1
|
Najjar F, Alsabe H, Sabbagh H, Al-Massarani G, Aljapawe A, Alamalla N, Banat I, Ikhtiar A. Endothelial progenitor cells as an angiogenic biomarker for the diagnosis and prognosis of lung cancer. Rep Pract Oncol Radiother 2024; 29:544-557. [PMID: 39759554 PMCID: PMC11698562 DOI: 10.5603/rpor.102618] [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: 05/04/2023] [Accepted: 09/04/2024] [Indexed: 01/07/2025] Open
Abstract
Background Angiogenesis is mediated by endothelial progenitor cells (EPCs) derived from bone-marrow. In this prospective study, we tried to investigate the clinical utility of circulating EPCs in lung cancer (LC) patients. Materials and methods Flow cytometry technique was used to assess circulating EPCs according to the immuno-phenotype CD45- CD34+ CD133+ CD146+ mononuclear cells. Sixty patients and 30 controls were included in this prospective study. Results The mean of baseline EPC numbers was significantly higher in LC patients than in controls (p =0.003). Pretreatment EPC values were significantly correlated with primary tumor size (p = 0.05) and tumor response (p = 0.04). Receiver operating characteristics (ROC) curves were plotted to discriminate EPC numbers between patients and controls. Using ROC analysis, the optimal cutoff value was 125 cells/mL with a sensitivity and a specificity for baseline EPCs of 76.7% and 63.3%, respectively. According to this cutoff value, basal EPC values were significantly correlated with primary tumor size (p = 0.047) and response to chemotherapy (p = 0.034). High EPC levels were significantly associated with longer progression-free survival (PFS) and overall survival (OS) duration (p = 0.0043 and p = 0.02, respectively). Conclusion Increased baseline EPC values seem to be a useful biomarker for the prediction of prognosis and tumor response in LC patients. Furthermore, high EPC levels at diagnosis might be an indicator of tumor growth and longer survival in LC patients.
Collapse
Affiliation(s)
- Fadi Najjar
- Biomarkers Laboratory, Department of Radiation Medicine, Atomic Energy Commission of Syria (AECS), Damascus, Syria
| | - Hassan Alsabe
- Division of Thoracic Oncology, Department of Oncology, Al-Bairouni University Hospital, Damascus, Syria
| | - Hussein Sabbagh
- Division of Thoracic Oncology, Department of Oncology, Al-Bairouni University Hospital, Damascus, Syria
| | - Ghassan Al-Massarani
- Biomarkers Laboratory, Department of Radiation Medicine, Atomic Energy Commission of Syria (AECS), Damascus, Syria
| | - Abdulmunim Aljapawe
- Radiobiology Laboratory, Department of Molecular Biology and Biotechnology, Atomic Energy Commission (AEC), Damascus, Syria
| | - Nissreen Alamalla
- Biomarkers Laboratory, Department of Radiation Medicine, Atomic Energy Commission of Syria (AECS), Damascus, Syria
| | - Issraa Banat
- Biomarkers Laboratory, Department of Radiation Medicine, Atomic Energy Commission of Syria (AECS), Damascus, Syria
| | - Adnan Ikhtiar
- Radiobiology Laboratory, Department of Molecular Biology and Biotechnology, Atomic Energy Commission (AEC), Damascus, Syria
| |
Collapse
|
2
|
Falleni M, Dal Lago M, Tosi D, Ghilardi G, De Pasquale L, Saibene AM, Felisati G, Cozzolino M, Gianelli U. Vascular mimicry and mosaic vessels in parathyroid tumours: a new diagnostic approach? J Clin Pathol 2024:jcp-2024-209703. [PMID: 39288990 DOI: 10.1136/jcp-2024-209703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
AIMS Evaluation of 'alternative' vascularisation in human cancer is considered an important prognostic parameter; the 2022 WHO classification of parathyroid tumours despite progresses in clinical triaging of patients strongly emphasises new histopathological parameters to properly stratify these lesions. 'Alternative' and 'classic' vessels were here investigated for the first time in parathyroid tumours for their possible histopathological and clinical relevance during progression. METHODS Using a double CD31/PAS staining, microvessel density (MVD, 'classic' CD31+ vessels), mosaic vessel density (MoVD, 'alternative' CD31+/-vessels) and vessel mimicry density (VMD, 'alternative' CD31-/PAS+ vessels) were evaluated in 4 normal parathyroid glands (N), 50 Adenomas (A), 35 Atypical Tumours (AT) and 10 Carcinomas (K). RESULTS Compared with N, MVD significantly increased in A (p=0.012) and decreased in K (p=0.013) with vessel counts lower than in AT and A (p<0.001). MoVs and VMs, absent in normal tissue, were documented in non-benign parathyroid lesions (AT, K) (p<0.001), with MoVs and VMs most represented in AT and K, respectively (p<0.001), in peripheral growing areas. Vessel distribution was correlated to neoplastic progression (r=-0.541 MVD; r=+0.760 MoVD, r=+0.733 VMD), with MVD decrease in AT and K inversely related to MoVD and VMD increase (r=-0.503 and r=-0.456). CONCLUSIONS 'Alternative' vessel identification in parathyroid tumours is crucial because it: (1) explains the paradox of non-angiogenic tumours, consisting in a new bloody non-endothelial vessel network and (2) helps pathologists to unmask worrisome lesions. Furthermore, detection of alternative vascular systems in human tumours might explain the limited success of antiangiogenic therapies and encourage new oncological studies.
Collapse
Affiliation(s)
- Monica Falleni
- Unit of Pathology, Department of Health Sciences, University of Milan, ASST Santi Paolo e Carlo, Milano, Italy
| | - Matteo Dal Lago
- Unit of Pathology, Department of Health Sciences, University of Milan, ASST Santi Paolo e Carlo, Milano, Italy
| | - Delfina Tosi
- Unit of Pathology, Department of Health Sciences, University of Milan, ASST Santi Paolo e Carlo, Milano, Italy
| | - Giorgio Ghilardi
- Surgical Unit, Department of Health Sciences, University of Milan, ASST Santi Paolo e Carlo, Milano, Italy
| | | | - Alberto M Saibene
- Otolaryngology Unit, Department of Health Sciences, University of Milan, ASST Santi Paolo e Carlo, Milano, Italy
| | - Giovanni Felisati
- Otolaryngology Unit, Department of Health Sciences, University of Milan, ASST Santi Paolo e Carlo, Milano, Italy
| | - Mario Cozzolino
- Renal Division, Department of Health Sciences, University of Milan, ASST Santi Paolo e Carlo, Milano, Italy
| | - Umberto Gianelli
- Unit of Pathology, Department of Health Sciences, University of Milan, ASST Santi Paolo e Carlo, Milano, Italy
| |
Collapse
|
3
|
Guo Z, Jing X, Sun X, Sun S, Yang Y, Cao Y. Tumor angiogenesis and anti-angiogenic therapy. Chin Med J (Engl) 2024; 137:2043-2051. [PMID: 39051171 PMCID: PMC11374217 DOI: 10.1097/cm9.0000000000003231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Indexed: 07/27/2024] Open
Abstract
ABSTRACT Anti-angiogenic drugs (AADs), which mainly target the vascular endothelial growth factor-A signaling pathway, have become a therapeutic option for cancer patients for two decades. During this period, tremendous clinical experience of anti-angiogenic therapy has been acquired, new AADs have been developed, and the clinical indications for AAD treatment of various cancers have been expanded using monotherapy and combination therapy. However, improvements in the therapeutic outcomes of clinically available AADs and the development of more effective next-generation AADs are still urgently required. This review aims to provide historical and perspective views on tumor angiogenesis to allow readers to gain mechanistic insights and learn new therapeutic development. We revisit the history of concept initiation and AAD discovery, and summarize the up-to-date clinical translation of anti-angiogenic cancer therapy in this field.
Collapse
Affiliation(s)
- Ziheng Guo
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xu Jing
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17177, Sweden
| | - Xiaoting Sun
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17177, Sweden
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vison and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shishuo Sun
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17177, Sweden
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17177, Sweden
| |
Collapse
|
4
|
van Luyk ME, Krotenberg Garcia A, Lamprou M, Suijkerbuijk SJE. Cell competition in primary and metastatic colorectal cancer. Oncogenesis 2024; 13:28. [PMID: 39060237 PMCID: PMC11282291 DOI: 10.1038/s41389-024-00530-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Adult tissues set the scene for a continuous battle between cells, where a comparison of cellular fitness results in the elimination of weaker "loser" cells. This phenomenon, named cell competition, is beneficial for tissue integrity and homeostasis. In fact, cell competition plays a crucial role in tumor suppression, through elimination of early malignant cells, as part of Epithelial Defense Against Cancer. However, it is increasingly apparent that cell competition doubles as a tumor-promoting mechanism. The comparative nature of cell competition means that mutational background, proliferation rate and polarity all factor in to determine the outcome of these processes. In this review, we explore the intricate and context-dependent involvement of cell competition in homeostasis and regeneration, as well as during initiation and progression of primary and metastasized colorectal cancer. We provide a comprehensive overview of molecular and cellular mechanisms governing cell competition and its parallels with regeneration.
Collapse
Affiliation(s)
- Merel Elise van Luyk
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Ana Krotenberg Garcia
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Maria Lamprou
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Saskia Jacoba Elisabeth Suijkerbuijk
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
5
|
Tátrai E, Ranđelović I, Surguta SE, Tóvári J. Role of Hypoxia and Rac1 Inhibition in the Metastatic Cascade. Cancers (Basel) 2024; 16:1872. [PMID: 38791951 PMCID: PMC11120288 DOI: 10.3390/cancers16101872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/03/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
The hypoxic condition has a pivotal role in solid tumors and was shown to correlate with the poor outcome of anticancer treatments. Hypoxia contributes to tumor progression and leads to therapy resistance. Two forms of a hypoxic environment might have relevance in tumor mass formation: chronic and cyclic hypoxia. The main regulators of hypoxia are hypoxia-inducible factors, which regulate the cell survival, proliferation, motility, metabolism, pH, extracellular matrix function, inflammatory cells recruitment and angiogenesis. The metastatic process consists of different steps in which hypoxia-inducible factors can play an important role. Rac1, belonging to small G-proteins, is involved in the metastasis process as one of the key molecules of migration, especially in a hypoxic environment. The effect of hypoxia on the tumor phenotype and the signaling pathways which may interfere with tumor progression are already quite well known. Although the role of Rac1, one of the small G-proteins, in hypoxia remains unclear, predominantly, in vitro studies performed so far confirm that Rac1 inhibition may represent a viable direction for tumor therapy.
Collapse
Affiliation(s)
- Enikő Tátrai
- The National Tumor Biology Laboratory, Department of Experimental Pharmacology, National Institute of Oncology, H-1122 Budapest, Hungary; (I.R.); (S.E.S.); (J.T.)
| | - Ivan Ranđelović
- The National Tumor Biology Laboratory, Department of Experimental Pharmacology, National Institute of Oncology, H-1122 Budapest, Hungary; (I.R.); (S.E.S.); (J.T.)
| | - Sára Eszter Surguta
- The National Tumor Biology Laboratory, Department of Experimental Pharmacology, National Institute of Oncology, H-1122 Budapest, Hungary; (I.R.); (S.E.S.); (J.T.)
- School of Ph. D. Studies, Semmelweis University, H-1085 Budapest, Hungary
| | - József Tóvári
- The National Tumor Biology Laboratory, Department of Experimental Pharmacology, National Institute of Oncology, H-1122 Budapest, Hungary; (I.R.); (S.E.S.); (J.T.)
- School of Ph. D. Studies, Semmelweis University, H-1085 Budapest, Hungary
| |
Collapse
|
6
|
Shao L, Yang X, Sun Z, Tan X, Lu Z, Hu S, Dou W, Duan S. Three-dimensional pseudo-continuous arterial spin-labelled perfusion imaging for diagnosing upper cervical lymph node metastasis in patients with nasopharyngeal carcinoma: a whole-node histogram analysis. Clin Radiol 2024; 79:e736-e743. [PMID: 38341343 DOI: 10.1016/j.crad.2024.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 02/12/2024]
Abstract
AIM To evaluate whole-node histogram parameters of blood flow (BF) maps derived from three-dimensional pseudo-continuous arterial spin-labelled (3D pCASL) imaging in discriminating metastatic from benign upper cervical lymph nodes (UCLNs) for nasopharyngeal carcinoma (NPC) patients. MATERIALS AND METHODS Eighty NPC patients with a total of 170 histologically confirmed UCLNs (67 benign and 103 metastatic) were included retrospectively. Pre-treatment 3D pCASL imaging was performed and whole-node histogram analysis was then applied. Histogram parameters and morphological features, such as minimum axis diameter (MinAD), maximum axis diameter (MaxAD), and location of UCLNs, were assessed and compared between benign and metastatic lesions. Predictors were identified and further applied to establish a combined model by multivariate logistic regression in predicting the probability of metastatic UCLNs. Receiver operating characteristic (ROC) curves were used to analyse the diagnostic performance. RESULTS Metastatic UCLNs had larger MinAD and MinAD/MaxAD ratio, greater energy and entropy values, and higher incidence of level II (upper jugular group), but lower BF10th value than benign nodes (all p<0.05). MinAD, BF10th, energy, and entropy were validated as independent predictors in diagnosing metastatic UCLNs. The combined model yielded an area under the curve (AUC) of 0.932, accuracy of 84.42 %, sensitivity of 80.6 %, and specificity of 90.29 %. CONCLUSIONS Whole-node histogram analysis on BF maps is a feasible tool to differentiate metastatic from benign UCLNs in NPC patients, and the combined model can further improve the diagnostic efficacy.
Collapse
Affiliation(s)
- L Shao
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi City, Jiangsu, China
| | - X Yang
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi City, Jiangsu, China
| | - Z Sun
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi City, Jiangsu, China.
| | - X Tan
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi City, Jiangsu, China
| | - Z Lu
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi City, Jiangsu, China
| | - S Hu
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi City, Jiangsu, China
| | - W Dou
- General Electric (GE) Healthcare, MR Research China, Beijing, China
| | - S Duan
- General Electric (GE) Healthcare China, Shanghai, China
| |
Collapse
|
7
|
Baghy K, Ladányi A, Reszegi A, Kovalszky I. Insights into the Tumor Microenvironment-Components, Functions and Therapeutics. Int J Mol Sci 2023; 24:17536. [PMID: 38139365 PMCID: PMC10743805 DOI: 10.3390/ijms242417536] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/25/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Similarly to our healthy organs, the tumor tissue also constitutes an ecosystem. This implies that stromal cells acquire an altered phenotype in tandem with tumor cells, thereby promoting tumor survival. Cancer cells are fueled by abnormal blood vessels, allowing them to develop and proliferate. Tumor-associated fibroblasts adapt their cytokine and chemokine production to the needs of tumor cells and alter the peritumoral stroma by generating more collagen, thereby stiffening the matrix; these processes promote epithelial-mesenchymal transition and tumor cell invasion. Chronic inflammation and the mobilization of pro-tumorigenic inflammatory cells further facilitate tumor expansion. All of these events can impede the effective administration of tumor treatment; so, the successful inhibition of tumorous matrix remodeling could further enhance the success of antitumor therapy. Over the last decade, significant progress has been made with the introduction of novel immunotherapy that targets the inhibitory mechanisms of T cell activation. However, extensive research is also being conducted on the stromal components and other cell types of the tumor microenvironment (TME) that may serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Kornélia Baghy
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary;
| | - Andrea Ladányi
- Department of Surgical and Molecular Pathology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary;
| | - Andrea Reszegi
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1091 Budapest, Hungary
| | - Ilona Kovalszky
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary;
| |
Collapse
|
8
|
Tisza A, Klikovits T, Benej M, Torok S, Szeitz B, Valko Z, Hoda MA, Hegedus B, Bonta M, Nischkauer W, Hoetzenecker K, Limbeck A, Schelch K, Laszlo V, Megyesfalvi Z, Dome B. Laser ablation-inductively coupled plasma-mass spectrometry analysis reveals differences in chemotherapeutic drug distribution in surgically resected pleural mesothelioma. Br J Clin Pharmacol 2023; 89:3364-3374. [PMID: 37272312 PMCID: PMC10952999 DOI: 10.1111/bcp.15813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/04/2023] [Accepted: 05/22/2023] [Indexed: 06/06/2023] Open
Abstract
AIMS Pleural mesothelioma (PM) is a highly aggressive thoracic tumour with poor prognosis. Although reduced tissue drug accumulation is one of the key features of platinum (Pt) resistance, little is known about Pt distribution in human PM. METHODS We assessed Pt levels of blood samples and surgically resected specimens from 25 PM patients who had received neoadjuvant Pt-based chemotherapy (CHT). Pt levels and tissue distributions were measured by laser ablation-inductively coupled plasma-mass spectrometry and correlated with clinicopathological features. RESULTS In surgically resected PM specimens, mean Pt levels of nontumourous (fibrotic) areas were significantly higher (vs tumourous regions, P = 0.0031). No major heterogeneity of Pt distribution was seen within the tumourous areas. Pt levels correlated neither with the microvessel area nor with apoptosis rate in the tumourous or nontumourous regions. A significant positive correlation was found between serum and both full tissue section and tumourous area mean Pt levels (r = 0.532, P = 0.006, 95% confidence interval [95% CI] 0.161-0.771 and r = 0.415, P = 0.039, 95% CI 0.011-0.702, respectively). Furthermore, a significant negative correlation was detected between serum Pt concentrations and elapsed time from the last cycle of CHT (r = -0.474, P = 0.017, 95% CI -0.738--0.084). Serum Pt levels correlated negatively with overall survival (OS) (P = 0.029). CONCLUSIONS There are major differences in drug distribution between tumourous and nontumourous areas of PM specimens. Serum Pt levels significantly correlate with full section and tumourous area average Pt levels, elapsed time from the last CHT cycle, and OS. Further studies investigating clinicopathological factors that modulate tissue Pt concentration and distribution are warranted.
Collapse
Affiliation(s)
- Anna Tisza
- Department of Tumor BiologyNational Korányi Institute of PulmonologyBudapestHungary
- Department of Pathology and Experimental Cancer ResearchSemmelweis UniversityBudapestHungary
| | - Thomas Klikovits
- Department of Thoracic Surgery, Comprehensive Cancer CenterMedical University of ViennaViennaAustria
- Karl‐Landsteiner‐Institute for Clinical and Translational Thoracic Surgery Research, Clinic FloridsdorfViennaAustria
| | - Michal Benej
- Karl‐Landsteiner‐Institute for Clinical and Translational Thoracic Surgery Research, Clinic FloridsdorfViennaAustria
| | - Szilvia Torok
- Department of Tumor BiologyNational Korányi Institute of PulmonologyBudapestHungary
| | - Beata Szeitz
- Division of Oncology, Department of Internal Medicine and OncologySemmelweis UniversityBudapestHungary
| | - Zsuzsanna Valko
- Department of Thoracic Surgery, Comprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Comprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - Balazs Hegedus
- Department of Thoracic Surgery, University Medicine Essen – RuhrlandklinikUniversity Duisburg‐EssenEssenGermany
- Department of Pathology, Forensic and Insurance MedicineSemmelweis UniversityBudapestHungary
| | - Maximilian Bonta
- Institute of Chemical Technologies and Analytics, Division of Instrumental Analytical ChemistryTU WienViennaAustria
| | - Winfried Nischkauer
- Institute of Chemical Technologies and Analytics, Division of Instrumental Analytical ChemistryTU WienViennaAustria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Comprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - Andreas Limbeck
- Institute of Chemical Technologies and Analytics, Division of Instrumental Analytical ChemistryTU WienViennaAustria
| | - Karin Schelch
- Department of Thoracic Surgery, Comprehensive Cancer CenterMedical University of ViennaViennaAustria
- Center for Cancer ResearchMedical University of ViennaViennaAustria
| | - Viktoria Laszlo
- Department of Tumor BiologyNational Korányi Institute of PulmonologyBudapestHungary
- Department of Thoracic Surgery, Comprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - Zsolt Megyesfalvi
- Department of Tumor BiologyNational Korányi Institute of PulmonologyBudapestHungary
- Department of Thoracic Surgery, Comprehensive Cancer CenterMedical University of ViennaViennaAustria
- Department of Thoracic SurgeryNational Institute of Oncology‐Semmelweis UniversityBudapestHungary
| | - Balazs Dome
- Department of Tumor BiologyNational Korányi Institute of PulmonologyBudapestHungary
- Department of Thoracic Surgery, Comprehensive Cancer CenterMedical University of ViennaViennaAustria
- Department of Thoracic SurgeryNational Institute of Oncology‐Semmelweis UniversityBudapestHungary
- Department of Translational MedicineLund UniversityLundSweden
| |
Collapse
|
9
|
Grizzi F, Hegazi MAAA, Zanoni M, Vota P, Toia G, Clementi MC, Mazzieri C, Chiriva-Internati M, Taverna G. Prostate Cancer Microvascular Routes: Exploration and Measurement Strategies. Life (Basel) 2023; 13:2034. [PMID: 37895416 PMCID: PMC10608780 DOI: 10.3390/life13102034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Angiogenesis is acknowledged as a pivotal feature in the pathology of human cancer. Despite the absence of universally accepted markers for gauging the comprehensive angiogenic activity in prostate cancer (PCa) that could steer the formulation of focused anti-angiogenic treatments, the scrutiny of diverse facets of tumoral blood vessel development may furnish significant understanding of angiogenic processes. Malignant neoplasms, encompassing PCa, deploy a myriad of strategies to secure an adequate blood supply. These modalities range from sprouting angiogenesis and vasculogenesis to intussusceptive angiogenesis, vascular co-option, the formation of mosaic vessels, vasculogenic mimicry, the conversion of cancer stem-like cells into tumor endothelial cells, and vascular pruning. Here we provide a thorough review of these angiogenic mechanisms as they relate to PCa, discuss their prospective relevance for predictive and prognostic evaluations, and outline the prevailing obstacles in quantitatively evaluating neovascularization via histopathological examinations.
Collapse
Affiliation(s)
- Fabio Grizzi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
| | - Mohamed A. A. A. Hegazi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
| | - Matteo Zanoni
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Paolo Vota
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Giovanni Toia
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Maria Chiara Clementi
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Cinzia Mazzieri
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Maurizio Chiriva-Internati
- Departments of Gastroenterology, Hepatology & Nutrition, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Gianluigi Taverna
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| |
Collapse
|
10
|
Kamal MV, Damerla RR, Dikhit PS, Kumar NAN. Prostaglandin-endoperoxide synthase 2 (PTGS2) gene expression and its association with genes regulating the VEGF signaling pathway in head and neck squamous cell carcinoma. J Oral Biol Craniofac Res 2023; 13:567-574. [PMID: 37559688 PMCID: PMC10407435 DOI: 10.1016/j.jobcr.2023.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 08/11/2023] Open
Abstract
Introduction The PTGS2 gene codes for the cyclooxygenase-2 (COX-2) enzyme that catalyzes the committed step in prostaglandin (PG) synthesis. Various in-vivo and in-vitro data suggest that prostaglandin E2 mediates as a signaling molecule for activating the VEGF signaling pathway (VSP), forming an association between COX-2 and VSP. Several chemotherapy regimens increasingly rely on preventing the synthesis of PGs. The targeted and metronomic chemotherapy agents, which suppress the COX-2 enzymes, have a major role in suppressing the oral cancer cascade. Hence, this study was designed to understand the pattern of PTGS2 expression and genes regulating VSP in head and neck cancers. Methods PTGS2 expression was analyzed in the TCGA database computationally with the help of the UALCAN web-server. The expression of VEGF signaling pathway genes was mined, and their expression pattern was determined. Co-expression analysis was done to elucidate the association between VEGF signaling genes and PTGS2. The ShineyGo web server was used for gene set enrichment. Results Significantly high PTGS2 expression was observed in tumor samples. Further genes regulating VEGF signaling were significantly overexpressed in tumor samples. Co-expression analysis results showed a significant positive correlation between PTGS2 and angiogenesis-regulating genes. The majority of the genes were enriched for angiogenesis pathways. Conclusion PTGS2 was significantly expressed in head and neck cancer, and its expression was associated with genes regulating angiogenesis.
Collapse
Affiliation(s)
- Mehta Vedant Kamal
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Rama Rao Damerla
- Department of Medical Genetics, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Punit Singh Dikhit
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Naveena AN Kumar
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
11
|
Tsilimigras DI, Ntanasis-Stathopoulos I, Pawlik TM. Molecular Mechanisms of Colorectal Liver Metastases. Cells 2023; 12:1657. [PMID: 37371127 DOI: 10.3390/cells12121657] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
The liver is the most frequently target for metastasis among patients with colorectal cancer mainly because of the portal vein circulation that directly connects the colon and rectum with the liver. The liver tumor microenvironment consists of different cell types each with unique characteristics and functions that modulate the antigen recognition and immune system activation. Primary tumors from other sites "prime" the liver prior to the seeding of cancer cells, creating a pre-metastatic niche. Following invasion into the liver, four different phases are key to the development of liver metastases: a microvascular phase in which cancer cells infiltrate and become trapped in sinusoidal vessels; an extravascular, pre-angiogenic phase; an angiogenic phase that supplies oxygen and nutrients to cancer cells; and a growth phase in which metastatic cells multiply and enlarge to form detectable tumors. Exosomes carry proteins, lipids, as well as genetic information that can create a pre-metastatic niche in distant sites, including the liver. The complexity of angiogenic mechanisms and the exploitation of the vasculature in situ by cancer cells have limited the efficacy of currently available anti-angiogenic therapies. Delineating the molecular mechanisms implicated in colorectal liver metastases is crucial to understand and predict tumor progression; the development of distant metastases; and resistance to chemotherapy, immunotherapy, and targeted treatment.
Collapse
Affiliation(s)
- Diamantis I Tsilimigras
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, 395 W. 12th Ave., Columbus, OH 43210, USA
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, 395 W. 12th Ave., Columbus, OH 43210, USA
| |
Collapse
|
12
|
Delgado-Bellido D, Oliver FJ, Vargas Padilla MV, Lobo-Selma L, Chacón-Barrado A, Díaz-Martin J, de Álava E. VE-Cadherin in Cancer-Associated Angiogenesis: A Deceptive Strategy of Blood Vessel Formation. Int J Mol Sci 2023; 24:ijms24119343. [PMID: 37298296 DOI: 10.3390/ijms24119343] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Tumor growth depends on the vascular system, either through the expansion of blood vessels or novel adaptation by tumor cells. One of these novel pathways is vasculogenic mimicry (VM), which is defined as a tumor-provided vascular system apart from endothelial cell-lined vessels, and its origin is partly unknown. It involves highly aggressive tumor cells expressing endothelial cell markers that line the tumor irrigation. VM has been correlated with high tumor grade, cancer cell invasion, cancer cell metastasis, and reduced survival of cancer patients. In this review, we summarize the most relevant studies in the field of angiogenesis and cover the various aspects and functionality of aberrant angiogenesis by tumor cells. We also discuss the intracellular signaling mechanisms involved in the abnormal presence of VE-cadherin (CDH5) and its role in VM formation. Finally, we present the implications for the paradigm of tumor angiogenesis and how targeted therapy and individualized studies can be applied in scientific analysis and clinical settings.
Collapse
Affiliation(s)
- Daniel Delgado-Bellido
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain
- Instituto de Salud Carlos III, CIBERONC, 28220 Madrid, Spain
- Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, 41013 Seville, Spain
| | - F J Oliver
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain
| | | | - Laura Lobo-Selma
- Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, 41013 Seville, Spain
| | | | - Juan Díaz-Martin
- Instituto de Salud Carlos III, CIBERONC, 28220 Madrid, Spain
- Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, 41013 Seville, Spain
| | - Enrique de Álava
- Instituto de Salud Carlos III, CIBERONC, 28220 Madrid, Spain
- Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, 41013 Seville, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain
| |
Collapse
|
13
|
Fitzgerald KN, Motzer RJ, Lee CH. Adjuvant therapy options in renal cell carcinoma - targeting the metastatic cascade. Nat Rev Urol 2023; 20:179-193. [PMID: 36369389 PMCID: PMC10921989 DOI: 10.1038/s41585-022-00666-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2022] [Indexed: 11/13/2022]
Abstract
Localized renal cell carcinoma (RCC) is primarily managed with nephrectomy, which is performed with curative intent. However, disease recurs in ~20% of patients. Treatment with adjuvant therapies is used after surgery with the intention of curing additional patients by disrupting the establishment, maturation or survival of micrometastases, processes collectively referred to as the metastatic cascade. Immune checkpoint inhibitors and vascular endothelial growth factor receptor (VEGFR)-targeting tyrosine kinase inhibitors (TKIs) have shown efficacy in the treatment of metastatic RCC, increasing the interest in the utility of these agents in the adjuvant setting. Pembrolizumab, an inhibitor of the immune checkpoint PD1, is now approved by the FDA and is under review by European regulatory agencies for the adjuvant treatment of patients with localized resected clear cell RCC based on the results of the KEYNOTE-564 trial. However, the optimal use of immunotherapy and VEGFR-targeting TKIs for adjuvant treatment of RCC is not completely understood. These agents disrupt the metastatic cascade at multiple steps, providing biological rationale for further investigating the applications of these therapeutics in the adjuvant setting. Clinical trials to evaluate adjuvant therapeutics in RCC are ongoing, and clinical considerations must guide the practical use of immunotherapy and TKI agents for the adjuvant treatment of localized resected RCC.
Collapse
Affiliation(s)
- Kelly N Fitzgerald
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chung-Han Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
14
|
Tímár J, Honn KV, Hendrix MJC, Marko-Varga G, Jalkanen S. Newly identified form of phenotypic plasticity of cancer: immunogenic mimicry. Cancer Metastasis Rev 2023; 42:323-334. [PMID: 36754910 PMCID: PMC10014767 DOI: 10.1007/s10555-023-10087-1] [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: 10/13/2022] [Accepted: 01/18/2023] [Indexed: 02/10/2023]
Abstract
Cancer plasticity is now a recognized new hallmark of cancer which is due to disturbances of cell differentiation programs. It is manifested not only in various forms like the best-known epithelial-mesenchymal transition (EMT) but also in vasculogenic and megakaryocytic mimicries regulated by EMT-specific or less-specific transcription factors such as HIF1a or STAT1/2. Studies in the past decades provided ample data that cancer plasticity can be manifested also in the expression of a vast array of immune cell genes; best-known examples are PDL1/CD274, CD47, or IDO, and we termed it immunogenic mimicry (IGM). However, unlike other types of plasticities which are epigenetically regulated, expression of IGM genes are frequently due to gene amplifications. It is important that the majority of the IGM genes are regulated by interferons (IFNs) suggesting that their protein expressions are regulated by the immune microenvironment. Most of the IGM genes have been shown to be involved in immune escape of cancers broadening the repertoire of these mechanisms and offering novel targets for immunotherapeutics.
Collapse
Affiliation(s)
- József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary.
| | - Kenneth V Honn
- Departments of Pathology, Oncology and Chemistry, Wayne State University, Detroit, MI, USA.,Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - Mary J C Hendrix
- Department of Biology, Shepherd University, Shepherdstown, WV, USA
| | - György Marko-Varga
- Clinical Protein Science and Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Sirpa Jalkanen
- Medicity Research Laboratories, Turku, Finland.,InFLAMES Flagship, University of Turku, Turku, Finland
| |
Collapse
|
15
|
Pathak S, Gupta R, Parkar H, Joshi N, Nagotu S, Kale A. The role of Colchicine on actin polymerization dynamics: as a potent anti-angiogenic factor. J Biomol Struct Dyn 2022; 40:11729-11743. [PMID: 34424806 DOI: 10.1080/07391102.2021.1965911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Over the years, cancer research has focused on different strategies to discover drugs and therapies to treat the metastatic stage of cancer. This stage depends upon the type, and the cause of cancer. One of the central facts about any cancer invasion is the formation of new blood vessels that provide nutrients to these uncontrollably dividing cells. This phenomenon is called angiogenesis and is responsible for tumor progression and metastasis. Tumor angiogenesis is a sequential process wherein various angiogenic factors produced by tumor cells bind to receptors of endothelial cells. This stimulates the cytoskeletal protein, especially actin to reorganize themselves and undergo the process of canalization. The driving force for such membrane transformation is spatially and temporally-regulated by polymerization of submembrane actin filaments. So far, Colchicine has been studied for its effectiveness in controlling microtubule reorganization during cell division, but its role is far from understood on actin polymerization. In our current study, we report the effect of Colchicine on actin polymerization dynamics using biophysical analysis like Right light scattering (RLS), Dynamic light scattering (DLS), Circular dichroism (CD) analysis, Scanning electron microscopy (SEM) study. Isothermal titration calorimetry (ITC) and kinetic measurements. Isothermal titration calorimetry (ITC) indicates multiple site binding for colchicine with actin aggregates. We have checked the in vivo effect of colchicine using end3 cells of Saccharomyces cerevisiae. We also report the anti-angiogenesis activity of colchicine via ex-ovo chicken chorioallantoic membrane (CAM) assay. We predict the target site of binding for the drug by docking studies. Based on our findings, we suggest the 'drug-repurposed' function for colchicine as a potential anti-angiogenic candidate.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Samridhi Pathak
- School of Chemical Sciences, UM-DAE Center for excellence in basic sciences, University of Mumbai, Mumbai, Maharashtra, India
| | - Rahul Gupta
- School of Chemical Sciences, UM-DAE Center for excellence in basic sciences, University of Mumbai, Mumbai, Maharashtra, India
| | - Haifa Parkar
- School of Chemical Sciences, UM-DAE Center for excellence in basic sciences, University of Mumbai, Mumbai, Maharashtra, India
| | - Neha Joshi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Shirisha Nagotu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Avinash Kale
- School of Chemical Sciences, UM-DAE Center for excellence in basic sciences, University of Mumbai, Mumbai, Maharashtra, India
| |
Collapse
|
16
|
Cuypers A, Truong ACK, Becker LM, Saavedra-García P, Carmeliet P. Tumor vessel co-option: The past & the future. Front Oncol 2022; 12:965277. [PMID: 36119528 PMCID: PMC9472251 DOI: 10.3389/fonc.2022.965277] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/04/2022] [Indexed: 12/11/2022] Open
Abstract
Tumor vessel co-option (VCO) is a non-angiogenic vascularization mechanism that is a possible cause of resistance to anti-angiogenic therapy (AAT). Multiple tumors are hypothesized to primarily rely on growth factor signaling-induced sprouting angiogenesis, which is often inhibited during AAT. During VCO however, tumors invade healthy tissues by hijacking pre-existing blood vessels of the host organ to secure their blood and nutrient supply. Although VCO has been described in the context of AAT resistance, the molecular mechanisms underlying this process and the profile and characteristics of co-opted vascular cell types (endothelial cells (ECs) and pericytes) remain poorly understood, resulting in the lack of therapeutic strategies to inhibit VCO (and to overcome AAT resistance). In the past few years, novel next-generation technologies (such as single-cell RNA sequencing) have emerged and revolutionized the way of analyzing and understanding cancer biology. While most studies utilizing single-cell RNA sequencing with focus on cancer vascularization have centered around ECs during sprouting angiogenesis, we propose that this and other novel technologies can be used in future investigations to shed light on tumor EC biology during VCO. In this review, we summarize the molecular mechanisms driving VCO known to date and introduce the models used to study this phenomenon to date. We highlight VCO studies that recently emerged using sequencing approaches and propose how these and other novel state-of-the-art methods can be used in the future to further explore ECs and other cell types in the VCO process and to identify potential vulnerabilities in tumors relying on VCO. A better understanding of VCO by using novel approaches could provide new answers to the many open questions, and thus pave the way to develop new strategies to control and target tumor vascularization.
Collapse
Affiliation(s)
- Anne Cuypers
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), Vlaams Instituut voor Biotechnologie (VIB) and Department of Oncology, Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Anh-Co Khanh Truong
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), Vlaams Instituut voor Biotechnologie (VIB) and Department of Oncology, Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Lisa M. Becker
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), Vlaams Instituut voor Biotechnologie (VIB) and Department of Oncology, Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Paula Saavedra-García
- Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), Vlaams Instituut voor Biotechnologie (VIB) and Department of Oncology, Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
17
|
Novruzov E, Mori Y, Antke C, Dabir M, Schmitt D, Kratochwil C, Koerber SA, Haberkorn U, Giesel FL. A Role of Non-FDG Tracers in Lung Cancer? Semin Nucl Med 2022; 52:720-733. [PMID: 35803770 DOI: 10.1053/j.semnuclmed.2022.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/11/2022]
Abstract
Since the introduction of PET/CT hybrid imaging about two decades ago the landscape of oncological imaging has fundamentally changed, opening a new era of molecular imaging with emphasis on functional characterization of biological processes such as metabolism, cellular proliferation, hypoxia, apoptosis, angiogenesis and immune response. The most commonly assessed functional hallmark of cancer is the increased metabolism in tumor cells due to well-known Warburg effect, because of which FDG has been the most employed radiotracer, the so-called pan-cancer agent, in oncological imaging. However, several limitations such as low specificity and low sensitivity for several histopathological forms of lung cancer as well as high background uptake in the normal tissue of FDG imaging lead to numerous serious pitfalls. This restricts its utilization and diagnostic value in lung cancer imaging, even though this is currently considered to be the method of choice in pulmonary cancer imaging. Accurate initial tumor staging and therapy response monitoring with respect to the TNM criteria plays a crucial role in therapy planning and management in patients with lung cancer. To this end, many efforts have been made for decades to develop novel PET radiopharmaceuticals with innovative approaches that go beyond the assessment of increased glycolytic activity alone. Radiopharmaceuticals targeting DNA synthesis, amino acid metabolism, angiogenesis, or hypoxia have been extensively studied, leading to the emergence of indications for specific clinical questions or as a complementary imaging tool alongside existing conventional or FDG imaging. Nevertheless, despite some initial encouraging results, these tracers couldn't gain a widespread use and acceptance in clinical routine. However, given its mechanism of action and some initial pilot studies regarding lung cancer imaging, FAPI has emerged as a very promising alternative tool that could provide superior or comparable diagnostic performance to FDG imaging in lung cancer entities. Thus, in this review article, we summarized the current PET radiopharmaceuticals, different imaging approaches and discussed the potential benefits and clinical applications of these agents in lung cancer imaging.
Collapse
Affiliation(s)
- Emil Novruzov
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Yuriko Mori
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Christina Antke
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Mardjan Dabir
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Dominik Schmitt
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan A Koerber
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Frederik L Giesel
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Dusseldorf, Dusseldorf, Germany.
| |
Collapse
|
18
|
Kovacs I, Bugyik E, Dezso K, Tarnoki-Zach J, Mehes E, Gulyas M, Czirok A, Lang E, Grusch M, Schelch K, Hegedus B, Horvath I, Barany N, Megyesfalvi Z, Tisza A, Lohinai Z, Hoda MA, Hoetzenecker K, Pezzella F, Paku S, Laszlo V, Dome B. Malignant pleural mesothelioma nodules remodel their surroundings to vascularize and grow. Transl Lung Cancer Res 2022; 11:991-1008. [PMID: 35832452 PMCID: PMC9271443 DOI: 10.21037/tlcr-21-828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 04/24/2022] [Indexed: 12/03/2022]
Abstract
Background The microanatomical steps of malignant pleural mesothelioma (MPM) vascularization and the resistance mechanisms to anti-angiogenic drugs in MPM are unclear. Methods We investigated the vascularization of intrapleurally implanted human P31 and SPC111 MPM cells. We also assessed MPM cell's motility, invasion and interaction with endothelial cells in vitro. Results P31 cells exhibited significantly higher two-dimensional (2D) motility and three-dimensional (3D) invasion than SPC111 cells in vitro. In co-cultures of MPM and endothelial cells, P31 spheroids permitted endothelial sprouting (ES) with minimal spatial distortion, whereas SPC111 spheroids repealed endothelial sprouts. Both MPM lines induced the early onset of submesothelial microvascular plexuses covering large pleural areas including regions distant from tumor colonies. The development of these microvascular networks occurred due to both intussusceptive angiogenesis (IA) and ES and was accelerated by vascular endothelial growth factor A (VEGF-A)-overexpression. Notably, SPC111 colonies showed different behavior to P31 cells. P31 nodules incorporated tumor-induced capillary plexuses from the earliest stages of tumor formation. P31 cells deposited a collagenous matrix of human origin which provided "space" for further intratumoral angiogenesis. In contrast, SPC111 colonies pushed the capillary plexuses away and thus remained avascular for weeks. The key event in SPC111 vascularization was the development of a desmoplastic matrix of mouse origin. Continuously invaded by SPC111 cells, this matrix transformed into intratumoral connective tissue trunks, providing a route for ES from the diaphragm. Conclusions Here, we report two distinct growth patterns of orthotopically implanted human MPM xenografts. In the invasive pattern, MPM cells invade and thus co-opt peritumoral capillary plexuses. In the pushing/desmoplastic pattern, MPM cells induce a desmoplastic response within the underlying tissue which allows the ingrowth of a nutritive vasculature from the pleura.
Collapse
Affiliation(s)
- Ildiko Kovacs
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Edina Bugyik
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Katalin Dezso
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | | | - Elod Mehes
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Biological Physics, Eotvos University, Budapest, Hungary
| | - Marton Gulyas
- Department of Biological Physics, Eotvos University, Budapest, Hungary
| | - Andras Czirok
- Department of Biological Physics, Eotvos University, Budapest, Hungary
- Department of Anatomy & Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Elisabeth Lang
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Michael Grusch
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Karin Schelch
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Balazs Hegedus
- Department of Thoracic Surgery, Ruhrlandklinik, University Clinic Essen, Essen, Germany
| | - Ildiko Horvath
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Nandor Barany
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Zsolt Megyesfalvi
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary
| | - Anna Tisza
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Zoltan Lohinai
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Francesco Pezzella
- Nuffield Division of Laboratory Science, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Sandor Paku
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Viktoria Laszlo
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Balazs Dome
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary
| |
Collapse
|
19
|
Mosteiro A, Pedrosa L, Ferrés A, Diao D, Sierra À, González JJ. The Vascular Microenvironment in Glioblastoma: A Comprehensive Review. Biomedicines 2022; 10:biomedicines10061285. [PMID: 35740307 PMCID: PMC9219822 DOI: 10.3390/biomedicines10061285] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma multiforme, the deadliest primary brain tumor, is characterized by an excessive and aberrant neovascularization. The initial expectations raised by anti-angiogenic drugs were soon tempered due to their limited efficacy in improving the overall survival. Intrinsic resistance and escape mechanisms against anti-VEGF therapies evidenced that tumor angiogenesis is an intricate multifaceted phenomenon and that vessels not only support the tumor but exert indispensable interactions for resistance and spreading. This holistic review covers the essentials of the vascular microenvironment of glioblastoma, including the perivascular niche components, the vascular generation patterns and the implicated signaling pathways, the endothelial–tumor interrelation, and the interconnection between vessel aberrancies and immune disarrangement. The revised concepts provide novel insights into the preclinical models and the potential explanations for the failure of conventional anti-angiogenic therapies, leading to an era of new and combined anti-angiogenic-based approaches.
Collapse
Affiliation(s)
- Alejandra Mosteiro
- Department of Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (A.F.); (J.J.G.)
- Correspondence:
| | - Leire Pedrosa
- Laboratory of Experimental Oncological Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (L.P.); (D.D.); (À.S.)
| | - Abel Ferrés
- Department of Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (A.F.); (J.J.G.)
| | - Diouldé Diao
- Laboratory of Experimental Oncological Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (L.P.); (D.D.); (À.S.)
| | - Àngels Sierra
- Laboratory of Experimental Oncological Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (L.P.); (D.D.); (À.S.)
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - José Juan González
- Department of Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (A.F.); (J.J.G.)
- Laboratory of Experimental Oncological Neurosurgery, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (L.P.); (D.D.); (À.S.)
| |
Collapse
|
20
|
Ippolitov D, Arreza L, Munir MN, Hombach-Klonisch S. Brain Microvascular Pericytes—More than Bystanders in Breast Cancer Brain Metastasis. Cells 2022; 11:cells11081263. [PMID: 35455945 PMCID: PMC9028330 DOI: 10.3390/cells11081263] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 01/27/2023] Open
Abstract
Brain tissue contains the highest number of perivascular pericytes compared to other organs. Pericytes are known to regulate brain perfusion and to play an important role within the neurovascular unit (NVU). The high phenotypic and functional plasticity of pericytes make this cell type a prime candidate to aid physiological adaptations but also propose pericytes as important modulators in diverse pathologies in the brain. This review highlights known phenotypes of pericytes in the brain, discusses the diverse markers for brain pericytes, and reviews current in vitro and in vivo experimental models to study pericyte function. Our current knowledge of pericyte phenotypes as it relates to metastatic growth patterns in breast cancer brain metastasis is presented as an example for the crosstalk between pericytes, endothelial cells, and metastatic cells. Future challenges lie in establishing methods for real-time monitoring of pericyte crosstalk to understand causal events in the brain metastatic process.
Collapse
Affiliation(s)
- Danyyl Ippolitov
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (D.I.); (L.A.); (M.N.M.)
| | - Leanne Arreza
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (D.I.); (L.A.); (M.N.M.)
| | - Maliha Nuzhat Munir
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (D.I.); (L.A.); (M.N.M.)
| | - Sabine Hombach-Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (D.I.); (L.A.); (M.N.M.)
- Department of Pathology, University of Manitoba, Winnipeg, MB R3E 0Z2, Canada
- Correspondence:
| |
Collapse
|
21
|
Ignatova EO, Kozlov E, Ivanov M, Mileyko V, Menshikova S, Sun H, Fedyanin M, Tryakin A, Stilidi I. Clinical significance of molecular subtypes of gastrointestinal tract adenocarcinoma. World J Gastrointest Oncol 2022; 14:628-645. [PMID: 35321271 PMCID: PMC8919013 DOI: 10.4251/wjgo.v14.i3.628] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/04/2021] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Adenocarcinomas of the gastrointestinal tract (esophagus, stomach, and colon) represent a heterogeneous group of diseases with distinct etiology, clinical features, treatment approaches, and prognosis. Studies are ongoing to isolate molecular genetic subtypes, perform complete biological characterization of the tumor, determine prognostic groups, and find predictive markers to the effectiveness of therapy. Separate molecular genetic classifications were created for esophageal adenocarcinoma [The Cancer Genome Atlas (TCGA)], stomach cancer (TCGA, Asian Cancer Research Group), and colon cancer (Colorectal Cancer Subtyping Consortium). In 2018, isolation of TCGA molecular genetic subtypes for adenocarcinomas of the gastrointestinal tract (esophagus, stomach, and colon) highlighted the need for further studies and clinical validation of subtyping of gastrointestinal adenocarcinomas. However, this approach has limitations. The aim of our work was to critically analyze integration of molecular genetic subtyping of gastrointestinal adenocarcinomas in clinical practice.
Collapse
Affiliation(s)
- Ekaterina Olegovna Ignatova
- Department of Second Chemotherapy, Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Moscow, Russia
- Department of Oncogenetics, Research Centre for Medical Genetics Research Centre for Medical Genetics, Moscow 115522, Moscow, Russia
| | | | - Maxim Ivanov
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Moscow 141700, Moscow, Russia
| | | | - Sofia Menshikova
- Department of Anticancer Drug Treatment, AO K31 City, Moscow 121552, Moscow, Russia
| | - Henian Sun
- Pirogov Russian National Research Medical University (Pirogov Medical University), Moscow 117997, Moscow, Russia
| | - Mikhail Fedyanin
- Department of Second Chemotherapy, Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Moscow, Russia
| | - Alexey Tryakin
- Department of Second Chemotherapy, Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Moscow, Russia
| | - Ivan Stilidi
- Department of Second Chemotherapy, Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Moscow, Russia
| |
Collapse
|
22
|
WISP2/CCN5 Suppresses Vasculogenic Mimicry through Inhibition of YAP/TAZ Signaling in Breast Cancer Cells. Cancers (Basel) 2022; 14:cancers14061487. [PMID: 35326638 PMCID: PMC8945957 DOI: 10.3390/cancers14061487] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Breast cancer is the most frequent malignancy in women worldwide. Advanced breast cancer with distant organ metastases is considered incurable with currently available therapies. The vasculogenic mimicry (VM) process is associated with an invasive and metastatic cancer phenotype and a poor prognosis for human breast cancer patients. Our aim was to study the effect of WISP2, a matricellular protein, on VM. We found that WISP2 inhibits VM through inhibition of CYR61 protein expression and YAP-TAZ signaling. Our finding may open promising candidates for blocking VM in breast cancer. Abstract Vasculogenic mimicry (VM) formed by aggressive tumor cells to create vascular networks connected with the endothelial cells, plays an important role in breast cancer progression. WISP2 has been considered as a tumor suppressor protein; however, the relationship between WISP2 and VM formation remains unclear. We used the in vitro tube formation assay and in vivo immunohistochemical analysis in a mouse model, and human breast tumors were used to evaluate the effect of WISP2 on VM formation. Here we report that WISP2 acts as a potent inhibitor of VM formation in breast cancer. Enforced expression of WISP2 decreased network formation while knockdown of WISP2 increased VM. Mechanistically, WISP2 increased retention of oncogenic activators YAP/TAZ in cytoplasm, leading to decreased expression of the angiogenic factor CYR61. Studies using an in vivo mouse model and human breast tumors confirmed the in vitro cell lines data. In conclusion, our results indicate that WISP2 may play a critical role in VM and highlight the critical role of WISP2 as a tumor suppressor.
Collapse
|
23
|
Tsujimoto Y, Matsumoto Y, Tanaka M, Imabayashi T, Uchimura K, Tsuchida T. Diagnostic Value of Bronchoscopy for Peripheral Metastatic Lung Tumors. Cancers (Basel) 2022; 14:cancers14020375. [PMID: 35053537 PMCID: PMC8773960 DOI: 10.3390/cancers14020375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Metastatic lung tumors are relatively common, and their pathological diagnosis is crucial for determining the appropriate treatment strategy. Although bronchoscopy is one biopsy method, its role has not been established for peripheral metastatic lung tumors. The present study aimed to investigate the value of bronchoscopy, using radial endobronchial ultrasound for diagnosis. We analyzed 235 lesions consecutively, and the overall diagnostic yield was 76.6%, which was slightly higher than the reported cumulative sensitivity for general peripheral pulmonary lesions. There were no serious complications. The results demonstrated that bronchoscopy is a valuable technique for peripheral metastatic lung tumors that combines diagnostic accuracy and safety. Moreover, the higher diagnostic yield was associated with the large lesion size, inner location, and visibility on radiography. These findings should contribute to the selection of biopsy methods for metastatic lung tumors and improve the diagnostic yield. Abstract Although lungs are one of the most frequent sites of metastasis for malignant tumors, little has been reported about the value of bronchoscopy for lung metastases presenting with peripheral pulmonary lesions (PPLs). This retrospective cohort study investigated the diagnostic value of bronchoscopy for peripheral metastatic lung tumors. Consecutive patients who underwent diagnostic bronchoscopy with radial endobronchial ultrasound for PPLs and were finally diagnosed with metastatic lung tumors from April 2012 to March 2019 were included. We analyzed 235 PPLs, with a median size of 18.8 mm. The overall diagnostic yield was 76.6%. In a multivariable analysis, large lesion size (>20.0 mm vs. <20.0 mm: 87.6% vs. 67.7%, p = 0.043, OR = 2.26), inner location (inner 2/3 vs. outer 1/3: 84.8% vs. 69.1%, p = 0.004, OR = 2.79), and visibility on radiography (visible vs. invisible: 83.2% vs. 56.1%, p = 0.015, OR = 3.29) significantly affected the diagnostic yield. Although a positive bronchus sign tended to have a higher yield, no significant difference was observed (81.8% vs. 70.6%, p = 0.063). Only one case of lung abscess was observed, with no serious complications. In conclusion, bronchoscopy is a valuable technique for peripheral metastatic lung tumors, with good diagnostic accuracy and safety.
Collapse
Affiliation(s)
- Yoshie Tsujimoto
- Department of Endoscopy, Respiratory Endoscopy Division, National Cancer Center Hospital, Tokyo 1040045, Japan; (Y.T.); (M.T.); (T.I.); (K.U.); (T.T.)
- Department of Respiratory Medicine, National Center for Global Health and Medicine, Tokyo 1628655, Japan
| | - Yuji Matsumoto
- Department of Endoscopy, Respiratory Endoscopy Division, National Cancer Center Hospital, Tokyo 1040045, Japan; (Y.T.); (M.T.); (T.I.); (K.U.); (T.T.)
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo 1040045, Japan
- Correspondence: ; Tel.: +813-3542-2511
| | - Midori Tanaka
- Department of Endoscopy, Respiratory Endoscopy Division, National Cancer Center Hospital, Tokyo 1040045, Japan; (Y.T.); (M.T.); (T.I.); (K.U.); (T.T.)
| | - Tatsuya Imabayashi
- Department of Endoscopy, Respiratory Endoscopy Division, National Cancer Center Hospital, Tokyo 1040045, Japan; (Y.T.); (M.T.); (T.I.); (K.U.); (T.T.)
| | - Keigo Uchimura
- Department of Endoscopy, Respiratory Endoscopy Division, National Cancer Center Hospital, Tokyo 1040045, Japan; (Y.T.); (M.T.); (T.I.); (K.U.); (T.T.)
| | - Takaaki Tsuchida
- Department of Endoscopy, Respiratory Endoscopy Division, National Cancer Center Hospital, Tokyo 1040045, Japan; (Y.T.); (M.T.); (T.I.); (K.U.); (T.T.)
| |
Collapse
|
24
|
Pezzella F, Kerbel RS. On coalescent angiogenesis and the remarkable flexibility of blood vessels. Angiogenesis 2022; 25:1-3. [PMID: 34993716 DOI: 10.1007/s10456-021-09825-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Francesco Pezzella
- Nuffield Division of Laboratory Science, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK.
| | - Robert S Kerbel
- Biological Sciences Platform, Department of Medical Biophysics, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| |
Collapse
|
25
|
Mukherjee S, Madamsetty VS. Nanomedicine: An Alternative Approach Towards Anti-angiogenic Cancer Therapy. SYNTHESIS LECTURES ON BIOMEDICAL ENGINEERING 2022:21-31. [DOI: 10.1007/978-3-031-11284-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
26
|
The Colorectal Cancer Tumor Microenvironment and Its Impact on Liver and Lung Metastasis. Cancers (Basel) 2021; 13:cancers13246206. [PMID: 34944826 PMCID: PMC8699466 DOI: 10.3390/cancers13246206] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Colorectal cancer (CRC) is the third most common cancer worldwide. Metastasis to secondary organs, such as the liver and lungs, is a key driver of CRC-related mortality. The tumor microenvironment, which consists of the primary cancer cells, as well as associated support and immune cells, significantly affects the behavior of CRC cells at the primary tumor site, as well as in metastatic lesions. In this paper, we review the role of the individual components of the tumor microenvironment on tumor progression, immune evasion, and metastasis, and we discuss the implications of these components on antitumor therapies. Abstract Colorectal cancer (CRC) is the third most common malignancy and the second most common cause of cancer-related mortality worldwide. A total of 20% of CRC patients present with distant metastases, most frequently to the liver and lung. In the primary tumor, as well as at each metastatic site, the cellular components of the tumor microenvironment (TME) contribute to tumor engraftment and metastasis. These include immune cells (macrophages, neutrophils, T lymphocytes, and dendritic cells) and stromal cells (cancer-associated fibroblasts and endothelial cells). In this review, we highlight how the TME influences tumor progression and invasion at the primary site and its function in fostering metastatic niches in the liver and lungs. We also discuss emerging clinical strategies to target the CRC TME.
Collapse
|
27
|
Cocola C, Magnaghi V, Abeni E, Pelucchi P, Martino V, Vilardo L, Piscitelli E, Consiglio A, Grillo G, Mosca E, Gualtierotti R, Mazzaccaro D, La Sala G, Di Pietro C, Palizban M, Liuni S, DePedro G, Morara S, Nano G, Kehler J, Greve B, Noghero A, Marazziti D, Bussolino F, Bellipanni G, D'Agnano I, Götte M, Zucchi I, Reinbold R. Transmembrane Protein TMEM230, a Target of Glioblastoma Therapy. Front Cell Neurosci 2021; 15:703431. [PMID: 34867197 PMCID: PMC8636015 DOI: 10.3389/fncel.2021.703431] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 10/12/2021] [Indexed: 11/13/2022] Open
Abstract
Glioblastomas (GBM) are the most aggressive tumors originating in the brain. Histopathologic features include circuitous, disorganized, and highly permeable blood vessels with intermittent blood flow. These features contribute to the inability to direct therapeutic agents to tumor cells. Known targets for anti-angiogenic therapies provide minimal or no effect in overall survival of 12–15 months following diagnosis. Identification of novel targets therefore remains an important goal for effective treatment of highly vascularized tumors such as GBM. We previously demonstrated in zebrafish that a balanced level of expression of the transmembrane protein TMEM230/C20ORF30 was required to maintain normal blood vessel structural integrity and promote proper vessel network formation. To investigate whether TMEM230 has a role in the pathogenesis of GBM, we analyzed its prognostic value in patient tumor gene expression datasets and performed cell functional analysis. TMEM230 was found necessary for growth of U87-MG cells, a model of human GBM. Downregulation of TMEM230 resulted in loss of U87 migration, substratum adhesion, and re-passaging capacity. Conditioned media from U87 expressing endogenous TMEM230 induced sprouting and tubule-like structure formation of HUVECs. Moreover, TMEM230 promoted vascular mimicry-like behavior of U87 cells. Gene expression analysis of 702 patients identified that TMEM230 expression levels distinguished high from low grade gliomas. Transcriptomic analysis of patients with gliomas revealed molecular pathways consistent with properties observed in U87 cell assays. Within low grade gliomas, elevated TMEM230 expression levels correlated with reduced overall survival independent from tumor subtype. Highest level of TMEM230 correlated with glioblastoma and ATP-dependent microtubule kinesin motor activity, providing a direction for future therapeutic intervention. Our studies support that TMEM230 has both glial tumor and endothelial cell intracellular and extracellular functions. Elevated levels of TMEM230 promote glial tumor cell migration, extracellular scaffold remodeling, and hypervascularization and abnormal formation of blood vessels. Downregulation of TMEM230 expression may inhibit both low grade glioma and glioblastoma tumor progression and promote normalization of abnormally formed blood vessels. TMEM230 therefore is both a promising anticancer and antiangiogenic therapeutic target for inhibiting GBM tumor cells and tumor-driven angiogenesis.
Collapse
Affiliation(s)
- Cinzia Cocola
- Institute for Biomedical Technologies, National Research Council, Milan, Italy.,Consorzio Italbiotec, Milan, Italy
| | - Valerio Magnaghi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Edoardo Abeni
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Paride Pelucchi
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Valentina Martino
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Laura Vilardo
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Eleonora Piscitelli
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Arianna Consiglio
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Giorgio Grillo
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Ettore Mosca
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Roberta Gualtierotti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Mazzaccaro
- Operative Unit of Vascular Surgery, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Gina La Sala
- Institute of Biochemistry and Cell Biology, Italian National Research Council, Rome, Italy
| | - Chiara Di Pietro
- Institute of Biochemistry and Cell Biology, Italian National Research Council, Rome, Italy
| | - Mira Palizban
- Department of Gynecology and Obstetrics, University Hospital of Münster, Münster, Germany
| | - Sabino Liuni
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Giuseppina DePedro
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Giovanni Nano
- Operative Unit of Vascular Surgery, IRCCS Policlinico San Donato, San Donato Milanese, Italy.,Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - James Kehler
- National Institutes of Health, NIDDK, Laboratory of Cell and Molecular Biology, Bethesda, MD, United States
| | - Burkhard Greve
- Department of Radiation Therapy and Radiation Oncology, University Hospital of Münster, Münster, Germany
| | - Alessio Noghero
- Lovelace Biomedical Research Institute, Albuquerque, NM, United States.,Department of Oncology, University of Turin, Orbassano, Italy
| | - Daniela Marazziti
- Institute of Biochemistry and Cell Biology, Italian National Research Council, Rome, Italy
| | - Federico Bussolino
- Department of Oncology, University of Turin, Orbassano, Italy.,Laboratory of Vascular Oncology Candiolo Cancer Institute - IRCCS, Candiolo, Italy
| | - Gianfranco Bellipanni
- Department of Biology, Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United States
| | - Igea D'Agnano
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Martin Götte
- Department of Gynecology and Obstetrics, University Hospital of Münster, Münster, Germany
| | - Ileana Zucchi
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Rolland Reinbold
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| |
Collapse
|
28
|
Footprints of microRNAs in Cancer Biology. Biomedicines 2021; 9:biomedicines9101494. [PMID: 34680611 PMCID: PMC8533183 DOI: 10.3390/biomedicines9101494] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs involved in post-transcriptional gene regulation. Over the past years, various studies have demonstrated the role of aberrant miRNA expression in the onset of cancer. The mechanisms by which miRNA exerts its cancer-promoting or inhibitory effects are apparent through the various cancer hallmarks, which include selective proliferative advantage, altered stress response, vascularization, invasion and metastasis, metabolic rewiring, the tumor microenvironment and immune modulation; therefore, this review aims to highlight the association between miRNAs and the various cancer hallmarks by dissecting the mechanisms of miRNA regulation in each hallmark separately. It is hoped that the information presented herein will provide further insights regarding the role of cancer and serve as a guideline to evaluate the potential of microRNAs to be utilized as biomarkers and therapeutic targets on a larger scale in cancer research.
Collapse
|
29
|
Akgol S, Kalkan BM, Yucel D, Kocabas F. SC1 limits tube formation, branching, migration, expansion and induce apoptosis of endothelial cells. Vascul Pharmacol 2021; 141:106903. [PMID: 34481979 DOI: 10.1016/j.vph.2021.106903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/02/2021] [Accepted: 08/27/2021] [Indexed: 11/30/2022]
Abstract
Endothelial cells (ECs) are essential in the growth and progression of the tumor cells by supplying nutrition and angiogenesis factors. Targeting ECs emerged as a major strategy to prevent the growth of tumors. Studies suggest that ERK1/2 signaling is important for endothelial cells, which could be specifically targeted by small molecule SC1. We aimed to study the effects of SC1 treatments on endothelial cell proliferation, angiogenesis, and death. To this end, we performed viability, apoptosis, cell cycle, gene expression, wound closure, tube formation, and western blot analysis in endothelial cells post SC1 treatments. Intriguingly, we found that SC1 has an antiangiogenic effect on endothelial cells, which limits the endothelial cell expansion, tube formation, branching, and migration. The proliferation is especially limited in dose dependent manner by SC1. In addition, we found that SC1 elevates the apoptosis of endothelial cells and associated pathways including BAK1, Stat1, Sox4, and Caspase1. We believe that these findings could contribute to the development of improved therapies based on the SC1 as an attractive candidate for anticancer clinical studies targeted to tumor angiogenesis.
Collapse
Affiliation(s)
- Sezer Akgol
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | | | - Dogacan Yucel
- Department of Medicine, University of Minnesota, USA
| | - Fatih Kocabas
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
30
|
Rosińska S, Gavard J. Tumor Vessels Fuel the Fire in Glioblastoma. Int J Mol Sci 2021; 22:6514. [PMID: 34204510 PMCID: PMC8235363 DOI: 10.3390/ijms22126514] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma, a subset of aggressive brain tumors, deploy several means to increase blood vessel supply dedicated to the tumor mass. This includes typical program borrowed from embryonic development, such as vasculogenesis and sprouting angiogenesis, as well as unconventional processes, including co-option, vascular mimicry, and transdifferentiation, in which tumor cells are pro-actively engaged. However, these neo-generated vascular networks are morphologically and functionally abnormal, suggesting that the vascularization processes are rather inefficient in the tumor ecosystem. In this review, we reiterate the specificities of each neovascularization modality in glioblastoma, and, how they can be hampered mechanistically in the perspective of anti-cancer therapies.
Collapse
Affiliation(s)
- Sara Rosińska
- CRCINA, Inserm, CNRS, Université de Nantes, 44000 Nantes, France;
| | - Julie Gavard
- CRCINA, Inserm, CNRS, Université de Nantes, 44000 Nantes, France;
- Integrated Center for Oncology, ICO, 44800 St. Herblain, France
| |
Collapse
|
31
|
Teuwen LA, De Rooij LPMH, Cuypers A, Rohlenova K, Dumas SJ, García-Caballero M, Meta E, Amersfoort J, Taverna F, Becker LM, Veiga N, Cantelmo AR, Geldhof V, Conchinha NV, Kalucka J, Treps L, Conradi LC, Khan S, Karakach TK, Soenen S, Vinckier S, Schoonjans L, Eelen G, Van Laere S, Dewerchin M, Dirix L, Mazzone M, Luo Y, Vermeulen P, Carmeliet P. Tumor vessel co-option probed by single-cell analysis. Cell Rep 2021; 35:109253. [PMID: 34133923 DOI: 10.1016/j.celrep.2021.109253] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 05/16/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022] Open
Abstract
Tumor vessel co-option is poorly understood, yet it is a resistance mechanism against anti-angiogenic therapy (AAT). The heterogeneity of co-opted endothelial cells (ECs) and pericytes, co-opting cancer and myeloid cells in tumors growing via vessel co-option, has not been investigated at the single-cell level. Here, we use a murine AAT-resistant lung tumor model, in which VEGF-targeting induces vessel co-option for continued growth. Single-cell RNA sequencing (scRNA-seq) of 31,964 cells reveals, unexpectedly, a largely similar transcriptome of co-opted tumor ECs (TECs) and pericytes as their healthy counterparts. Notably, we identify cell types that might contribute to vessel co-option, i.e., an invasive cancer-cell subtype, possibly assisted by a matrix-remodeling macrophage population, and another M1-like macrophage subtype, possibly involved in keeping or rendering vascular cells quiescent.
Collapse
Affiliation(s)
- Laure-Anne Teuwen
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium; Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp 2610, Belgium; Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Laura P M H De Rooij
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Anne Cuypers
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Katerina Rohlenova
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Sébastien J Dumas
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Melissa García-Caballero
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Elda Meta
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Jacob Amersfoort
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Federico Taverna
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Lisa M Becker
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Nuphar Veiga
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Anna Rita Cantelmo
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Vincent Geldhof
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Nadine V Conchinha
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Joanna Kalucka
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Lena-Christin Conradi
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Shawez Khan
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Tobias K Karakach
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Stefaan Soenen
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Leuven 3000, Belgium
| | - Stefan Vinckier
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Luc Schoonjans
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 510275, Guangzhou, Guangdong, P.R. China
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Steven Van Laere
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp 2610, Belgium; Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Luc Dirix
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp 2610, Belgium; Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, CCB, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Lars Bolund Institute of Regenerative Medicine, BGI-Qingdao, Qingdao 266555, P.R. China; BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank, BGI-Shenzhen, Shenzhen 518120, P.R. China.
| | - Peter Vermeulen
- Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp 2610, Belgium; Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 510275, Guangzhou, Guangdong, P.R. China; Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark.
| |
Collapse
|
32
|
Abstract
The development of tumors requires an initiator event, usually exposure to DNA damaging agents that cause genetic alterations such as gene mutations or chromosomal abnormalities, leading to deregulated cell proliferation. Although the mere stochastic accumulation of further mutations may cause tumor progression, it is now clear that an inflammatory microenvironment has a major tumor-promoting influence on initiated cells, in particular when a chronic inflammatory reaction already existed before the initiated tumor cell was formed. Moreover, inflammatory cells become mobilized in response to signals emanating from tumor cells. In both cases, the microenvironment provides signals that initiated tumor cells perceive by membrane receptors and transduce via downstream kinase cascades to modulate multiple cellular processes and respond with changes in cell gene expression, metabolism, and morphology. Cytokines, chemokines, and growth factors are examples of major signals secreted by immune cells, fibroblast, and endothelial cells and mediate an intricate cell-cell crosstalk in an inflammatory microenvironment, which contributes to increased cancer cell survival, phenotypic plasticity and adaptation to surrounding tissue conditions. Eventually, consequent changes in extracellular matrix stiffness and architecture, coupled with additional genetic alterations, further fortify the malignant progression of tumor cells, priming them for invasion and metastasis. Here, we provide an overview of the current knowledge on the composition of the inflammatory tumor microenvironment, with an emphasis on the major signals and signal-transducing events mediating different aspects of stromal cell-tumor cell communication that ultimately lead to malignant progression.
Collapse
|
33
|
Florea A, Mottaghy FM, Bauwens M. Molecular Imaging of Angiogenesis in Oncology: Current Preclinical and Clinical Status. Int J Mol Sci 2021; 22:5544. [PMID: 34073992 PMCID: PMC8197399 DOI: 10.3390/ijms22115544] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis is an active process, regulating new vessel growth, and is crucial for the survival and growth of tumours next to other complex factors in the tumour microenvironment. We present possible molecular imaging approaches for tumour vascularisation and vitality, focusing on radiopharmaceuticals (tracers). Molecular imaging in general has become an integrated part of cancer therapy, by bringing relevant insights on tumour angiogenic status. After a structured PubMed search, the resulting publication list was screened for oncology related publications in animals and humans, disregarding any cardiovascular findings. The tracers identified can be subdivided into direct targeting of angiogenesis (i.e., vascular endothelial growth factor, laminin, and fibronectin) and indirect targeting (i.e., glucose metabolism, hypoxia, and matrix metallo-proteases, PSMA). Presenting pre-clinical and clinical data of most tracers proposed in the literature, the indirect targeting agents are not 1:1 correlated with angiogenesis factors but do have a strong prognostic power in a clinical setting, while direct targeting agents show most potential and specificity for assessing tumour vascularisation and vitality. Within the direct agents, the combination of multiple targeting tracers into one agent (multimers) seems most promising. This review demonstrates the present clinical applicability of indirect agents, but also the need for more extensive research in the field of direct targeting of angiogenesis in oncology. Although there is currently no direct tracer that can be singled out, the RGD tracer family seems to show the highest potential therefore we expect one of them to enter the clinical routine.
Collapse
Affiliation(s)
- Alexandru Florea
- Department of Nuclear Medicine, University Hospital RWTH Aachen, 52074 Aachen, Germany; (A.F.); (M.B.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229HX Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht University, 6229HX Maastricht, The Netherlands
| | - Felix M. Mottaghy
- Department of Nuclear Medicine, University Hospital RWTH Aachen, 52074 Aachen, Germany; (A.F.); (M.B.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229HX Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht University, 6229HX Maastricht, The Netherlands
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229HX Maastricht, The Netherlands
| | - Matthias Bauwens
- Department of Nuclear Medicine, University Hospital RWTH Aachen, 52074 Aachen, Germany; (A.F.); (M.B.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229HX Maastricht, The Netherlands
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229HX Maastricht, The Netherlands
| |
Collapse
|
34
|
Sebestyén A, Kopper L, Dankó T, Tímár J. Hypoxia Signaling in Cancer: From Basics to Clinical Practice. Pathol Oncol Res 2021; 27:1609802. [PMID: 34257622 PMCID: PMC8262153 DOI: 10.3389/pore.2021.1609802] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/14/2021] [Indexed: 12/24/2022]
Abstract
Cancer hypoxia, recognized as one of the most important hallmarks of cancer, affects gene expression, metabolism and ultimately tumor biology-related processes. Major causes of cancer hypoxia are deficient or inappropriate vascularization and systemic hypoxia of the patient (frequently induced by anemia), leading to a unique form of genetic reprogramming by hypoxia induced transcription factors (HIF). However, constitutive activation of oncogene-driven signaling pathways may also activate hypoxia signaling independently of oxygen supply. The consequences of HIF activation in tumors are the angiogenic phenotype, a novel metabolic profile and the immunosuppressive microenvironment. Cancer hypoxia and the induced adaptation mechanisms are two of the major causes of therapy resistance. Accordingly, it seems inevitable to combine various therapeutic modalities of cancer patients by existing anti-hypoxic agents such as anti-angiogenics, anti-anemia therapies or specific signaling pathway inhibitors. It is evident that there is an unmet need in cancer patients to develop targeted therapies of hypoxia to improve efficacies of various anti-cancer therapeutic modalities. The case has been opened recently due to the approval of the first-in-class HIF2α inhibitor.
Collapse
Affiliation(s)
- Anna Sebestyén
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - László Kopper
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Titanilla Dankó
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - József Tímár
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
35
|
Abstract
Liver metastases are commonly detected in a range of malignancies including colorectal cancer (CRC), pancreatic cancer, melanoma, lung cancer and breast cancer, although CRC is the most common primary cancer that metastasizes to the liver. Interactions between tumour cells and the tumour microenvironment play an important part in the engraftment, survival and progression of the metastases. Various cells including liver sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, parenchymal hepatocytes, dendritic cells, resident natural killer cells as well as other immune cells such as monocytes, macrophages and neutrophils are implicated in promoting and sustaining metastases in the liver. Four key phases (microvascular, pre-angiogenic, angiogenic and growth phases) have been identified in the process of liver metastasis. Imaging modalities such as ultrasonography, CT, MRI and PET scans are typically used for the diagnosis of liver metastases. Surgical resection remains the main potentially curative treatment among patients with resectable liver metastases. The role of liver transplantation in the management of liver metastasis remains controversial. Systemic therapies, newer biologic agents (for example, bevacizumab and cetuximab) and immunotherapeutic agents have revolutionized the treatment options for liver metastases. Moving forward, incorporation of genetic tests can provide more accurate information to guide clinical decision-making and predict prognosis among patients with liver metastases.
Collapse
|
36
|
Alternative Vascularization Mechanisms in Tumor Resistance to Therapy. Cancers (Basel) 2021; 13:cancers13081912. [PMID: 33921099 PMCID: PMC8071410 DOI: 10.3390/cancers13081912] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Tumors rely on blood vessels to grow and metastasize. Malignant tumors can employ different strategies to create a functional vascular network. Tumor cells can use normal processes of vessel formation but can also employ cancer-specific mechanisms, by co-opting normal vessels present in tissues or by turning themselves into vascular cells. These different types of tumor vessels have specific molecular and functional characteristics that profoundly affect tumor behavior and response to therapies, including drugs targeting the tumor vasculature (antiangiogenic therapies). In this review, we discuss how vessels formed by different mechanisms affect the intrinsic sensitivity of tumors to therapy and, on the other hand, how therapies can affect tumor vessel formation, leading to resistance to drugs, cancer recurrence, and treatment failure. Potential strategies to avoid vessel-mediated resistance to antineoplastic therapies will be discussed. Abstract Blood vessels in tumors are formed through a variety of different mechanisms, each generating vessels with peculiar structural, molecular, and functional properties. This heterogeneity has a major impact on tumor response or resistance to antineoplastic therapies and is now emerging as a promising target for strategies to prevent drug resistance and improve the distribution and efficacy of antineoplastic treatments. This review presents evidence of how different mechanisms of tumor vessel formation (vasculogenesis, glomeruloid proliferation, intussusceptive angiogenesis, vasculogenic mimicry, and vessel co-option) affect tumor responses to antiangiogenic and antineoplastic therapies, but also how therapies can promote alternative mechanisms of vessel formation, contributing to tumor recurrence, malignant progression, and acquired drug resistance. We discuss the possibility of tailoring treatment strategies to overcome vasculature-mediated drug resistance or to improve drug distribution and efficacy.
Collapse
|
37
|
Sphyris N, King C, Hoar J, Werden SJ, Vijay GV, Miura N, Gaharwar A, Sarkar TR. Carcinoma cells that have undergone an epithelial-mesenchymal transition differentiate into endothelial cells and contribute to tumor growth. Oncotarget 2021; 12:823-844. [PMID: 33889304 PMCID: PMC8057273 DOI: 10.18632/oncotarget.27940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/25/2021] [Indexed: 12/23/2022] Open
Abstract
Hypoxia stimulates neoangiogenesis, promoting tumor outgrowth, and triggers the epithelial-mesenchymal transition (EMT), which bestows cells with mesenchymal traits and multi-lineage differentiation potential. Here, we investigated whether EMT can confer endothelial attributes upon carcinoma cells, augmenting tumor growth and vascularization. Following orthotopic implantation of MCF-7 human epithelial breast cancer cells into mice, tumors of different sizes were immunostained for markers of hypoxia and EMT. Larger tumors were well-vascularized with CD31-positive cells of human origin. Hypoxic regions, demarcated by HIF-1α staining, exhibited focal areas of E-cadherin loss and elevated levels of vimentin and the EMT-mediator FOXC2. Implantation of MCF-7 cells, co-mixed with human mammary epithelial (HMLE) cells overexpressing the EMT-inducer Snail, markedly potentiated tumor growth and vascularization, compared with MCF-7 cells injected alone or co-mixed with HMLE-vector cells. Intra-tumoral vessels contained CD31-positive cells derived from either donor cell type. FOXC2 knockdown abrogated the potentiating effects of HMLE-Snail cells on MCF-7 tumor growth and vascularization, and compromised endothelial transdifferentiation of mesenchymal cells cultured in endothelial growth medium. Hence, cells that have undergone EMT can promote tumor growth and neovascularization either indirectly, by promoting endothelial transdifferentiation of carcinoma cells, or directly, by acquiring an endothelial phenotype, with FOXC2 playing key roles in these processes.
Collapse
Affiliation(s)
- Nathalie Sphyris
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Present address: Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK.,These authors contributed equally to this work
| | - Cody King
- Department of Biochemistry, Texas A&M University, College Station, TX, USA
| | - Jonathan Hoar
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Steven J Werden
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Geraldine V Vijay
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoyuki Miura
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Akhilesh Gaharwar
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Tapasree Roy Sarkar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Biology, Texas A&M University, College Station, TX, USA.,These authors contributed equally to this work
| |
Collapse
|
38
|
Hashemzehi M, Yavari N, Rahmani F, Asgharzadeh F, Soleimani A, Shakour N, Avan A, Hadizadeh F, Fakhraie M, Marjaneh RM, Ferns GA, Reisi P, Ryzhikov M, Khazaei M, Hassanian SM. Inhibition of transforming growth factor-beta by Tranilast reduces tumor growth and ameliorates fibrosis in colorectal cancer. EXCLI JOURNAL 2021; 20:601-613. [PMID: 33883985 PMCID: PMC8056055 DOI: 10.17179/excli2020-2932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/03/2021] [Indexed: 12/15/2022]
Abstract
Transforming Growth Factor-beta (TGF-β) is dysregulated in colorectal cancer and there is growing evidence that it is associated with a poor prognosis and chemo-resistance in several malignances, including CRC. In this study we have explored the therapeutic potential of targeting TGF-β using Tranilast in colon cancer. The anti-proliferative activity of Tranilast was evaluated in 2- and 3-dimensional cells. We used a xenograft model of colon cancer to investigate the activity of Tranilast alone or in combination with 5-FU on tumor growth using histological staining and biochemical studies, as well as gene expression analyses using RT-PCR and Western blotting. Tranilast alone or in combination with 5-FU inhibited tumor growth and was associated with a reduction of TGF-β expression and CD31 positive endothelial cells. Histological evaluation showed that Tranilast increased tumor necrosis and reduced tumor density and angiogenesis. Tranilast increased MDA and ROS production. It was also found that Tranilast reduced total thiol concentration and reduced SOD and catalase activity. Tranilast plus 5-FU was also found to attenuate collagen deposition, reducing tumor fibrosis in tumor xenografts. Our results show that Tranilast, a TGF inhibitor, in combination with 5-FU reduces tumor growth by inhibiting fibrosis and inducting ROS, thus supporting this therapeutic approach in CRC treatment.
Collapse
Affiliation(s)
- Milad Hashemzehi
- Department of Medical Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Tropical and Communicable Diseases Research Centre, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Negar Yavari
- Department of Medical Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farzad Rahmani
- Tropical and Communicable Diseases Research Centre, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Fereshteh Asgharzadeh
- Department of Medical Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atena Soleimani
- Department of Clinical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Neda Shakour
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Fakhraie
- Department of Medical Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reyhaneh Moradi Marjaneh
- Department of Medical Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Parham Reisi
- Department of Medical Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Majid Khazaei
- Department of Medical Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
39
|
Chu B, He JM, Liu LL, Wu CX, You LL, Li XL, Wang S, Chen CS, Tu M. Proangiogenic Peptide Nanofiber Hydrogels for Wound Healing. ACS Biomater Sci Eng 2021; 7:1100-1110. [PMID: 33512985 DOI: 10.1021/acsbiomaterials.0c01264] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Rapid vascularization is vital for dermal regeneration, nutrient and nutrition transfer, metabolic waste removal, and prevention of infection. This study reports on a series of proangiogenic peptides designed to undergo self-assembly and promote angiogenesis and hence skin regeneration. The proangiogenic peptides comprised an angiogenic peptide segment, GEETEVTVEGLEPG, and a β-sheet structural peptide sequence. These peptides dissolved easily in ultrapure water and rapidly self-assembled into hydrogels in a pH-dependent manner, creating three-dimensional fibril network structures and nanofibers as revealed by a scanning microscope and a transmission electron microscope. In vitro experiments showed that the peptide hydrogels favored adhesion and proliferation of mouse fibroblasts (L929) and human umbilical vein endothelial cells (HUVECs). In particular, many connected tubes were formed in the HUVECs after 8 h of culture on the peptide hydrogels. In vivo experiments demonstrated that new blood vessels grew into the proangiogenic peptide hydrogels within 2 weeks after subcutaneous implantation in mice. Moreover, the proangiogenic-combined hydrogels exhibited faster repair cycles and better healing of skin defects. Collectively, the results indicate that the proangiogenic peptide hydrogels are a promising therapeutic option for skin regeneration.
Collapse
Affiliation(s)
- Bin Chu
- Department of Biomedical Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, P. R. China.,Key Laboratory of Biomedical Materials and Implant Devices, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, P. R. China
| | - Jin-Mei He
- Key Laboratory of Biomedical Materials and Implant Devices, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, P. R. China
| | - Lan-Lan Liu
- Key Laboratory of Biomedical Materials and Implant Devices, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, P. R. China
| | - Chao-Xi Wu
- Department of Biomedical Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, P. R. China
| | - Ling-Ling You
- Department of Biomedical Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, P. R. China
| | - Xiao-Li Li
- Key Laboratory of Biomedical Materials and Implant Devices, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, P. R. China
| | - Song Wang
- Key Laboratory of Biomedical Materials and Implant Devices, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, P. R. China
| | - Chang-Sheng Chen
- Key Laboratory of Biomedical Materials and Implant Devices, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, P. R. China
| | - Mei Tu
- Department of Biomedical Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, P. R. China
| |
Collapse
|
40
|
Xu C, Wang S, Wu Y, Sun X, Yang D, Wang S. Recent advances in understanding the roles of sialyltransferases in tumor angiogenesis and metastasis. Glycoconj J 2021; 38:119-127. [PMID: 33411077 DOI: 10.1007/s10719-020-09967-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 10/25/2020] [Accepted: 12/08/2020] [Indexed: 01/04/2023]
Abstract
Abnormal glycosylation is a common characteristic of cancer cells and there is a lot of evidence that glycans can regulate the biological behavior of tumor cells. Sialylation modification, a form of glycosylation modification, plays an important role in cell recognition, cell adhesion and cell signal transduction. Abnormal sialylation on the surface of tumor cells is related to tumor migration and invasion, with abnormal expression of sialyltransferases being one of the main causes of abnormal sialylation. Recent studies provide a better understanding of the importance of the sialyltransferases, and how they influences cancer cell angiogenesis, adhesion and Epithelial-Mesenchymal Transition (EMT). The present review will provide a direction for future studies in determining the roles of sialyltransferases in cancer metastasis, and abnormal sialyltransferases are likely to be potential biomarkers for cancer.
Collapse
Affiliation(s)
- Chunyan Xu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, People's Republic of China
| | - Shidan Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, People's Republic of China
| | - Yinshuang Wu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, People's Republic of China
| | - Xiaoxin Sun
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, People's Republic of China
| | - Deyong Yang
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, People's Republic of China.
| | - Shujing Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, People's Republic of China.
| |
Collapse
|
41
|
Daum S, Hagen H, Naismith E, Wolf D, Pircher A. The Role of Anti-angiogenesis in the Treatment Landscape of Non-small Cell Lung Cancer - New Combinational Approaches and Strategies of Neovessel Inhibition. Front Cell Dev Biol 2021; 8:610903. [PMID: 33469537 PMCID: PMC7813779 DOI: 10.3389/fcell.2020.610903] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor progression depends primarily on vascular supply, which is facilitated by angiogenic activity within the malignant tissue. Non-small cell lung cancer (NSCLC) is a highly vascularized tumor, and inhibition of angiogenesis was projected to be a promising therapeutic approach. Over a decade ago, the first anti-angiogenic agents were approved for advanced stage NSCLC patients, however, they only produced a marginal clinical benefit. Explanations why anti-angiogenic therapies only show modest effects include the highly adaptive tumor microenvironment (TME) as well as the less understood characteristics of the tumor vasculature. Today, advanced methods of in-depth characterization of the NSCLC TME by single cell RNA sequencing (scRNA-Seq) and preclinical observations enable a detailed characterization of individual cancer landscapes, allowing new aspects for a more individualized inhibition of angiogenesis to be identified. Furthermore, the tumor vasculature itself is composed of several cellular subtypes, which closely interact with other cellular components of the TME, and show distinct biological functions such as immune regulation, proliferation, and organization of the extracellular matrix. With these new insights, combinational approaches including chemotherapy, anti- angiogenic and immunotherapy can be developed to yield a more target-oriented anti-tumor treatment in NSCLC. Recently, anti-angiogenic agents were also shown to induce the formation of high endothelial venules (HEVs), which are essential for the formation of tertiary lymphoid structures, and key components in triggering anti-tumor immunity. In this review, we will summarize the current knowledge of tumor-angiogenesis and corresponding anti-angiogenic therapies, as well as new aspects concerning characterization of tumor-associated vessels and the resulting new strategies for anti-angiogenic therapies and vessel inhibition in NSCLC. We will further discuss why anti-angiogenic therapies form an interesting backbone strategy for combinational therapies and how anti-angiogenic approaches could be further developed in a more personalized tumor-oriented fashion with focus on NSCLC.
Collapse
Affiliation(s)
- Sophia Daum
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
| | - Hannes Hagen
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
| | - Erin Naismith
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
| | - Dominik Wolf
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
- Medical Clinic 3, Department of Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn (UKB), Bonn, Germany
| | - Andreas Pircher
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
42
|
The Dependence between Urinary Levels of Angiogenesis Factors, 8-Iso-prostaglandin F2 α, ɣ-Synuclein, and Interleukin-13 in Patients with Bladder Cancer: A Pilot Study. JOURNAL OF ONCOLOGY 2020; 2020:4848752. [PMID: 33343662 PMCID: PMC7725553 DOI: 10.1155/2020/4848752] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/09/2020] [Accepted: 11/21/2020] [Indexed: 02/06/2023]
Abstract
During the last decade, a significant increase in the incidence of bladder cancer (BC) has been observed. Angiogenesis plays a key role in the process of tumor growth and metastasis. Additionally, the participation of oxidative stress and chronic inflammation in BC pathogenesis is indicated. The aim of the study was to evaluate the urinary levels of parameters of angiogenesis, stimulating angiogenin (ANG) and inhibiting angiostatin (ANGST), 8-iso-prostaglandin F2α (8-iso-PGF2α) as a marker of oxidative stress, ɣ-synuclein (SNCG) as a cancer progression parameter, and interleukin-13 (IL-13) as an anti-inflammatory immunomodulator. The levels of ANG, ANGST, 8-iso-PGF2α, SNCG, and IL-13 in the urine of BC patients and healthy controls were measured by the enzyme-linked immunosorbent assay. These parameters were examined in the whole group of BC patients and in subgroups depending on the clinical stage: nonmuscle-invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC); histopathologic malignancy: low grade (LG) and high grade (HG) and in primary and recurrent BC. Significantly, higher urinary parameters were found in BC patients in comparison to controls. Levels of all parameters increased with the development of cancer, with the exception of 8-iso-prostaglandin F2α, in which the level was higher in the early stages of the disease, but these differences were not statistically significant. Some correlations have been demonstrated between parameters in BC patients. Based on the receiver operating characteristic curves, ANG and ANGST had the best diagnostic value for BC. The obtained results indicate the important role of the examined parameters of angiogenesis, oxidative stress, and inflammation in the pathogenesis and development of BC. It is reasonable to continue research in order to thoroughly assess the impact of various associated processes on the course of BC. It is also important to carry out similar tests in patients with other urological diseases.
Collapse
|
43
|
Ling W, Nie J, Zhang D, Yang Q, Jin H, Ou X, Ma X, Luo Y. Role of Contrast-Enhanced Ultrasound (CEUS) in the Diagnosis of Cervical Lymph Node Metastasis in Nasopharyngeal Carcinoma (NPC) Patients. Front Oncol 2020; 10:972. [PMID: 32766127 PMCID: PMC7379866 DOI: 10.3389/fonc.2020.00972] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/18/2020] [Indexed: 02/05/2023] Open
Abstract
Objective: The aim of the study was to evaluate the diagnostic value of contrast-enhanced ultrasound (CEUS) in distinguishing between benign and malignant cervical lymph nodes in patients with nasopharyngeal carcinoma (NPC). Material and Methods: A total of 144 NPC patients with enlarged superficial cervical lymph nodes underwent CEUS examination. The comparison of CEUS image characteristics between malignant and benign cervical lymph nodes was performed in this study as well. We analyzed parameters of the time-intensity curve (TIC), which includes time to peak (TP), area under the gamma curve (AUC), and peak intensity (PI). Furthermore, receiver operating characteristic (ROC) curve analysis was also investigated to evaluate the diagnostic value of CEUS. Result: We conducted 144 lymph node examinations in total, where 64 cases were biopsy-proven benign nodules and 80 cases were biopsy-proven metastatic nodules. The vast majority of the benign nodes displayed centrifugal perfusion (96.88%, 62/64) and homogeneous enhancement (93.75%, 60/64), while most of the malignant nodes showed centripetal perfusion (92.50%, 74/80) and inhomogeneous 80.00% (64/80). In addition, quantitative analysis showed that CEUS parameters including PI, TP, and AUC in benign lymph nodes (12.51 ± 2.15, 23.79 ± 11.80, and 1110.33 ± 286.17, respectively) were significantly higher than that in the malignant nodes (10.51 ± 2.98, 16.52 ± 6.95, and 784.09 ± 340.24, respectively). The assistance of the three aforementioned parameters and CEUS image characteristics would result in an acceptable diagnostic value. Conclusion: Our results suggest that imaging perfusion patterns as well as quantitative parameters obtained from CEUS provide valuable information for the evaluation of cervical lymph nodes in NPC patients.
Collapse
Affiliation(s)
- Wenwu Ling
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Ji Nie
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Dingyue Zhang
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Qianru Yang
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Hongyu Jin
- West China School of Medicine, Sichuan University, Chengdu, China
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xuejin Ou
- Department of Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yan Luo
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
44
|
Téglási V, Csűry DT, Dezső K, Bugyik E, Szabó V, Szállási Z, Paku S, Reiniger L. Origin and Distribution of Connective Tissue and Pericytes Impacting Vascularization in Brain Metastases With Different Growth Patterns. J Neuropathol Exp Neurol 2020; 78:326-339. [PMID: 30816955 DOI: 10.1093/jnen/nlz007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The impact of growth pattern on the distribution of connective tissue and on the vascularization of brain metastases (40 colon, lung and breast carcinoma samples) was analyzed. Most of the cases showed either a "pushing-type" (18/40, mostly colon and lung carcinomas) or a "papillary-type" (19/40, mostly breast carcinomas) growth pattern. There was a striking difference in the growth pattern and vascularization of colon/lung versus breast carcinoma metastases. Pushing-type brain metastases incorporated fewer vessels and accumulated more collagen in the adjacent brain parenchyma, whereas papillary-type brain metastases incorporated more vessels and accumulated collagen in the center of the tumor. We observed duplication of the PDGFRβ-positive pericyte layer accompanied by an increase in the amount of collagen within the vessel walls. The outer layer of pericytes and the collagen was removed from the vessel by invasive activity of the tumors, which occurred either peri- or intratumorally, depending on the growth pattern of the metastasis. Our findings suggest that pericytes are the main source of the connective tissue in brain metastases. Vascularization and connective tissue accumulation of the brain metastases largely depend on the growth pattern of the tumors.
Collapse
Affiliation(s)
- Vanda Téglási
- 1st Department of Pathology and Experimental Cancer Research
| | - Dániel T Csűry
- 1st Department of Pathology and Experimental Cancer Research
| | - Katalin Dezső
- 1st Department of Pathology and Experimental Cancer Research
| | - Edina Bugyik
- 1st Department of Pathology and Experimental Cancer Research
| | - Vanessza Szabó
- 1st Department of Pathology and Experimental Cancer Research
| | - Zoltán Szállási
- Brain Metastasis Research Group, Hungarian Academy of Sciences, 2nd Department of Pathology, Semmelweis University, Budapest, Hungary.,Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.,Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Sándor Paku
- 1st Department of Pathology and Experimental Cancer Research
| | - Lilla Reiniger
- 1st Department of Pathology and Experimental Cancer Research.,Brain Metastasis Research Group, Hungarian Academy of Sciences, 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
45
|
Ria R, Melaccio A, Racanelli V, Vacca A. Anti-VEGF Drugs in the Treatment of Multiple Myeloma Patients. J Clin Med 2020; 9:E1765. [PMID: 32517267 PMCID: PMC7355441 DOI: 10.3390/jcm9061765] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
The interaction between the bone marrow microenvironment and plasma cells plays an essential role in multiple myeloma progression and drug resistance. The vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) pathway in vascular endothelial cells activates and promotes angiogenesis. Moreover, VEGF activates and promotes vasculogenesis and vasculogenic mimicry when it interacts with VEGF receptors expressed in precursor cells and inflammatory cells, respectively. In myeloma bone marrow, VEGF and VEGF receptor expression are upregulated and hyperactive in the stromal and tumor cells. It has been demonstrated that several antiangiogenic agents can effectively target VEGF-related pathways in the preclinical phase. However, they are not successful in treating multiple myeloma, probably due to the vicarious action of other cytokines and signaling pathways. Thus, the simultaneous blocking of multiple cytokine pathways, including the VEGF/VEGFR pathway, may represent a valid strategy to treat multiple myeloma. This review aims to summarize recent advances in understanding the role of the VEGF/VEGFR pathway in multiple myeloma, and mainly focuses on the transcription pathway and on strategies that target this pathway.
Collapse
Affiliation(s)
- Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari “Aldo Moro” Medical School, 70124 Bari, Italy; (A.M.); (V.R.); (A.V.)
| | | | | | | |
Collapse
|
46
|
Zhao C, Zhao Z, Wang Z, Hu L, Wang H, Fang Z. Supervillin promotes tumor angiogenesis in liver cancer. Oncol Rep 2020; 44:674-684. [PMID: 32468064 PMCID: PMC7336518 DOI: 10.3892/or.2020.7621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/10/2020] [Indexed: 12/23/2022] Open
Abstract
Tumor angiogenesis is a hallmark of liver cancer and is necessary for tumor growth and progression. Supervillin (SVIL) is highly expressed and implicated in several malignant processes of liver cancer. However, the functional relationships between SVIL and tumor angiogenesis in liver cancer have not yet been fully elucidated. The present study was based on bioinformatics analysis, patient tissue sample detection, three-dimensional simulated blood vessel formation, a series of cytological experiments and mouse models. The results demonstrated the important role of SVIL in the progression of malignant liver cancer and tumor angiogenesis, both in terms of vasculogenic mimicry (VM) and endothelium-dependent vessel (EDV) development. SVIL knockdown inhibited VM formation and induced tumor cell apoptosis via the VEGF-p38 signaling axis and through various VM-associated transcriptional factors, including vascular endothelial-cadherin, matrix metalloproteinase 9/12 and migration-inducing protein 7. SVIL may therefore be considered a potential tumor vascular biomarker and a promising therapeutic target for patients with liver cancer.
Collapse
Affiliation(s)
- Chenggang Zhao
- Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China
| | - Zhiyang Zhao
- Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China
| | - Zhen Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China
| | - Lizhu Hu
- Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China
| | - Hongzhi Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China
| | - Zhiyou Fang
- Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China
| |
Collapse
|
47
|
Delgado‐Martín B, Medina MÁ. Advances in the Knowledge of the Molecular Biology of Glioblastoma and Its Impact in Patient Diagnosis, Stratification, and Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902971. [PMID: 32382477 PMCID: PMC7201267 DOI: 10.1002/advs.201902971] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/24/2020] [Indexed: 05/07/2023]
Abstract
Gliomas are the most common primary brain tumors in adults. They arise in the glial tissue and primarily occur in the brain. Low-grade tumors of World Health Organization (WHO) grade II tend to progress to high-grade gliomas of WHO grade III and, eventually, glioblastoma of WHO grade IV, which is the most common and deadly glioma, with a median survival of 12-15 months after final diagnosis. Knowledge of the molecular biology and genetics of glioblastoma has increased significantly in the past few years, giving rise to classification methods that can help in management and stratification of glioblastoma patients. However, glioblastoma remains an incurable disease. Glioblastoma cells have acquired genetic and metabolic adaptations in order to sustain tumor growth and progression, including changes in energetic metabolism, invasive capacity, migration, and angiogenesis, that make it very difficult to find suitable therapeutic targets and to develop effective drugs. The current standard of care for glioblastoma patients is surgery followed by radiotherapy plus concomitant and adjuvant chemotherapy with temozolomide. Although progress in glioblastoma therapies in recent years has been more limited than in other tumors, numerous drugs and targets are being proposed and many clinical trials are underway to develop effective subtype-specific treatments.
Collapse
Affiliation(s)
- Belén Delgado‐Martín
- Department of Molecular Biology and BiochemistryFaculty of SciencesCampus de Teatinos s/nUniversity of MálagaMálagaE‐29071Spain
| | - Miguel Ángel Medina
- Department of Molecular Biology and BiochemistryFaculty of SciencesCampus de Teatinos s/nUniversity of MálagaMálagaE‐29071Spain
- IBIMA (Biomedical Research Institute of Málaga)MálagaE‐29071Spain
- CIBER de Enfermedades Raras (CIBERER)MálagaE‐29071Spain
| |
Collapse
|
48
|
Mabeta P. Paradigms of vascularization in melanoma: Clinical significance and potential for therapeutic targeting. Biomed Pharmacother 2020; 127:110135. [PMID: 32334374 DOI: 10.1016/j.biopha.2020.110135] [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: 11/26/2019] [Revised: 03/16/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Melanoma is the most aggressive form of skin cancer. Malignant melanoma in particular has a poor prognosis and although treatment has improved, drug resistance continues to be a challenge. Angiogenesis, the formation of blood vessels from existing microvessels, precedes the progression of melanoma from a radial growth phase to a malignant phenotype. In addition, melanoma cells can form networks of vessel-like fluid conducting channels through vasculogenic mimicry (VM). Both angiogenesis and VM have been postulated to contribute to the development of resistance to treatment and to enable metastasis. Also, the metastatic spread of melanoma is highly dependent on lymphangiogenesis, the formation of lymphatic vessels from pre-existing vessels. Interestingly, the design and clinical testing of drugs that target VM and lymphangiogenesis lag behind that of angiogenesis inhibitors. Despite this, antiangiogenic drugs have not significantly improved the overall survival of melanoma patients, thus necessitating the targeting of alternative mechanisms. In this article, I review the roles of the three paradigms of tissue perfusion, namely, angiogenesis, VM and lymphangiogenesis, in promoting melanoma progression and metastasis. This article also explores the latest development and potential opportunities in the therapeutic targeting of these processes.
Collapse
Affiliation(s)
- Peace Mabeta
- Angiogenesis Laboratory, Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
49
|
O'Brien K, Saravanabavan S, Zhang JQJ, Wong ATY, Munt A, Burgess JS, Rangan GK. Regression of Peritubular Capillaries Coincides with Angiogenesis and Renal Cyst Growth in Experimental Polycystic Kidney Disease. Int J Nephrol Renovasc Dis 2020; 13:53-64. [PMID: 32280260 PMCID: PMC7132028 DOI: 10.2147/ijnrd.s238767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/06/2020] [Indexed: 12/12/2022] Open
Abstract
Background/Aim The natural history of the renal microvasculature changes in PKD is not known. The aim of this study was to test the hypothesis that angiogenesis is coupled with kidney cyst expansion, and the loss of peritubular capillary networks precedes the onset of interstitial fibrosis. Methods The renal microvasculature (RECA-1 and CD34) was evaluated in groups of Lewis polycystic kidney (LPK) rats and juvenile cystic kidney (jck) mice during the early, mid and late stage of disease. In addition, LPK rats and jck mice received sirolimus to determine if the reduction in renal cyst growth is in part mediated by the suppression of angiogenesis. Results In LPK rats, the loss of peritubular capillaries occurred in early-stage disease and paralleled cyst formation whereas in jck mice it was delayed to the mid stage. In both models, vasa recta were displaced by growing cysts and regressed in LPK rats with disease progression but lengthened in jck mice. Cortical and medullary capillary neoangiogenesis occurred during the early stage in both models and persisted with progression. Treatment with sirolimus reduced cyst enlargement but did not alter the progression of renal microvasculature changes in either model. Conclusion Regression of peritubular capillaries and disruption of vasa recta occur in parallel with angiogenesis and the progressive enlargement of kidney cysts. These data suggest that the regrowth of peritubular capillaries together with inhibition of angiogenesis are potential strategies to be considered in the treatment of PKD.
Collapse
Affiliation(s)
- Kristal O'Brien
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Sayanthooran Saravanabavan
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Jennifer Q J Zhang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Annette T Y Wong
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Alexandra Munt
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Jane S Burgess
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Gopala K Rangan
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| |
Collapse
|
50
|
Martí JM, Fernández-Cortés M, Serrano-Sáenz S, Zamudio-Martinez E, Delgado-Bellido D, Garcia-Diaz A, Oliver FJ. The Multifactorial Role of PARP-1 in Tumor Microenvironment. Cancers (Basel) 2020; 12:cancers12030739. [PMID: 32245040 PMCID: PMC7140056 DOI: 10.3390/cancers12030739] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/03/2020] [Accepted: 03/15/2020] [Indexed: 02/08/2023] Open
Abstract
Poly(ADP-ribose) polymerases (PARPs), represent a family of 17 proteins implicated in a variety of cell functions; some of them possess the enzymatic ability to synthesize and attach poly (ADP-ribose) (also known as PAR) to different protein substrates by a post-translational modification; PARPs are key components in the cellular response to stress with consequences for different physiological and pathological events, especially during neoplasia. In recent years, using PARP inhibitors as antitumor agents has raised new challenges in understanding their role in tumor biology. Notably, the function of PARPs and PAR in the dynamic of tumor microenvironment is only starting to be understood. In this review, we summarized the conclusions arising from recent studies on the interaction between PARPs, PAR and key features of tumor microenvironment such as hypoxia, autophagy, tumor initiating cells, angiogenesis and cancer-associated immune response.
Collapse
|