Abstract
Head and neck squamous cell carcinoma (HNSCC) is a common cancer characterized by increased angiogenesis. Vascular endothelial growth factor (VEGF) is a key regulator of angiogenesis and has not been extensively studied in HNSCC. This review aimed to provide a comprehensive overview of the VEGF family and its involvement in HNSCC. It discusses the significance of angiogenesis in HNSCC and the potential implications of VEGF family members, including VEGF-A, VEGF-B, VEGF-C, and VEGF-D, in tumor progression and angiogenic processes. The review highlights the need for further investigation to elucidate the specific functions and therapeutic implications of the VEGF family in HNSCC, which can ultimately contribute to development of novel therapeutic strategies for this type of cancer.
Head and neck squamous cell carcinoma (HNSCC) is a significant global health concern, accounting for a substantial proportion of cancer cases worldwide. It ranks as the sixth most common cancer globally, contributing to a considerable number of cancer cases. HNSCC has a significant impact on patient morbidity, mortality, and quality of life (1). The prevalence of HNSCC varies across different regions and populations. Certain geographical areas, such as Southeast Asia, Eastern Europe, and parts of South America, have higher incidence rates of HNSCC compared to other regions. However, it is important to note that HNSCC affects populations globally and is not limited to specific areas (2). Various risk factors contribute to the development of HNSCC, including tobacco and alcohol use, betel nut chewing, human papillomavirus (HPV) infection, poor oral hygiene, and exposure to certain occupational hazards such as asbestos and certain chemicals. Despite advances in early detection and treatment modalities, the overall prognosis for HNSCC remains relatively poor, with high rates of local recurrence and distant metastasis (3).
Tobacco use, particularly smoking cigarettes, is a prominent risk factor for HNSCC. It is associated with a dose-dependent relationship, and the risk diminishes upon smoking cessation. Similarly, alcohol consumption, especially heavy and prolonged use, significantly increases the risk of HNSCC. The synergistic effect of tobacco and alcohol further amplifies the risk (4). Betel quid chewing, prevalent in certain regions, is another well-established risk factor for HNSCC. The various components of betel quid, including areca nut and tobacco, contribute to the carcinogenicity (5). Human papillomavirus (HPV) infection, primarily with high-risk HPV types such as HPV-16 and HPV-18, has emerged as a significant risk factor for oropharyngeal cancers within the HNSCC spectrum. HPV-related HNSCC tends to affect younger individuals and is associated with better treatment outcomes (6). Genetic susceptibility plays a role in modifying an individual’s susceptibility to HNSCC. Certain genetic variations and polymorphisms have been associated with an increased risk of developing HNSCC. Family history of HNSCC also indicates a potential genetic predisposition (7). Additional risk factors include poor oral hygiene, chronic inflammation, and occupational and environmental exposures. Poor oral hygiene practices, such as inadequate dental care and persistent oral infections, contribute to HNSCC development (8). Chronic inflammation, often associated with conditions like oral lichen planus and gastroesophageal reflux disease (GERD), increases the risk of HNSCC. Occupational exposures to asbestos, certain chemicals, and dust have been implicated as potential risk factors, highlighting the importance of occupational safety measures (9).
The need for improved therapeutic approaches in HNSCC arises from several factors. Firstly, the disease often presents at advanced stages, resulting in limited treatment options and poorer outcomes. Secondly, the complex anatomical and functional structures of the head and neck region pose challenges in achieving effective local control while preserving critical functions such as speech and swallowing. Additionally, the heterogeneity of HNSCC at the molecular level contributes to treatment resistance and disease progression. Given these challenges, there is a critical need for the development of novel therapeutic approaches in HNSCC. Understanding the underlying molecular mechanisms driving tumor growth, invasion, and metastasis is crucial for identifying potential targets for intervention. The vascular endothelial growth factor (VEGF) family has emerged as one such target, playing a pivotal role in tumor angiogenesis and offering opportunities for therapeutic intervention in HNSCC (10).
Angiogenesis, the formation of new blood vessels, is essential for tumor growth and metastasis, providing oxygen and nutrients to support the expanding tumor mass. The VEGF family members are key regulators of this process, orchestrating the formation of new blood vessels from pre-existing ones. The newly formed blood vessels in the tumor microenvironment are often abnormal, characterized by increased permeability and irregular structure. This abnormal vasculature contributes to tumor progression by facilitating the entry of cancer cells into the circulation, promoting metastasis to distant sites. Additionally, the leaky vasculature provides a route for the delivery of immune cells, growth factors, and nutrients, further supporting tumor growth and survival (11).
Given the crucial role of the VEGF family in tumor angiogenesis and progression, targeting this pathway has emerged as a promising strategy for cancer therapy. Anti-VEGF therapies, such as monoclonal antibodies and tyrosine kinase inhibitors, have been developed to block VEGF signaling and inhibit angiogenesis. These therapies have shown efficacy in various cancers, including colorectal cancer, renal cell carcinoma, and non-small cell lung cancer. By disrupting tumor blood supply, anti-VEGF therapies can reduce tumor growth, inhibit metastasis, and enhance the effectiveness of other treatment modalities, such as chemotherapy and radiation therapy (12).
Molecular Biology of the VEGF Family
The vascular endothelial growth factor family is a group of proteins that play a crucial role in the regulation of blood vessel formation and maintenance (vascular homeostasis). These proteins are involved in various physiological and pathological processes, including embryonic development, wound healing, and tumor growth. The VEGF family consists of several members, including VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, and placental growth factor (PlGF) (13).
VEGF-A is the one most extensively studied and well-characterized member of the VEGF family. It exists in multiple isoforms generated through alternative splicing, such as VEGF-A121, VEGF-A165, and VEGF-A189. VEGF-A is a potent inducer of angiogenesis and is involved in various physiological and pathological processes, including embryonic development, wound healing, and tumor growth. VEGF-A is a potent inducer of angiogenesis and essential for the development of blood vessels in embryonic development and tissue repair processes (14). It promotes endothelial cell proliferation, migration, and tube formation to form new blood vessels. Additionally, VEGF-A increases vascular permeability, allowing plasma proteins and immune cells to extravasate into tissues. The expression and activity of VEGF-A are tightly regulated under normal physiological conditions. Various factors can influence VEGF-A expression, including hypoxia, growth factors, cytokines, and transcription factors. Hypoxia-inducible factor 1 (HIF-1) is a key transcription factor that upregulates VEGF-A expression in response to low oxygen levels. VEGF-A has important clinical implications. Its dysregulation is associated with various pathological conditions, including cancer, diabetic retinopathy, age-related macular degeneration, and inflammatory diseases. In cancer, increased VEGF-A expression promotes tumor angiogenesis, enabling tumor growth and metastasis (15).
VEGF-B is closely related to VEGF-A but has distinct functions. It is involved in the regulation of vascularization during embryonic development and plays a role in cardiac and skeletal muscle function. VEGF-B has also been implicated in ischemic diseases and acts as a survival factor for endothelial cells. VEGF-B has been investigated for its therapeutic potential in ischemic diseases, particularly those affecting the heart and skeletal muscles (16). Studies have shown that VEGF-B can enhance blood vessel growth, improve tissue perfusion, and protect against tissue damage in ischemic conditions. VEGF-B can also interact with neuropilin co-receptors, specifically neuropilin-1 (NRP1) and neuropilin-2 (NRP2). These interactions modulate VEGF-B signaling can influence angiogenesis and other cellular processes (17, 18).
VEGF-C and VEGF-D are primarily associated with lymphangiogenesis, the formation of lymphatic vessels. They play crucial roles in the development and maintenance of lymphatic vasculature. Additionally, VEGF-C and VEGF-D have been implicated in metastasis, as they can promote lymphatic vessel growth and facilitate the spread of cancer cells to lymph nodes. They are synthesized as precursor forms that undergo proteolytic cleavage to generate bioactive forms. The proteolytic processing is mediated by enzymes such as furin and plasmin. The processed forms contain regions called VEGF homology domains (VHDs) that are responsible for receptor binding and activation (19). VEGF-C and VEGF-D can also interact with co-receptors known as neuropilins, specifically neuropilin-2 (NRP2). These interactions enhance the binding affinity of VEGF-C and VEGF-D for VEGFR-3 and modulate their signaling and biological activities (20).
VEGF-E is a viral protein encoded by certain strains of the Orf virus and other parapox viruses. It stimulates angiogenesis and vascular permeability. VEGF-E has been used as a research tool to study angiogenesis and to develop models for vascular diseases. VEGF-E shows a strong angiogenic activity, promoting the formation of new blood vessels, and is also associated with increased vascular permeability. It stimulates proliferation, migration and tube formation of endothelial cells, similar to other members of the VEGF family (21).
Placental growth factor (PlGF) is structurally related to VEGF-A but interacts with VEGFR-1. It is involved in angiogenesis and vascular remodeling, particularly during pregnancy and placental development. PlGF can also contribute to pathological angiogenesis in diseases such as cancer. PlGF has angiogenic properties, but its effects on blood vessel formation are generally milder compared to VEGF-A. PlGF can induce endothelial cell proliferation, migration, and the formation of new blood vessels. It also plays a role in vascular remodeling and arterialization (22).
VEGF family members are secreted glycoproteins characterized by a conserved VEGF homology domain (VHD) that contains eight cysteine residues involved in disulfide bond formation. This domain is essential for receptor binding and activation. The VEGF proteins are typically dimeric or homodimeric, and they interact with specific cell surface receptors to initiate signaling cascades (23).
The primary receptors for VEGF family members are receptor tyrosine kinases known as VEGF receptors (VEGFRs). The VEGFR family consists of three members: VEGFR-1 (Flt-1), VEGFR-2 (KDR/Flk-1), and VEGFR-3 (Flt-4). VEGFR-1 and VEGFR-2 are mainly involved in angiogenesis, while VEGFR-3 plays a role in lymphangiogenesis. Upon binding of VEGF ligands, the receptors undergo dimerization, leading to activation of their intracellular kinase domains and subsequent initiation of downstream signaling pathways. The activation of VEGF receptors triggers a variety of intracellular signaling pathways. The major pathways include the phosphoinositide 3-kinase (PI3K)/Akt pathway, the mitogen-activated protein kinase (MAPK) pathway, and the phospholipase C (PLC)-γ pathway. These signaling cascades regulate endothelial cell proliferation, survival, migration, and vascular permeability (24).
VEGF family members exert their effects primarily on endothelial cells, which line the inner surface of blood vessels. They promote angiogenesis by stimulating endothelial cell proliferation and migration. Additionally, VEGF proteins enhance vascular permeability, allowing the extravasation of plasma proteins and immune cells into tissues. The expression and activity of VEGF family members are tightly regulated to maintain proper vascular function (25). Multiple factors, such as hypoxia, growth factors, cytokines, and transcription factors, can modulate VEGF expression. For example, hypoxia-inducible factor 1 (HIF-1) plays a crucial role in upregulating VEGF expression under low oxygen conditions. Additionally, negative regulators, including soluble VEGF receptors and specific inhibitors, help control the activity of VEGF proteins (26).
Not all VEGF proteins show a mandatory angiogenic activity. Angiogenic inhibitory isoforms of VEGF are specific variants or splice forms of VEGF that possess reduced or even inhibitory effects on angiogenesis. These isoforms differ from the pro-angiogenic isoforms of VEGF and can act as natural inhibitors of angiogenesis. One well-known example is VEGF-A165b, which is generated by alternative splicing of the VEGF-A gene. This alternative splicing event results in a shorter isoform that lacks the heparin-binding domain found in the pro-angiogenic isoforms. VEGF-A165b has been shown to have anti-angiogenic properties and can counteract the effects of pro-angiogenic VEGF isoforms. It acts by competing for binding to VEGF receptors (VEGFRs), thereby interfering with the signaling pathways involved in promoting angiogenesis. VEGF-A165b has been reported to inhibit endothelial cell proliferation, migration, and tube formation, which are essential steps in the process of angiogenesis. The balance between pro-angiogenic and anti-angiogenic isoforms of VEGF, including VEGF-A165b, is believed to play a critical role in regulating angiogenesis in various physiological and pathological conditions. Dysregulation of this balance, with an imbalance favoring pro-angiogenic isoforms, has been implicated in diseases characterized by excessive angiogenesis, such as cancer (27, 28). The discovery and characterization of angiogenic inhibitory isoforms of VEGF have sparked interest in their potential as therapeutic targets for diseases associated with excessive angiogenesis, including cancer. Further research is needed to fully understand the mechanisms and therapeutic implications of these isoforms in different disease contexts.
In the case of HNSCC, there have not been many studies on VEGF; however, altered expression patterns of VEGF-A isoforms, including VEGF-A165b, have been reported. The findings suggest that HNSCC tumors may have higher expression of VEGF-A165b compared to normal tissues or benign lesions. Increased levels of VEGF-A165b in HNSCC could act as a negative feedback mechanism to counterbalance the pro-angiogenic effects of other VEGF-A isoforms (29). The role of VEGF-A165b in HNSCC remains an active area of research, and its precise functions and implications are not yet fully elucidated.
The molecular biology of the VEGF family has significant implications for therapeutic interventions targeting angiogenesis-related diseases, such as cancer, ischemic diseases, and ocular disorders. Manipulating the VEGF signaling pathway has been an important strategy for developing anti-angiogenic therapies.
VEGF and Angiogenesis in HNSCC
Angiogenesis plays a crucial role in the development, progression, and metastasis of head and neck squamous cell carcinoma (HNSCC). Angiogenesis is a hallmark of solid tumors, including HNSCC. As the tumor grows, it demands an increased blood supply to sustain its metabolic needs and facilitate its expansion. Tumor angiogenesis involves the emergence of new blood vessels from nearby existing vessels into the tumor microenvironment (11).
Immunohistochemical methods provided valuable information about VEGF expression in HNSCC tumor cells. Despite the limited number of studies focusing specifically on VEGF expression in HNSCC tumor cells using immunohistochemical methods in the past 10 years, the available data suggest that VEGF expression is detectable in HNSCC tumor tissue. The exact pattern and intensity of VEGF expression may vary among different HNSCC subtypes and tumor stages (30).
Vascular endothelial growth factor family members, particularly VEGF-A, VEGF-C, and VEGF-D, are potent angiogenic factors involved in HNSCC angiogenesis. Overexpression of VEGF family members in HNSCC cells promotes endothelial cell proliferation, migration, and vessel formation. Other angiogenic factors, such as basic fibroblast growth factor (bFGF) and platelet-derived growth factor (PDGF), also contribute to angiogenesis in HNSCC. Hypoxia, a low oxygen environment, is a common feature of solid tumors, including HNSCC. Hypoxia-inducible factor (HIF) proteins are key regulators of the cellular response to hypoxia. In HNSCC, hypoxia induces the stabilization and activation of HIF, which in turn upregulates the expression of angiogenic factors, including VEGF-A. HIF activation promotes angiogenesis by enhancing the expression of pro-angiogenic genes (31).
The tumor microenvironment plays a critical role in promoting angiogenesis in HNSCC. Cancer cells and stromal cells, such as cancer-associated fibroblasts and immune cells, secrete various angiogenic factors and cytokines, creating a pro-angiogenic environment. This switch towards an angiogenic phenotype involves a delicate balance between pro-angiogenic factors and anti-angiogenic factors (32).
The extent of tumor angiogenesis in HNSCC has been associated with a poor prognosis and clinical outcomes. Increased microvessel density (MVD) or higher levels of angiogenesis-related markers, such as VEGF-A expression, have been correlated with tumor aggressiveness, metastasis, and decreased survival rates (33). Consequently, targeting angiogenesis has emerged as a therapeutic strategy in HNSCC. Anti-angiogenic agents, such as monoclonal antibodies against VEGF-A or small molecule inhibitors of VEGF receptors, have been investigated in clinical trials either as standalone therapies or in combination with conventional treatments (27).
HNSCC tumors often exhibit increased expression and secretion of VEGF-A. This can occur due to various factors, including genetic alterations, HIF activation in response to tumor hypoxia, and inflammatory signals within the tumor microenvironment. VEGF-A is a potent inducer of angiogenesis and acts as a major pro-angiogenic factor in HNSCC. Increased VEGF-A levels contribute to the formation of a dense and abnormal tumor vasculature network (34). VEGF-A interacts with its receptor [vascular endothelial growth factor receptor-2 (VEGFR-2)] on endothelial cells, initiating signaling cascades that promote endothelial cell proliferation, migration, and survival. In addition to promoting angiogenesis, VEGF-A is involved in lymphangiogenesis, can induce lymphatic endothelial cell proliferation and migration, contributing to lymphatic vessel expansion and lymph node metastasis in HNSCC. The overexpression of VEGF-A in HNSCC is associated with aggressive tumor behavior, increased invasiveness, and a higher likelihood of lymph node metastasis. It promotes tumor growth by providing the necessary blood supply and facilitating the spread of cancer cells through the lymphatic system (35).
Both VEGF-C and VEGF-D are involved in the induction of lymphangiogenesis, they bind to their receptor VEGFR-3 (vascular endothelial growth factor receptor 3) expressed on lymphatic endothelial cells. Elevated levels of VEGF-C and VEGF-D have been associated with lymph node metastasis in HNSCC. Increased expression of these factors facilitates the growth and expansion of lymphatic vessels, providing a pathway for tumor cells to spread to regional lymph nodes. Lymph node metastasis is an important prognostic factor in HNSCC and is associated with poor prognosis (36, 37).
Understanding the role of VEGF and angiogenesis in HNSCC is essential for developing targeted therapies and improving patient outcomes. Targeting the VEGF pathway and inhibiting angiogenesis have the potential to limit tumor growth, reduce metastasis, and enhance the efficacy of existing treatment modalities in HNSCC.
Therapeutic Targeting of VEGF in HNSCC
HNSCC is a highly prevalent and aggressive malignancy with limited treatment options. The dysregulated angiogenesis observed in HNSCC provides a rationale for targeting VEGF, a key driver of tumor vascularization. VEGF-targeted therapies have shown promise in preclinical models and clinical trials, but their overall efficacy, safety profile, and limitations need to be thoroughly evaluated (38).
Numerous preclinical studies have demonstrated the role of VEGF in promoting angiogenesis and tumor progression in HNSCC. These studies have shown that blocking VEGF signaling through monoclonal antibodies or small molecule inhibitors leads to reduced tumor angiogenesis, inhibition of tumor growth, and decreased metastatic potential in animal models of HNSCC (39). Studies have shown that VEGF inhibition can reduce the invasive potential of HNSCC cells, inhibit tumor cell migration, and prevent the formation of distant metastases (40). By blocking VEGF-induced angiogenesis, therapies targeting VEGF can potentially limit the dissemination of HNSCC tumor cells to distant sites. Preclinical models have also been utilized to investigate mechanisms of resistance to VEGF-targeted therapies in HNSCC. These studies have identified various resistance mechanisms, including alternative angiogenic pathways, compensatory signaling pathways, and genetic alterations in the VEGF pathway (41, 42). Understanding these mechanisms can help inform the development of strategies to overcome or circumvent resistance and improve the efficacy of VEGF-targeted therapies.
Numerous clinical trials have investigated the efficacy of VEGF-targeted therapies, including monoclonal antibodies and tyrosine kinase inhibitors, in HNSCC. These studies have demonstrated improvements in progression-free survival, overall survival, and objective response rates in patients receiving VEGF-targeted therapy, either as monotherapy or in combination with conventional treatments. However, the magnitude of the clinical benefit varies among different studies and patient populations (43, 44).
Monoclonal antibodies specifically bind to VEGF-A, preventing its interaction with VEGF receptors and inhibiting downstream angiogenic signaling pathways. This blockade of VEGF-mediated signaling impedes tumor angiogenesis, vascular permeability, and tumor growth in HNSCC. Clinical trials evaluating VEGF-targeted monoclonal antibodies, such as bevacizumab, have demonstrated promising results (Table I). These studies have shown improved progression-free survival, overall survival, and objective response rates when bevacizumab is combined with standard chemotherapy or radiation therapy. Notably, the benefits may vary depending on patient selection, treatment regimen, and tumor characteristics (45).
Anti-VEGF and VEGFR inhibitor therapy in HNSCC.
VEGF-targeted monoclonal antibodies are generally well-tolerated; however, they can be associated with specific adverse events. The most common side effects include hypertension, proteinuria, bleeding complications, and wound healing complications. Close monitoring and appropriate management strategies are necessary to ensure patient safety. Despite the promising efficacy, several challenges and limitations exist with VEGF-targeted monoclonal antibodies in HNSCC. Primary and acquired resistance mechanisms may limit treatment efficacy (46). Additionally, patient selection criteria, optimal treatment duration, and identification of predictive biomarkers for response to therapy need further investigation.
Combining VEGF-targeted monoclonal antibodies with other treatment modalities, such as immune checkpoint inhibitors, chemotherapy, or targeted agents, is a promising strategy to enhance therapeutic outcomes in HNSCC. Synergistic effects and improved response rates have been observed with these combination approaches, warranting further investigation (47).
Tyrosine kinase inhibitors (TKIs) have emerged as a promising class of drugs for inhibiting VEGF signaling in HNSCC. TKIs block the intracellular tyrosine kinase domain of VEGF receptors, preventing downstream signaling and inhibiting angiogenesis. By targeting VEGF receptors, TKIs disrupt tumor blood supply and inhibit tumor growth and metastasis in HNSCC. Clinical trials evaluating TKIs as VEGF-targeted therapies in HNSCC have shown mixed results (Table I). Some TKIs, such as sorafenib and sunitinib, have demonstrated modest clinical benefits, including improved progression-free survival and tumor response rates. However, the overall efficacy of TKIs in HNSCC varies, and patient selection, treatment regimens, and tumor characteristics may influence treatment outcomes (48).
TKIs are associated with specific adverse events, including skin rash, hypertension, diarrhea, and fatigue. Careful monitoring and appropriate management strategies are necessary to minimize treatment-related toxicity and optimize patient safety. The development of resistance to TKIs represents a significant challenge in HNSCC treatment. Mechanisms such as acquired mutations in the tyrosine kinase domain, activation of alternative signaling pathways, and tumor heterogeneity contribute to treatment failure. Combination approaches and the identification of predictive biomarkers may help overcome resistance and improve treatment efficacy (49).
VEGF-targeted therapies are associated with specific safety concerns, including hypertension, proteinuria, bleeding events, gastrointestinal disturbances, and wound healing complications. Adverse events are typically manageable but require careful monitoring and intervention. Additionally, the development of resistance to VEGF-targeted therapies remains a significant challenge, necessitating the exploration of combination approaches and predictive biomarkers to enhance treatment outcomes (56). The criteria for inclusion in antiangiogenic therapy in HNSCC are still being studied and refined. The decision to include patients in antiangiogenic therapy is typically based on a combination of clinical and molecular factors: tumor stage, biomarkers of angiogenesis, performance status of the patient, previous treatment history, comorbidities and patient factors (1, 57). It is important to note that the specific criteria for inclusion in antiangiogenic therapy may vary depending on the clinical trial protocol or treatment guidelines being followed. The decision to include patients in antiangiogenic therapy should be made on an individual basis, taking into account the available evidence, patient characteristics, and treatment goals, in consultation with a multidisciplinary team of oncologists.
VEGF Biomarkers as Predictive Indicators in HNSCC Treatment
Predictive biomarkers are valuable tools for guiding treatment decisions in VEGF-targeted therapy for HNSCC (Table II), unfortunately there are not many studies on VEGF biomarkers performed on squamous cell cancer of the head and neck area. Biomarkers that can accurately predict treatment response allow for personalized medicine approaches and improved clinical outcomes. Several potential biomarkers, including VEGF expression levels, genetic alterations, and circulating angiogenic factors, have been investigated (63). However, further validation and standardization are needed to implement these biomarkers in routine clinical practice.
Studies on VEGF biomarkers in HNSCC.
Molecular biomarkers of VEGF refer to specific genetic alterations or expression patterns within the VEGF pathway or related genes that have the potential to predict treatment response to VEGF-targeted therapies in head and neck squamous cell carcinoma. These biomarkers can provide valuable information about the activity and regulation of the VEGF signaling pathway (64).
It is important to note that although these molecular criteria are promising, their clinical utility in guiding decisions on anti-VEGF therapy in HNSCC is still under investigation. Further research and clinical trials are needed to establish robust and validated molecular biomarkers that can reliably predict treatment response and guide personalized therapeutic approaches in HNSCC. VEGF expression levels in tumor tissues or serum have been investigated as potential predictive biomarkers. High VEGF expression has been associated with increased response to VEGF-targeted therapy, suggesting its potential as a predictive marker (65). Overexpression of VEGF is associated with aggressive tumor behavior and poor prognosis. High VEGF expression levels in tumor tissues or serum have been linked to increased tumor growth, invasiveness, and a higher risk of metastasis. Consequently, targeting VEGF and its angiogenic effects has been explored as a therapeutic approach to inhibit tumor growth and progression in HNSCC (66). Polymorphisms in VEGF receptors, such as VEGFR-2, have been explored as potential biomarkers for treatment response prediction. VEGFR-2 is a key receptor for VEGF signaling, and certain genetic variants (polymorphisms) within the VEGFR-2 gene may influence the efficacy of VEGF-targeted therapy. Specific variations in the VEGFR-2 gene could affect the receptor’s affinity for VEGF, thus potentially impacting treatment response (67). Specific genetic variants may influence the efficacy of VEGF-targeted therapy and serve as predictive markers. Various angiogenesis-related biomarkers, including angiopoietins, microvessel density, and circulating endothelial cells, have been investigated for their predictive value in VEGF-targeted therapy. These biomarkers reflect the tumor vasculature and may help identify patients who are more likely to respond to treatment (65).
Molecular alterations within the VEGF pathway, such as VEGF gene amplification or mutations, may influence treatment response to VEGF-targeted therapy. VEGF gene amplification refers to the process by which there is an increased copy number of the VEGF gene in a cell or tumor. Amplification of the VEGF gene leads to an excessive production of the VEGF protein. In the case of VEGF gene amplification, cancer cells produce higher amounts of VEGF compared to normal cells, leading to an increase in the levels of VEGF protein in the tumor microenvironment. Elevated VEGF levels promote the growth and migration of endothelial cells – the building blocks of blood vessels – resulting in the formation of an extensive network of blood vessels around the tumor. This network of blood vessels supplies the tumor with nutrients and oxygen, enabling the cancer cells to thrive and proliferate (68). VEGF gene amplification is commonly observed in various types of cancers, including solid tumors like breast cancer, lung cancer, glioblastoma, and colorectal cancer, among others. In cancers where VEGF gene amplification is present, it is often associated with more aggressive tumor behavior, increased tumor growth, and a higher likelihood of metastasis (69, 70). Detection of these alterations may help identify patients who are more likely to benefit from VEGF inhibition.
Hypoxia, a common feature in solid tumors, plays a role in VEGF expression and angiogenesis. Hypoxia-related biomarkers, such as hypoxia-inducible factor 1-alpha (HIF-1α), have been studied as potential predictive markers for VEGF-targeted therapy (71).
HIF-1α is a critical transcription factor that plays a central role in the cellular response to low oxygen levels. Hypoxia is a common characteristic of the tumor microenvironment in solid tumors, including various types of cancer, where rapid cell proliferation outpaces the supply of oxygen through the tumor’s blood vessels (72).
Under hypoxic conditions, HIF-1α is stabilized and accumulates within the cell, leading to the formation of a complex with another protein, HIF-1β. This HIF-1α/β complex, known as HIF-1, acts as a transcription factor that binds to specific DNA sequences, called hypoxia-responsive elements (HREs), within the regulatory regions of target genes. By doing so, HIF-1 can activate or repress the transcription of numerous genes involved in various cellular processes, including angiogenesis. In the context of cancer, hypoxia can induce the upregulation of VEGF expression through HIF-1α. VEGF is one of the target genes of HIF-1, and its transcription is increased under hypoxic conditions (73).
The interplay between HIF-1α, VEGF, and angiogenesis is a critical component of the tumor microenvironment, and it has significant implications for cancer progression and response to therapy. In the context of VEGF-targeted therapy, tumors with high levels of HIF-1α and consequently elevated VEGF expression may be more dependent on VEGF-driven angiogenesis. Thus, such tumors could potentially be more responsive to VEGF-targeted therapies, as inhibiting VEGF signaling could disrupt the tumor’s blood supply and hinder its growth (73).
Understanding the role of HIF-1α in regulating VEGF expression and angiogenesis is essential for the development of targeted therapies aimed at disrupting angiogenesis and inhibiting tumor progression. Additionally, the assessment of HIF-1α levels in tumors may serve as a potential biomarker to identify patients who may benefit from VEGF-targeted therapy or other anti-angiogenic treatments.
The molecular biomarkers of VEGF hold great promise for guiding treatment decisions and optimizing therapeutic outcomes in head and neck squamous cell carcinoma. Combining multiple biomarkers, such as VEGF expression with genetic alterations or angiogenesis-related markers, may improve the predictive accuracy for VEGF-targeted therapy in HNSCC (63). The integration of different biomarkers can provide a comprehensive assessment of treatment response. Predictive biomarkers hold promise for optimizing patient selection and treatment response prediction in VEGF-targeted therapy for HNSCC.
A comprehensive approach that combines multiple biomarkers and integrates them with clinical characteristics may offer a more accurate prediction of treatment response and patient outcomes in VEGF-targeted therapy for HNSCC. By identifying patients who are more likely to benefit from VEGF inhibition, this personalized approach can enhance the effectiveness of VEGF-targeted therapies and potentially lead to better treatment outcomes.
However, it is crucial to exercise caution and continue thorough investigation in the selection and use of these molecular biomarkers to ensure their robustness and clinical significance. Collaborative efforts among researchers and clinicians are needed to advance our understanding of VEGF biology in HNSCC and identify reliable biomarkers that can guide treatment decisions and improve patient management in this aggressive malignancy. With continued research and ongoing clinical trials, we can maximize the potential of VEGF-targeted therapies and move closer to the goal of precision medicine in the treatment of HNSCC.
Future Directions and Challenges
Despite the advancements in VEGF-targeted therapies, several challenges remain. The heterogeneity of HNSCC, the complexity of tumor angiogenesis, and the development of resistance necessitate a deeper understanding of the underlying mechanisms and the exploration of combination strategies with immune checkpoint inhibitors, chemotherapy, and other targeted agents. Additionally, addressing the financial burden and access to these therapies is essential to ensure equitable treatment for all patients (74). Continued research is focused on the development of novel VEGF inhibitors with improved efficacy and safety profiles. This includes the investigation of small molecule inhibitors, antibody-drug conjugates, and bispecific antibodies targeting VEGF and other relevant pathways. The goal is to enhance the anti-angiogenic effects and overcome resistance mechanisms associated with current VEGF-targeted therapies (75).
Immunotherapeutic approaches, such as immune checkpoint inhibitors and adoptive cell therapies, have shown promising results in various cancers. Ongoing research is exploring the potential of immunotherapies in HNSCC, either as monotherapy or in combination with other treatment modalities. This includes targeting immune checkpoints such as PD-1/PD-L1, exploring tumor-specific antigens, and enhancing the activation and persistence of tumor-infiltrating lymphocytes (76).
Advancements in molecular profiling technologies have allowed for better characterization of HNSCC tumors. Future directions involve identifying predictive biomarkers to guide treatment decisions and improve patient outcomes. Personalized treatment approaches may involve selecting therapies based on specific genetic alterations, molecular subtypes, or immune profiles. This includes exploring targeted therapies against specific mutations, optimizing combination regimens, and utilizing biomarkers to guide treatment response assessment (77).
In head and neck squamous cell carcinoma, the molecular criteria for recommending anti-VEGF therapy are still being investigated, and the field is rapidly evolving. While there is no standardized molecular biomarker that exclusively determines the recommendation for anti-VEGF therapy in HNSCC, several molecular factors are being studied as potential indicators (78).
The identification and validation of robust biomarkers for HNSCC are crucial for the successful implementation of targeted therapies and personalized treatment approaches. Ongoing research efforts aim to discover biomarkers that can predict treatment response, identify patients at high risk of recurrence, and guide therapeutic decisions. This includes investigating genetic alterations, protein expression patterns, immune-related markers, and circulating tumor DNA as potential biomarkers. The tumor microenvironment plays a critical role in HNSCC progression and therapy resistance. Future research is focused on understanding the interactions between tumor cells, immune cells, stromal cells, and the extracellular matrix. Targeting components of the tumor microenvironment, such as fibroblasts, immune suppressive cells, and angiogenic factors, may offer new therapeutic avenues (79).
Several challenges need to be addressed to advance the field of HNSCC treatment. These include tumor heterogeneity, resistance mechanisms, appropriate patient selection, and optimizing combination strategies. Additionally, the development of effective drug delivery systems and the management of treatment-related toxicities remain significant challenges. Translational research efforts and well-designed clinical trials are vital for evaluating the efficacy and safety of emerging therapies in HNSCC. Collaborative efforts between basic scientists, translational researchers, and clinical investigators are essential to bridge the gap between laboratory discoveries and clinical practice.
Conclusion
The VEGF family plays a crucial role in the pathogenesis of HNSCC, and therapeutic targeting of VEGF has shown promise in improving treatment outcomes. Monoclonal antibodies and tyrosine kinase inhibitors have demonstrated efficacy in combination with standard treatment approaches. However, challenges such as tumor heterogeneity, resistance mechanisms, and the need for predictive biomarkers remain. Future research should focus on identifying robust biomarkers, developing novel VEGF inhibitors, exploring combination therapies, and implementing personalized treatment approaches based on specific molecular alterations and biomarkers. Overall, targeting the VEGF family holds potential for improving outcomes in HNSCC; further research and clinical trials are needed to optimize its efficacy and safety.
Acknowledgements
The Authors are grateful to the lecturers Pusa Nela Gaje, Amalia Raluca Ceausu and Cristian Silviu Suciu from the Department of Histology of the University of Medicine and Pharmacy Victor Babes Timisoara, Romania. For their critical evaluation of the manuscript.
Footnotes
Authors’ Contributions
CSD, designed and wrote of the manuscript; MR, revised the text and supervised the study.
Conflicts of Interest
The Authors declare no conflicts of interest.
- Received July 3, 2023.
- Revision received August 4, 2023.
- Accepted August 28, 2023.
- Copyright © 2023 The Author(s). Published by the International Institute of Anticancer Research.
This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).