Antiangiogenic mechanisms and factors in breast cancer treatment


Jessica M Castañeda-Gill1, Jamboor K Vishwanatha2
1 Department of Molecular and Medical Genetics, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
2 Department of Molecular and Medical Genetics, Graduate School of Biomedical Sciences; Institute for Cancer Research?, Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX, USA

Date of Submission 21-Dec-2015
Date of Acceptance 23-Jan-2016
Date of Web Publication 12-Feb-2016

 

Correspondence Address:
Jamboor K Vishwanatha
Department of Molecular and Medical Genetics, Graduate School of Biomedical Sciences; Institute for Cancer Research?, Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX
USA

Source of Support: None, Conflict of Interest: None

DOI: 10.4103/1477-3163.176223

Abstract

Breast cancer is known to metastasize in its latter stages of existence. The different angiogenic mechanisms and factors that allow for its progression are reviewed in this article. Understanding these mechanisms and factors will allow researchers to design drugs to inhibit angiogenic behaviors and control the rate of tumor growth.

Keywords: Angiogenesis, breast cancer, metastasis, therapy, tumor vasculature

How to cite this article:
Castañeda-Gill JM, Vishwanatha JK. Antiangiogenic mechanisms and factors in breast cancer treatment. J Carcinog 2016;15:1

How to cite this URL:
Castañeda-Gill JM, Vishwanatha JK. Antiangiogenic mechanisms and factors in breast cancer treatment. J Carcinog [serial online] 2016 [cited 2021 Oct 13];15:1. Available from: https://carcinogenesis.com/text.asp?2016/15/1/1/176223

 Introduction

The human body has the ability to form new blood vessels from existing vessels to ensure proper vascularization during embryonic growth, reproductive cycles, and proper wound healing. Vascularization from preexisting blood vessels is termed angiogenesis. While angiogenesis is beneficial in most processes, it is also what maintains tumor growth and malignancy. Cancer has the ability to grow and metastasize if ample vascularization is successfully maintained; hence, hindering cancer growth via anti-angiogenic mechanisms has generated research interest. To properly understand the anti-angiogenic mechanisms, it is imperative that pro-angiogenic mechanisms and growth factors that induce these processes be studied in detail.

Cancer cells need ample blood supply to enable metastatic processes including angiogenic factor induction and signaling, growth stimulating factor induction and signaling, enabling proteolytic enzymes, expressing cell adhesion proteins, and exerting resistance to immunogenic molecules. [1] To achieve the metastatic behavior, growth factors and other such molecules are released in order to induce nondifferentiated stromal cells to the site of tumor growth. These cells, also known as mesenchymal stem cells (MSCs), enable tumor growth by providing tumor cells with the necessary signals. MSCs, which are located in the bone marrow and can undergo osteogenesis; chondrogenesis; and adipogenesis if needed, are shown to increase breast cancer progression and metastasis by allowing cell motility via chemokine secretion. [2] Furthermore, MSCs have been shown to secrete vascular endothelial growth factor-A (VEGF-A) and interleukin-6 (IL), which are potent angiogenic factors in breast cancer growth. [3]

Angiogenic Mechanism

Understanding angiogenic mechanisms and studying the different factors which bring about healthy angiogenesis can allow researchers to inhibit angiogenic behaviors, which in turn can lead to a better awareness of how to induce tumor suppression. Through these studies, we can determine the role of angiogenesis and its impact on breast cancer tumor progression.

Distinguishing between regular angiogenesis and tumor-associated angiogenic processes is imperative for effectively treating breast cancer. In humans, regular angiogenesis can be defined as the integration of endothelial cell precursors that form capillary plexus and later become blood vessels. Some processes in which normal angiogenesis occurs include embryo nutrition, tissue repair, and physiological changes associated with growth. During these common processes, there is an intricate balance between pro- and anti-angiogenic signals that is rigorously maintained, in order for newly formed blood vessels to attain maturity and stability in a timely manner. [4] However, in tumor-associated angiogenesis, the balance between these angiogenic factors is lost, allowing blood vessels to develop uncontrollably. This negative shift leads to changes in normal vasculature characteristics including their physical properties. [4],[5]

Tumor Vasculature and Angiogenesis

Tumor vasculature is markedly distinct from normal vasculature in that blood vessels that supply tumor tissue are irregularly sized and arranged in a disorganized manner, where they share characteristics of arterioles, capillaries, and venules simultaneously. In normal tissue vessels, blood flow and density are controlled by the tissue’s metabolic needs to avoid over-feeding or under-feeding its cells. [6],[7] However, in tumor vasculature, sporadic blood flow is observed, leading to damaged capillary network systems. [7],[8]

Tumors are composed of various cellular components that allow for effective angiogenic growth. First, functional vasculature contains adipose tissue that is surrounded by stromal cells that provide a proper framework for the developing tumor’s vascular network. White adipose tissue (WAT) maintains vascular growth and has been shown to aid breast cancer development and progression in mouse models, [9] while brown adipose tissue (BAT) enables metabolic processes, such as efficient oxygen and nutrient transport that aids tumor growth. [10] WAT and BAT are both known to be in charge of producing angiogenic factors, with the most common ones in relation to breast cancer being VEGF A, B, and C; basic fibroblast growth factor (bFGF)/FGF-2; matrix metalloproteinases (MMPs); and IL-8. [10],[11] The release of these factors is strictly dependent upon the pro-versus antiangiogenic balance at the adipose tissue pad that is in proximity to the tumor tissue. [10]

Another element responsible for tumor vasculature formation and angiogenesis is vascular endothelial cells, which are recruited by growth factors such as VEGF and bFGF/FGF-2 to propagate, migrate and form tube-like structures. [12],[13] Endothelial precursor cells migrate from the bone marrow into the bloodstream, where they eventually settle into a niche and begin forming new blood vessels, with the help of VEGF, FGF, and platelet-derived growth factor. [7],[14],[15],[16] It is these newly formed blood vessels that provide vascular networks for the burgeoning tumor to grow, eliminate waste from its rapidly dividing cells, and allow metastasis. [17]

The earliest stage of tumor angiogenesis occurs when VEGFs cause vasodilation of existing capillaries that enables diffused plasma proteins to lay down a matrix for endothelial cells to migrate to and loosen the smooth pericyte covering of the blood vessel. This process is aided by the involvement of the tyrosine kinase receptor 2 (TIE2) and one of its ligands, angiopoietin-2 (ANG-2). [18] The vascular membrane and matrix are then degraded in order for endothelial cells to gain entrance into the lumenal space and migrate toward the pro-angiogenic stimuli. In normal angiogenic endothelial migration, pericyte processes decrease cell proliferation and decrease VEGF dependence. [13] This behavior is unseen in tumor tissue angiogenesis, where the pericyte influence on endothelial proliferation is curtailed or sometimes nonexistent. As is the case of VEGF-A inhibition by blood vessels, other cytokines such as ANG-1 and placental growth factor (PlGF) are known to send growth signals to endothelial cells. [4],[7],[19],[20] In addition, VEGF can interact with the tumor via different pathways such as PI3K/AKT and mitogen-activated protein kinase (MAPK), [21],[22] as well as express an E-cadherin repressor that is involved in breast cancer cell invasiveness. [23]

Angiogenic Factors and Their Roles in Breast Cancer Progression and Treatment

Tumor cells establish angiogenesis by secreting various angiogenic factors. The most studied and understood of these factors are VEGF and IL-8. VEGF and IL-8 secretion are especially apparent in breast cancer angiogenesis, [19],[22],[24],[25],[26],[27],[28] in addition to bFGF/FGF-2 and MMPs. Current research shows that tumor angiogenesis requires a combination of the aforementioned factors, if not all of them, in order for a tumor to efficiently undergo development and potential metastasis. [4],[29],[30]

Vascular endothelial growth factors

While VEGFs are most known for their role in endothelial cell proliferation and migration, their expression has been observed in macrophages; epidermal cells; thrombocytes; and tumor cells, with involvement in normal physiological processes such as development; osteogenesis; and wound healing, in addition to numerous pathologies. [31],[32],[33],[34] Five mammalian VEGF ligands exist, VEGF-A/B/C/D and PlGF, which can interact with three different VEGF receptors (VEGFR), VEGFR-1/2/3. [32],[35],[36],[37] Of the VEGF ligand-receptor interactions, the ones most involved in breast cancer development, progression, and metastasis are VEGF-A and VEGFR-1 or 2. [35],[36],[37],[38],[39],[40],[41],[42]

In tumor angiogenesis, each VEGF family member plays some role, either directly or indirectly, to enhance pathological progression. As previously stated, the most expressed VEGF member, VEGF-A, stimulates angiogenesis most potently via its interaction with VEGFR-2, [32],[33],[39],[40],[41],[43] even though it interacts with a higher affinity with VEGFR-1. VEGFR-1 can act as a decoy receptor that can control VEGF-A interaction with, and activation of, VEGFR-2, and ultimately impact angiogenesis and its involvement in tumor development. [32],[33],[37],[43]

VEGF-B, which is highly expressed in the brain and skeletal system, as well as breast tumors, promotes angiogenesis indirectly by binding VEGFR-1 on endothelial cells in order to initiate plasminogen activation, and eventually, metastasis. [17],[33],[34],[35],[42],[44],[45],[46] VEGF-C promotes angiogenesis and lymphangiogenesis via still unknown mechanisms through its binding with VEGFR-2 and VEGFR-3, respectively, while VEGF-D is known to ensure lymphatic vessel growth. [17],[33],[34],[35],[42] High overall VEGF expression levels are associated with breast cancer aggressiveness and metastasis, as well as poor treatment responsiveness. [36],[47]

Finally, PlGF, which only binds and activates VEGFR-1, is primarily known for its role in embryogenesis; however, recent studies have demonstrated its involvement in cancer development via its pro-inflammatory, pro-angiogenic actions that aid metastasis. [35],[48],[49]

Interleukins

ILs are a class of cytokines secreted by cells in response to various stimuli, in order to initiate the immune response to a pathological condition. IL-8, a member of the IL class, is produced by a variety of cells, including endothelial and tumor cells, in order to activate its receptors, CXCR1 and 2, and potentiate signaling pathways that can relate to cell migration and mobilization and angiogenesis, which are important for breast cancer progression and metastasis. [22],[26],[27],[28],[50],[51] Secretion of IL-8 is elicited in response to the presence of several chemokines and growth factors, including VEGF and MMPs, to facilitate the aforementioned physiological processes. [4],[28] Studies have shown that tumors, including breast carcinomas, expressing high levels of IL-8 are found to be more aggressive and invasive, and less responsive to traditional treatment protocols, making it a target to future antiangiogenic therapies. [4],[22],[26],[27],[51],[52]

Fibroblast growth factors

In addition to VEGF, FGFs are also known to be a family of potent angiogenic motivators with an association with breast cancer risk. [53] While FGF-1 is known as the acidic polypeptide, FGF-2 is the bFGF polypeptide and has been shown to aid in proliferation and differentiation of endothelial cells. [54],[55]

The bFGF/FGF-2 protein is known to exist in five isoforms as a result of multiple polyadenylation sites and altered mRNA translation, where they are categorized into low or high molecular weight forms. [56],[57],[58] The 18 kDa bFGF/FGF-2, which is the most common FGF, interacts with all four high-affinity FGF-receptors (FGFRs), but specifically FGFR-1 and FGFR-2-IIIC, with the help of heparan sulfate proteoglycans that are located on the cell surface. [59],[60],[61],[62] bFGF/FGF-2 activation is thought to occur following the degradation of heparan sulfate within the extracellular matrix (ECM), which leads to sprouting of new blood vessels. [63],[64] Molecules in the extracellular environment, such as heparan sulfate proteoglycans; integrin receptors; ECM proteins; cytokines; and serum components, can alter FGF-2/FGFR-2 interactions, which can enhance tumor progression and metastasis through regulation of angiogenesis, although less so than the aforementioned growth factors. [55],[59],[61],[62],[65]

Matrix metalloproteinases

Finally, a class of proteolytic enzymes called MMPs, is involved in angiogenesis through their ability to remodel the ECM. Some of the initial steps of angiogenic blood vessel formation, which include destabilization of the established blood vessel wall and degradation of matrix proteins in order for mobilization and migration of endothelial cells, are performed by MMPs. [66],[67],[68],[69],[70],[71],[72],[73],[74],[75] MMPs, which are divided into at least five categories based on the matrix substrates they destabilize-collagenases (MMP-1, MMP-8, and MMP-13), gelatinases (MMP-2 and MMP-9), stromelysins (MMP-3, MMP-10, and MMP-11), matrilysins (MMP-7), and elastase (MMP-12) are initially secreted as inactive zymogens and then activated extracellularly by other proteolytic enzymes. [66],[67],[68],[71],[73],[75] As a result, MMPs are currently being studied for their role in breast cancer invasion and metastasis. [67],[71],[72],[73],[76],[77],[78] MMPs are typically regulated by a group of natural inhibitors, known as tissue inhibitors of metalloproteinases, which are being investigated for their contribution to cancer development and as potential treatments. [4],[67],[73],[75],[79]

Examples of Angiogenic Inhibitors and their Modes of Action

Numerous antiangiogenic compounds exist in various phases of development to clinical practice, both endogenous and exogenous; however, this section will examine a select few of the most commonly studied inhibitors including interferons-α/β, endostatin, angiostatin, thrombospondin (TSP), and decorin.

Interferons-α/β

Interferons-α/β are types of class I interferons that regulate the immune system and control cell growth and differentiation, two important aspects of tumor formation. During the last two decades, research on interferons-α/β has shown that they have the ability to impair tumor angiogenesis via downregulation of mRNA expression of the growth factor bFGF/FGF-2 and reduced expression of the MMP-2 gene, [80],[81] making their development as potential antiangiogenic therapies crucial. [82]

Endostatin

Endostatin is a naturally occurring fragment of collagen XVIII generated by tumor cell proteases that inhibits endothelial cell proliferation and migration. [83],[84],[85],[86] Endostatin is thought to have several possible mechanisms of action in relation to the inhibition of tumor angiogenesis including prevention of tumor necrosis factor alpha (TNFα) activation of the JNK signaling pathway, [87] antagonism of the cell surface receptors a v – and a 5 -integrins, [86] and inhibition of cell cycle progression of endothelial cells. [85] The JNK signaling pathways, c-Jun NH2-terminal kinase, are activated by different types of cellular stresses and signals and utilizes TNFα to elicit cell death. [87]

As previously stated, endostatin inhibits TNF activation of JNK and pro-angiogenic genes dependent on it. [87] In reference to the cell cycle, endostatin causes G1 arrest by decreasing the phosphorylated state of retinoblastoma genes and cyclin D1 mRNA and protein expression. [88] While the protein itself has not shown any direct cytotoxicity in tumor cells, gene transfer has been hypothesized as a means of delivery in order to induce apoptosis and ultimately tumor regression/inhibition, [82],[85] which has proven successful in a breast cancer study that exhibited a 90% reduction in tumor growth compared to the untreated control. [89]

Angiostatin

As with endostatin, angiostatin is a proteolytic fragment of a larger protein, specifically, plasminogen, that acts as a metastatic suppressor by blocking the formation of blood vessels and is thought to impair tumor progression. [82],[85],[90],[91],[92] One mechanism by which angiostatin, whose receptors include adenosine triphosphate (ATP) synthase and integrin αvβ3 , is hypothesized to inhibit tumor growth is via the binding of ATP synthase at its catalytic subunit. [93] As a result, ATP synthesis is terminated, leading to inhibition of the cells’ uncontrolled proliferation. [92],[93]

In addition, prolonged angiostatin treatment has also been shown to inhibit the activation of the MAPK, extracellular-signal-regulated kinases-1 (ERK1) and ERK2, by FGF-2 or VEGF in human skin vascular cells. [94] The MAPK/ERK pathway functions in cellular proliferation, differentiation, and even survival following phosphorylation of specific threonine and tyrosine residues. [94] ERKs regulate growth factor-responsive targets in the cytosol and are also able to translocate to the nucleus, where they phosphorylate a number of transcription factors that regulate gene expression. The role of FGF-2, which is to stimulate phosphorylation of ERK-1 and 2, is blocked by angiostatin; [89],[94] this leads to a loss of integrity of the cell cycle and eventually impairment of angiogenesis.

Finally, other potential mechanisms of action by which angiostatin inhibits angiogenesis are through suppression of VEGF activities and arrest of the cell cycle at the G2-to-M transition. [89] As with endostatin, angiostatin has been considered as an antiangiogenic gene therapy due to issues with delivery and treatment with its protein form. [82],[85],[95]

Thrombospondin

TSPs are a family of five matricellular glycoproteins involved in cell proliferation, migration, and survival through their interactions with numerous cell surface and ECM proteins, indicating their importance in angiogenesis. Significant research shows that TSPs, which are identified in breast cancer, can act as potent endogenous antiangiogenic factors that inhibit the aforementioned cellular processes within endothelial cells, leading to tumor suppression. [96],[97],[98],[99],[101]

The most studied of the TSPs is TSP-1, which binds αvβ3 integrins, heparin sulfate proteoglycans, transforming growth factor-β (TGF-β), and other ECM proteins and proteases, and inversely regulates proliferation and migration of vascular smooth muscle and endothelial cells. [100.101.102] Through its various peptide binding sequences, TSP-1 acts as a tumor suppressor by inhibiting bFGF/FGF-2 and VEGF through binding heparin sulfate receptors on the cell surface and ECM or inhibiting MMP-9 activation, inhibiting angiogenesis via its interaction with the RGD sequence on αvβ3 integrins, inhibiting tumor growth through its binding and activation of TGF-β, and inducing apoptosis by inhibiting MMP-9 activation. [100],[102],[102],[103],[104],[105],[106],[107],[108],[109] TSP-2, which is structurally similar to TSP-1, demonstrates similar antiangiogenic, antitumor capabilities making it another option for drug development. [110],[111]

Decorin

Decorin is a member of a family of small leucine-rich proteoglycans involved in various cellular processes including matrix organization, formation/”decoration” of collagen fibrils, wound healing, and maintenance of cell proliferation through its interaction with other ECM proteins and growth factors. [112],[113],[114],[115],[116],[117],[118],[119],[120],[121],[122] Decorin, which is secreted by mesenchymal cells; connective tissue cells; and tumor stromal cells, inhibits tumor angiogenesis; progression; and metastasis through its interactions with such receptors and proteins as EGFRs; TGF-β; VEGF and VEGFR-2; and bFGF/FGF-2 making it a potential anticancer treatment. [116],[117],[119],[121],[122],[123],[124],[125],[126],[127],[128],[129],[130],[131],[132],[133],[134],[135],[136],[137],[138],[139],[140],[141],[142],[143],[144],[145],[146],[147],[148],[149],[150],[151],[152],[153],[154],[155],[156]

While one of decorin’s mechanisms of angiogenic and growth inhibition may involve the potential impeding of epidermal growth factor receptors (EGFRs) interaction with EGF due to an overlap in binding sites, [145] it is known that its interaction with EGFR leads to MAPK activation, utilization of intracellular calcium, upregulation of the cell cycle cyclin-dependent kinase inhibitor, p21, and a drastic reduction in EGFR activity, in addition to internalization and lysosomal degradation of the receptor. [116],[117],[119],[121],[122],[132],[134],[135],[140],[141],[142],[144],[146],[148],[157] In the case of breast cancer, which exhibits high EGFR expression, specifically ErbB2 (human epidermal growth factor receptor 2), decorin inhibits the receptor’s synthesis and activity, while reducing cell growth and migration, leading to a reduction in tumor size at the primary site, as well as the development and expansion of metastases within the lungs. [117],[119],[121],[122],[141],[144],[151],[154]

TGF-β is a growth factor secreted by most cells, including healthy and cancerous breast tissue that is multifaceted in its involvement in all aspects of cell survival and death. Depending on the stage of breast cancer development and progression that the growth factor is present, it can act as either a pro- or anti-oncogenic protein. In the case of late stage, aggressive, metastatic breast cancers, TGF-β is highly oncogenic making it a frequent target for therapies. In the presence of decorin binding, TGF-β, and subsequently, its receptor’s activity is impaired leading to tumor suppression. [119],[121],[122],[125],[127],[128],[130],[137] Decorin is also known to inhibit tumor angiogenesis and growth through its negative interactions with VEGF and its receptor and bFGF/FGF-2 by downregulating VEGF and bFGF/FGF-2 expression and activities, and by binding VEGFR-2 to impair its interaction with, and ultimate activation by, VEGF. [118],[121],[122],[136],[149],[155],[156]

Clinical Trials for Antiangiogenic Treatments in Breast Cancer

Numerous therapies to combat angiogenesis in breast cancer have passed from clinical trials to use with varying levels of success; however, they can all be divided into different categories based on what aspect of tumor angiogenesis they affect, including those that inhibit pro-angiogenic growth factors and/or their receptors (such as VEGF-A and VEGFR-1 and -2), ECM proteins (such as integrins), endothelial cell quantitative and spatial expansion/spreading, and proteases (such as MMPs). [17],[24],[158],[159],[160] A few examples of drugs that have undergone clinical investigation, and their mechanisms of action are described here.

Bevacizumab

Bevacizumab, also called Avastin, is a humanized monoclonal antibody that binds VEGF-A and impairs its activity and interactions with VEGFR-1 and -2, leading to a reduction in tumor growth. [33],[47],[158],[159],[161],[162],[163],[164],[165] As the first US Food and Drug Administration (FDA) approved antiangiogenic, it was utilized as a combination treatment with other chemotherapeutics for metastatic colon cancer, [166] with its single and combinatorial use expanded to nonsmall cell lung cancer, renal cell carcinoma, pancreatic cancer, ovarian cancer, advanced kidney cancer, glioma, leukemia, and breast cancer. [19],[47],[158],[159],[160],[167],[168],[169],[170],[171],[172],[173],[174],[175],[176],[177],[178],[179],[180],[181],[182],[183],[184],[185],[186],[187] In a phase II study testing only bevacizumab in 75 patients with metastatic breast cancer patients that had received treatment in the past, only 9.3% responded effectively with four being progression-free beyond a year. [158],[168] While treatment with bevacizumab alone or in combination with traditional chemotherapy was well-tolerated, it produced a range of side effects including bleeding and blood clots, abnormal excreted protein levels, and high blood pressure. [158],[167] Since completion of the phase II Cobleigh study (2003), additional phase II and phase III trials utilizing bevacizumab in combination with capecitabine demonstrated improvements in response rates, but no noticeable changes in progression-free or overall survivals rates, while its use with taxanes; cyclophosphamide; doxorubicin; and gemcitabine resulted in extended progression-free survival lengths. [154],[160],[187],[188],[189],[190],[191],[192],[193],[194],[195] While a majority of these studies were performed in later stage, metastatic breast cancer patients, other trials completed in individuals diagnosed with early breast cancer also exhibited increased response rates when compared to those receiving standard care protocols. [160],[196],[197] As November 2011, bevacizumab’s FDA approval for use as a metastatic breast cancer therapy was removed due to lack of significant improvements in response and survival; however, it continues to be investigated in various breast cancer treatment trials and prescribed in clinical practice. [198]

Cilengitide

Cilengitide is classified as an antiangiogenic drug due to its ability to inhibit the cellular adhesion receptors αvβ3 /αvβ5 integrins. [107],[164],[199],[200] Cilengitide has shown effectiveness in preclinical studies through its induction of cell detachment and reduced proliferation in a panel of four breast cancer cell lines, as well as enhanced treatment effectiveness and cell death and inhibited bone metastasis that was furthered when combined with radiotherapy when utilized in breast cancer xenografts. [200],[201],[202] Cilengitide has been investigated in glioblastoma, nonsmall cell lung cancer, and prostate cancer with noted tumor reduction and negligible toxicity; however, overall survival was not significantly increased compared to currently-available treatments. [203],[204],[205],[206] Currently, a phase I trial is underway to evaluate cilengitide as a combination therapy with paclitaxel in patients with advanced solid tumors, particularly those diagnosed with high-grade, aggressive breast cancers. [207]

Vitaxin

Vitaxin, also referred to as Abegrin, is a humanized monoclonal antibody that affects the vitronectin receptor, αvβ3 integrin, leading to impaired endothelial cell expansion and tumor growth. [158],[208],[209] Previous clinical studies have been performed with Vitaxin, but showed little results, leading to its modification to Abegrin. [209] In a small scale, phase II study investigating Abegrin’s effects in various metastatic cancer patients that had received prior treatment, including one individual breast cancer, the drug was well tolerated but did not produce clinically relevant results, possibly due to its specificity for αvβ3[208]

Conclusions and Future Prospective

Despite the fact that angiogenesis is a naturally occurring process that is necessary for activities ranging from embryonic development to wound healing, it also possesses detrimental aspects that are involved in disease occurrence and progression. When the angiogenic balance is disturbed and various signaling pathways are exploited, uncontrolled cell growth occurs, which can result in the propagation of cancer. Through the use of natural inhibitors such as endostatin, angiostatin, TSP, and decorin, the likelihood of tumor progression and metastasis are reduced, though not eliminated. Although most antiangiogenics have proven poorly effective against breast cancer, their development and investigation in clinical trials have provided a basis for identifying drug targets that can impair tumor angiogenesis, and potentially tumor growth and migration. While more effective treatments to combat tumor angiogenesis appear to be further in the future, continued investigation is necessary to establish potential new therapies, as well as to determine the possibility to repurpose old drugs.

Financial support and sponsorship

Funding for this publication was made possible (in part) by 5P20MD006882 and 1 R13 MD009663-01 from the NIMHD. The views expressed in this publication do not necessarily reflect the official policies of the Department of Health and Human Services; nor does mention by trade names, commercial practices, or organizations imply endorsement by the United States Government.

Conflicts of interest

There are no conflicts of interest.


References

1.
Boyce BF, Yoneda T, Guise TA. Factors regulating the growth of metastatic cancer in bone. Endocr Relat Cancer 1999;6:333-47.  Back to cited text no. 1
    
2.
Deans RJ, Moseley AB. Mesenchymal stem cells: Biology and potential clinical uses. Exp Hematol 2000;28:875-84.  Back to cited text no. 2
    
3.
De Luca A, Gallo M, Aldinucci D, Ribatti D, Lamura L, D′Alessio A, et al. Role of the EGFR ligand/receptor system in the secretion of angiogenic factors in mesenchymal stem cells. J Cell Physiol 2011;226:2131-8.  Back to cited text no. 3
    
4.
Papetti M, Herman IM. Mechanisms of normal and tumor-derived angiogenesis. Am J Physiol Cell Physiol 2002;282:C947-70.  Back to cited text no. 4
    
5.
Folberg R, Hendrix MJ, Maniotis AJ. Vasculogenic mimicry and tumor angiogenesis. Am J Pathol 2000;156:361-81.  Back to cited text no. 5
    
6.
Siemann DW. The unique characteristics of tumor vasculature and preclinical evidence for its selective disruption by tumor-vascular disrupting agents. Cancer Treat Rev 2011;37:63-74.  Back to cited text no. 6
    
7.
Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nat Rev Cancer 2003;3:401-10.  Back to cited text no. 7
    
8.
Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 1996;86:353-64.  Back to cited text no. 8
    
9.
Bertolini F, Petit JY, Kolonin MG. Stem cells from adipose tissue and breast cancer: Hype, risks and hope. Br J Cancer 2015;112:419-23.  Back to cited text no. 9
    
10.
Christiaens V, Lijnen HR. Angiogenesis and development of adipose tissue. Mol Cell Endocrinol 2010;318:2-9.  Back to cited text no. 10
    
11.
Yoshida S, Ono M, Shono T, Izumi H, Ishibashi T, Suzuki H, et al. Involvement of interleukin-8, vascular endothelial growth factor, and basic fibroblast growth factor in tumor necrosis factor alpha-dependent angiogenesis. Mol Cell Biol 1997;17:4015-23.  Back to cited text no. 11
    
12.
Ke LD, Shi YX, Im SA, Chen X, Yung WK. The relevance of cell proliferation, vascular endothelial growth factor, and basic fibroblast growth factor production to angiogenesis and tumorigenicity in human glioma cell lines. Clin Cancer Res 2000;6:2562-72.  Back to cited text no. 12
    
13.
Lamalice L, Le Boeuf F, Huot J. Endothelial cell migration during angiogenesis. Circ Res 2007;100:782-94.  Back to cited text no. 13
    
14.
Aghi M, Chiocca EA. Contribution of bone marrow-derived cells to blood vessels in ischemic tissues and tumors. Mol Ther 2005;12:994-1005.  Back to cited text no. 14
    
15.
Larrivée B, Karsan A. Involvement of marrow-derived endothelial cells in vascularization. In: Kauser K, Zeiher AM, editors. Bone Marrow-Derived Progenitors. Berlin, Germany: Springer Science & Business Media; 2007. p. 102-5.  Back to cited text no. 15
    
16.
Liu M, Yang L, Guo S, Gonzalez-Perez RR. Endothelial and cancer stem cell regulation of tumor angiogenesis. In: Gonzalez-Perez RR, Rueda BR, editors. Tumor Angiogenesis Regulators. Boca Raton, FL: CRC Press; 2013. p. 99-101.  Back to cited text no. 16
    
17.
Nishida N, Yano H, Nishida T, Kamura T, Kojiro M. Angiogenesis in cancer. Vasc Health Risk Manag 2006;2:213-9.  Back to cited text no. 17
    
18.
Holash J, Wiegand SJ, Yancopoulos GD. New model of tumor angiogenesis: Dynamic balance between vessel regression & growth mediated by angiopoietins and VEGF. Oncogene 1999;18:5356-62.  Back to cited text no. 18
    
19.
Pang RW, Poon RT. Clinical implications of angiogenesis in cancers. Vasc Health Risk Manag 2006;2:97-108.  Back to cited text no. 19
    
20.
Carmeliet P, Moons L, Luttun A, Vincenti V, Compernolle V, De Mol M, et al. Synergism between vascular endothelial growth factor and placental growth factor contributes to angiogenesis and plasma extravasation in pathological conditions. Nat Med 2001;7:575-83.  Back to cited text no. 20
    
21.
Price DJ, Miralem T, Jiang S, Steinberg R, Avraham H. Role of vascular endothelial growth factor in the stimulation of cellular invasion and signaling of breast cancer cells. Cell Growth Differ 2001;12:129-35.  Back to cited text no. 21
    
22.
Chelouche-Lev D, Miller CP, Tellez C, Ruiz M, Bar-Eli M, Price JE. Different signalling pathways regulate VEGF and IL-8 expression in breast cancer: Implications for therapy. Eur J Cancer 2004;40:2509-18.  Back to cited text no. 22
    
23.
Wanami LS, Chen HY, Peiró S, García de Herreros A, Bachelder RE. Vascular endothelial growth factor-A stimulates Snail expression in breast tumor cells: Implications for tumor progression. Exp Cell Res 2008;314:2448-53.  Back to cited text no. 23
    
24.
Gasparini G, Harris AL. Clinical importance of the determination of tumor angiogenesis in breast carcinoma: Much more than a new prognostic tool. J Clin Oncol 1995;13:765-82.  Back to cited text no. 24
    
25.
Toi M, Inada K, Suzuki H, Tominaga T. Tumor angiogenesis in breast cancer: Its importance as a prognostic indicator and the association with vascular endothelial growth factor expression. Breast Cancer Res Treat 1995;36:193-204.  Back to cited text no. 25
    
26.
Lin Y, Huang R, Chen L, Li S, Shi Q, Jordan C, et al. Identification of interleukin-8 as estrogen receptor-regulated factor involved in breast cancer invasion and angiogenesis by protein arrays. Int J Cancer 2004;109:507-15.  Back to cited text no. 26
    
27.
Marjon PL, Bobrovnikova-Marjon EV, Abcouwer SF. Expression of the pro-angiogenic factors vascular endothelial growth factor and interleukin-8/CXCL8 by human breast carcinomas is responsive to nutrient deprivation and endoplasmic reticulum stress. Mol Cancer 2004;3:4.  Back to cited text no. 27
    
28.
Razmkhah M, Jaberipour M, Hosseini A, Safaei A, Khalatbari B, Ghaderi A. Expression profile of IL-8 and growth factors in breast cancer cells and adipose-derived stem cells (ASCs) isolated from breast carcinoma. Cell Immunol 2010;265:80-5.  Back to cited text no. 28
    
29.
Fidler IJ, Ellis LM. Chemotherapeutic drugs – More really is not better. Nat Med 2000;6:500-2.  Back to cited text no. 29
[PUBMED]    
30.
Li A, Dubey S, Varney ML, Dave BJ, Singh RK. IL-8 directly enhanced endothelial cell survival, proliferation, and matrix metalloproteinases production and regulated angiogenesis. J Immunol 2003;170:3369-76.  Back to cited text no. 30
    
31.
Duffy AM, Bouchier-Hayes DJ, Harmey JH. Vascular endothelial growth factor (VEGF) and its role in non-endothelial cells: Autocrine signalling by VEGF. In: Harmey JH, editor. VEGF and Cancer. Georgetown, TX: Landes Bioscience; 2004. p. 133-61.  Back to cited text no. 31
    
32.
Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med 2003;9:669-76.  Back to cited text no. 32
    
33.
Roskoski R Jr. Vascular endothelial growth factor (VEGF) signaling in tumor progression. Crit Rev Oncol Hematol 2007;62:179-213.  Back to cited text no. 33
    
34.
Hoeben A, Landuyt B, Highley MS, Wildiers H, Van Oosterom AT, De Bruijn EA. Vascular endothelial growth factor and angiogenesis. Pharmacol Rev 2004;56:549-80.  Back to cited text no. 34
    
35.
Shibuya M. Vascular endothelial growth factor (VEGF) and its receptor (VEGFR) signaling in angiogenesis: A crucial target for anti- and pro-angiogenic therapies. Genes Cancer 2011;2:1097-105.  Back to cited text no. 35
    
36.
Longatto Filho A, Lopes JM, Schmitt FC. Angiogenesis and breast cancer. J Oncol 2010;2010:1-7.  Back to cited text no. 36
    
37.
Sa-Nguanraksa D, O-Charoenrat P. The role of vascular endothelial growth factor a polymorphisms in breast cancer. Int J Mol Sci 2012;13:14845-64.  Back to cited text no. 37
    
38.
Srabovic N, Mujagic Z, Mujanovic-Mustedanagic J, Softic A, Muminovic Z, Rifatbegovic A, et al. Vascular endothelial growth factor receptor-1 expression in breast cancer and its correlation to vascular endothelial growth factor A. Int J Breast Cancer 2013;2013:746749.  Back to cited text no. 38
    
39.
Guo S, Colbert LS, Fuller M, Zhang Y, Gonzalez-Perez RR. Vascular endothelial growth factor receptor-2 in breast cancer. Biochim Biophys Acta 2010;1806:108-21.  Back to cited text no. 39
    
40.
Lushnikova AA, Nasunova IB, Parokonnaya AA, Lyubchenko LN, Kampova-Polevaya EB. VEGFR-2 expression in tumor tissue of breast cancer patients. Dokl Biol Sci 2010;434:363-7.  Back to cited text no. 40
    
41.
Claesson-Welsh L, Welsh M. VEGFA and tumour angiogenesis. J Intern Med 2013;273:114-27.  Back to cited text no. 41
    
42.
Linardou H, Kalogeras KT, Kronenwett R, Kouvatseas G, Wirtz RM, Zagouri F, et al. The prognostic and predictive value of mRNA expression of vascular endothelial growth factor family members in breast cancer: A study in primary tumors of high-rish early breast cancer patients participating in a randomized Hellenic Cooperative Oncology Group trial. Breast Cancer Res 2012;14:R145.  Back to cited text no. 42
[PUBMED]    
43.
Goel HL, Mercurio AM. VEGF targets the tumour cell. Nat Rev Cancer 2013;13:871-82.  Back to cited text no. 43
    
44.
Gunningham SP, Currie MJ, Han C, Robinson BA, Scott PA, Harris AL, et al. VEGF-B expression in human primary breast cancers is associated with lymph node metastasis but not angiogenesis. J Pathol 2001;193:325-32.  Back to cited text no. 44
    
45.
Olofsson B, Korpelainen E, Pepper MS, Mandriota SJ, Aase K, Kumar V, et al. Vascular endothelial growth factor B (VEGF-B) binds to VEGF receptor-1 and regulates plasminogen activator activity in endothelial cells. Proc Natl Acad Sci U S A 1998;95:11709-14.  Back to cited text no. 45
    
46.
Yang X, Zhang Y, Hosaka K, Andersson P, Wang J, Tholander F, et al. VEGF-B promotes cancer metastasis through a VEGF-A-independent mechanism and serves as a marker of poor prognosis for cancer patients. Proc Natl Acad Sci U S A 2015;112:E2900-9.  Back to cited text no. 46
    
47.
Schneider BP, Miller KD. Angiogenesis of breast cancer. J Clin Oncol 2005;23:1782-90.  Back to cited text no. 47
    
48.
Taylor AP, Leon E, Goldenberg DM. Placental growth factor (PlGF) enhances breast cancer cell motility by mobilising ERK1/2 phosphorylation and cytoskeletal rearrangement. Br J Cancer 2010;103:82-9.  Back to cited text no. 48
    
49.
Kim KJ, Cho CS, Kim WU. Role of placenta growth factor in cancer and inflammation. Exp Mol Med 2012;44:10-9.  Back to cited text no. 49
    
50.
Akdis M, Burgler S, Crameri R, Eiwegger T, Fujita H, Gomez E, et al. Interleukins, from 1 to 37, and interferon-g: Receptors, functions, and roles in diseases. J Allergy Clin Immunol 2011;127:701-21.e1-70.  Back to cited text no. 50
    
51.
Bar-Eli M. Role of interleukin-8 in tumor growth and metastasis of human melanoma. Pathobiology 1999;67:12-8.  Back to cited text no. 51
    
52.
Kitadai Y, Takahashi Y, Haruma K, Naka K, Sumii K, Yokozaki H, et al. Transfection of interleukin-8 increases angiogenesis and tumorigenesis of human gastric carcinoma cells in nude mice. Br J Cancer 1999;81:647-53.  Back to cited text no. 52
    
53.
Slattery ML, John EM, Stern MC, Herrick J, Lundgreen A, Giuliano AR, et al. Associations with growth factor genes (FGF1, FGF2, PDGFB, FGFR2, NRG2, EGF, ERBB2) with breast cancer risk and survival: The Breast Cancer Health Disparities Study. Breast Cancer Res Treat 2013;140:587-601.  Back to cited text no. 53
    
54.
Partanen J, Mäkelä TP, Eerola E, Korhonen J, Hirvonen H, Claesson-Welsh L, et al. FGFR-4, a novel acidic fibroblast growth factor receptor with a distinct expression pattern. EMBO J 1991;10:1347-54.  Back to cited text no. 54
    
55.
Jianwu F, Siluo H, Huisheng L, Crepin M, Tao X, Jianfeng L. Role of FGF-2/FGFR signaling pathway in cancer and its signification in breast cancer. Chin Sci Bull 2003;48:1539-47.  Back to cited text no. 55
    
56.
Touriol C, Morillon A, Gensac MC, Prats H, Prats AC. Expression of human fibroblast growth factor 2 mRNA is post-transcriptionally controlled by a unique destabilizing element present in the 3′- untranslated region between alternative polyadenylation sites. J Biol Chem 1999;274:21402-8.  Back to cited text no. 56
    
57.
Sørensen V, Nilsen T, Wiedlocha A. Functional diversity of FGF-2 isoforms by intracellular sorting. Bioessays 2006;28:504-14.  Back to cited text no. 57
    
58.
Liao S, Bodmer J, Pietras D, Azhar M, Doetschman T, Schultz Jel J. Biological functions of the low and high molecular weight protein isoforms of fibroblast growth factor-2 in cardiovascular development and disease. Dev Dyn 2009;238:249-64.  Back to cited text no. 58
    
59.
Yiangou C, Gomm JJ, Coope RC, Law M, Luqmani YA, Shousha S, et al. Fibroblast growth factor 2 in breast cancer: Occurrence and prognostic significance. Br J Cancer 1997;75:28-33.  Back to cited text no. 59
    
60.
Powers CJ, McLeskey SW, Wellstein A. Fibroblast growth factors, their receptors and signaling. Endocr Relat Cancer 2000;7:165-97.  Back to cited text no. 60
    
61.
Jain VK, Turner NC. Challenges and opportunities in the targeting of fibroblast growth factor receptors in breast cancer. Breast Cancer Res 2012;14:208.  Back to cited text no. 61
    
62.
Brady N, Chuntova P, Bade LK, Schwertfeger KL. The FGF/FGFR axis as a therapeutic target in breast cancer. Expert Rev Endocrinol Metab 2013;8:391-402.  Back to cited text no. 62
    
63.
Vlodavsky I, Korner G, Ishai-Michaeli R, Bashkin P, Bar-Shavit R, Fuks Z. Extracellular matrix-resident growth factors and enzymes: Possible involvement in tumor metastasis and angiogenesis. Cancer Metastasis Rev 1990;9:203-26.  Back to cited text no. 63
    
64.
Granato AM, Nanni O, Falcini F, Folli S, Mosconi G, De Paola F, et al. Basic fibroblast growth factor and vascular endothelial growth factor serum levels in breast cancer patients and healthy women: Useful as diagnostic tools? Breast Cancer Res 2004;6:R38-45.  Back to cited text no. 64
    
65.
Partanen J, Mäkelä TP, Alitalo R, Lehväslaiho H, Alitalo K. Putative tyrosine kinases expressed in K-562 human leukemia cells. Proc Natl Acad Sci U S A 1990;87:8913-7.  Back to cited text no. 65
    
66.
Woessner JF Jr. The family of matrix metalloproteinases. Ann N Y Acad Sci 1994;732:11-21.  Back to cited text no. 66
    
67.
Heppner KJ, Matrisian LM, Jensen RA, Rodgers WH. Expression of most matrix metalloproteinase family members in breast cancer represents a tumor-induced host response. Am J Pathol 1996;149:273-82.  Back to cited text no. 67
    
68.
Nagase H, Woessner JF Jr. Matrix metalloproteinases. J Biol Chem 1999;274:21491-4.  Back to cited text no. 68
    
69.
McCawley LJ, Matrisian LM. Matrix metalloproteinases: Multifunctional contributors to tumor progression. Mol Med Today 2000;6:149-56.  Back to cited text no. 69
    
70.
Murray GI. Matrix metalloproteinases: A multifunctional group of molecules. J Pathol 2001;195:135-7.  Back to cited text no. 70
[PUBMED]    
71.
Bartsch JE, Staren ED, Appert HE. Matrix metalloproteinase expression in breast cancer. J Surg Res 2003;110:383-92.  Back to cited text no. 71
    
72.
Rundhaug JE. Matrix metalloproteinases, angiogenesis, and cancer. Clin Cancer Res 2004;9:551-4.  Back to cited text no. 72
    
73.
Kousidou OC, Roussidis AE, Theocharis AD, Karamanos NK. Expression of MMPs and TIMPs genes in human breast cancer epithelial cells depends on cell culture conditions and is associated with their invasive potential. Anticancer Res 2004;24:4025-30.  Back to cited text no. 73
    
74.
van Hinsbergh VW, Engelse MA, Quax PH. Pericellular proteases in angiogenesis and vasculogenesis. Arterioscler Thromb Vasc Biol 2006;26:716-28.  Back to cited text no. 74
    
75.
Verma RP, Hansch C. Matrix metalloproteinases (MMPs): Chemical-biological functions and (Q) SARs. Bioorg Med Chem 2007;15:2223-68.  Back to cited text no. 75
    
76.
Lee KS, Rha SY, Kim SJ, Kim JH, Roh JK, Kim BS, et al. Sequential activation and production of matrix metalloproteinase-2 during breast cancer progression. Clin Exp Metastasis 1996;14:512-9.  Back to cited text no. 76
    
77.
Brummer O, Athar S, Riethdorf L, Löning T, Herbst H. Matrix-metalloproteinases 1, 2, and 3 and their tissue inhibitors 1 and 2 in benign and malignant breast lesions: An in situ hybridization study. Virchows Arch 1999;435:566-73.  Back to cited text no. 77
    
78.
Merdad A, Karim S, Schulten HJ, Dallol A, Buhmeida A, Al-Thubaity F, et al. Expression of matrix metalloproteinases (MMPs) in primary human breast cancer: MMP-9 as a potential biomarker for cancer invasion and metastasis. Anticancer Res 2014;34:1355-66.  Back to cited text no. 78
    
79.
Bisacchi D, Benelli R, Vanzetto C, Ferrari N, Tosetti F, Albini A. Anti-angiogenesis and angioprevention: Mechanisms, problems and perspectives. Cancer Detect Prev 2003;27:229-38.  Back to cited text no. 79
    
80.
Singh RK, Gutman M, Bucana CD, Sanchez R, Llansa N, Fidler IJ. Interferons alpha and beta down-regulate the expression of basic fibroblast growth factor in human carcinomas. Proc Natl Acad Sci U S A 1995;92:4562-6.  Back to cited text no. 80
    
81.
Albini A, Marchisone C, Del Grosso F, Benelli R, Masiello L, Tacchetti C, et al. Inhibition of angiogenesis and vascular tumor growth by interferon-producing cells: A gene therapy approach. Am J Pathol 2000;156:1381-93.  Back to cited text no. 81
    
82.
Indraccolo S, Gola E, Rosato A, Minuzzo S, Habeler W, Tisato V, et al. Differential effects of angiostatin, endostatin and interferon-alpha(1) gene transfer on in vivo growth of human breast cancer cells. Gene Ther 2002;9:867-78.  Back to cited text no. 82
    
83.
O′Reilly MS, Boehm T, Shing Y, Fukai N, Vasios G, Lane WS, et al. Endostatin: An endogenous inhibitor of angiogenesis and tumor growth. Cell 1997;88:277-85.  Back to cited text no. 83
    
84.
Dhanabal M, Ramchandran R, Waterman MJ, Lu H, Knebelmann B, Segal M, et al. Endostatin induces endothelial cell apoptosis. J Biol Chem 1999;274:11721-6.  Back to cited text no. 84
    
85.
Scappaticci FA, Smith R, Pathak A, Schloss D, Lum B, Cao Y, et al. Combination angiostatin and endostatin gene transfer induces synergistic antiangiogenic activity in vitro and antitumor efficacy in leukemia and solid tumors in mice. Mol Ther 2001;3:186-96.  Back to cited text no. 85
    
86.
Rehn M, Veikkola T, Kukk-Valdre E, Nakamura H, Ilmonen M, Lombardo C, et al. Interaction of endostatin with integrins implicated in angiogenesis. Proc Natl Acad Sci U S A 2001;98:1024-9.  Back to cited text no. 86
    
87.
Yin G, Liu W, An P, Li P, Ding I, Planelles V, et al. Endostatin gene transfer inhibits joint angiogenesis and pannus formation in inflammatory arthritis. Mol Ther 2002;5(5 Pt 1):547-54.  Back to cited text no. 87
    
88.
Hanai J, Dhanabal M, Karumanchi SA, Albanese C, Waterman M, Chan B, et al. Endostatin causes G1 arrest of endothelial cells through inhibition of cyclin D1. J Biol Chem 2002;277:16464-9.  Back to cited text no. 88
    
89.
Harris AL, Generali D. Biological therapies for metastatic breast cancer: Antiangiogenesis. In: Piccart M, Wood WC, Hung MC, Solin LJ, Cardoso F, editors. Breast Cancer Management and Molecular Medicine: Towards Tailored Approaches. Berlin, Germany: Springer Science & Business Media; 2007. p. 685-7.  Back to cited text no. 89
    
90.
O′Reilly MS, Holmgren L, Shing Y, Chen C, Rosenthal RA, Moses M, et al. Angiostatin: A novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell 1994;79:315-28.  Back to cited text no. 90
    
91.
O′Reilly MS, Holmgren L, Chen C, Folkman J. Angiostatin induces and sustains dormancy of human primary tumors in mice. Nat Med 1996;2:689-92.  Back to cited text no. 91
    
92.
Wahl ML, Moser TL, Pizzo SV. Angiostatin and anti-angiogenic therapy in human disease. Recent Prog Horm Res 2004;59:73-104.  Back to cited text no. 92
    
93.
Moser TL, Stack MS, Asplin I, Enghild JJ, Højrup P, Everitt L, et al. Angiostatin binds ATP synthase on the surface of human endothelial cells. Proc Natl Acad Sci U S A 1999;96:2811-6.  Back to cited text no. 93
    
94.
Wajih N, Sane DC. Angiostatin selectively inhibits signaling by hepatocyte growth factor in endothelial and smooth muscle cells. Blood 2003;101:1857-63.  Back to cited text no. 94
    
95.
Tanaka T, Cao Y, Folkman J, Fine HA. Viral vector-targeted antiangiogenic gene therapy utilizing an angiostatin complementary DNA. Cancer Res 1998;58:3362-9.  Back to cited text no. 95
    
96.
Wong SY, Purdie AT, Han P. Thrombospondin and other possible related matrix proteins in malignant and benign breast disease. An immunohistochemical study. Am J Pathol 1992;140:1473-82.  Back to cited text no. 96
    
97.
Zabrenetzky V, Harris CC, Steeg PS, Roberts DD. Expression of the extracellular matrix molecule thrombospondin inversely correlates with malignant progression in melanoma, lung and breast carcinoma cell lines. Int J Cancer 1994;59:191-5.  Back to cited text no. 97
    
98.
Volpert OV, Stellmach V, Bouck N. The modulation of thrombospondin and other naturally occurring inhibitors of angiogenesis during tumor progression. Breast Cancer Res Treat 1995;36:119-26.  Back to cited text no. 98
    
99.
Streit M, Velasco P, Brown LF, Skobe M, Richard L, Riccardi L, et al. Overexpression of thrombospondin-1 decreases angiogenesis and inhibits the growth of human cutaneous squamous cell carcinomas. Am J Pathol 1999;155:441-52.  Back to cited text no. 99
    
100.
Lawler J. Thrombospondin-1 as an endogenous inhibitor of angiogenesis and tumor growth. J Cell Mol Med 2002;6:1-12.  Back to cited text no. 100
    
101.
Rice A, Quinn CM. Angiogenesis, thrombospondin, and ductal carcinoma in situ of the breast. J Clin Pathol 2002;55:569-74.  Back to cited text no. 101
    
102.
Chen H, Herndon ME, Lawler J. The cell biology of thrombospondin-1. Matrix Biol 2000;19:597-614.  Back to cited text no. 102
    
103.
Taraboletti G, Roberts D, Liotta LA, Giavazzi R. Platelet thrombospondin modulates endothelial cell adhesion, motility, and growth: A potential angiogenesis regulatory factor. J Cell Biol 1990;111:765-72.  Back to cited text no. 103
    
104.
Vogel T, Guo NH, Krutzsch HC, Blake DA, Hartman J, Mendelovitz S, et al. Modulation of endothelial cell proliferation, adhesion, and motility by recombinant heparin-binding domain and synthetic peptides from the type I repeats of thrombospondin. J Cell Biochem 1993;53:74-84.  Back to cited text no. 104
    
105.
Iruela-Arispe ML, Lombardo M, Krutzsch HC, Lawler J, Roberts DD. Inhibition of angiogenesis by thrombospondin-1 is mediated by 2 independent regions within the type 1 repeats. Circulation 1999;100:1423-31.  Back to cited text no. 105
    
106.
Yu H, Tyrrell D, Cashel J, Guo NH, Vogel T, Sipes JM, et al. Specificities of heparin-binding sites from the amino-terminus and type 1 repeats of thrombospondin-1. Arch Biochem Biophys 2000;374:13-23.  Back to cited text no. 106
    
107.
Brooks PC, Montgomery AM, Rosenfeld M, Reisfeld RA, Hu T, Klier G, et al. Integrin alpha v beta 3 antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell 1994;79:1157-64.  Back to cited text no. 107
    
108.
Murphy-Ullrich JE, Poczatek M. Activation of latent TGF-beta by thrombospondin-1: Mechanisms and physiology. Cytokine Growth Factor Rev 2000;11:59-69.  Back to cited text no. 108
    
109.
Rodriguez-Manzaneque JC, Lane TF, Ortega MA, Hynes RO, Lawler J, Iruela-Arispe ML. Thrombospondin-1 suppresses spontaneous tumor growth and inhibits activation of matrix metalloproteinase-9 and mobilization of vascular endothelial growth factor. Proc Natl Acad Sci U S A 2001;98:12485-90.  Back to cited text no. 109
    
110.
Volpert OV, Tolsma SS, Pellerin S, Feige JJ, Chen H, Mosher DF, et al. Inhibition of angiogenesis by thrombospondin-2. Biochem Biophys Res Commun 1995;217:326-32.  Back to cited text no. 110
    
111.
Streit M, Riccardi L, Velasco P, Brown LF, Hawighorst T, Bornstein P, et al. Thrombospondin-2: A potent endogenous inhibitor of tumor growth and angiogenesis. Proc Natl Acad Sci U S A 1999;96:14888-93.  Back to cited text no. 111
    
112.
Vogel KG, Paulsson M, Heinegård D. Specific inhibition of type I and type II collagen fibrillogenesis by the small proteoglycan of tendon. Biochem J 1984;223:587-97.  Back to cited text no. 112
    
113.
Scott JE. Proteoglycan-fibrillar collagen interactions. Biochem J 1988;252:313-23.  Back to cited text no. 113
    
114.
Hocking AM, Shinomura T, McQuillan DJ. Leucine-rich repeat glycoproteins of the extracellular matrix. Matrix Biol 1998;17:1-19.  Back to cited text no. 114
    
115.
Iozzo RV. Matrix proteoglycans: From molecular design to cellular function. Annu Rev Biochem 1998;67:609-52.  Back to cited text no. 115
    
116.
Iozzo RV, Moscatello DK, McQuillan DJ, Eichstetter I. Decorin is a biological ligand for the epidermal growth factor receptor. J Biol Chem 1999;274:4489-92.  Back to cited text no. 116
    
117.
Grant DS, Yenisey C, Rose RW, Tootell M, Santra M, Iozzo RV. Decorin suppresses tumor cell-mediated angiogenesis. Oncogene 2002;21:4765-77.  Back to cited text no. 117
    
118.
Reed CC, Iozzo RV. The role of decorin in collagen fibrillogenesis and skin homeostasis. Glycoconj J 2002;19:249-55.  Back to cited text no. 118
    
119.
Reed CC, Waterhouse A, Kirby S, Kay P, Owens RT, McQuillan DJ, et al. Decorin prevents metastatic spreading of breast cancer. Oncogene 2005;24:1104-10.  Back to cited text no. 119
    
120.
Kalamajski S, Oldberg A. The role of small leucine-rich proteoglycans in collagen fibrillogenesis. Matrix Biol 2010;29:248-53.  Back to cited text no. 120
    
121.
Sofeu Feugaing DD, Götte M, Viola M. More than matrix: The multifaceted role of decorin in cancer. Eur J Cell Biol 2013;92:1-11.  Back to cited text no. 121
    
122.
Järveläinen H, Sainio A, Wight TN. Pivotal role for decorin in angiogenesis. Matrix Biol 2015;43:15-26.  Back to cited text no. 122
    
123.
Adany R, Heimer R, Caterson B, Sorrell JM, Iozzo RV. Altered expression of chondroitin sulfate proteoglycan in the stroma of human colon carcinoma. Hypomethylation of PG-40 gene correlates with increased PG-40 content and mRNA levels. J Biol Chem 1990;265:11389-96.  Back to cited text no. 123
    
124.
Bianco P, Fisher LW, Young MF, Termine JD, Robey PG. Expression and localization of the two small proteoglycans biglycan and decorin in developing human skeletal and non-skeletal tissues. J Histochem Cytochem 1990;38:1549-63.  Back to cited text no. 124
    
125.
Yamaguchi Y, Mann DM, Ruoslahti E. Negative regulation of transforming growth factor-beta by the proteoglycan decorin. Nature 1990;346:281-4.  Back to cited text no. 125
    
126.
Järveläinen HT, Kinsella MG, Wight TN, Sandell LJ. Differential expression of small chondroitin/dermatan sulfate proteoglycans, PG-I/biglycan and PG-II/decorin, by vascular smooth muscle and endothelial cells in culture. J Biol Chem 1991;266:23274-81.  Back to cited text no. 126
    
127.
Ruoslahti E, Yamaguchi Y. Proteoglycans as modulators of growth factor activities. Cell 1991;64:867-9.  Back to cited text no. 127
    
128.
Border WA, Noble NA, Yamamoto T, Harper JR, Yamaguchi YU, Pierschbacher MD, et al. Natural inhibitor of transforming growth factor-beta protects against scarring in experimental kidney disease. Nature 1992;360:361-4.  Back to cited text no. 128
    
129.
Coppock DL, Kopman C, Scandalis S, Gilleran S. Preferential gene expression in quiescent human lung fibroblasts. Cell Growth Differ 1993;4:483-93.  Back to cited text no. 129
    
130.
Hildebrand A, Romarís M, Rasmussen LM, Heinegård D, Twardzik DR, Border WA, et al. Interaction of the small interstitial proteoglycans biglycan, decorin and fibromodulin with transforming growth factor beta. Biochem J 1994;302(Pt 2):527-34.  Back to cited text no. 130
    
131.
Mauviel A, Santra M, Chen YQ, Uitto J, Iozzo RV. Transcriptional regulation of decorin gene expression. Induction by quiescence and repression by tumor necrosis factor-alpha. J Biol Chem 1995;270:11692-700.  Back to cited text no. 131
    
132.
De Luca A, Santra M, Baldi A, Giordano A, Iozzo RV. Decorin-induced growth suppression is associated with up-regulation of p21, an inhibitor of cyclin-dependent kinases. J Biol Chem 1996;271:18961-5.  Back to cited text no. 132
    
133.
Santra M, Mann DM, Mercer EW, Skorski T, Calabretta B, Iozzo RV. Ectopic expression of decorin protein core causes a generalized growth suppression in neoplastic cells of various histogenetic origin and requires endogenous p21, an inhibitor of cyclin-dependent kinases. J Clin Invest 1997;100:149-57.  Back to cited text no. 133
    
134.
Moscatello DK, Santra M, Mann DM, McQuillan DJ, Wong AJ, Iozzo RV. Decorin suppresses tumor cell growth by activating the epidermal growth factor receptor. J Clin Invest 1998;101:406-12.  Back to cited text no. 134
    
135.
Patel S, Santra M, McQuillan DJ, Iozzo RV, Thomas AP. Decorin activates the epidermal growth factor receptor and elevates cytosolic Ca2+in A431 carcinoma cells. J Biol Chem 1998;273:3121-4.  Back to cited text no. 135
    
136.
Penc SF, Pomahac B, Winkler T, Dorschner RA, Eriksson E, Herndon M, et al. Dermatan sulfate released after injury is a potent promoter of fibroblast growth factor-2 function. J Biol Chem 1998;273:28116-21.  Back to cited text no. 136
    
137.
Ständer M, Naumann U, Dumitrescu L, Heneka M, Löschmann P, Gulbins E, et al. Decorin gene transfer-mediated suppression of TGF-beta synthesis abrogates experimental malignant glioma growth in vivo. Gene Ther 1998;5:1187-94.  Back to cited text no. 137
    
138.
Verbeek BS, Adriaansen-Slot SS, Vroom TM, Beckers T, Rijksen G. Overexpression of EGFR and c-erbB2 causes enhanced cell migration in human breast cancer cells and NIH3T3 fibroblasts. FEBS Lett 1998;425:145-50.  Back to cited text no. 138
    
139.
Nash MA, Loercher AE, Freedman RS. In vitro growth inhibition of ovarian cancer cells by decorin: Synergism of action between decorin and carboplatin. Cancer Res 1999;59:6192-6.  Back to cited text no. 139
    
140.
Csordás G, Santra M, Reed CC, Eichstetter I, McQuillan DJ, Gross D, et al. Sustained down-regulation of the epidermal growth factor receptor by decorin. A mechanism for controlling tumor growth in vivo. J Biol Chem 2000;275:32879-87.  Back to cited text no. 140
    
141.
Santra M, Eichstetter I, Iozzo RV. An anti-oncogenic role for decorin. Down-regulation of ErbB2 leads to growth suppression and cytodifferentiation of mammary carcinoma cells. J Biol Chem 2000;275:35153-61.  Back to cited text no. 141
    
142.
Swindle CS, Tran KT, Johnson TD, Banerjee P, Mayes AM, Griffith L, et al. Epidermal growth factor (EGF)-like repeats of human tenascin-C as ligands for EGF receptor. J Cell Biol 2001;154:459-68.  Back to cited text no. 142
    
143.
Nash MA, Deavers MT, Freedman RS. The expression of decorin in human ovarian tumors. Clin Cancer Res 2002;8:1754-60.  Back to cited text no. 143
    
144.
Reed CC, Gauldie J, Iozzo RV. Suppression of tumorigenicity by adenovirus-mediated gene transfer of decorin. Oncogene 2002;21:3688-95.  Back to cited text no. 144
    
145.
Santra M, Reed CC, Iozzo RV. Decorin binds to a narrow region of the epidermal growth factor (EGF) receptor, partially overlapping but distinct from the EGF-binding epitope. J Biol Chem 2002;277:35671-81.  Back to cited text no. 145
    
146.
Tralhão JG, Schaefer L, Micegova M, Evaristo C, Schönherr E, Kayal S, et al. In vivo selective and distant killing of cancer cells using adenovirus-mediated decorin gene transfer. FASEB J 2003;17:464-6.  Back to cited text no. 146
    
147.
Köninger J, Giese NA, di Mola FF, Berberat P, Giese T, Esposito I, et al. Overexpressed decorin in pancreatic cancer: Potential tumor growth inhibition and attenuation of chemotherapeutic action. Clin Cancer Res 2004;10:4776-83.  Back to cited text no. 147
    
148.
Zhu JX, Goldoni S, Bix G, Owens RT, McQuillan DJ, Reed CC, et al. Decorin evokes protracted internalization and degradation of the epidermal growth factor receptor via caveolar endocytosis. J Biol Chem 2005;280:32468-79.  Back to cited text no. 148
    
149.
Sulochana KN, Fan H, Jois S, Subramanian V, Sun F, Kini RM, et al. Peptides derived from human decorin leucine-rich repeat 5 inhibit angiogenesis. J Biol Chem 2005;280:27935-48.  Back to cited text no. 149
    
150.
Seidler DG, Goldoni S, Agnew C, Cardi C, Thakur ML, Owens RT, et al. Decorin protein core inhibits in vivo cancer growth and metabolism by hindering epidermal growth factor receptor function and triggering apoptosis via caspase-3 activation. J Biol Chem 2006;281:26408-18.  Back to cited text no. 150
    
151.
Goldoni S, Seidler DG, Heath J, Fassan M, Baffa R, Thakur ML, et al. An antimetastatic role for decorin in breast cancer. Am J Pathol 2008;173:844-55.  Back to cited text no. 151
    
152.
Salomäki HH, Sainio AO, Söderström M, Pakkanen S, Laine J, Järveläinen HT. Differential expression of decorin by human malignant and benign vascular tumors. J Histochem Cytochem 2008;56:639-46.  Back to cited text no. 152
    
153.
Hu Y, Sun H, Owens RT, Wu J, Chen YQ, Berquin IM, et al. Decorin suppresses prostate tumor growth through inhibition of epidermal growth factor and androgen receptor pathways. Neoplasia 2009;11:1042-53.  Back to cited text no. 153
    
154.
Shintani K, Matsumine A, Kusuzaki K, Morikawa J, Matsubara T, Wakabayashi T, et al. Decorin suppresses lung metastases of murine osteosarcoma. Oncol Rep 2008;19:1533-9.  Back to cited text no. 154
    
155.
Khan GA, Girish GV, Lala N, Di Guglielmo GM, Lala PK. Decorin is a novel VEGFR-2-binding antagonist for the human extravillous trophoblast. Mol Endocrinol 2011;25:1431-43.  Back to cited text no. 155
    
156.
Mohan RR, Tovey JC, Sharma A, Schultz GS, Cowden JW, Tandon A. Targeted decorin gene therapy delivered with adeno-associated virus effectively retards corneal neovascularization in vivo. PLoS One 2011;6:e26432.  Back to cited text no. 156
    
157.
Schwaller J, Koeffler HP, Niklaus G, Loetscher P, Nagel S, Fey MF, et al. Posttranscriptional stabilization underlies p53-independent induction of p21WAF1/CIP1/SDI1 in differentiating human leukemic cells. J Clin Invest 1995;95:973-9.  Back to cited text no. 157
    
158.
Miller KD. Recent translational research: Antiangiogenic therapy for breast cancer – Where do we stand? Breast Cancer Res 2004;6:128-32.  Back to cited text no. 158
    
159.
Samant RS, Shevde LA. Recent advances in anti-angiogenic therapy of cancer. Oncotarget 2011;2:122-34.  Back to cited text no. 159
    
160.
Fakhrejahani E, Toi M. Antiangiogenesis therapy for breast cancer: An update and perspectives from clinical trials. Jpn J Clin Oncol 2014;44:197-207.  Back to cited text no. 160
    
161.
Kim KJ, Li B, Winer J, Armanini M, Gillett N, Phillips HS, et al. Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature 1993;362:841-4.  Back to cited text no. 161
    
162.
Presta LG, Chen H, O′Connor SJ, Chisholm V, Meng YG, Krummen L, et al. Humanization of an anti-vascular endothelial growth factor monoclonal antibody for the therapy of solid tumors and other disorders. Cancer Res 1997;57:4593-9.  Back to cited text no. 162
    
163.
Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov 2004;3:391-400.  Back to cited text no. 163
    
164.
Rüegg C, Hasmim M, Lejeune FJ, Alghisi GC. Antiangiogenic peptides and proteins: From experimental tools to clinical drugs. Biochim Biophys Acta 2006;1765:155-77.  Back to cited text no. 164
    
165.
Hsu JY, Wakelee HA. Monoclonal antibodies targeting vascular endothelial growth factor: Current status and future challenges in cancer therapy. BioDrugs 2009;23:289-304.  Back to cited text no. 165
    
166.
Kabbinavar F, Hurwitz HI, Fehrenbacher L, Meropol NJ, Novotny WF, Lieberman G, et al. Phase II, randomized trial comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV) with FU/LV alone in patients with metastatic colorectal cancer. J Clin Oncol 2003;21:60-5.  Back to cited text no. 166
    
167.
Gordon MS, Margolin K, Talpaz M, Sledge GW Jr., Holmgren E, Benjamin R, et al. Phase I safety and pharmacokinetic study of recombinant human anti-vascular endothelial growth factor in patients with advanced cancer. J Clin Oncol 2001;19:843-50.  Back to cited text no. 167
    
168.
Cobleigh MA, Langmuir VK, Sledge GW, Miller KD, Haney L, Novotny WF, et al. A phase I/II dose-escalation trial of bevacizumab in previously treated metastatic breast cancer. Semin Oncol 2003;30 5 Suppl 16:117-24.  Back to cited text no. 168
    
169.
Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ, Topalian SL, et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med 2003;349:427-34.  Back to cited text no. 169
    
170.
Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004;350:2335-42.  Back to cited text no. 170
    
171.
Johnson DH, Fehrenbacher L, Novotny WF, Herbst RS, Nemunaitis JJ, Jablons DM, et al. Randomized phase II trial comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and paclitaxel alone in previously untreated locally advanced or metastatic non-small-cell lung cancer. J Clin Oncol 2004;22:2184-91.  Back to cited text no. 171
    
172.
Blagosklonny MV. How Avastin potentiates chemotherapeutic drugs: Action and reaction in antiangiogenic therapy. Cancer Biol Ther 2005;4:1307-10.  Back to cited text no. 172
    
173.
de Gramont A, Van Cutsem E. Investigating the potential of bevacizumab in other indications: Metastatic renal cell, non-small cell lung, pancreatic and breast cancer. Oncology 2005;69 Suppl 3:46-56.  Back to cited text no. 173
    
174.
Herbst RS, Johnson DH, Mininberg E, Carbone DP, Henderson T, Kim ES, et al. Phase I/II trial evaluating the anti-vascular endothelial growth factor monoclonal antibody bevacizumab in combination with the HER-1/epidermal growth factor receptor tyrosine kinase inhibitor erlotinib for patients with recurrent non-small-cell lung cancer. J Clin Oncol 2005;23:2544-55.  Back to cited text no. 174
    
175.
Miller KD, Chap LI, Holmes FA, Cobleigh MA, Marcom PK, Fehrenbacher L, et al. Randomized phase III trial of capecitabine compared with bevacizumab plus capecitabine in patients with previously treated metastatic breast cancer. J Clin Oncol 2005;23:792-9.  Back to cited text no. 175
    
176.
Hochster HS. Bevacizumab in combination with chemotherapy: First-line treatment of patients with metastatic colorectal cancer. Semin Oncol 2006;33 5 Suppl 10:S8-14.  Back to cited text no. 176
    
177.
Bukowski RM, Kabbinavar FF, Figlin RA, Flaherty K, Srinivas S, Vaishampayan U, et al. Randomized phase II study of erlotinib combined with bevacizumab compared with bevacizumab alone in metastatic renal cell cancer. J Clin Oncol 2007;25:4536-41.  Back to cited text no. 177
    
178.
Herbst RS, O′Neill VJ, Fehrenbacher L, Belani CP, Bonomi PD, Hart L, et al. Phase II study of efficacy and safety of bevacizumab in combination with chemotherapy or erlotinib compared with chemotherapy alone for treatment of recurrent or refractory non small-cell lung cancer. J Clin Oncol 2007;25:4743-50.  Back to cited text no. 178
    
179.
de Groot JF, Yung WK. Bevacizumab and irinotecan in the treatment of recurrent malignant gliomas. Cancer J 2008;14:279-85.  Back to cited text no. 179
    
180.
Dickler MN, Rugo HS, Eberle CA, Brogi E, Caravelli JF, Panageas KS, et al. A phase II trial of erlotinib in combination with bevacizumab in patients with metastatic breast cancer. Clin Cancer Res 2008;14:7878-83.  Back to cited text no. 180
    
181.
Li WW, Hutnik M, Gehr G. Antiangiogenesis in haematological malignancies. Br J Haematol 2008;143:622-31.  Back to cited text no. 181
    
182.
Manegold C. Bevacizumab for the treatment of advanced non-small-cell lung cancer. Expert Rev Anticancer Ther 2008;8:689-99.  Back to cited text no. 182
    
183.
Sachdev JC, Jahanzeb M. Evolution of bevacizumab-based therapy in the management of breast cancer. Clin Breast Cancer 2008;8:402-10.  Back to cited text no. 183
    
184.
Torrisi R, Bagnardi V, Cardillo A, Bertolini F, Scarano E, Orlando L, et al. Preoperative bevacizumab combined with letrozole and chemotherapy in locally advanced ER- and/or PgR-positive breast cancer: Clinical and biological activity. Br J Cancer 2008;99:1564-71.  Back to cited text no. 184
    
185.
Arkenau HT, Brunetto AT, Barriuso J, Olmos D, Eaton D, de Bono J, et al. Clinical benefit of new targeted agents in phase I trials in patients with advanced colorectal cancer. Oncology 2009;76:151-6.  Back to cited text no. 185
    
186.
Thomas MB, Morris JS, Chadha R, Iwasaki M, Kaur H, Lin E, et al. Phase II trial of the combination of bevacizumab and erlotinib in patients who have advanced hepatocellular carcinoma. J Clin Oncol 2009;27:843-50.  Back to cited text no. 186
    
187.
Greenberg S, Rugo HS. Triple-negative breast cancer: Role of antiangiogenic agents. Cancer J 2010;16:33-8.  Back to cited text no. 187
    
188.
Miller K, Wang M, Gralow J, Dickler M, Cobleigh M, Perez EA, et al. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med 2007;357:2666-76.  Back to cited text no. 188
    
189.
Aogi K, Masuda N, Ohno S, Oda T, Iwata H, Kashiwaba M, et al. First-line bevacizumab in combination with weekly paclitaxel for metastatic breast cancer: Efficacy and safety results from a large, open-label, single-arm Japanese study. Breast Cancer Res Treat 2011;129:829-38.  Back to cited text no. 189
    
190.
Brufsky AM, Hurvitz S, Perez E, Swamy R, Valero V, O′Neill V, et al. RIBBON-2: A randomized, double-blind, placebo-controlled, phase III trial evaluating the efficacy and safety of bevacizumab in combination with chemotherapy for second-line treatment of human epidermal growth factor receptor 2-negative metastatic breast cancer. J Clin Oncol 2011;29:4286-93.  Back to cited text no. 190
    
191.
Pivot X, Schneeweiss A, Verma S, Thomssen C, Passos-Coelho JL, Benedetti G, et al. Efficacy and safety of bevacizumab in combination with docetaxel for the first-line treatment of elderly patients with locally recurrent or metastatic breast cancer: Results from AVADO. Eur J Cancer 2011;47:2387-95.  Back to cited text no. 191
    
192.
Robert NJ, Diéras V, Glaspy J, Brufsky AM, Bondarenko I, Lipatov ON, et al. RIBBON-1: Randomized, double-blind, placebo-controlled, phase III trial of chemotherapy with or without bevacizumab for first-line treatment of human epidermal growth factor receptor 2-negative, locally recurrent or metastatic breast cancer. J Clin Oncol 2011;29:1252-60.  Back to cited text no. 192
    
193.
Smith IE, Pierga JY, Biganzoli L, Cortés-Funes H, Thomssen C, Pivot X, et al. First-line bevacizumab plus taxane-based chemotherapy for locally recurrent or metastatic breast cancer: Safety and efficacy in an open-label study in 2,251 patients. Ann Oncol 2011;22:595-602.  Back to cited text no. 193
    
194.
Pierga JY, Petit T, Delozier T, Ferrero JM, Campone M, Gligorov J, et al. Neoadjuvant bevacizumab, trastuzumab, and chemotherapy for primary inflammatory HER2-positive breast cancer (BEVERLY-2): An open-label, single-arm phase 2 study. Lancet Oncol 2012;13:375-84.  Back to cited text no. 194
    
195.
Gianni L, Romieu GH, Lichinitser M, Serrano SV, Mansutti M, Pivot X, et al. AVEREL: A randomized phase III Trial evaluating bevacizumab in combination with docetaxel and trastuzumab as first-line therapy for HER2-positive locally recurrent/metastatic breast cancer. J Clin Oncol 2013;31:1719-25.  Back to cited text no. 195
    
196.
Bear HD, Tang G, Rastogi P, Geyer CE Jr., Robidoux A, Atkins JN, et al. Bevacizumab added to neoadjuvant chemotherapy for breast cancer. N Engl J Med 2012;366:310-20.  Back to cited text no. 196
    
197.
von Minckwitz G, Eidtmann H, Rezai M, Fasching PA, Tesch H, Eggemann H, et al. Neoadjuvant chemotherapy and bevacizumab for HER2-negative breast cancer. N Engl J Med 2012;366:299-309.  Back to cited text no. 197
    
198.
U.S. Food and Drug Administration. FDA Commissioner Announces Avastin Decision. [Press Release]; 2011. Available from: http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm280536.htm. [Last accessed 2016 Feb 02].  Back to cited text no. 198
    
199.
Brooks PC, Clark RA, Cheresh DA. Requirement of vascular integrin alpha v beta 3 for angiogenesis. Science 1994;264:569-71.  Back to cited text no. 199
    
200.
Bäuerle T, Komljenovic D, Merz M, Berger MR, Goodman SL, Semmler W. Cilengitide inhibits progression of experimental breast cancer bone metastases as imaged noninvasively using VCT, MRI and DCE-MRI in a longitudinal in vivo study. Int J Cancer 2011;128:2453-62.  Back to cited text no. 200
    
201.
Lautenschlaeger T, Perry J, Peereboom D, Li B, Ibrahim A, Huebner A, et al. In vitro study of combined cilengitide and radiation treatment in breast cancer cell lines. Radiat Oncol 2013;8:246.  Back to cited text no. 201
    
202.
Burke PA, DeNardo SJ, Miers LA, Lamborn KR, Matzku S, DeNardo GL. Cilengitide targeting of alpha(v)beta(3) integrin receptor synergizes with radioimmunotherapy to increase efficacy and apoptosis in breast cancer xenografts. Cancer Res 2002;62:4263-72.  Back to cited text no. 202
    
203.
Stupp R, Hegi ME, Gorlia T, Erridge SC, Perry J, Hong YK, et al. Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071-22072 study): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2014;15:1100-8.  Back to cited text no. 203
    
204.
Manegold C, Vansteenkiste J, Cardenal F, Schuette W, Woll PJ, Ulsperger E, et al. Randomized phase II study of three doses of the integrin inhibitor cilengitide versus docetaxel as second-line treatment for patients with advanced non-small-cell lung cancer. Invest New Drugs 2013;31:175-82.  Back to cited text no. 204
    
205.
Bradley DA, Daignault S, Ryan CJ, Dipaola RS, Cooney KA, Smith DC, et al. Cilengitide (EMD 121974, NSC 707544) in asymptomatic metastatic castration resistant prostate cancer patients: A randomized phase II trial by the prostate cancer clinical trials consortium. Invest New Drugs 2011;29:1432-40.  Back to cited text no. 205
    
206.
Eskens FA, Dumez H, Hoekstra R, Perschl A, Brindley C, Böttcher S, et al. Phase I and pharmacokinetic study of continuous twice weekly intravenous administration of Cilengitide (EMD 121974), a novel inhibitor of the integrins alphavbeta3 and alphavbeta5 in patients with advanced solid tumours. Eur J Cancer 2003;39:917-26.  Back to cited text no. 206
    
207.
National Cancer Institute. Weekly Doses of Cilengitide and Paclitaxel in Treating Patients with Advanced Solid Tumors that Cannot be Removed by Surgery. In: ClinicalTrials.gov. Bethesda, MD: National Library of Medicine; 2000. NLMIdentifier: NCT01276496. Available from: http://www.clinicaltrials.gov/ct2/show/NCT01276496. [Last cited on 2015 Sep 29].  Back to cited text no. 207
    
208.
Posey JA, Khazaeli MB, DelGrosso A, Saleh MN, Lin CY, Huse W, et al. A pilot trial of Vitaxin, a humanized anti-vitronectin receptor (anti alpha v beta 3) antibody in patients with metastatic cancer. Cancer Biother Radiopharm 2001;16:125-32.  Back to cited text no. 208
    
209.
Liu Z, Wang F, Chen X. Integrin alpha(v)beta(3)-Targeted Cancer Therapy. Drug Dev Res 2008;69:329-39.  Back to cited text no. 209
    

 Authors


Jessica M. Castaρeda-Gill: Department of Molecular and Medical Genetics, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, TX, USA
Jamboor K. Vishwanatha: Institute for Cancer Research, Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX, USA