Vasculogenic mimicry

Last updated
Vascular mimicries (indicated by arrows) in melanoma A) H&E stain B) PanMelanoma cocktail stain. Vascular mimicry melanoma staining.jpg
Vascular mimicries (indicated by arrows) in melanoma A) H&E stain B) PanMelanoma cocktail stain.

Vasculogenic mimicry (VM) is a strategy used by tumors to ensure sufficient blood supply is brought to its cells through establishing new tumor vascularization. [1] [2] [3] This process is similar to tumor angiogenesis; on the other hand vascular mimicry is unique in that this process occurs independent of endothelial cells. Vasculature is instead developed de novo by cancer cells, which under stress conditions such as hypoxia, express similar properties to stem cells, capable of differentiating to mimic the function of endothelial cells and form vasculature-like structures. [1] [2] The ability of tumors to develop and harness nearby vasculature is considered one of the hallmarks of cancer disease development and is thought to be closely linked to tumor invasion and metastasis. [4] Vascular mimicry has been observed predominantly in aggressive and metastatic cancers and has been associated with negative tumor characteristics such as increased metastasis, increased tissue invasion, and overall poor outcomes for patient survival. [2] [3] Vascular mimicry poses a serious problem for current therapeutic strategies due to its ability to function in the presence of Anti-angiogenic therapeutic agents. In fact, such therapeutics have been found to actually drive VM formation in tumors, causing more aggressive and difficult to treat tumors to develop. [5]

Contents

The structure of tumor blood vessels formed through the known tumor angiogenesis pathways. Vasculogenesis, sprouting angiogenesis, Intussusceptive angiogenesis, vessel co-option, and both vascular mimicry tubular and patterned matrix types are shown. Types of Tumor Angiogenesis.jpg
The structure of tumor blood vessels formed through the known tumor angiogenesis pathways. Vasculogenesis, sprouting angiogenesis, Intussusceptive angiogenesis, vessel co-option, and both vascular mimicry tubular and patterned matrix types are shown.

Overview

Vascular mimicry was first discovered in 1999 by Maniotis et al. who identified blood supplying channels in malignant melanoma that were composed entirely of tumor cell based structures. They found that cancer cells had taken on endothelial cell properties and were forming blood conducting vessels independent of normal angiogenesis pathways. [2] This finding spurred interest within the research community to discover the cause of this pathway and its relevance to disease.[ citation needed ]

Researchers have since discovered that VM is closely linked to several signaling pathways including vascular signaling, embryogenesis, and the hypoxic response. [2]

Requirements for VM formation

In order for new vasculature to form through VM, several requirements must be met. These generally include aberrant expression of VE-cadherin, as well as changes to the density and composition of the extracellular matrix. [6] VE-cadherin is a transmembrane protein that is specifically expressed in endothelial cells to promote and maintain specific adhesion of endothelial cells critical to vascularization. However, aggressive and metastatic cancers have been found to have aberrant expression of VE-cadherin, allowing for formation of cancer cell specific vasculature. [7] VE-cadherin also regulates inter-cellular signaling pathways which promote invasion of cancer cells into nearby and distal tissues, a major component of metastatic disease. [7] VE-cadherin has also been linked to regulatory functions in cell proliferation, cell death, and the expression and function of VEGFRs. [8]

Visual representation of vascular mimicry formation under hypoxic conditions. EET (A subtype of EMT) drives cancer cells to take on endothelial-like properties to form vasculature. Vascular mimicry formation.webp
Visual representation of vascular mimicry formation under hypoxic conditions. EET (A subtype of EMT) drives cancer cells to take on endothelial-like properties to form vasculature.

Another major factor in the formation of VM in tumors is the adjustment of the extracellular matrix to promote vasculature formation. Aggressive tumors express elevated levels of proteins such as matrix metalloproteinases (MMPs), laminin5y2, and type-1 & 4 collagens. These glycoproteins secreted are into the ECM by cancer cells and act to clear space for new vessel formation, as well as promote recruitment and reorganization of cancer cells to form new blood conducting vessels. [6]

Identification and classification

Clinically, VM is diagnosed through immunohistochemistry (IHC) and Periodic acid-Schiff stain (PAS) of patient tumor biopsy. IHC staining identifies the expression of common biomarkers of endothelial cells such as CD31, while PAS staining marks the extracellular matrix for glycoproteins, laminin, proteoglycans, heparin sulfate and collagens, which are known to be a sign of VM. [2] Clinicians diagnose a tumor as having VM by CD31-/PAS+ expressing blood conducting vessels, indicating that there are no endothelial cells but still vasculature present. [2] [3]

Vascular mimicry may be divided into tubular and patterned matrix types. [9] [3] Tubular VM is characterized by channels surrounded by glycoprotein covered tumor cells where endothelial cells would normally sit. Patterned matrix type VM lacks the endothelial-like tumor cells and is instead tumor cells that are enveloped by PAS+ matrix/

Mechanisms and pathways

Epithelial-mesenchymal transition (EMT)

EMT is thought to be one of the major drivers of VM in cancer. [10] This is a mechanism through which cancer cells lose their epithelial properties resulting in a loss of cell-cell adhesion and a transition towards mesenchymal-like properties . EMT plays a diverse and essential role in adhesion, motility and morphology of cells under both normal and pathological conditions. When cells undergo EMT, they lose their polarity, ability to adhere to neighboring cells, and the tight contacts as result of losing expression of epithelial cell markers such as E-cadherin. Cells that have taken on mesenchymal properties are non-adherent, and thus promote invasion into nearby tissues. This process occurs normally in wound healing as well as embryonic development. [11] This transition process has been identified to occur in cancer cells and drives metastasis and invasion into other tissues. [11] EMT is associated with increasing and maintaining the VM in tumors through several pathways such as Twist transcription factor, and TGFB. [12] [13] Importantly to VM, the process of EMT results in the loss of E-cadherin, and the promotion of VE-cadherin transcription and expression, a critical factor in development of vascular mimicry. [3] EMT is also implicated in promotion of stem-like properties in cancer cells. [14]

Cancer stem cell signaling (CSCs)

Cancer stem cells (CSCs) is a theory which explains the heterogeneity of tumors. This theory states that tumors arise from a small subpopulation of cancer cells which have the ability to self-renew as well as differentiate. [15] These CSCs give rise to unique cell subpopulations within the tumor through differentiation, which express unique cell characteristics that further drive tumor growth and metastasis. CSCs have been linked to driving VM formation predominantly through the secretion of VEGF, which is a well establish regulator or VE-cadherin expression. [10] Within the CSCs, several studies have identified specific subtypes of cancer stem cells which are linked to promoting VM formation, including stem cells expressing CD133+ in acute leukemia, [10] melanoma, [10] and Triple negative breast cancer (TNBC). [3]

Further, the Notch and Nodal signaling pathways, both a part of the transforming growth factor β (TGF-β) protein superfamily that functions to reserve undifferentiated stem cells during early development to allow for future development, have been positively associated with VM formation. [7] Under physiological conditions, Nodal is rarely expressed in adult tissues. This changes however in cancer cells, where Nodal is significantly upregulated in numerous aggressive cancers. [1] Nodal signaling occurs unchecked in cancers through binding of activin-like kinase receptors ALK and signals downstream transcription associated with increased cell plasticity, an essential factor in VM formation. Notch function remains active in adult tissues and is an essential signaling pathway in stem cell differentiation, including the formation of vasculature. Notch modulates Nodal activation during embryonic stages during the establishment of positionality of specific cells in the embryo. This process is reactivated in cancers such as melanoma and has been shown to drive cancers cells to possess increased plasticity as well as overall cancer aggressiveness. [1] Specifically, increased Notch-4 expression was found across multiple aggressive melanoma cell lines, and the inhibition of Notch4 activity in melanoma was sufficient to prevent VM formation. [7]

The transition of tumors from angiogenesis to vascular mimicry under hypoxic conditions Vascular mimicry and angiogenesis.png
The transition of tumors from angiogenesis to vascular mimicry under hypoxic conditions

Hypoxia and tumor microenvironment

The major signaling pathways involved in vasculogenic mimicry. Vascular signaling (purple), Stem cell signaling (blue), and Hypoxia (green) are the major pathways shown. Vasculogenic mimicry signaling pathways.jpg
The major signaling pathways involved in vasculogenic mimicry. Vascular signaling (purple), Stem cell signaling (blue), and Hypoxia (green) are the major pathways shown.

Another major mechanism through which vasculogenic mimicry is induced is hypoxia. Hypoxia is very common in tumors due to insufficient vasculature to provide oxygen for the rapidly proliferating tumor issues within what is known as the tumor microenvironment. [16] To circumvent this problem, tumors utilize a diverse set of pathways to promote various angiogenesis strategies as well as vasculogenic mimicry. Hypoxia inducible factors (HIFs) are transcription factors which are able to remain functional during hypoxic conditions, bind to hypoxia responsive elements (HRE) on the genome. Gene targets with HRE sites bind HIF and mediate gene transcription. [16] The location of HRE sites on the genome varies among cancer cell types, however hypoxia based signaling has been found to activate VM related genes including VE-cadherin, COX-2, Twist, Nodal, EphA2, VEGF-A, and VEGR-1. [7] Hypoxia can also promote VM through other signaling pathways such as NF-κB, which upregulates Twist expression, an inducer of VM. [6] It is important to note as well that many chemotherapeutic agents used in the treatment of cancers intentionally drive hypoxia in tumors as a treatment strategy by inhibiting tumor angiogenesis. While these treatments vary in efficacy depending on cancer type and patient, it has been shown that such therapies may actually drive tumors down more aggressive routes through the promotion of VM based tumor survival. [9]

Major observed signaling pathways significant to VM formation or maintenance

Major signalling pathways involved in VM
NameClassificationRole in VM
EphA2 Ephrin tyrosine kinaseHighly expressed in malignant tumors, EphA2 thought to bind PI3K and increase VE-cadherin expression. [10]
VEGFR-1 & 2 vascular endothelial growth factor receptorBoth receptors signaling pathways have been linked to VM. VEGFR-1 drives VM formation through activation of PI3K pathway in melanoma, while VEGFR-2 has been determined to drive VM in glioblastoma. [17]
Notch/Nodal TGF-β protein superfamilyImportant to maintaining CSCs population responsible for VM, as well as increasing MMP-2 expression through activation of VEGFR-1 pathway. [17]
MMPs Matrix metalloproteinasesHigh expression considered a requirement for VM formation, functions as a proteolytic enzyme thought to interact with laminin to promote VM blood vessel formation. [3] Further, MMPs have a major role in degradation and reshaping of the extracellular matrix (ECM) and basement membrane, a process critical to provide space for new VM to form. [6]
HIF1-A Hypoxia inducible factorExpressed under hypoxic conditions. Binds to HER sites in the genome, several activated genes have been linked to VM formation. [7]
twist transcription factorImportant roles in VM including downregulation of E-Cadherin, [6] upregulation of VE-cadherin expression, [10] and driving EMT in tumor cells, [17]
COX-2 isozymeEstablished as a driver of VM in breast cancer. [12] High expression of COX-2 was associated with increased cancer cell stemness, driving of VM through PI3K/AKT pathways, and modulating NOTCH and WNT expression. [12]
non-coding RNAs Ribonucleic acid (Not translated)Multiple ncRNAs have been linked to VM formation. miRNA-193b expression regulates VM by inhibition of DDAH1, resulting in suppression of VM. miR-93 increases VM formation in TNBC, overexpression of miR-93 has been found to increase vascularization of the tumor. [18]

Cancer

Vascular mimicry has been noted in numerous cancers has been consistently associated with poor patient survival outcomes. Meta-analysis of over 3000 patients across 15 different cancers found that the 5-year survival of patients with VM positive tumors was 31%, while patients with VM negative tumors had a significantly better 5-year survival rate of 56%. [9] Further, this difference in outcome is greater when looking at later stage disease. [19] VM is also a significant factor in the development of tumor resistance to Anti-angiogenic chemotherapeutic strategies. [9]

Breast cancer

Left: the relationship between breast cancer cell stemness, vasculogenic mimicry, and prognosis. Right: provides a diagram of factors involved in formation of vascular mimicry in breast cancer. Breast cancer image.webp
Left: the relationship between breast cancer cell stemness, vasculogenic mimicry, and prognosis. Right: provides a diagram of factors involved in formation of vascular mimicry in breast cancer.

New strategies to overcome treatment resistance in breast cancers have looked to target tumor angiogenesis and the problem posed by vascular mimicry in treatment resistance. [18] The presence of VM in breast cancer has been associated with the more challenging to treat triple negative breast cancer (TNBC). In addition, VM has been found to be more predominant in HER2 positive breast cancer, increase the presence of Epithelial-mesenchymal transition markers, and increased stem-like properties of breast cancer cells. [18] In a study of malignant breast cancers, 35% of reported cases of TNBC were found to be VM+, while only 17.8% of non-TNBC were found to be VM+. [9]

Glioblastoma

Glioblastoma is characterized by hypervascularization, thus therapeutic strategies targeting tumor angiogenesis are often used clinically. [9] Vascular mimicry provides tumors with a strategy to resist anti-angiogenic therapies, which have been shown to actually increase VM formation in patients due to the hypoxia these treatments induce. [9]

Glioblastoma Glioblastoma multiforme.jpg
Glioblastoma

Melanoma

Melanoma is an aggressive skin cancer which has been shown to utilize both angiogenesis and vascular mimicry to drive metastasis and also increase treatment resistance. [1] Metastatic melanoma has very poor survival outcomes with a median survival of 6 months. [19] VM has been identified to be present only in metastatic and very aggressive melanomas. Major biomarkers of VM in melanoma include: VEGF-A, HIF-1A, and Nodal.

Urological tumors

This includes cancers of the urinary system such as prostate cancer, and clear renal cell carcinomas. [10]

Prostate cancer

Prostate cancer is cancer of the male reproductive organ and represents one of the most common solid tumor cancers in men. Metastatic prostate cancer is currently treated using androgen deprivation therapy, however post 18 months, metastatic prostate cancer develops resistance to therapy. Prostate cancer with VM has been shown to occur in patients with overall more aggressive disease including higher Gleason scores, and more metastatic and lymph node formation. Biomarkers of VM in prostate cancer include: HIF1α, ZEB1, EphA2, and Sp1. [10]

Renal cell carcinoma

Renal cell carcinoma (RCC) is cancer of the kidneys with a 5-year survival rate for early detection of 92.6%. Only 75% of patients with RCC are detected during early stages leaving 25% diagnosed as more aggressive metastatic stages of tumor development. RCC relies upon angiogenesis to develop, thus the use of the anti-angiogenic drug Sunitinib have been used to great effect. The use of Sunitinib has promising effects on restricting tumor growth, however this effect is diminished after several months of treatment. Retrospective studies of RCC reveal that VM+ patients have significantly decreased rates of disease-free survival when compared to VM- patients. Further, VM+ patients had a significantly higher risk for cancer recurrence. Biomarkers of VM in RCC include: MMPs, Vimentin, low PRRX1 expression, CIP2A, and ncRNAs. [10]

Ovarian cancer

Ovarian cancer has a standard treatment strategy of carboplatin and paclitaxel that has a positive effect in 80% of patients. Despite a strong initial response to chemotherapy, the majority of these patients will see disease recurrence as well as develop resistance to available chemotherapeutic strategies. [20] The use of Anti-angiogenic compounds in ovarian cancer has met with limited success largely due to the availability of VM signaling pathways in aggressive tumors. Due to poor strategies for early detection, ovarian cancer is generally caught at later stages of disease and thus difficult to treat. VM is a major alternative vascularization pathway in ovarian cancer that is currently poorly understood in the ovarian cancer context. [17] Major markers of VM in ovarian cancers include: VE-cadherin, VEGF-A, CD147, uPA, Twist1, hypoxia related factors, and CSC markers such as ALDH and CD133. [20]

Therapeutics

While there are few drugs currently developed that specifically target vascular mimicry, there are compounds in development to inhibit VM, as well as numerous researched compounds which have shown effective VM inhibition in animal and cell culture models.

(* Indicates the compound has FDA approval, NOTE: Not approved for specific use in treatment of VM)

Known compounds which inhibit VM
NameDrug targetCancer type tested in
PARP inhibition [7] VE-cadherin Melanoma [3]
*Rapamycin [7] mTOR/VEGF Ovarian cancer, [3] Gliomas [10]
Isoxanthohumol [7] TGF-β signalling Breast Cancer [3]
Resveratrol [7] VEGF-R1 & R2 Melanoma [3]
Ginsenoside Rg3 [7] VE-cadherin, MMPs, EphA2 Pancreatic cancer [3]
Brucine [18] EMT transition, MMP-2 and MMP-9 inhibition Triple negative breast cancer [3]
R8 modified epirubicin-dihydroartemisinin liposomes VE-cadherin, TGF-β signalling, MMP-2, HIF-1 Non-small cell lung cancer [3]
Chrysin [10] SPHK/HIF-1α Prostate Cancer [10]
*Sorafenib [10] VEGFR-2 Metastatic renal cell carcinoma [10]
*Panobinostat [19] HDAC/hypoxia Multiple myeloma
*Copanlisib [19] PI3K Follicular lymphoma

See also

Related Research Articles

<span class="mw-page-title-main">Angiogenesis</span> Blood vessel formation, when new vessels emerge from existing vessels

Angiogenesis is the physiological process through which new blood vessels form from pre-existing vessels, formed in the earlier stage of vasculogenesis. Angiogenesis continues the growth of the vasculature mainly by processes of sprouting and splitting, but processes such as coalescent angiogenesis, vessel elongation and vessel cooption also play a role. Vasculogenesis is the embryonic formation of endothelial cells from mesoderm cell precursors, and from neovascularization, although discussions are not always precise. The first vessels in the developing embryo form through vasculogenesis, after which angiogenesis is responsible for most, if not all, blood vessel growth during development and in disease.

<span class="mw-page-title-main">Metastasis</span> Spread of a disease inside a body

Metastasis is a pathogenic agent's spread from an initial or primary site to a different or secondary site within the host's body; the term is typically used when referring to metastasis by a cancerous tumor. The newly pathological sites, then, are metastases (mets). It is generally distinguished from cancer invasion, which is the direct extension and penetration by cancer cells into neighboring tissues.

<span class="mw-page-title-main">Endothelium</span> Layer of cells that lining inner surface of blood vessels

The endothelium is a single layer of squamous endothelial cells that line the interior surface of blood vessels and lymphatic vessels. The endothelium forms an interface between circulating blood or lymph in the lumen and the rest of the vessel wall. Endothelial cells form the barrier between vessels and tissue and control the flow of substances and fluid into and out of a tissue.

<span class="mw-page-title-main">Pericyte</span> Cells associated with capillary linings

Pericytes are multi-functional mural cells of the microcirculation that wrap around the endothelial cells that line the capillaries throughout the body. Pericytes are embedded in the basement membrane of blood capillaries, where they communicate with endothelial cells by means of both direct physical contact and paracrine signaling. The morphology, distribution, density and molecular fingerprints of pericytes vary between organs and vascular beds. Pericytes help to maintain homeostatic and hemostatic functions in the brain, one of the organs with higher pericyte coverage, and also sustain the blood–brain barrier. These cells are also a key component of the neurovascular unit, which includes endothelial cells, astrocytes, and neurons. Pericytes have been postulated to regulate capillary blood flow and the clearance and phagocytosis of cellular debris in vitro. Pericytes stabilize and monitor the maturation of endothelial cells by means of direct communication between the cell membrane as well as through paracrine signaling. A deficiency of pericytes in the central nervous system can cause increased permeability of the blood–brain barrier.

Vascular endothelial growth factor, originally known as vascular permeability factor (VPF), is a signal protein produced by many cells that stimulates the formation of blood vessels. To be specific, VEGF is a sub-family of growth factors, the platelet-derived growth factor family of cystine-knot growth factors. They are important signaling proteins involved in both vasculogenesis and angiogenesis.

An angiogenesis inhibitor is a substance that inhibits the growth of new blood vessels (angiogenesis). Some angiogenesis inhibitors are endogenous and a normal part of the body's control and others are obtained exogenously through pharmaceutical drugs or diet.

<span class="mw-page-title-main">Catenin</span>

Catenins are a family of proteins found in complexes with cadherin cell adhesion molecules of animal cells. The first two catenins that were identified became known as α-catenin and β-catenin. α-Catenin can bind to β-catenin and can also bind filamentous actin (F-actin). β-Catenin binds directly to the cytoplasmic tail of classical cadherins. Additional catenins such as γ-catenin and δ-catenin have been identified. The name "catenin" was originally selected because it was suspected that catenins might link cadherins to the cytoskeleton.

Vasculogenesis is the process of blood vessel formation, occurring by a de novo production of endothelial cells. It is sometimes paired with angiogenesis, as the first stage of the formation of the vascular network, closely followed by angiogenesis.

<span class="mw-page-title-main">Cancer stem cell</span> Cancer cells with features of normal cells

Cancer stem cells (CSCs) are cancer cells that possess characteristics associated with normal stem cells, specifically the ability to give rise to all cell types found in a particular cancer sample. CSCs are therefore tumorigenic (tumor-forming), perhaps in contrast to other non-tumorigenic cancer cells. CSCs may generate tumors through the stem cell processes of self-renewal and differentiation into multiple cell types. Such cells are hypothesized to persist in tumors as a distinct population and cause relapse and metastasis by giving rise to new tumors. Therefore, development of specific therapies targeted at CSCs holds hope for improvement of survival and quality of life of cancer patients, especially for patients with metastatic disease.

The epithelial–mesenchymal transition (EMT) is a process by which epithelial cells lose their cell polarity and cell–cell adhesion, and gain migratory and invasive properties to become mesenchymal stem cells; these are multipotent stromal cells that can differentiate into a variety of cell types. EMT is essential for numerous developmental processes including mesoderm formation and neural tube formation. EMT has also been shown to occur in wound healing, in organ fibrosis and in the initiation of metastasis in cancer progression.

<span class="mw-page-title-main">Angiopoietin</span> Protein family

Angiopoietin is part of a family of vascular growth factors that play a role in embryonic and postnatal angiogenesis. Angiopoietin signaling most directly corresponds with angiogenesis, the process by which new arteries and veins form from preexisting blood vessels. Angiogenesis proceeds through sprouting, endothelial cell migration, proliferation, and vessel destabilization and stabilization. They are responsible for assembling and disassembling the endothelial lining of blood vessels. Angiopoietin cytokines are involved with controlling microvascular permeability, vasodilation, and vasoconstriction by signaling smooth muscle cells surrounding vessels. There are now four identified angiopoietins: ANGPT1, ANGPT2, ANGPTL3, ANGPT4.

<span class="mw-page-title-main">CD146</span> Protein-coding gene in the species Homo sapiens

CD146 also known as the melanoma cell adhesion molecule (MCAM) or cell surface glycoprotein MUC18, is a 113kDa cell adhesion molecule currently used as a marker for endothelial cell lineage. In humans, the CD146 protein is encoded by the MCAM gene.

Stem-cell niche refers to a microenvironment, within the specific anatomic location where stem cells are found, which interacts with stem cells to regulate cell fate. The word 'niche' can be in reference to the in vivo or in vitro stem-cell microenvironment. During embryonic development, various niche factors act on embryonic stem cells to alter gene expression, and induce their proliferation or differentiation for the development of the fetus. Within the human body, stem-cell niches maintain adult stem cells in a quiescent state, but after tissue injury, the surrounding micro-environment actively signals to stem cells to promote either self-renewal or differentiation to form new tissues. Several factors are important to regulate stem-cell characteristics within the niche: cell–cell interactions between stem cells, as well as interactions between stem cells and neighbouring differentiated cells, interactions between stem cells and adhesion molecules, extracellular matrix components, the oxygen tension, growth factors, cytokines, and the physicochemical nature of the environment including the pH, ionic strength and metabolites, like ATP, are also important. The stem cells and niche may induce each other during development and reciprocally signal to maintain each other during adulthood.

Endothelial progenitor cell is a term that has been applied to multiple different cell types that play roles in the regeneration of the endothelial lining of blood vessels. Outgrowth endothelial cells are an EPC subtype committed to endothelial cell formation. Despite the history and controversy, the EPC in all its forms remains a promising target of regenerative medicine research.

<span class="mw-page-title-main">VE-cadherin</span> Protein-coding gene in the species Homo sapiens

Cadherin-5, or VE-cadherin, also known as CD144, is a type of cadherin. It is encoded by the human gene CDH5.

<span class="mw-page-title-main">EGFL7</span> Protein-coding gene in the species Homo sapiens

EGF-like domain-containing protein 7 is a protein that in humans is encoded by the EGFL7 gene. Intron 7 of EGFL7 hosts the miR-126 microRNA gene.

<span class="mw-page-title-main">ADH-1</span> Chemical compound

ADH-1 is a small, cyclic pentapeptide vascular-targeting drug. It was developed by Adherex Technologies.

<span class="mw-page-title-main">Tumor microenvironment</span> Surroundings of tumors including nearby cells and blood vessels

The tumor microenvironment is a complex ecosystem surrounding a tumor, composed of cancer cells, stromal tissue and the extracellular matrix. Mutual interaction between cancer cells and the different components of the tumor microenvironment support its growth and invasion in healthy tissues which correlates with tumor resistance to current treatments and poor prognosis. The tumor microenvironment is in constant change because of the tumor's ability to influence the microenvironment by releasing extracellular signals, promoting tumor angiogenesis and inducing peripheral immune tolerance, while the immune cells in the microenvironment can affect the growth and evolution of cancerous cells.

Migration inducting gene 7 is a gene that corresponds to a cysteine-rich protein localized to the cell membrane and cytoplasm. It is the first-in-class of novel proteins translated from what are thought to be long Non-coding RNAs.

<span class="mw-page-title-main">Tumor-associated endothelial cell</span>

Tumor-associated endothelial cells or tumor endothelial cells (TECs) refers to cells lining the tumor-associated blood vessels that control the passage of nutrients into surrounding tumor tissue. Across different cancer types, tumor-associated blood vessels have been discovered to differ significantly from normal blood vessels in morphology, gene expression, and functionality in ways that promote cancer progression. There has been notable interest in developing cancer therapeutics that capitalize on these abnormalities of the tumor-associated endothelium to destroy tumors.

References

  1. 1 2 3 4 5 Hendrix, Mary J.C.; Seftor, Elisabeth A.; Seftor, Richard E.B.; Chao, Jun-Tzu; Chien, Du-Shieng; Chu, Yi-Wen (March 2016). "Tumor cell vascular mimicry: Novel targeting opportunity in melanoma". Pharmacology & Therapeutics. 159: 83–92. doi:10.1016/j.pharmthera.2016.01.006. PMC   4779708 . PMID   26808163.
  2. 1 2 3 4 5 6 7 Wechman, Stephen L.; Emdad, Luni; Sarkar, Devanand; Das, Swadesh K.; Fisher, Paul B. (2020), "Vascular mimicry: Triggers, molecular interactions and in vivo models", Advances in Cancer Research, Elsevier, 148: 27–67, doi:10.1016/bs.acr.2020.06.001, ISBN   978-0-12-820327-9, PMC   7594199 , PMID   32723566
  3. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 Luo, Qingxi; Wang, Jun; Zhao, Wenyuan; Peng, Zhenzi; Liu, Xianyu; Li, Bin; Zhang, Heng; Shan, Bin; Zhang, Chunfang; Duan, Chaojun (December 2020). "Vasculogenic mimicry in carcinogenesis and clinical applications". Journal of Hematology & Oncology. 13 (1): 19. doi: 10.1186/s13045-020-00858-6 . ISSN   1756-8722. PMC   7071697 . PMID   32169087.
  4. Hanahan, Douglas (2022-01-01). "Hallmarks of Cancer: New Dimensions". Cancer Discovery. 12 (1): 31–46. doi: 10.1158/2159-8290.CD-21-1059 . ISSN   2159-8274. PMID   35022204.
  5. Pezzella, Francesco; Ribatti, Domenico (December 2022). "Vascular co‐option and vasculogenic mimicry mediate resistance to antiangiogenic strategies". Cancer Reports. 5 (12): e1318. doi:10.1002/cnr2.1318. ISSN   2573-8348. PMC   9780428 . PMID   33295149.
  6. 1 2 3 4 5 Wei, Xiaoxu; Chen, Yunhua; Jiang, Xianjie; Peng, Miao; Liu, Yiduo; Mo, Yongzhen; Ren, Daixi; Hua, Yuze; Yu, Boyao; Zhou, Yujuan; Liao, Qianjin; Wang, Hui; Xiang, Bo; Zhou, Ming; Li, Xiaoling (2021-01-04). "Mechanisms of vasculogenic mimicry in hypoxic tumor microenvironments". Molecular Cancer. 20 (1): 7. doi: 10.1186/s12943-020-01288-1 . ISSN   1476-4598. PMC   7784348 . PMID   33397409.
  7. 1 2 3 4 5 6 7 8 9 10 11 Delgado-Bellido, Daniel; Serrano-Saenz, Santiago; Fernández-Cortés, Mónica; Oliver, F. Javier (December 2017). "Vasculogenic mimicry signaling revisited: focus on non-vascular VE-cadherin". Molecular Cancer. 16 (1): 65. doi: 10.1186/s12943-017-0631-x . ISSN   1476-4598. PMC   5359927 . PMID   28320399.
  8. Vestweber, Dietmar (February 2008). "VE-Cadherin: The Major Endothelial Adhesion Molecule Controlling Cellular Junctions and Blood Vessel Formation". Arteriosclerosis, Thrombosis, and Vascular Biology. 28 (2): 223–232. doi: 10.1161/ATVBAHA.107.158014 . ISSN   1079-5642. PMID   18162609.
  9. 1 2 3 4 5 6 7 Angara, Kartik; Borin, Thaiz F.; Arbab, Ali S. (August 2017). "Vascular Mimicry: A Novel Neovascularization Mechanism Driving Anti-Angiogenic Therapy (AAT) Resistance in Glioblastoma". Translational Oncology. 10 (4): 650–660. doi: 10.1016/j.tranon.2017.04.007 . PMC   5496207 . PMID   28668763.
  10. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 Lin, Xinyu; Long, Sheng; Yan, Congcong; Zou, Xiaofeng; Zhang, Guoxi; Zou, Junrong; Wu, Gengqing (2023-09-21). "Therapeutic potential of vasculogenic mimicry in urological tumors". Frontiers in Oncology. 13. doi: 10.3389/fonc.2023.1202656 . ISSN   2234-943X. PMC   10551447 . PMID   37810976.
  11. 1 2 Sadrkhanloo, Mehrdokht; Entezari, Maliheh; Orouei, Sima; Ghollasi, Marzieh; fathi, Nikoo; Rezaei, Shamin; Hejazi, Elahe Sadat; Kakavand, Amirabbas; Saebfar, Hamidreza; Hashemi, Mehrdad; Goharrizi, Mohammad Ali Sheikh Beig; Salimimoghadam, Shokooh; Rashidi, Mohsen; Taheriazam, Afshin; Samarghandian, Saeed (August 2022). "STAT3-EMT axis in tumors: Modulation of cancer metastasis, stemness and therapy response". Pharmacological Research. 182: 106311. doi:10.1016/j.phrs.2022.106311. PMID   35716914. S2CID   249727974.
  12. 1 2 3 Morales-Guadarrama, Gabriela; García-Becerra, Rocío; Méndez-Pérez, Edgar Armando; García-Quiroz, Janice; Avila, Euclides; Díaz, Lorenza (2021-07-12). "Vasculogenic Mimicry in Breast Cancer: Clinical Relevance and Drivers". Cells. 10 (7): 1758. doi: 10.3390/cells10071758 . ISSN   2073-4409. PMC   8304745 . PMID   34359928.
  13. Ma, Xiao; Geng, Ziang; Wang, Siqi; Yu, Zhongxue; Liu, Tiancong; Guan, Shu; Du, Shaonan; Zhu, Chen (September 2023). "The driving mechanism and targeting value of mimicry between vascular endothelial cells and tumor cells in tumor progression". Biomedicine & Pharmacotherapy. 165: 115029. doi: 10.1016/j.biopha.2023.115029 .
  14. Kotiyal, Srishti; Bhattacharya, Susinjan (2015). "Epithelial Mesenchymal Transition and Vascular Mimicry in Breast Cancer Stem Cells". Critical Reviews in Eukaryotic Gene Expression. 25 (3): 269–280. doi:10.1615/CritRevEukaryotGeneExpr.2015014042. ISSN   1045-4403. PMID   26558950.
  15. Li, Yanyan; Wicha, Max S.; Schwartz, Steven J.; Sun, Duxin (September 2011). "Implications of cancer stem cell theory for cancer chemoprevention by natural dietary compounds". The Journal of Nutritional Biochemistry. 22 (9): 799–806. doi:10.1016/j.jnutbio.2010.11.001. PMC   3248810 . PMID   21295962.
  16. 1 2 Jing, Xinming; Yang, Fengming; Shao, Chuchu; Wei, Ke; Xie, Mengyan; Shen, Hua; Shu, Yongqian (December 2019). "Role of hypoxia in cancer therapy by regulating the tumor microenvironment". Molecular Cancer. 18 (1): 157. doi: 10.1186/s12943-019-1089-9 . ISSN   1476-4598. PMC   6844052 . PMID   31711497.
  17. 1 2 3 4 Ayala-Domínguez, Lízbeth; Olmedo-Nieva, Leslie; Muñoz-Bello, J. Omar; Contreras-Paredes, Adriana; Manzo-Merino, Joaquín; Martínez-Ramírez, Imelda; Lizano, Marcela (2019-09-27). "Mechanisms of Vasculogenic Mimicry in Ovarian Cancer". Frontiers in Oncology. 9: 998. doi: 10.3389/fonc.2019.00998 . ISSN   2234-943X. PMC   6776917 . PMID   31612116.
  18. 1 2 3 4 Chavoshi, Hadi; Poormolaie, Neda; Vahedian, Vahid; Kazemzadeh, Hamid; Mir, Amirabbas; Nejabati, Hamid Reza; Behroozi, Javad; Isazadeh, Alireza; Hajezimian, Saba; Nouri, Mohammad; Maroufi, Nazila Fathi (June 2022). "Vascular mimicry: A potential therapeutic target in breast cancer". Pathology - Research and Practice. 234: 153922. doi:10.1016/j.prp.2022.153922. PMID   35500501.
  19. 1 2 3 4 Mabeta, Peace (July 2020). "Paradigms of vascularization in melanoma: Clinical significance and potential for therapeutic targeting". Biomedicine & Pharmacotherapy. 127: 110135. doi: 10.1016/j.biopha.2020.110135 . hdl: 2263/76222 . PMID   32334374.
  20. 1 2 Lim, Dansaem; Do, Yeojin; Kwon, Byung Su; Chang, Woochul; Lee, Myeong-Sok; Kim, Jongmin; Cho, Jin Gu (2020-06-30). "Angiogenesis and vasculogenic mimicry as therapeutic targets in ovarian cancer". BMB Reports. 53 (6): 291–298. doi:10.5483/BMBRep.2020.53.6.060. ISSN   1976-670X. PMC   7330806 . PMID   32438972.

Further reading