1
|
Ribatti D. Mast cell proteases and metastasis. Pathol Res Pract 2024; 266:155801. [PMID: 39755049 DOI: 10.1016/j.prp.2024.155801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/12/2024] [Accepted: 12/25/2024] [Indexed: 01/06/2025]
Abstract
Mast cells exert multiple roles beyond their classical role in IgE-mediated allergic reactions. These cells secrete pro-inflammatory and anti-inflammatory agents and change from protective immune cells to pro-inflammatory cells, capable of influencing the progression of different pathological conditions, including tumors, in which they exert anti-tumorigenic and pro-tumorigenic roles. In this context, this article analyzes the potential role played by mast cell-derived proteases in tumor progression and more specifically in driving metastatic process and the potential therapeutic approaches that inhibiting the activation of these cells could help faith cancer spreading.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
2
|
Jiang Y, Lu L. New insight into the agonism of protease-activated receptors as an immunotherapeutic strategy. J Biol Chem 2024; 300:105614. [PMID: 38159863 PMCID: PMC10810747 DOI: 10.1016/j.jbc.2023.105614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024] Open
Abstract
The activation and mobilization of immune cells play a crucial role in immunotherapy. Existing therapeutic interventions, such as cytokines administration, aim to enhance immune cell activity. However, these approaches usually result in modest effectiveness and toxic side effects, thereby restricting their clinical application. Protease-activated receptors (PARs), a subfamily of G protein-coupled receptors, actively participate in the immune system by directly activating immune cells. The activation of PARs by proteases or synthetic ligands can modulate immune cell behavior, signaling, and responses to treat immune-related diseases, suggesting the significance of PARs agonism in immunotherapy. However, the agonism of PARs in therapeutical applications remains rarely discussed, since it has been traditionally considered that PARs activation facilitates disease progressions. This review aims to comprehensively summarize the activation, rather than inhibition, of PARs in immune-related physiological responses and diseases. Additionally, we will discuss the emerging immunotherapeutic potential of PARs agonism, providing a new strategic direction for PARs-mediated immunotherapy.
Collapse
Affiliation(s)
- Yuhong Jiang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China.
| | - Lei Lu
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
3
|
Shen K, Ke S, Chen B, Gao W. Integrated analysis of single-cell and bulk RNA-sequencing reveals the poor prognostic value of ABCA1 in gastric adenocarcinoma. Discov Oncol 2023; 14:189. [PMID: 37874419 PMCID: PMC10597929 DOI: 10.1007/s12672-023-00807-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023] Open
Abstract
PURPOSE ATP-binding cassette A1 (ABCA1) is a potential prognostic marker for various tumor types. However, the biological effects and prognostic value of ABCA1 in gastric adenocarcinoma (GAC) remain unknown. METHODS GAC-associated single-cell RNA and bulk RNA-sequencing (bulk-seq) data were obtained from the Gene Expression Omnibus and The Cancer Genome Atlas databases, respectively. The differential expression of ABCA1 between GAC and normal gastric tissues was analyzed based on the bulk-seq data. Additionally, the relationship between ABCA1 expression and various clinicopathological features was explored. Furthermore, Kaplan-Meier survival and Cox regression analyses were performed to establish the prognostic value of ABCA1. The relationships between ABCA1 expression and anti-tumor drug sensitivity and immune checkpoints were also explored. Finally, the biological functions of ABCA1 were evaluated at the single-cell level, and in vitro studies were performed to assess the effects of ABCA1 on GAC cell proliferation and invasion. RESULTS ABCA1 expression is significantly elevated in GAC samples compared with that in normal gastric tissues. Clinical features and survival analysis revealed that high ABCA1 expression is associated with poor clinical phenotypes and prognosis, whereas Cox analysis identified ABCA1 as an independent risk factor for patients with GAC. Furthermore, high ABCA1 expression suppresses sensitivity to various chemotherapeutic drugs, including cisplatin and mitomycin, while upregulating immune checkpoints. ABCA1-overexpressing macrophages are associated with adverse clinical phenotypes in GAC and express unique ligand-receptor pairs that drive GAC progression. In vitro, ABCA1-knockdown GAC cells exhibit significantly inhibited proliferative and invasive properties. CONCLUSION High ABCA1 expression promotes an adverse immune microenvironment and low survival rates in patients with GAC. Furthermore, ABCA1 and ABCA1-producing macrophages may serve as novel molecular targets in GAC treatment.
Collapse
Affiliation(s)
- Kaiyu Shen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shuaiyi Ke
- Department of Internal Medicine, Affiliated Xianju's Hospital, XianJu People's Hospital, Zhejiang Southeast Campus of Zhejiang Provincial People's Hospital, Hangzhou Medical College, XianJu, 317399, China
| | - Binyu Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Wencang Gao
- Department of Oncology, the Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| |
Collapse
|
4
|
Targeted Therapy for Hepatocellular Carcinoma: Old and New Opportunities. Cancers (Basel) 2022; 14:cancers14164028. [PMID: 36011021 PMCID: PMC9406380 DOI: 10.3390/cancers14164028] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 12/05/2022] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is the most frequent primitive cancer of the liver, accounting for 90% of all recorded cases. HCC is the third most common cause of cancer-related death, with a 5-year survival rate of just 3%. In terms of the advanced stages, systemic treatments have allowed patients to achieve clinical benefits, although the prognosis remains very poor. In the past few decades, new molecular targeted therapies have been developed and clinically evaluated with interesting results. However, on the basis of the poor prognoses and the meager benefits deriving from the available systemic therapies, research into new treatments is extremely necessary. In this review, we focus on the available systemic therapies for advanced HCC, with a look toward the future. Abstract Hepatocellular carcinoma (HCC) is the most frequent primitive cancer of the liver, accounting for 90% of all recorded cases. HCC is the third most common cause of cancer-related death, with a 5-year survival rate of just 3%. In the advanced stages, systemic treatments allow doctors to obtain clinical benefits, although the prognosis remains very poor. In the past few decades, new molecular targeted therapies against receptor tyrosine kinases have been developed and clinically evaluated. Sorafenib was the first oral tyrosine kinase inhibitor (TKI) approved for the treatment of advanced HCC in 2007. Subsequently, other TKIs, including Cabozantinib, Regorafenib, Lenvatinib, and vascular endothelial growth factor receptor (VEGFR) inhibitors such as Ramucirumab and VEGF inhibitors such as Bevacizumab have been approved as first- or second-line treatments. More recently, the combination of immune checkpoint inhibitors and VEGF inhibitors (Atezolizumab plus Bevacizumab) have been analyzed and approved for the treatment of advanced HCC. On the basis of the poor prognoses and the meager benefits deriving from the available systemic therapies, research into new treatments is extremely necessary. In this review, we focus on the available systemic therapies for advanced HCC, with a look toward the future.
Collapse
|
5
|
Huang L, Zhao Y, Shi Y, Hu W, Zhang J. Bone Metastasis From Gastric Adenocarcinoma-What Are the Risk Factors and Associated Survival? A Large Comprehensive Population-Based Cohort Study. Front Oncol 2022; 12:743873. [PMID: 35402215 PMCID: PMC8989732 DOI: 10.3389/fonc.2022.743873] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 02/28/2022] [Indexed: 01/19/2023] Open
Abstract
Background While bone metastasis is not common in gastric adenocarcinoma (GaC), it can have important impacts on prognosis. This large cohort study aimed at exploring factors associated with bone metastasis in GaC and investigating the time-dependent cumulative mortalities and prognostic factors in GaC patients with bone metastasis at the population level. Methods Data on patients with GaC diagnosed in 2010–2016 were retrieved from a large population-based database. We explored factors associated with bone metastasis using the multivariable-adjusted logistic model. We then calculated the time-dependent cancer-specific mortalities in GaC patients with bone metastasis using the cumulative incidence function and compared mortalities across subgroups using Gray’s test. We further assessed factors associated with mortality using the multivariable-adjusted Fine–Gray subdistribution hazard model. Results Together 11,072 eligible patients with metastatic GaC were enrolled, which comprised 1,511 (14%) people with bone metastasis and 9,561 (86%) with other metastasis, encompassing 6,999 person-years of follow-up. Bone metastasis was more frequently detected in 2014 or later, in younger patients, in patients with gastric cardia cancers, in people with signet-ring cell carcinoma, and in those with poorly differentiated/undifferentiated cancers; it was less commonly observed in black patients. Bone metastasis was associated with more frequent brain and lung metastases. The median survival of patients with bone metastasis was 4 months; the 6-month and 3-year cancer-specific cumulative mortalities were 56% and 85%, respectively. In patients receiving chemotherapy, American Indians/Alaskan Natives, patients with gastric antrum/pylorus cancers, and those with positive lymph nodes had higher mortality risks, while those undergoing resection had lower mortality hazards. Conclusion In GaC patients, bone metastasis was associated with various clinicopathologic factors including age, ethnicity, tumor location, histology, differentiation, and metastasis to other sites. Patients with bone metastasis had poor prognosis which was associated with ethnicity, tumor location, lymph node involvement, and treatment. Our findings provide important hints for tailed patient management and for further mechanistic investigations.
Collapse
Affiliation(s)
- Lei Huang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yajie Zhao
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Shi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiguo Hu
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
6
|
Analysis of Related Risk Factors and Prognostic Factors of Gastric Cancer with Bone Metastasis: A SEER-Based Study. J Immunol Res 2022; 2022:3251051. [PMID: 35211630 PMCID: PMC8863473 DOI: 10.1155/2022/3251051] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/06/2022] [Indexed: 12/17/2022] Open
Abstract
Background Gastric cancer is among the most common malignant tumors at home and abroad, because its early symptoms are mostly insidious, which leads to distant metastasis when gastric cancer is first diagnosed. The common metastatic sites of gastric cancer are mainly the liver, lung, and peritoneum, but bone metastasis is relatively rare, and the prognosis of gastric cancer bone metastasis is very poor. Therefore, this study is built on the SEER database to analyze the related risk factors of gastric cancer bone metastasis and related factors affecting the prognosis of gastric cancer patients, aiming at improving clinicians' understanding of clinical diagnosis and prognosis of bone metastasis of gastric cancer, thus reducing misdiagnosis and missed diagnosis. Methods The SEER database was collected to screen out patients with gastric cancer bone metastases and nonbone metastases matched with them from 2010 to 2016, and the Kaplan-Meier method was used to draw survival curves, and the comparison between survival curves was performed by Log-rank test to analyze the overall survival of the two groups of patient's time. Logistic regression analysis was used to analyze the related risk factors of gastric cancer bone metastasis, and the Cox regression proportional hazard model was used to analyze the relationship between gastric cancer bone metastasis and patient prognosis. Results Using Kaplan-Meier survival curve to analyze the 1, 3, and 5-year survival rates of gastric cancer patients with bone metastasis and non-metastasis groups were 14.2%, 1.8%, 0.6% and 71.4%, 44.3%, 36.4%, respectively; the average survival rate of the metastatic group was The time was 4.0 months (95%CI: 3.475~4.525), and the average survival time of the non-metastatic group was 30.0 months (95%CI: 26.778~33.222). The difference between the two groups was statistically significant (χ2 = 1076.866, P < 0.001). Multivariate logistic regression analysis showed that race (P = 0.007, OR = 1.296), grade (P < 0.001, OR = 0.575), marital status (P < 0.001, OR = 0.040), tumor size (P = 0.006, OR = 0.752), TNM stage (P < 0.001), T stage (P = 0.023, OR = 0.882), and M stage (P < 0.001, OR = 44.958) are independent risk factors for gastric cancer bone metastasis. The Cox univariate analysis suggests that gastric cancer bone metastasis is a risk factor for the prognosis of gastric cancer patients. The Cox multivariate analysis validates that gastric cancer bone metastasis (HR = 0.584, 95% CI: 0.497~0.688, P < 0.001) is independent of the overall survival rate of gastric cancer patients. Conclusions Race, grade, marital status, tumor size, TNM stage, T stage, and M stage are independent risk factors for gastric cancer bone metastasis; and gastric cancer bone metastasis is an independent risk factor that affects the prognosis of gastric cancer patients. Therefore, for such high-risk groups, large range screening of the above indicators can effectively improve the prognosis of gastric cancer patients to a certain extent.
Collapse
|
7
|
Alsamraae M, Cook LM. Emerging roles for myeloid immune cells in bone metastasis. Cancer Metastasis Rev 2021; 40:413-425. [PMID: 33855680 DOI: 10.1007/s10555-021-09965-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/08/2021] [Indexed: 12/11/2022]
Abstract
Metastasis, especially bone metastasis, is a major cause of cancer-related deaths, which is associated with long-term pain due to skeletal-related events and poor quality of life. Tumor cells alter the bone microenvironment through aberrant activation of osteoclasts and osteoblasts which induces bone osteolysis and release of growth factors leading to cancer growth. Though this phenomenon has been well characterized, bone-targeted therapies have shown little improvement in patient survival. Recent evidence indicates a growing appreciation for the complex bone environment, in addition to bone-remodeling stromal cells, which includes an abundance of myeloid immune cells that can either protect against or contribute to the progression of the disease within the bone cavity. Additionally, myeloid cells are recruited into primary tumor sites, where they promote development of the pre-metastatic niche and also can regulate tumor progression within the tumor-bone microenvironment through a milieu of complex mechanisms and involving heterogeneous myeloid populations. In this review, we have highlighted the complex roles of myeloid immunity in bone metastasis and hope to bring attention to the potential of novel immunotherapeutic interventions for the elimination of bone metastasis.
Collapse
Affiliation(s)
- Massar Alsamraae
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Leah M Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA. .,Fred & Pamela Buffett Cancer Center, Omaha, NE, USA.
| |
Collapse
|
8
|
Mast Cells Positive for c-Kit Receptor and Tryptase Correlate with Angiogenesis in Cancerous and Adjacent Normal Pancreatic Tissue. Cells 2021; 10:cells10020444. [PMID: 33669751 PMCID: PMC7923170 DOI: 10.3390/cells10020444] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/16/2021] [Accepted: 02/16/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Mast cells (MCs) contain proangiogenic factors, in particular tryptase, associated with increased angiogenesis in several tumours. With special reference to pancreatic cancer, few data have been published on the role of MCs in angiogenesis in both pancreatic ductal adenocarcinoma tissue (PDAT) and adjacent normal tissue (ANT). In this study, density of mast cells positive for c-Kit receptor (MCDP-c-KitR), density of mast cells positive for tryptase (MCDPT), area of mast cells positive for tryptase (MCAPT), and angiogenesis in terms of microvascular density (MVD) and endothelial area (EA) were evaluated in a total of 45 PDAT patients with stage T2–3N0–1M0. Results: For each analysed tissue parameter, the mean ± standard deviation was evaluated in both PDAT and ANT and differences were evaluated by Student’s t-test (p ranged from 0.001 to 0.005). Each analysed tissue parameter was then correlated to each other one by Pearson t-test analysis (p ranged from 0.01 to 0.03). No other correlation among MCDP-c-KitR, MCDPT, MCAPT, MVD, EA and the main clinical–pathological characteristics was found. Conclusions: Our results suggest that tissue parameters increased from ANT to PDAT and that mast cells are strongly associated with angiogenesis in PDAT. On this basis, the inhibition of MCs through tyrosine kinase inhibitors, such as masitinib, or inhibition of tryptase by gabexate mesylate may become potential novel antiangiogenetic approaches in pancreatic cancer therapy.
Collapse
|
9
|
Duguay BA, Lu L, Arizmendi N, Unsworth LD, Kulka M. The Possible Uses and Challenges of Nanomaterials in Mast Cell Research. THE JOURNAL OF IMMUNOLOGY 2020; 204:2021-2032. [PMID: 32253270 DOI: 10.4049/jimmunol.1800658] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 12/19/2019] [Indexed: 11/19/2022]
Abstract
Mast cells are tissue-resident immune cells that are involved in inflammation and fibrosis but also serve beneficial roles, including tissue maintenance, angiogenesis, pathogen clearance, and immunoregulation. Their multifaceted response and the ability of their mediators to target multiple organs and tissues means that mast cells play important roles in numerous conditions, including asthma, atopic dermatitis, drug sensitivities, ischemic heart disease, Alzheimer disease, arthritis, irritable bowel syndrome, infections (parasites, bacteria and viruses), and cancer. As a result, mast cells have become an important target for drug discovery and diagnostic research. Recent work has focused on applying novel nanotechnologies to explore cell biology. In this brief review, we will highlight the use of nanomaterials to modify mast cell functions and will discuss the potential of these technologies as research tools for understanding mast cell biology.
Collapse
Affiliation(s)
- Brett A Duguay
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, Alberta T6G 2M9, Canada
| | - Lei Lu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, People's Republic of China
| | - Narcy Arizmendi
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, Alberta T6G 2M9, Canada
| | - Larry D Unsworth
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada; and
| | - Marianna Kulka
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, Alberta T6G 2M9, Canada.,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| |
Collapse
|
10
|
Li K, Fan J, Qin X, Wei Q. Novel therapeutic compounds for prostate adenocarcinoma treatment: An analysis using bioinformatic approaches and the CMap database. Medicine (Baltimore) 2020; 99:e23768. [PMID: 33371142 PMCID: PMC7748316 DOI: 10.1097/md.0000000000023768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 11/17/2020] [Indexed: 01/15/2023] Open
Abstract
INTRODUCTION Prostate adenocarcinoma is the most frequently diagnosed malignancy, particularly for people >70 years old. The main challenge in the treatment of advanced neoplasm is bone metastasis and therapeutic resistance for known oncology drugs. Novel treatment methods to prolong the survival time and improve the life quality of these specific patients are required. The present study attempted to screen potential therapeutic compounds for the tumor through bioinformatics approaches, in order to provide conceptual treatment for this malignant disease. METHODS Differentially expressed genes were obtained from the Gene Expression Omnibus database and submitted into the Connectivity Map database for the detection of potentially associated compounds. Target genes were extracted from the search results. Functional annotation and pathway enrichment were performed for the confirmation. Survival analysis was used to measure potential therapeutic effects. RESULTS It was revealed that 3 compounds (vanoxerine, tolnaftate, and gabexate) may help to prolong the disease-free survival time from tumor metastasis of patients with the tumor. A total of 6 genes [also-keto reductase family 1 member C3 (AKR1C3), collagen type III α 1 chain (COL3A1), lipoprotein lipase (LPL), glucuronidase, β pseudogene 11 (GUSBP11), apolipoprotein E (APOE), and collagen type I α 1 chain (COL1A1)] were identified to be the potential therapeutic targets for the aforementioned compounds. CONCLUSION In the present study, it was speculated that 3 compounds may function as the potential therapeutic drugs of bone metastatic prostate adenocarcinoma; however, further studies verifying vitro and in vivo are necessary.
Collapse
Affiliation(s)
- Kai Li
- Departments of Orthopedics, The First Affiliated Hospital, Guangxi Medical University
| | - Jingyuan Fan
- Departments of Orthopedics, The First Affiliated Hospital, Guangxi Medical University
| | - Xinyi Qin
- Graduate School of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Qingjun Wei
- Departments of Orthopedics, The First Affiliated Hospital, Guangxi Medical University
| |
Collapse
|
11
|
Sammarco G, Varricchi G, Ferraro V, Ammendola M, De Fazio M, Altomare DF, Luposella M, Maltese L, Currò G, Marone G, Ranieri G, Memeo R. Mast Cells, Angiogenesis and Lymphangiogenesis in Human Gastric Cancer. Int J Mol Sci 2019; 20:E2106. [PMID: 31035644 PMCID: PMC6540185 DOI: 10.3390/ijms20092106] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/15/2019] [Accepted: 04/19/2019] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer is diagnosed in nearly one million new patients each year and it remains the second leading cause of cancer-related deaths worldwide. Although gastric cancer represents a heterogeneous group of diseases, chronic inflammation has been shown to play a role in tumorigenesis. Cancer development is a multistep process characterized by genetic and epigenetic alterations during tumour initiation and progression. The stromal microenvironment is important in maintaining normal tissue homeostasis or promoting tumour development. A plethora of immune cells (i.e., lymphocytes, macrophages, mast cells, monocytes, myeloid-derived suppressor cells, Treg cells, dendritic cells, neutrophils, eosinophils, natural killer (NK) and natural killer T (NKT) cells) are components of gastric cancer microenvironment. Mast cell density is increased in gastric cancer and there is a correlation with angiogenesis, the number of metastatic lymph nodes and the survival of these patients. Mast cells exert a protumorigenic role in gastric cancer through the release of angiogenic (VEGF-A, CXCL8, MMP-9) and lymphangiogenic factors (VEGF-C and VEGF-F). Gastric mast cells express the programmed death ligands (PD-L1 and PD-L2) which are relevant as immune checkpoints in cancer. Several clinical undergoing trials targeting immune checkpoints could be an innovative therapeutic strategy in gastric cancer. Elucidation of the role of subsets of mast cells in different human gastric cancers will demand studies of increasing complexity beyond those assessing merely mast cell density and microlocalization.
Collapse
Affiliation(s)
- Giuseppe Sammarco
- Department of Health Science, General Surgery, Magna Graecia University, Medicine School of Germaneto, 88100 Catanzaro, Italy.
| | - Gilda Varricchi
- Department of Translational Medical Sciences (DISMET) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy.
- WAO Center of Excellence, 80131 Naples, Italy.
| | - Valentina Ferraro
- Department of Biomedical Sciences and Human Oncology, Unit of Endocrine, Digestive and Emergency Surgery, Aldo Moro University, 74124 Bari, Italy.
| | - Michele Ammendola
- Department of Health Science, General Surgery, Magna Graecia University, Medicine School of Germaneto, 88100 Catanzaro, Italy.
| | - Michele De Fazio
- Department of Emergency and Organ Transplantation, Aldo Moro University, 74124 Bari, Italy.
| | | | - Maria Luposella
- Cardiovascular Disease Unit, San Giovanni di Dio Hospital, 88900 Crotone, Italy.
| | - Lorenza Maltese
- Pathology Unit, Pugliese-Ciaccio Hospital, 88100 Catanzaro, Italy.
| | - Giuseppe Currò
- Department of Health Science, General Surgery, Magna Graecia University, Medicine School of Germaneto, 88100 Catanzaro, Italy.
- Department of Human Pathology of Adult and Evolutive Age G. Barresi, University of Messina, 98122 Messina, Italy.
| | - Gianni Marone
- Department of Translational Medical Sciences (DISMET) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy.
- WAO Center of Excellence, 80131 Naples, Italy.
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), 80131 Naples, Italy.
| | - Girolamo Ranieri
- Interventional Oncology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, 74124 Bari, Italy.
| | - Riccardo Memeo
- Department of Emergency and Organ Transplantation, Aldo Moro University, 74124 Bari, Italy.
| |
Collapse
|
12
|
Park SH, Eber MR, Widner DB, Shiozawa Y. Role of the Bone Microenvironment in the Development of Painful Complications of Skeletal Metastases. Cancers (Basel) 2018; 10:cancers10050141. [PMID: 29747461 PMCID: PMC5977114 DOI: 10.3390/cancers10050141] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 01/02/2023] Open
Abstract
Cancer-induced bone pain (CIBP) is the most common and painful complication in patients with bone metastases. It causes a significant reduction in patient quality of life. Available analgesic treatments for CIBP, such as opioids that target the central nervous system, come with severe side effects as well as the risk of abuse and addiction. Therefore, alternative treatments for CIBP are desperately needed. Although the exact mechanisms of CIBP have not been fully elucidated, recent studies using preclinical models have demonstrated the role of the bone marrow microenvironment (e.g., osteoclasts, osteoblasts, macrophages, mast cells, mesenchymal stem cells, and fibroblasts) in CIBP development. Several clinical trials have been performed based on these findings. CIBP is a complex and challenging condition that currently has no standard effective treatments other than opioids. Further studies are clearly warranted to better understand this painful condition and develop more effective and safer targeted therapies.
Collapse
Affiliation(s)
- Sun H Park
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| | - Matthew R Eber
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| | - D Brooke Widner
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| | - Yusuke Shiozawa
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
13
|
Marech I, Ammendola M, Leporini C, Patruno R, Luposella M, Zizzo N, Passantino G, Sacco R, Farooqi AA, Zuccalà V, Leo S, Dentamaro R, Porcelli M, Gadaleta P, De Sarro G, Gadaleta CD, Ranieri G. C-Kit receptor and tryptase expressing mast cells correlate with angiogenesis in breast cancer patients. Oncotarget 2017; 9:7918-7927. [PMID: 29487702 PMCID: PMC5814269 DOI: 10.18632/oncotarget.23722] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 10/28/2017] [Indexed: 12/21/2022] Open
Abstract
C-Kit protein is a transmembrane tyrosine kinase (TK) receptor (c-KitR-TK), which is predominantly expressed on mast cells (MCs) playing a role in tumor angiogenesis. It could be also expressed on epithelial breast cancer cells (EBCCs), but no data have been published regarding the correlation between mast cells positive to c-KitR (MCs-c-KitR), EBCCs positive to c-KitR (EBCCs-c-KitR), BC angiogenesis in terms of microvessel density (MVD) and the main clinic-pathological features. This study aims to evaluate the above parameters and their correlations in a series of selected 121 female early BC patients. It has been found a strong correlation between MVD and MCDPT, and MCs-c-KitR, MVD and MCs density positive to tryptase (MCDPT), and MCs-c-KitR and MCDPT by Pearson correlation. These data suggest an involvement of both MCDPT and MCs-c-KitR in BC tumor angiogenesis. Furthermore, BC tissue expressing c-KitR could be a putative predictive factor to c-KitR-TK inhibitors. In this way, selected patients with higher MCs-c-KitR could be candidate to receive c-KitR-TK inhibitors (e.g. masitinib, sunitinib) or tryptase inhibitors (e.g. nafamostat mesilate, gabexate mesilate).
Collapse
Affiliation(s)
- Ilaria Marech
- Interventional and Medical Oncology Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy
| | - Michele Ammendola
- Department of Medical and Surgery Science Medical School, Clinical Surgery Unit, Magna Graecia University, 88100 Catanzaro, Italy
| | - Christian Leporini
- Department of Health Science, Clinical Pharmacology and Pharmacovigilance Unit, Pharmacovigilance's Centre Calabria Region, Magna Graecia University, Germaneto, 88100 Catanzaro, Italy
| | - Rosa Patruno
- Chair of Pathology, Veterinary Medical School, Aldo Moro University, 70010 Valenzano, Italy
| | - Maria Luposella
- Cardiovascular Disease Unit, San Giovanni di Dio Hospital, 88900 Crotone, Italy
| | - Nicola Zizzo
- Chair of Pathology, Veterinary Medical School, Aldo Moro University, 70010 Valenzano, Italy
| | - Giuseppe Passantino
- Chair of Pathology, Veterinary Medical School, Aldo Moro University, 70010 Valenzano, Italy
| | - Rosario Sacco
- Department of Medical and Surgery Science Medical School, Clinical Surgery Unit, Magna Graecia University, 88100 Catanzaro, Italy
| | - Ammad Ahmad Farooqi
- Laboratory for Translational and Personalized Medicine, Rashid Latif Medical College, University of Lahore, 44000 Islamabad, Pakistan
| | - Valeria Zuccalà
- Pathology Unit, Pugliese-Ciaccio Hospital, 88100 Catanzaro, Italy
| | - Silvana Leo
- Medical Oncology Unit, Vito Fazzi Hospital, Piazzetta Muratore, 73100 Lecce, Italy
| | - Rosalba Dentamaro
- Senology Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy
| | - Mariangela Porcelli
- Interventional and Medical Oncology Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy
| | - Pietro Gadaleta
- Interventional and Medical Oncology Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy
| | - Giovambattista De Sarro
- Department of Health Science, Clinical Pharmacology and Pharmacovigilance Unit, Pharmacovigilance's Centre Calabria Region, Magna Graecia University, Germaneto, 88100 Catanzaro, Italy
| | - Cosmo Damiano Gadaleta
- Interventional and Medical Oncology Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy
| | - Girolamo Ranieri
- Interventional and Medical Oncology Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy
| |
Collapse
|
14
|
Paolino G, Corsetti P, Moliterni E, Corsetti S, Didona D, Albanesi M, Mattozzi C, Lido P, Calvieri S. Mast cells and cancer. GIORN ITAL DERMAT V 2017; 154:650-668. [PMID: 29192477 DOI: 10.23736/s0392-0488.17.05818-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mast cells (MCs) are a potent proangiogenic factor in tumors, they product several pro-angiogenic factors such as fibroblast growth factor 2 (FGF-2), vascular epithelial growth factor (VEGF), tryptase and chymase. Tryptase is a serine protease classified as α-tryptase and β-tryptase, both produced by MCs. Tryptase degrades the tissues, playing an important role in angiogenesis and in the development of metastases. Serum tryptase increases with age, with increased damage to cells and risk of developing a malignancy and it could be considered the expression of a fundamental role of MCs in tumor growth or, on the contrary, in the antitumor response. Many biomarkers have been developed in clinical practice for improving diagnosis and prognosis of some neoplasms. Elevated tryptase levels are found in subgroups of patients with haematologic and solid cancers. In the current review, we want to update the perspectives of tryptase as a potential biomarker in daily practice in different neoplasms.
Collapse
Affiliation(s)
| | | | | | - Serena Corsetti
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, San Vito al Tagliamento, Pordenone, Italy -
| | - Dario Didona
- First Division of Dermatology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Marcello Albanesi
- Department of Emergency and Organ Transplantation, School of Allergology and Clinical Immunology, University of Bari Aldo Moro, Bari, Italy
| | | | - Paolo Lido
- Internal Medicine Residency Program, Tor Vergata University, Rome, Italy
| | | |
Collapse
|
15
|
Becerra-Pedraza LC, Carlos NG, Carmona GRC, Martínez-Piña DA. Uncommon Initial Presentation of Gastric Cancer with Bone Metastases: a Case Report. J Gastrointest Cancer 2017; 50:334-337. [PMID: 29047043 DOI: 10.1007/s12029-017-0015-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Luis Cuitláhuac Becerra-Pedraza
- General Hospital of Morelia Dr. Miguel Silva, Morelia, Michoacán, Mexico. .,, Av. Insurgentes Sur. # 3877. Col Fama, Tlalpan, Mexico City, Mexico.
| | | | | | | |
Collapse
|
16
|
Ammendola M, Gadaleta CD, Frampton AE, Piardi T, Memeo R, Zuccalà V, Luposella M, Patruno R, Zizzo N, Gadaleta P, Pessaux P, Sacco R, Sammarco G, Ranieri G. The density of mast cells c-Kit + and tryptase + correlates with each other and with angiogenesis in pancreatic cancer patients. Oncotarget 2017; 8:70463-70471. [PMID: 29050294 PMCID: PMC5642569 DOI: 10.18632/oncotarget.19716] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 06/24/2017] [Indexed: 12/16/2022] Open
Abstract
Literature data suggest that inflammatory cells such as mast cells (MCs) are involved in angiogenesis. MCs can stimulate angiogenesis by releasing of well identified pro-angiogenic cytokines stored in their cytoplasm. In particular, MCs can release tryptase, a potent in vivo and in vitro pro-angiogenic factor. Nevertheless, few data are available concerning the role of MCs positive to tryptase in primary pancreatic cancer angiogenesis. This study analyzed the correlation between mast cells positive to c-Kit receptor (c-Kit+ MCs), the density of MCs expressing tryptase (MCD-T) and microvascular density (MVD) in primary tumor tissue from patients affected by pancreatic ductal adenocarcinoma (PDAC). A series of 35 PDAC patients with stage T2-3N0-1M0 (by AJCC for Pancreas Cancer Staging 7th Edition) were selected and then undergone to surgery. Tumor tissue samples were evaluated by mean of immunohistochemistry and image analysis methods in terms of number of c-Kit+ MCs, MCD-T and MVD. The above parameters were related each other and with the most important main clinico-pathological features. A significant correlation between c-Kit+ MCs, MCD-T and MVD groups each other was found by Pearson t-test analysis (r ranged from 0.75 to 0.87; p-value ranged from 0.01 to 0.04). No other significant correlation was found. Our in vivo preliminary data, suggest that tumor microenvironmental MCs evaluated in terms of c-Kit+ MCs and MCD-T may play a role in PDAC angiogenesis and they could be further evaluated as a novel tumor biomarker and as a target of anti-angiogenic therapy.
Collapse
Affiliation(s)
- Michele Ammendola
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University of Catanzaro "Magna Graecia" Medical School, Viale Europa-Germaneto, Catanzaro, Italy
| | - Cosmo Damiano Gadaleta
- Interventional Radiology Unit with Integrated Section of Traslational Medical Oncology, National Cancer Research Centre, "Giovanni Paolo II", Bari, Italy
| | - Adam Enver Frampton
- HPB Surgical Unit, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital, London, UK
| | - Tullio Piardi
- Department of General, Digestive and Endocrine Surgery, Hopital Robert Debre, Centre Hospitalier Universitaire de Reims, Universite de Reims Champagne-Ardenne, Reims, France
| | - Riccardo Memeo
- Hepato-Biliary and Pancreatic Surgical Unit, General, Digestive and Endocrine Surgery, IRCAD, IHU Mix-Surg, Institute for Minimally Invasive Image-Guided Surgery, University of Strasbourg, 1 place de l'Hôpital, Strasbourg, France
| | - Valeria Zuccalà
- Pathology Unit, "Pugliese-Ciaccio" Hospital, Catanzaro, Italy
| | - Maria Luposella
- Cardiovascular Disease Unit, "San Giovanni di Dio" Hospital, Crotone, Italy
| | - Rosa Patruno
- Chair of Pathology, Veterinary Medical School, University "Aldo Moro", Bari, Italy
| | - Nicola Zizzo
- Chair of Pathology, Veterinary Medical School, University "Aldo Moro", Bari, Italy
| | - Pietro Gadaleta
- Interventional Radiology Unit with Integrated Section of Traslational Medical Oncology, National Cancer Research Centre, "Giovanni Paolo II", Bari, Italy
| | - Patrick Pessaux
- Hepato-Biliary and Pancreatic Surgical Unit, General, Digestive and Endocrine Surgery, IRCAD, IHU Mix-Surg, Institute for Minimally Invasive Image-Guided Surgery, University of Strasbourg, 1 place de l'Hôpital, Strasbourg, France
| | - Rosario Sacco
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University of Catanzaro "Magna Graecia" Medical School, Viale Europa-Germaneto, Catanzaro, Italy
| | - Giuseppe Sammarco
- Department of Medical and Surgical Sciences, Clinical Surgery Unit, University of Catanzaro "Magna Graecia" Medical School, Viale Europa-Germaneto, Catanzaro, Italy
| | - Girolamo Ranieri
- Interventional Radiology Unit with Integrated Section of Traslational Medical Oncology, National Cancer Research Centre, "Giovanni Paolo II", Bari, Italy
| |
Collapse
|
17
|
Sheng WZ, Chen YS, Tu CT, He J, Zhang B, Gao WD. ANGPTL2 expression in gastric cancer tissues and cells and its biological behavior. World J Gastroenterol 2016; 22:10364-10370. [PMID: 28058016 PMCID: PMC5175248 DOI: 10.3748/wjg.v22.i47.10364] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/25/2016] [Accepted: 09/14/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To explore expression of angiopoietin-like protein 2 (ANGPTL2) and its effect on biological behavior such as proliferation and invasiveness in gastric cancer.
METHODS Western blotting was used to detect expression of ANGPTL2 in 60 human normal gastric tissues, 60 human gastric cancer tissues and gastric cell lines including GES-1, N87, SGC7901, BGC823 and PAMC82. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and Transwell assay were used to detect the proliferation and invasive ability of gastric cancer cells.
RESULTS Compared to normal tissues, ANGPTL2 protein levels were significantly upregulated in gastric tissues, and this level was closely correlated with gastric tumor grade, clinical stage and lymph node metastasis. Compared to GES-1 cells, ANGPTL2 mRNA and protein levels were significantly increased in gastric cancer cells including N87, SGC7901, BGC823 and PAMC82. The expression of ANGPTL2 in highly malignant gastric cancer cell lines BGC823 and PAMC82 was significantly higher than in low malignancy gastric cancer cell lines N87 and SGC7901. MTT and Transwell experiments indicated that the proliferation rate and invasive ability of stable overexpressed gastric cancer cells was faster than in cells transfected with Lv-NC and blank control cells, and the invasive ability of stable overexpressed gastric cancer cells was higher than that of cells transfected with Lv-NC and blank control cells.
CONCLUSION ANGPTL2 contributed to proliferation and invasion of gastric cancer cells. In clinical treatment, ANGPTL2 may become a new target for treatment of gastric cancer.
Collapse
|
18
|
Nowara E, Huszno J. Masitinib plus gemcitabine for personalized treatment of PDAC patients with overexpression of ACOX1. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2016. [DOI: 10.1080/23808993.2016.1257911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Elzbieta Nowara
- Clinical and Experimental Chemotherapy Department, Cancer Center and Institution of Oncology, Gliwice, Poland
| | - Joanna Huszno
- Clinical and Experimental Chemotherapy Department, Cancer Center and Institution of Oncology, Gliwice, Poland
| |
Collapse
|
19
|
Liu Z, Ran X, Liu J, Du Y, Ren J, Qu X. Non-toxic lead sulfide nanodots as efficient contrast agents for visualizing gastrointestinal tract. Biomaterials 2016; 100:17-26. [DOI: 10.1016/j.biomaterials.2016.05.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 04/18/2016] [Accepted: 05/17/2016] [Indexed: 01/15/2023]
|
20
|
Ammendola M, Sacco R, Sammarco G, Luposella M, Patruno R, Gadaleta CD, Sarro GD, Ranieri G. Mast Cell-Targeted Strategies in Cancer Therapy. Transfus Med Hemother 2016; 43:109-13. [PMID: 27330532 DOI: 10.1159/000444942] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/18/2016] [Indexed: 02/06/2023] Open
Abstract
Mast cells (MCs) are cells that originate in the bone marrow from pluripotent CD34+ hematopoietic stem cells. Precursors of MCs migrate through the circulation to their target tissues, completing their maturation process into granulated cells under the influence of several microenvironment growth factors. The most important of these factors is the ligand for the c-Kit receptor (c-Kit-R) namely stem cell factor (SCF), secreted mainly by fibroblasts and endothelial cells (ECs). SCF also regulates development, survival and de novo proliferation of MCs. It has already been demonstrated that gain-of-function mutations of gene c-Kit encoding c-Kit-R result in the development of some tumors. Furthermore, MCs are able also to modulate both innate and adaptive immune response and to express the high-affinity IgE receptor following IgE activation. Among the other IgE-independent MC activation mechanisms, a wide variety of other surface receptors for cytokines, chemokines, immunoglobulins, and complement are also described. Interestingly, MCs can stimulate angiogenesis by releasing of several pro-angiogenic cytokines stored in their cytoplasm. Studies published in the last year suggest that angiogenesis stimulated by MCs may play an important role in tumor growth and progression. Here, we aim to focus several biological features of MCs and to summarize new anti-cancer MC-targeted strategies with potential translation in human clinical trials.
Collapse
Affiliation(s)
- Michele Ammendola
- Department of Medical and Surgical Sciences, General Surgery Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro, Italy
| | - Rosario Sacco
- Department of Medical and Surgical Sciences, General Surgery Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro, Italy
| | - Giuseppe Sammarco
- Department of Medical and Surgical Sciences, General Surgery Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro, Italy
| | - Maria Luposella
- Cardiovascular Disease Unit, 'San Giovanni di Dio' Hospital, Crotone, Italy
| | - Rosa Patruno
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, 'Giovanni Paolo II', Bari, Italy
| | - Cosmo Damiano Gadaleta
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, 'Giovanni Paolo II', Bari, Italy
| | - Giovambattista De Sarro
- Department of Health Science, Clinical Pharmacology and Pharmacovigilance Unit and Pharmacovigilance's Centre Calabria Region, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro, Italy
| | - Girolamo Ranieri
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, 'Giovanni Paolo II', Bari, Italy
| |
Collapse
|