Minireviews Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Aug 15, 2022; 14(8): 1469-1477
Published online Aug 15, 2022. doi: 10.4251/wjgo.v14.i8.1469
Angiogenesis in gastrointestinal stromal tumors: From bench to bedside
Stavros P Papadakos, First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens 10679, Greece
Christos Tsagkaris, Faculty of Medicine, University of Crete, Heraklion 71003, Greece
Marios Papadakis, University Hospital Witten-Herdecke, University of Witten-Herdecke, Wuppertal 42283, Germany
Andreas S Papazoglou, Dimitrios V Moysidis, First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki 54636, Greece
Constantinos G Zografos, First Department of Surgery, Athens Medical School, National and Kapodistrian University of Athens, Laikon General Hospital, Athens 11527, Greece
Stamatios Theocharis, First Department of Pathology, Medical School, University of Athens, Athens 11527, Greece
ORCID number: Stavros P Papadakos (0000-0003-1583-1125); Christos Tsagkaris (0000-0002-4250-574X); Marios Papadakis (0000-0002-9020-874X); Andreas S Papazoglou (0000-0003-4981-8121); Dimitrios V Moysidis (0000-0001-9083-0267); Constantinos G Zografos (0000-0002-8203-6407).
Author contributions: Papadakos SP and Tsagkaris C contributed equally; Papadakos SP and Tsagkaris C contributed to the onceptualization and study design; Papadakos SP wrote the first draft; Moysidis DV, Papazoglou AS, Papadakis M and Zografos CG wrote the second draft; Tsagkaris C, Papadakis M and Theocharis S contributed to the critical revision; Papadakis M and Theocharis S contributed to the supervision
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Christos Tsagkaris, MD, Academic Fellow, Faculty of Medicine, University of Crete, Voutes Camp, Andrea Kalokairinou, Heraklion 71003, Greece. chriss20x@gmail.com
Received: March 20, 2022
Peer-review started: March 20, 2022
First decision: April 25, 2022
Revised: May 15, 2022
Accepted: July 18, 2022
Article in press: July 18, 2022
Published online: August 15, 2022
Processing time: 142 Days and 14.1 Hours

Abstract

Gastrointestinal stromal tumors (GISTs) are rare neoplasms with an estimated incidence from 0.78 to 1-1.5 patients per 100000. They most commonly occur in the elderly during the eighth decade of life affecting predominantly the stomach, but also the small intestine, the omentum, mesentery and rectosigmoid. The available treatments for GIST are associated with a significant rate of recurrent disease and adverse events. Thorough understanding of GIST’s pathophysiology and translation of this knowledge into novel regimens or drug repurposing is essential to counter this challenge. The present review summarizes the existing evidence about the role of angiogenesis in GIST’s development and progression and discusses its clinical underpinnings.

Key Words: Gastrointestinal stromal tumor, Cancer, Oncology, Angiogenesis, Gastrointestinal oncology, Stromal tumors

Core Tip: Thorough understanding of gastrointestinal stromal tumors (GISTs)’s pathophysiology and translation of this knowledge into novel regimens or drug repurposing is essential to counter this challenge. The present review summarizes the existing evidence about the role of angiogenesis in GIST’s development and progression and discusses its clinical underpinnings.



INTRODUCTION

Gastrointestinal stromal tumors (GISTs) are rare neoplasms with an estimated incidence from 0.78 patients to 2 patients per 100000[1,2]. Their highest prevalence is noted during the eight decade of age, when they affect up to 3.06 individuals per 100000[3]. GIST typically present as subepithelial masses mainly in stomach (60%) and small intestine (20%-30%) with omentum, mesentery and rectosigmoid areas being less-frequently involved areas[4]. According to their primary location, GISTs could clinically present as gastrointestinal hemorrhage, anemia, dyspepsia or vomiting when the upper gastrointestinal tract is involved and as bowel obstruction, frequent urination or diarrhea in implication of the lower gastrointestinal tract[5]. The metastatic disease principally concerns the liver, omentum, and peritoneum presenting as abdominal pain or constipation while extra-intestinal metastases to lymph nodes (LN) and lungs are infrequent[4]. The pathological diagnosis relies on the tissue’s morphological and molecular characteristics. Based on their morphology, GISTs are classified into three groups according to the predominant cell type: Spindle cell type (70%), epithelioid cell type (20%) and a mixed type (10%). CD117 comprises a transmembrane protein which is the end-product of the c-kit expression[6]. The KIT (CD117) positivity in immunohistochemistry (IHC) in tissues which are morphologically consistent with GIST establishes the diagnosis in the 95% of the cases. In KIT negative cases, the discovered on GIST 1 (DOG1) and CD34, which is an antigen of the myeloid progenitor cells, staining or the documentation of KIT or platelet-derived growth factor receptor (PDGFRA) gene mutations are sufficient to institute a diagnosis. Seldom in pediatric and young populations, GIST formation arises in the context of succinate dehydrogenase-deficiency in conjunction with paragangliomas and pulmonary chondromas[7,8].

The pharmacologic targeting of angiogenesis in cancer therapeutics was introduced as a groundbreaking approach. Nevertheless, the anti-vascular endothelial growth factor (VEGF) targeting alone or in conjunction with chemotherapy displayed only modest benefit in overall survival in solid tumors indicating the complexity of the mechanisms that regulate tumor angiogenesis[9]. Τhus, the necessity arose to develop a broad spectrum of anti-angiogenic treatments such as: Direct VEGFR2 antagonists (ramucirumab), VEGF-Traps (aflibercept), several receptor tyrosine kinases inhibitors targeting the PDGF-R, CD117 (c-KIT), fibroblast growth factor receptors (FGFR), epidermal growth factor receptor, RET, RAF kinases and the repurposing of drugs like the mammalian target of rapamycin inhibitors and lenalidomide[9,10]. In fact, anti-angiogenetic therapy has gained ground in the management of advanced, unresectable disease. Imatinib, an abl, c-KIT and PDGF-R tyrosine kinase inhibitor (TKI), constitutes the empiric treatment when the mutational status of the disease remains unknown and the first line of treatment in KIT and PDGFRA positive metastatic, inoperable GISTs. The D842V mutation in PDGFRA comprises a therapeutic exception and is being treated with avapritinib while KIT and PDGFRA wild type tumors are treated with sunitinib or regorafinib[11].

All the above mentioned drugs achieve, at least partially, their cytotoxicity disrupting signaling pathways which are implicated in angiogenesis, as it would be further analyzed below. This suggests that angiogenesis might be of paramount importance for the carcinogenesis process in GISTs and an attempt to summarize all the pre-clinical and clinical data would be of great value.

THE ROLE OF ANGIOGENESIS IN GIST’S DEVELOPMENT AND PROGRESSION
The molecular mechanisms of angiogenesis in GISTs–preclinical data

The regulation of angiogenesis is necessary for cancer cells initially to cope with their increased metabolic needs and in the process to promote their metastatic potential. Its significance was firstly recognized by Folkman[12], which stated that the magnified rate of neovascularization compared with wound healing and inflammation as a result of an interplay between tumor cells and endothelial cells was a prerequisite in order to achieve tumor growth[12]. Presently, it is widely known that the angiogenic process is being coordinated by the balance of several angiogenesis inducers and inhibitors in tumor’s microenvironment. The dominance of the pro-angiogenetic factors, a phenomenon called “angiogenic switch”[13], triggers the angiogenesis and could result either as result of the consequent hypoxia from the increased tumor proliferation or by the immune cell infiltration[14]. The primary induction phase with the undeveloped vessels paves the way for the remodeling phase when the blood vessel generation is sustained[15]. Several models of angiogenesis have been described explaining partially the poor outcomes of the selective angiogenic blockage as certain tumors can utilize alternative modes of angiogenesis[14]. Their analytical presentation has been done elsewhere[14,16,17] and goes beyond the scope of this review but a brief presentation in Table 1 would be helpful.

Table 1 The basic mechanisms of angiogenesis.
Angiogenetic mechanism
Function
Implicated signaling/ pathways
Sprouting angiogenesisVessel formation from a parental one as a sprout outgrowthVEGF, Dll4/notch pathways and neuropilins
Intussusceptive AngiogenesisSplitting of a parental vessel into two newly formedVEGF, PDGF pathways and erythropoietin
Vasculogenesis/Endothelial progenitor cellsVessel formation from endothelial progenitor cells differentiating into mature endothelial cellsVEGF pathway, chemokines
Vasculogenic mimicryVessel-like formations without endothelial cellsHGFR
Trans-differentiation of CSCsCSC give rise to endothelial cellsTie-2, TGF-, CXCL12/CXCR4

Xenograft studies in mice constitute an invaluable source of evidence about the angiogenetic mechanisms in GISTs. Our fundamental conceptualization about the orchestration of the angiogenetic process descended from Giner et al[18]. They utilized an intensely CD117, DOG1 and CD34-positive GIST with continual Ki-67 expression in about 15% of the tumor’s mass. The neovascularization experiments demonstrated the propagation of the induction phase during the first 96 h after implantation which proceeded by the remodeling phase. The induction phase was guided by the VEGF, VEGFC, PDGFA, PDGFB gene expression in conformity with their receptors. In more detail, the IHC data indicate that the VEGF ligand and the VEGFR2, VEGFR3 were positive at day 4 after the xenografting. As regards the chemokine expression, CXCL9, CXCL10, GRO and their receptors CXCR3, CXCR2 were stained in tumor cells and stroma soon after the implantation with a slight staining predominance of the chemokine receptors. These effects are possibly orchestrated by hypoxia-inducible factor (HIF)1α and the CXCL12/CXCR4 axis, which are constantly expressed[18].

The angiogenetic process in GIST has been further delineated and several regulatory molecules have been identified. CCL2 represents a chemokine expressed by the tumor cells to attract CCR2-expressing endothelial progenitor cells from the circulation as documented in HER-2/neu-driven breast cancer[19]. On the other hand, the VEGF-induced nuclear factor kappa B (NF-kB) upregulation is frequently utilized to attract inflammatory cell into tumor to stimulate the angiogenesis[20]. The bromodomain and extraterminal domain family mediates immunity regulating several signaling pathways[21]. In GISTs, the BRD4 upregulation enhanced the migratory and invasion processes regulating angiogenesis through the NF-kB/CCL2 signaling pathway. The BRD4-expressing cells attract tumor-associated macrophages via the expression of CCL2 potentiating the tumor’s microvessel density and secrete various pro-angiogenic molecules such as VEGFA, LOX and MMP9[22,23]. Towards the same direction, mutations of the protein phosphatase 2, regulatory subunit A, alpha (PPP2R1A) affect the carcinogenesis process[24,25]. In GISTs, mutations in PPP2R1A gene are found in nearly 20% of the cases and correlate with a more aggressive tumor phenotype. They result in increased growth rate via enhancing phosphorylation of c-kit, Akt1/2, ERK1/2 and WNK1. The latter seems to mediate the regulation of the angiogenetic process[26,27]. A further analysis of the specific mechanisms would be of great value and it should be applied.

Furthermore, while the contribution of epigenetic mechanisms in the GIST progression is well established, its impact in the angiogenetic mechanisms could be further delineated. Several gaps in our understanding that remain unaddressed by the subdivisions according to the driver gene mutation status could be further elucidated by the tumor’s epigenetic landscape. The alterations in the tumor’s methylation profile are associated with a more aggressive phenotype[28] and the methylation status of the CD133 could reshape the management of the disease and it would be presented below in more depth[29]. The KDM4 family members (KDM4A-D) reshaping the structure of chromatin are implicated in the pathogenesis of a wide variety of cancers[30]. In GIST, the upregulation of KDM4D potentiates the angiogenesis in vivo, as indicated by the overexpression of CD31 in IHC. These effects are mediated by the HIF1β/VEGFA pathway in the presence of demethylation in the promoters of the H3K9me3 and H3K36me3 genes[31].

Finally, it is worth mentioning that several multi-TKIs exert their anti-tumor efficacy at least partially by the inhibition of angiogenesis. Cabozantinib exerts it’s activity inhibiting the receptor tyrosine kinases MET, VEGFR2, Flt-3, c-Kit and RET[32,33] while sorafenib inhibits the signaling of VEGFR-1, VEGFR-2, VEGFR-3, PDGFR-β, Flt-3, c-Kit and the Raf serine/threonine kinases[34]. Both diminish the tumor’s microvascular density as evidenced by CD31 IHC[35,36].

The association between imaging data and angiogenesis in GISTs

There have been several classification systems to stratify the malignant potential of GISTs such as: The National Institutes of Healthv consensus criteria (Fletcher's criteria), the Armed Forces Institute of Pathology criteria (Miettinen's criteria) or the International Union against Cancer TNM classification. Their main drawback constitute the inability to validate the tumor’s aggressiveness without surgical resection and detailed pathologic examination of the entire tumor to estimate the mitotic count[37,38] . Although taking into consideration the current therapeutic trends, the management of the advanced, unresectable disease is unequivocal, there are margins for improvement in the management of primary localized disease, especially in small-sized tumors. It could not be emphasized enough that even small GISTs could develop malignant behavior. Τhereat, it could provide us a wealth of valuable predictive and prognostic information an attempt to incorporate imaging data about the vascularization of the tumor such as the vessels’ irregularity or the blood perfusion[39].

The above mentioned gap was attempted to be filled by a landmark study by Iannicelli et al[40], the computed tomography (CT) constitutes the fundamental imaging modality in patients presenting with the clinical manifestations of GIST. Reviewing past literature, several studies have documented that aim to associate certain imaging features with pathologic parameters[41,42]. Iannicelli et al[40] presented that GISTs with irregular margins tended to have superior mitotic rate than tumor with regular margins. Furthermore, a heterogonous pattern of contrast enhancement (CE), the angiogenesis and necrosis correlated with an increased tumor size and a more aggressive clinical behavior. It worth mentioning that the intensity of CE although it represents a novel mark of biologic activity, was not correlated with neither the number of mitoses nor the tumor’s risk stratification[40]. The above comprise an indirect link between tumor’s margins and mitotic rate, which is essential in order to stratify before surgery the clinical behavior of the tumor and highlight the importance of angiogenesis in disease progression. The latter could also be deduced by dynamic positron emission tomography analysis. Strauss et al[43] reported an association between the rate in which the F-18-fluorodeoxyglucose diffused into the tumor with the expression of VEGF-A[43]. The main limitation of CT comprises it’s low sensitivity as regards the imaging of vascularity in small sized tumors[39]. This divergence could be addressed by the endoscopic ultrasound (EUS) technology.

The utilization of EUS has emerged during the last decades. Its ability to evade the intervention of the abdominal fat and gastrointestinal gas in conjunction with the capability of FNA biopsy render it a useful tool towards a more personalized approach in the management of GIST. In EUS the GISTs are visualized as hypoechoic masses arising from the muscularis propria or the muscularis mucosae. The presence of irregular margins, cystic areas or malignant LN herald bad prognosis[44]. The usage of contrast media enhances further the diagnostic capacity of the EUS and promotes the tumor’s vascularity as a valuable prognostic biomarker. The role of CE-EUS in the management has been extensively reviewed elsewhere[45] and we intend to delineate the fundamentals. Sakamoto et al[39] classified the tumor’s vascularity into two subgroups according to the pattern of perfusion (homogenous or heterogeneous) and vessel appearance (regular or irregular). The homogenous perfusion with regular vessels were considered as signs of mild clinical behavior. Furthermore, they compared the diagnostic sensitivity of contrast-enhanced harmonic US, Power-Doppler EUS and CE-multidetector CT to visualize tumor vessels. In GISTs larger than 3 cm their sensitivities were 100%, 75% and 42% respectively. The differences became more emphatic in tumors less than 3 cm: 100%, 25% and 0%, respectively. It was noteworthy that every malignant lesion less than 3 cm in the cohort had been detected by the CEH-EUS before surgery[39]. The above indicate that CE-US comprises a powerful tool to visualize vascularity. Taking a step further, Yamashita et al[46] demonstrated an association between the imaging findings on CE-US and the pathologic risk stratification. In more depth, the large vessels lacked elastic tissue, indicating that neovascularization constitutes the underlying pathogenetic mechanism, and expressed VEGF[46].

It becomes evident that the imaging findings of vascularity might be sensational and practice changing in a subset of patients with small sized tumors (< 3 cm) and aggressive phenotype. A more substantial body of evidence should be collected in order to address properly those dilemmas.

Angiogenesis mediators as biomarkers in GIST–clinical data

The development of biomarkers comprises an essential step towards the individualization of medical practice. Liquid biopsy provides a cutting-edge, non-invasive technology to access predictive information to guide the therapeutic management in a wide variety of diseases[47-51]. It’s application in GIST treatment has been started to emerge[52,53]. Reviewing subsequent and more recent literature, an extensive number of studies has been found associating molecules implicated in angiogenesis with pathologic features. Although there are several limitations in the above mentioned research, the importance of angiogenesis in GIST’s malignant progression is delineated. In Table 2 are summarized the most significant data.

Table 2 A brief presentation of several angiogenetic molecules in disease progression.
Ref.
Sample size
Molecule/methods
Outcomes
Zhao et al[59]124 patients-62, 50% in stomach, 22.6% in small intestineHIF-1α/IHCAssociation with disease-free survival (P = 0.03)
VEGF/IHCAssociation with disease-free survival (P = 0.002)
MVD/IHCAssociation with disease-free survival (P < 0.001)
Kang et al[60]213 patients-63% in stomach, 25.3% in small intestine634G/CSuperior OS than 634 G/G (P = 0.054)
Superior RFS than 634 G/G (P = 0.082)
Mu et al[22]20 patientsBRD4/mRNA, IHCIncreased BRD4 expression compared with normal tissue
BRD4/IHCAssociated with poor OS (P < 0.01)
Associated with poor DFS (P < 0.01)
Toda-Ishii et al[61]94 patients–mean follow-up period 65 moPPP2R1A mutations/PCRLower OS (P < 0.05)
Lower DFS (P < 0.05)
Liu et al[62]52 patients–27 malignant cases–11 borderline–14 benignMMP-9, COX-2, VEGF/IHCEnhance metastasis (P = 0.014, P = 0.010, P = 0.032 respectively)
Higher mitotic count (P = 0.021, P = 0.027, P = 0.009 respectively)
Higher incidence of central necrosis (P < 0.01)
Takahashi et al[63]53 patients: 21 cases < 30 mm-9 cases with liver metastasisVEGF/IHCAssociation with liver metastasis (P < 0.01)
VEGF/IHCPoor 10-yr OS (P < 0.05)
MVD/IHCAssociation with liver metastasis (P < 0.05)
Verboom et al[64]227 patients-36 SNPs-18 genes, median PFS 39 mo–median OS 86.5 mors1570360 polymorphism in VEGFA geneAssociation with poorer PFS (P = 0.015)
rs1870377 polymorphism in VEGFR2 geneAssociation with lower PFS (P = 0.037)
Chen et al[65]62 patients: 31 high risk–31 low riskHIF-1α/IHCAssociation with high risk disease (P < 0.0001)
Association with GIST recurrence or metastasis (P = 0.009)
Basilio-de-Oliveira and Pannain[66]54 patientsVEGF/IHCAssociation with survival (P < 0.001)
CD105/IHCAssociation with prognosis (P < 0.001)
Imamura et al[67]95 patients: 64 cases in stomach–31 in small intestineMVD/IHCAssociation with tumor grade (P = 0.036)
Association with VEGF expression (P < 0.0001)
Association with DFS after surgery (P = 0.0028)
Wang et al[68]68 patients: 20 low risk cases–48 high risk casesSoluble VEGFAssociation with lower DSS (P < 0.05)
VEGF/IHC
MVD/IHC
CONCLUSION

As highlighted above, angiogenesis mediates an extensive proportion of GIST’s malignant dynamics. Several signaling pathways are implicated in the regulation of angiogenesis such as: The VEGF, the fibroblast growth factor-2 (FGF2), the PDGF, the angiopoietins, the Eph/ephrin signaling, the Apelin/ APLNR pathway, the HIFs and several chemokines[14]. The VEGF signaling comprises the most well-studied pathway in GIST angiogenesis.

The FGF2/R2 signaling has been extensively studied in GIST as a drug resistance mechanism. Sergei et al[54] and Boichuk et al[55] demonstrated that the blockage of FGFR2 signaling could enhance the responsiveness to DNA-Topoisomerase II inhibitors[54] while the downregulation of FGF2 signaling might stimulate the response to imatinib[55]. It’s contribution in GIST progression has been reviewed[56] but data about potential effects in GIST vascularization process are missing. Towards the same direction, the Eph/ephrin system has been investigated in carcinogenesis[57,58]. It would be of paramount importance an attempt to outline its contribution in GIST angiogenesis.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Oncology

Country/Territory of origin: Greece

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Lewitowicz P, Poland; Messias LHD S-Editor: Fan JR L-Editor: A P-Editor: Fan JR

References
1.  van der Graaf WTA, Tielen R, Bonenkamp JJ, Lemmens V, Verhoeven RHA, de Wilt JHW. Nationwide trends in the incidence and outcome of patients with gastrointestinal stromal tumour in the imatinib era. Br J Surg. 2018;105:1020-1027.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 28]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
2.  Casali PG, Blay JY, Abecassis N, Bajpai J, Bauer S, Biagini R, Bielack S, Bonvalot S, Boukovinas I, Bovee JVMG, Boye K, Brodowicz T, Buonadonna A, De Álava E, Dei Tos AP, Del Muro XG, Dufresne A, Eriksson M, Fedenko A, Ferraresi V, Ferrari A, Frezza AM, Gasperoni S, Gelderblom H, Gouin F, Grignani G, Haas R, Hassan AB, Hindi N, Hohenberger P, Joensuu H, Jones RL, Jungels C, Jutte P, Kasper B, Kawai A, Kopeckova K, Krákorová DA, Le Cesne A, Le Grange F, Legius E, Leithner A, Lopez-Pousa A, Martin-Broto J, Merimsky O, Messiou C, Miah AB, Mir O, Montemurro M, Morosi C, Palmerini E, Pantaleo MA, Piana R, Piperno-Neumann S, Reichardt P, Rutkowski P, Safwat AA, Sangalli C, Sbaraglia M, Scheipl S, Schöffski P, Sleijfer S, Strauss D, Strauss SJ, Hall KS, Trama A, Unk M, van de Sande MAJ, van der Graaf WTA, van Houdt WJ, Frebourg T, Gronchi A, Stacchiotti S; ESMO Guidelines Committee, EURACAN and GENTURIS. Gastrointestinal stromal tumours: ESMO-EURACAN-GENTURIS Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2022;33:20-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 204]  [Article Influence: 102.0]  [Reference Citation Analysis (0)]
3.  Ma GL, Murphy JD, Martinez ME, Sicklick JK. Epidemiology of gastrointestinal stromal tumors in the era of histology codes: results of a population-based study. Cancer Epidemiol Biomarkers Prev. 2015;24:298-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 152]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
4.  Nishida T, Blay JY, Hirota S, Kitagawa Y, Kang YK. The standard diagnosis, treatment, and follow-up of gastrointestinal stromal tumors based on guidelines. Gastric Cancer. 2016;19:3-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 234]  [Cited by in F6Publishing: 293]  [Article Influence: 36.6]  [Reference Citation Analysis (0)]
5.  Caterino S, Lorenzon L, Petrucciani N, Iannicelli E, Pilozzi E, Romiti A, Cavallini M, Ziparo V. Gastrointestinal stromal tumors: correlation between symptoms at presentation, tumor location and prognostic factors in 47 consecutive patients. World J Surg Oncol. 2011;9:13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 57]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
6.  Sarlomo-Rikala M, Kovatich AJ, Barusevicius A, Miettinen M. CD117: a sensitive marker for gastrointestinal stromal tumors that is more specific than CD34. Mod Pathol. 1998;11:728-734.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Joensuu H, Hohenberger P, Corless CL. Gastrointestinal stromal tumour. Lancet. 2013;382:973-983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 396]  [Cited by in F6Publishing: 437]  [Article Influence: 39.7]  [Reference Citation Analysis (0)]
8.  Miettinen M, Wang ZF, Sarlomo-Rikala M, Osuch C, Rutkowski P, Lasota J. Succinate dehydrogenase-deficient GISTs: a clinicopathologic, immunohistochemical, and molecular genetic study of 66 gastric GISTs with predilection to young age. Am J Surg Pathol. 2011;35:1712-1721.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 273]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
9.  Jain RK. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell. 2014;26:605-622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1120]  [Cited by in F6Publishing: 1084]  [Article Influence: 108.4]  [Reference Citation Analysis (0)]
10.  Teng LS, Jin KT, He KF, Zhang J, Wang HH, Cao J. Clinical applications of VEGF-trap (aflibercept) in cancer treatment. J Chin Med Assoc. 2010;73:449-456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 34]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
11.  Klug LR, Khosroyani HM, Kent JD, Heinrich MC. New treatment strategies for advanced-stage gastrointestinal stromal tumours. Nat Rev Clin Oncol. 2022;19:328-341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 32]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
12.  Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285:1182-1186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5115]  [Cited by in F6Publishing: 5696]  [Article Influence: 107.5]  [Reference Citation Analysis (1)]
13.  Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 1996;86:353-364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4848]  [Cited by in F6Publishing: 4711]  [Article Influence: 168.3]  [Reference Citation Analysis (0)]
14.  Lugano R, Ramachandran M, Dimberg A. Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol Life Sci. 2020;77:1745-1770.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 422]  [Cited by in F6Publishing: 851]  [Article Influence: 170.2]  [Reference Citation Analysis (0)]
15.  Llombart-Bosch A, López-Guerrero JA, Carda Batalla C, Ruíz Suarí A, Peydró-Olaya A. Structural basis of tumoral angiogenesis. Adv Exp Med Biol. 2003;532:69-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
16.  Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011;473:298-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3471]  [Cited by in F6Publishing: 3746]  [Article Influence: 288.2]  [Reference Citation Analysis (0)]
17.  Kuczynski EA, Vermeulen PB, Pezzella F, Kerbel RS, Reynolds AR. Vessel co-option in cancer. Nat Rev Clin Oncol. 2019;16:469-493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 258]  [Article Influence: 51.6]  [Reference Citation Analysis (0)]
18.  Giner F, Machado I, Lopez-Guerrero JA, Mayordomo-Aranda E, Llombart-Bosch A. High-risk gastrointestinal stromal tumour (GIST) and synovial sarcoma display similar angiogenic profiles: a nude mice xenograft study. Ecancermedicalscience. 2017;11:726.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
19.  Chen X, Wang Y, Nelson D, Tian S, Mulvey E, Patel B, Conti I, Jaen J, Rollins BJ. CCL2/CCR2 Regulates the Tumor Microenvironment in HER-2/neu-Driven Mammary Carcinomas in Mice. PLoS One. 2016;11:e0165595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 35]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
20.  Jiang BH, Liu LZ. PI3K/PTEN signaling in angiogenesis and tumorigenesis. Adv Cancer Res. 2009;102:19-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 338]  [Cited by in F6Publishing: 408]  [Article Influence: 27.2]  [Reference Citation Analysis (0)]
21.  Wang N, Wu R, Tang D, Kang R. The BET family in immunity and disease. Signal Transduct Target Ther. 2021;6:23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 122]  [Article Influence: 40.7]  [Reference Citation Analysis (0)]
22.  Mu J, Sun P, Ma Z. BRD4 promotes tumor progression and NF-κB/CCL2-dependent tumor-associated macrophage recruitment in GIST. Cell Death Dis. 2019;10:935.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
23.  Liu N, Ling R, Tang X, Yu Y, Zhou Y, Chen D. Post-Translational Modifications of BRD4: Therapeutic Targets for Tumor. Front Oncol. 2022;12:847701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 11]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
24.  Calin GA, di Iasio MG, Caprini E, Vorechovsky I, Natali PG, Sozzi G, Croce CM, Barbanti-Brodano G, Russo G, Negrini M. Low frequency of alterations of the alpha (PPP2R1A) and beta (PPP2R1B) isoforms of the subunit A of the serine-threonine phosphatase 2A in human neoplasms. Oncogene. 2000;19:1191-1195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 164]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
25.  Janssens V, Goris J. Protein phosphatase 2A: a highly regulated family of serine/threonine phosphatases implicated in cell growth and signalling. Biochem J. 2001;353:417-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 407]  [Cited by in F6Publishing: 924]  [Article Influence: 40.2]  [Reference Citation Analysis (0)]
26.  Xie J, Yoon J, Yang SS, Lin SH, Huang CL. WNK1 protein kinase regulates embryonic cardiovascular development through the OSR1 signaling cascade. J Biol Chem. 2013;288:8566-8574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
27.  Lai JG, Tsai SM, Tu HC, Chen WC, Kou FJ, Lu JW, Wang HD, Huang CL, Yuh CH. Zebrafish WNK lysine deficient protein kinase 1 (wnk1) affects angiogenesis associated with VEGF signaling. PLoS One. 2014;9:e106129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
28.  Okamoto Y, Sawaki A, Ito S, Nishida T, Takahashi T, Toyota M, Suzuki H, Shinomura Y, Takeuchi I, Shinjo K, An B, Ito H, Yamao K, Fujii M, Murakami H, Osada H, Kataoka H, Joh T, Sekido Y, Kondo Y. Aberrant DNA methylation associated with aggressiveness of gastrointestinal stromal tumour. Gut. 2012;61:392-401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 51]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
29.  Geddert H, Braun A, Kayser C, Dimmler A, Faller G, Agaimy A, Haller F, Moskalev EA. Epigenetic Regulation of CD133 in Gastrointestinal Stromal Tumors. Am J Clin Pathol. 2017;147:515-524.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
30.  Lee DH, Kim GW, Jeon YH, Yoo J, Lee SW, Kwon SH. Advances in histone demethylase KDM4 as cancer therapeutic targets. FASEB J. 2020;34:3461-3484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 62]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
31.  Hu F, Li H, Liu L, Xu F, Lai S, Luo X, Hu J, Yang X. Histone demethylase KDM4D promotes gastrointestinal stromal tumor progression through HIF1β/VEGFA signalling. Mol Cancer. 2018;17:107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 26]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
32.  Yakes FM, Chen J, Tan J, Yamaguchi K, Shi Y, Yu P, Qian F, Chu F, Bentzien F, Cancilla B, Orf J, You A, Laird AD, Engst S, Lee L, Lesch J, Chou YC, Joly AH. Cabozantinib (XL184), a novel MET and VEGFR2 inhibitor, simultaneously suppresses metastasis, angiogenesis, and tumor growth. Mol Cancer Ther. 2011;10:2298-2308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 825]  [Cited by in F6Publishing: 935]  [Article Influence: 71.9]  [Reference Citation Analysis (0)]
33.  Grüllich C. Cabozantinib: a MET, RET, and VEGFR2 tyrosine kinase inhibitor. Recent Results Cancer Res. 2014;201:207-214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 49]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
34.  Wilhelm SM, Adnane L, Newell P, Villanueva A, Llovet JM, Lynch M. Preclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and VEGF and PDGF receptor tyrosine kinase signaling. Mol Cancer Ther. 2008;7:3129-3140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1000]  [Cited by in F6Publishing: 1061]  [Article Influence: 66.3]  [Reference Citation Analysis (0)]
35.  Gebreyohannes YK, Schöffski P, Van Looy T, Wellens J, Vreys L, Cornillie J, Vanleeuw U, Aftab DT, Debiec-Rychter M, Sciot R, Wozniak A. Cabozantinib Is Active against Human Gastrointestinal Stromal Tumor Xenografts Carrying Different KIT Mutations. Mol Cancer Ther. 2016;15:2845-2852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
36.  Huynh H, Lee JW, Chow PK, Ngo VC, Lew GB, Lam IW, Ong HS, Chung A, Soo KC. Sorafenib induces growth suppression in mouse models of gastrointestinal stromal tumor. Mol Cancer Ther. 2009;8:152-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 46]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
37.  Agaimy A. Gastrointestinal stromal tumors (GIST) from risk stratification systems to the new TNM proposal: more questions than answers? Int J Clin Exp Pathol. 2010;3:461-471.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Takahashi T, Nakajima K, Nishitani A, Souma Y, Hirota S, Sawa Y, Nishida T. An enhanced risk-group stratification system for more practical prognostication of clinically malignant gastrointestinal stromal tumors. Int J Clin Oncol. 2007;12:369-374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 114]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
39.  Sakamoto H, Kitano M, Matsui S, Kamata K, Komaki T, Imai H, Dote K, Kudo M. Estimation of malignant potential of GI stromal tumors by contrast-enhanced harmonic EUS (with videos). Gastrointest Endosc. 2011;73:227-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 80]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
40.  Iannicelli E, Carbonetti F, Federici GF, Martini I, Caterino S, Pilozzi E, Panzuto F, Briani C, David V. Evaluation of the Relationships Between Computed Tomography Features, Pathological Findings, and Prognostic Risk Assessment in Gastrointestinal Stromal Tumors. J Comput Assist Tomogr. 2017;41:271-278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
41.  Horton KM, Juluru K, Montogomery E, Fishman EK. Computed tomography imaging of gastrointestinal stromal tumors with pathology correlation. J Comput Assist Tomogr. 2004;28:811-817.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 71]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
42.  Baheti AD, Shinagare AB, O'Neill AC, Krajewski KM, Hornick JL, George S, Ramaiya NH, Tirumani SH. MDCT and clinicopathological features of small bowel gastrointestinal stromal tumours in 102 patients: a single institute experience. Br J Radiol. 2015;88:20150085.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 30]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
43.  Strauss LG, Dimitrakopoulou-Strauss A, Koczan D, Pan L, Hohenberger P. Correlation of dynamic PET and gene array data in patients with gastrointestinal stromal tumors. ScientificWorldJournal. 2012;2012:721313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
44.  Palazzo L, Landi B, Cellier C, Cuillerier E, Roseau G, Barbier JP. Endosonographic features predictive of benign and malignant gastrointestinal stromal cell tumours. Gut. 2000;46:88-92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 244]  [Cited by in F6Publishing: 261]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
45.  Chhoda A, Jain D, Surabhi VR, Singhal S. Contrast Enhanced Harmonic Endoscopic Ultrasound: A Novel Approach for Diagnosis and Management of Gastrointestinal Stromal Tumors. Clin Endosc. 2018;51:215-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
46.  Yamashita Y, Kato J, Ueda K, Nakamura Y, Abe H, Tamura T, Itonaga M, Yoshida T, Maeda H, Moribata K, Niwa T, Maekita T, Iguchi M, Tamai H, Ichinose M. Contrast-enhanced endoscopic ultrasonography can predict a higher malignant potential of gastrointestinal stromal tumors by visualizing large newly formed vessels. J Clin Ultrasound. 2015;43:89-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
47.  Hadjimichael AC, Pergaris A, Kaspiris A, Foukas AF, Theocharis SE. Liquid Biopsy: A New Translational Diagnostic and Monitoring Tool for Musculoskeletal Tumors. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
48.  Masaoutis C, Korkolopoulou P, Theocharis S. Exosomes in sarcomas: Tiny messengers with broad implications in diagnosis, surveillance, prognosis and treatment. Cancer Lett. 2019;449:172-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
49.  Lone SN, Nisar S, Masoodi T, Singh M, Rizwan A, Hashem S, El-Rifai W, Bedognetti D, Batra SK, Haris M, Bhat AA, Macha MA. Liquid biopsy: a step closer to transform diagnosis, prognosis and future of cancer treatments. Mol Cancer. 2022;21:79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 210]  [Article Influence: 105.0]  [Reference Citation Analysis (0)]
50.  Heydari R, Abdollahpour-Alitappeh M, Shekari F, Meyfour A. Emerging Role of Extracellular Vesicles in Biomarking the Gastrointestinal Diseases. Expert Rev Mol Diagn. 2021;21:939-962.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
51.  Koulouris A, Tsagkaris C, Messaritakis I, Gouvas N, Sfakianaki M, Trypaki M, Spyrou V, Christodoulakis M, Athanasakis E, Xynos E, Tzardi M, Mavroudis D, Souglakos J. Resectable Colorectal Cancer: Current Perceptions on the Correlation of Recurrence Risk, Microbiota and Detection of Genetic Mutations in Liquid Biopsies. Cancers (Basel). 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
52.  Ko TK, Lee E, Ng CC, Yang VS, Farid M, Teh BT, Chan JY, Somasundaram N. Circulating Tumor DNA Mutations in Progressive Gastrointestinal Stromal Tumors Identify Biomarkers of Treatment Resistance and Uncover Potential Therapeutic Strategies. Front Oncol. 2022;12:840843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
53.  Li J, Guo S, Sun Z, Fu Y. Noncoding RNAs in Drug Resistance of Gastrointestinal Stromal Tumor. Front Cell Dev Biol. 2022;10:808591.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
54.  Sergei B, Pavel D, Aigul G, Firyuza B, Ilmira N, Ilshat M, Aida A, Refat K, Natalia A, Elena S, Vera G. Inhibition of FGFR2-Signaling Attenuates a Homology-Mediated DNA Repair in GIST and Sensitizes Them to DNA-Topoisomerase II Inhibitors. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
55.  Boichuk S, Galembikova A, Mikheeva E, Bikinieva F, Aukhadieva A, Dunaev P, Khalikov D, Petrov S, Kurtasanov R, Valeeva E, Kireev I, Dugina V, Lushnikova A, Novikova M, Kopnin P. Inhibition of FGF2-Mediated Signaling in GIST-Promising Approach for Overcoming Resistance to Imatinib. Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
56.  Napolitano A, Ostler AE, Jones RL, Huang PH. Fibroblast Growth Factor Receptor (FGFR) Signaling in GIST and Soft Tissue Sarcomas. Cells. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
57.  Pergaris A, Danas E, Goutas D, Sykaras AG, Soranidis A, Theocharis S. The Clinical Impact of the EPH/Ephrin System in Cancer: Unwinding the Thread. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 26]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
58.  Papadakos SP, Petrogiannopoulos L, Pergaris A, Theocharis S. The EPH/Ephrin System in Colorectal Cancer. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
59.  Zhao Y, Wang Q, Deng X, Zhao Y. Altered angiogenesis gene expression in gastrointestinal stromal tumors: potential use in diagnosis, outcome prediction, and treatment. Neoplasma. 2012;59:384-392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
60.  Kang BW, Kim JG, Chae YS, Bae HI, Kwon O, Chung HY, Yu W, Song HS, Kang YN, Ryu SW, Lee KH, Bae YK, Choi JH, Kim SW, Ryoo HM, Cho CH, Chae HD, Park KW, Gu MJ, Bae BJ. Clinical significance of vascular endothelial growth factor and vascular endothelial growth factor receptor-2 gene polymorphisms in patients with gastrointestinal stromal tumors. Asia Pac J Clin Oncol. 2014;10:e40-e45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
61.  Toda-Ishii M, Akaike K, Suehara Y, Mukaihara K, Kubota D, Kohsaka S, Okubo T, Mitani K, Mogushi K, Takagi T, Kaneko K, Yao T, Saito T. Clinicopathological effects of protein phosphatase 2, regulatory subunit A, alpha mutations in gastrointestinal stromal tumors. Mod Pathol. 2016;29:1424-1432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 14]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
62.  Liu N, Huang J, Sun S, Zhou Z, Zhang J, Gao F, Sun Q. Expression of matrix metalloproteinase-9, cyclooxygenase-2 and vascular endothelial growth factor are increased in gastrointestinal stromal tumors. Int J Clin Exp Med. 2015;8:6495-6501.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Takahashi R, Tanaka S, Kitadai Y, Sumii M, Yoshihara M, Haruma K, Chayama K. Expression of vascular endothelial growth factor and angiogenesis in gastrointestinal stromal tumor of the stomach. Oncology. 2003;64:266-274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 78]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
64.  Verboom MC, Kloth JSL, Swen JJ, van der Straaten T, Bovée JVMG, Sleijfer S, Reyners AKL, Mathijssen RHJ, Guchelaar HJ, Steeghs N, Gelderblom H. Genetic polymorphisms in angiogenesis-related genes are associated with worse progression-free survival of patients with advanced gastrointestinal stromal tumours treated with imatinib. Eur J Cancer. 2017;86:226-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 10]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
65.  Chen WT, Huang CJ, Wu MT, Yang SF, Su YC, Chai CY. Hypoxia-inducible factor-1alpha is associated with risk of aggressive behavior and tumor angiogenesis in gastrointestinal stromal tumor. Jpn J Clin Oncol. 2005;35:207-213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 57]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
66.  Basilio-de-Oliveira RP, Pannain VL. Prognostic angiogenic markers (endoglin, VEGF, CD31) and tumor cell proliferation (Ki67) for gastrointestinal stromal tumors. World J Gastroenterol. 2015;21:6924-6930.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 54]  [Cited by in F6Publishing: 55]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
67.  Imamura M, Yamamoto H, Nakamura N, Oda Y, Yao T, Kakeji Y, Baba H, Maehara Y, Tsuneyoshi M. Prognostic significance of angiogenesis in gastrointestinal stromal tumor. Mod Pathol. 2007;20:529-537.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 45]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
68.  Wang TB, Qiu WS, Wei B, Deng MH, Wei HB, Dong WG. Serum vascular endothelial growth factor and angiogenesis are related to the prognosis of patients with gastrointestinal stromal tumors. Ir J Med Sci. 2009;178:315-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]