Fact-checked by Grok 2 weeks ago

Neovascularization

Neovascularization refers to the formation of new vessels, regardless of their or type, often driven by physiological needs or pathological conditions. This process primarily involves , the sprouting or splitting of new vessels from pre-existing vasculature, and , the assembly of endothelial cells into primitive vascular networks—primarily during embryonic development but also possible in adults. In physiological contexts, neovascularization supports tissue growth and repair, such as during embryonic development, , exercise-induced muscle adaptation, or expansion in response to weight gain. For instance, it ensures that no tissue is more than a few hundred micrometers from a , maintaining oxygen and nutrient delivery under metabolic demands. Key regulators include (VEGF), which promotes endothelial cell proliferation and , alongside hemodynamic and hypoxic signals that balance angiogenic and anti-angiogenic factors. Pathologically, neovascularization contributes to numerous diseases by enabling abnormal vessel growth that exacerbates tissue damage. In cancer, it sustains tumor progression by providing essential nutrients and oxygen, as hypothesized by in 1971, leading to therapeutic strategies like inhibitors. Ocular disorders, including , age-related macular degeneration, and , feature aberrant vessel invasion that causes vision loss through hemorrhage or . Similarly, in , plaque neovascularization heightens vulnerability to rupture and , while in , it fuels chronic joint destruction. These roles underscore neovascularization's dual nature, where it is harnessed for treatments in ischemic conditions but targeted for inhibition in proliferative diseases.

Definition and Overview

Definition

Neovascularization refers to the formation of new blood vessels, a term derived from the "neo-" (meaning new) combined with "vascularization" (the process of developing blood vessels), with its earliest documented use appearing in in 1952. The underlying biological process of generating functional vascular networks has been studied extensively since the early , particularly in the context of embryonic development, where systematic descriptions of vessel were pioneered by researchers like Florence Sabin through histological analyses of avian and mammalian embryos. At its core, neovascularization involves the creation or adaptive remodeling of vessels to meet demands for oxygen and nutrients in organisms. This occurs through the , migration, proliferation, and assembly of endothelial cells or their precursors into capillary-like tubes, often initiated by environmental cues such as , which stabilizes hypoxia-inducible factors to upregulate angiogenic growth factors, alongside inflammatory mediators that recruit supporting cells. (VEGF) serves as a primary stimulator in this orchestration. Key cellular and structural elements include endothelial cells, which form the luminal lining and respond directly to signaling cues; pericytes, which envelop vessels to stabilize them, regulate blood flow, and prevent leakage; and the extracellular matrix, a dynamic network of proteins like and that provides mechanical support, guides via , and modulates bioavailability during vessel assembly and maturation. Although neovascularization is sometimes used broadly to describe any new vessel growth, it is conceptually distinct from related processes: entails the initial differentiation of angioblasts (endothelial precursors) into primitive vascular plexuses, primarily during embryogenesis; involves the sprouting, branching, or splitting of capillaries from established vessels in response to local gradients; and arteriogenesis focuses on the shear stress-induced enlargement and maturation of preexisting arterial collaterals into functional conduits.

Physiological vs. Pathological Neovascularization

Neovascularization serves essential functions in normal physiology, where it is tightly regulated to support tissue development and repair in adults, but becomes detrimental when dysregulated in disease states. In physiological contexts, triggers such as transient during activate hypoxia-inducible factor-1α (HIF-1α), which upregulates (VEGF) and other pro-angiogenic factors to promote vessel formation. This process is balanced by endogenous inhibitors like thrombospondin-1 (TSP-1), which suppresses endothelial cell proliferation, migration, and survival through interactions with and receptors, ensuring controlled vascular expansion. As a result, physiological neovascularization yields stable, organized vasculature with proper coverage and low permeability, facilitating efficient oxygen delivery and tissue homeostasis. In contrast, pathological neovascularization arises from chronic or severe triggers, including prolonged tumor , ischemia in ischemic diseases, and sustained , which lead to excessive HIF-1α stabilization and unchecked VEGF production. Chronic , for instance, recruits leukocytes that release pro-angiogenic , amplifying vessel sprouting in an uncontrolled manner, while tumor induces an "angiogenic switch" favoring rapid but aberrant growth. Ischemia in non-healing wounds or vascular occlusions similarly drives dysregulated responses, often without adequate inhibitory counterbalance from factors like TSP-1. Consequently, pathological vessels are fragile, tortuous, and highly permeable due to incomplete recruitment and disrupted endothelial junctions, promoting hemorrhage, , and tumor . In , particularly cancer, tumors hijack the angiogenic program—either by inducing new vessels or co-opting existing ones—to sustain uncontrolled . This transforms a beneficial into one that exacerbates progression, underscoring the fine line between regulated and pathological exploitation.

Mechanisms of Neovascularization

Vasculogenesis

Vasculogenesis refers to the de novo formation of the initial vascular network during embryonic , where mesodermal precursor cells differentiate into endothelial progenitor cells, known as angioblasts, which subsequently assemble into primitive vascular plexuses. This process begins with the commitment of to the endothelial lineage, followed by the migration, proliferation, and coalescence of angioblasts to form cord-like structures that eventually develop lumens and connect into a functional plexus. Unlike later vascular expansion, vasculogenesis establishes the foundational architecture of the without reliance on pre-existing vessels. The modern understanding of vasculogenesis as an in situ process emerged in the late through pioneering studies using quail-chick models, which demonstrated that endothelial cells arise locally from al precursors rather than solely migrating from extraembryonic sites. In these experiments, interspecific grafts revealed that somitic and splanchnopleural in the avian embryo generates endothelial cells on site, forming blood islands and vascular tubes independently in different territories. This work, building on earlier histological observations, solidified as a distinct of origination in the , distinguishing it from migratory contributions. Central to vasculogenesis is the hemangioblast, a bipotent derived from that gives rise to both endothelial and hematopoietic lineages, as evidenced by clonal analyses and marker expression in avian and murine models. Key molecular regulators include (VEGF), which promotes angioblast and via its receptors VEGFR-1 and VEGFR-2; fibroblast growth factor-2 (FGF-2), which induces mesodermal commitment to the endothelial fate and supports proliferation; and angiopoietins, particularly Ang-1, which stabilize nascent vessels through Tie-2 receptor signaling in coordination with VEGF. These factors act in a spatiotemporal , with VEGF and FGF-2 initiating hemangioblast specification and angiopoietins aiding maturation. In vitro models of vasculogenesis have been instrumental in dissecting these processes, particularly through the differentiation of embryonic stem (ES) cells into embryoid bodies, which recapitulate blood island formation and vascular channel development. When murine ES cells aggregate into cystic embryoid bodies, they spontaneously generate endothelial cells expressing markers like PECAM-1 and Flk-1, assembling into tube-like structures in a VEGF-dependent manner, mirroring embryonic vasculogenesis. These systems allow manipulation of signaling pathways, confirming the roles of FGF-2 and angiopoietins in enhancing network complexity without external scaffolds.

Angiogenesis

Angiogenesis is the process by which new blood vessels form from pre-existing vasculature through and branching, enabling tissue and growth in both physiological and pathological contexts. This highly regulated mechanism involves the coordinated activation, migration, and proliferation of endothelial cells, primarily driven by angiogenic signals in response to or injury. Unlike de novo vessel formation, remodels mature vessels to expand the microvascular network. The process begins with the degradation of the () surrounding existing vessels, facilitated by matrix metalloproteinases (MMPs) such as MMP-2 and MMP-9, which are secreted by endothelial cells and to create pathways for sprout invasion. Specialized endothelial tip cells then emerge at the sprout front, extending —actin-rich protrusions that sense gradients of angiogenic factors like ()—to guide directional migration. Trailing stalk cells proliferate to elongate the sprout, driven by VEGF signaling, while subsequent lumen formation occurs through endothelial cell rearrangement and vacuole fusion, establishing a patent vessel tube. and cells later associate with the nascent vessel to promote maturation and stability. Key stimulators of angiogenesis include VEGF-A, which binds to its primary receptor VEGFR-2 on endothelial s, activating downstream PI3K/Akt pathways that enhance , , and . Angiopoietin-1 (Ang-1), acting via the Tie2 receptor, complements VEGF by recruiting s and stabilizing walls, preventing leakage and supporting long-term functionality. Inhibitors such as endostatin and angiostatin, derived from XVIII and plasminogen respectively, counteract these effects by suppressing endothelial and , often through interference with signaling. Additionally, Notch signaling mediates between tip and stalk s, restricting excessive branching and ensuring orderly sprout patterning via Delta-like 4 (Dll4) ligands.00043-4) Vessel density resulting from angiogenesis is quantitatively assessed using microvessel density (MVD), a histological method that counts CD31- or von Willebrand factor-positive endothelial structures in tumor or tissue sections to gauge angiogenic activity and correlate it with disease progression. High MVD often indicates robust neovascularization, providing a prognostic marker in contexts like cancer.

Arteriogenesis

Arteriogenesis refers to the adaptive growth and remodeling of pre-existing arterioles into larger arteries, primarily in response to hemodynamic forces such as increased fluid following vascular . This process enhances conductance vessel function to bypass obstructions and restore blood flow, occurring predominantly in non-ischemic regions where vessels experience elevated tangential forces. Unlike other forms of vascular , arteriogenesis involves structural enlargement, with arteries potentially increasing in diameter by up to 20-fold through coordinated cellular and molecular events. The process begins with endothelial cell activation by , leading to the expression of adhesion molecules and that facilitate recruitment into the vessel wall. Recruited monocytes differentiate into macrophages, which release proteases to degrade the (ECM) and promote smooth muscle cell (SMC) and , enabling vessel wall thickening and lumen expansion. ECM remodeling further supports this growth through the deposition of new and , driven by matrix metalloproteinases (MMPs) from macrophages and SMCs, resulting in a more robust arterial structure capable of withstanding higher pressures. Key regulators include monocyte chemoattractant protein-1 (MCP-1), which mediates monocyte chemotaxis via the CCR2 receptor to initiate inflammatory infiltration. Platelet-derived growth factor-BB (PDGF-BB) promotes and SMC recruitment for vessel stabilization, while (bFGF) stimulates endothelial and SMC proliferation to drive outward remodeling. These factors act synergistically under to amplify arteriogenic responses. Arteriogenesis differs from by targeting pre-existing conductance vessels rather than forming new , with success often measured by the collateral flow index, which quantifies improved arterial independent of capillary density. Although both can be triggered by ischemic conditions, arteriogenesis emphasizes flow-mediated over hypoxia-driven . Experimental models, such as hindlimb ischemia induced by ligation in mice, demonstrate flow-dependent collateral artery growth, where shear stress in upstream arterioles leads to partial restoration of within weeks through monocyte-dependent mechanisms. In these models, genetic ablation of MCP-1 impairs collateral development, underscoring its essential role.

Physiological Roles

Embryonic and Developmental Vasculogenesis

, the de novo formation of blood vessels from endothelial progenitor cells known as angioblasts, initiates during early embryogenesis to establish the primary vascular network. In humans, this process begins around week 3 of , coinciding with , when mesodermal cells in the extraembryonic differentiate into hemangioblasts—bipotent precursors capable of giving rise to both endothelial and hematopoietic lineages. These hemangioblasts aggregate to form blood islands, which develop into the initial vascular structures, including the vasculature and the primitive that will contribute to the great vessels of the . Spatially, embryonic vasculogenesis exhibits precise patterning driven by mesodermal origins and migratory cues. The paired dorsal aortae, which form the central axial vessels, primarily arise from angioblasts in the splanchnic lateral plate , with additional contributions from the paraxial , particularly the dermomyotome, where endothelial precursors delaminate and migrate to incorporate into the aortic . Intersomitic vessels, which supply the developing somites, emerge via vasculogenesis from angioblasts in the segmental plate—an unsegmented region of the paraxial —before transitioning to patterns along the trunk. This organized spatial assembly ensures efficient coverage of the , with the vessels facilitating early nutrient exchange and the intraembryonic networks supporting . Genetic regulation orchestrates hemangioblast specification and vascular tube formation through key transcription factors. The TAL1/SCL factor is indispensable for committing mesodermal progenitors to the , activating downstream genes like FLK1 and promoting endothelial ; its absence in model organisms results in complete failure of and embryonic lethality due to lack of blood vessels. Mutations or disruptions in TAL1/SCL and related pathways, such as those involving or factors, are associated with congenital vascular malformations, including hypoplastic vessels and disorganized networks observed in developmental syndromes. Following primary , the process largely gives way to for vascular expansion, though remnants persist postnatally. In adults, vasculogenesis is rare and confined to specific contexts, such as reactivation within stem cell niches like the bone marrow, where endothelial progenitor cells (EPCs) derived from hematopoietic stem cells contribute to vessel formation under regenerative demands. These postnatal EPCs mirror embryonic hemangioblasts in function, integrating into existing vasculature or forming new segments in response to ischemic or hypoxic signals, highlighting a conserved from .

Wound Healing and Tissue Repair

Neovascularization plays a critical role in the proliferative phase of , which typically spans days 4 to 21 post-injury, where it facilitates the formation of through . During this phase, endothelial cells proliferate and migrate to form new capillaries, providing essential oxygen and nutrients to support tissue regeneration and re-epithelialization. The process begins with the degradation of the provisional matrix by proteases, allowing endothelial sprouts to invade and organize into functional vessels within the . Key regulators of this neovascularization include hypoxia-induced (VEGF) secreted by macrophages, which promotes endothelial , migration, and tube formation in the oxygen-deprived wound bed. Transforming growth factor-beta (TGF-β), released by various cells including platelets and fibroblasts, aids in matrix integration by enhancing production and stabilizing newly formed vessels with and cells. As healing progresses into the remodeling phase, excess vessels regress through endothelial cell , restoring a balanced vascular network and preventing chronic inflammation. Clinically, impaired neovascularization contributes to delayed closure in diabetic patients, often due to diminished VEGF responsiveness and hyperglycemia-induced . Conversely, excessive neovascularization can lead to hypertrophic scarring in keloids, where persistent angiogenic signaling sustains abnormal tissue growth. Experimental models, such as the ear chamber , have been instrumental in quantifying vessel ingrowth rates. Additionally, bone marrow-derived endothelial progenitor cells contribute vasculogenesis-like processes to support this repair.

Other Physiological Contexts

Neovascularization also supports adaptation in during endurance exercise training, where increased capillary density enhances oxygen delivery and metabolic efficiency. This is driven by from elevated blood flow and hypoxic signals, leading to of endothelial cells and improved endurance capacity, as observed in human studies showing up to 20-30% increases in capillary-to-fiber ratios after prolonged training. In adipose tissue expansion, such as during weight gain, neovascularization is essential for supplying nutrients to growing adipocytes, preventing hypoxia and fibrosis. Angiogenic factors like VEGF promote vessel sprouting within the expanding fat pads, with disruptions linked to unhealthy obesity phenotypes characterized by inflammation and metabolic dysfunction.

Pathological Conditions

Ocular Neovascularization Disorders

Ocular neovascularization disorders encompass a range of conditions characterized by the abnormal growth of blood vessels in the retina, choroid, or iris, primarily driven by retinal ischemia and hypoxia. In diabetic retinopathy (DR), chronic hyperglycemia leads to retinal capillary occlusion and ischemia, which upregulates vascular endothelial growth factor (VEGF) expression in retinal glial cells, promoting neovascularization. Similarly, in retinopathy of prematurity (ROP), hyperoxia-induced vaso-obliteration followed by relative hypoxia in premature infants triggers VEGF release, leading to aberrant vessel proliferation. These processes primarily involve angiogenesis, where new vessels sprout from existing capillaries in response to hypoxic signals. The clinical manifestations of these disorders often result in significant vision impairment through several mechanisms. Neovascular vessels are fragile and prone to leakage, causing that distorts central vision. More severe complications include vitreous hemorrhage, where blood from ruptured vessels obscures the visual axis, and tractional due to fibrovascular proliferation pulling on the . These features collectively contribute to progressive vision loss, with potential for sudden and profound deficits if untreated. Diagnosis relies on advanced imaging modalities to visualize and characterize neovascular activity. (FA) is a cornerstone technique, revealing hyperfluorescent leakage patterns from abnormal vessels and areas of non-perfusion indicative of ischemia. (OCT), including OCT angiography (OCTA), provides high-resolution cross-sectional images and non-invasive vessel quantification, detecting neovascular membranes and associated edema without dye injection. These tools enable early identification and monitoring of disease progression. Epidemiologically, proliferative DR, a key neovascular form, affects approximately 3.5% of diabetic adults in the United States as of 2021, with vision-threatening diabetic retinopathy (including neovascular complications) impacting 5.06% of the diabetic population. The incidence of PDR has declined by nearly 300% since 2002, but overall DR cases continue to rise with increasing diabetes prevalence, estimated at around 10 million affected individuals in the US by 2025 amid aging populations and , with projections indicating a substantial global burden through the 2020s.

Cancer-Associated Neovascularization

Cancer-associated neovascularization refers to the process by which tumors stimulate the formation of new blood vessels to support their growth and progression. This phenomenon was first hypothesized by in 1971, who proposed that tumor expansion is limited by the need for and that inhibiting this process could serve as a therapeutic strategy. Folkman's seminal work laid the foundation for understanding tumors as "wounds that do not heal," highlighting the dependency of solid tumors on neovascularization for nutrient supply and oxygen delivery beyond a microscopic size of approximately 1-2 mm³. A pivotal event in this process is the angiogenic switch, where the balance shifts from an avascular, dormant state to one dominated by pro-angiogenic signals that exceed anti-angiogenic inhibitors. This switch is often triggered by oncogenes such as mutant , which upregulate the expression of key pro-angiogenic factors including (VEGF) and (bFGF). For instance, oncogenic H-RAS stimulates tumor angiogenesis through both autocrine VEGF production in tumor cells and paracrine bFGF secretion from stromal cells, enabling rapid vessel formation and tumor expansion. The resulting imbalance promotes endothelial cell proliferation, migration, and tube formation, transforming the tumor from a preclinical, non-invasive into a malignant one capable of sustained growth. Within the tumor microenvironment, hypoxic conditions in the expanding tumor core play a central role in driving neovascularization. Hypoxia-inducible factor-1α (HIF-1α), stabilized under low oxygen levels, transactivates genes encoding pro-angiogenic molecules like VEGF, further amplifying vessel recruitment. Additionally, hypoxic tumors secrete stromal cell-derived factor-1 (SDF-1), which mobilizes endothelial progenitor cells (EPCs) from the to incorporate into nascent vessels, enhancing neovascularization and tumor perfusion. This recruitment of EPCs, often CD45+ myeloid cells, integrates into the tumor vasculature, supporting its immaturity and leakiness, which facilitates immune evasion and nutrient delivery. Clinically, cancer-associated neovascularization enables by providing a vascular route for tumor and is associated with adverse outcomes. High microvessel density (MVD), a histological marker of assessed via or CD105 staining, correlates with poor in various cancers; for example, elevated MVD in predicts shorter disease-free survival and increased metastatic risk. Similarly, in colon cancer, high intratumoral MVD is linked to reduced overall survival, particularly in advanced stages, underscoring its role as an independent prognostic indicator. These vessels not only sustain growth but also create a permissive niche for circulating tumor cells to extravasate and colonize distant sites, exacerbating disease progression.

Cardiovascular Ischemic Diseases

In ischemic heart disease, neovascularization primarily manifests as collateral arteriogenesis, which is triggered by increased tangential fluid on endothelial cells following (). This process enlarges pre-existing arterioles into functional collateral vessels, providing alternative blood flow to ischemic myocardium and reducing infarct size. However, arteriogenesis is often impaired in clinical settings, serving as an adaptive response to restore but limited by factors such as advanced age and comorbidities like , which reduce endothelial progenitor cell () recruitment and . In (PAD), neovascularization through and arteriogenesis represents an adaptive mechanism to bypass occlusions in lower limb arteries, potentially alleviating . Pro-angiogenic factors, such as vascular endothelial growth factor (VEGF), hold therapeutic promise by stimulating capillary sprouting and collateral maturation to enhance limb perfusion. Yet, this response can become maladaptive, as excessive VEGF-induced vessel permeability increases the risk of hemorrhage and edema, complicating treatment in patients with critical limb ischemia. Key regulators modulate these processes; adipokines like enhance EPC mobilization from , promoting their incorporation into ischemic neovessels and improving post-ischemic recovery. Similarly, statins augment arteriogenesis by upregulating endothelial (eNOS) activity, which supports endothelial cell survival and shear stress-mediated vessel remodeling without directly affecting levels in this context. Recent advances in the 2020s, including follow-up analyses of VEGF trials for , demonstrate mixed outcomes, with some improvements in ankle-brachial index and but inconsistent reductions in rates or sustained benefits. These findings highlight the need for optimized methods to balance adaptive neovascularization against maladaptive vascular instability.

Therapeutic Strategies

Anti-Neovascularization Therapies

Anti-neovascularization therapies primarily target the (VEGF) pathway, a key driver of pathological , by blocking VEGF signaling to inhibit excessive vessel formation in diseases like cancer and ocular disorders. Monoclonal antibodies such as (Avastin), the first FDA-approved anti-VEGF agent in 2004, directly bind to VEGF-A isoforms, preventing their interaction with VEGF receptors (VEGFRs) on endothelial cells and thereby suppressing endothelial proliferation, migration, and survival. This mechanism has been foundational, as VEGF-A is the predominant isoform promoting neovascularization in pathological contexts. In , is commonly used in combination regimens to enhance efficacy against solid tumors by normalizing tumor vasculature and improving drug delivery. For instance, in metastatic , combined with fluorouracil-based extended median overall survival by 4.7 months (from 15.6 to 20.3 months) in the pivotal AVF2107g , establishing its role in first-line therapy. Similarly, in , intravitreal injections of (Lucentis), a fragment derived from the same parent as and FDA-approved in 2006, bind VEGF-A with high affinity to treat neovascular age-related (wet ), reducing vascular leakage and stabilizing or improving (losing fewer than 15 letters) in about 94.5% of patients, with approximately 34.7% gaining 15 or more letters, over two years. These applications highlight the therapy's specificity for ocular neovascularization, where localized delivery minimizes systemic exposure. Despite their success, therapies face challenges including acquired resistance, often mediated by upregulation of alternative angiogenic pathways such as (FGF) signaling, which can compensate for VEGF inhibition and sustain tumor progression. Additionally, common side effects like , , and increased risk of arterial arise from systemic VEGF blockade, affecting up to 20-30% of patients on and necessitating careful monitoring. Emerging strategies address these limitations through multi-kinase inhibitors that target multiple receptors involved in neovascularization. (Sutent), approved by the FDA in 2006 for advanced , inhibits VEGFR-1/2/3, PDGFR-β, and other kinases, achieving objective response rates of approximately 7% but clinical benefit rates (including stable disease) of around 53% in gastrointestinal stromal tumors resistant to prior therapies by broadly disrupting angiogenic signaling. These agents offer a complementary approach, potentially overcoming single-pathway resistance, though they introduce additional toxicities like hand-foot syndrome.

Pro-Neovascularization Therapies

Pro-neovascularization therapies aim to stimulate the formation of new blood vessels in ischemic tissues, such as those affected by cardiovascular diseases, to improve and promote tissue repair. These approaches primarily target the delivery of angiogenic factors like (VEGF) and (FGF) to enhance neovascularization in conditions where natural is insufficient. Unlike anti-angiogenic strategies, these therapies seek to harness physiological vessel growth mechanisms for therapeutic benefit in regenerative contexts. Gene therapy represents a key approach, involving the intramuscular or epicardial injection of plasmids encoding pro-angiogenic factors to induce local expression. For instance, the pCMV-VEGF165 plasmid (Neovasculgen), which carries the VEGF-165 , has been used to stimulate in ischemic diseases through direct plasmid injection, demonstrating in preclinical and early clinical models by increasing density. Similarly, NV1FGF, a plasmid-based encoding FGF-1, has been administered via intramuscular injection in patients with critical limb ischemia, showing improved limb and reduced rates in phase II trials, although a subsequent phase III trial (TAMARIS) did not confirm the reduction in major rates as its primary endpoint. Protein delivery methods complement by providing direct administration of recombinant angiogenic factors; recombinant FGF-1, for example, has been tested in preclinical models of hindlimb ischemia, where daily intramuscular injections led to significant improvements in blood flow and collateral vessel formation. In applications for refractory angina, adenoviral vector-based therapies like AdVEGF-D have shown promise in clinical trials during the and beyond. The phase I KAT301 trial demonstrated the safety and feasibility of intramyocardial AdVEGF-D injection, resulting in improved exercise tolerance and reduced episodes in patients with no-option refractory , with long-term follow-up confirming sustained benefits up to five years post-treatment. The more recent EXACT phase II trial using XC001 (AdVEGFXC1), an adenoviral VEGF construct, reported enhancements in exercise time, ischemia reduction, and symptom relief in 32 patients with refractory . Stem cell-based strategies, particularly involving endothelial progenitor cells (), have been applied for myocardial repair following ; EPCs home to ischemic sites, secrete paracrine factors to promote neovascularization, and contribute to vascular regeneration, as evidenced in preclinical models where EPC transplantation increased density and improved cardiac function. Despite these advances, pro-neovascularization therapies face significant challenges, including transient effects from limited duration—plasmid-based vectors often yield short-lived activity, reducing long-term —and off-target growth risks, such as unintended that could exacerbate tumors or cause . Regulatory hurdles persist, with no VEGF-based gene therapies approved by the FDA for pro-angiogenic indications as of 2025, due to inconsistent clinical outcomes and safety concerns in large-scale trials. Recent developments in the have focused on delivery systems to enable sustained release of pro-angiogenic factors, addressing transience issues in preclinical studies. For example, nanoporous silica loaded with VEGF have demonstrated controlled release over weeks in ischemic models, enhancing stable neovascularization and tissue perfusion without bolus-related side effects. Similarly, hydrogel-embedded VEGF have shown improved angiogenic outcomes in hindlimb ischemia models, promoting mature vessel formation with reduced leakage. These innovations hold potential for more targeted and durable therapies in ischemic cardiovascular conditions.

References

  1. [1]
    Overview of Angiogenesis - NCBI - NIH
    The term neovascularization means the formation of any blood vessel in the adult regardless of its size or type. Go to: 1.3. THE ANGIOGENIC PROCESS. 1.3.1 ...<|control11|><|separator|>
  2. [2]
    Neovascularization (Pathology) - an overview | ScienceDirect Topics
    Neovascularization (NV) is a process whereby new vasculature forms in avascular tissues or tissue space via a combination of vasculogenesis and angiogenesis.
  3. [3]
    neovascularization, n. meanings, etymology and more
    neovascularization is formed within English, by compounding. Etymons: neo- comb. form, vascularization n. See etymology ...
  4. [4]
    Patterning of embryonic blood vessels - Developmental Dynamics
    Jul 23, 2003 · The morphogenesis of embryonic blood vessels has been studied since the beginning of the twentieth century. Sabin was one of the first to ...
  5. [5]
    Hypoxia-Induced Angiogenesis: Good and Evil - PMC
    Hypoxia promotes vessel growth by upregulating multiple pro-angiogenic pathways that mediate key aspects of endothelial, stromal, and vascular support cell ...The Major Oxygen Homeostasis... · Figure 1 · Hypoxia And Hif Signaling In...<|control11|><|separator|>
  6. [6]
    Vascular endothelial growth factor signaling in health and disease
    May 19, 2025 · One of the most significant regulatory mechanisms of VEGF-A transcription is hypoxia, which is mediated by hypoxia-inducible factor 1 (HIF-1).
  7. [7]
    The role of endothelial cell–pericyte interactions in vascularization ...
    Endothelial cells (ECs) and pericytes (PCs) play synergistic roles in angiogenesis. ECs promote vascular development by releasing signaling molecules, such as ...
  8. [8]
    Extracellular Matrix Regulation of Vascular Morphogenesis ...
    The extracellular matrix represents a critical regulator of tissue vascularization during embryonic development and postnatal life.
  9. [9]
    Hypoxia-Inducible Factor-1 in Physiological and Pathophysiological ...
    The HIF-1 pathway was found to be a master regulator of angiogenesis. Whether the process is physiological or pathological, HIF-1 seems to participate in ...
  10. [10]
    Molecular Basis for the Regulation of Angiogenesis by ...
    Thrombospondins TSP-1 and TSP-2 are potent endogenous inhibitors of angiogenesis. They inhibit angiogenesis through direct effects on endothelial cell ...
  11. [11]
    Pathophysiology of Tumor Neovascularization - PubMed Central - NIH
    The present review focuses on features of neovascularization under physiological and pathological conditions, with an emphasis on tumor neovascularization.
  12. [12]
    Hallmarks of Cancer: New Dimensions | Cancer Discovery
    Jan 12, 2022 · Given the growing appreciation that tumors can become sufficiently vascularized either by switching on angiogenesis or by co-opting normal ...Unlocking Phenotypic... · Transdifferentiation · Senescent Cells<|separator|>
  13. [13]
    Vasculogenesis and Angiogenesis: Molecular and Cellular Controls
    Schematically, vasculogenesis, defined as the differentiation of an embryonic stem cell, precursor of the endothelial cell or angioblast, can be distinguished ...
  14. [14]
    Mechanisms of Endothelial Differentiation in Embryonic ...
    Consistent with its central importance in embryonic development, the history of research into vascular development is populated with seminal histologists such ...Missing: neovascularization | Show results with:neovascularization
  15. [15]
    Vasculogenesis - an overview | ScienceDirect Topics
    Vasculogenesis is the process of forming de novo blood vessels through the differentiation of progenitor cells to endothelial cells (ECs) and their organization ...Missing: history | Show results with:history
  16. [16]
    Relationship between vasculogenesis, angiogenesis and ...
    Mar 1, 1989 · Quail-chick intracoelomic grafts of organ rudiments were used to study the origin of endothelia and haemopoietic cells during avian ...
  17. [17]
    Relationship between vasculogenesis, angiogenesis and ... - PubMed
    This study demonstrates that, in ontogeny, vascularization obeys different rules depending on which germ layer the mesoderm is associated with.
  18. [18]
    [PDF] Vasculogenesis and angiogenesis in the mouse embryo studied ...
    Using the quail/chick chimera model (Le Douarin, 1969), angiogenesis was shown to occur during development of the vessels of the brain (Stewart and Wiley ...Missing: historical discovery 1990s
  19. [19]
    Embryonic vasculogenesis and hematopoietic specification - PMC
    Vasculogenesis is the process by which blood vessels are formed de novo. This process first occurs in the embryonic yolk sac of mammalian embryos.
  20. [20]
    Signal Transduction in Vasculogenesis and Developmental ...
    The process of vessel formation at this early stage of development is vasculogenesis, a term coined by Risau and colleagues in 1988 (Risau and Lemmon, 1988; ...
  21. [21]
    The role of FGF and VEGF in angioblast induction and migration ...
    Nov 16, 2000 · Quail/chick chimeras have proven very useful to address the role of FGF-2 in vasculogenesis. ... In the quail, two VEGF receptor-related molecules ...
  22. [22]
    In Vitro Models of Vasculogenesis and Angiogenesis - Nature
    Apr 1, 2001 · Vasculogenesis is defined as the differentiation of precursor cells (angioblasts) into endothelial cells and the de novo formation of a ...
  23. [23]
    Embryonic stem cell-derived cystic embryoid bodies form vascular ...
    Feb 1, 1992 · Murine embryonic stem cells can differentiate in vitro to form cystic embryoid bodies (CEB) that contain different structures and cell types ...
  24. [24]
    Model of Vasculogenesis from Embryonic Stem Cells for Vascular ...
    Jun 5, 2006 · Embryonic stem cell-derived cystic embryoid bodies form vascular channels: an in vitro model of blood vessel development. Development. 1992 ...
  25. [25]
    Cell behaviors and dynamics during angiogenesis | Development
    Jul 1, 2016 · During developmental angiogenesis, the formation of angiogenic sprouts can be divided into three major phases: (1) tip cell selection, during ...
  26. [26]
    “Sprouting angiogenesis”, a reappraisal - ScienceDirect.com
    Dec 15, 2012 · Angiogenesis is defined as a new blood vessel sprouting from pre-existing vessels. This highly regulated process take place through two non-exclusive events.
  27. [27]
    Complex role of matrix metalloproteinases in angiogenesis - Nature
    Sep 1, 1998 · Interstitial collagenase (MMP-1) is also required for angiogenesis in vitro. Vascular endothelial growth factor (VEGF) induces MMP-1 expression ...Missing: paper | Show results with:paper
  28. [28]
    VEGF guides angiogenic sprouting utilizing endothelial tip cell ...
    We show here that VEGF-A controls angiogenic sprouting in the early postnatal retina by guiding filopodial extension from specialized endothelial cells situated ...Tip Cell Filopodia Extend On... · Vegf-A Stimulates Tip Cell... · Vegf Stimulates Tip Cell...
  29. [29]
    Mechanisms of Vessel Branching: Filopodia on Endothelial Tip Cells ...
    Mar 5, 2009 · “Stalk cells” trail behind the tip cell and elongate the stalk of the sprout; they proliferate, form junctions, lay down extracellular matrix, ...Missing: steps | Show results with:steps
  30. [30]
    Molecular Bases of VEGFR-2-Mediated Physiological Function and ...
    The VEGF-activated VEGFR-2 stimulates and mediates variety of signaling transduction, biological responses, and pathological processes in angiogenesis.
  31. [31]
    Requisite Role of Angiopoietin-1, a Ligand for the TIE2 Receptor ...
    Angiopoietin-1 is a primary physiologic ligand for TIE2 and that it has critical in vivo angiogenic actions that are distinct from VEGF.
  32. [32]
    An Endogenous Inhibitor of Angiogenesis and Tumor Growth
    Jan 24, 1997 · Endostatin is a specific inhibitor of endothelial proliferation and is a potent angiogenesis inhibitor. Systemic therapy with endostatin causes a nearly ...
  33. [33]
    Microvessel Density as a Prognostic Factor in Women with Breast ...
    To conclude, our meta-analysis found a statistically significant inverse relationship between angiogenesis, assessed by MVD, and survival, confirming that human ...Abstract · Introduction · Materials and Methods · Results
  34. [34]
    Vascular Growth in Health and Disease - Frontiers
    It may prove helpful to go back to the lab to accurately distinguish between vasculogenesis, angiogenesis, and arteriogenesis contributing to the disease in ...
  35. [35]
  36. [36]
    New Directions in Therapeutic Angiogenesis and Arteriogenesis in ...
    This review will focus on what has been learned to advance medical revascularization for PAD and how that information might lead to novel approaches for ...
  37. [37]
    Lecture - Early Vascular Development - UNSW Embryology
    Sep 7, 2017 · This lecture has 2 parts; firstly introducing an overview of early vascular development, secondly the key events in heart development.
  38. [38]
    Dorsal aorta polarization and haematopoietic stem cell emergence
    Jan 5, 2023 · The paraxial (somitic) mesoderm provides ECs for the vascularization of the trunk and limbs, while the splanchnopleural mesoderm provides ...
  39. [39]
    Dermomyotome-derived endothelial cells migrate to the dorsal aorta ...
    Sep 11, 2023 · A rare population of endothelial precursors delaminates from the dermomyotome to incorporate exclusively into the developing dorsal aorta.
  40. [40]
    Three-Dimensional Analysis of Vascular Development in the Mouse ...
    Intersomitic (intersegmental) vessels are a useful model for sprouting angiogenesis and vessel pathfinding. They are the first vessels in the embryo to form by ...
  41. [41]
    Specification of hematopoietic and vascular development by the ...
    Oct 15, 1999 · Among the nuclear regulators critical to the proper development of hematopoietic and vascular cells is the transcription factor SCL/tal-1/tcl-5 ...
  42. [42]
    Transcription factors regulating vasculogenesis and angiogenesis
    Feb 16, 2023 · Role of SCL/Tal-1, GATA, and ets transcription factor binding sites for the regulation of flk-1 expression during murine vascular development.
  43. [43]
    The Role of Endothelial Progenitor Cells in Postnatal Vasculogenesis
    EPCs are mainly located within the stem cell niche in BM, along with some circulating populations in the peripheral blood. The process by which EPCs contribute ...Missing: remnants | Show results with:remnants
  44. [44]
    Human endothelial stem/progenitor cells, angiogenic factors and ...
    Sep 15, 2010 · Evidence for the potential location of vasculogenic niches within vessel walls after birth is discussed below. ... postnatal angiogenesis and ...Human Endothelial... · 2. Endothelial Progenitor... · 3.2. Cell Therapies And...<|control11|><|separator|>
  45. [45]
    Wound Healing Phases - StatPearls - NCBI Bookshelf
    The proliferative phase is characterized by the formation of granulation tissue, reepithelialization, and neovascularization. This phase can last several weeks.
  46. [46]
    MicroRNA-132 enhances transition from inflammation to ... - NIH
    The subsequent proliferative phase (4–21 days after injury) focuses on reepithelialization, formation of granulation tissue, and restoration of the vascular ...<|control11|><|separator|>
  47. [47]
    Angiogenesis and wound repair: when enough is enough - PMC - NIH
    Macrophages, activated epithelial cells, and other inflammatory cells can release proangiogenic mediators such as VEGF and CXC chemokines, supporting robust ...<|control11|><|separator|>
  48. [48]
    Inflammation and wound healing: The role of the macrophage - PMC
    The hypoxic environment of the wound also promotes inflammation, as hypoxia stimulates numerous cell types, including macrophages, to produce mediators ...
  49. [49]
    Signalling by Transforming Growth Factor Beta Isoforms in Wound ...
    Transforming growth factor beta (TGFβ) signalling is essential for wound healing, including both non-specific scar formation and tissue-specific regeneration.
  50. [50]
    Angiogenesis in Wound Repair: Too Much of a Good Thing? - PMC
    During resolution, most of the newly formed vessels regress. The apoptotic endothelial cells that are formed as the vascular bed is pruned can be engulfed by ...
  51. [51]
    Research progress on the mechanism of angiogenesis in wound ...
    This paper comprehensively integrates the regulatory mechanisms of new blood vessel formation in wound healing, focusing on the proliferation and migration of ...
  52. [52]
    Characterization of the skin keloid microenvironment - PMC - NIH
    Aug 16, 2023 · Neovascularization is critical for transporting immune cells, oxygen, cytokines and nutrients to the microenvironment in wound healing, and ...<|control11|><|separator|>
  53. [53]
    Current methods for assaying angiogenesis in vitro and in vivo - PMC
    The rabbit ear chamber is expensive for routine use, and a period of 4–6 weeks must pass after surgery before the test substance can be placed into the chamber.
  54. [54]
    The Role of Vascular Endothelial Growth Factor in Wound Healing
    VEGF induces endothelial cell migration in wound healing through two primary mechanisms, chemotaxis and vasodilatation. In the initial phase of angiogenesis, ...
  55. [55]
    Targeting Neovascularization in Ischemic Retinopathy
    Hypoxia-induced VEGF expression in retinal glial cells plays a key role in formation of new retinal vessels [40]. The capacity of Müller cells to preserve the ...
  56. [56]
    Retinopathy of prematurity: A review of pathophysiology and ...
    Nov 22, 2022 · In phase 2, these vascular changes result in retinal ischemia and trigger the release of growth factors leading to abnormal intravitreal ...
  57. [57]
    Macular Edema - StatPearls - NCBI Bookshelf
    Macular edema is a retinal condition characterized by fluid accumulation in the macula, or central part of the retina, responsible for sharp, central vision.
  58. [58]
    Ocular Neovascularization - PMC - PubMed Central - NIH
    When traction retinal detachment involves the macula, which is responsible for reading and driving vision, severe visual loss occurs. Figure 2. Fluorescein ...
  59. [59]
    Mechanisms of Acquired Resistance to Anti-VEGF Therapy ... - IOVS
    The hyperglycemia-induced ischemia present in DR is considered a major contributing factor to the upregulation of VEGF, which in turn plays a central role in ...<|control11|><|separator|>
  60. [60]
    Fluorescein Angiography in Neovascular AMD
    Aug 25, 2008 · Fluorescein angiography is a powerful imaging modality to identify the presence, location and size of the neovascular complex.
  61. [61]
    Choroidal Neovascularization: OCT Angiography Findings - EyeWiki
    Sep 18, 2025 · Type 1 CNV demonstrates occult leakage on fluorescein angiography. Polypoidal choroidal vasculoplathy (PCV) is a subtype of Type 1 CNV that is ...Disease Entity · Classification · Diagnosis · Diagnostic Procedures
  62. [62]
    Prevalence of Diabetic Retinopathy in the US in 2021 - PMC - NIH
    Jun 15, 2023 · The study team estimated that 9.60 million people in the US (26.43% of those with diabetes) had diabetic retinopathy and 1.84 million people (5.06% of those ...
  63. [63]
    Global Prevalence of Diabetic Retinopathy Over the Last Decade ...
    Jul 24, 2025 · Results Preliminary synthesis indicates an overall global DR prevalence averaging approximately 27%, with higher rates reported in Africa (up to ...Missing: 2020s | Show results with:2020s
  64. [64]
    A review of Judah Folkman's remarkable achievements in biomedicine
    Sep 9, 2008 · In 1971, Folkman proposed a hypothesis that tumor growth is angiogenesis-dependent (2). In that report, Folkman showed preliminary evidence that ...Sign Up For Pnas Alerts · Molecular Basis Of The... · Clinical Drug DeliveryMissing: original | Show results with:original
  65. [65]
    Oncogenic H-ras stimulates tumor angiogenesis by two ... - PNAS
    The angiogenic switch is a critical control point for tumor expansion. The ability of a tumor to become neovascularized permits rapid expansion of tumor growth ...
  66. [66]
    Oncogenes and Tumor Angiogenesis: Differential Modes of ...
    A possible link between oncogenes and tumor angiogenesis has been implicated by the finding that expression of various oncogenes,particularly mutant ras, ...Missing: bFGF | Show results with:bFGF
  67. [67]
    Angiogenic signaling pathways and anti-angiogenic therapy for cancer
    May 11, 2023 · VEGF-C and VEGF-D encourage tumor growth and metastasis through lymphangiogenesis and lymphatic metastasis, which is mediated by VEGFR-3.
  68. [68]
    Hypoxic microenvironment in cancer: molecular mechanisms and ...
    Feb 17, 2023 · Hypoxia-induced HIF-1α can promote programmed death ligand-1 (PD-L1) expression in cancer cells and suppress immune effects. Moreover, hypoxia ...
  69. [69]
    HIF1α Induces the Recruitment of Bone Marrow-Derived Vascular ...
    We demonstrate that HIF1α, the direct effector of hypoxia, partly through increases in SDF1α, induces recruitment of bone marrow-derived CD45+ myeloid cells.
  70. [70]
    Tumor Hypoxia Regulates Immune Escape/Invasion - Frontiers
    Hypoxia in the tumor microenvironment promotes tumor angiogenesis and abnormal vascularization through the activation of HIF-1α and downstream effector genes, ...
  71. [71]
    Microvessel density and VEGF expression are prognostic factors in ...
    Jun 13, 2006 · Microvessel density (MVD), as a surrogate marker of tumoral angiogenesis, has been proposed to identify patients at high risk of recurrence.
  72. [72]
    Prognostic value of microvessel density in stage II and III colon ... - NIH
    Aug 16, 2019 · MVD is an independent prognostic factor associated with poor DFS in stage II colon cancer patients, and with better DFS in stage III colon cancer patients.
  73. [73]
    The collateral circulation of the heart - BMC Medicine - BioMed Central
    Jun 4, 2013 · The most important trigger for collateral growth, called arteriogenesis, is tangential fluid shear stress at the endothelial level along with ...
  74. [74]
    Diabetic dyslipidemia impairs coronary collateral formation: An update
    Aug 21, 2022 · Coronary collateralization is substantially impaired in patients with type 2 diabetes and occlusive coronary artery disease, which leads to aggravated ...Missing: comorbidities | Show results with:comorbidities
  75. [75]
    Effects of Aging on the Collateral Circulation, and Therapeutic ...
    Aug 7, 2025 · Decreased vascular reserve and weak collateral presence are factors that accelerate infarct growth in the elderly, [26] For similar reasons, we ...
  76. [76]
    Gene Therapeutic Strategies for Peripheral Artery Disease and New ...
    This article discusses gene therapy approaches in the context of PAD, both intermittent claudication and critical limb ischemia, and the promise of adeno- ...
  77. [77]
    Therapeutic angiogenesis and tissue revascularization in ischemic ...
    Feb 16, 2023 · This review will comprehensively describe the main pathogenesis of vascular injury in ischemic vascular disease, the therapeutic function of the above three ...
  78. [78]
    Adiponectin promotes endothelial progenitor cell number and function
    Here, we investigated the effects of adiponectin on EPCs. EPC levels did not increase in adiponectin deficient (APN-KO) in response to hindlimb ischemia.
  79. [79]
    Endothelial nitric oxide synthase is essential for the HMG‐CoA ...
    Sep 17, 2001 · Thus, it is plausible that activation of eNOS by statins favors the process of arteriogenesis in response to tissue ischemia. Consistent ...
  80. [80]
  81. [81]
    Human Studies of Angiogenic Gene Therapy - PMC - NIH
    The first investigation to use VEGF165 plasmid for myocardial gene therapy was an open-label study in which the plasmid was administered by epicardial injection ...Vegf-C · Fibroblast Growth Factor... · Hepatocyte Growth Factor...
  82. [82]
    Gene Therapeutic Drug pCMV-VEGF165 Plasmid ('Neovasculgen ...
    Sep 17, 2024 · Plasmids carrying the VEGF-165 gene to stimulate angiogenesis are widely used in the therapy of ischemic diseases, both in animal models and ...Missing: neovascularization therapies
  83. [83]
    NV1FGF, a pCOR plasmid-based angiogenic gene therapy for the ...
    Preclinical studies using NV1FGF demonstrated restoration in capillary and arteriolar density in rabbit and hamster models of hind limb ischemia. In phase I and ...
  84. [84]
    Naked plasmid DNA encoding fibroblast growth factor type 1 for the ...
    The therapeutic effect of FGF-1 was studied in a rabbit model of hind limb ischemia. Daily intramuscular injection of recombinant FGF-1 (4 mg) for a ...
  85. [85]
    Long-term safety and efficacy of intramyocardial adenovirus ... - Nature
    Oct 1, 2021 · Phase I KAT301 trial was the first study that investigated the safety and efficacy of intramyocardial adenovirus-mediated VEGF-DΔNΔC (AdVEGF-D) ...
  86. [86]
    Angiogenic Gene Therapy for Refractory Angina: Results of the ...
    May 2, 2024 · The degree of benefit also exceeds that observed in the REVASC trial (Randomized Evaluation of VEGF for Angiogenesis), which also utilized open- ...
  87. [87]
    Endothelial progenitor cells in neovascularization of infarcted ...
    After ischemic insult, EPCs are believed to home to sites of neovascularization, where they contribute to vascular regeneration by forming a structural ...
  88. [88]
    Gene Therapy in Cardiac Surgery: Clinical Trials, Challenges, and ...
    Transient Gene Expression. One of the main factors limiting the positive effect of gene therapy is transient therapeutic gene expression [48]. This problem ...
  89. [89]
    Clinical Trials in Coronary Angiogenesis: Issues, Problems ...
    A key issue is that prolonged exposure to angiogenic stimulation may have considerable safety implications given the critical role of angiogenesis in ...
  90. [90]
    Pro-Angiogenic Bioactive Molecules in Vascular Morphogenesis
    Several VEGF mimetic peptides, gene therapy vectors, and small-molecule pro-angiogenic drugs are in clinical trials, but none have been FDA-approved.
  91. [91]
    Sustained delivery of functional vascular endothelial growth factor ...
    Jun 26, 2025 · As drug carriers, nanoporous silica nanoparticles (NPSNPs) were chosen and loaded with VEGF by the interaction with grafted amino groups.
  92. [92]
    Translational Studies on the Potential of a VEGF Nanoparticle ...
    This VEGF-nano in a HA-TA hydrogel delivery system is successfully integrated with an appropriate device for clinical use, demonstrates promising angiogenic ...