3,855 results on '"TUMOR ANGIOGENESIS"'
Search Results
552. Angiogenesis in human gliomas: Prognostic and therapeutic implications
- Author
-
Johnson, J. P., Bruce, J. N., Goldberg, Itzhak D., editor, and Rosen, Eliot M., editor
- Published
- 1997
- Full Text
- View/download PDF
553. Overexpression of Receptor Tyrosine Kinase EphB4 Triggers Tumor Growth and Hypoxia in A375 Melanoma Xenografts: Insights from Multitracer Small Animal Imaging Experiments
- Author
-
Christin Neuber, Birgit Belter, Sebastian Meister, Frank Hofheinz, Ralf Bergmann, Hans-Jürgen Pietzsch, and and Jens Pietzsch
- Subjects
Eph receptor tyrosine kinase family ,Ephrin ligands ,tumor microenvironment ,malignant melanoma ,small animal positron emission tomography ,tumor angiogenesis ,tumor hypoxia ,Organic chemistry ,QD241-441 - Abstract
Experimental evidence has associated receptor tyrosine kinase EphB4 with tumor angiogenesis also in malignant melanoma. Considering the limited in vivo data available, we have conducted a systematic multitracer and multimodal imaging investigation in EphB4-overexpressing and mock-transfected A375 melanoma xenografts. Tumor growth, perfusion, and hypoxia were investigated by positron emission tomography. Vascularization was investigated by fluorescence imaging in vivo and ex vivo. The approach was completed by magnetic resonance imaging, radioluminography ex vivo, and immunohistochemical staining for blood and lymph vessel markers. Results revealed EphB4 to be a positive regulator of A375 melanoma growth, but a negative regulator of tumor vascularization. Resulting in increased hypoxia, this physiological characteristic is considered as highly unfavorable for melanoma prognosis and therapy outcome. Lymphangiogenesis, by contrast, was not influenced by EphB4 overexpression. In order to distinguish between EphB4 forward and EphrinB2, the natural EphB4 ligand, reverse signaling a specific EphB4 kinase inhibitor was applied. Blocking experiments show EphrinB2 reverse signaling rather than EphB4 forward signaling to be responsible for the observed effects. In conclusion, functional expression of EphB4 is considered a promising differentiating characteristic, preferentially determined by non-invasive in vivo imaging, which may improve personalized theranostics of malignant melanoma.
- Published
- 2018
- Full Text
- View/download PDF
554. Significance of anti-angiogenic therapy in head and neck cancer—Heterogeneity of tumor endothelium
- Author
-
Kyoko Hida, Noritaka Ohga, Yasuhiro Hida, and Masanobu Shindoh
- Subjects
Tumor angiogenesis ,Endothelium ,Anti-angiogenic therapy ,Dentistry ,RK1-715 - Abstract
Tumor angiogenesis is necessary for solid tumor progression and metastasis. Thus, targeting tumor blood vessels is an important strategy for cancer therapy. Especially, it would give large benefit to head and neck cancer patients if ideal anti-angiogenic drug is developed. Tumor blood vessels have been shown to differ from their normal counterparts, for example, by changes in morphology. An important concept in tumor angiogenesis is that tumor endothelial cells are assumed to be genetically normal, even though these endothelial cells are structurally and functionally abnormal. To date, many anti-angiogenic drugs have been developed, but it has been also reported to cause toxic side effects. To develop ideal anti-angiogenic therapies, understanding tumor endothelial cell abnormalities is important. We have isolated tumor endothelial cells from mouse tumor xenografts and have shown that tumor endothelial cells are abnormal. Tumor endothelial cells upregulate many genes, such as epidermal growth factor receptor. Tumor endothelial cells are also more sensitive to EGF. Unexpectedly, tumor endothelial cells were cytogenetically abnormal. In marked contrast, freshly isolated normal endothelial cells were diploid. We conclude that tumor endothelial cells can acquire cytogenetic abnormalities while in the tumor microenvironment. Here, we provide an overview of the current studies on tumor endothelial cell abnormalities.
- Published
- 2010
- Full Text
- View/download PDF
555. Angiogenesis in breast cancer
- Author
-
Weidner, Noel, Freireich, Emil J., editor, Dickson, Robert B., editor, and Lippman, Marc E., editor
- Published
- 1996
- Full Text
- View/download PDF
556. Regulation of Tumor Angiogenesis by Organ-Specific Cytokines
- Author
-
Singh, R. K., Fidler, I. J., Compans, R. W., editor, Cooper, M., editor, Koprowski, H., editor, Melchers, F., editor, Oldstone, M., editor, Olsnes, S., editor, Potter, M., editor, Saedler, H., editor, Vogt, P. K., editor, Wagner, H., editor, Günthert, Ursula, editor, and Birchmeier, Walter, editor
- Published
- 1996
- Full Text
- View/download PDF
557. Oxidative stress in tumor microenvironment—Its role in angiogenesis
- Author
-
Armando ROJAS, Raúl SILVA, Héctor FIGUEROA, and Miguel A MORALES
- Subjects
Oxidative stress ,Tumor angiogenesis ,NOX enzymes ,Nitric oxide ,VEGF ,Cadherins ,Advanced glycation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The tumor angiogenesis process is believed to be dependent on an "angiogenic switch" formed by a cascade of biologic events as a consequence of the "cross-talk" between tumor cells and several components of local microenvironment including endothelial cells, macrophages, mast cells and stromal components. Oxidative stress represents an important stimulus that widely contributes to this angiogenic switch, which is particularly relevant in lungs, where oxidative stress is originated from different sources including the incomplete reduction of oxygen during respiration, exposure to hypoxia/reoxygenation, stimulated resident or chemoattracted immune cells to lung tissues, as well as by a variety of chemicals compounds. In the present review we highlight the role of oxidative stress in tumor angiogenesis as a key signal linked to other relevant actors in this complex process.
- Published
- 2008
558. Tumor-Derived Extracellular Vesicles Induce Abnormal Angiogenesis via TRPV4 Downregulation and Subsequent Activation of YAP and VEGFR2
- Author
-
Brianna Guarino, Venkatesh Katari, Ravi Adapala, Neha Bhavnani, Julie Dougherty, Mahmood Khan, Sailaja Paruchuri, and Charles Thodeti
- Subjects
Histology ,vascular endothelial growth factor receptor 2 ,Biomedical Engineering ,Bioengineering ,tumor angiogenesis ,extracellular vesicles ,transient receptor potential vanilloid 4 ,endothelial cells ,TP248.13-248.65 ,Biotechnology - Abstract
Tumor angiogenesis is initiated and maintained by the tumor microenvironment through secretion of autocrine and paracrine factors, including extracellular vesicles (EVs). Although tumor-derived EVs (t-EVs) have been implicated in tumor angiogenesis, growth and metastasis, most studies on t-EVs are focused on proangiogenic miRNAs and growth factors. We have recently demonstrated that conditioned media from human lung tumor cells (A549) downregulate TRPV4 channels and transform normal endothelial cells to a tumor endothelial cell-like phenotype and induce abnormal angiogenesis in vitro, via t-EVs. However, the underlying molecular mechanism of t-EVs on endothelial cell phenotypic transition and abnormal angiogenesis in vivo remains unknown. Here, we demonstrate that t-EVs downregulate TRPV4 expression post-translationally and induce abnormal angiogenesis by activating Rho/Rho kinase/YAP/VEGFR2 pathways. Further, we demonstrate that t-EVs induce abnormal vessel formation in subcutaneously implanted Matrigel plugs in vivo (independent of tumors), which are characterized by increased VEGFR2 expression and reduced pericyte coverage. Taken together, our findings demonstrate that t-EVs induce abnormal angiogenesis via TRPV4 downregulation-mediated activation of Rho/Rho kinase/YAP/VEGFR2 pathways and suggest t-EVs and TRPV4 as novel targets for vascular normalization and cancer therapy.
- Published
- 2021
- Full Text
- View/download PDF
559. Donafenib-Loaded Callispheres Beads Embolization in a VX2 Liver Tumor: Investigating Efficacy, Safety, and Improvement of Tumor Angiogenesis After Embolization
- Author
-
Songjiang Huang, Chen Zhou, Tongqiang Li, Qin Shi, Chongtu Yang, Shuguang Ju, Chaoyang Wang, Jiacheng Liu, Bin Xiong, Yaowei Bai, Yingliang Wang, and Yang Chen
- Subjects
safety ,Tumor angiogenesis ,Liver tumor ,business.industry ,medicine.medical_treatment ,efficacy ,hepatocellular carcinoma ,tumor angiogenesis ,medicine.disease ,donafenib ,medicine ,Cancer research ,Embolization ,business ,Journal of Hepatocellular Carcinoma ,Original Research ,transcatheter arterial chemoembolization - Abstract
Tongqiang Li,1,2,* Qin Shi,1,2,* Jiacheng Liu,1,2,* Yingliang Wang,1,2 Chen Zhou,1,2 Chaoyang Wang,1,2 Shuguang Ju,1,2 Songjiang Huang,1,2 Chongtu Yang,1,2 Yang Chen,1,2 Yaowei Bai,1,2 Bin Xiong1,2 1Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Peopleâs Republic of China; 2Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, Peopleâs Republic of China*These authors contributed equally to this workCorrespondence: Bin XiongDepartment of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue #1277, Wuhan, 430022, Peopleâs Republic of ChinaEmail herr_xiong@126.comObjective: To investigate the efficiency and safety of callispheres beads loaded with donafenib (DCBs) for embolization in a VX2 liver tumor, as well as the improvement of tumor angiogenesis following embolization.Methods: Forty New Zealand white rabbit VX2 liver tumors were treated with four different drugs via the hepatic artery: NS (normal saline), CB (blank callispheres beads), ACB (adriamycin-loaded callispheres beads) and DCB (DCBs). Hematoxylin-eosin staining was performed to assess tumor necrosis, while MRI was employed to detect the changes in tumor size. The safety was evaluated by the liver and kidney function parameters, and the immunofluorescence and immunohistochemical staining were performed to reflect the tumor hypoxia and tumor angiogenesis following embolization.Results: The DCB group had the smallest tumor growth rate, but the tumor necrosis rate was the highest of the four groups. Compared to the CB and ACB groups, the DCB group did not aggravate the liver damage and had no influence on kidney function. The staining results showed that, although the tumor hypoxia deteriorated after DCBs embolization, the expression of VEGF (vascular endothelial growth factor) reduced, thus inhibiting tumor angiogenesis.Conclusion: DCB administration via hepatic artery is an effective and safe treatment for a preclinical liver cancer model, with the unique benefit of suppressing tumor angiogenesis following embolization.Keywords: hepatocellular carcinoma, donafenib, transcatheter arterial chemoembolization, tumor angiogenesis, efficacy, safety
- Published
- 2021
560. Neutrophil and Natural Killer Cell Interactions in Cancers: Dangerous Liaisons Instructing Immunosuppression and Angiogenesis
- Author
-
Maria Teresa Palano, Matteo Gallazzi, Martina Cucchiara, Andrea De Lerma Barbaro, Daniela Gallo, Barbara Bassani, Antonino Bruno, and Lorenzo Mortara
- Subjects
Pharmacology ,natural killer cells ,immunosuppression ,Immunology ,Review ,tumor angiogenesis ,Immunosuppression ,Natural killer cells ,Neutrophil-NK cell crosstalk ,Neutrophils ,Tumor angiogenesis ,Tumor microenvironment ,Infectious Diseases ,neutrophils ,Drug Discovery ,tumor microenvironment ,Medicine ,Pharmacology (medical) ,neutrophil-NK cell crosstalk - Abstract
The tumor immune microenvironment (TIME) has largely been reported to cooperate on tumor onset and progression, as a consequence of the phenotype/functional plasticity and adaptation capabilities of tumor-infiltrating and tumor-associated immune cells. Immune cells within the tumor micro (tissue-local) and macro (peripheral blood) environment closely interact by cell-to-cell contact and/or via soluble factors, also generating a tumor-permissive soil. These dangerous liaisons have been investigated for pillars of tumor immunology, such as tumor associated macrophages and T cell subsets. Here, we reviewed and discussed the contribution of selected innate immunity effector cells, namely neutrophils and natural killer cells, as “soloists” or by their “dangerous liaisons”, in favoring tumor progression by dissecting the cellular and molecular mechanisms involved.
- Published
- 2021
561. New Insights Into the Regulatory Roles of Extracellular Vesicles in Tumor Angiogenesis and Their Clinical Implications
- Author
-
Maohua Huang, Yuhe Lei, Yinqin Zhong, Chiwing Chung, Mei Wang, Min Hu, and Lijuan Deng
- Subjects
Cell and Developmental Biology ,QH301-705.5 ,Mini Review ,miRNAs ,lncRNAs ,Cell Biology ,tumor angiogenesis ,Biology (General) ,extracellular vesicles ,CircRNAs ,proteins ,Developmental Biology - Abstract
Angiogenesis is required for tumor growth and development. Extracellular vesicles (EVs) are important signaling entities that mediate communication between diverse types of cells and regulate various cell biological processes, including angiogenesis. Recently, emerging evidence has suggested that tumor-derived EVs play essential roles in tumor progression by regulating angiogenesis. Thousands of molecules are carried by EVs, and the two major types of biomolecules, noncoding RNAs (ncRNAs) and proteins, are transported between cells and regulate physiological and pathological functions in recipient cells. Understanding the regulation of EVs and their cargoes in tumor angiogenesis has become increasingly important. In this review, we summarize the effects of tumor-derived EVs and their cargoes, especially ncRNAs and proteins, on tumor angiogenesis and their mechanisms, and we highlight the clinical implications of EVs in bodily fluids as biomarkers and as diagnostic, prognostic, and therapeutic targets in cancer patients.
- Published
- 2021
- Full Text
- View/download PDF
562. A Comprehensive Review on the Pleotropic Role of Nitric Oxide, a Well-Known Cytotoxin
- Author
-
Ankit Sharma, Michael Babich, Estevan Limón López, and James A. Radosevich
- Subjects
Tumor angiogenesis ,Cancer ,Human physiology ,Biology ,medicine.disease ,Nitric oxide ,chemistry.chemical_compound ,Histone ,chemistry ,microRNA ,DNA methylation ,Cancer research ,medicine ,biology.protein ,Epigenetics - Abstract
Free radical nitric oxide (NO\(\bullet\)) plays a dual role in human physiology and pathophysiology. At low levels NO\(\bullet\) is considered to play a protective role for cells; however, at higher levels it becomes cytotoxic and contributes to tumor angiogenesis and cancer progression. Although earlier studies investigating the role of NO\(\bullet\) in cancer have been tissue-specific, this review focuses on the underlying similarities of NO\(\bullet\) exposure that present across a variety of site-specific cancers. We also try to explain the role of NO\(\bullet\)and the cellular protecting enzyme glutathione-transferase. This article emphasizes the emerging role of NO\(\bullet\)in three main areas related to epigenetics: DNA methylation, microRNAs, and histone modifications. In conclusion, we describe the recent advancement of a model cell line system in which cells demonstrate a high level of NO\(\bullet\) nitric oxide adaptation, and propose that these adapted cell lines can act as useful tools to study the role of NO\(\bullet\) in cancer biology.
- Published
- 2021
- Full Text
- View/download PDF
563. Real-Time Longitudinal Evaluation of Tumor Blood Vessels Using a Compact Preclinical Fluorescence Imaging System
- Author
-
Seo-Young Kim, Hoibin Jeong, Yujung Kang, Su-Hyeon Cho, Kil-Nam Kim, Huisu Kim, and Song-Rae Kim
- Subjects
Tumor angiogenesis ,Fluorescence-lifetime imaging microscopy ,indocyanine green ,Angiogenesis ,Clinical Biochemistry ,Angiogenesis Inhibitors ,Article ,Metastasis ,Mice ,optical imaging ,chemistry.chemical_compound ,preclinical study ,Neoplasms ,medicine ,Animals ,blood flow ,tumor angiogenesis ,Neovascularization, Pathologic ,business.industry ,General Medicine ,Blood flow ,medicine.disease ,medicine.anatomical_structure ,Pharmaceutical Preparations ,chemistry ,Tumor progression ,business ,Indocyanine green ,TP248.13-248.65 ,Biotechnology ,Biomedical engineering ,Blood vessel - Abstract
Tumor angiogenesis is enhanced in all types of tumors to supply oxygen and nutrients for their growth and metastasis. With the development of anti-angiogenic drugs, the importance of technology that closely monitors tumor angiogenesis has also been emerging. However, to date, the technology for observing blood vessels requires specialized skills with expensive equipment, thereby limiting its applicability only to the laboratory setting. Here, we used a preclinical optical imaging system for small animals and, for the first time, observed, in real time, the entire process of blood vessel development in tumor-bearing mice injected with indocyanine green. Time-lapse sequential imaging revealed blood vessel volume and blood flow dynamics on a microscopic scale. Upon analyzing fluorescence dynamics at each stage of tumor progression, vessel volume and blood flow were found to increase as the tumor developed. Conversely, these vascular parameters decreased when the mice were treated with angiogenesis inhibitors, which suggests that the effects of drugs targeting angiogenesis can be rapidly and easily screened. The results of this study may help evaluate the efficacy of angiogenesis-targeting drugs by facilitating the observation of tumor blood vessels easily in a laboratory unit without large and complex equipment.
- Published
- 2021
- Full Text
- View/download PDF
564. Role of STK33 in the tumor angiogenesis program supported by HSP90 chaperone
- Author
-
Liu, Yang, Seufferlein, Thomas, and Brunner, Cornelia
- Subjects
Hypoxie ,Protein-serine-threonine kinases ,Angiogenesis inhibitors ,Therapeutic use ,Tumor hypoxia ,HIF-1�� ,Vascular endothelial growth factor A ,HIF-1α ,Tumorwachstum ,ddc:610 ,DDC 610 / Medicine & health ,Angiogenese ,Tumor angiogenesis - Abstract
Lately, the HSP90 client serine/threonine kinase STK33 emerged to be required by cancer cells for their viability and proliferation. However, its mechanistic contribution to carcinogenesis is not clearly understood. Here we report that elevated STK33 expression correlates with advanced stages of human pancreatic and colorectal carcinomas. Impaired proliferation and augmented apoptosis associated with genetic abrogation of STK33 were paralleled by decreased vascularization in tumor xenografts. In line with this, ectopic STK33 not only promoted tumor growth after pharmacologic inhibition of HSP90 using structurally divergent small molecules currently in clinical development, but also restored blood vessel formation in vivo. Mechanistic studies demonstrated that HSP90-stabilized STK33 interacts with and regulates hypoxia-driven accumulation and activation of HIF-1�� as well as secretion of VEGF-A in hypoxic cancer cells. In addition, our study reveals a putative cooperation between STK33 and other HSP90 client protein kinases involved in molecular and cellular events through which cancer cells ensure their survival by securing the oxygen and nutrient supply. Altogether, our findings indicate that STK33 interferes with signals from hypoxia and HSP90 to promote tumor angiogenesis and tumor growth.
- Published
- 2021
565. Estrogens Regulate Placental Angiogenesis in Horses
- Author
-
Alejandro Esteller-Vico, Peter Daels, Mats H.T. Troedsson, Edward L. Squires, Pouya Dini, Shingo Haneda, Barry A. Ball, Kirsten E. Scoggin, and Yasuo Nambo
- Subjects
Vascular Endothelial Growth Factor A ,Angiogenesis ,letrozole ,MARES ,Androgen ,chemistry.chemical_compound ,angiogenesis ,Receptors ,estrogen ,Developmental ,Testosterone ,Biology (General) ,Aromatase ,First ,ENZYME-IMMUNOASSAY ,Spectroscopy ,equine ,biology ,PROLIFERATION ,placental development ,Gene Expression Regulation, Developmental ,Steroid 17-alpha-Hydroxylase ,General Medicine ,MEDIATED ANGIOGENESIS ,17 beta-estradiol ,Computer Science Applications ,Chemistry ,medicine.anatomical_structure ,Receptors, Androgen ,Maternal-Fetal Relations ,embryonic structures ,GROWTH ,Female ,Pregnancy Trimester ,pregnancy ,medicine.medical_specialty ,placenta ,QH301-705.5 ,medicine.drug_class ,CORPUS-LUTEUM ,Neovascularization, Physiologic ,Catalysis ,Article ,TUMOR ANGIOGENESIS ,Inorganic Chemistry ,FACTOR EXPRESSION ,chorioallantois ,Vasculogenesis ,Estrone sulfate ,Internal medicine ,Placenta ,medicine ,Angiopoietin-1 ,Animals ,Horses ,Veterinary Sciences ,Physical and Theoretical Chemistry ,Physiologic ,QD1-999 ,Molecular Biology ,Neovascularization ,Fetus ,Organic Chemistry ,Androstenedione ,17β-estradiol ,Estrogens ,Pregnancy Trimester, First ,Endocrinology ,Gene Expression Regulation ,chemistry ,Estrogen ,biology.protein ,EARLY-PREGNANCY ,Hormone ,ESTRADIOL INCREASES VEGF - Abstract
A sufficient vascular network within the feto-maternal interface is necessary for placental function. Several pregnancy abnormalities have been associated with abnormal vascular formations in the placenta. We hypothesized that growth and expansion of the placental vascular network in the equine (Equus caballus) placenta is regulated by estrogens (estrogen family hormones), a hormone with a high circulating concentration during equine gestation. Administration of letrozole, a potent and specific inhibitor of aromatase, during the first trimester (D30 to D118), decreased circulatory estrone sulfate concentrations, increased circulatory testosterone and androstenedione concentrations, and tended to reduce the weight of the fetus (p <, 0.1). Moreover, the gene expression of CYP17A1 was increased, and the expression of androgen receptor was decreased in the D120 chorioallantois (CA) of letrozole-treated mares in comparison to that of the control mares. We also found that at D120, the number of vessels tended to decrease in the CAs with letrozole treatment (p = 0.07). In addition, expression of a subset of angiogenic genes, such as ANGPT1, VEGF, and NOS2, were altered in the CAs of letrozole-treated mares. We further demonstrated that 17β-estradiol increases the expression of ANGPT1 and VEGF and increases the angiogenic activity of equine endothelial cells in vitro. Our results from the estrogen-suppressed group demonstrated an impaired placental vascular network, suggesting an estrogen-dependent vasculogenesis in the equine CA during the first trimester.
- Published
- 2021
- Full Text
- View/download PDF
566. Diagnostic value of various vascular features of breast cancer by age
- Author
-
Wenqiang Lin, Liang Xu, Jia Lin, and Teng Lin
- Subjects
Tumor angiogenesis ,medicine.medical_specialty ,Diagnostic methods ,Physiology ,Breast imaging ,Breast Neoplasms ,Sensitivity and Specificity ,Metastasis ,Diagnosis, Differential ,Breast cancer ,Age groups ,Physiology (medical) ,Medicine ,Humans ,Breast ,Aged ,Ultrasonography ,business.industry ,Ultrasound ,Hematology ,Flow pattern ,medicine.disease ,Microvessels ,Female ,Radiology ,Ultrasonography, Mammary ,Cardiology and Cardiovascular Medicine ,business - Abstract
BACKGROUND: Tumor angiogenesis plays a critical role in the growth and metastasis of breast cancer. Evaluating the added value of vascular features to Breast Imaging Reporting and Data System (BI-RADS) in differentiating malignant masses from benign ones is essential. Micro-flow Imaging (MFI) is a promising noninvasive diagnostic method for the microvessels in breast tumors, but its precise value is still uncertain. OBJECTIVES: Understanding whether malignant tumor vascular characteristics by MFI are associated with breast cancer and whether the diagnostic efficiency varies by age. MATERIALS AND METHODS: B-mode ultrasound and MFI were performed for 153 solid breast lesions. The vessels images by MFI were reviewed and assessed by two investigators, respectively. Then the diagnostic efficacy of different vascular features combined with BI-RADS was evaluated in different age groups. RESULTS: The mean size of lesions is 19.4 (range 18–78) mm. There were 94 breast masses in benign, while 59 breast masses in malignant by pathology. III Adler classification, penetrating vessels, and complex flow pattern showed a positive association with a high risk of malignant breast lesions (p
- Published
- 2021
567. Strategies for Remodeling the Tumor Microenvironment Using Active Ingredients of Ginseng-A Promising Approach for Cancer Therapy
- Author
-
Mo Li, Xin Wang, Ying Wang, Shunchao Bao, Qing Chang, Linlin Liu, Shuai Zhang, and Liwei Sun
- Subjects
Pharmacology ,tumor stem cell ,tumor microenvironment ,cancer therapy ,food and beverages ,Pharmacology (medical) ,Review ,tumor angiogenesis ,Therapeutics. Pharmacology ,RM1-950 ,complex mixtures ,immune response ,Panax ginseng (C.A. Meyer) - Abstract
The tumor microenvironment (TME) plays a key role in promoting the initiation and progression of tumors, leading to chemoradiotherapy resistance and immunotherapy failure. Targeting of the TME is a novel anti-tumor therapeutic approach and is currently a focus of anti-tumor research. Panax ginseng C. A. Meyer (ginseng), an ingredient of well-known traditional Asia medicines, exerts beneficial anti-tumor effects and can regulate the TME. Here, we present a systematic review that describes the current status of research efforts to elucidate the functions and mechanisms of ginseng active components (including ginsenosides and ginseng polysaccharides) for achieving TME regulation. Ginsenosides have variety effects on TME, such as Rg3, Rd and Rk3 can inhibit tumor angiogenesis; Rg3, Rh2 and M4 can regulate the function of immune cells; Rg3, Rd and Rg5 can restrain the stemness of cancer stem cells. Ginseng polysaccharides (such as red ginseng acidic polysaccharides and polysaccharides extracted from ginseng berry and ginseng leaves) can regulate TME mainly by stimulating immune cells. In addition, we propose a potential mechanistic link between ginseng-associated restoration of gut microbiota and the tumor immune microenvironment. Finally, we describe recent advances for improving ginseng efficacy, including the development of a nano-drug delivery system. Taken together, this review provides novel perspectives on potential applications for ginseng active ingredients as anti-cancer adjuvants that achieve anti-cancer effects by reshaping the tumor microenvironment.
- Published
- 2021
568. Epstein-Barr Virus Promotes Tumor Angiogenesis by Activating STIM1-Dependent Ca2+ Signaling in Nasopharyngeal Carcinoma
- Author
-
Ting Que, Xiaojian Zhang, Jiazhang Wei, Jiaxiang Ye, Jinyan Zhang, Fei Liu, Yayan Deng, Xiaoling Luo, and Yue Luo
- Subjects
Microbiology (medical) ,CD31 ,inorganic chemicals ,Angiogenesis ,chemistry.chemical_compound ,Epidermal growth factor ,medicine ,otorhinolaryngologic diseases ,Immunology and Allergy ,Epstein-Barr virus ,Molecular Biology ,Tube formation ,Gene knockdown ,General Immunology and Microbiology ,nasopharyngeal carcinoma ,tumor angiogenesis ,medicine.disease ,Vascular endothelial growth factor ,stomatognathic diseases ,Infectious Diseases ,chemistry ,Nasopharyngeal carcinoma ,Cell culture ,Cancer research ,Medicine ,stromal interaction molecule 1 - Abstract
Epstein-Barr virus (EBV) promotes tumor angiogenesis in nasopharyngeal carcinoma (NPC) by activating store-operated Ca2+ entry. Since such entry has been linked to stromal interaction molecule 1 (STIM1), we examined whether the virus acts via STIM1-dependent Ca2+ signaling to promote tumor angiogenesis in NPC. STIM1 expression was detected in NPC cell lines HK1 and CNE2 that were negative or positive for EBV. STIM1 was knocked down in EBV-positive cells using recombinant lentivirus, then cytosolic Ca2+ levels were measured based on fluorescence resonance energy transfer. Cells were also exposed to epidermal growth factor (EGF), and secretion of vascular endothelial growth factor (VEGF) was measured using an enzyme-linked immunosorbent assay. Endothelial tube formation was quantified in an in vitro angiogenesis assay. Growth of CNE2-EBV xenografts was measured in mice, and angiogenesis was assessed based on immunohistochemical staining against CD31. Paraffin-embedded NPC tissues from patients were assayed for CD31 and STIM1. EGFR and ERK signaling pathways were assessed in NPC cell lines. STIM1 expression was higher in EBV-positive than in EBV-negative NPC cell lines. STIM1 knockdown in EBV-positive NPC cells significantly reduced Ca2+ influx and VEGF production after EGF treatment. STIM1 knockdown also inhibited xenograft growth and angiogenesis. Moreover, CD31 expression level was higher in EBV-positive than EBV-negative NPC tissues, and high expression of CD31 co-localized with high expression of STIM1 in EBV-positive tissues from NPC patients. Viral infection of NPC cells led to higher levels of phosphorylated ERK1/2 after EGF treatment, which STIM1 knockdown partially reversed. Our results suggest that EBV promotes EGF-induced ERK1/2 signaling by activating STIM1-dependent Ca2+ signaling, and that blocking such signaling may inhibit EBV-promoted angiogenesis in NPC.
- Published
- 2021
- Full Text
- View/download PDF
569. Expression of CCRL2 Inhibits Tumor Growth by Concentrating Chemerin and Inhibiting Neoangiogenesis
- Author
-
Valérie Wittamer, Ingrid Dubois-Vedrenne, Annalisa Del Prete, Diana Al Delbany, Silvano Sozzani, Anne Lefort, Marc Parmentier, Ayoub Radi, Frédérick Libert, Maxime Vernimmen, and Virginie Robert
- Subjects
Cancer Research ,DMBA ,CMKLR1 ,medicine.disease_cause ,Article ,GPR1 ,ChemR23 ,Chemokine receptor ,medicine ,Chemerin ,Rarres2 ,Tumor angiogenesis ,Receptor ,RC254-282 ,biology ,Chemistry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,tumor angiogenesis ,Sciences bio-médicales et agricoles ,Phenotype ,Oncology ,biology.protein ,Cancer research ,Carcinogenesis ,chemerin - Abstract
CCRL2 belongs to the G protein-coupled receptor family and is one of the three chemerin receptors. It is considered as a non-signaling receptor, presenting chemerin to cells expressing the functional chemerin receptor ChemR23/CMKLR1 and possibly GPR1. In the present work, we investigate the role played by CCRL2 in mouse cancer models. Loss of function of Ccrl2 accelerated the development of papillomas in a chemical model of skin carcinogenesis (DMBA/TPA), whereas the growth of B16 and LLC tumor cell grafts was delayed. Delayed tumor growth was also observed when B16 and LLC cells overexpress CCRL2, while knockout of Ccrl2 in tumor cells reversed the consequences of Ccrl2 knockout in the host. The phenotypes associated with CCRL2 gain or loss of function were largely abrogated by knocking out the chemerin or Cmklr1 genes. Cells harboring CCRL2 could concentrate bioactive chemerin and promote the activation of CMKLR1-expressing cells. A reduction of neoangiogenesis was observed in tumor grafts expressing CCRL2, mimicking the phenotype of chemerin-expressing tumors. This study demonstrates that CCRL2 shares functional similarities with the family of atypical chemokine receptors (ACKRs). Its expression by tumor cells can significantly tune the effects of the chemerin/CMKLR1 system and act as a negative regulator of tumorigenesis., info:eu-repo/semantics/published
- Published
- 2021
570. Towards integration of time-resolved confocal microscopy of a 3D in vitro microfluidic platform with a hybrid multiscale model of tumor angiogenesis
- Author
-
Lima Eabf, Thomas E. Yankeelov, Angela M. Jarrett, Marissa Nichole Rylander, Caleb Phillips, and Manasa Gadde
- Subjects
Tumor angiogenesis ,Ecology ,Chemistry ,Dynamics (mechanics) ,Microfluidics ,In vitro ,law.invention ,Endothelial stem cell ,Vascular endothelial growth factor ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,medicine.anatomical_structure ,Computational Theory and Mathematics ,Confocal microscopy ,law ,Modeling and Simulation ,Genetics ,medicine ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Blood vessel ,Biomedical engineering - Abstract
The goal of this study is to calibrate a multiscale model of tumor angiogenesis with time-resolved data to allow for systematic testing of mathematical predictions of vascular sprouting. The multi-scale model consists of an agent-based description of tumor and endothelial cell dynamics coupled to a continuum model of vascular endothelial growth factor concentration. First, we calibrate ordinary differential equation models to time-resolved protein expression data to estimate the rates of secretion and consumption of vascular endothelial growth factor by endothelial and tumor cells, respectively. These parameters are then input into the multiscale tumor angiogenesis model, and the remaining model parameters are then calibrated to time resolved confocal microscopy images obtained within a 3D vascularized microfluidic platform. The microfluidic platform mimics a functional blood vessel with a surrounding collagen matrix seeded with inflammatory breast cancer cells, which induce tumor angiogenesis. Once the multi-scale model is fully parameterized, we forecast the spatiotemporal distribution of vascular sprouts at future time points and directly compare the predictions to experimentally measured data. We assess the ability of our model to globally recapitulate angiogenic vasculature density, resulting in an average relative calibration error of 17.7% ± 6.3% and an average prediction error of 20.2% ± 4% and 21.7% ± 3.6% using one and four calibrated parameters, respectively. We then assess the model’s ability to predict local vessel morphology (individualized vessel structure as opposed to global vascular density), initialized with the first time point and calibrated with two intermediate time points. To the best of our knowledge, this represents the first study to integrate well-controlled, experimental data into a mechanism-based, multiscale, mathematical model of angiogenic sprouting to make specific, testable predictions.
- Published
- 2021
- Full Text
- View/download PDF
571. The Dual Effect of the BMP9–ALK1 Pathway in Blood Vessels: An Opportunity for Cancer Therapy Improvement?
- Author
-
Blanca Ayuso-Íñigo, Lucía Méndez-García, Miguel Pericacho, and José M. Muñoz-Félix
- Subjects
Cancer Research ,Tumor hypoxia ,Kinase ,business.industry ,Angiogenesis ,medicine.medical_treatment ,bone morphogenetic protein 9 ,High endothelial venules ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunosuppression ,Review ,tumor angiogenesis ,ALK1 ,Oncology ,Blood vessel maturation ,medicine ,Cancer research ,cancer therapy ,business ,Adverse effect ,Receptor ,RC254-282 - Abstract
Simple Summary The modulation of tumor blood vessels is a great opportunity for improving cancer therapies. Understanding the cellular and molecular players that regulate the biology of tumor blood vessels and tumor angiogenesis is necessary for the development of new anti-tumor strategies. Bone morphogenetic protein 9 (BMP9) is a circulating factor with multiple effects in vascular biology through its receptor activin receptor-like kinase 1 (ALK1). In this review, we give an overview of the possible benefits of modulating BMP9–ALK1 functions for cancer therapy improvement. Abstract The improvement of cancer therapy efficacy, the extension of patient survival and the reduction of adverse side effects are major challenges in cancer research. Targeting blood vessels has been considered a promising strategy in cancer therapy. Since the tumor vasculature is disorganized, leaky and triggers immunosuppression and tumor hypoxia, several strategies have been studied to modify tumor vasculature for cancer therapy improvement. Anti-angiogenesis was first described as a mechanism to prevent the formation of new blood vessels and prevent the oxygen supply to tumor cells, showing numerous limitations. Vascular normalization using low doses of anti-angiogenic drugs was purposed to overcome the limitations of anti-angiogenic therapies. Other strategies such as vascular promotion or the induction of high endothelial venules are being studied now to improve cancer therapy. Bone morphogenetic protein 9 (BMP9) exerts a dual effect through the activin receptor-like kinase 1 (ALK1) receptor in blood vessel maturation or activation phase of angiogenesis. Thus, it is an interesting pathway to target in combination with chemotherapies or immunotherapies. This review manuscript explores the effect of the BMP9–ALK1 pathway in tumor angiogenesis and the possible usefulness of targeting this pathway in anti-angiogenesis, vascular normalization or vascular promotion therapies.
- Published
- 2021
572. Mechanical Aspects of Angiogenesis
- Author
-
Daniel Rüdiger, Stefan Zahler, and Maibritt Kretschmer
- Subjects
Tumor angiogenesis ,Cancer Research ,mechanical cues ,ECM ,cell migration ,Angiogenesis ,Chemistry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,vessel stabilization ,Cell migration ,Context (language use) ,Adhesion ,Review ,tumor angiogenesis ,endothelial cells ,cancer treatment ,Cell biology ,Extracellular matrix ,Endothelial stem cell ,angiogenesis ,stiffness ,Oncology ,Tumor growth ,RC254-282 ,tip/stalk cells - Abstract
Simple Summary The formation of new blood vessels from already existing ones is a process of high clinical relevance, since it is of great importance for both physiological and pathological processes. In regard to tumors, the process is crucial, since it ensures the supply with nutrients and the growth of the tumor. The influence of mechanical factors on this biological process is an emerging field. Until now, the shear force of the blood flow has been considered the main mechanical parameter during angiogenesis. This review article provides an overview of further mechanical cues, with particular focus on the surrounding extracellular matrix impacting the cell behavior and, thus, regulating angiogenesis. This underlines the enormous importance of the mechanical properties of the extracellular matrix on cell biological processes and shows how changing the mechanics of the extracellular matrix could be used as a possible therapeutic approach in cancer therapy. Abstract Angiogenesis is of high clinical relevance as it plays a crucial role in physiological (e.g., tissue regeneration) and pathological processes (e.g., tumor growth). Besides chemical signals, such as VEGF, the relationship between cells and the extracellular matrix (ECM) can influence endothelial cell behavior during angiogenesis. Previously, in terms of the connection between angiogenesis and mechanical factors, researchers have focused on shear forces due to blood flow. However, it is becoming increasingly important to include the direct influence of the ECM on biological processes, such as angiogenesis. In this context, we focus on the stiffness of the surrounding ECM and the adhesion of cells to the ECM. Furthermore, we highlight the mechanical cues during the main stages of angiogenesis: cell migration, tip and stalk cells, and vessel stabilization. It becomes clear that the different stages of angiogenesis require various chemical and mechanical cues to be modulated by/modulate the stiffness of the ECM. Thus, changes of the ECM during tumor growth represent additional potential dysregulations of angiogenesis in addition to erroneous biochemical signals. This awareness could be the basis of therapeutic approaches to counteract specific processes in tumor angiogenesis.
- Published
- 2021
573. Nanosized zingerone-triggered anti-angiogenesis contributes to tumor suppression in human hepatocellular carcinoma.
- Author
-
Kung, Mei-Lang, Huang, Shih-Tsung, Tsai, Kuo-Wang, Chu, Tian-Huei, and Hsieh, Shuchen
- Subjects
- *
NEOVASCULARIZATION , *HEPATOCELLULAR carcinoma , *ENDOTHELIAL cells , *CELL growth , *TUMORS , *CELL lines - Abstract
Suppression of tumor angiogenesis has become one of the most important means of antitumorigenesis, tumor development, and metastasis. Therefore, developing novel anti-angiogenic agents for tumor therapeutics is an important issue. Phytochemicals possess numerous effective antitumorigenesis properties and can inhibit cell growth and metastasis and are thus regarded as promising candidates for use as chemopreventive agents and chemotherapeutic adjuvants. In our previous study, we fabricated phytochemical-derived carbon dot-zingerone nanoparticles (zingerone NPs) and demonstrated their potent antitumorigenesis in vivo and in vitro against human hepatocellular carcinoma (HCC). In this study, we further validated the roles of zingerone NPs in antitumor angiogenesis. Here, two human endothelial cell lines, EA.hy293 and HUVECs, and two hepatocellular carcinoma cell lines, SK-Hep-1 and Huh7, were utilized to investigate the effects of zingerone NPs on antitumor angiogenesis. Our data demonstrated that zingerone NPs significantly inhibited the viability of human endothelial EA.hy296 cells and HUVECs. Moreover, zingerone NPs revealed superior anti-angiogenic effects on cell invasiveness, in vitro tube formation, and ex vivo rat aorta ring angiogenesis. By verifying zingerone NP-mediated molecular mechanisms of anti-angiogenesis, we found that zingerone NPs significantly decreased the protein expression of VEGF and VEGFR in endothelial cells. Moreover, zingerone NPs downregulated VEGF expression in hepatoma cell lines. The hepatoma Huh7 xenograft model also showed that zingerone NPs markedly attenuated tumor angiogenesis through inhibition of VEGF and CD31 expression. In addition, we also demonstrated that zingerone NPs significantly suppressed tumorigenesis by inhibiting VEGF-mediated PI3K/Akt/mTOR axis signaling and Akt/eNOS signaling in endothelial and hepatocellular carcinoma cells. These results suggest that the phytochemical-derived carbon dots-zingerone NPs possess superior characteristics in antitumor angiogenesis and antitumorigenesis. Altogether, our study provides recent advances in zingerone NPs as anti-angiogenic agents to enhance chemopreventive and anticancer effects for future tumor therapeutic strategies. [Display omitted] • Zingerone NP-mediated anti-angiogenesis in vitro and ex vivo in human hepatoma. • Zingerone significantly inhibited endothelial cell activity and cell motility. • Zingerone NP-mediated downregulation of VEGF-elicited angiogenesis. • PI3K/Akt/mTOR axis signaling is involved in zingerone NP-mediated anti-angiogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
574. PBRM1 and KDM5C cooperate to define high-angiogenesis tumors and increased antiangiogenic response in renal cancer.
- Author
-
Santos M, Lanillos J, Caleiras E, Valdivia C, Roldan-Romero JM, Laínez N, Puente J, Beuselinck B, Oudard S, Zucman-Rossi J, Navarro P, Robledo M, Castellano D, de Velasco G, García-Donas J, and Rodriguez-Antona C
- Abstract
Vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) are key antiangiogenic drugs for renal cancer treatment. While Von Hippel-Lindau dysfunction constitutes the base for VEGFR-TKIs sensitivity, the role for individual and concurrent mutations in the genes encoding for the chromatin remodelers Polybromo-1 ( PBRM1 ) and Lysine Demethylase 5C ( KDM5C ) is poorly understood. Here, we analyzed the tumor mutational and expression profiles of 155 unselected clear cell RCC (ccRCC) cases treated with first-line VEGFR-TKIs and the ccRCC cases of IMmotion151 trial were used for validation. We found that concurrent PBRM1 and KDM5C ( PBRM1 & KDM5C ) mutations occurred in 4-9% of cases and were enriched in Memorial Sloan Kettering Cancer Center favorable-risk patients. In our cohort, tumors only mutated in PBRM1 or concurrently mutated in PBRM1 and KDM5C had increased angiogenesis (P=0.0068 and 0.039; respectively), and tumors only mutated in KDM5C showed a similar trend. Best response to VEGFR-TKIs corresponded to PBRM1 & KDM5C mutated cases, followed by those mutated only in KDM5C or only in PBRM1 (P=0.050, 0.040 and 0.027 versus non-mutated cases, respectively), with a trend for longer progression free survival (PFS) in the group with only PBRM1 mutated (HR=0.64; P=0.059). Validation in the IMmotion151 trial revealed a similar correlation with increased angiogenesis and the PFS of patients in the VEGFR-TKI-arm was the longest in PBRM1 & KDM5C mutated cases, intermediate for only PBRM1 or only KDM5C mutated patients and the shortest in non-mutated cases (P=0.009 and 0.025, for PBRM1 & KDM5C and PBRM1 versus non-mutated cases). In conclusion, somatic PBRM1 and KDM5C mutations are common in patients with metastatic ccRCC and likely cooperate increasing tumor angiogenesis and VEGFR-TKI-based antiangiogenic therapy benefit., Competing Interests: None., (AJCR Copyright © 2023.)
- Published
- 2023
575. ARL13B promotes angiogenesis and glioma growth by activating VEGFA-VEGFR2 signaling.
- Author
-
Chen L, Xie X, Wang T, Xu L, Zhai Z, Wu H, Deng L, Lu Q, Chen Z, Yang X, Lu H, Chen YG, and Luo S
- Subjects
- Humans, Mice, Animals, Hedgehog Proteins metabolism, Signal Transduction, Neovascularization, Pathologic metabolism, Cell Proliferation, Vascular Endothelial Growth Factor Receptor-2 metabolism, Vascular Endothelial Growth Factor A metabolism, ADP-Ribosylation Factors metabolism, ADP-Ribosylation Factors pharmacology, Endothelial Cells metabolism, Glioma pathology
- Abstract
Background: Tumor angiogenesis is essential for solid tumor progression, invasion and metastasis. The aim of this study was to identify potential signaling pathways involved in tumor angiogenesis., Methods: Genetically engineered mouse models were used to investigate the effects of endothelial ARL13B(ADP-ribosylation factor-like GTPase 13B) over-expression and deficiency on retinal and cerebral vasculature. An intracranially transplanted glioma model and a subcutaneously implanted melanoma model were employed to examine the effects of ARL13B on tumor growth and angiogenesis. Immunohistochemistry was used to measure ARL13B in glioma tissues, and scRNA-seq was used to analyze glioma and endothelial ARL13B expression. GST-fusion protein-protein interaction and co-immunoprecipitation assays were used to determine the ARL13B-VEGFR2 interaction. Immunobloting, qPCR, dual-luciferase reporter assay and functional experiments were performed to evaluate the effects of ARL13B on VEGFR2 activation., Results: Endothelial ARL13B regulated vascular development of both the retina and brain in mice. Also, ARL13B in endothelial cells regulated the growth of intracranially transplanted glioma cells and subcutaneously implanted melanoma cells by controlling tumor angiogenesis. Interestingly, this effect was attributed to ARL13B interaction with VEGFR2, through which ARL13B regulated the membrane and ciliary localization of VEGFR2 and consequently activated its downstream signaling in endothelial cells. Consistent with its oncogenic role, ARL13B was highly expressed in human gliomas, which was well correlated with the poor prognosis of glioma patients. Remarkably, ARL13B, transcriptionally regulated by ZEB1, enhanced the expression of VEGFA by activating Hedgehog signaling in glioma cells., Conclusions: ARL13B promotes angiogenesis and tumor growth by activating VEGFA-VEGFR2 signaling. Thus, targeting ARL13B might serve as a potential approach for developing an anti-glioma or anti-melanoma therapy., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
576. Cooperation between Prostaglandin E2 and Epidermal Growth Factor Receptor in Cancer Progression: A Dual Target for Cancer Therapy.
- Author
-
Finetti F, Paradisi L, Bernardi C, Pannini M, and Trabalzini L
- Abstract
It is recognized that prostaglandin E2 (PGE2) is one key lipid mediator involved in chronic inflammation, and it is directly implicated in tumor development by regulating cancer cell growth and migration, apoptosis, epithelial-mesenchymal transition, angiogenesis, and immune escape. In addition, the expression of the enzymes involved in PGE2 synthesis, cyclooxygenase 2 (COX-2) and microsomal prostaglandin E synthase 1 (mPGES1), positively correlates with tumor progression and aggressiveness, clearly indicating the crucial role of the entire pathway in cancer. Moreover, several lines of evidence suggest that the COX2/mPGES1/PGE2 inflammatory axis is involved in the modulation of epidermal growth factor receptor (EGFR) signaling to reinforce the oncogenic drive of EGFR activation. Similarly, EGFR activation promotes the induction of COX2/mPGES1 expression and PGE2 production. In this review, we describe the interplay between COX2/mPGES1/PGE2 and EGFR in cancer, and new therapeutic strategies that target this signaling pathway, to outline the importance of the modulation of the inflammatory process in cancer fighting.
- Published
- 2023
- Full Text
- View/download PDF
577. Neuroglobin plays as tumor suppressor by disrupting the stability of GPR35 in colorectal cancer.
- Author
-
Xiang Q, Zhou D, Xiang X, Le X, Deng C, Sun R, Li C, Pang H, He J, Zheng Z, Tang J, Peng W, Peng X, He X, Wu F, Qiu J, Xu Y, and Xiang T
- Subjects
- Humans, Neuroglobin genetics, Neuroglobin metabolism, Epigenesis, Genetic, Cell Line, Tumor, Biomarkers metabolism, Gene Expression Regulation, Neoplastic, Cell Proliferation, Tumor Microenvironment, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, DNA Methylation, Colorectal Neoplasms pathology
- Abstract
Background: The incidence of colorectal cancer (CRC) has increased in recent years. Identification of accurate tumor markers has become the focus of CRC research. Early and frequent DNA methylation tends to occur in cancer. Thus, identifying accurate methylation biomarkers would improve the efficacy of CRC treatment. Neuroglobin (NGB) is involved in neurological and oncological diseases. However, there are currently no reports on epigenetic regulation involvement of NGB in CRC., Results: NGB was downregulated or silenced in majority CRC tissues and cell lines. The hypermethylation of NGB was detected in tumor tissue, but no or a very low methylation frequency in normal tissues. Overexpression of NGB induced G2/M phase arrest and apoptosis, suppressed proliferation, migration, invasion in vitro, and inhibited CRC tumor growth and angiogenesis in vivo. Isobaric tag for relative and absolute quantitation (Itraq)-based proteomics identified approximately 40% proteins related to cell-cell adhesion, invasion, and tumor vessel formation in the tumor microenvironment, among which GPR35 was proved critical for NGB-regulated tumor angiogenesis suppression in CRC., Conclusions: NGB, an epigenetically silenced factor, inhibits metastasis through the GPR35 in CRC. It is expected to grow into a potential cancer risk assessment factor and a valuable biomarker for early diagnosis and prognosis assessment of CRC., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
578. NEDD8-Activating Enzyme Inhibitor MLN4924 Inhibits Both the Tumor Stroma and Angiogenesis in Pancreatic Cancer via Gli1 and REDD1.
- Author
-
Mao W, Zhang L, Rong Y, Kuang T, Wang D, Xu X, Lou W, and Li J
- Subjects
- Humans, Zinc Finger Protein GLI1 genetics, Transcription Factors genetics, Enzyme Inhibitors pharmacology, Cell Line, Tumor, Apoptosis, NEDD8 Protein, Pancreatic Neoplasms, Endothelial Cells pathology, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms pathology
- Abstract
Purpose: Pancreatic cancer is characterized by a dense desmoplasia stroma, which hinders efficient drug delivery and plays a critical role in tumor progression and metastasis. MLN4924 is a first-in-class NEDD8-activating enzyme inhibitor that exhibits anti-tumor activities toward pancreatic cancer, and given the comprehensive effects that MLN4924 could have, we ask what impact MLN4924 would have on the stroma of pancreatic cancer and its underlying mechanisms., Methods: Primary pancreatic stellate cells (PSCs) and human HMEC-1 cells were treated with MLN4924 in vitro. The proliferation and extracellular matrix protein levels of PSCs were tested, and their relationship with transcription factor Gli1 in PSCs was investigated. The angiogenic phenotypes of HMEC-1 cells were evaluated using capillary-like tube formation assay, and their relationship with REDD1 in HMEC-1 cells was investigated., Results: In this study, we found that MLN4924 inhibited the proliferation of pancreatic stellate cells and their secretion of collagen and CXCL-1, and the collagen secretion inhibiting effect of MLN4924 was related with transcription factor Gli1. MLN4924 inhibited multiple angiogenic phenotypes of HMEC-1 cells, and mTOR agonist partially relieved the inhibition of MLN4924 on HEMCs. MLN4924 increased the expression of REDD1 and REDD1 knockdown promoted the angiogenic phenotypes of HMEC-1 cells., Conclusions: Our study suggests that MLN4924 inhibits both the tumor stroma and angiogenesis in pancreatic cancer, and the inhibition effect is related with Gli1 in pancreatic stellate cells and REDD1 in vascular endothelial cells, respectively., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
579. Angiogenesis and Angiogenesis Factors in Stages of Carcinogenesis
- Author
-
Conti, Claudio J., D’Alessandro, Natale, editor, Mihich, Enrico, editor, Rausa, Luciano, editor, Tapiero, Haim, editor, and Tritton, Thomas R., editor
- Published
- 1993
- Full Text
- View/download PDF
580. Targeting OPA1-Mediated Mitochondrial Fusion Contributed to Celastrol’s Anti-Tumor Angiogenesis Effect
- Author
-
Gaofu Li, Lei Zhou, Huifang Deng, Congshu Huang, Ningning Wang, Lanxin Yue, Pengfei Zhang, Yongqiang Zhou, Wei Zhou, and Yue Gao
- Subjects
celastrol ,tumor angiogenesis ,mitochondrion-targeted medicine ,OPA1 ,Pharmaceutical Science - Abstract
Celastrol, an active triterpenoid extracted from one of the most famous traditional Chinese medicines (TCMs), Tripterygium wilfordii Hook.f., is a novel anti-cancer drug with significant anti-angiogenesis activity. However, the exact molecular mechanisms underlying its anti-tumor angiogenesis effect remain unclear. The process of angiogenesis needs lots of energy supply, which mostly derives from mitochondria, the “energy factory” in our body. This study shows that celastrol exerts visible suppression on tumor growth and angiogenesis in a cell-derived xenograft (CDX). Likewise, it reduced the tube formation and migration of human umbilical vein endothelial cells (HUVECs), suppressed the energy metabolism of mitochondria in the Seahorse XF Mito Stress Test, and triggered mitochondrial fragmentation and NF-κB activation. Mechanically, celastrol downregulated the expression of mitochondrial-sharping protein optic atrophy protein 1 (OPA1), which was further estimated by the OPA1 knockdown model of HUVECs. Specifically, celastrol directly suppressed OPA1 at the mRNA level by inhibiting the phosphorylation of STAT3, and stattic (STAT3 inhibitor) showed the same effects on OPA1 suppression and anti-angiogenesis activity. Overall, this study indicates that celastrol inhibits tumor angiogenesis by suppressing mitochondrial function and morphology via the STAT3/OPA1/P65 pathway and provides new insight for mitochondrion-targeted cancer therapy.
- Published
- 2022
- Full Text
- View/download PDF
581. Scoping Review on Platelets and Tumor Angiogenesis: Do We Need More Evidence or Better Analysis?
- Author
-
Arianna Filippelli, Cinzia Del Gaudio, Vittoria Simonis, Valerio Ciccone, Andrea Spini, and Sandra Donnini
- Subjects
Pathologic ,Blood Platelets ,Neovascularization, Pathologic ,Organic Chemistry ,tumor angiogenesis ,General Medicine ,Catalysis ,platelets ,tumor microenvironment ,Humans ,Tumor Microenvironment ,Neoplasms ,Computer Science Applications ,Inorganic Chemistry ,Physical and Theoretical Chemistry ,Molecular Biology ,Neovascularization ,Spectroscopy - Abstract
Platelets are an active component of the tumor microenvironment (TME), involved in the regulation of multiple tumor processes, including angiogenesis. They are generated rich in angiogenic factors in their granules to actively participate in the hemostatic process by megakaryocytes and further enriched in angiogenic factors by all components of the tumor microenvironment to control the angiogenic process because of their preferential relationship with the endothelial component of vessels. In recent decades, the literature has reported a great deal of evidence on the role of platelets in tumor angiogenesis; however, it is unclear whether the number or mean volume of platelets and/or their content and localization in TME may have clinical relevance in the choice and management of therapy for the cancer patient. In this scoping review, we collected and critically reviewed the scientific evidence supporting a close relationship between platelets, cancer, and angiogenesis. The aim of this work was to define the landscape of platelet-activated angiogenesis in cancer progression and analyze what and how much evidence is present in the last 20 years in the literature at both the preclinical and clinical levels, to answer whether platelets could be a useful determinant for analyzing tumor angiogenesis. In conclusion, this scoping review indicates that there is much evidence, both preclinical and clinical, but in the preclinical context, studies demonstrate the direct involvement of platelets in tumor angiogenesis; in the clinical context the evidence is indirect, though strong, and the indication of how and to what extent platelet content contributes to tumor angiogenesis is lacking. So, do we need more evidence or better analysis? More molecular and quali-quantitative data is needed to translate the results obtained in preclinical studies into the clinical setting. This information about platelets, if correlated with tumor type and its biology, including tumor vasculature, type of angiogenesis, and patient characteristics (age, sex, comorbidities, drug treatments for chronic diseases) could be an important pa- rameter for correlating platelet biology to angiogenesis, for personalizing cancer therapy, and for clinical prognosis.
- Published
- 2022
- Full Text
- View/download PDF
582. Slit/Robo Signaling Pathway in Cancer; a New Stand Point for Cancer Treatment
- Author
-
Koohini, Zahra, Koohini, Zohreh, and Teimourian, Shahram
- Published
- 2019
- Full Text
- View/download PDF
583. A Moving Mesh Method for Mathematical Model of Capillary Formation in Tumor Angiogenesis
- Author
-
Bagherpoorfard, Mina and Soheili, Ali R.
- Published
- 2019
- Full Text
- View/download PDF
584. Clinically relevant concentrations of lidocaine inhibit tumor angiogenesis through suppressing VEGF/VEGFR2 signaling
- Author
-
Gao, Jin, Hu, Huimin, and Wang, Xuesong
- Published
- 2019
- Full Text
- View/download PDF
585. Ropivacaine inhibits tumor angiogenesis via sodium-channel-independent mitochondrial dysfunction and oxidative stress
- Author
-
Yang, Jingwen, Li, Guangting, Bao, Kaibei, Liu, Weihua, Zhang, Yaozhi, and Ting, Weijen
- Published
- 2019
- Full Text
- View/download PDF
586. Dynamic Contrast-Enhanced MRI Perfusion Parameters as Imaging Biomarkers of Angiogenesis.
- Author
-
Kim, Sung Hun, Lee, Hyeon Sil, Kang, Bong Joo, Song, Byung Joo, Kim, Hyun-Bin, Lee, Hyunyong, Jin, Min-Sun, and Lee, Ahwon
- Subjects
- *
NEOVASCULARIZATION , *PERFUSION , *MAGNETIC resonance imaging , *BIOMARKERS , *TUMOR microenvironment , *DUCTAL carcinoma , *VASCULAR endothelial growth factors , *PROGNOSIS - Abstract
Hypoxia in the tumor microenvironment is the leading factor in angiogenesis. Angiogenesis can be identified by dynamic contrast-enhanced breast MRI (DCE MRI). Here we investigate the relationship between perfusion parameters on DCE MRI and angiogenic and prognostic factors in patients with invasive ductal carcinoma (IDC). Perfusion parameters (Ktrans, kep and ve) of 81 IDC were obtained using histogram analysis. Twenty-fifth, 50th and 75th percentile values were calculated and were analyzed for association with microvessel density (MVD), vascular endothelial growth factor (VEGF) and conventional prognostic factors. Correlation between MVD and ve50 was positive (r = 0.33). Ktrans50 was higher in tumors larger than 2 cm than in tumors smaller than 2 cm. In multivariate analysis, Ktrans50 was affected by tumor size and MVD with 12.8% explanation. There was significant association between Ktrans50 and tumor size and MVD. Therefore we conclude that DCE MRI perfusion parameters are potential imaging biomarkers for prediction of tumor angiogenesis and aggressiveness. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
587. Tumor-Derived Factors and Reduced p53 Promote Endothelial Cell Centrosome Over-Duplication.
- Author
-
Yu, Zhixian, Mouillesseaux, Kevin P., Kushner, Erich J., and Bautch, Victoria L.
- Subjects
- *
HYPOXEMIA , *ENDOTHELIAL cells , *CENTROSOMES , *DRUG resistance in cancer cells , *P53 antioncogene , *THERAPEUTICS - Abstract
Approximately 30% of tumor endothelial cells have over-duplicated (>2) centrosomes, which may contribute to abnormal vessel function and drug resistance. Elevated levels of vascular endothelial growth factor A induce excess centrosomes in endothelial cells, but how other features of the tumor environment affect centrosome over-duplication is not known. To test this, we treated endothelial cells with tumor-derived factors, hypoxia, or reduced p53, and assessed centrosome numbers. We found that hypoxia and elevated levels of bone morphogenetic protein 2, 6 and 7 induced excess centrosomes in endothelial cells through BMPR1A and likely via SMAD signaling. In contrast, inflammatory mediators IL-8 and lipopolysaccharide did not induce excess centrosomes. Finally, down-regulation in endothelial cells of p53, a critical regulator of DNA damage and proliferation, caused centrosome over-duplication. Our findings suggest that some tumor-derived factors and genetic changes in endothelial cells contribute to excess centrosomes in tumor endothelial cells. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
588. Normal Wound Healing and Tumor Angiogenesis as a Game of Competitive Inhibition.
- Author
-
Kareva, Irina, Abou-Slaybi, Abdo, Dodd, Oliver, Dashevsky, Olga, and Klement, Giannoula Lakka
- Subjects
- *
NEOVASCULARIZATION , *WOUND healing , *TUMOR growth , *TUMOR treatment , *GLYCOSAMINOGLYCANS , *PLATELET-derived growth factor - Abstract
Both normal wound healing and tumor angiogenesis are mitigated by the sequential, carefully orchestrated release of growth stimulators and inhibitors. These regulators are released from platelet clots formed at the sites of activated endothelium in a temporally and spatially controlled manner, and the order of their release depends on their affinity to glycosaminoglycans (GAG) such as heparan sulfate (HS) within the extracellular matrix, and platelet open canallicular system. The formation of vessel sprouts, triggered by angiogenesis regulating factors with lowest affinities for heparan sulfate (e.g. VEGF), is followed by vessel-stabilizing PDGF-B or bFGF with medium affinity for HS, and by inhibitors such as PF-4 and TSP-1 with the highest affinities for HS. The invasive wound-like edge of growing tumors has an overabundance of angiogenesis stimulators, and we propose that their abundance out-competes angiogenesis inhibitors, effectively preventing inhibition of angiogenesis and vessel maturation. We evaluate this hypothesis using an experimentally motivated agent-based model, and propose a general theoretical framework for understanding mechanistic similarities and differences between the processes of normal wound healing and pathological angiogenesis from the point of view of competitive inhibition. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
589. Dendritic cells loading autologous tumor lysate promote tumor angiogenesis.
- Author
-
Yang, Yi, Lu, Jing, Liu, Hangfan, Jin, Guoguo, Bai, Ruihua, Li, Xiang, Wang, Dongyu, Zhao, Jimin, Huang, Youtian, Liu, Kangdong, Xing, Ying, and Dong, Ziming
- Abstract
Dendritic cells (DC) have been exploited for vaccination against cancer for years. DC loading autologous tumor lysate (ATL-DC) have been assessed in ongoing clinical trials, but frequently do not meet expectation. In this study, we found that mice immunized with ATL-DC induced less protective anti-tumor effect than immunized with DC alone. The percentage of CD8 T cells and the lysis efficiency of CTLs to auto tumor cells in ATL-DC vaccination group was less than that of DC group. Moreover, vaccination of mice with ATL-DC also promoted tumor angiogenesis by analyzing the CD31 positive microvessel density and hemoglobin content of tumor specimens. Human umbilical vein endothelial cells (HUVEC) have been proved effective in the anti-angiogenesis immunity against cancer. However, in the following research we found that the anti-tumor effect was attenuated while immunized mice with HUVEC combined with ATL-DC (HUVEC + ATL-DC). Furthermore, immunized mice with HUVEC + ATL-DC profoundly increased the tumor angiogenesis by analyzing the microvessel density and hemoglobin content of tumor specimens. These data suggest that vaccination using ATL-DC antagonized HUVEC induced anti-angiogenesis effect. Our research for the first time indicated that ATL-DC have the potential to promote the process of tumor angiogenesis in vivo . As vaccines based on DC loading autologous tumor lysate have been used in clinical, this find warned that the safety of this kind of vaccine should be taken into consideration seriously. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
590. Oxygen Distributions—Evaluation of Computational Methods, Using a Stochastic Model for Large Tumour Vasculature, to Elucidate the Importance of Considering a Complete Vascular Network.
- Author
-
Lagerlöf, Jakob H. and Bernhardt, Peter
- Subjects
- *
TUMORS , *STOCHASTIC analysis , *ALGORITHMS , *MICHAELIS-Menten equation , *THRESHOLDING algorithms , *MATHEMATICAL models - Abstract
Purpose: To develop a general model that utilises a stochastic method to generate a vessel tree based on experimental data, and an associated irregular, macroscopic tumour. These will be used to evaluate two different methods for computing oxygen distribution. Methods: A vessel tree structure, and an associated tumour of 127 cm3, were generated, using a stochastic method and Bresenham’s line algorithm to develop trees on two different scales and fusing them together. The vessel dimensions were adjusted through convolution and thresholding and each vessel voxel was assigned an oxygen value. Diffusion and consumption were modelled using a Green’s function approach together with Michaelis-Menten kinetics. The computations were performed using a combined tree method (CTM) and an individual tree method (ITM). Five tumour sub-sections were compared, to evaluate the methods. Results: The oxygen distributions of the same tissue samples, using different methods of computation, were considerably less similar (root mean square deviation, RMSD≈0.02) than the distributions of different samples using CTM (0.001< RMSD<0.01). The deviations of ITM from CTM increase with lower oxygen values, resulting in ITM severely underestimating the level of hypoxia in the tumour. Kolmogorov Smirnov (KS) tests showed that millimetre-scale samples may not represent the whole. Conclusions: The stochastic model managed to capture the heterogeneous nature of hypoxic fractions and, even though the simplified computation did not considerably alter the oxygen distribution, it leads to an evident underestimation of tumour hypoxia, and thereby radioresistance. For a trustworthy computation of tumour oxygenation, the interaction between adjacent microvessel trees must not be neglected, why evaluation should be made using high resolution and the CTM, applied to the entire tumour. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
591. Coupled Hybrid Continuum-Discrete Model of Tumor Angiogenesis and Growth.
- Author
-
Lyu, Jie, Cao, Jinfeng, Zhang, Peiming, Liu, Yang, and Cheng, Hongtao
- Subjects
- *
TUMOR growth , *TUMOR blood vessels , *NEOVASCULARIZATION , *TUMOR microenvironment , *MATHEMATICAL continuum , *COMPUTATIONAL mathematics - Abstract
The processes governing tumor growth and angiogenesis are codependent. To study the relationship between them, we proposed a coupled hybrid continuum-discrete model. In this model, tumor cells, their microenvironment (extracellular matrixes, matrix-degrading enzymes, and tumor angiogenic factors), and their network of blood vessels, described by a series of discrete points, were considered. The results of numerical simulation reveal the process of tumor growth and the change in microenvironment from avascular to vascular stage, indicating that the network of blood vessels develops gradually as the tumor grows. Our findings also reveal that a tumor is divided into three regions: necrotic, semi-necrotic, and well-vascularized. The results agree well with the previous relevant studies and physiological facts, and this model represents a platform for further investigations of tumor therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
592. Candesartan attenuates angiogenesis in hepatocellular carcinoma via downregulating AT1R/VEGF pathway.
- Author
-
Fan, Fangtian, Tian, Chao, Tao, Li, Wu, Hongyan, Liu, Zhaoguo, Shen, Cunsi, Jiang, Guorong, and Lu, Yin
- Subjects
- *
CANDESARTAN , *LIVER cancer , *ANGIOTENSIN II , *ANTINEOPLASTIC agents , *PROTEIN expression , *LABORATORY mice - Abstract
Angiotensin II type 1 receptor (AT1R) was reported to express in many types of tumors, promoting tumor growth and angiogenesis. We herein examined AT1R expression in liver cancer and the potential antitumor effects of AT1R antagonist Candesartan in liver cancer. We found that AT1R expression was positively correlated with VEGF-A expression and microvascular density (MVD) in 40 HCC patients. Angiotensin II and Candesartan neither had effects on the proliferation of liver cancer cells in vitro. However, Angiotensin II upregulated AT1R protein expression and promoted production of VEGF-A in liver cancer cells in a dose-dependent manner. Candesartan was able to reverse this process in a dose-dependent manner. Moreover, Candesartan downregulated the expression of VEGF-A in SMMC-7721 bearing xenografts in mice and inhibited tumor growth and angiogenesis in vivo. Our data suggested that AT1R antagonist Candesartan might be useful to suppress liver cancer by inhibiting angiogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
593. Leptin-induced transphosphorylation of vascular endothelial growth factor receptor increases Notch and stimulates endothelial cell angiogenic transformation.
- Author
-
Lanier, Viola, Gillespie, Corey, Leffers, Merle, Daley-Brown, Danielle, Milner, Joy, Lipsey, Crystal, Webb, Nia, Anderson, Leonard M., Newman, Gale, Waltenberger, Johannes, and Gonzalez-Perez, Ruben Rene
- Subjects
- *
NOTCH signaling pathway , *LEPTIN , *VASCULAR endothelial growth factor receptors , *CELL proliferation , *ENDOTHELIAL cells , *NEOVASCULARIZATION inhibitors - Abstract
Leptin increases vascular endothelial growth factor (VEGF), VEGF receptor-2 (VEGFR-2), and Notch expression in cancer cells, and transphosphorylates VEGFR-2 in endothelial cells. However, the mechanisms involved in leptin’s actions in endothelial cells are not completely known. Here we investigated whether a leptin-VEGFR-Notch axis is involved in these leptin’s actions. To this end, human umbilical vein and porcine aortic endothelial cells (wild type and genetically modified to overexpress VEGFR-1 or -2) were cultured in the absence of VEGF and treated with leptin and inhibitors of Notch (gamma-secretase inhibitors: DAPT and S2188, and silencing RNA), VEGFR (kinase inhibitor: SU5416, and silencing RNA) and leptin receptor, OB-R (pegylated leptin peptide receptor antagonist 2: PEG-LPrA2). Interestingly, in the absence of VEGF, leptin induced the expression of several components of Notch signaling pathway in endothelial cells. Inhibition of VEGFR and Notch signaling significantly decreased leptin-induced S-phase progression, proliferation, and tube formation in endothelial cells. Moreover, leptin/OB-R induced transphosphorylation of VEGFR-1 and VEGFR-2 was essential for leptin’s effects. These results unveil for the first time a novel mechanism by which leptin could induce angiogenic features via upregulation/trans-activation of VEGFR and downstream expression/activation of Notch in endothelial cells. Thus, high levels of leptin found in overweight and obese patients might lead to increased angiogenesis by activating VEGFR-Notch signaling crosstalk in endothelial cells. These observations might be highly relevant for obese patients with cancer, where leptin/VEGFR/Notch crosstalk could play an important role in cancer growth, and could be a new target for the control of tumor angiogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
594. The metronomic therapy with prednisone, etoposide, and cyclophosphamide reduces the serum levels of VEGF and circulating endothelial cells and improves response rates and progression-free survival in patients with relapsed or refractory non-Hodgkin's lymphoma.
- Author
-
Zeng, Jiangzheng, Yang, Liangxia, Huang, Fen, Hong, Tao, He, Zhihui, Lei, Junhua, Sun, Huamao, Lu, Yanda, and Hao, Xinbao
- Subjects
- *
BLOOD serum analysis , *VASCULAR endothelial growth factors , *ENDOTHELIAL cells , *CANCER chemotherapy , *PROGRESSION-free survival , *CANCER relapse , *ANTINEOPLASTIC agents , *DRUG resistance in cancer cells , *DRUG administration , *EPITHELIAL cells , *ETOPOSIDE , *LYMPHOMAS , *PREDNISONE , *PROGNOSIS , *DISEASE relapse , *CYCLOPHOSPHAMIDE - Abstract
Purpose: There is an urgent need for a better strategy in the management of relapsed or refractory non-Hodgkin's lymphoma (NHL). The present study was designed to evaluate the efficacy and safety of the regimen using metronomic prednisone, etoposide, and cyclophosphamide in the treatment of patients with relapsed or refractory NHL, in comparison with conventional salvage chemotherapy.Methods: Eligible patients were randomized to the test group (n = 23) receiving metronomic prednisone, etoposide, and cyclophosphamide or the control group (n = 21) receiving conventional salvage chemotherapy. The serum levels of circulating endothelial cells (CECs) and vascular endothelial growth factor (VEGF) were measured before and after two cycles of treatment; overall response rate (ORR) and disease control rate (DCR) were evaluated at cycles 2, 4, 6, and 12 months after treatment.Results: After two cycles of treatment, the ORRs of the test and control groups were statistically similar, while the DCR of the test group (87.0 %) was significantly higher than that of the control group (57.1 %). At 12 months after treatment, the ORR and DCR of the test group (47.8 and 69.6 %, respectively) were significantly higher than that of the control group (19.0 and 33.3 %, respectively). The serum CECs and VEGF levels in the test group after treatment were significantly lower than that before treatment or that of the control group. In the patients with ORR and DCR in the test group, the serum CECs and VEGF levels remained relatively low at cycles 2, 4, and 6 and at 12 months after treatment. There was a progression-free survival (PFS) benefit of 6.5 months in the test group, compared with the control group.Conclusion: Metronomic chemotherapy with prednisone, etoposide, and cyclophosphamide resulted in higher ORR and DCR, fewer adverse effects, and longer PFS in patients with relapsed or refractory NHL, with significant reduction in serum CECs and VEGF levels. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
595. Classical and non-classical proangiogenic factors as a target of antiangiogenic therapy in tumor microenvironment.
- Author
-
Marech, Ilaria, Leporini, Christian, Ammendola, Michele, Porcelli, Mariangela, Gadaleta, Cosmo Damiano, Russo, Emilio, De Sarro, Giovambattista, and Ranieri, Girolamo
- Subjects
- *
TUMOR microenvironment , *NEOVASCULARIZATION , *CANCER cell proliferation , *CANCER cell migration , *CANCER treatment , *CELLULAR signal transduction , *GENETIC overexpression - Abstract
Angiogenesis is sustained by classical and non-classical proangiogenic factors (PFs) acting in tumor microenvironment and these factors are also potential targets of antiangiogenic therapies. All PFs induce the overexpression of several signaling pathways that lead to migration and proliferation of endothelial cells contributing to tumor angiogenesis and survival of cancer cells. In this review, we have analyzed each PF with its specific receptor/s and we have summarized the available antiangiogenic drugs (e.g. monoclonal antibodies) targeting these PFs, some of these agents have already been approved, others are currently in development for the treatment of several human malignancies. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
596. DCE-MRI-Derived Parameters in Evaluating Abraxane-Induced Early Vascular Response and the Effectiveness of Its Synergistic Interaction with Cisplatin.
- Author
-
Sun, Xilin, Yang, Lili, Yan, Xuefeng, Sun, Yingying, Zhao, Dongliang, Ji, Yang, Wang, Kai, Chen, Xiaoyuan, and Shen, Baozhong
- Subjects
- *
PACLITAXEL , *DRUG efficacy , *CANCER treatment , *DRUG synergism , *CISPLATIN , *BLOOD vessels - Abstract
Our previous studies revealed molecular alterations of tumor vessels, varying from immature to mature alterations, resulting from Abraxane, and demonstrated that the integrin-specific PET tracer 18F-FPPRGD2 can be used to noninvasively monitor such changes. However, changes in the tumor vasculature at functional levels such as perfusion and permeability are also important for monitoring Abraxane treatment outcomes in patients with cancer. The purpose of this study is to further investigate the vascular response during Abraxane therapy and the effectiveness of its synergistic interaction with cisplatin using Dynamic contrast enhanced-magnetic resonance imaging (DCE-MRI). Thirty MDA-MB-435 tumor mice were randomized into three groups: PBS control (C group), Abraxane only (A group), and sequential treatment with Abraxane followed by cisplatin (A-P group). Tumor volume was monitored based on caliper measurements. A DCE-MRI protocol was performed at baseline and day 3. The Ktrans, Kep and Ve were calculated and compared with CD31, α-SMA, and Ki67 histology data. Sequential treatment with Abraxane followed by cisplatin produced a significantly greater inhibition of tumor growth during the three weeks of the observation period. Decreases in Ktrans and Kep for the A and A-P groups were observed on day 3. Immunohistological staining suggested vascular remodeling during the Abraxane therapy. The changes in Ktrans and Kep values were correlated with alterations in the permeability of the tumor vasculature induced by the Abraxane treatment. In conclusion, Abraxane-mediated permeability variations in tumor vasculature can be quantitatively visualized by DCE-MRI, making this a useful method for studying the effects of early cancer treatment, especially the early vascular response. Vascular remodeling by Abraxane improves the efficiency of cisplatin delivery and thus results in a favorable treatment outcome. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
597. Simulation of tumor induced angiogenesis using an analytical adaptive modeling including dynamic sprouting and blood flow modeling.
- Author
-
Naghavi, Nadia, Hosseini, Farideh.S., Sardarabadi, Mohammad, and Kalani, Hadi
- Subjects
- *
NEOVASCULARIZATION , *GERMINATION , *BLOOD flow , *GROWTH factors , *VASCULAR endothelial growth factors , *STOCHASTIC analysis , *THERAPEUTICS - Abstract
In this paper, an adaptive model for tumor induced angiogenesis is developed that integrates generation and diffusion of a growth factor originated from hypoxic cells, adaptive sprouting from a parent vessel, blood flow and structural adaptation. The proposed adaptive sprout spacing model (ASS) determines position, time and number of sprouts which are activated from a parent vessel and also the developed vascular network is modified by a novel sprout branching prediction algorithm. This algorithm couples local vascular endothelial growth factor (VEGF) concentrations, stresses due to the blood flow and stochastic branching to the structural reactions of each vessel segment in response to mechanical and biochemical stimuli. The results provide predictions for the time-dependent development of the network structure, including the position and diameters of each segment and the resulting distributions of blood flow and VEGF. Considering time delays between sprout progressions and number of sprouts activated at different time durations provides information about micro-vessel density in the network. Resulting insights could be useful for motivating experimental investigations of vascular pattern in tumor induced angiogenesis and development of therapies targeting angiogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
598. Reprogramming of Normal Fibroblasts into Cancer-Associated Fibroblasts by miRNAs-Mediated CCL2/VEGFA Signaling.
- Author
-
Shen, Hua, Yu, Xiaobo, Yang, Fengming, Zhang, Zhihua, Shen, Jianxin, Sun, Jin, Choksi, Swati, Jitkaew, Siriporn, and Shu, Yongqian
- Subjects
- *
FIBROBLASTS , *CONNECTIVE tissue cells , *MICRORNA , *DISEASE progression , *CANCER cells , *CYTOKINES , *CELLULAR immunity - Abstract
Cancer-associated fibroblasts (CAFs), the most common constituent of the tumor stoma, are known to promote tumor initiation, progression and metastasis. However, the mechanism of how cancer cells transform normal fibroblasts (NFs) into CAFs is largely unknown. In this study, we determined the contribution of miRNAs in the transformation of NFs into CAFs. We found that miR-1 and miR-206 were down-regulated, whereas miR-31 was up-regulated in lung CAFs when compared with matched NFs. Importantly, modifying the expression of these three deregulated miRNAs induced a functional conversion of NFs into CAFs and vice versa. When the miRNA-reprogrammed NFs and CAFs were co-cultured with lung cancer cells (LCCs), a similar pattern of cytokine expression profiling were observed between two groups. Using a combination of cytokine expression profiling and miRNAs algorithms, we identified VEGFA/CCL2 and FOXO3a as direct targets of miR-1, miR-206 and miR-31, respectively. Importantly, systemic delivery of anti-VEGFA/CCL2 or pre-miR-1, pre-miR-206 and anti-miR-31 significantly inhibited tumor angiogenesis, TAMs accumulation, tumor growth and lung metastasis. Our results show that miRNAs-mediated FOXO3a/VEGF/CCL2 signaling plays a prominent role in LCCs-mediated NFs into CAFs, which may have clinical implications for providing novel biomarker(s) and potential therapeutic target(s) of lung cancer in the future. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
599. Synthetic Site-Selectively Mono-6-O-Sulfated Heparan Sulfate Dodecasaccharide Shows Anti-Angiogenic Properties In Vitro and Sensitizes Tumors to Cisplatin In Vivo.
- Author
-
Avizienyte, Egle, Cole, Claire L., Rushton, Graham, Miller, Gavin J., Bugatti, Antonella, Presta, Marco, Gardiner, John M., and Jayson, Gordon C.
- Subjects
- *
HEPARAN sulfate , *SULFATES , *CISPLATIN , *GLYCOSAMINOGLYCANS , *GLUCOSAMINE , *ENDOTHELIAL cells - Abstract
Heparan sulphate (HS), a ubiquitously expressed glycosaminoglycan (GAG), regulates multiple cellular functions by mediating interactions between numerous growth factors and their cell surface cognate receptors. However, the structural specificity of HS in these interactions remains largely undefined. Here, we used completely synthetic, structurally defined, alternating N-sulfated glucosamine (NS) and 2-O-sulfated iduronate (IS) residues to generate dodecasaccharides ([NSIS]6) that contained no, one or six glucosamine 6-O-sulfates (6S). The aim was to address how 6S contributes to the potential of defined HS dodecasaccharides to inhibit the angiogenic growth factors FGF2 and VEGF165, in vitro and in vivo. We show that the addition of a single 6S at the non-reducing end of [NSIS]6, i.e. [NSIS6S]-[NSIS]5, significantly augments the inhibition of FGF2-dependent endothelial cell proliferation, migration and sprouting in vitro when compared to the non-6S variant. In contrast, the fully 6-O-sulfated dodecasaccharide, [NSIS6S]6, is not a potent inhibitor of FGF2. Addition of a single 6S did not significantly improve inhibitory properties of [NSIS]6 when tested against VEGF165-dependent endothelial cell functions.In vivo, [NSIS6S]-[NSIS]5 blocked FGF2-dependent blood vessel formation without affecting tumor growth. Reduction of non-FGF2-dependent ovarian tumor growth occurred when [NSIS6S]-[NSIS]5 was combined with cisplatin. The degree of inhibition by [NSIS6S]-[NSIS]5 in combination with cisplatin in vivo equated with that induced by bevacizumab and sunitinib when administered with cisplatin. Evaluation of post-treatment vasculature revealed that [NSIS6S]-[NSIS]5 treatment had the greatest impact on tumor blood vessel size and lumen formation. Our data for the first time demonstrate that synthetic, structurally defined oligosaccharides have potential to be developed as active anti-angiogenic agents that sensitize tumors to chemotherapeutic agents. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
600. Construction of a disulfide-stabilized diabody against fibroblast growth factor-2 and the inhibition activity in targeting breast cancer.
- Author
-
Cai, Yaxiong, Zhang, Jinxia, Lao, Xuejun, Jiang, Haowu, Yu, Yunfei, Deng, Yanrui, Zhong, Jiangchuan, Liang, Yiye, Xiong, Likuan, and Deng, Ning
- Abstract
Fibroblast growth factor-2 ( FGF-2) is one of the most important angiogenic factors to promote tumor growth, progression and metastasis. Neutralizing antibodies against FGF-2 may suppress the growth of tumor cells by blocking the FGF-2 signaling pathway. In this study, a disulfide-stabilized diabody (ds-Diabody) that specifically targets FGF-2 was designed. Compared to its parent antibody, the introduction of disulphide bonds in the diabody could significantly increase the stability of ds-Diabody and maintain its antigen binding activity. The ds-Diabody against FGF-2 could effectively inhibit the tube formation and migration of vascular endothelial cells and block the proliferation and invasion of human breast cancer cells. In the mouse model of breast cancer xenograft tumors, the ds-Diabody against FGF-2 could significantly inhibit the growth of tumor cells. Moreover, the densities of microvessels stained with CD31 and lymphatic vessels stained with LYVE1 in tumors showed a significant decrease following treatment with the ds-Diabody against FGF-2. Our data indicated that the ds-Diabody against FGF-2 could inhibit tumor angiogenesis, lymphangiogenesis and tumor growth. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.