Endostatin Lowers Blood Pressure Via Nitric Oxide and Prevents Hypertension Associated with VEGF Inhibition

Total Page:16

File Type:pdf, Size:1020Kb

Endostatin Lowers Blood Pressure Via Nitric Oxide and Prevents Hypertension Associated with VEGF Inhibition Endostatin lowers blood pressure via nitric oxide and prevents hypertension associated with VEGF inhibition Sarah B. Sunshinea,1, Susan M. Dallabridaa, Ellen Duranda, Nesreen S. Ismaila, Lauren Bazineta, Amy E. Birsnera, Regina Sohnb, Sadakatsu Ikedac, William T. Puc, Matthew H. Kulked, Kashi Javaheriana, David Zurakowskie,f, Judah M. Folkmana,2, and Maria Rupnicka,b Divisions of aVascular Biology and cCardiology and Departments of eAnesthesia and fSurgery, Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115; bDivision of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115; and dDepartment of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215 Edited by Robert Langer, Massachusetts Institute of Technology, Cambridge, MA, and approved May 28, 2012 (received for review April 2, 2012) Antiangiogenesis therapy has become a vital part of the arma- control. VEGF stimulates endothelial NO synthase (eNOS), mentarium against cancer. Hypertension is a dose-limiting toxicity resulting in NO production and lower BP (21, 22). Inhibiting for VEGF inhibitors. Thus, there is a pressing need to address the VEGF in animal studies reduces eNOS expression, leading to associated adverse events so these agents can be better used. The vasoconstriction and hypertension (23). In patients, VEGF in- hypertension may be mediated by reduced NO bioavailability fusion causes rapid NO release and hypotension (24). resulting from VEGF inhibition. We proposed that the hyperten- Endostatin (ES), a fragment of collagen XVIII on chromosome sion may be prevented by coadministration with endostatin (ES), 21, is an endogenous angiogenesis inhibitor (25, 26). This 183- an endogenous angiogenesis inhibitor with antitumor effects amino acid fragment causes tumor regression in a number of shown to increase endothelial NO production in vitro. We de- animal models (27, 28). Although the molecular pathways are not termined that Fc-conjugated ES promoted NO production in fully defined, major effects of ES signaling include inhibition of endothelial and smooth muscle cells. ES also lowered blood endothelial cell migration and survival and angiogenesis. In ad- pressure in normotensive mice and prevented hypertension in- dition, ES induces NO release by cultured endothelial cells and duced by anti-VEGF antibodies. This effect was associated with relaxation of ex vivo vascular rings (29, 30). Down syndrome higher circulating nitrate levels and was absent in eNOS-knockout patients have an extra copy of chromosome 21 and a negligible mice, implicating a NO-mediated mechanism. Retrospective study incidence of solid tumors (31). Although several genes likely MEDICAL SCIENCES of patients treated with ES in a clinical trial revealed a small but contribute to this cancer protection (32), it is intriguing to note significant reduction in blood pressure, suggesting that the find- that these patients have ES levels 1.6 times higher than those of ings may translate to the clinic. Coadministration of ES with VEGF the general population (33). Further, their BP is lower than age- inhibitors may offer a unique strategy to prevent drug-related matched controls (34, 35). These data suggested to us that ES may hypertension and enhance antiangiogenic tumor suppression. enhance the antiangiogenic benefits and lessen the hypertensive effects of VEGF inhibition. Such a finding would offer an ap- nhibiting angiogenesis has proven to be effective in treating proach to improve tolerance to VEGF inhibitors, enabling long- Idiseases dependent on new blood vessel growth. In cancer term treatment with reduced risk of cardiovascular adverse events. patients, antiangiogenic agents prolong progression-free survival Here we show that murine Fc-conjugated ES lowers BP in and improve response rates when used in combination with cy- mice via an NO-mediated mechanism and blocks the hyperten- totoxic chemotherapy (1). In macular degeneration and diabetic sive response to anti-VEGF antibodies. Further, we found fi retinopathy these agents reduce vision loss (2, 3). Consequently, a small but signi cant reduction in BP in patients treated with ES fi angiogenesis inhibitors have been approved in 29 countries thus as part of a clinical trial, suggesting that the nding in mice may far (4), and new applications continue to be explored. be translatable. These results support further investigation into VEGF is a potent angiogenesis stimulator clinically estab- antitumor effects of combined therapy. fi fi lished as an ef cacious target for inhibition. The rst Food and Results Drug Administration-approved angiogenesis inhibitor was bev- acizumab (Avastin), a monoclonal anti-VEGF antibody now ES Lowers BP in C57BL/6 Mice. Normotensive C57BL/6 mice were used to treat several types of cancer (colon, lung, renal, breast) treated with ES at various doses and schedules, and BP and ocular neovascularization. Unfortunately, the enthusiasm for responses were compared with vehicle (saline)-treated controls. All mice were injected daily for 5 d with either ES or saline; bevacizumab and other such inhibitors is tempered by the after a 2-d intervening break, the cycle was repeated. A slight emergence of treatment-limiting adverse cardiovascular effects. reduction in BP at the onset of the study was noted in all the Hypertension is the most common dose-limiting toxicity of groups, likely resulting from further acclimation. Thereafter, BP VEGF inhibitors (5–9). Incidence ranges from 15% to 60%, in the control group remained unchanged, whereas BP of the depending on drug- and patient-related factors still being defined ES-treated groups decreased in relation to dose and schedule (10–14). Early and aggressive initiation of antihypertensive ther- (Fig. 1). Mice receiving ES (4 mg/kg) biweekly had the smallest apy can help maintain the treatment schedule (15) and reduce complications (16, 17). However, baseline blood pressures (BP) often are not reestablished (18). Further, it appears that nearly all Author contributions: S.B.S., S.M.D., J.M.F., and M.R. designed research; S.B.S., S.M.D., patients experience some increase in BP, even if not frank hy- E.D., N.S.I., L.B., A.E.B., R.S., S.I., W.T.P., M.H.K., K.J., J.M.F., and M.R. performed re- pertension (19). This finding is concerning, given that changes in search; S.B.S., S.M.D., N.S.I., M.H.K., D.Z., J.M.F., and M.R. analyzed data; and S.B.S. and BP of as little as 5 mm Hg can significantly impact mortality (20). M.R. wrote the paper. As life expectancy for patients maintained on these newer anti- The authors declare no conflict of interest. tumor agents continues to improve, complications from the ac- This article is a PNAS Direct Submission. companying chronic BP elevations will likely accumulate. 1To whom correspondence should be addressed. E-mail: [email protected]. One widely held explanation for angiogenesis inhibitor-asso- 2Deceased January 14, 2008. ciated hypertension is based on the role of VEGF in NO regu- This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. lation. NO is a potent vasodilator that plays a critical role in BP 1073/pnas.1203275109/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1203275109 PNAS Early Edition | 1of6 Downloaded by guest on September 24, 2021 Fig. 1. Murine Fc-ES resulted in a dose-dependent reduction in systolic BP in C57BL/6 mice. The low-dose group received 4 mg/kg, 2 d/wk (on days 0, 3, 7, 10, 14, and 17) and had the smallest decrease in BP. The high-dose group received 4 mg/kg, 5 d/wk (on days 0–4, 7–11, and 14–18) and had the most stable BP reduction. The concentrated-dose group received the cumulative high weekly dose of 20 mg/kg administered in a single concentrated bolus once a week (on days 0, 7, and 14) and had the greatest declines in BP on the days following treatment; BP levels recovered thereafter. *P < 0.05. BP reductions, with nadirs of ∼10 mm Hg below controls on the day following treatment (e.g., days 1 and 18). BP rebounded to about 5 mm Hg below controls between drug doses. Mice treated with ES (4 mg/kg) for 5 d consecutively exhibited the most stable reduction in BP, to an average of 10 mm Hg below controls, and did not rebound during the 2-d treatment breaks. The last group, which received a cumulative weekly dose of 20 mg/kg as a single, concentrated bolus and had the most sig- nificant decline in BP, to nearly 20 mm Hg below control fol- lowing drug injections. The BP then recovered to baseline before the next dose. Endogenous and Exogenous ES Increases NO. Physiologic concen- trations of ES have been shown to increase NO production in endothelial cells and to decrease vascular tone in ex vivo studies Fig. 2. Murine Fc-ES administration (4 mg/kg, 5 d/wk) is associated with (A) (29). We found that murine Fc-ES significantly increased nitrite higher circulating nitrate levels (a measure of NO) and (B) lower systolic BP in production in microvascular (MS1) endothelial cells (P = 0.005) C57BL/6 mice as compared with saline-treated controls. (C) Circulating NO (Fig. S1A) and smooth muscle cells (P = 0.0004) (Fig. S1B) levels were higher in ES-overexpressing mice and were lower in collagen compared with the matched vehicle (saline) controls. Elevated XVIII/ES-knockout mice than in wild-type control mice. These findings cor- NO production by these vascular cells in response to ES in vivo related NO levels with endogenous and exogenous ES levels. would lead to lower BP. fi Serum nitrate levels were signi cantly higher in mice treated nearly 130 mm Hg compared with 105 mm Hg typically seen in P A with ES ( = 0.0003) (Fig.
Recommended publications
  • Endostatin: a Novel Inhibitor of Androgen Receptor Function in Prostate Cancer
    Endostatin: A novel inhibitor of androgen receptor function in prostate cancer Joo Hyoung Leea, Tatyana Isayevaa, Matthew R. Larsonb, Anandi Sawanta, Ha-Ram Chaa, Diptiman Chandaa, Igor N. Chesnokovc, and Selvarangan Ponnazhagana,1 Departments of aPathology and cBiochemistry and Molecular Genetics, University of Alabama, Birmingham, AL 35294; and bDepartment of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, MI 48109 Edited* by Louise T. Chow, University of Alabama at Birmingham, Birmingham, AL, and approved December 29, 2014 (received for review September 12, 2014) Acquired resistance to androgen receptor (AR)-targeted therapies a C-terminal LBD. Like other nuclear receptors (NRs), AR is a compels the development of novel treatment strategies for castra- transcription factor regulating target-gene expression in a ligand- tion-resistant prostate cancer (CRPC). Here, we report a profound dependent manner (2, 16). Cognate ligand binding induces effect of endostatin on prostate cancer cells by efficient intracellular conformational changes predominantly in helix 12 of AR trafficking, direct interaction with AR, reduction of nuclear AR level, LBD, which enhances transcriptional activity by forming a ligand- and down-regulation of AR-target gene transcription. Structural dependent AF-2 binding interface for coactivators (17). Wilson modeling followed by functional analyses further revealed that and colleagues demonstrated that the interdomain interaction phenylalanine-rich α1-helix in endostatin—which shares struc- between AF-1 in NTD and AF-2 in LBD (N/C interaction) leads tural similarity with noncanonical nuclear receptor box in AR— to AR stabilization and slower ligand dissociation (18, 19). antagonizes AR transcriptional activity by occupying the activation Functional activity of AR largely depends on AF-2 that function (AF)-2 binding interface for coactivators and N-terminal accommodates the binding of various AR coactivators by rec- AR AF-1.
    [Show full text]
  • Therapeutic Inhibition of VEGF Signaling and Associated Nephrotoxicities
    REVIEW www.jasn.org Therapeutic Inhibition of VEGF Signaling and Associated Nephrotoxicities Chelsea C. Estrada,1 Alejandro Maldonado,1 and Sandeep K. Mallipattu1,2 1Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York; and 2Renal Section, Northport Veterans Affairs Medical Center, Northport, New York ABSTRACT Inhibition of vascular endothelial growth factor A (VEGFA)/vascular endothelial with hypertension and proteinuria. Re- growth factor receptor 2 (VEGFR2) signaling is a common therapeutic strategy in ports describe histologic changes in the oncology, with new drugs continuously in development. In this review, we consider kidney primarily as glomerular endothe- the experimental and clinical evidence behind the diverse nephrotoxicities associ- lial injury with thrombotic microangiop- ated with the inhibition of this pathway. We also review the renal effects of VEGF athy (TMA).8 Nephrotic syndrome has inhibition’s mediation of key downstream signaling pathways, specifically MAPK/ also been observed,9 with the clinical ERK1/2, endothelial nitric oxide synthase, and mammalian target of rapamycin manifestations varying according to (mTOR). Direct VEGFA inhibition via antibody binding or VEGF trap (a soluble decoy mechanism and direct target of VEGF receptor) is associated with renal-specific thrombotic microangiopathy (TMA). Re- inhibition. ports also indicate that tyrosine kinase inhibition of the VEGF receptors is prefer- Current VEGF inhibitors can be clas- entially associated with glomerulopathies such as minimal change disease and FSGS. sifiedbytheirtargetofactioninthe Inhibition of the downstream pathway RAF/MAPK/ERK has largely been associated VEGFA-VEGFR2 pathway: drugs that with tubulointerstitial injury. Inhibition of mTOR is most commonly associated with bind to VEGFA, sequester VEGFA, in- albuminuria and podocyte injury, but has also been linked to renal-specificTMA.In hibit receptor tyrosine kinases (RTKs), all, we review the experimentally validated mechanisms by which VEGFA-VEGFR2 or inhibit downstream pathways.
    [Show full text]
  • MINIREVIEW Complex Role of Matrix Metalloproteinases in Angiogen- Esis
    Cell Research (1998), 8, 171-177 MINIREVIEW Complex role of matrix metalloproteinases in angiogen- esis SANG QING XIANG AMY* Biochemistry Division, Department of Chemistry, Florida State University, Tallahassee, Florida 32306-4390, USA ABSTRACT Matrix metalloproteinases (MMPs) and tissue in- hibitors of metalloproteinases (TIMPs) play a significant role in regulating angiogenesis, the process of new blood vessel formation. Interstitial collagenase (MMP-1), 72 kDa gelatinase A/type IV collagenase (MMP-2), and 92 kDa gelatinase B/type IV collagenase (MMP-9) dissolve ex- tracellular matrix (ECM) and may initiate and promote angiogenesis. TIMP-1, TIMP-2, TIMP-3, and possibly, TIMP-4 inhibit neovascularization. A new paradigm is emerging that matrilysin (MMP-7), MMP-9, and metal- loelastase (MMP-12) may block angiogenesis by convert- ing plasminogen to angiostatin, which is one of the most potent angiogenesis antagonists. MMPs and TIMPs play a complex role in regulating angiogenesis. An understanding of the biochemical and cellular pathways and mechanisms of angiogenesis will provide important information to al- low the control of angiogenesis, e.g. the stimulation of angiogenesis for coronary collateral circulation formation; while the inhibition for treating arthritis and cancer. Key word s: Collagenases, tissue inhibitors of metallo- proteinases, neovascularization, plasmino- gen angiostatin converting enzymes, ex- tracellular matrix. * Corresponding author: Professor Qing Xiang Amy Sang, Department of Chemistry, 203 Dittmer Laboratory of Chemistry Building, Florida State University, Tallahassee, Florida 32306-4390, USA Phone: (850) 644-8683 Fax: (850) 644-8281 E-mail: [email protected]. 171 MMPs in angiogenesis Significance of matrix metalloproteinases in angiogenesis Matrix metalloproteinases (MMPs) are a family of highly homologous zinc en- dopeptidases that cleave peptide bonds of the extracellular matrix (ECM) proteins, such as collagens, laminins, elastin, and fibronectin[1, 2, 3].
    [Show full text]
  • Integrins: Roles in Cancer Development and As Treatment Targets
    British Journal of Cancer (2004) 90, 561 – 565 & 2004 Cancer Research UK All rights reserved 0007 – 0920/04 $25.00 www.bjcancer.com Minireview Integrins: roles in cancer development and as treatment targets 1 ,1,2 H Jin and J Varner* 1John and Rebecca Moores Comprehensive Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0912, USA; 2Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0912, USA The integrin family of cell adhesion proteins promotes the attachment and migration of cells on the surrounding extracellular matrix (ECM). Through signals transduced upon integrin ligation by ECM proteins or immunoglobulin superfamily molecules, this family of proteins plays key roles in regulating tumour growth and metastasis as well as tumour angiogenesis. Several integrins play key roles in promoting tumour angiogenesis and tumour metastasis. Antagonists of several integrins (a5b1, avb3 and avb5) are now under evaluation in clinical trials to determine their potential as therapeutics for cancer and other diseases. British Journal of Cancer (2004) 90, 561 – 565. doi:10.1038/sj.bjc.6601576 www.bjcancer.com & 2004 Cancer Research UK Keywords: angiogenesis; metastasis; apoptosis; integrin a5b1; integrin avb3 During the last 10 years, novel insights into the mechanisms sequences (e.g., integrin a4b1 recognises EILDV and REDV in that regulate cell survival as well as cell migration and invasion alternatively spliced CS-1 fibronectin). Inhibitors of integrin have led to the development of novel integrin-based therapeutics function include function-blocking monoclonal antibodies, pep- for the treatment of cancer. Several integrins play important tide antagonists and small molecule peptide mimetics matrix roles in promoting cell proliferation, migration and survival (reviewed in Hynes, 1992; Cheresh, 1993).
    [Show full text]
  • Matrix Metalloproteinases in Angiogenesis: a Moving Target for Therapeutic Intervention
    Matrix metalloproteinases in angiogenesis: a moving target for therapeutic intervention William G. Stetler-Stevenson J Clin Invest. 1999;103(9):1237-1241. https://doi.org/10.1172/JCI6870. Perspective Angiogenesis is the process in which new vessels emerge from existing endothelial lined vessels. This is distinct from the process of vasculogenesis in that the endothelial cells arise by proliferation from existing vessels rather than differentiating from stem cells. Angiogenesis is an invasive process that requires proteolysis of the extracellular matrix and, proliferation and migration of endothelial cells, as well as synthesis of new matrix components. During embryonic development, both vasculogenesis and angiogenesis contribute to formation of the circulatory system. In the adult, with the single exception of the reproductive cycle in women, angiogenesis is initiated only in response to a pathologic condition, such as inflammation or hypoxia. The angiogenic response is critical for progression of wound healing and rheumatoid arthritis. Angiogenesis is also a prerequisite for tumor growth and metastasis formation. Therefore, understanding the cellular events involved in angiogenesis and the molecular regulation of these events has enormous clinical implications. This understanding is providing novel therapeutic targets for the treatment of a variety of diseases, including cancer. Whatever the pathologic condition, an initiating stimulus results in the formation of a migrating solid column of endothelial cells called the vascular sprout. The advancing front of this endothelial cell column presumably focuses proteolytic activity to create a defect in the extracellular matrix, through which the advancing and proliferating column of endothelial […] Find the latest version: https://jci.me/6870/pdf Matrix metalloproteinases in angiogenesis: Perspective a moving target for therapeutic intervention SERIES Topics in angiogenesis David A.
    [Show full text]
  • Tumor Angiogenesis and Anti-Angiogenic Strategies for Cancer Treatment
    Journal of Clinical Medicine Review Tumor Angiogenesis and Anti-Angiogenic Strategies for Cancer Treatment Raluca Ioana Teleanu 1, Cristina Chircov 2,3 , Alexandru Mihai Grumezescu 3,* and Daniel Mihai Teleanu 4 1 “Victor Gomoiu” Clinical Children’s Hospital, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; [email protected] 2 Faculty of Engineering in Foreign Languages, 060042 Bucharest, Romania; [email protected] 3 Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania 4 Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; [email protected] * Correspondence: [email protected]; Tel.: +40-21-402-39-97 Received: 4 December 2019; Accepted: 19 December 2019; Published: 29 December 2019 Abstract: Angiogenesis is the process through which novel blood vessels are formed from pre-existing ones and it is involved in both physiological and pathological processes of the body. Furthermore, tumor angiogenesis is a crucial factor associated with tumor growth, progression, and metastasis. In this manner, there has been a great interest in the development of anti-angiogenesis strategies that could inhibit tumor vascularization. Conventional approaches comprise the administration of anti-angiogenic drugs that target and block the activity of proangiogenic factors. However, as their efficacy is still a matter of debate, novel strategies have been focusing on combining anti-angiogenic agents with chemotherapy or immunotherapy. Moreover, nanotechnology has also been investigated for the potential of nanomaterials to target and release anti-angiogenic drugs at specific sites. The aim of this paper is to review the mechanisms involved in angiogenesis and tumor vascularization and provide an overview of the recent trends in anti-angiogenic strategies for cancer therapy.
    [Show full text]
  • The Procollagen N-Proteinases ADAMTS2, 3 and 14 in Pathophysiology
    Review The procollagen N-proteinases ADAMTS2, 3 and 14 in pathophysiology Mourad Bekhouche and Alain Colige Laboratory of Connective Tissues Biology, GIGA-R, University of Liège, B-4000 Sart Tilman, Belgium Correspondence to Alain Colige: Laboratory of Connective Tissues Biology, University of Liège, GIGA-Research, Tour de Pathologie B23/3, Avenue de l'Hôpital, 3, B-4000 Sart Tilman, Belgium. [email protected] http://dx.doi.org/10.1016/j.matbio.2015.04.001 Edited by W.C. Parks and S. Apte Abstract Collagen fibers are the main components of most of the extracellular matrices where they provide a structural support to cells, tissues and organs. Fibril-forming procollagens are synthetized as individual chains that associate to form homo- or hetero-trimers. They are characterized by the presence of a central triple helical domain flanked by amino and carboxy propeptides. Although there are some exceptions, these two propeptides have to be proteolytically removed to allow the almost spontaneous assembly of the trimers into collagen fibrils and fibers. While the carboxy-propeptide is mainly cleaved by proteinases from the tolloid family, the amino-propeptide is usually processed by procollagen N-proteinases: ADAMTS2, 3 and 14. This review summarizes the current knowledge concerning this subfamily of ADAMTS enzymes and discusses their potential involvement in physiopathological processes that are not directly linked to fibrillar procollagen processing. © 2015 Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). Introduction determine the cause of dermatosparaxis, a rare genetic disease that appeared in Belgian cattle herds during an Fibrillar collagens are the most abundant proteins inbreeding program [2,3].
    [Show full text]
  • C11, a Novel Fibroblast Growth Factor Receptor 1 (FGFR1)
    www.nature.com/aps ARTICLE C11, a novel fibroblast growth factor receptor 1 (FGFR1) inhibitor, suppresses breast cancer metastasis and angiogenesis Zhuo Chen1,2, Lin-jiang Tong1, Bai-you Tang1, Hong-yan Liu1, Xin Wang2, Tao Zhang1, Xian-wen Cao2, Yi Chen1, Hong-lin Li2, Xu-hong Qian2, Yu-fang Xu2, Hua Xie1 and Jian Ding1 The fibroblast growth factor receptors (FGFRs) are increasingly considered attractive targets for therapeutic cancer intervention due to their roles in tumor metastasis and angiogenesis. Here, we identified a new selective FGFR inhibitor, C11, and assessed its antitumor activities. C11 was a selective FGFR1 inhibitor with an IC50 of 19 nM among a panel of 20 tyrosine kinases. C11 inhibited cell proliferation in various tumors, particularly bladder cancer and breast cancer. C11 also inhibited breast cancer MDA-MB-231 cell migration and invasion via suppression of FGFR1 phosphorylation and its downstream signaling pathway. Suppression of matrix metalloproteinases 2/9 (MMP2/9) was associated with the anti-motility activity of C11. Furthermore, the anti-angiogenesis activity of C11 was verified in endothelial cells and chicken chorioallantoic membranes (CAMs). C11 inhibited the migration and tube formation of HMEC-1 endothelial cells and inhibited angiogenesis in a CAM assay. In sum, C11 is a novel selective FGFR1 inhibitor that exhibits potent activity against breast cancer metastasis and angiogenesis. Keywords: C11; FGFR1 inhibitor; antitumor; breast cancer; metastasis; angiogenesis Acta Pharmacologica Sinica (2019) 40:823–832; https://doi.org/10.1038/s41401-018-0191-7 INTRODUCTION typically inhibit a broad range of additional kinases (e.g., VEGFRs The fibroblast growth factor receptor (FGFR) family comprises 4 and PDGFRs), and approved multitarget FGFR TKIs examined in members: FGFR1–4[1].
    [Show full text]
  • An Angiogenesis Inhibitor
    Proc. Natl. Acad. Sci. USA Vol. 77, No. 7, pp. 4331-4335, July 1980 Medical Sciences Control of tumor growth in animals by infusion of an angiogenesis inhibitor (cartilage/neovascularization/antiangiogenesis/B16 melanoma/V2 carcinoma) ROBERT LANGER*tI, HOWARD CONN*, JOSEPH VACANTI*, CHRISTIAN HAUDENSCHILD§, AND JUDAH FOLKMAN*¶ *Department of Surgery, Children's Hospital Medical Center and lHarvard Medical School, Boston, Massachusetts 02115; tDepartment of Nutrition and Food Science, Massachusetts Institute of Technology, Boston, Massachusetts 02139; and the WMallory Institute of Pathology, Boston University School of Medicine, Boston, Massachusetts 02118 Communicated by Earl P. Benditt, April 21, 1980 ABSTRACT Angiogenesis and tumor growth were inhibited distance (approximately 1.0 mm) by implanting the tumor i in two different animal models by regional infusion of a par- the rabbit cornea. This system permitted daily measurements tially purified cartilage extract. In rabbits bearing corneal im- demonstrated that vascular plants of V2 carcinoma and receiving the inhibitor, vascular of vessel growth rate and prolifer- growth rates were <3% of those in control animals receiving ation was inhibited and, subsequently, tumor growth was either Ringer's solution or bovine trypsin inhibitor (Trasylol). controlled. Subconjunctival B16 melanoma implants-in mice receiving the In a second set of experiments, the tumor (B16 melanoma) inhibitor weighed <2.5% of implants in mice receiving Ringer's was implanted directly onto a dense capillary bed in the mouse solution, Trasylol, or albumin. Histplogic study of major organs conjunctiva. This contiguous relationship between tumor and and standard blood tests revealed no toxic effects in any ofthe vascular bed more closely resembles most human and animal animals.
    [Show full text]
  • Increased Endostatin/Collagen XVIII Expression Correlates with Elevated VEGF Level and Poor Prognosis in Hepatocellular Carcinoma
    Modern Pathology (2005) 18, 663–672 & 2005 USCAP, Inc All rights reserved 0893-3952/05 $30.00 www.modernpathology.org Increased endostatin/collagen XVIII expression correlates with elevated VEGF level and poor prognosis in hepatocellular carcinoma Tsung-Hui Hu1,*, Chao-Cheng Huang2,*, Chia-Ling Wu3, Pey-Ru Lin4, Shang-Yun Liu2, Jui-Wei Lin2, Jiin-Haur Chuang3 and Ming Hong Tai4,5 1Division of Hepatology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; 2Division of Pathology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; 3Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; 4Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan and 5Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan Liver is the primary source for collagen XVIII, the precursor of angiogenesis inhibitor, endostatin. However, the role of endostatin/collagen XVIII expression during liver carcinogenesis remains elusive. Therefore, we studied its expression in five hepatoma cell lines and 105 hepatocellular carcinoma specimens. The poorly differentiated hepatoma cell lines exhibited increased endostatin/collagen XVIII levels compared with the well-differentiated ones. In hepatoma tissues, endostatin/collagen XVIII expression was detected in various types of liver cells and was significantly stronger in adjacent nontumor tissues than that in tumors (Po0.001). Endostatin/collagen XVIII expression in nontumor tissues correlated with tumor stages (P ¼ 0.014) and expression of vascular endothelial growth factor (P ¼ 0.007), but not the stages of hepatic fibrosis (P40.05). Kaplan–Meier analysis showed that patients with higher endostatin/collagen XVIII expression had significantly shorter overall survival (P ¼ 0.011) and disease-free survival (P ¼ 0.0034).
    [Show full text]
  • Angiogenesis Inhibitor Induced Thromboembolism in Cancer Patients Neeharik Mareedu and Carmen P Escalante*
    ISSN: 2378-3419 Mareedu and Escalante. Int J Cancer Clin Res 2017, 4:080 DOI: 10.23937/2378-3419/1410080 Volume 4 | Issue 1 International Journal of Open Access Cancer and Clinical Research REVIEW ARTICLE Angiogenesis Inhibitor Induced Thromboembolism in Cancer Patients Neeharik Mareedu and Carmen P Escalante* Department of General Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA *Corresponding author: Carmen P Escalante MD, Professor and Chair, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, 1400 Pressler St. Unit 1465 Houston, TX 77030-4004, USA, Tel: 713-792- 2148, Fax: 713-792-0365, E-mail: [email protected] and with fewer side effects when compared to conven- Abstract tional agents. Therapy with Angiogenesis Inhibitors (AIs) is a newer tar- geted approach in treating cancers. It gained popularity in Angiogenesis Inhibitors (AIs) are one of the classes of the past decade because of better outcomes and fewer side targeted therapy being used in the treatment of various effects when compared to conventional chemotherapeutic agents. These agents act by inhibiting the VEGF pathway cancers. Adverse effects profile of AIs is completely dif- and inhibit vasculogenesis, thus shutting off the nutrient ferent when compared to traditional chemotherapeu- supply to the continuously multiplying tumor cells. Growth tic agents owing to their different mechanism of action of blood vessels is a dynamic process in the human body on tumor cells. Due to more promising results with AIs, which is required for the various other processes through- an increasing number of oncologists are utilizing these out the human life.
    [Show full text]
  • Binding and Endothelial Tube Formation
    Proc. Natl. Acad. Sci. USA Vol. 95, pp. 7275–7280, June 1998 Biochemistry Defining the domains of type I collagen involved in heparin- binding and endothelial tube formation SHAWN M. SWEENEY†,CYNTHIA A. GUY‡,GREGG B. FIELDS§, AND JAMES D. SAN ANTONIO†¶ †Department of Medicine and the Cardeza Foundation for Hematologic Research, Jefferson Medical College of Thomas Jefferson University, Philadelphia, PA 19107; ‡Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455; and §Department of Chemistry and Biochemistry, and the Center for Molecular Biology and Biotechnology, Florida Atlantic University, Boca Raton, FL 33431 Edited by Darwin J. Prockop, MCP-Hahnemann Medical School, Philadelphia, PA, and approved April 14, 1998 (received for review January 12, 1998) ABSTRACT Cell surface heparan sulfate proteoglycan To localize more precisely the heparin-binding regions on type (HSPG) interactions with type I collagen may be a ubiquitous I collagen, we studied complexes between collagen monomers cell adhesion mechanism. However, the HSPG binding sites on and heparin–albumin–gold particles by electron microscopy type I collagen are unknown. Previously we mapped heparin (6), and we observed heparin binding primarily to a region on binding to the vicinity of the type I collagen N terminus by the triple helix near the procollagen N terminus. In collagen electron microscopy. The present study has identified type I fibrils, heparin–gold bound to the a bands region within each collagen sequences used for heparin binding and endothelial D-period, which is consistent with an N-terminal heparin- cell–collagen interactions. Using affinity coelectrophoresis, binding site on its monomers. The resolution of the mapping we found heparin to bind as follows: to type I collagen with technique was insufficient to assign heparin-binding function high affinity (Kd ' 150 nM); triple-helical peptides (THPs) to any particular protein sequence.
    [Show full text]