101
|
Wang H, Pan J, Barsky L, Jacob JC, Zheng Y, Gao C, Wang S, Zhu W, Sun H, Lu L, Jia H, Zhao Y, Bruns C, Vago R, Dong Q, Qin L. Characteristics of pre-metastatic niche: the landscape of molecular and cellular pathways. MOLECULAR BIOMEDICINE 2021; 2:3. [PMID: 35006432 PMCID: PMC8607426 DOI: 10.1186/s43556-020-00022-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/30/2020] [Indexed: 02/08/2023] Open
Abstract
Metastasis is a major contributor to cancer-associated deaths. It involves complex interactions between primary tumorigenic sites and future metastatic sites. Accumulation studies have revealed that tumour metastasis is not a disorderly spontaneous incident but the climax of a series of sequential and dynamic events including the development of a pre-metastatic niche (PMN) suitable for a subpopulation of tumour cells to colonize and develop into metastases. A deep understanding of the formation, characteristics and function of the PMN is required for developing new therapeutic strategies to treat tumour patients. It is rapidly becoming evident that therapies targeting PMN may be successful in averting tumour metastasis at an early stage. This review highlights the key components and main characteristics of the PMN and describes potential therapeutic strategies, providing a promising foundation for future studies.
Collapse
Affiliation(s)
- Hao Wang
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Junjie Pan
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Livnat Barsky
- Avram and Stella Goldstein-Goren, Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Yan Zheng
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Chao Gao
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Shun Wang
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Wenwei Zhu
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Haoting Sun
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Lu Lu
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Huliang Jia
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Yue Zhao
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Cologne, Germany
| | - Razi Vago
- Avram and Stella Goldstein-Goren, Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Qiongzhu Dong
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China.
| | - Lunxiu Qin
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China.
| |
Collapse
|
102
|
Wang X, Guo J, Yu P, Guo L, Mao X, Wang J, Miao S, Sun J. The roles of extracellular vesicles in the development, microenvironment, anticancer drug resistance, and therapy of head and neck squamous cell carcinoma. J Exp Clin Cancer Res 2021; 40:35. [PMID: 33478586 PMCID: PMC7819156 DOI: 10.1186/s13046-021-01840-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the main malignant tumours affecting human health, mainly due to delayed diagnosis and high invasiveness. Extracellular vehicles (EVs) are membranous vesicles released by cells into the extracellular matrix that carry important signalling molecules and stably and widely exist in various body fluids, such as plasma, saliva, cerebrospinal fluid, breast milk, urine, semen, lymphatic fluid, synovial fluid, amniotic fluid, and sputum. EVs transport almost all types of bioactive molecules (DNA, mRNAs, microRNAs (miRNAs), proteins, metabolites, and even pharmacological compounds). These "cargoes" can act on recipient cells, reshaping the surrounding microenvironment and altering distant targets, ultimately affecting their biological behaviour. The extensive exploration of EVs has deepened our comprehensive understanding of HNSCC biology. In this review, we not only summarized the effect of HNSCC-derived EVs on the tumour microenvironment but also described the role of microenvironment-derived EVs in HNSCC and discussed how the "mutual dialogue" between the tumour and microenvironment mediates the growth, metastasis, angiogenesis, immune escape, and drug resistance of tumours. Finally, the clinical application of EVS in HNSCC was assessed.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Head and Neck Tumors, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, 150000, Harbin, Heilongjiang, People's Republic of China
| | - Junnan Guo
- The First Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, 150000, Harbin, Heilongjiang, People's Republic of China
| | - Pingyang Yu
- Department of Head and Neck Tumors, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, 150000, Harbin, Heilongjiang, People's Republic of China
| | - Lunhua Guo
- Department of Head and Neck Tumors, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, 150000, Harbin, Heilongjiang, People's Republic of China
| | - Xionghui Mao
- Department of Head and Neck Tumors, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, 150000, Harbin, Heilongjiang, People's Republic of China
| | - Junrong Wang
- Department of Head and Neck Tumors, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, 150000, Harbin, Heilongjiang, People's Republic of China
| | - Susheng Miao
- Department of Head and Neck Tumors, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, 150000, Harbin, Heilongjiang, People's Republic of China.
| | - Ji Sun
- Department of Head and Neck Tumors, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, 150000, Harbin, Heilongjiang, People's Republic of China.
| |
Collapse
|
103
|
Ritchie S, Reed DA, Pereira BA, Timpson P. The cancer cell secretome drives cooperative manipulation of the tumour microenvironment to accelerate tumourigenesis. Fac Rev 2021; 10:4. [PMID: 33659922 PMCID: PMC7894270 DOI: 10.12703/r/10-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cellular secretions are a fundamental aspect of cell-cell and cell-matrix interactions in vivo. In malignancy, cancer cells have an aberrant secretome compared to their non-malignant counterparts, termed the "cancer cell secretome". The cancer cell secretome can influence every stage of the tumourigenic cascade. At the primary site, cancer cells can secrete a multitude of factors that facilitate invasion into surrounding tissue, allowing interaction with the local tumour microenvironment (TME), driving tumour development and progression. In more advanced disease, the cancer cell secretome can be involved in extravasation and metastasis, including metastatic organotropism, pre-metastatic niche (PMN) preparation, and metastatic outgrowth. In this review, we will explore the latest advances in the field of cancer cell secretions, including its dynamic and complex role in activating the TME and potentiating invasion and metastasis, with comments on how these secretions may also promote therapy resistance.
Collapse
Affiliation(s)
- Shona Ritchie
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Daniel A Reed
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Brooke A Pereira
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Paul Timpson
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| |
Collapse
|
104
|
Role of myeloid-derived suppressor cells in metastasis. Cancer Metastasis Rev 2021; 40:391-411. [PMID: 33411082 DOI: 10.1007/s10555-020-09947-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023]
Abstract
The spread of primary tumor cells to distant organs, termed metastasis, is the principal cause of cancer mortality and is a critical therapeutic target in oncology. Thus, a better understanding of metastatic progression is critical for improved therapeutic approaches requiring insight into the timing of tumor cell dissemination and seeding of distant organs, which can lead to the formation of occult lesions. However, due to limitations in imaging techniques, primary tumors can only be detected when they reach a relatively large size (e.g., > 1 cm3), which, based on our understanding of tumor evolution, is 10 to 20 years (30 doubling times) following tumor initiation. Recent insights into the timing of metastasis are based on the genomic profiling of paired primary tumors and metastases, suggesting that tumor cell seeding of secondary sites occurs early during tumor progression and years prior to diagnosis. Following seeding, tumor cells may remain in a dormant state as single cells or micrometastases before emerging as overt lesions. This timeline and the role of metastatic dormancy are regulated by interactions between the tumor, its microenvironment, and tumor-specific T cell responses. An improved understanding of the mechanisms and interactions responsible for immune evasion and tumor cell release from dormancy would support the development of novel targeted therapeutics. We posit herein that the immunosuppressive mechanisms mediated by myeloid-derived suppressor cells (MDSCs) are a major contributor to tumor progression, and that these mechanisms promote tumor cell escape from dormancy. Thus, while extensive studies have demonstrated a role for MDSCs in the escape from adoptive and innate immune responses (T-, natural killer (NK)-, and B cell responses), facilitating tumor progression and metastasis, few studies have considered their role in dormancy. In this review, we discuss the role of MDSC expansion, driven by tumor burden, and its role in escape from dormancy, resulting in occult metastases, and the potential for MDSC inhibition as an approach to prolong the survival of patients with advanced malignancies.
Collapse
|
105
|
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
|
106
|
Wang N, Wang S, Wang X, Zheng Y, Yang B, Zhang J, Pan B, Gao J, Wang Z. Research trends in pharmacological modulation of tumor-associated macrophages. Clin Transl Med 2021; 11:e288. [PMID: 33463063 PMCID: PMC7805405 DOI: 10.1002/ctm2.288] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
As one of the most abundant immune cell populations in the tumor microenvironment (TME), tumor-associated macrophages (TAMs) play important roles in multiple solid malignancies, including breast cancer, prostate cancer, liver cancer, lung cancer, ovarian cancer, gastric cancer, pancreatic cancer, and colorectal cancer. TAMs could contribute to carcinogenesis, neoangiogenesis, immune-suppressive TME remodeling, cancer chemoresistance, recurrence, and metastasis. Therefore, reprogramming of the immune-suppressive TAMs by pharmacological approaches has attracted considerable research attention in recent years. In this review, the promising pharmaceutical targets, as well as the existing modulatory strategies of TAMs were summarized. The chemokine-chemokine receptor signaling, tyrosine kinase receptor signaling, metabolic signaling, and exosomal signaling have been highlighted in determining the biological functions of TAMs. Besides, both preclinical research and clinical trials have suggested the chemokine-chemokine receptor blockers, tyrosine kinase inhibitors, bisphosphonates, as well as the exosomal or nanoparticle-based targeting delivery systems as the promising pharmacological approaches for TAMs deletion or reprogramming. Lastly, the combined therapies of TAMs-targeting strategies with traditional treatments or immunotherapies as well as the exosome-like nanovesicles for cancer therapy are prospected.
Collapse
Affiliation(s)
- Neng Wang
- The Research Center for Integrative MedicineSchool of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Shengqi Wang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Xuan Wang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Yifeng Zheng
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Bowen Yang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Juping Zhang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Bo Pan
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Jianli Gao
- Academy of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Zhiyu Wang
- The Research Center for Integrative MedicineSchool of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| |
Collapse
|
107
|
Butler G, Keeton SJ, Johnson LJ, Dash PR. A phenotypic switch in the dispersal strategy of breast cancer cells selected for metastatic colonization. Proc Biol Sci 2020; 287:20202523. [PMID: 33259764 DOI: 10.1098/rspb.2020.2523] [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] [Indexed: 12/12/2022] Open
Abstract
An important question in cancer evolution concerns which traits make a cell likely to successfully metastasize. Cell motility phenotypes, mediated by cell shape change, are strong candidates. We experimentally evolved breast cancer cells in vitro for metastatic capability, using selective regimes designed to simulate stages of metastasis, then quantified their motility behaviours using computer vision. All evolved lines showed changes to motility phenotypes, and we have identified a previously unknown density-dependent motility phenotype only seen in cells selected for colonization of decellularized lung tissue. These cells increase their rate of morphological change with an increase in migration speed when local cell density is high. However, when the local cell density is low, we find the opposite relationship: the rate of morphological change decreases with an increase in migration speed. Neither the ancestral population, nor cells selected for their ability to escape or invade extracellular matrix-like environments, displays this dynamic behavioural switch. Our results suggest that cells capable of distant-site colonization may be characterized by dynamic morphological phenotypes and the capacity to respond to the local social environment.
Collapse
Affiliation(s)
- George Butler
- School of Biological Sciences, University of Reading, Reading, UK
| | - Shirley J Keeton
- School of Biological Sciences, University of Reading, Reading, UK
| | - Louise J Johnson
- School of Biological Sciences, University of Reading, Reading, UK
| | - Philip R Dash
- School of Biological Sciences, University of Reading, Reading, UK
| |
Collapse
|
108
|
Möller A, Lobb RJ. The evolving translational potential of small extracellular vesicles in cancer. Nat Rev Cancer 2020; 20:697-709. [PMID: 32958932 DOI: 10.1038/s41568-020-00299-w] [Citation(s) in RCA: 336] [Impact Index Per Article: 67.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
Cancer-derived extracellular vesicles (EVs) are regarded as having promising potential to be used as therapeutics and disease biomarkers. Mechanistically, EVs have been shown to function in most, if not all, steps of cancer progression. Cancer EVs, including small EVs (sEVs), contain unique biomolecular cargo, consisting of protein, nucleic acid and lipids. Through progress in the identification of this specific cargo, cancer biomarkers have been identified and developed, opening up novel and interesting opportunities for cancer diagnosis and prognosis. Intriguingly, we still lack a comprehensive understanding of the cancer-specific pathways that govern EV biogenesis in cancer cells. Filling this knowledge gap will rapidly improve cancer EV biomarkers, as it will also allow discrimination of the procancer and anticancer actions of those EVs. Even more promising is uncovering therapeutically targetable, tumour-specific EV pathways and content, which will generate novel classes of cancer therapies. This Review highlights the progress the cancer sEV field has made in the areas of biomarker discovery and validation as well as sEV-based therapeutics, highlights the challenges we are facing and identifies gaps in our knowledge, which currently prevent us from developing the full potential of sEVs in cancer diagnostic and therapy.
Collapse
Affiliation(s)
- Andreas Möller
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.
| | - Richard J Lobb
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia
| |
Collapse
|
109
|
He X, Zhong X, Hu Z, Zhao S, Wei P, Li D. An insight into small extracellular vesicles: Their roles in colorectal cancer progression and potential clinical applications. Clin Transl Med 2020; 10:e249. [PMID: 33377655 PMCID: PMC7733319 DOI: 10.1002/ctm2.249] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers and a leading cause of mortality worldwide. Small extracellular vesicles (sEVs) are nano-sized extracellular vesicles containing a variety of bioactive molecules, such as nucleic acids, proteins, lipids, and metabolites. Recent evidence from CRC has revealed that sEVs contribute to tumorigenesis, progression, and drug resistance, and serve as a tool for "liquid biopsy" and a drug delivery system for therapy. In this review, we summarize information about the roles of sEVs in the proliferation, invasion, migration, epithelial-mesenchymal transition, formation of the premetastatic niche, and drug resistance to elucidate the mechanisms governing sEVs in CRC and to identify novel targets for therapy and prognostic and diagnostic biomarkers.
Collapse
Affiliation(s)
- Xuefeng He
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xinyang Zhong
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zijuan Hu
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
- Institute of PathologyFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Senlin Zhao
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Ping Wei
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
- Institute of PathologyFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Dawei Li
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
110
|
Li R, Qi Y, Han M, Geng B, Wang G, Han M. Computed tomography reveals microenvironment changes in premetastatic lung. Eur Radiol 2020; 31:4340-4349. [PMID: 33219849 DOI: 10.1007/s00330-020-07500-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 10/03/2020] [Accepted: 11/11/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Microenvironment changes had occurred in the metastatic organs before the arriving of the metastatic tumor cells. In this study, we evaluated the effectiveness of computed tomography (CT) images in quantifying the microenvironment changes in the premetastatic lung under both laboratory and clinical conditions. METHOD Free-breathing Balb/c mice underwent micro-CT repeatedly after the implantation of 4T1 breast tumor. CT-derived indicators (aerated lung volume, lung tissue volume, total lung volume, mean lung density, and the ratio of aerated lung volume to the total lung volume) were quantified. Hematoxylin-eosin staining was used to display the microenvironment changes in premetastatic lung. Moreover, we examined healthy adult women, adult women with histopathologically confirmed primary breast cancer, and adult women with histopathologically confirmed primary breast cancer and lung metastases in our institution to test whether the indicators derived from lung CT images changed with the growth of breast cancer. RESULTS In 4T1 tumor-bearing mice, lung density is increased before lung masses can be recognized on CT images and is correlated with the severity of inflammation in the lung microenvironment. In primary breast tumor-bearing patients, lung density is also increased before the clinical diagnosis of pulmonary metastasis and is correlated with disease score, which represents tumor progression. CONCLUSIONS CT is a reliable and quantitative tool that yields dynamic information on metastatic processes. Microenvironmental changes had occurred in patients' lung tissue before the clinical diagnosis of pulmonary metastasis. Our research will provide new insight for clinical research on the premetastatic niche. KEY POINTS • CT, which provides dynamic information on metastatic processes, is a reliable and quantitative tool to bridge laboratory and clinical studies of the premetastatic niche. • We confirmed that microenvironmental changes occurred in patients' lung tissue before clinicians could diagnose pulmonary metastasis. • Our results provide evidence for the study of the premetastatic niche by analyzing information obtained from CT images.
Collapse
Affiliation(s)
- Ranran Li
- Cancer Therapy and Research Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, People's Republic of China
| | - Yana Qi
- Cancer Therapy and Research Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, People's Republic of China
| | - Meng Han
- School of Basic Medical Sciences, Shandong First Medical University, Jinan, People's Republic of China
| | - Baocheng Geng
- Cancer Therapy and Research Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, People's Republic of China
| | - Guangyu Wang
- Cancer Therapy and Research Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, People's Republic of China
| | - Mingyong Han
- Cancer Therapy and Research Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, People's Republic of China.
| |
Collapse
|
111
|
Abstract
The spine is a frequent location for metastatic disease. As local control of primary tumor pathology continues to improve, survival rates improve and, by extension, the opportunity for metastasis increases. Breast, lung, and prostate cancer are the leading contributors to spinal metastases. Spinal metastases can manifest as bone pain, pathologic fractures, spinal instability, nerve root compression, and, in its most severe form, spinal cord compression. The global extent of disease, the spinal burden, neurologic status, and life expectancy help to categorize patients as to their candidacy for treatment options. Efficient identification and workup of those with spinal metastases will expedite the treatment cascade and improve quality of life.
Collapse
Affiliation(s)
- Joshua T Wewel
- Atlanta Brain and Spine Care, Piedmont Healthcare, Atlanta, Georgia
| | - John E O'Toole
- Department of Neurosurgery, University Medical Center, Chicago, Illinois, US
| |
Collapse
|
112
|
Hurtado P, Martínez-Pena I, Piñeiro R. Dangerous Liaisons: Circulating Tumor Cells (CTCs) and Cancer-Associated Fibroblasts (CAFs). Cancers (Basel) 2020; 12:E2861. [PMID: 33027902 PMCID: PMC7599894 DOI: 10.3390/cancers12102861] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/02/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023] Open
Abstract
The crosstalk between cancer cells and the tumor microenvironment (TME) is a key determinant of cancer metastasis. Cancer-associated fibroblasts (CAFs), one of the main cellular components of TME, promote cancer cell invasion and dissemination through mechanisms including cell-cell interactions and the paracrine secretion of growth factors, cytokines and chemokines. During metastasis, circulating tumor cells (CTCs) are shed from the primary tumor to the bloodstream, where they can be detected as single cells or clusters. The current knowledge about the biology of CTC clusters positions them as key actors in metastasis formation. It also indicates that CTCs do not act alone and that they may be aided by stromal and immune cells, which seem to shape their metastatic potential. Among these cells, CAFs are found associated with CTCs in heterotypic CTC clusters, and their presence seems to increase their metastatic efficiency. In this review, we summarize the current knowledge on the role that CAFs play on metastasis and we discuss their implication on the biogenesis, metastasis-initiating capacity of CTC clusters, and clinical implications. Moreover, we speculate about possible therapeutic strategies aimed to limit the metastatic potential of CTC clusters involving the targeting of CAFs as well as their difficulties and limitations.
Collapse
Affiliation(s)
- Pablo Hurtado
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (P.H.); (I.M.-P.)
| | - Inés Martínez-Pena
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (P.H.); (I.M.-P.)
| | - Roberto Piñeiro
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (P.H.); (I.M.-P.)
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, 28029 Madrid, Spain
| |
Collapse
|
113
|
Wang D, Xiao F, Feng Z, Li M, Kong L, Huang L, Wei Y, Li H, Liu F, Zhang H, Zhang W. Sunitinib facilitates metastatic breast cancer spreading by inducing endothelial cell senescence. Breast Cancer Res 2020; 22:103. [PMID: 32993785 PMCID: PMC7526390 DOI: 10.1186/s13058-020-01346-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/21/2020] [Indexed: 02/08/2023] Open
Abstract
Background Sunitinib, a receptor tyrosine kinase (RTK) inhibitor that targets multiple receptors such as vascular endothelial growth factor receptors (VEGFRs), was approved for cancer treatment in 2006. However, it was unsuccessful in treating certain cancers, particularly metastatic breast cancer (MBC), and the mechanism underlying this “sunitinib resistance” remains unclear. Herein, we investigated whether the sunitinib-associated inferior survival benefit in MBC was due to sunitinib-induced endothelial cell (EC) injury or EC senescence. Methods 4T1 murine breast cancer cells were used as the main breast tumor model for it produces a highly metastatic solid tumor that can spontaneously metastasize to the lung, which closely mimics highly metastatic human breast cancer. Senescence-associated β-galactosidase (SA-β-Gal, immunohistochemistry [IHC]-staining), P16, P53, and P57 (immunoblotting) were used as markers of cell senescence. A protein array containing 25 senescence-associated chemokines and the transwell chemotaxis assay were used to examine whether sunitinib increases inflammatory chemokine secretion which attracts tumor cells via chemokinesis. Flow cytometry and IHC were used to detect whether the sunitinib-induced senescent ECs recruit cancer-associated inflammatory myeloid cells. Finally, the spontaneous metastatic model was used to monitor whether sunitinib causes the formation of “pre-metastatic niche” which promotes MBC to metastasize to the lungs. Results We demonstrated that sunitinib induced a senescence-like endothelial cell (EC) phenotype. Inflammatory chemokine secretion and VCAM1 expression were significantly increased in senescent ECs, resulting in tumor cell (TC) chemotaxis and TC/EC interactions. Meanwhile, EC senescence caused loosening of EC junctions, facilitating TC transmigration through the endothelial barrier. Sunitinib-induced senescent ECs also recruited cancer-associated myeloid cells to form a “pre-metastatic niche”-like microenvironment. Alterations at the molecular level and in the tissue environment ultimately led to an increase in distant metastasis. Conclusion Although sunitinib was designed to target the EC directly, the increase in tumor metastasis may ironically be due to sunitinib “correctly” playing its role. Our findings suggest that we should carefully weigh the pros and cons before using sunitinib and other antiangiogenic drugs that directly target the ECs.
Collapse
Affiliation(s)
- Denian Wang
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 1, Ke Yuan 4th Road, Gao Peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Fei Xiao
- Department of Intensive Care Unit of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhongxue Feng
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 1, Ke Yuan 4th Road, Gao Peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Min Li
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 1, Ke Yuan 4th Road, Gao Peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Lingmiao Kong
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 1, Ke Yuan 4th Road, Gao Peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Luping Huang
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 1, Ke Yuan 4th Road, Gao Peng Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yong'gang Wei
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hongyu Li
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Fei Liu
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Haili Zhang
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Wei Zhang
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 1, Ke Yuan 4th Road, Gao Peng Street, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
114
|
Deng L, Li T, Liao Y, Liu S, Xie Z, Huang Z, Dai H, Li J, Lei X. Peritumoral activated hepatic stellate cells are associated with hepatic recurrence for resectable colorectal adenocarcinoma liver metastasis following resection. Oncol Lett 2020; 20:287. [PMID: 33014165 PMCID: PMC7520724 DOI: 10.3892/ol.2020.12150] [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: 11/05/2019] [Accepted: 07/14/2020] [Indexed: 12/24/2022] Open
Abstract
The formation of the pre-metastatic niche (PMN), which precedes the establishment of tumor lesions, plays a critical role in cancer recurrence and metastasis. Hepatic stellate cells (HSCs), a critical liver stromal cell component, can be induced to facilitate metastasis by modeling liver PMN formation. In the present study, activated HSCs were observed in the peritumor non-cancerous liver tissues (PNLT) colorectal adenocarcinoma liver metastasis (CRALM), and the density of activated HSCs was higher in PNLT compared with that in normal liver tissues (NLT). High density of activated HSC in the PNLT was positively associated with the number of tumor liver metastases (P=0.036), maximum diameter of liver metastases (P=0.002), and recurrence following synchronous radical resection (P=0.003). High density of activated HSCs in the PNLT was identified as a significant and independent prognostic factor for disease-free survival (HR, 2.083; 95% CI, 1.504–2.885; P=0.016) and overall survival (HR, 2.039; 95% CI, 1.312–3.169; P=0.019). Functionally, in vitro assays revealed that activated HSCs facilitated colorectal adenocarcinoma (CRA) cells to colonize the liver. Molecularly, it was demonstrated that the pro-recurrence of activated HSCs depended on paracrine hepatic growth factor. Taken together, the present results showed that high density of activated HSCs in the PNLT was an independent predictor for CRALM recurrence following resection, and they exerted their roles via their effect on CRA cell recruitment and proliferation by paracrine HGF.
Collapse
Affiliation(s)
- Li Deng
- Ultrasonic Department, Jiangxi Pingxiang People's Hospital, Pingxiang, Jiangxi 337000, P.R. China
| | - Taiyuan Li
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China.,Gastrointeral Surgical Institute of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yuanyuan Liao
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shuang Liu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhen Xie
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhixiang Huang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hua Dai
- Department of Pathology, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jianfeng Li
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiong Lei
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China.,Gastrointeral Surgical Institute of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
115
|
Nabariya DK, Pallu R, Yenuganti VR. Exosomes: The protagonists in the tale of colorectal cancer? Biochim Biophys Acta Rev Cancer 2020; 1874:188426. [PMID: 32956762 DOI: 10.1016/j.bbcan.2020.188426] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/11/2020] [Accepted: 09/12/2020] [Indexed: 02/07/2023]
Abstract
Exosomes, which facilitate intercellular communication, antigen presentation and shuttling of biological agents, were initially thought as the cell's garbage cargo but today, after about 40 years of their discovery, we are now beginning to understand their potential role in diagnosis and therapy of several diseases including cancers. Various studies over the decades have signified the role of exosomes in different stages of cancer. Exosomes play a key role in colorectal cancer initiation (CRC), promotion of anti- apoptotic signaling pathways, regulating tumor microenvironment, enhancing tumorigenicity, promotion of angiogenesis, stem cell proliferation and endothelial cell migration, establishment of immune suppressive environment, formation of pre- metastatic niche and metastasis. Exosomes also elicits drug resistance. Since, they have the ability to cross the biological barrier, exosomes are now being explored as an efficient target specific drug delivery system that facilitates the shipping of different biomolecules and therapeutic drugs. However, cautious and strong investigative approaches are required before approving exosomes as therapeutics or drug delivery systems. In this review, we summarize the role of exosomes in different stages of CRC and also elaborate on the applications of exosomes in diagnosis and therapy with respect to CRC.
Collapse
Affiliation(s)
- Deepti Kailash Nabariya
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Reddanna Pallu
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Vengala Rao Yenuganti
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India.
| |
Collapse
|
116
|
Tang H, Chu Y, Huang Z, Cai J, Wang Z. The metastatic phenotype shift toward myofibroblast of adipose-derived mesenchymal stem cells promotes ovarian cancer progression. Carcinogenesis 2020; 41:182-193. [PMID: 31046126 DOI: 10.1093/carcin/bgz083] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 04/06/2019] [Accepted: 04/30/2019] [Indexed: 12/26/2022] Open
Abstract
Ovarian cancer metastasizes to organs in the abdominal cavity, such as the omentum that is a rich source of adipose-derived mesenchymal stem cells (ADSCs). In present, ADSCs have received more and more attention for their roles in the development of cancer. In this study, we examined α-smooth muscle actin (α-SMA) expression and carcinoma-associated fibroblast (CAF)-like differentiation capabilities in ADSCs from omentum of different patients. The effects of ADSCs on the proliferation and invasion of epithelial ovarian cancer cells (EOCCs) were also assessed in vitro and in vivo. Our results showed that ADSCs from omentum of ovarian cancer patients, no matter whether metastasis or not, expressed higher levels of α-SMA than ADSCs from patients with benign gynecologic disease. Using direct and indirect co-culture system, we found that EOCCs induced ADSCs to express CAF markers, including α-SMA and fibroblast activation protein, via the transforming growth factor beta 1 (TGF-β1) signaling pathway. Moreover, co-cultured ADSCs exhibited functional properties similar to those of CAFs, including the ability to promote EOCCs proliferation, progression and metastasis both in vitro and in vivo. Furthermore, blocking the TGF-β1 pathway can counteract the CAF-like differentiation and tumor promotion effect of ADSCs. Our results suggest that ADSCs are a source of CAFs and that they participate in the interaction between EOCCs and the omental microenvironment. EOCCs could induce ADSCs in the omentum to differentiate before ovarian cancer metastasis, which participate in the formation of omental metastatic niches and promote the proliferation and invasion of ovarian cancer.
Collapse
Affiliation(s)
- Huijuan Tang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yijing Chu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zaiju Huang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
117
|
Zhuyan J, Chen M, Zhu T, Bao X, Zhen T, Xing K, Wang Q, Zhu S. Critical steps to tumor metastasis: alterations of tumor microenvironment and extracellular matrix in the formation of pre-metastatic and metastatic niche. Cell Biosci 2020; 10:89. [PMID: 32742634 PMCID: PMC7388444 DOI: 10.1186/s13578-020-00453-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
For decades, cancer metastasis has been a heated topic for its high mortality. Previous research has shown that pre-metastatic niche and metastatic niche are the 2 crucial steps in cancer metastasis, assisting cancerous cells' infiltration, survival, and colonization at target sites. More recent studies have unraveled details about the specific mechanisms related to the modification of pro-invasion environments. Here, we will review literatures on extracellular matrix (ECM) alterations, general cancer metastasis, organ specificity, pre-metastatic niche, metastatic niche, colony formation and impact on the course of metastasis. Respectively, the metastatic mechanisms like effect of hypoxia or inflammation on pre-metastatic niche construction, as well as the interaction between cancer cells and local milieu will be discussed. Based on the evidences of metastatic niches, we revisit and discussed the "Seed and Soil" hypothesis by Paget. This review will seek to provide insight into the mechanism of metastatic organ specificity which pre-metastatic niche and metastatic niche might suggest from an evolutionary aspect.
Collapse
Affiliation(s)
- Jianan Zhuyan
- School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438 China
- Shanghai Starriver Bilingual School, Shanghai, 201100 China
| | - Mingyu Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai, 200040 China
| | - Tianhao Zhu
- Shanghai Starriver Bilingual School, Shanghai, 201100 China
| | - Xunxia Bao
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093 China
| | - Timing Zhen
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093 China
| | - Kaichen Xing
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093 China
| | - Qiubo Wang
- Department of Clinical Laboratory, Wuxi 9th Affiliated Hospital of Soochow University, No.999 Liangxi Road, Wuxi, China
| | - Sibo Zhu
- School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438 China
| |
Collapse
|
118
|
D'Angelo E, Lindoso RS, Sensi F, Pucciarelli S, Bussolati B, Agostini M, Collino F. Intrinsic and Extrinsic Modulators of the Epithelial to Mesenchymal Transition: Driving the Fate of Tumor Microenvironment. Front Oncol 2020; 10:1122. [PMID: 32793478 PMCID: PMC7393251 DOI: 10.3389/fonc.2020.01122] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/04/2020] [Indexed: 12/11/2022] Open
Abstract
The epithelial to mesenchymal transition (EMT) is an evolutionarily conserved process. In cancer, EMT can activate biochemical changes in tumor cells that enable the destruction of the cellular polarity, leading to the acquisition of invasive capabilities. EMT regulation can be triggered by intrinsic and extrinsic signaling, allowing the tumor to adapt to the microenvironment demand in the different stages of tumor progression. In concomitance, tumor cells undergoing EMT actively interact with the surrounding tumor microenvironment (TME) constituted by cell components and extracellular matrix as well as cell secretome elements. As a result, the TME is in turn modulated by the EMT process toward an aggressive behavior. The current review presents the intrinsic and extrinsic modulators of EMT and their relationship with the TME, focusing on the non-cell-derived components, such as secreted metabolites, extracellular matrix, as well as extracellular vesicles. Moreover, we explore how these modulators can be suitable targets for anticancer therapy and personalized medicine.
Collapse
Affiliation(s)
- Edoardo D'Angelo
- First Surgical Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- LIFELAB Program, Consorzio per la Ricerca Sanitaria–CORIS, Veneto Region, Padua, Italy
- Institute of Pediatric Research, Fondazione Citta della Speranza, Padua, Italy
| | - Rafael Soares Lindoso
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine–REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Francesca Sensi
- Institute of Pediatric Research, Fondazione Citta della Speranza, Padua, Italy
- Department of Molecular Sciences and Nanosystems, Cà Foscari University of Venice, Venice, Italy
| | - Salvatore Pucciarelli
- First Surgical Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Benedetta Bussolati
- Department of Medical Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Marco Agostini
- First Surgical Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- LIFELAB Program, Consorzio per la Ricerca Sanitaria–CORIS, Veneto Region, Padua, Italy
- Institute of Pediatric Research, Fondazione Citta della Speranza, Padua, Italy
| | - Federica Collino
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Biomedical Sciences, University of Padova, Padua, Italy
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione Ca' Granda, IRCCS Policlinico di Milano, Milan, Italy
| |
Collapse
|
119
|
Selectively-Packaged Proteins in Breast Cancer Extracellular Vesicles Involved in Metastasis. Int J Mol Sci 2020; 21:ijms21144990. [PMID: 32679759 PMCID: PMC7403963 DOI: 10.3390/ijms21144990] [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: 06/19/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 01/02/2023] Open
Abstract
Cancer-derived extracellular vesicles are known to play a role in the progression of the disease. In this rapidly-growing field, there are many reports of phenotypic changes in cells following exposure to cancer-derived extracellular vesicles. This study examines the protein contents of vesicles derived from three well-known breast cancer cell lines, MCF-7, MDA-MB-231 and T47D, using peptide-centric LC-MS/MS and cytokine multiplex immunoassay analysis to understand the molecular basis of these changes. Through these techniques a large number of proteins within these vesicles were identified. A large proportion of these proteins are known to be important in cancer formation and progression and associated with cancer signaling, angiogenesis, metastasis and invasion and immune regulation. This highlights the importance of extracellular vesicles (EVs) in cancer communications and shows some of the mechanisms the vesicles use to assist in cancer progression.
Collapse
|
120
|
Gajos-Michniewicz A, Czyz M. WNT Signaling in Melanoma. Int J Mol Sci 2020; 21:E4852. [PMID: 32659938 PMCID: PMC7402324 DOI: 10.3390/ijms21144852] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
WNT-signaling controls important cellular processes throughout embryonic development and adult life, so any deregulation of this signaling can result in a wide range of pathologies, including cancer. WNT-signaling is classified into two categories: β-catenin-dependent signaling (canonical pathway) and β-catenin-independent signaling (non-canonical pathway), the latter can be further divided into WNT/planar cell polarity (PCP) and calcium pathways. WNT ligands are considered as unique directional growth factors that contribute to both cell proliferation and polarity. Origin of cancer can be diverse and therefore tissue-specific differences can be found in WNT-signaling between cancers, including specific mutations contributing to cancer development. This review focuses on the role of the WNT-signaling pathway in melanoma. The current view on the role of WNT-signaling in cancer immunity as well as a short summary of WNT pathway-related drugs under investigation are also provided.
Collapse
Affiliation(s)
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92–215 Lodz, Poland;
| |
Collapse
|
121
|
Tvaroška I, Selvaraj C, Koča J. Selectins-The Two Dr. Jekyll and Mr. Hyde Faces of Adhesion Molecules-A Review. Molecules 2020; 25:molecules25122835. [PMID: 32575485 PMCID: PMC7355470 DOI: 10.3390/molecules25122835] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/27/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Selectins belong to a group of adhesion molecules that fulfill an essential role in immune and inflammatory responses and tissue healing. Selectins are glycoproteins that decode the information carried by glycan structures, and non-covalent interactions of selectins with these glycan structures mediate biological processes. The sialylated and fucosylated tetrasaccharide sLex is an essential glycan recognized by selectins. Several glycosyltransferases are responsible for the biosynthesis of the sLex tetrasaccharide. Selectins are involved in a sequence of interactions of circulated leukocytes with endothelial cells in the blood called the adhesion cascade. Recently, it has become evident that cancer cells utilize a similar adhesion cascade to promote metastases. However, like Dr. Jekyll and Mr. Hyde’s two faces, selectins also contribute to tissue destruction during some infections and inflammatory diseases. The most prominent function of selectins is associated with the initial stage of the leukocyte adhesion cascade, in which selectin binding enables tethering and rolling. The first adhesive event occurs through specific non-covalent interactions between selectins and their ligands, with glycans functioning as an interface between leukocytes or cancer cells and the endothelium. Targeting these interactions remains a principal strategy aimed at developing new therapies for the treatment of immune and inflammatory disorders and cancer. In this review, we will survey the significant contributions to and the current status of the understanding of the structure of selectins and the role of selectins in various biological processes. The potential of selectins and their ligands as therapeutic targets in chronic and acute inflammatory diseases and cancer will also be discussed. We will emphasize the structural characteristic of selectins and the catalytic mechanisms of glycosyltransferases involved in the biosynthesis of glycan recognition determinants. Furthermore, recent achievements in the synthesis of selectin inhibitors will be reviewed with a focus on the various strategies used for the development of glycosyltransferase inhibitors, including substrate analog inhibitors and transition state analog inhibitors, which are based on knowledge of the catalytic mechanism.
Collapse
Affiliation(s)
- Igor Tvaroška
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic
- Institute of Chemistry, Slovak Academy of Sciences, 84538 Bratislava, Slovak Republic
- Correspondence: (I.T.); (J.K.); Tel.: +421-948-535-601 (I.T.); +420-731-682-606 (J.K.)
| | - Chandrabose Selvaraj
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic
| | - Jaroslav Koča
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic
- Correspondence: (I.T.); (J.K.); Tel.: +421-948-535-601 (I.T.); +420-731-682-606 (J.K.)
| |
Collapse
|
122
|
|
123
|
Sellner F. Isolated Pancreatic Metastases of Renal Cell Carcinoma-A Paradigm of a Seed and Soil Mechanism: A Literature Analysis of 1,034 Observations. Front Oncol 2020; 10:709. [PMID: 32547940 PMCID: PMC7273884 DOI: 10.3389/fonc.2020.00709] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/15/2020] [Indexed: 12/15/2022] Open
Abstract
Previously documented arguments, in favor of the suspected impact of a seed and soil mechanism, in the development and progression of isolated pancreatic metastasis of renal cell carcinomas (isPM) are: (1) uniform and independent from the side of the primary tumor distribution of isPM within the pancreas and, (2) the similar survival rates for singular and multiple isPM. In addition, the present study adds new arguments that further confirm the importance of an seed and soil mechanism in isPM: (1) Within the singular isPM, the size of the metastasis does not affect the overall survival; (2) Within the group of multiple isPMs, the overall survival does not depend on the number of metastases; (3) For synchronous and metachronous isPM, survival rates are also not different, and (4) Within the group of metachronous isPM there is also no correlation between the overall survival and interval until metastases occurs. This unusual ineffectiveness of otherwise known risk factors of solid cancers can be explained plausibly by the hypothesis of a very selective seed and soil mechanism in isPM. It only allows embolized renal carcinoma cells in the pancreas to complete all steps required to grow into clinically manifest metastases. In all other organs, on the other hand, the body is able to eliminate the embolized tumor cells or at least put them into a dormant state for many years. This minimizes the risk of occult micrometastases in distant organs, which could later—after isPM treatment—grow into clinically manifest metastases, so that the prognosis of the isPM is only determined by an adequate therapy of the pancreatic foci, and prognostic factors, such as total tumor burden or interval until the occurrence of the isPM remain ineffective.
Collapse
Affiliation(s)
- Franz Sellner
- Surgical Department, Kaiser Franz Josef Hospital, Vienna, Austria
| |
Collapse
|
124
|
Kumar A, Deep G. Exosomes in hypoxia-induced remodeling of the tumor microenvironment. Cancer Lett 2020; 488:1-8. [PMID: 32473240 DOI: 10.1016/j.canlet.2020.05.018] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/06/2020] [Accepted: 05/17/2020] [Indexed: 12/13/2022]
Abstract
Exosomes are structurally and functionally pleiotropic nano-sized (~30-150 nm in diameter) extracellular vesicles (EVs) with endosomal origin. These vesicles are secreted by almost all cells and play a significant role in intercellular communication and bio-waste disposal. To a great extent, exosomes represent biological "snapshot" of parent cells, and their cargos (protein, nucleotides, lipids, and metabolites) are loaded uniquely under different pathophysiological conditions. For example, most cancerous cells secrete a higher amount of exosomes loaded with distinct cargos under stressful low oxygen condition i.e. hypoxia, a key characteristic of solid tumors responsible for disease aggressiveness and poor survival. Exosomes secreted under hypoxia (ExoHypoxic) play a vital role in aiding cancer cells crosstalk with its microenvironment constituents to create conditions advantageous for cancer growth and metastatic spread. In this review article, we have highlighted the effects of ExoHypoxic on various tumor microenvironment components involved in angiogenesis, survival, proliferation, pre-metastatic niches preparation, immunomodulation, epithelial-to-mesenchymal transition, invasion, metastasis, and drug resistance. We have also described key ExoHypoxic cargos (miRNA, proteins, etc) and their targets in the receipt cells, responsible for various biological effects. Finally, we have emphasized the applicability of ExoHypoxic as a biomarker of tumor hypoxia and disease prognosis.
Collapse
Affiliation(s)
- Ashish Kumar
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA; Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA; Department of Urology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
125
|
Gui J, Zahedi F, Ortiz A, Cho C, Katlinski KV, Alicea-Torres K, Li J, Todd L, Zhang H, Beiting DP, Sander C, Kirkwood JM, Snow BE, Wakeham AC, Mak TW, Diehl JA, Koumenis C, Ryeom SW, Stanger BZ, Puré E, Gabrilovich DI, Fuchs SY. Activation of p38α stress-activated protein kinase drives the formation of the pre-metastatic niche in the lungs. ACTA ACUST UNITED AC 2020; 1:603-619. [PMID: 34124690 DOI: 10.1038/s43018-020-0064-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Primary tumor-derived factors (TDFs) act upon normal cells to generate a pre-metastatic niche, which promotes colonization of target organs by disseminated malignant cells. Here we report that TDFs-induced activation of the p38α kinase in lung fibroblasts plays a critical role in the formation of a pre-metastatic niche in the lungs and subsequent pulmonary metastases. Activation of p38α led to inactivation of type I interferon signaling and stimulation of expression of fibroblast activation protein (FAP). FAP played a key role in remodeling of the extracellular matrix as well as inducing the expression of chemokines that enable lung infiltration by neutrophils. Increased activity of p38 in normal cells was associated with metastatic disease and poor prognosis in human melanoma patients whereas inactivation of p38 suppressed lung metastases. We discuss the p38α-driven mechanisms stimulating the metastatic processes and potential use of p38 inhibitors in adjuvant therapy of metastatic cancers.
Collapse
Affiliation(s)
- Jun Gui
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; USA
| | - Farima Zahedi
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; USA
| | - Angelica Ortiz
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; USA
| | - Christina Cho
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; USA
| | - Kanstantsin V Katlinski
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; USA
| | - Kevin Alicea-Torres
- Immunology, Microenvironment, and Metastasis, Wistar Institute, Philadelphia, PA 19104; USA
| | - Jinyang Li
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104, USA
| | - Leslie Todd
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; USA
| | - Hongru Zhang
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; USA
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cindy Sander
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - John M Kirkwood
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Bryan E Snow
- The Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Andrew C Wakeham
- The Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Tak W Mak
- The Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - J Alan Diehl
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104, USA
| | - Sandra W Ryeom
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104, USA
| | - Ben Z Stanger
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104, USA
| | - Ellen Puré
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; USA
| | - Dmitry I Gabrilovich
- Immunology, Microenvironment, and Metastasis, Wistar Institute, Philadelphia, PA 19104; USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; USA
| |
Collapse
|
126
|
Peeney D, Jensen SM, Castro NP, Kumar S, Noonan S, Handler C, Kuznetsov A, Shih J, Tran AD, Salomon DS, Stetler-Stevenson WG. TIMP-2 suppresses tumor growth and metastasis in murine model of triple-negative breast cancer. Carcinogenesis 2020; 41:313-325. [PMID: 31621840 PMCID: PMC7221506 DOI: 10.1093/carcin/bgz172] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/12/2019] [Accepted: 10/11/2019] [Indexed: 12/30/2022] Open
Abstract
Metastasis is the primary cause of treatment failures and mortality in most cancers. Triple-negative breast cancer (TNBC) is refractory to treatment and rapidly progresses to disseminated disease. We utilized an orthotopic mouse model that molecularly and phenotypically resembles human TNBC to study the effects of exogenous, daily tissue inhibitor of metalloproteinase-2 (TIMP-2) treatment on tumor growth and metastasis. Our results demonstrated that TIMP-2 treatment maximally suppressed primary tumor growth by ~36-50% and pulmonary metastasis by >92%. Immunostaining assays confirmed disruption of the epithelial to mesenchymal transition (EMT) and promotion of vascular integrity in primary tumor tissues. Immunostaining and RNA sequencing analysis of lung tissue lysates from tumor-bearing mice identified significant changes associated with metastatic colony formation. Specifically, TIMP-2 treatment disrupts periostin localization and critical cell-signaling pathways, including canonical Wnt signaling involved in EMT, as well as PI3K signaling, which modulates proliferative and metastatic behavior through p27 phosphorylation/localization. In conclusion, our study provides evidence in support of a role for TIMP-2 in suppression of triple-negative breast cancer growth and metastasis through modulation of the epithelial to mesenchymal transition, vascular normalization, and signaling pathways associated with metastatic outgrowth. Our findings suggest that TIMP-2, a constituent of the extracellular matrix in normal tissues, may have both direct and systemic antitumor and metastasis suppressor effects, suggesting potential utility in the clinical management of breast cancer progression.
Collapse
Affiliation(s)
- David Peeney
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sandra M Jensen
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Nadia P Castro
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Sarvesh Kumar
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Silvia Noonan
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Chenchen Handler
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Alex Kuznetsov
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Joanna Shih
- Biostatistics Branch, National Cancer Institute, Rockville, MD, USA
| | - Andy D Tran
- Confocal Core Facility, National Cancer Institute, Bethesda, MD, USA
| | - David S Salomon
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - William G Stetler-Stevenson
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
127
|
Azizi M, Dianat-Moghadam H, Salehi R, Farshbaf M, Iyengar D, Sau S, Iyer AK, Valizadeh H, Mehrmohammadi M, Hamblin MR. Interactions Between Tumor Biology and Targeted Nanoplatforms for Imaging Applications. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910402. [PMID: 34093104 PMCID: PMC8174103 DOI: 10.1002/adfm.201910402] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Indexed: 05/04/2023]
Abstract
Although considerable efforts have been conducted to diagnose, improve, and treat cancer in the past few decades, existing therapeutic options are insufficient, as mortality and morbidity rates remain high. Perhaps the best hope for substantial improvement lies in early detection. Recent advances in nanotechnology are expected to increase the current understanding of tumor biology, and will allow nanomaterials to be used for targeting and imaging both in vitro and in vivo experimental models. Owing to their intrinsic physicochemical characteristics, nanostructures (NSs) are valuable tools that have received much attention in nanoimaging. Consequently, rationally designed NSs have been successfully employed in cancer imaging for targeting cancer-specific or cancer-associated molecules and pathways. This review categorizes imaging and targeting approaches according to cancer type, and also highlights some new safe approaches involving membrane-coated nanoparticles, tumor cell-derived extracellular vesicles, circulating tumor cells, cell-free DNAs, and cancer stem cells in the hope of developing more precise targeting and multifunctional nanotechnology-based imaging probes in the future.
Collapse
Affiliation(s)
- Mehdi Azizi
- Proteomics Research Centre, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran
| | - Hassan Dianat-Moghadam
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 5165665621, Iran
| | - Roya Salehi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, Tabriz 516615731, Iran
| | - Masoud Farshbaf
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 6581151656, Iran
| | - Disha Iyengar
- U-BiND Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Samaresh Sau
- U-BiND Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Arun K Iyer
- U-BiND Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Hadi Valizadeh
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, Tabriz 516615731, Iran
| | | | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
128
|
Fu LQ, Du WL, Cai MH, Yao JY, Zhao YY, Mou XZ. The roles of tumor-associated macrophages in tumor angiogenesis and metastasis. Cell Immunol 2020; 353:104119. [PMID: 32446032 DOI: 10.1016/j.cellimm.2020.104119] [Citation(s) in RCA: 246] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/06/2020] [Accepted: 05/01/2020] [Indexed: 12/14/2022]
Abstract
Tumor associated macrophages (TAMs) are the most frequent immune cells within tumor microenvironment (TME). There is growing evidence that TAMs are involved in tumor progression via multiple mechanisms. TAMs create an immunosuppressive TME by producing growth factors, chemokines, and cytokines which modulate recruitment of immune cells and inhibit anti-tumor responses. They also serve as angiogenesis promoting cells by production of pro-angiogenic factors and matrix metalloproteinases (MMPs) and vascular constructing which guarantee supplying oxygen and nutrients to solid tumor cells. Furthermore, TAMs play important functions in tumor metastasis through contributing to invasion, extravasation, survival, intravasation, and colonization of tumor cells. In this review, we summarized macrophage classification, TAMs polarization, and mechanisms underlying TAM-promoting angiogenesis and metastasis.
Collapse
Affiliation(s)
- Luo-Qin Fu
- Department of General Surgery, Chun'an First People's Hospital (Zhejiang Provincial People's Hospital Chun'an Branch), Hangzhou 311700, Zhejiang Province, China
| | - Wen-Lin Du
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, China
| | - Mao-Hua Cai
- Department of General Surgery, Chun'an First People's Hospital (Zhejiang Provincial People's Hospital Chun'an Branch), Hangzhou 311700, Zhejiang Province, China
| | - Jia-Yu Yao
- Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, China
| | - Yuan-Yuan Zhao
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, China; Department of Neurosurgery, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), No. 158 Shangtang Road, Hangzhou 310014, Zhejiang Province, China.
| | - Xiao-Zhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, China.
| |
Collapse
|
129
|
Bandopadhyay M, Bharadwaj M. Exosomal miRNAs in hepatitis B virus related liver disease: a new hope for biomarker. Gut Pathog 2020; 12:23. [PMID: 32346400 PMCID: PMC7183117 DOI: 10.1186/s13099-020-00353-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 03/31/2020] [Indexed: 02/06/2023] Open
Abstract
The World Health Organisation, in its 2019 progress report on HIV, viral hepatitis and STDs indicates that 257 million people are afflicted with chronic HBV infections, of which, 1 million patients lose their lives every year due to HBV related chronic liver diseases including serious complications such as liver cirrhosis and hepatocellular carcinoma. The course of HBV infection and associated liver injury depend on several host factors, genetic variability of the virus, and the host viral interplay. The challenge of medical science is the early diagnosis/identification of the potential for development of fatal complications like liver cirrhosis and HCC so that timely medical intervention can improve the chances of survival. Currently, neither the vaccination regime nor the diagnostic methods are completely effective as reflected in the high number of annual deaths. It is evident from numerous publications that microRNAs (miRNAs) are the critical regulators of gene expression and various cellular processes like proliferation, development, differentiation, apoptosis and tumorigenesis. Expressions of these diminutive RNAs are significantly affected in cancerous tissues as a result of numerous genomic and epigenetic modifications. Exosomes are membrane-derived vesicles (30–100 nm) secreted by normal as well as malignant cells, and are present in all body fluids. They are recognized as critical molecules in intercellular communication between cells through horizontal transfer of information via their cargo, which includes selective proteins, mRNAs and miRNAs. Exosomal miRNAs are transferred to recipient cells where they can regulate target gene expression. This provides an insight into the elementary biology of cancer progression and therefore the development of therapeutic approaches. This concise review outlines various on-going research on miRNA mediated regulation of HBV pathogenesis with special emphasis on association of exosomal miRNA in advanced stage liver disease like hepatocellular carcinoma. This review also discusses the possible use of exosomal miRNAs as biomarkers in the early detection of HCC and liver cirrhosis.
Collapse
Affiliation(s)
- Manikankana Bandopadhyay
- Molecular Genetics and Biochemistry, National Institute of Cancer Prevention and Research (NICPR), Indian Council of Medical Research (ICMR), Noida, Uttar Pradesh 201301 India
| | - Mausumi Bharadwaj
- Molecular Genetics and Biochemistry, National Institute of Cancer Prevention and Research (NICPR), Indian Council of Medical Research (ICMR), Noida, Uttar Pradesh 201301 India
| |
Collapse
|
130
|
Tang Q, Yin D, Wang Y, Du W, Qin Y, Ding A, Li H. Cancer Stem Cells and Combination Therapies to Eradicate Them. Curr Pharm Des 2020; 26:1994-2008. [PMID: 32250222 DOI: 10.2174/1381612826666200406083756] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/13/2020] [Indexed: 12/23/2022]
Abstract
Cancer stem cells (CSCs) show self-renewal ability and multipotential differentiation, like normal stem or progenitor cells, and which proliferate uncontrollably and can escape the effects of drugs and phagocytosis by immune cells. Traditional monotherapies, such as surgical resection, radiotherapy and chemotherapy, cannot eradicate CSCs, however, combination therapy may be more effective at eliminating CSCs. The present review summarizes the characteristics of CSCs and several promising combination therapies to eradicate them.
Collapse
Affiliation(s)
- Qi Tang
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, China.,Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Dan Yin
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Yao Wang
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, China
| | - Wenxuan Du
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, China
| | - Yuhan Qin
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, China
| | - Anni Ding
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, China
| | - Hanmei Li
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, China
| |
Collapse
|
131
|
Ge Y, Mu W, Ba Q, Li J, Jiang Y, Xia Q, Wang H. Hepatocellular carcinoma-derived exosomes in organotropic metastasis, recurrence and early diagnosis application. Cancer Lett 2020; 477:41-48. [PMID: 32112905 DOI: 10.1016/j.canlet.2020.02.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/21/2019] [Accepted: 02/06/2020] [Indexed: 12/17/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, despite improvements in the clinical trial and diagnosis, HCC still remains high mortality due to the 70% recurrence and lung metastasis after surgical resection. Exosomes are small membrane vesicles, which are shuttled from donor cells to recipient cells, contributing to the recruitment and reprogramming of constituents via an autocrine or paracrine fashion. HCC derived exosomes could redirect metastasis of tumor cells which lack the capacity to metastasize to a specific organ via generating pre-metastatic niche. These findings emphasize a practical and potentially feasible role of exosomes in the treatment of patients with HCC, both as a target and a vehicle for drug design. We herein summarize recent findings that implicate oncogenes and non-canonical signaling of HCC exosomes, as well as the impact of exosomal bioactive molecules in high recurrence induced by organ-specific metastasis. The aim of review is to illustrate the underlying mechanism of exosomes in tumor metastasis, immune evasion, and the potential application of prognostic biomarker in HCC process.
Collapse
Affiliation(s)
- Yang Ge
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Wei Mu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Qian Ba
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Jingquan Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yiguo Jiang
- School of Public Health, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qiang Xia
- Organ Transplantation Center, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China.
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200032, China.
| |
Collapse
|
132
|
Trovato R, Canè S, Petrova V, Sartoris S, Ugel S, De Sanctis F. The Engagement Between MDSCs and Metastases: Partners in Crime. Front Oncol 2020; 10:165. [PMID: 32133298 PMCID: PMC7040035 DOI: 10.3389/fonc.2020.00165] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 01/30/2020] [Indexed: 12/18/2022] Open
Abstract
Tumor metastases represent the major cause of cancer-related mortality, confirming the urgent need to identify key molecular pathways and cell-associated networks during the early phases of the metastatic process to develop new strategies to either prevent or control distal cancer spread. Several data revealed the ability of cancer cells to establish a favorable microenvironment, before their arrival in distant organs, by manipulating the cell composition and function of the new host tissue where cancer cells can survive and outgrow. This predetermined environment is termed “pre-metastatic niche” (pMN). pMN development requires that tumor-derived soluble factors, like cytokines, growth-factors and extracellular vesicles, genetically and epigenetically re-program not only resident cells (i.e., fibroblasts) but also non-resident cells such as bone marrow-derived cells. Indeed, by promoting an “emergency” myelopoiesis, cancer cells switch the steady state production of blood cells toward the generation of pro-tumor circulating myeloid cells defined as myeloid-derived suppressor cells (MDSCs) able to sustain tumor growth and dissemination. MDSCs are a heterogeneous subset of myeloid cells with immunosuppressive properties that sustain metastatic process. In this review, we discuss current understandings of how MDSCs shape and promote metastatic dissemination acting in each fundamental steps of cancer progression from primary tumor to metastatic disease.
Collapse
Affiliation(s)
- Rosalinda Trovato
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Stefania Canè
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Varvara Petrova
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Silvia Sartoris
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Stefano Ugel
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Francesco De Sanctis
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
133
|
Models for Monocytic Cells in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 32036607 DOI: 10.1007/978-3-030-35723-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Monocytes (Mos) are immune cells that critically regulate cancer, enabling tumor growth and modulating metastasis. Mos can give rise to tumor-associated macrophages (TAMs) and Mo-derived dendritic cells (moDCs), all of which shape the tumor microenvironment (TME). Thus, understanding their roles in the TME is key for improved immunotherapy. Concurrently, various biological and mechanical factors including changes in local cytokines, extracellular matrix production, and metabolic changes in the TME affect the roles of monocytic cells. As such, relevant TME models are critical to achieve meaningful insight on the precise functions, mechanisms, and effects of monocytic cells. Notably, murine models have yielded significant insight into human Mo biology. However, many of these results have yet to be confirmed in humans, reinforcing the need for improved in vitro human TME models for the development of cancer interventions. Thus, this chapter (1) summarizes current insight on the tumor biology of Mos, TAMs, and moDCs, (2) highlights key therapeutic applications relevant to these cells, and (3) discusses various TME models to study their TME-related activity. We conclude with a perspective on the future research trajectory of this topic.
Collapse
|
134
|
Ghouse SM, Vadrevu SK, Manne S, Reese B, Patel J, Patel B, Silwal A, Lodhi N, Paterson Y, Srivastava SK, Karbowniczek M, Markiewski MM. Therapeutic Targeting of Vasculature in the Premetastatic and Metastatic Niches Reduces Lung Metastasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:990-1000. [PMID: 31900334 PMCID: PMC7012400 DOI: 10.4049/jimmunol.1901208] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023]
Abstract
In the metastasis-targeted organs, angiogenesis is essential for the progression of dormant micrometastases to rapidly growing and clinically overt lesions. However, we observed changes suggesting angiogenic switching in the mouse lungs prior to arrival of tumor cells (i.e., in the premetastatic niche) in the models of breast carcinoma. This angiogenic switching appears to be caused by myeloid-derived suppressor cells recruited to the premetastatic lungs through complement C5a receptor 1 signaling. These myeloid cells are known to secrete several proangiogenic factors in tumors, including IL-1β and matrix metalloproteinase-9, and we found upregulation of these genes in the premetastatic lungs. Blockade of C5a receptor 1 synergized with antiangiogenic Listeria monocytogenes-based vaccines to decrease the lung metastatic burden by reducing vascular density and improving antitumor immunity in the lungs. This was mediated even when growth of primary breast tumors was not affected by these treatments. This work provides initial evidence that angiogenesis contributes to the premetastatic niche in rapidly progressing cancers and that inhibiting this process through immunotherapy is beneficial for reducing or even preventing metastasis.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Cancer Vaccines/administration & dosage
- Cell Line, Tumor
- Combined Modality Therapy/methods
- Complement C5a/immunology
- Complement C5a/metabolism
- Female
- Humans
- Immunotherapy/methods
- Listeria monocytogenes/immunology
- Lung/blood supply
- Lung/immunology
- Lung/pathology
- Lung Neoplasms/blood supply
- Lung Neoplasms/immunology
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
- Matrix Metalloproteinase 9/metabolism
- Mice
- Mice, Knockout
- Myeloid-Derived Suppressor Cells/immunology
- Myeloid-Derived Suppressor Cells/metabolism
- Neoplasm Metastasis/immunology
- Neoplasm Metastasis/therapy
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/therapy
- Receptor, Anaphylatoxin C5a/antagonists & inhibitors
- Receptor, Anaphylatoxin C5a/genetics
- Receptor, Anaphylatoxin C5a/metabolism
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Shanawaz M Ghouse
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Surya K Vadrevu
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Sasikanth Manne
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Britney Reese
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Jalpa Patel
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Bhaumik Patel
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Ashok Silwal
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Niraj Lodhi
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Yvonne Paterson
- Department of Microbiology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Sanjay K Srivastava
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Magdalena Karbowniczek
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601
| | - Maciej M Markiewski
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601;
| |
Collapse
|
135
|
Liu X, Xu D, Liu Z, Li Y, Zhang C, Gong Y, Jiang Y, Xing B. THBS1 facilitates colorectal liver metastasis through enhancing epithelial-mesenchymal transition. Clin Transl Oncol 2020; 22:1730-1740. [PMID: 32052380 DOI: 10.1007/s12094-020-02308-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/21/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Liver metastasis is one of the major causes of cancer-related death in patients with colorectal cancer (CRC). The purpose of this study was to identify specific molecules which are involved in colorectal liver metastasis (CRLM). MATERIALS AND METHODS In this study, we employed TMT (tandem mass tags)-labeling combined with liquid chromatography-mass spectrometry technology to do comparative analyses of proteomics between the primary tumor specimens derived from colorectal cancer patients with or without liver metastasis. Pathway enrichment analyses were performed using DAVID database. The crucial molecules were identified through protein-protein interaction network. Immunohistochemistry (IHC) was employed to analyze the expression of THBS1 (thrombospondin-1) in CRC tissues. Finally, transwell cell migration and invasion assays were performed to explore the roles of THBS1 in CRC cell migration and invasion. RESULTS We found that the expression of 311 proteins was dysregulated in CRLM using quantitative proteomics. Among these proteins, we identified FN1, TIMP1, THBS1, POSTN and VCAN as five crucial proteins in CRLM by analysis in silico. IHC assay revealed that increased THBS1 expression was significantly correlated with liver metastasis as well as poor prognosis of CRC patients. GEO data analysis also suggests that upregulated mRNA level of THBS1 is also associated with shorter overall survival of CRC patients. Moreover, THBS1 depletion inhibited migration and invasion of CRC cells through attenuating epithelial-mesenchymal transition. Co-expression analyses with TCGA data indicated that THBS1 is co-expressed with mesenchymal markers, including Vimentin, N-cadherin, Snail1 and Twist1 in CRC tissues. CONCLUSIONS By collecting the omics data with functional studies, the present results reveal that THBS1 facilitates colorectal liver metastasis through promoting epithelial-mesenchymal transition. This understanding of molecular roles of THBS1 in CRLM may be promising to develop targeted therapies to prolong survival in CRC patients.
Collapse
Affiliation(s)
- X Liu
- Hepatopancreatobiliary Surgery Department I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - D Xu
- Hepatopancreatobiliary Surgery Department I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Z Liu
- Hepatopancreatobiliary Surgery Department I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Y Li
- Hepatopancreatobiliary Surgery Department I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - C Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Y Gong
- Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Y Jiang
- Department of Medical Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - B Xing
- Hepatopancreatobiliary Surgery Department I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
136
|
Rafaeva M, Erler JT. Framing cancer progression: influence of the organ- and tumour-specific matrisome. FEBS J 2020; 287:1454-1477. [PMID: 31972068 DOI: 10.1111/febs.15223] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/16/2019] [Accepted: 01/20/2020] [Indexed: 12/19/2022]
Abstract
The extracellular matrix (ECM) plays a crucial role in regulating organ homeostasis. It provides mechanical and biochemical cues directing cellular behaviour and, therefore, has control over the progression of diseases such as cancer. Recent efforts have greatly enhanced our knowledge of the protein composition of the ECM and its regulators, the so-called matrisome, in healthy and cancerous tissues; yet, an overview of the common signatures and organ-specific ECM in cancer is missing. Here, we address this by taking a detailed approach to review why cancer grows in certain organs, and focus on the influence of the matrisome at primary and metastatic tumour sites. Our in-depth and comprehensive review of the current literature and general understanding identifies important commonalities and distinctions, providing insight into the biology of metastasis, which could pave the way to improve future diagnostics and therapies.
Collapse
Affiliation(s)
- Maria Rafaeva
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen (UCPH), Denmark
| | - Janine T Erler
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen (UCPH), Denmark
| |
Collapse
|
137
|
Charan M, Dravid P, Cam M, Setty B, Roberts RD, Houghton PJ, Cam H. Tumor secreted ANGPTL2 facilitates recruitment of neutrophils to the lung to promote lung pre-metastatic niche formation and targeting ANGPTL2 signaling affects metastatic disease. Oncotarget 2020; 11:510-522. [PMID: 32082485 PMCID: PMC7007290 DOI: 10.18632/oncotarget.27433] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/28/2019] [Indexed: 12/28/2022] Open
Abstract
The pre-metastatic niche (PMN) represents an abnormal microenvironment devoid of cancer cells, but favoring tumor growth. Little is known about the mechanisms that generate the PMN or their effects on host cells within metastasis-prone organs. Here, we investigated by using spontaneous metastatic models whether lung epithelial cells are essential for primary tumor induced neutrophil recruitment in lung and subsequently initiating PMN formation in osteosarcoma. We found that serum levels of ANGPTL2 in osteosarcoma patients are significantly higher compared to those in healthy controls and that ANGPTL2 secretion by tumor cells plays an essential role in osteosarcoma metastasis. We determined that tumor-derived ANGPTL2 stimulates lung epithelial cells, which is essential for primary tumor-induced neutrophil recruitment in lung and subsequent pre-metastatic niche formation. Lastly, we identified that a p63 isoform, ΔNp63, drives high level of ANGPTL2 secretion and pharmaceutical inhibition of ANGPTL2 signaling by a non–RGD-based integrin binding peptide (ATN-161) diminished metastatic load in lungs likely due to reduction of the lung pre-metastatic niche formation.
Collapse
Affiliation(s)
- Manish Charan
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Piyush Dravid
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Maren Cam
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Bhuvana Setty
- Department of Hematology & Oncology, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Ryan D Roberts
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Peter J Houghton
- Greehey Children's Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Hakan Cam
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
138
|
Deep G, Jain A, Kumar A, Agarwal C, Kim S, Leevy WM, Agarwal R. Exosomes secreted by prostate cancer cells under hypoxia promote matrix metalloproteinases activity at pre-metastatic niches. Mol Carcinog 2020; 59:323-332. [PMID: 31943365 DOI: 10.1002/mc.23157] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/28/2019] [Accepted: 01/02/2020] [Indexed: 12/21/2022]
Abstract
Approximately, 30 000 men die from prostate cancer (PCa) every year in the United States, mainly due to the metastasis. Thus, the key events associated with PCa metastasis are under rigorous investigation, with recent studies showing that preparation of pre-metastatic niches (PMN) in distant organs is an important step. However, the molecular basis for PMN preparation is still unclear. Hypoxia in primary tumors promotes aggressiveness; however, its precise role in metastasis is not clear. We recently reported that exosomes secreted by PCa cells under hypoxia promote stemness and invasiveness in naïve PCa cells; however, whether these extracellular vesicles also influence PMN remains unknown. In the present study, we isolated exosomes from human PCa PC3 cells under normoxic (21% O2 , exosomes secreted under normoxic condition [ExoNormoxic ]) and hypoxic (1% O2 , exosomes secreted under hypoxic condition [ExoHypoxic ]) conditions, and characterized their effect (10 µg exosomes, intraperitoneal (IP) treatment every 48 hours for 4 weeks) on key biomarkers associated with PMN in nude mice. Whole animal fluorescence imaging showed that ExoHypoxic treatment promotes matrix metalloproteinases (MMPs) activity in several putative metastatic sites. Histological studies confirmed that ExoHypoxic treatment enhanced the level of MMP2, MMP9, and extracellular matrix proteins (fibronectin and collagen) as well as increased the number of CD11b+ cells at selective PMN sites. Furthermore, proteomic profiling of exosomes by liquid chromatography/mass spectrometry identified cargo proteins in ExoNormoxic and ExoHypoxic as well as distinct canonical pathways targeted by them. These results suggest that exosomes secreted by PCa cells under hypoxia plausibly remodel distant PMN, and thus, could be a potential target to control metastatic PCa.
Collapse
Affiliation(s)
- Gagan Deep
- Department of Cancer Biology, Winston-Salem, North Carolina.,Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina.,Department of Urology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Anil Jain
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado
| | - Ashish Kumar
- Department of Cancer Biology, Winston-Salem, North Carolina
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado.,University of Colorado Cancer Center, University of Colorado, Aurora, Colorado
| | - Susy Kim
- Department of Cancer Biology, Winston-Salem, North Carolina
| | - W Matthew Leevy
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado.,University of Colorado Cancer Center, University of Colorado, Aurora, Colorado
| |
Collapse
|
139
|
Rodríguez Zorrilla S, García García A, Blanco Carrión A, Gándara Vila P, Somoza Martín M, Gallas Torreira M, Pérez Sayans M. Exosomes in head and neck cancer. Updating and revisiting. J Enzyme Inhib Med Chem 2020; 34:1641-1651. [PMID: 31496355 PMCID: PMC6746279 DOI: 10.1080/14756366.2019.1662000] [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] [Indexed: 12/12/2022] Open
Abstract
Exosomes have gone from being considered simple containers of intracellular waste substances to be considered important carriers of cellular signals. Its broad capacity to promote tumour growth, both in situ and metastatic, has greatly intensified scientific research on them. In the same way and depending on its content, its tumour suppressive properties have opened a window of light and hope in the fight against cancer. In the present review we try to gather in a simple and understandable way the most relevant knowledge to date on the role of exosomes in oral squamous cell carcinoma, helping to understand their process of formation, release and activity on the tumour microenvironment.
Collapse
Affiliation(s)
- Samuel Rodríguez Zorrilla
- Oral Surgery and Implantology Unit, School of Medicine and Dentistry, University of Santiago de Compostela , Santiago de Compostela , Spain
| | - Abel García García
- Oral Surgery and Implantology Unit, School of Medicine and Dentistry, Instituto de Investigación Sanitaria de Santiago (IDIS) , Santiago de Compostela , Spain
| | - Andrés Blanco Carrión
- Oral Surgery and Implantology Unit, School of Medicine and Dentistry, University of Santiago de Compostela , Santiago de Compostela , Spain
| | - Pilar Gándara Vila
- Oral Surgery and Implantology Unit, School of Medicine and Dentistry, University of Santiago de Compostela , Santiago de Compostela , Spain
| | - Manuel Somoza Martín
- Oral Surgery and Implantology Unit, School of Medicine and Dentistry, University of Santiago de Compostela , Santiago de Compostela , Spain
| | - Mercedes Gallas Torreira
- Oral Surgery and Implantology Unit, School of Medicine and Dentistry, University of Santiago de Compostela , Santiago de Compostela , Spain
| | - Mario Pérez Sayans
- Oral Surgery and Implantology Unit, School of Medicine and Dentistry, Instituto de Investigación Sanitaria de Santiago (IDIS) , Santiago de Compostela , Spain
| |
Collapse
|
140
|
Raskov H, Orhan A, Salanti A, Gögenur I. Premetastatic niches, exosomes and circulating tumor cells: Early mechanisms of tumor dissemination and the relation to surgery. Int J Cancer 2020; 146:3244-3255. [PMID: 31808150 DOI: 10.1002/ijc.32820] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/15/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022]
Abstract
The physiological stress response to surgery promotes wound healing and functional recovery and includes the activation of neural, inflammatory and proangiogenic signaling pathways. Paradoxically, the same pathways also promote metastatic spread and growth of residual cancer. Human and animal studies show that cancer surgery can increase survival, migration and proliferation of residual tumor cells. To secure the survival and growth of disseminated tumor cells, the formation of premetastatic niches in target organs involves a complex interplay between microenvironment, immune system, circulating tumor cells, as well as chemical mediators and exosomes secreted by the primary tumor. This review describes the current understanding of the early mechanisms of dissemination, as well as how surgery may facilitate disease progression.
Collapse
Affiliation(s)
- Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ali Salanti
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
141
|
Kobold S, Reck M. Interleukin-1β und pro-tumorale Inflammation – Zentrale Faktoren bei der Entstehung von Krebserkrankungen. Oncol Res Treat 2020. [DOI: 10.1159/000510547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
142
|
Circulating Extracellular Vesicle MicroRNA as Diagnostic Biomarkers in Early Colorectal Cancer-A Review. Cancers (Basel) 2019; 12:cancers12010052. [PMID: 31878015 PMCID: PMC7016718 DOI: 10.3390/cancers12010052] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/11/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies in the developed world, with global deaths expected to double in the next decade. Disease stage at diagnosis is the single greatest prognostic indicator for long-term survival. Unfortunately, early stage CRC is often asymptomatic and diagnosis frequently occurs at an advanced stage, where long-term survival can be as low as 14%. Circulating microRNAs encapsulated in extracellular vesicles (EVs) have recently come to prominence as novel diagnostic markers for cancer. EV-miRNAs are dysregulated in the circulation of CRC patients compared to healthy controls, and several specific miRNA candidates have been posited as diagnostic markers, including miR-21, miR-23a, miR-1246, and miR-92a. This review outlines the current landscape of EV-miRNAs as potential diagnostic markers for CRC, with a specific focus on those able to detect early stage disease.
Collapse
|
143
|
Kong J, Tian H, Zhang F, Zhang Z, Li J, Liu X, Li X, Liu J, Li X, Jin D, Yang X, Sun B, Guo T, Luo Y, Lu Y, Lin B, Liu T. Extracellular vesicles of carcinoma-associated fibroblasts creates a pre-metastatic niche in the lung through activating fibroblasts. Mol Cancer 2019; 18:175. [PMID: 31796058 PMCID: PMC6892147 DOI: 10.1186/s12943-019-1101-4] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023] Open
Abstract
Objectives Carcinoma-associated fibroblasts (CAFs) have been known to promote cancer progression by modifying the primary tumor microenvironment. We aimed to elucidate the intercellular communication between CAFs and secondary organs in salivary adenoid cystic carcinoma (SACC) metastasis. Methods Pre-metastatic and metastatic animal models of SACC were established using extracellular vesicles (EVs) from CAFs and SACC cells. Lung fibroblasts (LFs) were treated with EVs and their transcriptomic alterations were identified by RNA sequencing. ITRAQ were performed to analyze EV cargos. TC I-15 was used to inhibit EV uptake by LFs and SACC lung metastasis in vivo. Results Here, we show that CAF EVs induced lung pre-metastatic niche formation in mice and consequently increased SACC lung metastasis. The pre-metastatic niche induced by CAF EVs was different from that induced by SACC EVs. CAF EVs presented a great ability for matrix remodeling and periostin is a potential biomarker characterizing the CAF EV-induced pre-metastatic niche. We found that lung fibroblast activation promoted by CAF EVs was a critical event at the pre-metastatic niche. Integrin α2β1 mediated CAF EV uptake by lung fibroblasts, and its blockage by TC I-15 prevented lung pre-metastatic niche formation and subsequent metastasis. Plasma EV integrin β1 was considerably upregulated in the mice bearing xenografts with high risk of lung metastasis. Conclusions We demonstrated that CAF EVs participated in the pre-metastatic niche formation in the lung. Plasma EV integrin β1 might be a promising biomarker to predict SACC metastasis at an early stage. An integrated strategy targeting both tumor and stromal cells is necessary to prevent SACC metastasis.
Collapse
Affiliation(s)
- Jing Kong
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Hongzhu Tian
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Fuyin Zhang
- Department of Oral Surgery, the Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Zebing Zhang
- Department of Oral Pathology, College of Stomatology, Jilin University, Changchun, China
| | - Jiao Li
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Xue Liu
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Xiancheng Li
- Department of Urology, the Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jing Liu
- Sino-UK Regenerative Medicine Center, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiaojie Li
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Dong Jin
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Xuesong Yang
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, China
| | - Bo Sun
- Department of Oral Surgery, the Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yong Luo
- Faculty of Chemical, Environmental and Biological Science and Technology, Dalian Technology University, Dalian, China
| | - Yao Lu
- Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Bingcheng Lin
- Faculty of Chemical, Environmental and Biological Science and Technology, Dalian Technology University, Dalian, China.,Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Tingjiao Liu
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China.
| |
Collapse
|
144
|
Herrera M, Galindo-Pumariño C, García-Barberán V, Peña C. A Snapshot of The Tumor Microenvironment in Colorectal Cancer: The Liquid Biopsy. Int J Mol Sci 2019; 20:ijms20236016. [PMID: 31795332 PMCID: PMC6929174 DOI: 10.3390/ijms20236016] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022] Open
Abstract
The molecular profile of liquid biopsies is emerging as an alternative to tissue biopsies in the clinical management of malignant diseases. In colorectal cancer, significant liquid biopsy-based biomarkers have demonstrated an ability to discriminate between asymptomatic cancer patients and healthy controls. Furthermore, this non-invasive approach appears to provide relevant information regarding the stratification of tumors with different prognoses and the monitoring of treatment responses. This review focuses on the tumor microenvironment components which are detected in blood samples of colorectal cancer patients and might represent potential biomarkers. Exosomes released by tumor and stromal cells play a major role in the modulation of cancer progression in the primary tumor microenvironment and in the formation of an inflammatory pre-metastatic niche. Stromal cells-derived exosomes are involved in driving mechanisms that promote tumor growth, migration, metastasis, and drug resistance, therefore representing substantial signaling mediators in the tumor-stroma interaction. Besides, recent findings of specifically packaged exosome cargo in Cancer-Associated Fibroblasts of colorectal cancer patients identify novel exosomal biomarkers with potential clinical applicability. Furthermore, additional different signals emitted from the tumor microenvironment and also detectable in the blood, such as soluble factors and non-tumoral circulating cells, arise as novel promising biomarkers for cancer diagnosis, prognosis, and treatment response prediction. The therapeutic potential of these factors is still limited, and studies are in their infancy. However, innovative strategies aiming at the inhibition of tumor progression by systemic exosome depletion, exosome-mediated circulating tumor cell capturing, and exosome-drug delivery systems are currently being studied and may provide considerable advantages in the near future.
Collapse
Affiliation(s)
- Mercedes Herrera
- Department of Oncology-Pathology, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - Cristina Galindo-Pumariño
- Medical Oncology Department, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, 28034 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), 28029 Madrid, Spain
| | - Vanesa García-Barberán
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), 28029 Madrid, Spain
- Laboratorio de Oncología Molecular, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
- Correspondence: (V.G.-B.); (C.P.)
| | - Cristina Peña
- Medical Oncology Department, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, 28034 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), 28029 Madrid, Spain
- Correspondence: (V.G.-B.); (C.P.)
| |
Collapse
|
145
|
Exosomes: Biogenesis, Composition, Functions, and Their Role in Pre-metastatic Niche Formation. BIOTECHNOL BIOPROC E 2019. [DOI: 10.1007/s12257-019-0170-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
146
|
The CXCL5/CXCR2 axis is sufficient to promote breast cancer colonization during bone metastasis. Nat Commun 2019; 10:4404. [PMID: 31562303 PMCID: PMC6765048 DOI: 10.1038/s41467-019-12108-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 08/16/2019] [Indexed: 12/28/2022] Open
Abstract
Bone is one of the most common sites for metastasis across cancers. Cancer cells that travel through the vasculature and invade new tissues can remain in a non-proliferative dormant state for years before colonizing the metastatic site. Switching from dormancy to colonization is the rate-limiting step of bone metastasis. Here we develop an ex vivo co-culture method to grow cancer cells in mouse bones to assess cancer cell proliferation using healthy or cancer-primed bones. Profiling soluble factors from conditioned media identifies the chemokine CXCL5 as a candidate to induce metastatic colonization. Additional studies using CXCL5 recombinant protein suggest that CXCL5 is sufficient to promote breast cancer cell proliferation and colonization in bone, while inhibition of its receptor CXCR2 with an antagonist blocks proliferation of metastatic cancer cells. This study suggests that CXCL5 and CXCR2 inhibitors may have efficacy in treating metastatic bone tumors dependent on the CXCL5/CXCR2 axis.
Collapse
|
147
|
Li R, Qi Y, Jiang M, Zhang T, Wang H, Wang L, Han M. Primary tumor-secreted VEGF induces vascular hyperpermeability in premetastatic lung via the occludin phosphorylation/ubiquitination pathway. Mol Carcinog 2019; 58:2316-2326. [PMID: 31553086 DOI: 10.1002/mc.23120] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/03/2019] [Accepted: 09/10/2019] [Indexed: 12/31/2022]
Abstract
Primary tumor can induce the formation of premetastatic niche. The hyperpermeability of the vessels in the premetastatic niche is the first step in the development of metastasis. However, the cellular and molecular mechanisms of vascular hyperpermeability remain to be elucidated. In this study, 4T1 breast cells were injected into the breasts of mice to establish a tumor model. Our results showed that primary tumors induced hyperpermeability of the vessels in the premetastatic lung. Subsequent studies showed that the level of vascular endothelial growth factor (VEGF) was elevated in the tumor-bearing mice serum and the levels of tight junction (TJ) proteins occludin and ZO-1 were decreased in the premetastatic lung. In vitro studies demonstrated that VEGF increased the permeability of dextran and decreased the levels of occludin and ZO-1 in human umbilical vein endothelial cells. Moreover, the hyperpermeability of vessels and the degradation of occludin was blocked by bevacizumab. Overexpression of occludin alleviated the VEGF-induced hyperpermeability. Further investigations revealed that VEGF-induced occludin phosphorylation at Ser-490 and ubiquitination. Finally, we showed that VEGF accelerated the process of occludin degradation through the ubiquitin-proteasome system. In conclusion, primary tumor-secrete VEGF induce the occludin phosphorylation/ubiquitination and downregulation, resulting in the disruption of TJs and hyperpermeability of vessels in premetastatic lung. The occludin phosphorylation/ubiquitination pathway may be the mechanism of VEGF-induced vascular hyperpermeability in the lung premetastatic niche.
Collapse
Affiliation(s)
- Ranran Li
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yana Qi
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Man Jiang
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Tiehong Zhang
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Hongwei Wang
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Liguang Wang
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Mingyong Han
- Cancer Therapy and Research Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
148
|
Sellner F. Observations on Solitary Versus Multiple Isolated Pancreatic Metastases of Renal Cell Carcinoma: Another Indication of a Seed and Soil Mechanism? Cancers (Basel) 2019; 11:E1379. [PMID: 31533220 PMCID: PMC6770877 DOI: 10.3390/cancers11091379] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 12/15/2022] Open
Abstract
Isolated pancreas metastases are a rare type of metastasis of renal cell carcinoma, characterized by the presence of pancreatic metastases, while all other organs remain unaffected. In a previous study, we determined arguments from the literature which (a) indicate a systemic-haematogenic metastasis route (uniform distribution of the metastases across the pancreas and independence of the metastatic localization in the pancreas of the side of the renal carcinoma); and (b) postulate a high impact of a seed and soil mechanism (SSM) on isolated pancreatic metastasis of renal cell carcinoma (isPM) as an explanation for exclusive pancreatic metastases, despite a systemic haematogenous tumor cell embolization. The objective of the study presented was to search for further arguments in favor of an SSM with isPM. For that purpose, the factor's histology, grading, and singular/multiple pancreas metastases were analyzed on the basis of 814 observations published up to 2018. While histology and grading allowed for no conclusions regarding the importance of an SSM, the comparison of singular/multiple pancreas metastases produced arguments in favor of an SSM: 1. The multiple pancreas metastases observed in 38.1% prove that multiple tumor cell embolisms occur with isPM, the exclusive "maturation" of which in the pancreas requires an SSM; 2. The survival rates (SVR), which are consistent with singular and multiple pancreas metastases (despite the higher total tumor load with the latter), prove that the metastasized tumor cells are not able to survive in all other organs because of an SSM, which results in identical SVR when the pancreatic foci are treated adequately.
Collapse
Affiliation(s)
- Franz Sellner
- Surgical Department, Kaiser-Franz-Josef-Hospital, 1100 Wien, Austria.
| |
Collapse
|
149
|
The double inhibition of PDK1 and STAT3-Y705 prevents liver metastasis in colorectal cancer. Sci Rep 2019; 9:12973. [PMID: 31506552 PMCID: PMC6736869 DOI: 10.1038/s41598-019-49480-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 08/23/2019] [Indexed: 12/20/2022] Open
Abstract
As a key glycolysis enzyme, the significance of pyruvate dehydrogenase kinase 1 (PDK1) in the development of colorectal cancer (CRC) remains unknown. This study revealed that the prognosis of CRC patients with high levels of PDK1 was poor, and PDK1 knockdown significantly reduced liver metastasis of CRC in both nude mice and immune competent BALB/C mice. When combined with cryptotanshinone (CPT), an inhibitor of STAT3-p-Y705, the liver metastasis was further inhibited. PDK1 knockdown obviously increased reactive oxygen species level in anoikis conditions and subsequently resulted in an elevated anoikis, but the combination of PDK1 knockdown and CPT showed a reduced effect on anoikis. Based on this discrepancy, the adherence ability of CRC cells to matrix protein fibronectin was further detected. It showed that PDK1 knockdown significantly decreased the adherence of CRC cells to fibronectin when combined with CPT. These results suggest that inhibition of PDK1 can decrease the surviving CRC cells in blood circulation via up-regulation of anoikis, and inhibition of STAT3-p-Y705 can prevent it to settle down on the liver premetastatic niche, which ultimately reduces liver metastasis.
Collapse
|
150
|
Bosiljcic M, Cederberg RA, Hamilton MJ, LePard NE, Harbourne BT, Collier JL, Halvorsen EC, Shi R, Franks SE, Kim AY, Banáth JP, Hamer M, Rossi FM, Bennewith KL. Targeting myeloid-derived suppressor cells in combination with primary mammary tumor resection reduces metastatic growth in the lungs. Breast Cancer Res 2019; 21:103. [PMID: 31488209 PMCID: PMC6727565 DOI: 10.1186/s13058-019-1189-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 08/16/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Solid tumors produce proteins that can induce the accumulation of bone marrow-derived cells in various tissues, and these cells can enhance metastatic tumor growth by several mechanisms. 4T1 murine mammary tumors are known to produce granulocyte colony-stimulating factor (G-CSF) and increase the numbers of immunosuppressive CD11b+Gr1+ myeloid-derived suppressor cells (MDSCs) in tissues such as the spleen and lungs of tumor-bearing mice. While surgical resection of primary tumors decreases MDSC levels in the spleen, the longevity and impact of MDSCs and other immune cells in the lungs after tumor resection have been less studied. METHODS We used mass cytometry time of flight (CyTOF) and flow cytometry to quantify MDSCs in the spleen, peripheral blood, and lungs of mice bearing orthotopic murine mammary tumors. We also tested the effect of primary tumor resection and/or gemcitabine treatment on the levels of MDSCs, other immune suppressor and effector cells, and metastatic tumor cells in the lungs. RESULTS We have found that, similar to mice with 4T1 tumors, mice bearing metastatic 4T07 tumors also exhibit accumulation of CD11b+Gr1+ MDSCs in the spleen and lungs, while tissues of mice with non-metastatic 67NR tumors do not contain MDSCs. Mice with orthotopically implanted 4T1 tumors have increased granulocytic (G-) MDSCs, monocytic (M-) MDSCs, macrophages, eosinophils, and NK cells in the lungs. Resection of primary 4T1 tumors decreases G-MDSCs, M-MDSCs, and macrophages in the lungs within 48 h, but significant numbers of functional immunosuppressive G-MDSCs persist in the lungs for 2 weeks after tumor resection, indicative of an environment that can promote metastatic tumor growth. The chemotherapeutic agent gemcitabine depletes G-MDSCs, M-MDSCs, macrophages, and eosinophils in the lungs of 4T1 tumor-bearing mice, and we found that treating mice with gemcitabine after primary tumor resection decreases residual G-MDSCs in the lungs and decreases subsequent metastatic growth. CONCLUSIONS Our data support the development of therapeutic strategies to target MDSCs and to monitor MDSC levels before and after primary tumor resection to enhance the effectiveness of immune-based therapies and improve the treatment of metastatic breast cancer in the clinic.
Collapse
Affiliation(s)
- Momir Bosiljcic
- Integrative Oncology Department, BC Cancer Research Centre, Room 10-108, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada.,Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rachel A Cederberg
- Integrative Oncology Department, BC Cancer Research Centre, Room 10-108, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada.,Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Melisa J Hamilton
- Integrative Oncology Department, BC Cancer Research Centre, Room 10-108, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada
| | - Nancy E LePard
- Integrative Oncology Department, BC Cancer Research Centre, Room 10-108, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada
| | - Bryant T Harbourne
- Integrative Oncology Department, BC Cancer Research Centre, Room 10-108, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada.,Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jenna L Collier
- Integrative Oncology Department, BC Cancer Research Centre, Room 10-108, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada
| | - Elizabeth C Halvorsen
- Integrative Oncology Department, BC Cancer Research Centre, Room 10-108, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada.,Interdisciplinary Oncology Program, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rocky Shi
- Integrative Oncology Department, BC Cancer Research Centre, Room 10-108, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada
| | - S Elizabeth Franks
- Integrative Oncology Department, BC Cancer Research Centre, Room 10-108, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada
| | - Ada Y Kim
- Integrative Oncology Department, BC Cancer Research Centre, Room 10-108, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada.,Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Judit P Banáth
- Integrative Oncology Department, BC Cancer Research Centre, Room 10-108, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada
| | - Mark Hamer
- Biomedical Research Centre, University of British Columbia, 2222 Health Sciences Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Fabio M Rossi
- Biomedical Research Centre, University of British Columbia, 2222 Health Sciences Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Kevin L Bennewith
- Integrative Oncology Department, BC Cancer Research Centre, Room 10-108, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada. .,Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada. .,Interdisciplinary Oncology Program, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|