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Rizhao Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Liming Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- The Second Affiliated Hospital, Department of General Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Haitao Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Baofu Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Tan Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zhehao Shi
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Boyang Gao
- Alberta Institute, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junyan Jiang
- Alberta Institute, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yunfeng Shan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zhengping Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yuepeng Jin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Yi Wang
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Jinglin Xia
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Liver Cancer Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325005, China.
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Liver Cancer Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325005, China.
| |
Collapse
|
5
|
Yu L, Fan G, Wang Q, Zhu Y, Zhu H, Chang J, Wang Z, Zhan S, Hua X, She D, Huang J, Wang Y, Zhao J, Zhang CY, Chen X, Zhou G. In vivo self-assembly and delivery of VEGFR2 siRNA-encapsulated small extracellular vesicles for lung metastatic osteosarcoma therapy. Cell Death Dis 2023; 14:626. [PMID: 37739958 PMCID: PMC10516902 DOI: 10.1038/s41419-023-06159-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/02/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
The prognosis of lung metastatic osteosarcoma (OS) remains disappointing. siRNA-based gene silencing of VEGFR2 is a promising treatment strategy for lung metastatic OS, but there is a lack of safe and efficient delivery systems to encapsulate siRNAs for in vivo administration. This study presented a synthetic biological strategy that remolds the host liver with synthesized genetic circuits for efficient in vivo VEGFR2 siRNA delivery. After being taken-up by hepatocytes, the genetic circuit (in the form of a DNA plasmid) reprogrammed the liver to drive the autonomous intrahepatic assembly and encapsulation of VEGFR2 siRNAs into secretory small extracellular vesicles (sEVs), thus allowing for the transport of self-assembled VEGFR2 siRNAs towards the lung. The results showed that our strategy was superior to the positive medicine (Apatinib) for OS lung metastasis in terms of therapeutic efficacy and toxic adverse effects and may provide a feasible and viable therapeutic solution for lung metastatic OS.
Collapse
Affiliation(s)
- Lingfeng Yu
- Department of Orthopedics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Gentao Fan
- Department of Orthopedics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Qingyan Wang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Yan Zhu
- Department of Orthopedics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Hao Zhu
- Department of Orthopedics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Jiang Chang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, 210029, China
| | - Zhen Wang
- Department of Orthopedics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Shoubin Zhan
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Xianming Hua
- Department of Orthopedics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Diankun She
- Department of Orthopedics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Jianhao Huang
- Department of Orthopedics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Yicun Wang
- Department of Orthopedics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Jianning Zhao
- Department of Orthopedics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Chen-Yu Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210023, China.
| | - Xi Chen
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210023, China.
| | - Guangxin Zhou
- Department of Orthopedics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210002, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, 210023, China.
- Wuxi Xishan NJU Institute of Applied Biotechnology, Wuxi, Jiangsu, 214101, China.
| |
Collapse
|
6
|
Dudley AC, Griffioen AW. Pathological angiogenesis: mechanisms and therapeutic strategies. Angiogenesis 2023; 26:313-347. [PMID: 37060495 PMCID: PMC10105163 DOI: 10.1007/s10456-023-09876-7] [Citation(s) in RCA: 126] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/26/2023] [Indexed: 04/16/2023]
Abstract
In multicellular organisms, angiogenesis, the formation of new blood vessels from pre-existing ones, is an essential process for growth and development. Different mechanisms such as vasculogenesis, sprouting, intussusceptive, and coalescent angiogenesis, as well as vessel co-option, vasculogenic mimicry and lymphangiogenesis, underlie the formation of new vasculature. In many pathological conditions, such as cancer, atherosclerosis, arthritis, psoriasis, endometriosis, obesity and SARS-CoV-2(COVID-19), developmental angiogenic processes are recapitulated, but are often done so without the normal feedback mechanisms that regulate the ordinary spatial and temporal patterns of blood vessel formation. Thus, pathological angiogenesis presents new challenges yet new opportunities for the design of vascular-directed therapies. Here, we provide an overview of recent insights into blood vessel development and highlight novel therapeutic strategies that promote or inhibit the process of angiogenesis to stabilize, reverse, or even halt disease progression. In our review, we will also explore several additional aspects (the angiogenic switch, hypoxia, angiocrine signals, endothelial plasticity, vessel normalization, and endothelial cell anergy) that operate in parallel to canonical angiogenesis mechanisms and speculate how these processes may also be targeted with anti-angiogenic or vascular-directed therapies.
Collapse
Affiliation(s)
- Andrew C Dudley
- Department of Microbiology, Immunology and Cancer Biology, The University of Virginia, Charlottesville, VA, 22908, USA.
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Wu YN, Su X, Wang XQ, Liu NN, Xu ZW. The roles of phospholipase C-β related signals in the proliferation, metastasis and angiogenesis of malignant tumors, and the corresponding protective measures. Front Oncol 2023; 13:1231875. [PMID: 37576896 PMCID: PMC10419273 DOI: 10.3389/fonc.2023.1231875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
PLC-β is widely distributed in eukaryotic cells and is the key enzyme in phosphatidylinositol signal transduction pathway. The cellular functions regulated by its four subtypes (PLC-β1, PLC-β2, PLC-β3, PLC-β4) play an important role in maintaining homeostasis of organism. PLC-β and its related signals can promote or inhibit the occurrence and development of cancer by affecting the growth, differentiation and metastasis of cells, while targeted intervention of PLC-β1-PI3K-AKT, PLC-β2/CD133, CXCR2-NHERF1-PLC-β3, Gαq-PLC-β4-PKC-MAPK and so on can provide new strategies for the precise prevention and treatment of malignant tumors. This paper reviews the mechanism of PLC-β in various tumor cells from four aspects: proliferation and differentiation, invasion and metastasis, angiogenesis and protective measures.
Collapse
Affiliation(s)
- Yu-Nuo Wu
- Department of Clinical Medical, the First Clinical Medical College of Anhui Medical University, Hefei, Anhui, China
| | - Xing Su
- Department of Clinical Medical, the First Clinical Medical College of Anhui Medical University, Hefei, Anhui, China
| | - Xue-Qin Wang
- Department of Clinical Medical, the First Clinical Medical College of Anhui Medical University, Hefei, Anhui, China
| | - Na-Na Liu
- Department of Emergency Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhou-Wei Xu
- Department of Emergency Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui, China
| |
Collapse
|
8
|
Yoshimura T, Li C, Wang Y, Matsukawa A. The chemokine monocyte chemoattractant protein-1/CCL2 is a promoter of breast cancer metastasis. Cell Mol Immunol 2023:10.1038/s41423-023-01013-0. [PMID: 37208442 DOI: 10.1038/s41423-023-01013-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/17/2023] [Indexed: 05/21/2023] Open
Abstract
Breast cancer is the most prevalent cancer worldwide, and metastasis is the leading cause of death in cancer patients. Human monocyte chemoattractant protein-1 (MCP-1/CCL2) was isolated from the culture supernatants of not only mitogen-activated peripheral blood mononuclear leukocytes but also malignant glioma cells based on its in vitro chemotactic activity toward human monocytes. MCP-1 was subsequently found to be identical to a previously described tumor cell-derived chemotactic factor thought to be responsible for the accumulation of tumor-associated macrophages (TAMs), and it became a candidate target of clinical intervention; however, the role of TAMs in cancer development was still controversial at the time of the discovery of MCP-1. The in vivo role of MCP-1 in cancer progression was first evaluated by examining human cancer tissues, including breast cancers. Positive correlations between the level of MCP-1 production in tumors and the degree of TAM infiltration and cancer progression were established. The contribution of MCP-1 to the growth of primary tumors and metastasis to the lung, bone, and brain was examined in mouse breast cancer models. The results of these studies strongly suggested that MCP-1 is a promoter of breast cancer metastasis to the lung and brain but not bone. Potential mechanisms of MCP-1 production in the breast cancer microenvironment have also been reported. In the present manuscript, we review studies in which the role of MCP-1 in breast cancer development and progression and the mechanisms of its production were examined and attempt to draw a consensus and discuss the potential use of MCP-1 as a biomarker for diagnosis.
Collapse
Affiliation(s)
- Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan.
| | - Chunning Li
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Yuze Wang
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Akihiro Matsukawa
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| |
Collapse
|
9
|
Wälchli T, Bisschop J, Carmeliet P, Zadeh G, Monnier PP, De Bock K, Radovanovic I. Shaping the brain vasculature in development and disease in the single-cell era. Nat Rev Neurosci 2023; 24:271-298. [PMID: 36941369 PMCID: PMC10026800 DOI: 10.1038/s41583-023-00684-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 03/23/2023]
Abstract
The CNS critically relies on the formation and proper function of its vasculature during development, adult homeostasis and disease. Angiogenesis - the formation of new blood vessels - is highly active during brain development, enters almost complete quiescence in the healthy adult brain and is reactivated in vascular-dependent brain pathologies such as brain vascular malformations and brain tumours. Despite major advances in the understanding of the cellular and molecular mechanisms driving angiogenesis in peripheral tissues, developmental signalling pathways orchestrating angiogenic processes in the healthy and the diseased CNS remain incompletely understood. Molecular signalling pathways of the 'neurovascular link' defining common mechanisms of nerve and vessel wiring have emerged as crucial regulators of peripheral vascular growth, but their relevance for angiogenesis in brain development and disease remains largely unexplored. Here we review the current knowledge of general and CNS-specific mechanisms of angiogenesis during brain development and in brain vascular malformations and brain tumours, including how key molecular signalling pathways are reactivated in vascular-dependent diseases. We also discuss how these topics can be studied in the single-cell multi-omics era.
Collapse
Affiliation(s)
- Thomas Wälchli
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland.
- Group of Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada.
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada.
| | - Jeroen Bisschop
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Group of Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB & Department of Oncology, KU Leuven, Leuven, Belgium
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gelareh Zadeh
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Philippe P Monnier
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Donald K. Johnson Research Institute, Krembil Research Institute, Krembil Discovery Tower, Toronto, ON, Canada
- Department of Ophthalmology and Vision Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Science and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Ivan Radovanovic
- Group of Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
10
|
Pontis F, Roz L, Fortunato O, Bertolini G. The metastatic niche formation: focus on extracellular vesicle-mediated dialogue between lung cancer cells and the microenvironment. Front Oncol 2023; 13:1116783. [PMID: 37207158 PMCID: PMC10189117 DOI: 10.3389/fonc.2023.1116783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/21/2023] [Indexed: 05/21/2023] Open
Abstract
Lung cancer is the deadliest cancer in the world, with the majority of patients presenting with advanced or metastatic disease at first diagnosis. The lungs are also one of the most common sites of metastasis from lung cancer and other tumors. Understanding the mechanisms that regulate metastasis formation from primary lung cancer and in the lungs is therefore fundamental unmet clinical need. One of the first steps during the establishment of lung cancer metastases includes the formation of the pre-metastatic niche (PMN) at distant organs, which may occur even during the early phases of cancer development. The PMN is established through intricate cross-talk between primary tumor-secreted factors and stromal components at distant sites. Mechanisms controlling primary tumor escape and seeding of distant organs rely on specific properties of tumor cells but are also tightly regulated by interactions with stromal cells at the metastatic niche that finally dictate the success of metastasis establishment. Here, we summarize the mechanisms underlying pre-metastatic niche formation starting from how lung primary tumor cells modulate distant sites through the release of several factors, focusing on Extracellular Vesicles (EVs). In this context, we highlight the role of lung cancer-derived EVs in the modulation of tumor immune escape. Then, we illustrate the complexity of Circulating Tumor Cells (CTCs) that represent the seeds of metastasis and how interactions with stromal and immune cells can help their metastatic dissemination. Finally, we evaluate the contribution of EVs in dictating metastasis development at the PMN through stimulation of proliferation and control of disseminated tumor cell dormancy. Overall, we present an overview of different steps in the lung cancer metastatic cascade, focusing on the EV-mediated interactions between tumor cells and stromal/immune cells.
Collapse
|
11
|
PRSS2 remodels the tumor microenvironment via repression of Tsp1 to stimulate tumor growth and progression. Nat Commun 2022; 13:7959. [PMID: 36575174 PMCID: PMC9794699 DOI: 10.1038/s41467-022-35649-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
The progression of cancer from localized to metastatic disease is the primary cause of morbidity and mortality. The interplay between the tumor and its microenvironment is the key driver in this process of tumor progression. In order for tumors to progress and metastasize they must reprogram the cells that make up the microenvironment to promote tumor growth and suppress endogenous defense systems, such as the immune and inflammatory response. We have previously demonstrated that stimulation of Tsp-1 in the tumor microenvironment (TME) potently inhibits tumor growth and progression. Here, we identify a novel tumor-mediated mechanism that represses the expression of Tsp-1 in the TME via secretion of the serine protease PRSS2. We demonstrate that PRSS2 represses Tsp-1, not via its enzymatic activity, but by binding to low-density lipoprotein receptor-related protein 1 (LRP1). These findings describe a hitherto undescribed activity for PRSS2 through binding to LRP1 and represent a potential therapeutic strategy to treat cancer by blocking the PRSS2-mediated repression of Tsp-1. Based on the ability of PRSS2 to reprogram the tumor microenvironment, this discovery could lead to the development of therapeutic agents that are indication agnostic.
Collapse
|
12
|
Thurgur H, Penny J, Pinteaux E. Endothelial cell activation by interleukin-1 and extracellular matrix laminin-10 occurs via the YAP signalling pathway. J Neuroimmunol 2022; 373:577993. [PMID: 36327619 DOI: 10.1016/j.jneuroim.2022.577993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/29/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
Abstract
Laminin-10 (LM-10) is a key regulator of blood-brain barrier (BBB) repair after hypoxia and inflammation. Here we investigated the signalling mechanisms regulated by LM-10 in human brain endothelial cell line hCMEC/D3 in response to interleukin(IL)-1beta(β) in vitro. LM-10 promoted endothelial proliferation and repair of an endothelial monolayer after scratch injury, and upregulated IL-1β-induced ICAM-1 and VCAM-1 expression. IL-1β and LM-10 regulated YAP signalling pathway in endothelial cells leading to differential expression of YAP target genes, ctgf and serpine-1, providing evidence that the YAP signalling pathway could be a new therapeutic target for the treatment of BBB dysfunction in CNS diseases.
Collapse
Affiliation(s)
- Hannah Thurgur
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, Manchester, United Kingdom; Division of Neuroscience, Faculty of Biology, Medicine and Health; A.V. Hill Building, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Stopford Building, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Emmanuel Pinteaux
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, Manchester, United Kingdom; Division of Neuroscience, Faculty of Biology, Medicine and Health; A.V. Hill Building, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom.
| |
Collapse
|
13
|
Transient Receptor Potential (TRP) Channels in Tumor Vascularization. Int J Mol Sci 2022; 23:ijms232214253. [PMID: 36430727 PMCID: PMC9692925 DOI: 10.3390/ijms232214253] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Tumor diseases are unfortunately quick spreading, even though numerous studies are under way to improve early diagnosis and targeted treatments that take into account both the different characteristics associated with the various tumor types and the conditions of individual patients. In recent years, studies have focused on the role of ion channels in tumor development, as these proteins are involved in several cellular processes relevant to neoplastic transformation. Among all ion channels, many studies have focused on the superfamily of Transient Receptor Potential (TRP) channels, which are non-selective cation channels mediating extracellular Ca2+ influx. In this review, we examined the role of different endothelial TRP channel isoforms in tumor vessel formation, a process that is essential in tumor growth and metastasis.
Collapse
|
14
|
Chen K, Yong J, Zauner R, Wally V, Whitelock J, Sajinovic M, Kopecki Z, Liang K, Scott KF, Mellick AS. Chondroitin Sulfate Proteoglycan 4 as a Marker for Aggressive Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:5564. [PMID: 36428658 PMCID: PMC9688099 DOI: 10.3390/cancers14225564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/27/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Chondroitin sulfate (CS) proteoglycan 4 (CSPG4) is a cell surface proteoglycan that is currently under investigation as a marker of cancer malignancy, and as a potential target of anticancer drug treatment. CSPG4 acts as a driver of tumourigenesis by regulating turnover of the extracellular matrix (ECM) to promote tumour cell invasion, migration as well as inflammation and angiogenesis. While CSPG4 has been widely studied in certain malignancies, such as melanoma, evidence is emerging from global gene expression studies, which suggests a role for CSPG4 in squamous cell carcinoma (SCC). While relatively treatable, lack of widely agreed upon diagnostic markers for SCCs is problematic, especially for clinicians managing certain patients, including those who are aged or infirm, as well as those with underlying conditions such as epidermolysis bullosa (EB), for which a delayed diagnosis is likely lethal. In this review, we have discussed the structure of CSPG4, and quantitatively analysed CSPG4 expression in the tissues and pathologies where it has been identified to determine the usefulness of CSPG4 expression as a diagnostic marker and therapeutic target in management of malignant SCC.
Collapse
Affiliation(s)
- Kathryn Chen
- Ingham Institute for Applied Medical Research, Medicine, University of New South Wales, Liverpool, NSW 2170, Australia
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Joel Yong
- Ingham Institute for Applied Medical Research, Medicine, University of New South Wales, Liverpool, NSW 2170, Australia
- School of Chemical Engineering, University of New South Wales, Kensington, NSW 2033, Australia
| | - Roland Zauner
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology & Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Verena Wally
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology & Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - John Whitelock
- Ingham Institute for Applied Medical Research, Medicine, University of New South Wales, Liverpool, NSW 2170, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Kensington, NSW 2033, Australia
| | - Mila Sajinovic
- Ingham Institute for Applied Medical Research, Medicine, University of New South Wales, Liverpool, NSW 2170, Australia
| | - Zlatko Kopecki
- Future Industries Institute, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Kang Liang
- Ingham Institute for Applied Medical Research, Medicine, University of New South Wales, Liverpool, NSW 2170, Australia
- School of Chemical Engineering, University of New South Wales, Kensington, NSW 2033, Australia
| | - Kieran Francis Scott
- Ingham Institute for Applied Medical Research, Medicine, University of New South Wales, Liverpool, NSW 2170, Australia
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Albert Sleiman Mellick
- Ingham Institute for Applied Medical Research, Medicine, University of New South Wales, Liverpool, NSW 2170, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Kensington, NSW 2033, Australia
| |
Collapse
|
15
|
Lucotti S, Kenific CM, Zhang H, Lyden D. Extracellular vesicles and particles impact the systemic landscape of cancer. EMBO J 2022; 41:e109288. [PMID: 36052513 PMCID: PMC9475536 DOI: 10.15252/embj.2021109288] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/16/2022] [Accepted: 03/23/2022] [Indexed: 11/09/2022] Open
Abstract
Intercellular cross talk between cancer cells and stromal and immune cells is essential for tumor progression and metastasis. Extracellular vesicles and particles (EVPs) are a heterogeneous class of secreted messengers that carry bioactive molecules and that have been shown to be crucial for this cell-cell communication. Here, we highlight the multifaceted roles of EVPs in cancer. Functionally, transfer of EVP cargo between cells influences tumor cell growth and invasion, alters immune cell composition and function, and contributes to stromal cell activation. These EVP-mediated changes impact local tumor progression, foster cultivation of pre-metastatic niches at distant organ-specific sites, and mediate systemic effects of cancer. Furthermore, we discuss how exploiting the highly selective enrichment of molecules within EVPs has profound implications for advancing diagnostic and prognostic biomarker development and for improving therapy delivery in cancer patients. Altogether, these investigations into the role of EVPs in cancer have led to discoveries that hold great promise for improving cancer patient care and outcome.
Collapse
Affiliation(s)
- Serena Lucotti
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - Candia M Kenific
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - Haiying Zhang
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - David Lyden
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| |
Collapse
|
16
|
Jubelin C, Muñoz-Garcia J, Griscom L, Cochonneau D, Ollivier E, Heymann MF, Vallette FM, Oliver L, Heymann D. Three-dimensional in vitro culture models in oncology research. Cell Biosci 2022; 12:155. [PMID: 36089610 PMCID: PMC9465969 DOI: 10.1186/s13578-022-00887-3] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 08/18/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractCancer is a multifactorial disease that is responsible for 10 million deaths per year. The intra- and inter-heterogeneity of malignant tumors make it difficult to develop single targeted approaches. Similarly, their diversity requires various models to investigate the mechanisms involved in cancer initiation, progression, drug resistance and recurrence. Of the in vitro cell-based models, monolayer adherent (also known as 2D culture) cell cultures have been used for the longest time. However, it appears that they are often less appropriate than the three-dimensional (3D) cell culture approach for mimicking the biological behavior of tumor cells, in particular the mechanisms leading to therapeutic escape and drug resistance. Multicellular tumor spheroids are widely used to study cancers in 3D, and can be generated by a multiplicity of techniques, such as liquid-based and scaffold-based 3D cultures, microfluidics and bioprinting. Organoids are more complex 3D models than multicellular tumor spheroids because they are generated from stem cells isolated from patients and are considered as powerful tools to reproduce the disease development in vitro. The present review provides an overview of the various 3D culture models that have been set up to study cancer development and drug response. The advantages of 3D models compared to 2D cell cultures, the limitations, and the fields of application of these models and their techniques of production are also discussed.
Collapse
|
17
|
Arsenic Nanoparticles are Effective in Reducing 3-Methylcholanthrene Induced Carcinogenesis in Murine Fibrosarcoma by Promoting Anti-tumorigenic Inflammation. BIONANOSCIENCE 2022. [DOI: 10.1007/s12668-021-00920-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
18
|
Qiu J, Li Y, Wang B, Sun X, Qian D, Ying Y, Zhou J. The Role and Research Progress of Inhibitor of Differentiation 1 in Atherosclerosis. DNA Cell Biol 2022; 41:71-79. [PMID: 35049366 PMCID: PMC8863915 DOI: 10.1089/dna.2021.0745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 12/23/2022] Open
Abstract
Inhibitor of differentiation 1 has a helix-loop-helix (HLH) structure, belongs to a class of molecules known as the HLH trans-acting factor family, and plays an important role in advancing the cell cycle, promoting cell proliferation and inhibiting cell differentiation. Recent studies have confirmed that inhibitor of differentiation 1 plays an important role in the endothelial-mesenchymal transition of vascular endothelial cells, angiogenesis, reendothelialization after injury, and the formation and rupture of atherosclerotic plaques. An in-depth understanding of the role of inhibitor of differentiation 1 in atherosclerosis will provide new ideas and strategies for the treatment of related diseases.
Collapse
Affiliation(s)
- Jun Qiu
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Cardiology, Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo, China
| | - Youhong Li
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
| | - BingYu Wang
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Cardiology, Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo, China
| | - XinYi Sun
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Cardiology, Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo, China
| | - Dingding Qian
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Yuchen Ying
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Jianqing Zhou
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| |
Collapse
|
19
|
Parker KA, Robinson NJ, Schiemann WP. The role of RNA processing and regulation in metastatic dormancy. Semin Cancer Biol 2022; 78:23-34. [PMID: 33775829 PMCID: PMC8464634 DOI: 10.1016/j.semcancer.2021.03.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023]
Abstract
Tumor dormancy is a major contributor to the lethality of metastatic disease, especially for cancer patients who develop metastases years-to-decades after initial diagnosis. Indeed, tumor cells can disseminate during early disease stages and persist in new microenvironments at distal sites for months, years, or even decades before initiating metastatic outgrowth. This delay between primary tumor remission and metastatic relapse is known as "dormancy," during which disseminated tumor cells (DTCs) acquire quiescent states in response to intrinsic (i.e., cellular) and extrinsic (i.e., microenvironmental) signals. Maintaining dormancy-associated phenotypes requires DTCs to activate transcriptional, translational, and post-translational mechanisms that engender cellular plasticity. RNA processing is emerging as an essential facet of cellular plasticity, particularly with respect to the initiation, maintenance, and reversal of dormancy-associated phenotypes. Moreover, dysregulated RNA processing, particularly that associated with alternative RNA splicing and expression of noncoding RNAs (ncRNAs), can occur in DTCs to mediate intrinsic and extrinsic metastatic dormancy. Here we review the pathophysiological impact of alternative RNA splicing and ncRNAs in promoting metastatic dormancy and disease recurrence in human cancers.
Collapse
Affiliation(s)
- Kimberly A. Parker
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nathaniel J. Robinson
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - William P. Schiemann
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA,Corresponding Author: William P. Schiemann, Case Comprehensive Cancer Center, Case Western Reserve University, Wolstein Research Building, 2103 Cornell Road, Cleveland, OH 44106 Phone: 216-368-5763.
| |
Collapse
|
20
|
Lüönd F, Sugiyama N, Bill R, Bornes L, Hager C, Tang F, Santacroce N, Beisel C, Ivanek R, Bürglin T, Tiede S, van Rheenen J, Christofori G. Distinct contributions of partial and full EMT to breast cancer malignancy. Dev Cell 2021; 56:3203-3221.e11. [PMID: 34847378 DOI: 10.1016/j.devcel.2021.11.006] [Citation(s) in RCA: 200] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 09/13/2021] [Accepted: 11/05/2021] [Indexed: 12/13/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a transient, reversible process of cell de-differentiation where cancer cells transit between various stages of an EMT continuum, including epithelial, partial EMT, and mesenchymal cell states. We have employed Tamoxifen-inducible dual recombinase lineage tracing systems combined with live imaging and 5-cell RNA sequencing to track cancer cells undergoing partial or full EMT in the MMTV-PyMT mouse model of metastatic breast cancer. In primary tumors, cancer cells infrequently undergo EMT and mostly transition between epithelial and partial EMT states but rarely reach full EMT. Cells undergoing partial EMT contribute to lung metastasis and chemoresistance, whereas full EMT cells mostly retain a mesenchymal phenotype and fail to colonize the lungs. However, full EMT cancer cells are enriched in recurrent tumors upon chemotherapy. Hence, cancer cells in various stages of the EMT continuum differentially contribute to hallmarks of breast cancer malignancy, such as tumor invasion, metastasis, and chemoresistance.
Collapse
Affiliation(s)
- Fabiana Lüönd
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Nami Sugiyama
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland.
| | - Ruben Bill
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Laura Bornes
- Division of Molecular Pathology, Oncode Institute, Netherlands Cancer Institute, 1006 BE Amsterdam, the Netherlands
| | - Carolina Hager
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Fengyuan Tang
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Natascha Santacroce
- Department of Biosystems Science and Engineering, ETH Zürich, 4058 Basel, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zürich, 4058 Basel, Switzerland
| | - Robert Ivanek
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Thomas Bürglin
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Stefanie Tiede
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Jacco van Rheenen
- Division of Molecular Pathology, Oncode Institute, Netherlands Cancer Institute, 1006 BE Amsterdam, the Netherlands
| | | |
Collapse
|
21
|
García-Jiménez I, Cervantes-Villagrana RD, Del-Río-Robles JE, Castillo-Kauil A, Beltrán-Navarro YM, García-Román J, Reyes-Cruz G, Vázquez-Prado J. Gβγ mediates activation of Rho guanine nucleotide exchange factor ARHGEF17 that promotes metastatic lung cancer progression. J Biol Chem 2021; 298:101440. [PMID: 34808208 PMCID: PMC8703085 DOI: 10.1016/j.jbc.2021.101440] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 11/10/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022] Open
Abstract
Metastatic lung cancer is a major cause of death worldwide. Dissemination of cancer cells can be facilitated by various agonists within the tumor microenvironment, including by lysophosphatidic acid (LPA). We postulate that Rho guanine nucleotide exchange factors (RhoGEFs), which integrate signaling cues driving cell migration, are critical effectors in metastatic cancer. Specifically, we addressed the hypothetical role of ARHGEF17, a RhoGEF, as a potential effector of Gβγ in metastatic lung cancer cells responding to LPA. Here, we show that ARHGEF17, originally identified as a tumor endothelial marker, is involved in tumor growth and metastatic dissemination of lung cancer cells in an immunocompetent murine model. Gene expression–based analysis of lung cancer datasets showed that increased levels of ARHGEF17 correlated with reduced survival of patients with advanced-stage tumors. Cellular assays also revealed that this RhoGEF participates in the invasive and migratory responses elicited by Gi protein–coupled LPA receptors via the Gβγ subunit complex. We demonstrate that this signaling heterodimer promoted ARHGEF17 recruitment to the cell periphery and actin fibers. Moreover, Gβγ allosterically activates ARHGEF17 by the removal of inhibitory intramolecular restrictions. Taken together, our results indicate that ARHGEF17 may be a valid potential target in the treatment of metastatic lung cancer.
Collapse
|
22
|
Lee S, Wang SW, Yu CL, Tai HC, Yen JY, Tuan YL, Wang HH, Liu YT, Chen SS, Lee HY. Effect of phenylurea hydroxamic acids on histone deacetylase and VEGFR-2. Bioorg Med Chem 2021; 50:116454. [PMID: 34634618 DOI: 10.1016/j.bmc.2021.116454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/23/2021] [Accepted: 09/25/2021] [Indexed: 01/23/2023]
Abstract
A series of phenylurea hydroxamic acids incorporating pharmacophores of inhibitors of HDAC inhibitors and VEGFR-2 has been designed. Most of the compounds show antiproliferative activity comparable to that of Vorinostat and Sorafenib, and better EPC inhibitory activity. Enzymatic assays and Western blotting results indicated that compound 14 not only inhibits HDAC but also has slight VEGFR-2 inhibitory activity. A docking study revealed that the polar hydroxamic acid retains the interaction with HDAC through a zinc ion and also interacts with some residues of the active site of VEGFR-2. Despite 14 displaying a weaker VEGFR-2 activity, a possible route to develop potent HDAC/VEGFR-2 inhibitors is suggested.
Collapse
Affiliation(s)
- Szu Lee
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Shih-Wei Wang
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, Taiwan; Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chen-Lin Yu
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Huai-Ching Tai
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan; Department of Urology, Fu-Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Juei-Yu Yen
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Yu-Lien Tuan
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Hsueh-Hsiao Wang
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Yi-Ting Liu
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Shiou-Sheng Chen
- Division of Urology, Taipei City Hospital Zhong Xiao Branch, Taipei, Taiwan; Commission for General Education, National Taiwan University of Science and Technology, Taipei, Taiwan; Department of Urology, College of Medicine and Shu-Tien Urological Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; General Education Center, University of Taipei, Taipei, Taiwan.
| | - Hsueh-Yun Lee
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan; Master Program in Clinical Pharmacogenomics and Pharmacoproteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
23
|
Discovery of novel ID2 antagonists from pharmacophore-based virtual screening as potential therapeutics for glioma. Bioorg Med Chem 2021; 49:116427. [PMID: 34600240 DOI: 10.1016/j.bmc.2021.116427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/29/2022]
Abstract
Glioma, especially the most aggressive type glioblastoma multiforme, is a malignant cancer of the central nervous system with a poor prognosis. Traditional treatments are mainly surgery combined with radiotherapy and chemotherapy, which is still far from satisfactory. Therefore, it is of great clinical significance to find new therapeutic agents. Serving as an inhibitor of differentiation, protein ID2 (inhibitor of DNA binding 2) plays an important role in neurogenesis, neovascularization and malignant development of gliomas. It has been shown that ID2 affects the malignant progression of gliomas through different mechanisms. In this study, a pharmacophore-based virtual screening was carried out and 16 hit compounds were purchased for pharmacological evaluations on their ID2 inhibitory activities. Based on the cytotoxicity of these small-molecule compounds, two compounds were shown to effectively inhibit the viability of glioma cells in the micromolar range. Among them, AK-778-XXMU was chosen for further study due to its better solubility in water. A SPR (Surface Plasma Resonance) assay proved the high affinity between AK-778-XXMU and ID2 protein with the KD value as 129 nM. The plausible binding mode of ID2 was studied by molecular docking and it was found to match AGX51 very well in the same binding site. Subsequently, the cancer-suppressing potency of the compound was characterized both in vitro and in vivo. The data demonstrated that compound AK-778-XXMU is a potent ID2 antagonist which has the potential to be developed as a therapeutic agent against glioma.
Collapse
|
24
|
Tsakogiannis D, Nikolakopoulou A, Zagouri F, Stratakos G, Syrigos K, Zografos E, Koulouris N, Bletsa G. Update Overview of the Role of Angiopoietins in Lung Cancer. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:medicina57111191. [PMID: 34833409 PMCID: PMC8625006 DOI: 10.3390/medicina57111191] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 12/27/2022]
Abstract
Angiogenesis is a biological process that involves the formation of new blood vessels from the existing vasculature, and it plays a fundamental role in the development and progression of several types of cancer, including lung cancer. The angiopoietin/Tie2 ligand/receptor system orchestrates vascular integrity. In particular, Angiopoietin-1 activates the endothelial cell (EC)-specific receptor tyrosine kinase, Tie2, which is essential for preserving endothelial quiescence. On the other hand, Angiopoietin-2 acts as an inhibitor of the Angiopoietin-1/Tie2 signaling pathways, thus facilitating the destabilization of quiescent endothelium in cases of inflammation and cancer. Clinical studies have proven that high levels of Angiopoietin-2 indicate the development of non-small-cell lung carcinomas (NSCLC), while high levels of Angiopoietin-2 are strongly related to tumor angiogenesis, lymphangiogenesis, metastasis, and poor prognosis. Interestingly, the association of Angiopoietin-2 levels with the type of surgical approach makes Angiopoietin-2 a valuable factor in selecting the most suitable therapeutic strategy for lung cancer patients. The role of the Angiopoietin-1 and Angiopoietin-4 levels in NSCLC development requires further investigation. The present review focuses on the clinical impact of the Angiopoietin-1, Angiopoietin-2, and Angiopoietin-4 levels in patients diagnosed with NSCLC, emphasizing the interaction between them, and how they affect the development, progression, and metastasis of lung disease. Finally, it estimates the role of angiopoietins levels in the effective therapy of lung cancer patients.
Collapse
Affiliation(s)
| | - Asimina Nikolakopoulou
- Intensive Care Unit, 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.N.); (G.S.); (N.K.)
| | - Flora Zagouri
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (F.Z.); (E.Z.)
| | - Grigorios Stratakos
- Intensive Care Unit, 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.N.); (G.S.); (N.K.)
| | - Konstantinos Syrigos
- Third Department of Internal Medicine, Sotiria Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Eleni Zografos
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (F.Z.); (E.Z.)
| | - Nikolaos Koulouris
- Intensive Care Unit, 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.N.); (G.S.); (N.K.)
| | - Garyfalia Bletsa
- Research Center, Hellenic Anticancer Institute, 10680 Athens, Greece;
- Correspondence: or ; Tel./Fax: +30-21-03643723
| |
Collapse
|
25
|
Chen H, Stoltzfus KC, Lehrer EJ, Horn SR, Siva S, Trifiletti DM, Meng MB, Verma V, Louie AV, Zaorsky NG. The Epidemiology of Lung Metastases. Front Med (Lausanne) 2021; 8:723396. [PMID: 34616754 PMCID: PMC8488106 DOI: 10.3389/fmed.2021.723396] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/11/2021] [Indexed: 12/31/2022] Open
Abstract
Introduction: Lung metastasis is usually associated with poor outcomes in cancer patients. This study was performed to characterize and analyze the population of patients with de novo (synchronous) lung metastases using the Surveillance, Epidemiology and End Results (SEER) database. Materials and Methods: Baseline characteristics of lung metastasis patients were obtained from SEER case listings. Incidence rates and counts of synchronous lung metastasis were also obtained using the SEER*Stat software. Survival outcomes were analyzed using univariate and multivariable Cox regressions, controlling for confounders. An alpha threshold of 0.05 was used for statistical significance and p-values were subject to correction for multiple comparisons. Results: The age-adjusted incidence rate of synchronous lung metastasis was 17.92 per 100,000 between 2010 and 2015. Synchronous lung metastases most commonly arose from primary lung cancers, colorectal cancers, kidney cancers, pancreatic cancers and breast cancers. During this time period, 4% of all cancer cases presented with synchronous lung metastasis. The percentage of patients presenting with synchronous lung metastasis ranged from 0.5% of all prostate cancers to 13% of all primary lung cancers. The percentage of all cancer cases presenting with synchronous lung metastasis increased over time. De novo metastatic patients with lung metastases had worse overall survival [hazard ratio = 1.22 (1.21–1.23), p < 0.001] compared to those with only extrapulmonary metastases, controlling for potential confounders. Conclusions: Synchronous lung metastasis occurs frequently and is an independent predictors of poor patient outcomes. As treatment for lung metastases becomes more complicated, patients with synchronous lung metastasis represent a high-risk population.
Collapse
Affiliation(s)
- Hanbo Chen
- Department of Radiation Oncology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Kelsey C Stoltzfus
- Department of Radiation Oncology, Penn State Cancer Institute, Hershey, PA, United States.,Department of Public Health Sciences, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Eric J Lehrer
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Samantha R Horn
- Department of Radiation Oncology, Penn State Cancer Institute, Hershey, PA, United States.,Department of Public Health Sciences, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Shankar Siva
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Daniel M Trifiletti
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, United States
| | - Mao-Bin Meng
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Vivek Verma
- Department of Radiation Oncology, Allegheny General Hospital, Pittsburgh, PA, United States
| | - Alexander V Louie
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Nicholas G Zaorsky
- Department of Radiation Oncology, Penn State Cancer Institute, Hershey, PA, United States.,Department of Public Health Sciences, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, United States
| |
Collapse
|
26
|
ITLN1 inhibits tumor neovascularization and myeloid derived suppressor cells accumulation in colorectal carcinoma. Oncogene 2021; 40:5925-5937. [PMID: 34363021 DOI: 10.1038/s41388-021-01965-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/15/2021] [Indexed: 02/07/2023]
Abstract
Low levels of ITLN1 have been correlated with obesity-related colorectal carcinogenesis, however, the specific functions and underlying mechanisms remain unclear. Thus, we sought to explore the inhibitory role of ITLN1 in the tumor-permissive microenvironment that exists during the first occurrence and subsequent development of colorectal carcinoma (CRC). Results indicated that ITLN1 was frequently lost in CRC tissues and ITLN1 to be an independent prognostic predictor of CRC. Orthotopic and subcutaneous tumor xenograft approaches were then used to further confirm the protective role of ITLN1 during tumor progression. Increased ITLN1 expression in CRC cells significantly inhibited local pre-existing vessels sprouting, EPC recruitment and the infiltration of immunosuppressive myeloid-derived suppressor cells (MDSCs) into tumor tissues without affecting the behavior of CRC cells in vitro. Comparatively, ITLN1-derived MDSCs had a lower suppressive effect on T cell proliferation, NOS2 expression, and ROS production. In addition, ITLN1 overexpression markedly suppressed bone marrow (BM)-derived hematopoietic progenitor cells (HPC) differentiation into MDSCs as well as NOS2 activity on MDSCs. Using H-2b+YFP + chimerism through bone marrow transplantation, increased ITLN1 in HCT116 significantly reduced the BM-derived EPCs and MDSCs in vivo mobilization. Mechanistically, results indicated ITLN1 inhibited tumor-derived IL-17D and CXCL2 (MIP2) through the KEAP1/Nrf2/ROS/IL-17D and p65 NF-ĸB/CXCL2 signaling cascades dependent on PI3K/AKT/GSK3ß. This effect was reversed by the PI3K selective inhibitor LY294002. Collectively, ITLN1 synergistically suppressed IL-17D and CXCL2-mediated tumor vascularization, bone marrow derived EPC recruitment, as well as MDSCs generation and trafficking. Thus, ITLN1 potentially serves as a critical prognostic and therapeutic target for CRC.
Collapse
|
27
|
Thibault B, Ramos‐Delgado F, Pons‐Tostivint E, Therville N, Cintas C, Arcucci S, Cassant‐Sourdy S, Reyes‐Castellanos G, Tosolini M, Villard AV, Cayron C, Baer R, Bertrand‐Michel J, Pagan D, Ferreira Da Mota D, Yan H, Falcomatà C, Muscari F, Bournet B, Delord J, Aksoy E, Carrier A, Cordelier P, Saur D, Basset C, Guillermet‐Guibert J. Pancreatic cancer intrinsic PI3Kα activity accelerates metastasis and rewires macrophage component. EMBO Mol Med 2021; 13:e13502. [PMID: 34033220 PMCID: PMC8261517 DOI: 10.15252/emmm.202013502] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 04/17/2021] [Accepted: 04/23/2021] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) patients frequently suffer from undetected micro-metastatic disease. This clinical situation would greatly benefit from additional investigation. Therefore, we set out to identify key signalling events that drive metastatic evolution from the pancreas. We searched for a gene signature that discriminate localised PDAC from confirmed metastatic PDAC and devised a preclinical protocol using circulating cell-free DNA (cfDNA) as an early biomarker of micro-metastatic disease to validate the identification of key signalling events. An unbiased approach identified, amongst actionable markers of disease progression, the PI3K pathway and a distinctive PI3Kα activation signature as predictive of PDAC aggressiveness and prognosis. Pharmacological or tumour-restricted genetic PI3Kα-selective inhibition prevented macro-metastatic evolution by hindering tumoural cell migratory behaviour independently of genetic alterations. We found that PI3Kα inhibition altered the quantity and the species composition of the produced lipid second messenger PIP3 , with a selective decrease of C36:2 PI-3,4,5-P3 . Tumoural PI3Kα inactivation prevented the accumulation of pro-tumoural CD206-positive macrophages in the tumour-adjacent tissue. Tumour cell-intrinsic PI3Kα promotes pro-metastatic features that could be pharmacologically targeted to delay macro-metastatic evolution.
Collapse
Affiliation(s)
- Benoit Thibault
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- LABEX TouCANToulouseFrance
| | - Fernanda Ramos‐Delgado
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- LABEX TouCANToulouseFrance
| | - Elvire Pons‐Tostivint
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- LABEX TouCANToulouseFrance
| | - Nicole Therville
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- LABEX TouCANToulouseFrance
| | - Celia Cintas
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- LABEX TouCANToulouseFrance
| | - Silvia Arcucci
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- LABEX TouCANToulouseFrance
| | - Stephanie Cassant‐Sourdy
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- LABEX TouCANToulouseFrance
| | | | - Marie Tosolini
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
| | - Amelie V Villard
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- LABEX TouCANToulouseFrance
| | - Coralie Cayron
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- LABEX TouCANToulouseFrance
| | - Romain Baer
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- LABEX TouCANToulouseFrance
| | | | - Delphine Pagan
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
| | - Dina Ferreira Da Mota
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- Institut Universitaire du Cancer de Toulouse – Oncopole (IUCT‐O)Hopitaux de ToulouseInstitut Claudius Regaud ToulouseFrance
| | - Hongkai Yan
- Division of Translational Cancer ResearchGerman Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany
- Chair of Translational Cancer Research and Institute of Experimental Cancer TherapyKlinikum rechts der IsarSchool of MedicineTechnische Universität MünchenMunichGermany
| | - Chiara Falcomatà
- Division of Translational Cancer ResearchGerman Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany
- Chair of Translational Cancer Research and Institute of Experimental Cancer TherapyKlinikum rechts der IsarSchool of MedicineTechnische Universität MünchenMunichGermany
| | - Fabrice Muscari
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- Institut Universitaire du Cancer de Toulouse – Oncopole (IUCT‐O)Hopitaux de ToulouseInstitut Claudius Regaud ToulouseFrance
| | - Barbara Bournet
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- Institut Universitaire du Cancer de Toulouse – Oncopole (IUCT‐O)Hopitaux de ToulouseInstitut Claudius Regaud ToulouseFrance
| | - Jean‐Pierre Delord
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- Institut Universitaire du Cancer de Toulouse – Oncopole (IUCT‐O)Hopitaux de ToulouseInstitut Claudius Regaud ToulouseFrance
| | - Ezra Aksoy
- Centre for Biochemical PharmacologyWilliam Harvey Research InstituteQueen Mary University of LondonLondonUK
| | - Alice Carrier
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli‐Calmettes, CRCMMarseilleFrance
| | - Pierre Cordelier
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
| | - Dieter Saur
- Division of Translational Cancer ResearchGerman Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany
- Chair of Translational Cancer Research and Institute of Experimental Cancer TherapyKlinikum rechts der IsarSchool of MedicineTechnische Universität MünchenMunichGermany
| | - Celine Basset
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- LABEX TouCANToulouseFrance
- Institut Universitaire du Cancer de Toulouse – Oncopole (IUCT‐O)Hopitaux de ToulouseInstitut Claudius Regaud ToulouseFrance
| | - Julie Guillermet‐Guibert
- Centre de Recherches en Cancérologie de ToulouseInserm, CNRSUniversité de ToulouseToulouseFrance
- LABEX TouCANToulouseFrance
| |
Collapse
|
28
|
Wojnarowicz PM, Escolano MG, Huang YH, Desai B, Chin Y, Shah R, Xu S, Yadav S, Yaklichkin S, Ouerfelli O, Soni RK, Philip J, Montrose DC, Healey JH, Rajasekhar VK, Garland WA, Ratiu J, Zhuang Y, Norton L, Rosen N, Hendrickson RC, Zhou XK, Iavarone A, Massague J, Dannenberg AJ, Lasorella A, Benezra R. Anti-tumor effects of an ID antagonist with no observed acquired resistance. NPJ Breast Cancer 2021; 7:58. [PMID: 34031428 PMCID: PMC8144414 DOI: 10.1038/s41523-021-00266-0] [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: 11/03/2020] [Accepted: 04/15/2021] [Indexed: 12/19/2022] Open
Abstract
ID proteins are helix-loop-helix (HLH) transcriptional regulators frequently overexpressed in cancer. ID proteins inhibit basic-HLH transcription factors often blocking differentiation and sustaining proliferation. A small-molecule, AGX51, targets ID proteins for degradation and impairs ocular neovascularization in mouse models. Here we show that AGX51 treatment of cancer cell lines impairs cell growth and viability that results from an increase in reactive oxygen species (ROS) production upon ID degradation. In mouse models, AGX51 treatment suppresses breast cancer colonization in the lung, regresses the growth of paclitaxel-resistant breast tumors when combined with paclitaxel and reduces tumor burden in sporadic colorectal neoplasia. Furthermore, in cells and mice, we fail to observe acquired resistance to AGX51 likely the result of the inability to mutate the binding pocket without loss of ID function and efficient degradation of the ID proteins. Thus, AGX51 is a first-in-class compound that antagonizes ID proteins, shows strong anti-tumor effects and may be further developed for the management of multiple cancers.
Collapse
Affiliation(s)
- Paulina M Wojnarowicz
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marta Garcia Escolano
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yun-Han Huang
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell/Sloan Kettering/Rockefeller Tri-Institutional MD-PhD Program, New York, NY, 10065, USA
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, New York, NY, 10065, USA
| | - Bina Desai
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yvette Chin
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Riddhi Shah
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sijia Xu
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Saurabh Yadav
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sergey Yaklichkin
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ouathek Ouerfelli
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rajesh Kumar Soni
- Proteomics & Microchemistry Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John Philip
- Proteomics & Microchemistry Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David C Montrose
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - John H Healey
- Orthopedics Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Jeremy Ratiu
- Department of Immunology, Duke University, Durham, NC, USA
| | - Yuan Zhuang
- Department of Immunology, Duke University, Durham, NC, USA
| | - Larry Norton
- Evelyn H. Lauder Breast Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Neal Rosen
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronald C Hendrickson
- Proteomics & Microchemistry Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xi Kathy Zhou
- Department of Healthcare Policy and Research Weill Cornell Medical College, New York, NY, USA
| | - Antonio Iavarone
- Department of Neurology, Department of Pathology, Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Joan Massague
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Anna Lasorella
- Department of Pediatrics, Department of Pathology, Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Robert Benezra
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
29
|
Ferlini L, Peluso L, Lolli V, Gaspard N, Lefranc F. Prognosis of patients treated in a single neurosurgical reference centre for brain metastasis caused by dormant disseminated cells. Oncol Lett 2021; 21:454. [PMID: 33907564 PMCID: PMC8063273 DOI: 10.3892/ol.2021.12715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/30/2020] [Indexed: 11/06/2022] Open
Abstract
Brain metastasis (BM) is a frequent complication of systemic cancer usually associated with poor prognosis. Survival depends on numerous factors, which complicates prognosis and treatment. It has been suggested that BM growing from previously dormant disseminated tumour cells (DTCs) may exhibit a milder phenotype than BM derived from continuously progressing metastatic cells; however, to the best of our knowledge, the prognosis of patients presenting with BM from dormant DTCs is unknown. The present study retrospectively compared survival data, collected from a single neurosurgical centre, between patients presenting with BM from previously dormant DTCs and patients with non-dormant BM. A total of 262 medical records were reviewed. In the univariate Cox regression analysis, the median survival of the dormant BM group was statistically longer than that of the non-dormant group (P=0.048); a trend towards a longer survival persisted after correcting for age, presence of breast cancer and treatment options (P=0.057), which are all factors known to influence outcome. The improved outcome of these patients could be considered in models for prognostication. Moreover, the development of therapies able to eradicate dormant DTCs could provide a new promising strategy to prolong the survival of patients with a favourable prognosis.
Collapse
Affiliation(s)
- Lorenzo Ferlini
- Department of Neurology, Erasmus Hospital, Free University of Bruxelles, B-1070 Brussels, Belgium
| | - Lorenzo Peluso
- Department of Intensive Care, Erasmus Hospital, Free University of Bruxelles, B-1070 Brussels, Belgium
| | - Valentina Lolli
- Department of Radiology, Erasmus Hospital, Free University of Bruxelles, B-1070 Brussels, Belgium
| | - Nicolas Gaspard
- Department of Neurology, Erasmus Hospital, Free University of Bruxelles, B-1070 Brussels, Belgium
| | - Florence Lefranc
- Department of Neurosurgery, Erasmus Hospital, Free University of Bruxelles, B-1070 Brussels, Belgium
| |
Collapse
|
30
|
Xiong A, Li C, Xu J, Yang X, Nie W, Zhong H, Chu T, Zhang W, Zhong R, Pan F, Shen Y, Lou Y, Zhang B, Han B, Zhang X. Solid subtype predicts early bone metastases in sensitive EGFR-mutated lung adenocarcinoma patients after surgery. Lung Cancer 2021; 154:124-130. [PMID: 33657514 DOI: 10.1016/j.lungcan.2021.02.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/17/2021] [Accepted: 02/21/2021] [Indexed: 12/21/2022]
Abstract
This study aimed to explore the prognostic significance of solid pattern for bone metastases (BM) in epidermal growth factor receptor (EGFR)-mutated lung adenocarcinoma patients after surgery. A total of 237 stage I-III lung adenocarcinoma patients with EGFR mutation were analyzed after procedure. The patients were divided into four groups: the solid-present patients with BM, the solid-absent patients with BM, the solid-present patients without BM and the solid-absent patients without BM. The bone disease-free survival (bDFS), systemic disease-free survival (DFS) and overall survival (OS) were assessed. The results revealed that the patients with solid pattern had shorter DFS (15 months vs. 19 months; P < 0.001) and OS (47 months vs. 77 months; P = 0.001). Moreover, bDFS of solid-present patients was significantly shorter than solid-absent patients (27 months vs. 14 months; P < 0.001). In addition, patients with solid component had worsened bDFS, no matter with BM as first-site development (12.5 months vs. 16.5 months; P = 0.016) or non-first-site development (16.5 months vs. 45.5 months; P < 0.001). These findings suggested that solid pattern predicted worse DFS and OS and also showed shortened interval between surgery and BM.
Collapse
Affiliation(s)
- Anning Xiong
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China
| | - Changhui Li
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China
| | - Jianlin Xu
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China
| | - Xiaohua Yang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China
| | - Wei Nie
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China
| | - Hua Zhong
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China
| | - Tianqing Chu
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China
| | - Wei Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China
| | - Runbo Zhong
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China
| | - Feng Pan
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China
| | - Yinchen Shen
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China
| | - Yuqing Lou
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China
| | - Bo Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China
| | - Baohui Han
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China.
| | - Xueyan Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, China.
| |
Collapse
|
31
|
Ge Y, Zhou L, Chen Z, Mao Y, Li T, Tong P, Shan L. Identification of differentially expressed genes, signaling pathways and immune infiltration in rheumatoid arthritis by integrated bioinformatics analysis. Hereditas 2021; 158:5. [PMID: 33397492 PMCID: PMC7784358 DOI: 10.1186/s41065-020-00169-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 12/08/2020] [Indexed: 12/21/2022] Open
Abstract
Background The disability rate associated with rheumatoid arthritis (RA) ranks high among inflammatory joint diseases. However, the cause and potential molecular events are as yet not clear. Here, we aimed to identify differentially expressed genes (DEGs), pathways and immune infiltration involved in RA utilizing integrated bioinformatics analysis and investigating potential molecular mechanisms. Materials and methods The expression profiles of GSE55235, GSE55457, GSE55584 and GSE77298 were downloaded from the Gene Expression Omnibus database, which contained 76 synovial membrane samples, including 49 RA samples and 27 normal controls. The microarray datasets were consolidated and DEGs were acquired and further analyzed by bioinformatics techniques. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of DEGs were performed using R (version 3.6.1) software, respectively. The protein-protein interaction (PPI) network of DEGs were developed utilizing the STRING database. Finally, the CIBERSORT was used to evaluate the infiltration of immune cells in RA. Results A total of 828 DEGs were recognized, with 758 up-regulated and 70 down-regulated. GO and KEGG pathway analyses demonstrated that these DEGs focused primarily on cytokine receptor activity and relevant signaling pathways. The 30 most firmly related genes among DEGs were identified from the PPI network. The principal component analysis showed that there was a significant difference between the two tissues in infiltration immune. Conclusion This study shows that screening for DEGs, pathways and immune infiltration utilizing integrated bioinformatics analyses could aid in the comprehension of the molecular mechanisms involved in RA development. Besides, our study provides valuable data related to DEGs, pathways and immune infiltration of RA and may provide new insight into the understanding of molecular mechanisms.
Collapse
Affiliation(s)
- Yanzhi Ge
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Zuxiang Chen
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Yingying Mao
- Department of Epidemiology and Biostatistics, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Ting Li
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, PR China
| | - Peijian Tong
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| |
Collapse
|
32
|
Abstract
Tumor cells frequently disseminate to distant organ sites, where they encounter permissive or restrictive environments that enable them to grow and colonize or enter a dormant state. Tumor dormancy is not strictly defined, but generally describes a tumor cell that is non-proliferative or in a state of balanced equilibrium, in which the proliferation rate of the tumor cell or cells is equal to its rate of cell death. The mechanisms that regulate tumor cell entry into and exit from dormancy are poorly understood, but microenvironmental features as well as tumor cell intrinsic factors play an important role in mediating this transition. Upon homing to distant metastatic sites, tumor cells may disseminate into various niches, most frequently the perivascular, hematopoietic stem cell, or endosteal/osteogenic niche. Tumor cells sense the cytokines, growth factors, and chemo-attractants from each of these niches, and tumor cell expression of cognate ligands and receptors can determine whether a tumor cell enters or exits dormancy. In addition to the secreted factors and cell-cell interactions that regulate dormancy, the cellular milieu also impacts upon disseminated tumor cells to promote or restrain their growth in distant metastatic sites. In this chapter we will discuss the role of the osteogenic and perivascular niche on dormant tumor cells, as well as the impact of hypoxia (low oxygen tensions) and the immune system on the restriction and outgrowth of dormant, disseminated tumor cells.
Collapse
|
33
|
Abstract
Defined by its potential for self-renewal, differentiation and tumorigenicity, cancer stem cells (CSCs) are considered responsible for drug resistance and relapse. To understand the behavior of CSC, the effects of the microenvironment in each tissue are a matter of great concerns for scientists in cancer biology. However, there are many complicated obstacles in the mimicking the microenvironment of CSCs even with current advanced technology. In this context, novel biomaterials have widely been assessed as in vitro platforms for their ability to mimic cancer microenvironment. These efforts should be successful to identify and characterize various CSCs specific in each type of cancer. Therefore, extracellular matrix scaffolds made of biomaterial will modulate the interactions and facilitate the investigation of CSC associated with biological phenomena simplifying the complexity of the microenvironment. In this review, we summarize latest advances in biomaterial scaffolds, which are exploited to mimic CSC microenvironment, and their chemical and biological requirements with discussion. The discussion includes the possible effects on both cells in tumors and microenvironment to propose what the critical factors are in controlling the CSC microenvironment focusing the future investigation. Our insights on their availability in drug screening will also follow the discussion.
Collapse
|
34
|
Kadioglu O, Saeed MEM, Mahmoud N, Hussein Azawi SS, Rincic M, Liehr T, Efferth T. Identification of metastasis-related genes by genomic and transcriptomic studies in murine melanoma. Life Sci 2020; 267:118922. [PMID: 33358905 DOI: 10.1016/j.lfs.2020.118922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 01/10/2023]
Abstract
AIMS We systematically characterized metastatic murine B16-F10 melanoma, a sub-line derived from murine melanoma B16-F1 cells. MATERIALS AND METHODS RNA-sequencing and network analyses (Ingenuity Pathway Analysis) were performed to identify novel potential metastasis mechanisms. Chromosomal aberrations were identified by multicolor fluorescence in situ hybridization (mFISH) using all 21 murine whole chromosome painting probes. KEY FINDINGS Numerous genes were overexpressed in B16-F10 cells, some of which have been already described as being metastasis-linked. Nr5a1/sf1, a known prognostic marker for adrenal tumors, was 177-fold upregulated in B16-F10 cells compared to B16-F1 cells. Hoxb8 was 75-fold upregulated, which was previously associated with gastric cancer progression and metastasis. Ptk7, which is linked with tumorigenesis and metastasis of esophageal squamous carcinoma, was 67-fold upregulated. B16-F10 cells acquired additional chromosomal aberrations compared to B16-F1 cells, including dic(4)(pter->qter:qter->pter), +dic(6;15), +der(10)t(10;?1;16). SIGNIFICANCE In addition to well-known metastatic genes, numerous novel genes and genomic aberrations were identified, which may serve as targets for treatment in the future. Transcriptomic and genetic analyses in B16-F10 cells unraveled a range of novel metastasis mechanisms, which may also have important implications for future treatment strategies.
Collapse
Affiliation(s)
- Onat Kadioglu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Mohamed E M Saeed
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Nuha Mahmoud
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Shaymaa S Hussein Azawi
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Jena, Germany
| | - Martina Rincic
- Croatian Institute for Brain Research, School of Medicine University of Zagreb, Zagreb, Croatia
| | - Thomas Liehr
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Jena, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
35
|
The Role of Complement in Angiogenesis. Antibodies (Basel) 2020; 9:antib9040067. [PMID: 33271774 PMCID: PMC7709120 DOI: 10.3390/antib9040067] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022] Open
Abstract
The link of the complement system to angiogenesis has remained circumstantial and speculative for several years. Perhaps the most clinically relevant example of possible involvement of complement in pathological neovascularization is age-related macular degeneration. Recent studies, however, provide more direct and experimental evidence that indeed the complement system regulates physiological and pathological angiogenesis in models of wound healing, retinal regeneration, age-related macular degeneration, and cancer. Interestingly, complement-dependent mechanisms involved in angiogenesis are very much context dependent, including anti- and proangiogenic functions. Here, we discuss these new developments that place complement among other important regulators of homeostatic and pathological angiogenesis, and we provide the perspective on how these newly discovered complement functions can be targeted for therapy.
Collapse
|
36
|
Dai H, Tong C, Shi D, Chen M, Guo Y, Chen D, Han X, Wang H, Wang Y, Shen P. Efficacy and biomarker analysis of CD133-directed CAR T cells in advanced hepatocellular carcinoma: a single-arm, open-label, phase II trial. Oncoimmunology 2020; 9:1846926. [PMID: 33312759 PMCID: PMC7714531 DOI: 10.1080/2162402x.2020.1846926] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Expressed by cancer stem cells of various epithelial cell origins and hepatocellular carcinoma (HCC), CD133 is an attractive therapeutic target for HCC. The marker CD133 is highly expressed in endothelial progenitor cells (EPC). EPCs circulate in increased numbers in the peripheral blood of patients with highly vascularized HCC and contribute to angiogenesis and neovascularization. This phase II study investigated CD133-directed chimeric antigen receptor (CAR) T (CART-133) cells in adults with HCC. Patients with histologically confirmed and measurable advanced HCC and adequate hematologic, hepatic, and renal functions received CART-133 cell infusions. The primary endpoints were safety in phase I and progression-free survival (PFS) and overall survival (OS) in phase II. Other endpoints included biomarkers for CART-133 T cell therapy. Between June 1, 2015, and September 1, 2017, this study enrolled 21 patients who subsequently received CART-133 T cells across phases I and II. The median OS was 12 months (95% CI, 9.3–15.3 months) and the median PFS was 6.8 months (95% CI, 4.3–8.4 months). Of 21 evaluable patients, 1 had a partial response, 14 had stable disease for 2 to 16.3 months, and 6 progressed after T-cell infusion. The most common high-grade adverse event was hyperbilirubinemia. Outcome was correlated with the baseline levels of vascular endothelial growth factor (VEGF), soluble VEGF receptor 2 (sVEGFR2), stromal cell-derived factor (SDF)-1, and EPC counts. Changes in EPC counts, VEGF, SDF-1, sVEGFR2, and interferon (IFN)-γ after cell infusion were associated with survival. In patients with previously treated advanced HCC, CART-133 cell therapy demonstrates promising antitumor activity and a manageable safety profile. We identified early changes in circulating molecules as potential biomarkers of response to CART-133 cells. The predictive value of these proangiogenic and inflammatory factors as potential biomarkers of CART-133 cell therapy in HCC will be explored in prospective trials. This study is registered at ClinicalTrials.gov (NCT02541370)
Collapse
Affiliation(s)
- Hanren Dai
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,State Key Laboratory of Pharmaceutical Biotechnology, Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Science, Nanjing University, Nanjing, China.,Biotherapeutic Department, The First Medical Centre, Beijing, China
| | - Chuan Tong
- Biotherapeutic Department, The First Medical Centre, Beijing, China
| | - Daiwei Shi
- Department of General Surgery, The Second People's Hospital of Hefei, Hefei, China
| | - Meixia Chen
- Biotherapeutic Department, The First Medical Centre, Beijing, China
| | - Yelei Guo
- Biotherapeutic Department, The First Medical Centre, Beijing, China
| | - Deyun Chen
- Biotherapeutic Department, The First Medical Centre, Beijing, China
| | - Xiao Han
- Biotherapeutic Department, The First Medical Centre, Beijing, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yao Wang
- Biotherapeutic Department, The First Medical Centre, Beijing, China
| | - Pingping Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Science, Nanjing University, Nanjing, China
| |
Collapse
|
37
|
Solimando AG, Summa SD, Vacca A, Ribatti D. Cancer-Associated Angiogenesis: The Endothelial Cell as a Checkpoint for Immunological Patrolling. Cancers (Basel) 2020; 12:cancers12113380. [PMID: 33203154 PMCID: PMC7696032 DOI: 10.3390/cancers12113380] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/08/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary A clinical decision and study design investigating the level and extent of angiogenesis modulation aimed at vascular normalization without rendering tissues hypoxic is key and represents an unmet medical need. Specifically, determining the active concentration and optimal times of the administration of antiangiogenetic drugs is crucial to inhibit the growth of any microscopic residual tumor after surgical resection and in the pre-malignant and smolder neoplastic state. This review uncovers the pre-clinical translational insights crucial to overcome the caveats faced so far while employing anti-angiogenesis. This literature revision also explores how abnormalities in the tumor endothelium harm the crosstalk with an effective immune cell response, envisioning a novel combination with other anti-cancer drugs and immunomodulatory agents. These insights hold vast potential to both repress tumorigenesis and unleash an effective immune response. Abstract Cancer-associated neo vessels’ formation acts as a gatekeeper that orchestrates the entrance and egress of patrolling immune cells within the tumor milieu. This is achieved, in part, via the directed chemokines’ expression and cell adhesion molecules on the endothelial cell surface that attract and retain circulating leukocytes. The crosstalk between adaptive immune cells and the cancer endothelium is thus essential for tumor immune surveillance and the success of immune-based therapies that harness immune cells to kill tumor cells. This review will focus on the biology of the endothelium and will explore the vascular-specific molecular mediators that control the recruitment, retention, and trafficking of immune cells that are essential for effective antitumor immunity. The literature revision will also explore how abnormalities in the tumor endothelium impair crosstalk with adaptive immune cells and how targeting these abnormalities can improve the success of immune-based therapies for different malignancies, with a particular focus on the paradigmatic example represented by multiple myeloma. We also generated and provide two original bio-informatic analyses, in order to sketch the physiopathology underlying the endothelial–neoplastic interactions in an easier manner, feeding into a vicious cycle propagating disease progression and highlighting novel pathways that might be exploited therapeutically.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine ‘G. Baccelli’, University of Bari Medical School, 70124 Bari, Italy;
- Istituto di Ricovero e Cura a Carattere Scientifico-IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy
- Correspondence: (A.G.S.); (D.R.); Tel.: +39-3395626475 (A.G.S.); +39-080-5478326 (D.R.)
| | - Simona De Summa
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy;
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine ‘G. Baccelli’, University of Bari Medical School, 70124 Bari, Italy;
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (A.G.S.); (D.R.); Tel.: +39-3395626475 (A.G.S.); +39-080-5478326 (D.R.)
| |
Collapse
|
38
|
Zhao G, Weiner AI, Neupauer KM, de Mello Costa MF, Palashikar G, Adams-Tzivelekidis S, Mangalmurti NS, Vaughan AE. Regeneration of the pulmonary vascular endothelium after viral pneumonia requires COUP-TF2. SCIENCE ADVANCES 2020; 6:eabc4493. [PMID: 33239293 PMCID: PMC7688336 DOI: 10.1126/sciadv.abc4493] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 10/09/2020] [Indexed: 05/08/2023]
Abstract
Acute respiratory distress syndrome is associated with a robust inflammatory response that damages the vascular endothelium, impairing gas exchange. While restoration of microcapillaries is critical to avoid mortality, therapeutic targeting of this process requires a greater understanding of endothelial repair mechanisms. Here, we demonstrate that lung endothelium possesses substantial regenerative capacity and lineage tracing reveals that native endothelium is the source of vascular repair after influenza injury. Ablation of chicken ovalbumin upstream promoter-transcription factor 2 (COUP-TF2) (Nr2f2), a transcription factor implicated in developmental angiogenesis, reduced endothelial proliferation, exacerbating viral lung injury in vivo. In vitro, COUP-TF2 regulates proliferation and migration through activation of cyclin D1 and neuropilin 1. Upon influenza injury, nuclear factor κB suppresses COUP-TF2, but surviving endothelial cells ultimately reestablish vascular homeostasis dependent on restoration of COUP-TF2. Therefore, stabilization of COUP-TF2 may represent a therapeutic strategy to enhance recovery from pathogens, including H1N1 influenza and SARS-CoV-2.
Collapse
Affiliation(s)
- Gan Zhao
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Aaron I Weiner
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katherine M Neupauer
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria Fernanda de Mello Costa
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gargi Palashikar
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephanie Adams-Tzivelekidis
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nilam S Mangalmurti
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew E Vaughan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
39
|
Sistigu A, Musella M, Galassi C, Vitale I, De Maria R. Tuning Cancer Fate: Tumor Microenvironment's Role in Cancer Stem Cell Quiescence and Reawakening. Front Immunol 2020; 11:2166. [PMID: 33193295 PMCID: PMC7609361 DOI: 10.3389/fimmu.2020.02166] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cell dormancy is a common feature of human tumors and represents a major clinical barrier to the long-term efficacy of anticancer therapies. Dormant cancer cells, either in primary tumors or disseminated in secondary organs, may reawaken and relapse into a more aggressive disease. The mechanisms underpinning dormancy entry and exit strongly resemble those governing cancer cell stemness and include intrinsic and contextual cues. Cellular and molecular components of the tumor microenvironment persistently interact with cancer cells. This dialog is highly dynamic, as it evolves over time and space, strongly cooperates with intrinsic cell nets, and governs cancer cell features (like quiescence and stemness) and fate (survival and outgrowth). Therefore, there is a need for deeper insight into the biology of dormant cancer (stem) cells and the mechanisms regulating the equilibrium quiescence-versus-proliferation are vital in our pursuit of new therapeutic opportunities to prevent cancer from recurring. Here, we review and discuss microenvironmental regulations of cancer dormancy and its parallels with cancer stemness, and offer insights into the therapeutic strategies adopted to prevent a lethal recurrence, by either eradicating resident dormant cancer (stem) cells or maintaining them in a dormant state.
Collapse
Affiliation(s)
- Antonella Sistigu
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy.,Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Martina Musella
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudia Galassi
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ilio Vitale
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS Candiolo (TO), Candiolo, Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Ruggero De Maria
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| |
Collapse
|
40
|
Flow cytometric analysis of circulating endothelial cells and endothelial progenitor cells in pediatric solid tumors: prognostic impact on treatment response and survival. Cancer Immunol Immunother 2020; 70:755-761. [PMID: 32945943 DOI: 10.1007/s00262-020-02719-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/01/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND AIM Solid tumors, including pediatric malignancies, depend on angiogenesis for tumor growth, invasion, and metastases. We aimed to evaluate the prognostic impact of circulating endothelial cells (CECs) and endothelial progenitor cells (EPCs) on treatment response and survival of pediatric patients with solid tumors. METHODS A prospective study included 70 patients with different pediatric solid tumors treated with different types of chemotherapy and 20 age and sex-matched healthy children as controls. Blood samples collected at diagnosis then on day 7 and day 21 after chemotherapy. CECs and EPCs were evaluated using flow cytometry. RESULTS The mean levels of CECs and EPCs of patients at diagnosis were significantly higher than controls (85.29 ± 24.78 and 26.1 ± 9.11 versus 20.08 ± 6.65; and EPCs; 2.78 ± 1.48, respectively; P < 0.001 for both). The highest levels of CECs were observed in patients with rhabdomyosarcoma (RMS). An overall increase was reported in CECs, and after the first cycle of chemotherapy, that was significantly correlated to treatment response and overall survival. CONCLUSION Pediatric patients with solid tumors have elevated levels of CECs and EPCs with more elevation after chemotherapy. The magnitude of increase of CECs occurred on day 7 after chemotherapy may be considered as an early predictor of response to therapy and outcome in pediatric patients with solid tumors.
Collapse
|
41
|
Teo WS, Holliday H, Karthikeyan N, Cazet AS, Roden DL, Harvey K, Konrad CV, Murali R, Varghese BA, Thankamony AP, Chan CL, McFarland A, Junankar S, Ye S, Yang J, Nikolic I, Shah JS, Baker LA, Millar EKA, Naylor MJ, Ormandy CJ, Lakhani SR, Kaplan W, Mellick AS, O'Toole SA, Swarbrick A, Nair R. Id Proteins Promote a Cancer Stem Cell Phenotype in Mouse Models of Triple Negative Breast Cancer via Negative Regulation of Robo1. Front Cell Dev Biol 2020; 8:552. [PMID: 32766238 PMCID: PMC7380117 DOI: 10.3389/fcell.2020.00552] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/10/2020] [Indexed: 01/02/2023] Open
Abstract
Breast cancers display phenotypic and functional heterogeneity and several lines of evidence support the existence of cancer stem cells (CSCs) in certain breast cancers, a minor population of cells capable of tumor initiation and metastatic dissemination. Identifying factors that regulate the CSC phenotype is therefore important for developing strategies to treat metastatic disease. The Inhibitor of Differentiation Protein 1 (Id1) and its closely related family member Inhibitor of Differentiation 3 (Id3) (collectively termed Id) are expressed by a diversity of stem cells and are required for metastatic dissemination in experimental models of breast cancer. In this study, we show that ID1 is expressed in rare neoplastic cells within ER-negative breast cancers. To address the function of Id1 expressing cells within tumors, we developed independent murine models of Triple Negative Breast Cancer (TNBC) in which a genetic reporter permitted the prospective isolation of Id1+ cells. Id1+ cells are enriched for self-renewal in tumorsphere assays in vitro and for tumor initiation in vivo. Conversely, depletion of Id1 and Id3 in the 4T1 murine model of TNBC demonstrates that Id1/3 are required for cell proliferation and self-renewal in vitro, as well as primary tumor growth and metastatic colonization of the lung in vivo. Using combined bioinformatic analysis, we have defined a novel mechanism of Id protein function via negative regulation of the Roundabout Axon Guidance Receptor Homolog 1 (Robo1) leading to activation of a Myc transcriptional programme.
Collapse
Affiliation(s)
- Wee S. Teo
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Holly Holliday
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Nitheesh Karthikeyan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Aurélie S. Cazet
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Daniel L. Roden
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Kate Harvey
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | | | - Reshma Murali
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Binitha Anu Varghese
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Archana P. Thankamony
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Manipal Academy of Higher Education, Manipal, India
| | - Chia-Ling Chan
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Andrea McFarland
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Simon Junankar
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Sunny Ye
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Jessica Yang
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Iva Nikolic
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Jaynish S. Shah
- Gene & Stem Cell Therapy Program, Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Laura A. Baker
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Ewan K. A. Millar
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Department of Anatomical Pathology, NSW Health Pathology, St George Hospital, Kogarah, NSW, Australia
- School of Medical Sciences, UNSW Sydney, Kensington, NSW, Australia
- School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Matthew J. Naylor
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Discipline of Physiology & Bosch Institute, University of Sydney, Sydney, NSW, Australia
| | - Christopher J. Ormandy
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Sunil R. Lakhani
- UQ Centre for Clinical Research, School of Medicine and Pathology Queensland, Royal Brisbane & Women's Hospital, The University of Queensland, Herston, QLD, Australia
| | - Warren Kaplan
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Peter Wills Bioinformatics Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Albert S. Mellick
- UNSW Medicine, University of NSW, Kensington, NSW, Australia
- Medical Oncology Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School UNSW & CONCERT Translational Cancer Research Centre, Liverpool, NSW, Australia
| | - Sandra A. O'Toole
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Alexander Swarbrick
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Radhika Nair
- Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| |
Collapse
|
42
|
Robinson NJ, Parker KA, Schiemann WP. Epigenetic plasticity in metastatic dormancy: mechanisms and therapeutic implications. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:903. [PMID: 32793747 DOI: 10.21037/atm.2020.02.177] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The overwhelming majority of cancer-associated morbidity and mortality can be ascribed to metastasis. Metastatic disease frequently presents in a delayed fashion following initial diagnosis and treatment, requiring that disseminated cancer cells (DCCs) spread early in tumor progression and persist in a dormant state at metastatic sites. To accomplish this feat, DCCs exhibit substantial phenotypic plasticity that is mediated by the epigenetic regulation of dormancy programs in response to intrinsic (i.e., cellular) and extrinsic (i.e., microenvironmental) cues. The epigenome is a dynamic landscape that encompasses transcriptional regulation via alteration of chromatin architecture, posttranscriptional RNA processing, and the diverse functions carried out by noncoding RNAs. Signals converging on DCCs are transduced through epigenetic effectors. Conversely, epigenetic regulation of gene expression controls the crosstalk between DCCs and cells of the metastatic niche, a phenomenon that is essential for the institution of dormant phenotypes. Importantly, epigenetic effectors can be targeted therapeutically, and the development of novel epigenetic therapies may provide new inroads to combating recurrent metastatic disease. Here we provide an overview of the dynamics of metastatic dormancy and summarize our current understanding of the intersections between dormancy and the epigenome, both mechanistically and therapeutically.
Collapse
Affiliation(s)
| | - Kimberly A Parker
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - William P Schiemann
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
43
|
Nasrollahzadeh E, Razi S, Keshavarz-Fathi M, Mazzone M, Rezaei N. Pro-tumorigenic functions of macrophages at the primary, invasive and metastatic tumor site. Cancer Immunol Immunother 2020; 69:1673-1697. [PMID: 32500231 DOI: 10.1007/s00262-020-02616-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/16/2020] [Indexed: 12/19/2022]
Abstract
The tumor microenvironment (TME) not only facilitates cancer progression from the early formation to distant metastasis, but also it differs itself from time to time alongside the tumor evolution. Tumor-associated macrophages (TAMs), whether as pre-existing tissue-resident macrophages or recruited monocytes, are an inseparable part of this microenvironment. As their parents are broadly classified into a dichotomic, simplistic M1 and M2 subtypes, TAMs also exert paradoxical and diverse phenotypes as they are settled in different regions of TME and receive different microenvironmental signals. Briefly, M1 macrophages induce an inflammatory precancerous niche and flame the early oncogenic mutations, whereas their M2 counterparts are reprogrammed to release various growth factors and providing an immunosuppressive state in TME as long as abetting hypoxic cancer cells to set up a new vasculature. Further, they mediate stromal micro-invasion and co-migrate with invasive cancer cells to invade the vascular wall and neural sheath, while another subtype of TAMs prepares suitable niches much earlier than metastatic cells arrive at the target tissues. Accordingly, at the neoplastic transformation, during the benign-to-malignant transition and through the metastatic cascade, macrophages are involved in shaping the primary, micro-invasive and pre-metastatic TMEs. Whether their behavioral plasticity is derived from distinct genotypes or is fueled by microenvironmental cues, it could define these cells as remarkably interesting therapeutic targets.
Collapse
Affiliation(s)
- Elaheh Nasrollahzadeh
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer Biology, VIB, KU Leuven, Louvain, B3000, Belgium
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, 14194, Tehran, Iran. .,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
44
|
Circulating cells and exosomes in acute myelogenous leukemia and their role in disease progression and survival. Clin Immunol 2020; 217:108489. [PMID: 32492479 DOI: 10.1016/j.clim.2020.108489] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 02/12/2020] [Accepted: 05/29/2020] [Indexed: 11/20/2022]
Abstract
Acute myelogenous leukemia (AML) is an aggressive hematological malignancy associated with high rates of mortality. This incidence is due to the complexity in which the AML cells interact with other healthy human cells. These phenomena create an environment that favors the expansion of leukemic cells, which will affect the patient's prognosis. An important aspect is the ability of AML cells to evade immune responses via targeting and signaling immune cells to suppress anti-tumor responses. Many studies have reported that associations among components in the peripheral bloodstream might modulate leukemic progression because AML survival is a fundamental step for recolonizing bone marrow after allogeneic hematopoietic stem cell (HSC) transplantation or chemotherapy. Therefore, we collected the most important data about components that circulate with leukemic blasts and contribute to their survival and proliferation. We also discuss clinical approaches that could be conducted to more effectively treat the disease.
Collapse
|
45
|
Hutchings G, Janowicz K, Moncrieff L, Dompe C, Strauss E, Kocherova I, Nawrocki MJ, Kruszyna Ł, Wąsiatycz G, Antosik P, Shibli JA, Mozdziak P, Perek B, Krasiński Z, Kempisty B, Nowicki M. The Proliferation and Differentiation of Adipose-Derived Stem Cells in Neovascularization and Angiogenesis. Int J Mol Sci 2020; 21:ijms21113790. [PMID: 32471255 PMCID: PMC7312564 DOI: 10.3390/ijms21113790] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022] Open
Abstract
Neovascularization and angiogenesis are vital processes in the repair of damaged tissue, creating new blood vessel networks and increasing oxygen and nutrient supply for regeneration. The importance of Adipose-derived Mesenchymal Stem Cells (ASCs) contained in the adipose tissue surrounding blood vessel networks to these processes remains unknown and the exact mechanisms responsible for directing adipogenic cell fate remain to be discovered. As adipose tissue contains a heterogenous population of partially differentiated cells of adipocyte lineage; tissue repair, angiogenesis and neovascularization may be closely linked to the function of ASCs in a complex relationship. This review aims to investigate the link between ASCs and angiogenesis/neovascularization, with references to current studies. The molecular mechanisms of these processes, as well as ASC differentiation and proliferation are described in detail. ASCs may differentiate into endothelial cells during neovascularization; however, recent clinical trials have suggested that ASCs may also stimulate angiogenesis and neovascularization indirectly through the release of paracrine factors.
Collapse
Affiliation(s)
- Greg Hutchings
- The School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (G.H.); (K.J.); (L.M.)
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (I.K.); (M.J.N.); (B.K.)
| | - Krzysztof Janowicz
- The School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (G.H.); (K.J.); (L.M.)
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (I.K.); (M.J.N.); (B.K.)
| | - Lisa Moncrieff
- The School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (G.H.); (K.J.); (L.M.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
| | - Claudia Dompe
- The School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (G.H.); (K.J.); (L.M.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
- Correspondence:
| | - Ewa Strauss
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland;
- Department of Vascular, Endovascular Surgery, Angiology and Phlebology Poznan University of Medical Sciences, 61-701 Poznan, Poland; (L.K.); (Z.K.)
| | - Ievgeniia Kocherova
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (I.K.); (M.J.N.); (B.K.)
| | - Mariusz J. Nawrocki
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (I.K.); (M.J.N.); (B.K.)
| | - Łukasz Kruszyna
- Department of Vascular, Endovascular Surgery, Angiology and Phlebology Poznan University of Medical Sciences, 61-701 Poznan, Poland; (L.K.); (Z.K.)
| | - Grzegorz Wąsiatycz
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland; (G.W.); (P.A.)
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland; (G.W.); (P.A.)
| | - Jamil A. Shibli
- Department of Periodontology and Oral Implantology, Dental Research Division, University of Guarulhos, São Paulo 07023-070, Brazil;
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Raleigh, NC 27695, USA;
| | - Bartłomiej Perek
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, 61-848 Poznań, Poland;
| | - Zbigniew Krasiński
- Department of Vascular, Endovascular Surgery, Angiology and Phlebology Poznan University of Medical Sciences, 61-701 Poznan, Poland; (L.K.); (Z.K.)
| | - Bartosz Kempisty
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (I.K.); (M.J.N.); (B.K.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland; (G.W.); (P.A.)
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 601 77 Brno, Czech Republic
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
| |
Collapse
|
46
|
Bhuniya A, Guha I, Ganguly N, Saha A, Dasgupta S, Nandi P, Das A, Ghosh S, Ghosh T, Haque E, Banerjee S, Bose A, Baral R. NLGP Attenuates Murine Melanoma and Carcinoma Metastasis by Modulating Cytotoxic CD8 + T Cells. Front Oncol 2020; 10:201. [PMID: 32211313 PMCID: PMC7076076 DOI: 10.3389/fonc.2020.00201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Neem leaf glycoprotein (NLGP), a natural immunomodulator, attenuates murine carcinoma and melanoma metastasis, independent of primary tumor growth and alterations in basic cellular properties (cell proliferation, cytokine secretion, etc.). Colonization event of invasion–metastasis cascade was primarily inhibited by NLGP, with no effect on metastasis-related invasion, migration, and extravasation. High infiltration of interferon γ (IFN-γ)–secreting cytotoxic CD8+ T cells [CD44+, CD69+, GranB+, IFN-γ+, and interleukin 2+] was documented in the metastatic site of NLGP-treated mice. Systemic CD8+ T cell depletion abolished NLGP-mediated metastasis inhibition and reappeared upon adoptive transfer of NLGP-activated CD8+ T cells. Interferon γ-secreting from CD8+ T cells inhibit the expression of angiogenesis regulatory vascular endothelial growth factor and transforming growth factor β and have an impact on the prevention of colonization. Neem leaf glycoprotein modulates dendritic cells (DCs) for proper antigen presentation by its DC surface binding and upregulation of MHC-I/II, CD86, and CCR7. Neem leaf glycoprotein–treated DCs specifically imprint CXCR3 and CCR4 homing receptors on activated CD8+ T cells, which helps to infiltrate into metastatic sites to restrain colonization. Such NLGP's effect on DCs is translation dependent and transcription independent. Studies using ovalbumin, OVA257−264, and crude B16F10 antigen indicate MHC-I upregulation depends on the quantity of proteasome degradable peptide and only stimulates CD8+ T cells in the presence of antigen. Overall data suggest NLGP inhibits metastasis, in conjunction with tumor growth restriction, and thus might appear as a promising next-generation cancer immunotherapeutic.
Collapse
Affiliation(s)
- Avishek Bhuniya
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Ipsita Guha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Nilanjan Ganguly
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Akata Saha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Shayani Dasgupta
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Partha Nandi
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Arnab Das
- RNA Biology and Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sarbari Ghosh
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Tithi Ghosh
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Enamul Haque
- Department of Zoology, Barasat Government College, Barasat, India
| | - Saptak Banerjee
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Anamika Bose
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| |
Collapse
|
47
|
Takeshita T, Yan L, Asaoka M, Rashid O, Takabe K. Late recurrence of breast cancer is associated with pro-cancerous immune microenvironment in the primary tumor. Sci Rep 2019; 9:16942. [PMID: 31729458 PMCID: PMC6858361 DOI: 10.1038/s41598-019-53482-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
The fact that 20–40% of all breast cancer (BC) patients develop recurrence when 5 year survival is 90% strongly suggests that late recurrence, i.e. more than 5 years after diagnosis, is the remaining challenge to decrease the absolute number of BC deaths. Better understanding late recurrence is an essential first step to address this issue. We hypothesized that primary tumors with a distinctive tumor immune microenvironment will develop late recurrence. Accordingly, we evaluated the relationship between the timing of cancer recurrence, clinical factors, gene expression profiles, and immune status utilizing two published large cohorts. 308 primary BCs in TCGA were analyzed and categorized as: recurrence ≤2 years (Early, n = 49), between 2–5 years (Mid, n = 54), recurrence >5 years (Late, n = 20), and no recurrence >5 years (Survivors, n = 185). 1,727 primary BCs in METABRIC were analyzed and categorized similarly: Early, n = 170; distant (D), n = 19; local (L), Mid, n = 213; D, n = 21; L, Late, n = 199; D, n = 57, L, and Survivors, n = 1048. Utilizing pre-ranked GSEA, we showed that primary tumors with Survivors were associated with anti-cancer signaling such as INF-α/-γ response and TNF-α signaling, compared with all recurrence groups in pre-ranked GSEA. Furtherrmore, we found that host defense immunity (leukocyte fraction, lymphocyte infiltration, and macrophage fractions) was decreased in primary tumors with Late recurrence compared with Survivors. Utilizing the CIBERSORT algorithm, we showed anti-cancer lymphocytes, memory CD4+ T cells and γδT cells, were significantly lower, and pro-cancerous regulatory T cells were significantly higher in Late tumors compared with Survivors. In agreement, cytolytic activity score that assesses immune cell cytolytic activity was significantly lower in Late compared with Survivors. We demonstrated that not only host defense immunity, but also pro-cancerous immune cells and immune cell cytolytic activity in primary BC was associated with late recurrence.
Collapse
Affiliation(s)
- Takashi Takeshita
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mariko Asaoka
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Omar Rashid
- Holy Cross Hospital, Trinity Health, Fort Lauderdale, FL, USA.,Massachusetts General Hospital, Boston, MA, USA.,University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kazuaki Takabe
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA. .,Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, the State University of New York, Buffalo, NY, USA. .,Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan. .,Department of Surgery, Yokohama City University, Yokohama, Japan. .,Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan. .,Department of Breast Surgery, Fukushima Medical University, Fukushima, Japan.
| |
Collapse
|
48
|
Takeshita T, Yan L, Asaoka M, Rashid O, Takabe K. Late recurrence of breast cancer is associated with pro-cancerous immune microenvironment in the primary tumor. Sci Rep 2019. [PMID: 31729458 DOI: 10.1038/s41598‐019‐53482‐x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The fact that 20-40% of all breast cancer (BC) patients develop recurrence when 5 year survival is 90% strongly suggests that late recurrence, i.e. more than 5 years after diagnosis, is the remaining challenge to decrease the absolute number of BC deaths. Better understanding late recurrence is an essential first step to address this issue. We hypothesized that primary tumors with a distinctive tumor immune microenvironment will develop late recurrence. Accordingly, we evaluated the relationship between the timing of cancer recurrence, clinical factors, gene expression profiles, and immune status utilizing two published large cohorts. 308 primary BCs in TCGA were analyzed and categorized as: recurrence ≤2 years (Early, n = 49), between 2-5 years (Mid, n = 54), recurrence >5 years (Late, n = 20), and no recurrence >5 years (Survivors, n = 185). 1,727 primary BCs in METABRIC were analyzed and categorized similarly: Early, n = 170; distant (D), n = 19; local (L), Mid, n = 213; D, n = 21; L, Late, n = 199; D, n = 57, L, and Survivors, n = 1048. Utilizing pre-ranked GSEA, we showed that primary tumors with Survivors were associated with anti-cancer signaling such as INF-α/-γ response and TNF-α signaling, compared with all recurrence groups in pre-ranked GSEA. Furtherrmore, we found that host defense immunity (leukocyte fraction, lymphocyte infiltration, and macrophage fractions) was decreased in primary tumors with Late recurrence compared with Survivors. Utilizing the CIBERSORT algorithm, we showed anti-cancer lymphocytes, memory CD4+ T cells and γδT cells, were significantly lower, and pro-cancerous regulatory T cells were significantly higher in Late tumors compared with Survivors. In agreement, cytolytic activity score that assesses immune cell cytolytic activity was significantly lower in Late compared with Survivors. We demonstrated that not only host defense immunity, but also pro-cancerous immune cells and immune cell cytolytic activity in primary BC was associated with late recurrence.
Collapse
Affiliation(s)
- Takashi Takeshita
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mariko Asaoka
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Omar Rashid
- Holy Cross Hospital, Trinity Health, Fort Lauderdale, FL, USA.,Massachusetts General Hospital, Boston, MA, USA.,University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kazuaki Takabe
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA. .,Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, the State University of New York, Buffalo, NY, USA. .,Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan. .,Department of Surgery, Yokohama City University, Yokohama, Japan. .,Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan. .,Department of Breast Surgery, Fukushima Medical University, Fukushima, Japan.
| |
Collapse
|
49
|
Zheng W, Zhang H, Zhao D, Zhang J, Pollard JW. Lung Mammary Metastases but Not Primary Tumors Induce Accumulation of Atypical Large Platelets and Their Chemokine Expression. Cell Rep 2019; 29:1747-1755.e4. [PMID: 31722193 PMCID: PMC6919330 DOI: 10.1016/j.celrep.2019.10.016] [Citation(s) in RCA: 6] [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: 12/21/2018] [Revised: 09/12/2019] [Accepted: 10/03/2019] [Indexed: 01/11/2023] Open
Abstract
The tumor microenvironment (TME) at the metastatic site consists of multiple components with considerable cellular heterogeneity. To test whether endothelial cells (ECs) associated with lung metastases express a distinct gene expression program that promotes metastatic growth, we isolated CD31+/CD45- cells from lung mammary cancer metastases for RNA sequencing and found CD44 upregulation. Unexpectedly, the CD44+ subset did not comprise authentic ECs nor were they bone-marrow-derived CD45- endothelial progenitor cells. Instead, they were a population of large platelets that are distinct from regular small platelets. These CD44+ large platelets were enriched in lung metastases but not primary mammary tumors and upregulated myeloid cell-regulating chemokines indicative of potential regulation of metastasis via indirect mechanisms. Identification of this cellular player in the TME of metastasis suggests a role for the recently identified lung-resident megakaryocytes (MKs) and offers an unexplored route to discover novel mechanisms and an opportunity for therapeutic interventions.
Collapse
Affiliation(s)
- Wei Zheng
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA.
| | - Hui Zhang
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA
| | - Dejian Zhao
- Yale Center for Genome Analysis, Yale University, New Haven, CT 06510, USA
| | - Jinghang Zhang
- Department of Microbiology & Immunology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA
| | - Jeffrey W Pollard
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA; MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
50
|
Zhou G, Han X, Wu Z, Shi Q, Bao X. Rosiglitazone accelerates wound healing by improving endothelial precursor cell function and angiogenesis in db/db mice. PeerJ 2019; 7:e7815. [PMID: 31637120 PMCID: PMC6800979 DOI: 10.7717/peerj.7815] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/02/2019] [Indexed: 01/18/2023] Open
Abstract
Background & Aims Endothelial precursor cell (EPC) dysfunction is one of the risk factors for diabetes mellitus (DM) which results in delayed wound healing. Rosiglitazone (RSG) is a frequently prescribed oral glucose-lowering drug. Previous studies have shown the positive effects of RSG on ameliorating EPC dysfunction in diabetic patients. Interestingly, knowledge about RSG with regard to the wound healing process caused by DM is scarce. Therefore, in this study, we investigated the possible actions of RSG on wound healing and the related mechanisms involved in db/db diabetic mice. Methods Db/db mice with spontaneous glucose metabolic disorder were used as a type 2 DM model. RSG (20 mg/kg/d, i.g.,) was administered for 4 weeks before wound creation and bone marrow derived EPC (BM-EPC) isolation. Wound closure was assessed by wound area and CD31 staining. Tubule formation and migration assays were used to judge the function of the BM-EPCs. The level of vascular endothelial growth factor (VEGF), stromal cell derived factor-1α (SDF-1α) and insulin signaling was determined by ELISA. Cell viability of the BM-EPCs was measured by CCK-8 assay. Results RSG significantly accelerated wound healing and improved angiogenesis in db/db mice. Bioactivities of tube formation and migration were decreased in db/db mice but were elevated by RSG. Level of both VEGF and SDF-1α was increased by RSG in the BM-EPCs of db/db mice. Insulin signaling was elevated by RSG reflected in the phosphorylated-to-total AKT in the BM-EPCs. In vitro, RSG improved impaired cell viability and tube formation of BM-EPCs induced by high glucose, but this was prevented by the VEGF inhibitor avastin. Conclusion Our data demonstrates that RSG has benefits for wound healing and angiogenesis in diabetic mice, and was partially associated with improvement of EPC function through activation of VEGF and stimulation of SDF-1α in db/db mice.
Collapse
Affiliation(s)
- Guoliang Zhou
- Department of Pharmacy, School of Life and Health Sciences, Anhui Science and Technology University, Fengyang, Anhui, China
| | - Xue Han
- Laboratory Animal Center, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang Province, China
| | - Zhiheng Wu
- School of Clinical Medicine, Wannan Medicial Colledge, Wuhu, Anhui, China
| | - Qiaojuan Shi
- Laboratory Animal Center, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang Province, China
| | - Xiaogang Bao
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|