Open access peer-reviewed chapter

Tumour Angiogenesis in Breast Cancer

Written By

Pooja G. Singh, Kanthesh M. Basalingappa, T.S. Gopenath and B.V. Sushma

Submitted: 28 January 2022 Reviewed: 31 January 2022 Published: 14 May 2022

DOI: 10.5772/intechopen.102944

From the Edited Volume

Tumor Angiogenesis and Modulators

Edited by Ke Xu

Chapter metrics overview

239 Chapter Downloads

View Full Metrics

Abstract

Since the last comprehensive assessment of antiangiogenic therapy was published in Breast Cancer Research 3 years ago, clinical trials in a variety of tumour types, including breast cancer, have underscored the key relevance of tumour neovascularization. Bevacizumab, a drug designed to target vascular endothelial cell growth factor, was utilised in many of these studies (VEGF). Clinical trials using antiangiogenic treatment in breast cancer have highlighted the critical role of tumour neovascularization. Personalised medicine will become increasingly important to generate maximum therapeutic benefit to the patient but also to realise the optimal economic advantage from the finite resources available, according to a report by the US Department of Health and Human Services (HHS) and the National Institute for Occupational and Environmental Health (NIH). This overview covers the history of breast tumour neovascularization in both in situ and invasive breast cancer, the processes by which it occurs, and the impact of the microenvironment, with a focus on hypoxia. The regulation of angiogenesis, as well as the antivascular drugs employed in antiangiogenic dosing schedules, both innovative and traditional, are discussed.

Keywords

  • angiogenesis
  • VEGF
  • breast cancer

1. Introduction

Cancer has the potential to spread to nearby or distant organs, posing a life-threatening threat. For the metastatic spread of cancer tissue, the growth of the vascular network is crucial. Angiogenesis and lymphangiogenesis are the processes by which new blood and lymphatic vessels originate.

Cancer has the potential to spread to nearby or distant organs, posing a life-threatening threat. Tumour cells can enter blood or lymphatic vessels, circulate through the intravascular stream, and then spread to a new location (metastasis) [1]. The growth of the vascular network is crucial for cancer tissue metastatic dissemination. Angiogenesis and lymphangiogenesis are the processes that result in the formation of new blood and lymphatic vessels. Both are necessary for the formation of a new vascular network that will provide nutrients, oxygen, and immune cells while also removing waste. In tumour vascularization studies, angiogenic and lymphangiogenic factors are gaining popularity.

1.1 Angiogenesis in cancer

Endothelial cells, epithelial cells, mesothelial cells, and leucocytes, as well as cancer cells and host cells, all release chemicals that aid in angiogenesis. Plateletderived endothelial cell growth factor (I’D-ECGF), plateletderived growth factor (PDGF).

Angiogenesis is a series of events that are triggered by microvascular endothelial cells. Angiogenesis and lymphangiogenesis are essential for tumour growth and metastasis, and are triggered by chemical signals from tumour cells in the early stages of development. In a prior study, Muthukkaruppan and colleagues [2] looked at how cancer cells behaved when they were placed in different parts of the same organ. Blood circulation was present in the iris, but not in the anterior chamber [2]. Cancer cells without blood circulation grew to a diameter of 1–2 mm3 and then stopped growing when placed in an area where angiogenesis was possible, but they grew to a diameter of more than 2 mm3 when placed in an area where angiogenesis was possible.

If there is insufficient blood flow, tumours can become necrotic or even apoptotic [3, 4]. Angiogenesis thus aids cancer progression. The neovascularization stage of tumour angiogenesis is one of four steps in the process. Local injury to the basement membrane occurs first in tissues. Destruction and hypoxia take place almost immediately. Angiogenic chemicals cause endothelial cells to become activated and move. Endothelial cells multiply and settle in the third step of the process. Angiogenesis is still influenced by angiogenic stimuli, according to the fourth point.

Every 1000 days on average, vascular endothelial cells divide [5]. When tumour tissues need nutrition and oxygen, angiogenesis is induced. Activators and inhibitors of angiogenesis regulate the process. On the other hand, increasing angiogenic factor activity is insufficient to enhance neoplasm angiogenesis. Negative regulators or vascular growth inhibitors must also be inhibited [6].

1.2 Breast cancer: tumour angiogenesis

Clinical studies in a range of tumour types, including breast cancer, have proven the vital role of tumour neovascularization in the 3 years after the last comprehensive review of antiangiogenic therapy was published in Breast Cancer Research [7]. Bevacizumab (AvastinTM; Genentech, South San Francisco, CA, USA) was utilised in many of these trials since it was particularly intended to target vascular endothelial cell growth factor (VEGF). Bevacizumab is a recombinant VEGF antibody that binds to all known isoforms of VEGF-A and blocks receptor interaction, inhibiting angiogenesis and tumour growth. It was made from a mouse monoclonal antibody that had been humanised. One of the successes of antiangiogenic treatment, which was first suggested by Judah Folkman more than 35 years ago, is the critical contribution of this angiogenic factor in controlling many of the processes involved in angiogenesis, as well as its importance as a paradigm for the rational design of an anticancer agent.

Because all tumours (including liquid tumours like leukaemias) are angiogenesis-dependent, angiogenesis is highly restricted in adults, the endothelium of the vessels is accessible, and any treatment would be amplified through subsequent tumour infarction, the antiangiogenic approach has always appealed to researchers. Furthermore, because endothelial cells are non-neoplastic and should have a stable genome, cancer resistance should no longer be an issue [8].

To grow larger than a few centimetres in diameter, breast cancer, like other solid tumours, requires the formation of new blood vessels (neovascularization). The extra veins not only supply more nutrients to the tumour, but they also provide possible pathways for tumour dispersal and metastasis [9].

Tumour-induced angiogenesis first develops in pre-invasive high-grade ductal carcinoma in situ. In this case, a distinctive ring of microvessels emerges around the ducts, which are packed with proliferating epithelial cells. As the tumour grows, the amount of neovascularization increases [10]. Increased microvascular density or development, as well as variables that encourage new vessel growth, have been associated to poor breast cancer prognosis.

As a result, a significant amount of study has been focused on identifying the factors in the tumour microenvironment that promote and maintain angiogenesis in the hopes of limiting neovascularization and, as a result, tumour development and dissemination. Furthermore, unlike tumour cells, which are genetically unstable and can develop resistance to many therapeutic medications fast, normal vascular endothelium lacks mutations that would allow drug resistance [11, 12]. Both research lines are investigated in this paper.

Although the presence of axillary lymph nodes is the most important prognostic marker in operable breast cancer, it does not entirely explain for the wide range of disease outcomes. More precise prognostic indications would aid in the identification of patients at high risk of illness recurrence and mortality who would benefit from systemic adjuvant therapy [13]. Microvessel density (count or grade) in invasive breast cancer (a measure of tumour angiogenesis) is associated with metastasis and so may be a prognostic sign, according to recent research.

Breast tumour growth requires angiogenesis, or the rapid formation of new blood vessels, in order to acquire enough oxygen and nutrients [14]. Breast cancer cells, like all other biological tissues, rely on a vascular network of capillaries to provide food and oxygen on a regular basis. Endothelial cells (ECs), which line the interior surface of blood vessels, do not reproduce, hence capillaries do not proliferate. Hypoxia (low oxygen) triggers a variety of transcriptional responses that are mediated by transcription factors called hypoxia-inducible factors (HIFs) [15, 16, 17, 18]. HIFs are transcription factors that regulate the expression of genes involved in physiological processes like metabolism, angiogenesis, and cell division. Local angiogenesis is one of the tumour microenvironment’s long-term major responses to low O2 levels [19, 20].

It is the fusion of EC precursors that leads to the creation of capillary plexus, which thereafter evolves into blood vessels. Angiogenesis is required for a variety of normal processes, including embryonic development, growth, and wound healing [21].

As a result, the tumour activates an angiogenic switch and enters an irreversible active angiogenic state. Because of the tumour’s newly acquired status, it can recruit new capillaries, restoring oxygen and nutrients to both angiogenic and non-angiogenic cells, resulting in rapid tumour growth [9, 22, 23, 24]. Despite the fact that surgical excision of tumours is the current standard of care for breast cancer, adjuvant therapy, such as anti-angiogenic therapy, has been used after surgery in advanced disease stages when surgery is no longer an option [25].

Angiogenic growth factors, such as vascular endothelial growth factor (VEGF) and fibroblast growth factors, are primarily involved in the initiation and progression of tumour angiogenesis (FGF). Angiogenic factor levels, as well as the number of vascular networks created as a result, have been shown to predict breast cancer survival in many studies. To put it another way, high levels imply that the tumour cells are aggressive and are linked to a poor prognosis. The rate and degree to which blood vessels permeate are controlled by these variables in connection with beginning angiogenesis [26, 27, 28, 29]. Angiogenesis-targeting compounds have recently received a lot of attention in breast cancer research.

Bevacizumab, a humanised anti-VEGF monoclonal antibody, has been the most extensively investigated molecule [30, 31, 32, 33]. After promising results in preclinical trials targeting VEGF, the FDA authorised bevacizumab in 2008 for the treatment of metastatic HER2-negative breast cancer [34, 35].

Following that, multiple anti-angiogenic medicines targeting VEGF or blocking its receptor’s action were licenced, and they are now routinely utilised in the treatment of various malignancies [36, 37, 38, 39, 40]. The FDA, however, revoked its certification in 2011 due to conflicting results from earlier trials and allegations of increased toxicity as a result [41, 42, 43, 44].

While the discovery of these anti-angiogenic drugs and small molecules was heralded as a potential victory in one aspect of the cancer fight, the agents’ modest activities, such as their inability to arrest recurrent tumours in a latent state and the moderate improvement in overall patient survival, dampened the celebration.

1.3 The angiogenic cycle

Endothelial cells in normal, quiescent capillaries are in contact with a laminin-rich basement membrane and a layer of supportive pericytes that is 1- to 2-cell thick. Angiogenesis necessitates the weakening of connections between nearby pericytes as well as the degradation of the basement membrane [45]. The integrin adhesion receptors help endothelial cells re-enter the cell cycle and infiltrate the surrounding stromal matrix. Endothelial cells begin to resynthesize a basement membrane, which aids in cell cycle exit and promotes the creation of a capillary-like morphology [46]. Pericytes are then recruited to newly formed capillaries to help mature arteries stabilise [47]. Chronic exposure to angiogenic factors in the tumour microenvironment that promote basement membrane proteolysis or antagonise endothelial–pericyte interactions leads to the formation of a relatively unstable, highly permeable network of vessels that does not fully mature but can supply nutrients to meet the tumour’s growing metabolic demands. Increased arterial permeability is thought to encourage tumour cell extravasation and, eventually, spread [48, 49].

Advertisement

2. Factors that promote angiogenesis

2.1 Hypoxia

Hypoxia has long been suspected as a significant angiogenic stimulator within the tumour microenvironment. Densely packed, quickly proliferating cells with limited nutritional inputs are the source of low tissue oxygen tension [50]. In recent years, researchers have made tremendous progress in understanding the biochemical and molecular reactions to hypoxia, as well as how the tissue senses low oxygen tension [51]. It was discovered that the hypoxia-inducible factor (HIF), a heterodimeric transcription factor made up of the hypoxic response factor (HIF-1) and the constitutively expressed aryl hydrocarbon receptor nuclear translocator (ARNT or HIF-1), is particularly significant [52, 53].

HIF-1 binds to the von Hippel-Lindau (VHL) protein in oxygenated circumstances, causing ubiquitination and fast destruction [54]. In hypoxic settings, on the other hand, this factor is stabilised: it is unable to associate with VHL protein because prolyl hydroxylase, an enzyme that typically alters HIF-1 to facilitate its interactions with VHL protein, is inactive. As a result, the oxygen sensor has been proposed as prolyl hydroxylase [55, 56, 57, 58, 59].

Animals lacking HIF-1 had markedly reduced angiogenic responses, indicating that it plays a vital role in experimental tumour growth and tumour-associated angiogenesis. Human ductal carcinomas overexpress HIF-1, whereas benign tumours with little angiogenesis do not. In the hypoxic tumour microenvironment, stabilised HIF-1 induces the expression of a variety of proangiogenic mediators, including vascular endothelial growth factor (VEGF) and one of its receptors, VEGF receptor 1 (VEGFR1) [60, 61, 62].

2.2 Vascular endothelial growth factor

VEGF is a powerful and selective endothelium mitogen that can produce a rapid and full angiogenic response, as its name suggests. VEGF (VEGF-A), the most investigated and implicated in tumour-induced angiogenesis, is a family of glycoproteins (VEGF-A, -B, -C, and -D) that are linked to VEGF (VEGF-A). The lymphatic endothelium responds to VEGF-C and -D in a big way [63].

VEGF is produced and released by a range of normal cell types, but its expression is dramatically increased in tumour cells, including a variety of breast malignancies, as well as reactive breast tumour stromal cells [64]. In contrast to other cytokines produced by tumour cells, VEGF functions almost exclusively on endothelial cells because expression of the major VEGF receptor, VEGFR2, is confined to such cells. Interfering with VEGF or VEGFR2 allows for the specific targeting of tumour endothelium [65]. VEGFR1, on the other hand, is expressed by endothelial cells, monocytes, and macrophages, and its role was unknown until recently.

When VEGF binds to its receptor, it activates an intracellular signalling cascade that causes gene expression modifications that promote endothelial cell migration and proliferation [66]. Furthermore, because VEGF not only functions as an endothelium mitogen but also increases capillary permeability, it’s not surprising that the leakiness of tumour arteries is a fundamental distinguishing feature.

2.3 VEGF and breast tumour angiogenesis

An increase in VEGF synthesis by tumour cells and cells in the tumour stroma has been connected to angiogenesis induced by breast tumours, as previously mentioned. VEGFR2 expression was also shown to be greater in the endothelial cells of the adjacent breast tumour. Indeed, higher VEGF expression correlates with the first detectable breast-tumour driven angiogenesis in pre-invasive high grade ductal carcinoma in situ [67].

The elevated expression of VEGF in the breast tumour environment is thought to be due to a number of causes. Hypoxia and HIF-1 are clearly important factors. The fact that premenopausal women had higher levels of VEGF expression than postmenopausal women suggests that steroid hormones may also boost VEGF expression [68, 69]. Estradiol has long been known to be angiogenic, and evidence suggests that oestrogen effects may be mediated through VEGF induction. In certain breast cancer cell lines, estrogens increase VEGF expression whereas progestins lower it. Tamoxifen, an oestrogen receptor inhibitor, has recently been found to reduce VEGF transcription. However, whether oestrogen receptor expression is linked to VEGF expression and vascular density has to be determined.

VEGF production is also influenced by changes in the tumour environment. Matrix metalloproteinases, for example, are frequently secreted by numerous tumour cells, including human breast cancers [70]. Matrix metalloproteinase (MMP)-9, which is produced by tumour cells and expressed at high levels in human breast cancers, is one member of this family that has attracted a lot of attention. MMP-9 has been found to proteolyze the surrounding extracellular matrix, releasing trapped VEGF and thereby enhancing its bioavailability.

The expression of HER2 is another significant alteration in breast cancers. HER2 is a tyrosine kinase receptor that belongs to the epidermal growth factor receptor family and is expressed by the ERB2 gene [71, 72]. It signals in the lack of a known ligand. Furthermore, HER2 overexpression or heregulin stimulation causes an increase in VEGF mRNA, whereas treatment of breast tumours with an anti-HER2 neutralising antibody inhibits VEGF synthesis in a dose-dependent manner. Furthermore, HER2 was found to boost the rate of HIF-1 protein production in a new, rapamycin-dependent mechanism, rather than by blocking degradation as seen during hypoxia [73, 74].

VEGF production can also be boosted by changes in epithelial gene expression linked to tumorigenicity. The 644 integrin, which typically facilitates connections between breast epithelium and basement membrane, is upregulated and mislocalized in breast carcinoma cells, promoting tumour cell invasiveness. According to recent research, 644 signalling causes the inactivation of eIF-4E, a translational repressor, which enhances VEGF translation and, in turn, tumour cell survival [75, 76, 77]. The 644 signalling pathway, which enhances VEGF translation, converges on a rapamycin-sensitive route, similar to the HER2-mediated increases in HIF-1 and VEGF. Importantly, the tumour cells’ increased VEGF production has been shown to act in an autocrine manner, promoting epithelial cell survival directly.

2.4 Mechanisms of angiogenesis

Tumour development and metastasis are dependent on angiogenesis. Necrosis occurs when a tumour’s blood supply is cut off, preventing it from growing. After a while, any further metastatic spread into the systemic circulation is stopped. Scientists have been studying angiogenesis and the different variables that regulate it in order to better understand how it affects breast cancer and develop a strategy to limit tumour progression [25, 29]. Because of the dual nature of this process, it’s critical to understand and distinguish between normal angiogenesis processes, such as wound healing, normal growth, and embryo nutrition, and tumour-related angiogenesis mechanisms.

Angiogenesis, which involves communicating between tumour cells and a variety of other cell types within the tumour microenvironment, is initiated by some compounds known as angiogenic activators because of their capacity to stimulate cell proliferation in vitro. The generation of pro-angiogenic growth factors by tumour cells, which impact the existing vasculature, has been shown to be necessary for the induction of this process [21]. To generate and stabilise newly created blood vessels, a delicate signal balance between pro- and anti-angiogenic factors is vigorously maintained in the microenvironment during these closely regulated processes [78]. As a result, numerous investigations have demonstrated that these angiogenic activators are critical in the growth of malignancies.

Certain tumour cells express both pro- and anti-angiogenic proteins, which encourage and inhibit angiogenesis, according to previous research. Tumours are thought to turn on the angiogenic switch by reversing the balance of angiogenesis inducers and inhibitors [29, 37]. This switch can be made by altering gene transcription, as seen in various cancers where VEGF and/or FGF levels are higher than in healthy tissue. The levels of endogenous inhibitors are lowered in some cancers, on the other hand. The intricate mechanism that drives these alterations in the regulators’ balances, on the other hand, remains a fascinating subject of research (Figure 1).

Figure 1.

Angiogenesis is a physiological process that results in the formation of new blood vessels from existing ones. From pre-existing capillaries, new blood vessels emerge. The tumour receives crucial nutrients for growth from the new blood vessels that have sprouted near and within the tumour. Angiogenesis in healthy tissues is regulated by a balance of anti- and pro-angiogenic factors (bottom), but the presence of angiogenic factors in tumours disrupts this balance, resulting in abnormal blood vessel structure and function, as well as hypoxia. The vasculature is normalised and the balance is restored.

The tumours ability to switch on angiogenesis is determined by the balance of this switch. Further research revealed that a decrease in anti-angiogenic protein production activates the tumour angiogenic switch, promoting tumour growth and metastasis [79, 80, 81]. Stimulating angiogenesis in a tumour and forming the endothelial tubes that result is a multistep process governed by hypoxia at each stage. This pathway is heavily reliant on ECs expressing HIF-1, a heterodimeric transcription factor. Under hypoxic conditions, the HIF-1 protein is stabilised and forms a heterodimer with HIF-1, and this pair promotes the transcription of multiple target genes to adapt to the hypoxic environment in human cancer cells.

HIF-1, in conjunction with other members of the HIF family, has been demonstrated in certain studies to govern practically every element of angiogenesis, making the HIF pathway a master regulator of angiogenesis [82]. In various malignancies, HIF-1 and HIF-2 expression has also been linked to a poor prognosis and metastatic illness. As a result, it’s regarded as a promising therapeutic target for a variety of medical conditions (Figure 2).

Figure 2.

This figure depicts the balance hypothesis of the angiogenic switch. Angiogenesis switch mechanism is assumed to be in charge of normal angiogenesis (formation of new capillaries). By utilising angiogenesis inducers and inhibitors, which flip the switch, this balance can be tilted in favour of enhanced blood vessel formation. Reduced inhibitor levels (thrombospondin-1, 16 kD prolactin, interferon, platelet factor-4, Angiostatin, and others) or increased activator levels (aFGF, bFGF, VEGF, and others) can tip the balance and activate the switch, resulting in the formation of new blood vessels.

Hypoxia and activation of the HIF pathway in cancer cells are required for the sprouting and formation of new blood vessels because they control the expression of several pro-angiogenic genes [83]. Some of the most powerful cytokines are VEGF, an endothelial mitogen and pro-angiogenic factor, angiopoietin-1, angiopoietin-2, platelet-derived growth factor (PDGF), and basic fibroblast growth factor (bFGF) [84, 85, 86, 87, 88].

The FGF and VEGF families of angiogenetic growth factors have gotten more attention than the others. In 1983, the protein VEGF-A (vascular endothelial growth factor) was identified and sequenced. It was the first cytokine to be identified as a key contributor to tumour angiogenesis, was purified from tumour cell ascites as vascular permeability factor (VPF), and was also revealed to have pharmacological effects on EC mitogenesis; consequently, VPF is referred to as VEGF (Figure 3) [89, 90].

Figure 3.

This diagram depicts the receptor binding selectivity and signalling pathways of members of the vascular endothelial growth factor (VEGF) family. VEGF family members bind to VEGFR-1, VEGFR-2, and VEGFR-3 receptor tyrosine kinases, which activate a variety of signalling pathways and allow them to exert their physiological effects.

In vivo and in vitro, VEGF is now known to be a multifunctional peptide capable of triggering receptor-mediated endothelial cell proliferation and angiogenesis. The VEGF family contains at least five members, each of which has three VEGF receptors (VEGFR) [91, 92, 93]. These receptors use transmembrane receptor tyrosine kinases to communicate with the cell’s interior (RTKs). The VEGF gene is subject to complex transcriptional control, and four distinct RNA isoforms are produced with varying biological features as a result of alternative splicing of its pre-mRNAs. VEGF-B, VEGF-C, VEGF-D, VEGF-E, and platelet-derived growth factor are all produced as a result of this process (PDGF).

By attaching to VEGF receptors and ligands, VEGF, for example, can trigger angiogenesis. The effects of vascular endothelial growth factor (VEGF), as well as acidic and basic fibroblast growth factors (FGF1/2), can be employed to investigate the induction and progression of angiogenesis at various phases of tumour development. VEGF binds to its receptor (VEGFR) and ligands on the surface of ECs. It causes dimerization, autophosphorylation, and activation of the downstream signalling cascade after binding to and activating the transmembrane tyrosine kinase receptors on the cell’s surface [94, 95, 96]. Tube development and sprouting follow EC survival, proliferation, migration, and apoptosis avoidance through several cascade phases. Over time, this process results in the development of a complex network of new blood vessels. Vasodilation and vascular permeability, a key feature of tissue inflammation and the tumour microenvironment, are also induced by VEGF [97, 98, 99, 100, 101, 102, 103].

The activity of the ECs outlined above is caused by an increase in pro-angiogenic factors such as VEGF and proteolytic enzymes, as well as a decrease in anti-angiogenic factors. Finally, a capillary network is successfully established, supplying enough nutrition and oxygen to the growing tumour. Taking advantage of this new vascular bed, the tumour cell may reach the systemic circulation and induce distant metastases. As a result, the number of metastasis sites is proportional to the amount of cancer cells that enter the circulation at the outset [104, 105, 106, 107, 108, 109, 110, 111].

Angiogenic inducers have been implicated in the regulating process of angiogenesis in malignancies since their discovery a decade ago. Anti-angiogenic treatment decreases tumour vascular growth by interfering with VEGF and VEGFR intracellular signalling [112, 113, 114, 115, 116].

Angiogenesis was originally linked to cancer, arthritis, and psoriasis. However, the impact it has on a variety of other disorders has been documented. Tumours are innately primed for successful angiogenic development due to their nature and composition. An active vascular system is made up of adipose tissue that is encased in stromal cells and serves as a scaffold for the tumour’s vascular system to emerge [117, 118, 119].

Brown adipose tissue (made up of cells with numerous mitochondria) promotes tumour growth by supplying a steady supply of oxygen and nutrients, whereas white adipose tissue promotes the formation and progression of breast cancer in a mouse model. Both types of adipose tissues, which have been associated to breast cancer, produce angiogenic factors such as VEGF A, B, and C, basic fibroblast growth factor (bFGF)/FGF-2, matrix metalloproteinases (MMPs), and IL-8. This aberrant blood vessel creation has been linked to cardiovascular illness, cancer, blindness, and diabetic ulcers [120, 121, 122].

2.5 Non-angiogenic functions of VEGF in breast cancer

VEGF increases the formation of new blood vessels and lymphatics, as well as increasing vascular permeability, and has a variety of tumour-related effects. The importance of VEGF in vascular and lymphangiogenesis has dominated research in breast and other cancers [123]. The importance of VEGF in cancer behaviour cannot be overstated. The presence of hypoxic patches in most malignancies, on the other hand, implies that VEGF-induced angiogenesis is insufficient to alleviate hypoxia [124]. Hypoxia works as a strong selection pressure, allowing only the most aggressive and metastatic cells to thrive. Understanding the mechanisms that allow tumour cells to survive under hypoxia is therefore critical for interpreting cancer biology and developing therapeutic approaches [125, 126].

VEGF produced by tumour or stromal cells interacts to VEGF receptors on tumour cells, producing a signalling response that supports survival in the face of hypoxia and other apoptotic triggers, according to our and other labs’ research [127]. This process, which most likely operates in tandem with p53 inactivation, provides self-sufficiency to tumour cells, making it simpler for them to form tumours and increasing the possibility that they will spread to other parts of the body [128, 129]. To put it another way, we believe that hypoxia favours cells that can signal VEGF, and that the most aggressive tumour cells (metastatic cells) are determined by their dependency on VEGF.

A side effect of VEGF signalling in breast cancer cells is that it can help them move and invade more easily.

Advertisement

3. Breast carcinoma cells and VEGF signalling

3.1 Survival signalling by autocrine VEGF

Tumour cells receive signals from various sources as a result of the complex microenvironment of solid tumours, and these signals alter the activity of other cells. However, it is becoming obvious that cancer cells can attain a certain level of self-sufficiency by creating autocrine signalling pathways that aid critical tasks such as growth, survival, and invasion [130] within this web of paracrine signalling. As tumours develop towards invasive and metastatic illness, autocrine pathways become more critical as the tumour’s environment becomes increasingly hostile. As a result, autocrine signalling pathways are a major target for anti-tumour therapy. Our study on invasive breast carcinoma cell lines provided one of the first indications that VEGF may have autocrine functions in cancer [73, 131].

We discovered that a 50% reduction in VEGF expression resulted in a considerable increase in apoptosis, even in the presence of 10% serum, when we utilised an antisense oligonucleotide approach to limit VEGF expression. This evidence backs with the theory that these cells were selected in vivo because they rely on VEGF to survive [132]. The importance of VEGF in carcinoma and other cancer cell survival has now been validated by research from our lab and others.

Because it increased VEGF expression in invasive breast cancer cell lines, hypoxia inhibited apoptosis caused by serum deprivation. The mechanism by which autocrine VEGF maintains the survival of breast carcinoma cells appears to involve constitutive activation of the PI3-kinase pathway, as evidenced by the findings that reducing VEGF expression results in a significant decrease in PI3-kinase basal activity, hypoxia stimulates Akt activity, and inhibition of PI3-kinase induces apoptosis [133]. According to previous studies, VEGF inhibits apoptosis in breast cancer cells via upregulating the anti-apoptotic protein Bcl-2.

3.2 The role of VEGF in breast carcinoma migration and invasion

Carcinoma cells acquire the ability to migrate and infiltrate tissues as a result of malignant transformation and development. Although chemoattractant gradients may enhance carcinoma migration and invasion, it has been established that cells’ ability to form autocrine signalling pathways might boost their sensitivity to external stimuli [134]. Depleting VEGF expression in the presence of caspase inhibitors, which prevent apoptosis caused by VEGF expression loss, allowed us to find a role for autocrine VEGF in the migration and invasion of breast cancer cells towards chemokines. The capacity of breast cancer cells to migrate and invade in response to chemotactic stimuli is considerably diminished in such circumstances.

One mechanism for VEGF’s involvement in these events is its ability to alter the expression of the chemokine receptor CXCR4 [135]. This finding is significant for breast cancer growth since stromal-derived factor-1, the receptor’s ligand, is abundant in tumour stroma as well as organs such as the lymph and lung, which are the primary targets of invasive breast carcinoma cells, and CXCR4 inhibitors impede metastasis [136].

In addition to its survival benefits, VEGF autocrine signalling may contribute to tumour growth by boosting chemokine receptor expression and allowing tumour cells to migrate towards chemokine gradients [137].

3.3 Perspective

The revelation that breast cancer cells produce VEGF receptors is significant, but further research is needed to understand how these receptors are expressed as a result of transformation and progression, including EMT, and the mechanisms through which these receptors regulate tumour cell behaviour. Despite having inherent signalling capabilities, little is known about how NP-1 enhances VEGF165 signalling on breast cancer cells. In endothelial cells, it appears to work with either VEGFR1 or VEGFR2, although this has yet to be validated in breast cancer cells.

Another hypothesis is that NP-1 transmits NP-1 signals in neurons via interacting with non-VEGF receptors in cancer cells, such as plexins. Our findings reveal that plexin A1 is expressed in breast cancer cells and can affect cell motility. The study of plexin involvement in NP-1 signalling will require a much more in-depth understanding of plexin expression and function in breast and other cancers. In addition, more exact data on the location and relative expression of NP-1 in the mammary gland and human breast malignancies is needed.

VEGF-C and VEGF-D, for example, have been linked to angiogenesis and lymphangiogenesis in breast tumours. It’s critical to figure out whether these VEGFs have a paracrine or autocrine effect on breast cancer cells. Some data suggests that breast cancer cells can respond to VEGF-D autocrinely, although additional research is needed to confirm this.

References

  1. 1. Folkman J. Tumor angiogenesis theraperutic implications. The New England Journal of Medicine. 1971;285:1182-1186
  2. 2. Muthukkaruppan VR, Kubai L, Auerbach R. Tumor-induced neovascularization in the mouse eye. Journal of the National Cancer Institute. 1982;69:699-708
  3. 3. Holmgren L, O’Reilly MS, Folkman J. Dormancy of micrometastases: Balance proliferation and apoptosis in the presence of angiogenesis suppression. Nature Medicine. 1995;1:149-153
  4. 4. Parangi S, O’Reilly M, Christofori G, et al. Angiogenesis therapy of transgenic mice impairs de novo tumor growth. Proceedings of the National Academy of Sciences of the United States of America. 1996;93:2002-2007
  5. 5. Denekamp J. Angiogenesis, neovascular proliferation and vascular pathophysiology as targets for cancer therapy. The British Journal of Radiology. 1993;66:181-196
  6. 6. Dameron KM, Volpert OV, Tainsky MA, et al. Control of angiogenesis in fibroblasts by p53 regulation of thorombospongin-1. Science. 1994;265:1582-1584
  7. 7. Miller KD. Recent translational research: Antiangiogenic therapy for breast cancer: Where do we stand? Breast Cancer Research. 2004;6:128-132
  8. 8. Kerbel RS. A cancer therapy resistant to resistance. Nature. 1997;390:335-336
  9. 9. Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nature Medicine. 1995;1:27-31
  10. 10. Engels K, Fox SB, Whitehouse RM, Gatter KC, Harris AL. Distinct angiogenic patterns are associated with high-grade in situ ductal carcinomas of the breast. The Journal of Pathology. 1997;181:207-212
  11. 11. Gasparini G, Harris AL. Clinical importance of the determination of tumor angiogenesis in breast carcinoma: Much more than a new prognostic tool. Journal of Clinical Oncology. 1995;13:765-782
  12. 12. Linderholm B, Lindh B, Tavelin B, Grankvist K, Henriksson R. p53 and vascular endothelial growth factor expression predicts outcome in 833 patients with primary breast carconoma. International Journal of Cancer. 2000;89:51-62
  13. 13. Siourdsson H, Baldetorp B, Boro A, et al. Indicators of prognosis in node-negative breast cancer. The New England Journal of Medicine. 1990;322:1045-1053
  14. 14. American Cancer Society. Available from: https://www.cancer.org/research/cancer-facts-statistics/breast-cancer-facts-figures.html
  15. 15. DeSantis C, Ma J, Bryan L, Jemal A. Breast cancer statistics, 2013. CA Cancer Journal. 2014;64:52-62
  16. 16. American Cancer Society. Breast Cancer Facts & Figures 2015-2016. Atlanta: American Cancer Society, Inc.; 2015
  17. 17. Castañeda-Gill JM, Vishwanatha JK. Antiangiogenic mechanisms and factors in breast cancer treatment. Journal of Carcinogenesis. 2016;15:1
  18. 18. Coelho AL, Gomes MP, Catarino RJ, et al. Angiogenesis in NSCLC: Is vessel co-option the trunk that sustains the branches? Oncotarget. 2017;8:39795-39804
  19. 19. Sim EK, Zhang L, Shim WS, Lim YL, Ge R. Therapeutic angiogenesis for coronary artery disease. Journal of Cardiac Surgery. 2002;17:350-354
  20. 20. Risau W, Flamme I. Vasculogenesis. Annual Review of Cell and Developmental Biology. 1995;11:73-91
  21. 21. Hanahan D, Weinberg RA. Weinberg, Hallmarks of cancer: The next generation. Cell. 2011;144:646-674
  22. 22. Folkman J, Browder T, Palmblad J. Angiogenesis research: Guidelines for translation to clinical application. Thrombosis and Haemostasis. 2001;86:23-33
  23. 23. Tannock IF. The relation between cell proliferation and the vascular system in a transplanted mouse mammary tumour. British Journal of Cancer. 1968;22:258-273
  24. 24. Carmeliet P. Angiogenesis in life, disease and medicine. Nature. 2005;438:932-936
  25. 25. Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 1996;86:353-364
  26. 26. Grimm D, Wehland M, Pietsch J, et al. Drugs interfering with apoptosis in breast cancer. Current Pharmaceutical Design. 2011;17:272-283. DOI: 10.2174/138161211795049723
  27. 27. Grimm D, Bauer J, Schönberger J. Blockade of neoangiogenesis, a new and promising technique to control the growth of malignant tumors and its metastases. Current Vascular Pharmacology. 2009;7:347-357
  28. 28. Folkman J, Shing Y. Angiogenesis. The Journal of Biological Chemistry. 1992;267:10931-10934
  29. 29. Folkman J. Tumour angiogenesis: Therapeutic implications. The New England Journal of Medicine. 1971;285:1182-1186
  30. 30. Fong G, Rossant J, Gartsenstein M, et al. Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature. 1995;376:67-70
  31. 31. Shalably F, Rossant J, Yamaguchi TP, et al. Failure of blood island formation and vasculogenesis in FLK-1 deficient mice. Nature. 1995;376:62-66
  32. 32. Horak ER, Klenk N, Leek R, et al. Angiogenesis, assessed by platelet/EC adhesion molecule antibodies, as indicator of node metastases and survival in breast cancer. Lancet. 1992;340:1120-1124
  33. 33. Vartanian RK, Weidner N. Correlation of intratumoral EC proliferation with microvessel density (tumor angiogenesis) and tumor cell proliferation in breast carcinoma. The American Journal of Pathology. 1994;144:1188-1194
  34. 34. Weidner N, Semple JP, Welch WR, et al. Tumor angiogenesis and metastasis– correlation in invasive breast carcinoma. The New England Journal of Medicine. 1991;324:1-8
  35. 35. Linderholm B, Tavelin B, Grankvist K, et al. Does vascular endothelial growth factor (VEGF) predict local relapse and survival in radiotherapytreated node-negative breast cancer? British Journal of Cancer. 1999;81:727-732
  36. 36. George ML, Tutton MG, Janssen F, et al. VEGF-A, VEGF-C, and VEGF-D in colorectal cancer progression. Neoplasia. 2001;3:420-427
  37. 37. Holmgren L, O’Reilly MS, Folkman J. Dormancy of micrometastases: Balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nature Medicine. 1995;1:149-153
  38. 38. Linderholm BK, Hellborg H, Johansson U, et al. Significantly higher levels of vascular endothelial growth factor (VEGF) and shorter survival times for patients with primary operable triple-negative breast cancer. Annals of Oncology. 2009;20:1639-1646
  39. 39. FDA Approval for Bevacizumab. 2015. Available from: http://www.cancer.gov/cancertopics/druginfo/fda-bevacizumab
  40. 40. Tarallo V, De Falco S. The vascular endothelial growth factors and receptors family: Up to now the only target for anti-angiogenesis therapy. The International Journal of Biochemistry & Cell Biology. 2015;64:185-189
  41. 41. Bellou S, Pentheroudakis G, Murphy C, et al. Anti-angiogenesis in cancer therapy: Hercules and Hydra. Cancer Letters. 2013;338:219-228
  42. 42. Ivy SP, Wick JY, Kaufman BM. An overview of small-molecule inhibitors of VEGFR signaling. Nature Reviews: Clinical Oncology. 2009;6:569-579
  43. 43. Miles DW, Chan A, Dirix LY, et al. Phase III study of bevacizumab plus docetaxel compared with placebo plus docetaxel for the first-line treatment of human epidermal growth factor receptor 2-negative metastatic breast cancer. Journal of Clinical Oncology. 2010;28:3239-3247
  44. 44. Robert NJ, Dieras V, Glaspy J, 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. Journal of Clinical Oncology. 2011;29:1252-1260
  45. 45. Elcieri BP, Cheresh DA. Adhesion events in angiogenesis. Current Opinion in Cell Biology. 2001;13:563-568
  46. 46. Kubota Y, Kleinman HK, Martin GR, Lawley TJ. Role of laminin and basement membrane in the morphological differentiation of human endothelial cells into capillary-like structures. The Journal of Cell Biology. 1988;107:1589-1596
  47. 47. Wang GL, Jiang BH, Rue EA, Semenza GL. Hypoxia inducible factor is a basic helix-loop-helix PAS heterodimer regulated by cellular oxygen tension. Proceedings of the National Academy of Science USA. 1995;92:5510-5514
  48. 48. Masson N, Willam C, Maxwell PH, Pugh CW, Ratcliffe PJ. Independent function of two destruction domains in hypoxiainducible factor-alpha chains activated by prolyl hydroxylation. The EMBO Journal. 2001;20:5197-5206
  49. 49. Maxwell PH, Dachs GU, Gleadle JM, Nicholls LG, Harris AL, Stratford IJ, et al. Hypoxia-inducible factor-1 modulates gene expression in solid tumors and influences both angiogenesis and tumor growth. Proceedings of the National Academy of Science USA. 1997;94:8104-8109
  50. 50. Carmeliet P, Dor Y, Herbert JM, Fukumura D, Brusselmans K, Dewerchin M, et al. Role of Hif-1alpha in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature. 1998;394:485-490
  51. 51. Bos R, Zhong H, Hanrahan CF, Mommers EC, Semenza GL, Pinedo HM, et al. Levels of hypoxia-inducible factor-1 alpha during breast carcinogenesis. Journal of the National Cancer Institute. 2001;93:309-314. DOI: 10.1093/jnci/93.4.309
  52. 52. Forsythe JA, Jiang BH, Iyer NV, Agani F, Leung SW, Koos RD, et al. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Molecular and Cellular Biology. 1996;16:4604-4613
  53. 53. Gerber HP, Condorelli F, Park J, Ferrara N. Differential transcriptional regulation of the two vascular endothelial growth factor receptor genes. Flt-1, but not Flk-1/KDR, is up-regulated by hypoxia. The Journal of Biological Chemistry. 1997;272:23659-23667
  54. 54. Olofsson B, Jeltsch M, Eriksson U, Alitalo K. Current biology of Vegf-B and Vegf-C. Current Opinion in Biotechnology. 1999;10:528-535
  55. 55. Brown LF, Guidi AJ, Schnitt SJ, Van De Water L, Iruela-Arispe ML, Yeo TK, et al. Vascular stroma formation in carcinoma in situ, invasive carcinoma, and metastatic carcinoma of the breast. Clinical Cancer Research. 1999;5:1041-1056
  56. 56. Hattori K, Heissig B, Wu Y, Dias S, Tejada R, Ferris B, et al. Placental growth factor reconstitutes hematopoiesis by recruiting VEGFR1(+) stem cells from bonemarrow microenvironment. Nature Medicine. 2002;8:841-849
  57. 57. Luttun A, Tjwa M, Moons L, Wu Y, Angelillo-Scherrer A, Liao F, et al. Revascularization of ischemic tissues by PlGF treatment, and inhibition of tumor angiogenesis, arthritis and atherosclerosis by anti-Flt1. Nature Medicine. 2002;8:831-840
  58. 58. Dvorak HF, Sioussat TM, Brown LF, Berse B, Nagy JA, Sotrel A, et al. Distribution of vascular permeability factor (vascular endothelial growth factor) in tumors: Concentration in tumor blood vessels. The Journal of Experimental Medicine. 1991;174:1275-1278. DOI: 10.1084/jem.174.5.1275
  59. 59. Hashizume H, Baluk P, Morikawa S, McLean JW, Thurston G, Roberge S, et al. Openings between defective endothelial cells explain tumor vessel leakiness. The American Journal of Pathology. 2000;156:1363-1380. DOI: 10.1016/s0002-9440(10)65006-7
  60. 60. Guidi AJ, Schnitt SJ, Fischer L, Tognazzi K, Harris JR, Dvorak HF, et al. Vascular permeability factor (vascular endothelial growth factor) expression and angiogenesis in patients with ductal carcinoma in situ of the breast. Cancer. 1997;80:1945-1953
  61. 61. Greb RR, Maier I, Wallwiener D, Kiesel L. Vascular endothelial growth factor a (Vegf-a) mRNA expression levels decrease after menopause in normal breast tissue but not in breast cancer lesions. British Journal of Cancer. 1999;81:225-231
  62. 62. Losordo DW, Isner JM. Estrogen and angiogenesis: A review. Arteriosclerosis, Thrombosis, and Vascular Biology. 2001;21:6-12
  63. 63. Hyder SM, Murthy L, Stancel GM. Progestin regulation of vascular endothelial growth factor in human breast cancer cells. Cancer Research. 1998;58:392-395
  64. 64. Ruohola JK, Valve EM, Karkkainen MJ, Joukov V, Alitalo K, Harkonen PL. Vascular endothelial growth factors are differentially regulated by steroid hormones and antiestrogens in breast cancer cells. Molecular and Cellular Endocrinology. 1999;149:29-40
  65. 65. Buteau-Lozano H, Ancelin M, Lardeux B, Milanini J, PerrotApplanat M. Transcriptional regulation of vascular endothelial growth factor by estradiol and tamoxifen in breast cancer cells: A complex interplay between estrogen receptors alpha and beta. Cancer Research. 2002;62:4977-4984
  66. 66. Takei H, Lee ES, Jordan CV. In vitro regulation of vascular endothelial growth factor by estrogens and antiestrogens in estrogen-receptor positive breast cancer. Breast Cancer. 2002;9:39-42
  67. 67. Scorilas A, Karameris A, Arnogiannaki N, Ardavanis A, Bassilopoulos P, Trangas T, et al. Overexpression of matrix-metalloproteinase-9 in human breast cancer: A potential favourable indicator in node-negative patients. British Journal of Cancer. 2001;84:1488-1496
  68. 68. Bergers G, Brekken R, McMahon G, Vu TH, Itoh T, Tamaki K, et al. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nature Cell Biology. 2000;2:737-744
  69. 69. Bagheri-Yarmand R, Vadlamudi RK, Wang RA, Mendelsohn J, Kumar R. Vascular endothelial growth factor up-regulation via p21-activated kinase-1 signaling regulates heregulin-beta1- mediated angiogenesis. The Journal of Biological Chemistry. 2000;275:39451-39457
  70. 70. Yen L, You XL, Al Moustafa AE, Batist G, Hynes NE, Mader S, et al. Heregulin selectively upregulates vascular endothelial growth factor secretion in cancer cells and stimulates angiogenesis. Oncogene. 2000;19:3460-3469
  71. 71. Petit AM, Rak J, Hung MC, Rockwell P, Goldstein N, Fendly B, et al. Neutralizing antibodies against epidermal growth factor and ErbB-2/neu receptor tyrosine kinases down-regulate vascular endothelial growth factor production by tumor cells in vitro and in vivo: Angiogenic implications for signal transduction therapy of solid tumors. The American Journal of Pathology. 1997;151:1523-1530
  72. 72. Laughner E, Taghavi P, Chiles K, Mahon PC, Semenza GL. HER2 (neu) signaling increases the rate of hypoxia-inducible factor 1alpha (HIF-1alpha) synthesis: Novel mechanism for HIF-1- mediated vascular endothelial growth factor expression. Molecular and Cellular Biology. 2001;21:3995-4004
  73. 73. Chung J, Bachelder RE, Lipscomb EA, Shaw LM, Mercurio AM. Integrin (alpha 6 beta 4) regulation of eIF-4E activity and VEGF translation: A survival mechanism for carcinoma cells. The Journal of Cell Biology. 2002;158:165-174
  74. 74. Edel MJ, Harvey JM, Papadimitriou JM. Comparison of vascularity and angiogenesis in primary invasive mammary carcinomas and in their respective axillary lymph node metastases. Clinical & Experimental Metastasis. 2000;18:695-702
  75. 75. Monsky WL, Mouta Carreira C, Tsuzuki Y, Gohongi T, Fukumura D, Jain RK. Role of host microenvironment in angiogenesis and microvascular functions in human breast cancer xenografts: Mammary fat pad versus cranial tumors. Clinical Cancer Research. 2002;8:1008-1013
  76. 76. Lee JC, Kim DC, Gee MS, Saunders HM, Sehgal CM, Feldman MD, et al. Interleukin-12 inhibits angiogenesis and growth of transplanted but not in situ mouse mammary tumor virus-induced mammary carcinomas. Cancer Research. 2002;62:747-755
  77. 77. Animal models to study mammary gland development, physiology and tumorigenesis. Available from: http://mammary.nih.gov/models/index.html
  78. 78. Fidler I, Ellis L. The implications of angiogenesis for the biology and therapy of cancer metastasis. Cell. 1994;79:185-188
  79. 79. Papetti M, Herman IM. Mechanisms of normal and tumor-derived angiogenesis. American Journal of Physiology: Cell Physiology. 2002;282:947-970
  80. 80. Liekens S, De Clercq E, Neyts J. Angiogenesis: Regulators and clinical applications. Biochemical Pharmacology. 2001;61:253-270
  81. 81. Maj E, Papiernik D, Wietrzyk J. Antiangiogenic cancer treatment: The great discovery and greater complexity (review). International Journal of Oncology. 2016;49:1773-1784
  82. 82. Singh RK, Gutman M, Bucana CD, et al. Sequential development of an angiogenic phenotype by human fibroblasts progressing to tumorigenicity. Proceedings of the National Academy of Science USA. 1995;92:4562-4566
  83. 83. Kandel J, Bossy-Wetzel E, Radvanyi F, et al. Neovascularization is associated with a switch to the export of bFGF in the multistep development of fibrosarcoma. Cell. 1991;66:1095-1104
  84. 84. Folkman J, Hanahan D. Switch to the angiogenic phenotype during tumorigenesis. Princess Takamatsu Symposia. 1991;22:339-347
  85. 85. Good DJ, Polverini PJ, Rastinejad F, et al. A tumor suppressor–dependent inhibitor of angiogenesis is immunologically and functionally indistinguishable from a fragment of thrombospondin. Proceedings of the National Academy of Science USA. 1990;87:6624-6628
  86. 86. Rastinejad F, Polverini PJ, Bouck NP. Regulation of the activity of a new inhibitor of angiogenesis by a cancer suppressor gene. Cell. 1989;56:345-355
  87. 87. Tang N, Wang L, Esko J, et al. Loss of HIF-1alpha in endothelial cells disrupts a hypoxia-driven VEGF autocrine loop necessary for tumorigenesis. Cancer Cell. 2004;6:485-495
  88. 88. Harris AL. Hypoxia—A key regulatory factor in tumour growth. Nature Reviews: Cancer. 2002;2:38-47. DOI: 10.1038/nrc704
  89. 89. Semenza GL. HIF-1 and tumor progression: Pathophysiology and therapeutics. Trends in Molecular Medicine. 2002;8:S62-S67
  90. 90. Zhong H, De Marzo AM, Laughner E, et al. Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Research. 1999;59:5830-5835
  91. 91. Bertout JA, Patel SA, Simon MC. The impact of O2 availability on human cancer. Nature Reviews: Cancer. 2008;8:967-975
  92. 92. Mazure NM, Brahimi-Horn MC, Berta MA, Benizri E, Bilton RL, Dayan F, et al. HIF-1: Master and commander of the hypoxic world: A pharmacological approach to its regulation by siRNAs. Biochemical Pharmacology. 2004;68:971-980
  93. 93. Semenza GL. Targeting HIF-1 for cancer therapy. Nature Reviews: Cancer. 2003;3:721-732
  94. 94. Claesson-Welsh L, Welsh MJ. VEGFA and tumour angiogenesis. Internal Medicine. 2013;273:114-127
  95. 95. Mendelsohn J, Howley P, Israel M, Liotta L. The Molecular Basis of Cancer. Philadelphia: W. B. Saunders; 1995. pp. 206-232
  96. 96. Senger DR, Galli SJ, Dvorak AM, et al. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science. 1983;219:983-985
  97. 97. Felmeden DC, Blann AD, Lip GYH. Angiogenesis: Basic pathophysiology and implications for disease. European Heart Journal. 2003;24:586-603
  98. 98. Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science. 1989;246:1306-1309
  99. 99. Keck PJ, Hauser SD, Krivi G, et al. Vascular permeability factor, an endothelial cell mitogen related to PDGF. Science. 1989;246:1309-1312
  100. 100. Connolly DT, Heuvelman DM, Nelson R, et al. Tumor vascular permeability factor stimulates endothelial cell growth and angiogenesis. The Journal of Clinical Investigation. 1989;84:1470-1478
  101. 101. Tischer E, Gospodarowicz D, Mitchell R, et al. Vascular endothelial growth factor: A new member of the platelet derived growth factor gene family. Biochemical and Biophysical Research. 1989;165:1198-1206
  102. 102. Ferrara N, Houck K, Jakeman L, et al. Molecular and biological properties of vascular endothelial growth factor family of protein. Endocrine Reviews. 1992;13:18-32
  103. 103. Kristensen TB, Knutsson MLT, Wehland M, et al. Anti-vascular endothelial growth factor therapy in breast cancer. International Journal of Molecular Sciences. 2014;15:23024-23041
  104. 104. Maxwell PH, Wiesener MS, Chang G-W, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399:271-275
  105. 105. Park JE, Keller GA, Ferrara N. The vascular endothelial growth factor (VEGF) isoforms: Differential deposition into the subepithelial extracellular matrix and bioactivity of extracellular matrix-bound VEGF. Molecular Biology of the Cell. 1993;4:1317-1326
  106. 106. Li B, Leung DW, et al. The vascular endothelial growth factor family: Identification of a fourth molecular species and characterization of alternative splicing of RNA. Molecular Endocrinology. 1991;5:1806-1814
  107. 107. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nature Medicine. 2003;9:669-676
  108. 108. Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor signalling—In control of vascular function. Nature Reviews: Molecular Cell Biology. 2006;7:359-371
  109. 109. Bottaro DP, Liotta LA. Cancer: Out of air is not out of action. Nature. 2003;423:593-595
  110. 110. Zhao Y, Adjei AA. Targeting angiogenesis in cancer therapy: Moving beyond vascular endothelial growth factor. The Oncologist. 2015;20:660-673
  111. 111. Koch S, Claesson-Welsh L. Signal transduction by vascular endothelial growth factor receptors. Cold Spring Harbor Perspectives in Medicine. July 2012;2(7):a006502
  112. 112. Pepper MS, Ferrara N, Orci L, Montesano R. Potent synergism between vascular endothelial growth factor and basic fibroblast growth factor in the induction of angiogenesis in vitro. Biochemical and Biophysical Research Communications. 1992;189:824-831
  113. 113. Korpanty G, Smyth E, Carney DN. Update on anti-angiogenic therapy in non-small cell lung cancer: Are we making progress? Journal of Thoracic Disease. 2011;3:19-29
  114. 114. Heusschen R, van Gink M, Griffioen AW, Thijssen VL. MicroRNAs in the tumor endothelium: Novel controls on the angioregulatory switchboard. Biochimica et Biophysica Acta. 2010;1805:87-96
  115. 115. Wahl ML, Moser TL, Pizzo SV. Angiostatin and anti-angiogenic therapy in human disease. Recent Progress in Hormone Research. 2004;59:73-104
  116. 116. Lee SH, Jeung IC, Park TW, et al. Extension of the in vivo half-life of endostatin and its improved anti-tumor activities upon fusion to a humanized antibody against tumor-associated glycoprotein 72 in a mouse model of human colorectal carcinoma. Oncotarget. 2015;6:7182-7194
  117. 117. Caporali A, Emanueli C. MicroRNA regulation in angiogenesis. Vascular Pharmacology. 2011;55:79-86
  118. 118. Butler TP, Gullino PM. Quantitation of cell shedding into efferent blood of mammary adenocarcinoma. Cancer Research. 1975;35:512-516
  119. 119. Bouck N, Stellmach V, Hsu S. How tumors become angiogenic. Advances in Cancer Research. 1996;69:135-174
  120. 120. Folkman J. Tumor angiogenesis. In: The Molecular Basis of Cancer. 1995. Available from: https://www.nature.com/articles/nm0195-27#auth-Judah-Folkman. DOI: 10.1016/s0065-230x(08)60946-x
  121. 121. Folkman J. Clinical applications of research on angiogenesis. The New England Journal of Medicine. 1995;333:1757-1763. DOI: 10.1056/NEJM199512283332608
  122. 122. Wehland M, Bauer J, Magnusson NE, et al. Biomarkers for anti-angiogenic therapy in cancer. International Journal of Molecular Sciences. 2013;14:9338-9364. DOI: 10.3390%2Fijms14059338
  123. 123. Dvorak HF, Nagy JA, Feng D, Brown LF, Dvorak AM. Vascular permeability factor/vascular endothelial growth factor and the significance of microvascular hyperpermeability in angiogenesis. Current Topics in Microbiology and Immunology. 1999;237:97-132
  124. 124. Hockel M, Vaupel P. Tumor hypoxia: Definitions and current clinical, biologic, and molecular aspects. Journal of the National Cancer Institute. 2001;93:266-276
  125. 125. Bachelder RE, Crago A, Chung J, Wendt MA, Shaw LM, Robinson G, et al. Vascular endothelial growth factor is an autocrine survival factor for neuropilin-expressing breast carcinoma cells. Cancer Research. 2001;61:5736-5740
  126. 126. Harmey JH, Bouchier-Hayes D. Vascular endothelial growth factor (VEGF), a survival factor for tumour cells: Implications for anti-angiogenic therapy. BioEssays. 2002;24:280-283
  127. 127. Bates RC, Goldsmith JD, Bachelder RE, Brown C, Shibuya M, Oettgen P, et al. Flt-1 (VEGFR-1)-dependent survival characterizes the epithelial-mesenchymal transition of colonic organoids. Current Biology. 2003;13:1721-1727
  128. 128. Sharieff W. Bevacizumab in colorectal cancer. New England Journal of Medicine. 2004;351:1690-1691
  129. 129. Brusselmans K, Bono F, Collen D, Herbert JM, Carmeliet P, Dewerchin M. A novel role for vascular endothelial growth factor as an autocrine survival factor for embryonic stem cells during hypoxia. The Journal of Biological Chemistry. 2005;280:3493-3499
  130. 130. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57-70
  131. 131. Masood R, Cai J, Zheng T, Smith DL, Hinton DR, Gill PS. Vascular endothelial growth factor (VEGF) is an autocrine growth factor for VEGF receptor-positive human tumors. Blood. 2001;98:1904-1913
  132. 132. Dias S, Hattori K, Heissig B, Zhu Z, Wu Y, Witte L, et al. Inhibition of both paracrine and autocrine VEGF/ VEGFR-2 signaling pathways is essential to induce long-term remission of xenotransplanted human leukemias. Proceedings of the National Academy of Science USA. 2001;98:10857-10862
  133. 133. Bachelder RE, Wendt MA, Mercurio AM. Vascular endothelial growth factor promotes breast carcinoma invasion in an autocrine manner by regulating the chemokine receptor CXCR4. Cancer Research. 2002;62:7203-7206
  134. 134. Shvartsman SY, Hagan MP, Yacoub A, Dent P, Wiley HS, Lauffenburger DA. Autocrine loops with positive feedback enable context-dependent cell signaling. American Journal of Physiology: Cell Physiology. 2002;282:C545-C559
  135. 135. Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, et al. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001;410:50-56
  136. 136. Luo Y, Raible D, Raper JA. Collapsin: A protein in brain that induces the collapse and paralysis of neuronal growth cones. Cell. 1993;75:217-227
  137. 137. Bachelder RE, Lipscomb EA, Lin X, Wendt MA, Chadborn NH, Eickholt BJ, et al. Competing autocrine pathways involving alternative neuropilin-1 ligands regulate chemotaxis of carcinoma cells. Cancer Research. 2003;63:5230-5233

Written By

Pooja G. Singh, Kanthesh M. Basalingappa, T.S. Gopenath and B.V. Sushma

Submitted: 28 January 2022 Reviewed: 31 January 2022 Published: 14 May 2022