Open Access

Evaluation of VEGF and VEGFR gene expression as prognostic markers in low and intermediate‑1 risk patients with myelodysplastic syndromes

  • Authors:
    • Nikolay Kalitin
    • Galina Dudina
    • Natalia Kostritsa
    • Anastasiya Sivirinova
    • Aida Karamysheva
  • View Affiliations

  • Published online on: January 26, 2023     https://doi.org/10.3892/ol.2023.13681
  • Article Number: 95
  • Copyright: © Kalitin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Vascular endothelial growth factors (VEGFs) are angiogenic factors playing a key role in tumor development. VEGFs are produced by different normal and tumor cells, including platelets, lymphocytes and mononuclear cells of peripheral blood. VEGF (VEGF‑A, VEGF‑C and VEGF‑D) and VEGFR (VEGFR1, VEGFR2 and VEGFR3) gene expression was studied in patients with myelodysplastic syndrome (MDS) to evaluate the possible prognostic role of the expression of these genes. Gene expression levels were determined using peripheral blood samples of 51 patients with MDS and 15 healthy volunteers by quantitative PCR. Expression of all VEGF and VEGFR genes was elevated in patients with MDS compared with healthy volunteers. No association of VEGF‑A expression with the hemoglobin content in peripheral blood was found. The analyses of gene expression in patients with MDS stratified by risk groups according to the International Prognostic Scoring System showed progressive augmentation of VEGF‑A gene expression from low to high‑risk groups and VEGFR1 and VEGFR2 expression from intermediate‑1 to high‑risk groups. The statistically significant difference in survival time of patients with high and low levels of VEGFR1 expression was revealed. VEGF‑A/VEGFR1 expression may be important for risk evaluation of patients with MDS.

Introduction

Myelodysplastic syndromes (MDSs) are a heterogeneous group of blood disorders characterized by peripheral blood cytopenia due to ineffective hematopoiesis, dysplasia in ≥1 hematopoietic cell lineages and increased risk of transformation to acute myeloid leukemia (AML). MDSs are hypothesized to be clonal stem cell disorders arising from accumulation of multiple gene abnormalities, such as somatic point mutations, copy-number alterations and chromosomal aberrations (1). Genomic and chromosomal instability and variable molecular mechanisms contribute to pathogenesis and prognosis of MDS (2).

Accumulation of bone marrow (BM) blasts is a key feature and one of the main risk criterion and prognostic factors in patients with MDS (3). However, the majority of patients with MDS die from causes intrinsic to the disease, such as infection, pneumonia, sepsis and bleeding, which are not associated with leukemic transformation (4). This is particularly relevant to patients with lower risk MDS, which is defined as low or intermediate-1-risk according to the International Prognostic Scoring System (IPSS) (5).

VEGFs and their receptors, VEGFRs, are involved in regulation of proliferation, migration, invasion and differentiation of normal and cancer cells (69). VEGFs exert their effects by binding to receptors of the VEGFR family that consists of tyrosine kinase receptors, VEGFR1, VEGFR2 and VEGFR3 (10). The role of VEGF-A/VEGFR1 and VEGF-A/VEGFR2 signaling pathways has been evaluated in certain hematological malignancies, such as multiple myeloma, lymphoma and myeloproliferative neoplasms (1113). However, a potential role of VEGF-A-dependent signaling in MDS pathogenesis has been poorly studied and the results are contradictory. Studies have demonstrated that BM blasts express VEGF-A and modulate VEGF-A-dependent autocrine loop signaling in MDS (14,15). However, Aguayo et al (16) found no prognostic significance of plasma VEGF-A levels in patients with MDS but suggested that VEGF-A plays a role as a prognostic factor in patients with AML. Verstovsek et al (17) showed that increased VEGF-A expression in BM specimens is inversely associated with survival time in patients with MDS or AML, whereas VEGFR1 and VEGFR2 expression levels have no prognostic impact.

To the best of our knowledge, there is no published data on the role of VEGF-C, VEGF-D ligands and VEGFR3 in MDS pathogenesis. However, a previous study revealed that activation of the VEGF-C/VEGFR3 pathway promotes cancer cell mobility to induce metastasis and an increase in VEGF-C and/or VEGFR3 expression may be associated with a shorter survival in numerous types of malignancy (18). The aim of the present study was to evaluate VEGF and VEGFRs expression as putative prognostic markers for MDS.

Patients and methods

Patients and controls

The study group consisted of 51 patients with verified MDS (31 female and 20 male) with a median age of 69.8 years (range, 59–77 years) who were diagnosed between January 1, 2011 and August 31, 2018 and treated at A.S. Loginov Moscow Clinical Scientific Center (Moscow, Russia). The diagnosis of MDS was based on cytological examination of peripheral blood cells. The patients were followed-up from MDS diagnosis until May 31, 2019. The transformation to AML or the death of patients was considered, if they occurred during the follow-up period. The control group consisted of 15 volunteers (8 females and 7 male) free of neoplasms or any other abnormality. The median age and range were 65.3 and 61–68 years, respectively. Clinical and hematological variables (hemoglobin content and platelet and leukocyte count) in the control group were within normal ranges.

Laboratory procedures

Hemoglobin concentration, as well as platelet and leukocyte counts were measured using the automated hematology analyzer ADVIA 2120i according to the manufacturer's recommendations (Siemens Healthineers AG).

Inclusion and exclusion criteria

Inclusion criteria were de novo female and male patients with MDS aged ≥18 years old or patients with MDS who only received prior supportive care (such as red blood cell and/or platelet transfusions for severe anemia and severe thrombocytopenia improvement). Patients treated with erythropoiesis-stimulating agents (in patients with chromosome 5q deletion) were also included. Patients with BM blast cells=5% were excluded. Patients previously treated with hypomethylating agents and/or who received immunosuppressive therapy were excluded. Patients with the hypoplastic variant of MDS as well as patients that refused to participate were not included.

The patient risk stratification according to the 2017 World Health Organization (WHO) classification of tumors of hematopoietic and lymphoid tissue (19) and IPSS (20), as well as other patient characteristics are presented in Table I. Karyotype was classified using the International System for Human Cytogenetic Nomenclature (20,21).

Table I.

Clinical variables of 51 patients with MDS.

Table I.

Clinical variables of 51 patients with MDS.

Clinical variableValue
Median age (range), years69.80 (59.00–77.00)
Sex, n
  Female31.00
  Male20.00
WHO classification, n
  MDS-SLD8.00
  MDS-RS2.00
  MDS-MLD11.00
  MDS-EBa22.00
  MDS-del(5q)8.00
IPSS classification, n
  Low15.00
  Intermediate-112.00
  Intermediate-29.00
  High15.00
Karyotypeb, n
  Good30.00
  Intermediate2.00
  Poor6.00
  n/d13.00
Mean hemoglobin count ± SEM, g/dl6.59±1.60
Mean platelets count ± SEM, ×109/l118.33±60.85
Mean leukocyte count ± SEM, ×109/l4.67±3.53
Bone marrow blasts, n
  >5%24.00
  <5%27.00
AML progression, n14.00
Death, n26.00
Survival time ± SD, months24.80±22.68

a MDS-EB is EB1 + EB2.

b Cytogenetic subgroups: Good, normal karyotype. Isolated-Y, del(5q) or del(20q); poor, ≥3 abnormalities or chromosome 7 anomalies; intermediate, other abnormalities; n/d, not determined; MDS-SLD, myelodysplastic syndrome with single-lineage dysplasia; MDS-RS, MDS with ring sideroblasts; MDS-MLD, MDS with multilineage dysplasia; MDS-EB, MDS with excess blasts; MDS-del(5q), MDS with isolated 5q-deletion; IPSS, International Prognosis Scoring System; AML, acute myeloid leukemia.

Mononuclear cell preparation and cDNA synthesis

The peripheral blood specimens (5–10 ml) were separated using a Ficoll® density gradient (PanEko) and the obtained mononuclear cell fraction was used for further study. Total RNA was isolated by TRI Reagent® (Molecular Research Center). All procedures were as previously described (22). Briefly, cDNA synthesis reaction mixture contained 1 µg purified total RNA, 1 µl random 6 primers (Syntol), 2.5 mM dNTP mixture (Thermo Fisher Scientific, Inc.), 0.4 units RNase inhibitor (Thermo Fisher Scientific, Inc.) and 2 units M-MuLVplus reverse transcriptase (Thermo Fisher Scientific, Inc.). The mixture volume was 25 µl. The synthesis was performed in Terzic’ thermocycler (DNA technology, Russia) at 42°С for 50 min with pre-incubating for 10 min at 25°С. The reaction was stopped by heating at 70°С for 10 min.

Quantitative PCR (qPCR)

The amplification of cDNA was performed in a Bio-Rad CFX (Bio-Rad Laboratories, Inc.) detection system using EvaGreen® dye (Biotium) and qPCR master mix (Syntol) according to the manufacture's protocol. PCR conditions for all genes were 95°С for 5 min followed by 39 cycles of 95°С for 20 sec, 59°С for 25 sec and 72°С for 20 sec. Each sample was measured in triplicate. For data standardization, the 60S subunit of the RPL27 gene was used. The relative expression was determined according to the 2−∆∆Cq equation [∆Cq=Cq (RPL27)-Cq (test gene), where Cq is the threshold cycle of the gene in the exponential phase of the amplification curve] (23). The following primers were used: VEGF-A forward, 5′-AGGGCAGAATCATCACGAAGT-3′ and reverse, 5′-AGGGCTTCGATTGGATGGCA-3′; VEGF-C forward, 5′-GAGGAGCAGTTACGGTCTGTG-3′ and reverse, 5′-tcctttccttagctgacacttgt-3′; VEGF-D forward, 5′-TCCCATCGGTCCACTAGGTTT-3′ and reverse, 5′-AGGGCTGCACTGAGTTCTTTG-3′; VEGFR1 forward, 5′-TTTGCCTGAAATGGTGAGTAAGG-3′ and reverse, 5′-TGGTTTGCTTGAGCTGTGTTC-3′; VEGFR2 forward, 5′-GGCCCAATAATCAGAGTGGCA-3′ and reverse, 5′-CCAGTGTCATTTCCGATCACTTT-3′; VEGFR3 forward, 5′-TGCACGAGGTACATGCCAAC-3′ and reverse, 5′-GCTGCTCAAAGTCTCTCACGAA-3′ and RPL27 forward, 5′-ACCGCTACCCCCGCAAAGTG-3′ and reverse, 5′-CCCGTCGGGCCTTGCGTTTA-3′.

Statistical analysis

All qPCR experiments were performed in triplicate. Data are presented as the mean ± SEM. Correlation was analyzed using Pearson's rank test. Overall survival was estimated by Kaplan-Meier method with log-rank test. To assess diagnostic value of VEGF and VEGFRs gene expression as candidate biomarkers was performed receiver operating characteristic (ROC) analysis. Area under ROC curve (AUC) was used to compare the discriminatory performance of putative markers to determine their utility as a novel diagnostic test. Statistical significance was analyzed using an unpaired two-tailed Student's t test. P<0.05 was considered to indicate a statistically significant difference. All statistical calculations were performed using GraphPad Prism for Windows program, Version 5.00 (Trial), 2007 (Dotmatics).

Results

VEGF and VEGFR expression is elevated in patients with MDS

VEGF (VEGF-A, VEGF-C and VEGF-D) and VEGFR (VEGFR1, VEGFR2 and VEGFR3) gene expression levels were studied in the peripheral blood samples of 51 patients with MDS and 15 healthy donors. Gene expression varied considerably in patients with MDS compared with healthy donors (Fig. 1A). Although no statistical difference between patients with MDS and controls in VEGFR1 and VEGFR3 expression was found, relative VEGFR1 expression was 0.4-20.0×10−3 in patients with MDS and from 1.5×10−3 to 4.2×10−3 in control group (mean, 2.71±0.46×10−3 vs. 2.82±0.19) ×10−3. Similarly, VEGFR3 relative expression varied from 0.1-8.0×10−3 in patients with MDS and from 0 to 3.4×10−3 in controls (mean values: (1.78±0.24) ×10−3 vs. 1.02±0.30×10−3. VEGF-A and VEGF-C expression were higher in patients with MDS than in controls. The mean values of relative VEGFA expression in patients with MDS and in control group were (19.73±2.84) ×10−3 and 11.07±0.99×10−3. The mean values of relative VEGF-C expression in MDS patients and control group were (22.50±3.88) ×10−3 and 9.23±1.69×10−3. VEGFR2 expression levels were low in the group of healthy volunteers (mean value 0.08±0.04) and upregulated in patients with MDS (mean value 0.82±1.39), indicating that VEGFR2-dependent signaling was stimulated in patients with MDS. VEGF-D expression was absent in control group and varied from 0 to 1.4×10−3 in patients with MDS.

VEGF-A is activated under hypoxic conditions (24). To evaluate the association between VEGF-A expression and hypoxia, levels of VEGF-A expression and hemoglobin content in the peripheral blood samples of patients with MDS was compared. No correlation was found between these two variables (Fig. 1B). Moreover, the mean value of hemoglobin was almost the same in patients with MDS with different levels of VEGF-A expression (above and below the mean level of 19.62×10−3 of relative VEGF-A gene expression; Fig. 1C). Thus, VEGF-A expression was not dependent on the content of hemoglobin in peripheral blood of patients with MDS.

To determine if there was an association between clinical characteristics of patients with MDS and gene expression, levels of hemoglobin, platelets and leukocytes were compared with VEGF and VEGFR gene expression levels. Significant associations were found between levels of platelets and the expression levels of VEGF-C and VEGF-D (Fig. 1D and E).

VEGF and VEGFR expression in patients with MDS with different risk of disease development

All patients with MDS were stratified in the present study, according to the IPSS. Patients were assigned to low (15 patients), intermediate-1 (12 patients), intermediate-2 (9 patients) and high (15 patients) risk groups and the survival time and gene expression in these groups were compared. No statistical difference in survival time between any of these risk groups was found. Nevertheless, the median survival time diminished from the intermediate-1 (51 months) and intermediate-2 (49 months) risk groups to the high-risk group (23 months). The median survival time in the low-risk group was not reached (Fig. 2A).

VEGF-A expression was progressively elevated from the low to the high-risk groups (Fig. 2). A statistically significant difference was found for VEGF-A expression between the low and intermediate-2 risk groups, as well as between the low and high-risk groups. No statistically significant difference between risk groups in VEGF-C, VEGF-D, VEGFR1 and VEGFR2 expression was found. Expression of VEGFR1 and VEGFR2 genes was notably elevated from the intermediate-1 to the high-risk group. VEGF-C and VEGF-D expression was notably higher in the low-risk group of patients compared with the other groups. VEGFR3 expression was lowest in the high risk group. The only statistically significant difference in gene expression was found between the intermediate-2 and high risk groups for VEGFR3.

VEGF and VEGFR expression in patients with MDS with different levels of BM blasts

A key prognostic factor characterizing patients with MDS is the percentage of BM blast cells. BM blast cells <5% is usually associated with good prognosis (20). In the present study, all patients from low and intermediate-1 risk groups had BM blast levels <5% and patients from intermediate-2 and high-risk groups had BM blast levels >5%. The survival time and gene expression levels of MDS patients with BM blast levels <5% (27 patients) and >5% (24 patients) were compared. No significant difference in survival time between these patients was found, although the median survival times were different (51 vs. 28 months for <5% and >5% BM blast levels, respectively; Fig. 3A).

Although there was no statistically significant difference, the mean VEGF-A, VEGFR1 and VEGFR2 expression levels were elevated in patients with MDS with >5% BM blast levels (Fig. 3B and C). VEGF-C, VEGF-D and VEGFR3 expression was decreased in the group of patients with MDS with BM blast levels >5%.

The prognostic evaluation of potential clinical outcomes in MDS patients with BM blast levels <5% is complicated. Some of these patients die within a few months of developing AML transformation or other complications of bone marrow failure, while others can survive for a long time (25). Survival time of patients with BM blast levels <5% (group 1, 9 patients) did not differ from that of patients with BM blast levels >5% (17 vs. 14 months, respectively), while patients of a group 2 with BM blast levels <5% had an improved survival (group 2, 18 patients; Fig. 3A). VEGF and VEGFRs gene expression levels in groups 1 and 2 were compared (Fig. 3D and E) and VEGF-A, VEGF-C, VEGFR1 and VEGFR2 expression in these groups was very similar to that in patients with MDS with BM blast levels >5 and <5% (Fig. 3B and C).

Survival of MDS patients with different VEGF-A and VEGFR1 expression levels

As both VEGF-A and VEGFR1 gene expression levels were elevated in patients with MDS with a worse prognosis, the survival of patients was compared. No statistically significant difference was found between patients with MDS with high and low levels of VEGF-A expression, although the median survival times in patients with VEGF-A expression levels above (18 patients) and below (33 patients) the mean value (1.96х10−3) were different (23 and 43 months, respectively; Fig. 4A). Patients with MDS with VEGFR1 expression levels exceeding the mean relative VEGFR1 gene expression (2.69х10−3; 15 patients) had statistically worse survival (log-rank test; Fig. 4B) with a median survival time of 17 months compared with 48 months for the group with low levels of VEGFR1 expression (36 patients).

To evaluate the diagnostic value of VEGF-A and VEGFR1 gene expression levels as candidate biomarkers of MDS, receiver operating characteristic (ROC) curve analysis was applied. Area under the curve (AUC) was 0.57 for the VEGF-A gene, but the result was not statistically significant (Fig. 5A). However, VEGFR1 expression levels discriminated between patients with MDS and the healthy controls (AUC=0.684), suggesting its potential diagnostic value (Fig. 5B).

Discussion

Due to the heterogeneity of MDSs at morphological, clinical and molecular levels, the accurate diagnosis of these diseases has certain problems (subjectivity of morphological assessment of bone marrow aspirate and biopsy, as well as heterogeneity of cytogenetic alterations) and the expected clinical outcome for patients with MDS is different. Whereas some patients transform to AML or die from complications of BM transplant failure within a few months, other patients with MDS survive for years without major hematological problems (26). A precise diagnosis is important for the prediction of patient survival and risk of AML transformation and selection of appropriate therapy.

Several classification systems have been presented to evaluate MDS diagnosis, the most commonly used being the French-American-British (FAB) (19) and IPSS (20) classification. In the present study, patients with MDS were stratified according to IPSS scoring. The diagnosis of MDS is often based on morphological characteristics, and due to the subjectivity of this evaluation, discrepancy in MDS diagnoses exists. Naqvi et al (27) analyzed discordance between the diagnosis of 915 patients with MDS referred according to FAB or IPSS and final diagnosis, and found that 12% of diagnoses (109/915 patients) were reclassified. Most of the reclassified patients (67% classified according to FAB and 77% classified by IPSS) had higher risk disease. According to the WHO 2017 classification, a correct diagnosis of MDS requires the integration of not only morphological but also clinical, cytogenetic and, potentially, molecular biology parameters, thus providing an improved classification of MDS diagnosis (19). Diagnosis is more obvious in patients with excess BM blasts: Increased blast percentages (>5–10%) indicate an increase in risk of leukemic transformation and of death (28). The patients with early low-risk disease need additional diagnostic tools, including cytogenetic evaluation, flow cytometry and DNA sequencing, to define the diagnosis and predict outcomes (29). New biological markers may also be helpful to stratify patients with MDS.

VEGF and one of its receptors, VEGFR2, are prognostic factors for a number of solid tumors such as primary (30) and non-small cell lung cancer (31), neuroblastoma (32) and bladder (33), colon (34) and breast cancer (35). To the best of our knowledge, however, the data on VEGF-dependent signaling and its role in MDS development are scarce. A study showed an increase in BM microvessel density (MVD) in refractory anemia with excess blasts in transformation (RAEB-T), chronic myelomonocytic leukemia and AML compared with patients with RA, RA with ring sideroblasts and RAEB (36). VEGF expression in MDS specimens is especially high in RAEB ± T and AML samples (14). VEGFR1 and, to a lesser extent, VEGFR2 are expressed in patients with MDS and exhibit autocrine cytokine interaction (14,17).

Based on expression data of VEGF and its receptors in MDS, it was suggested that VEGF-dependent signaling could be a potential therapeutic target (7,17). A number of agents interfering with VEGF signaling, such as bevacizumab (37), SU5416 (38), AG-013736 (39), Vatalanib (40), Aflibercept/NSC 724770 (41), have been tested in patients with MDS, but yielded no or small clinical responses and clinical applicability is limited by toxicity and side effects.

The results of investigations on the VEGF prognostic impact in patients with MDS are also not encouraging. The prognostic significance of VEGF plasma levels in patients with AML and MDS has been studied (16): VEGF plasma levels are associated with a shorter survival in patients with AML, but not MDS. By contrast, elevated BM cellular VEGF levels are significantly associated with shorter survival in patients with MDS (17). Cheng et al (42) studied the potential prognostic value of VEGF-A, VEGF-C, angiopoietin-1 (Ang-1), Ang-2 and receptor Tie-2 expression in the BM of patients with MDS. Ang-1, but not Ang-2, VEGFs or Tie-2, was shown to be an independent poor prognostic factor for patients with MDS.

Despite elevated BM MVD in patients with MDS, the treatment with anti-VEGF or its receptor agents does not produce any appreciable therapeutic effects. The attempts to evaluate the potential of VEGF or its receptors to be prognostic factors for patients with MDS also were not fruitful.

In the present study, relative mRNA expression levels of VEGF (VEGF-A, VEGF-C and VEGF-D) and VEGF receptors (VEGFR1, VEGFR2 and VEGFR3) in peripheral blood samples of patients with MDS were augmented compared with healthy control samples, suggesting that VEGF-dependent signaling was activated in patients with MDS. Although VEGF-A is activated under hypoxia, no association between VEGF-A expression and hemoglobin content in peripheral blood samples was found. The comparison of VEGF and VEGFR gene expression levels in patients with MDS subdivided according to IPSS risk revealed increased VEGF-A, VEGFR1 and VEGFR2 expression in higher risk groups, with the most significant difference for VEGFR1 expression.

A key predictor of MDS development is levels of BM blast cells in patients (28) Patients with MDS with BM blast levels <5% included all patients from the IPSS system low and intermediate-1-risk groups; patients with MDS with BM blast levels >5% comprised the intermediate-2 and high-risk groups.

The difference in overall survival of patients stratified by BM blast levels (patients with >5% vs. patients with <5% blasts) was not statistically significant, but median survival time varied considerably (51 vs. 28 months, respectively). VEGF-A, VEGFR1 and VEGFR2 gene expression was upregulated in patients with MDS with BM blast levels >5% compared with patients with BM blast levels <5%. By contrast, VEGF-C, VEGF-D and VEGFR3 gene expression levels were elevated in patients with MDS with BM blast levels <5%. This suggested that VEGF-A/VEGFR1 and VEGFR2 signaling was activated and VEGF-C and VEGF-D/VEGFR3 signaling was suppressed in patients with BM blast levels >5%.

In the present study, according to the survival curve of patients with MDS with BM blast levels <5%, two groups of patients were discriminated. The survival curve of a subgroup of these patients (group 1) largely coincided with the survival curve of patients with BM blast levels >5%, while the survival curve of patients with BM blast levels <5% (group 2) differed. The comparison of VEGF and VEGFR gene expression levels in groups 1 and 2 revealed similar gene expression, as in groups of patients with BM blast levels >5 and <5%, with the exception of VEGF-D and VEGFR3 genes. As such, the elevation of certain VEGF (VEGF-A) and VEGFR (VEGFR1 and VEGFR2) gene expression levels in patients with MDS with BM blast levels <5% may indicate the intensification of disease.

As the most prominent difference in gene expression levels between groups concerned VEGF-A and VEGFR1, the survival of patients with MDS subdivided by these gene expression levels was analyzed. A significant difference in survival was found for subgroups by VEGFR1 expression. The survival rate of patients with MDS with VEGFR1 expression below the mean, but not the median level of expression, was higher than in patients with higher levels of this gene expression. The difference in survival of patients subdivided by VEGF-A expression was not significant, but the median survival times in groups with higher VEGF-A expression differed significantly from those in groups with lower expression (23 vs. 43 months in groups subdivided by the mean expression and 17 vs. 48 months in groups subdivided by the median level of expression).

VEGF-A exerts its function through binding with its two specific receptors-VEGFR1 and VEGFR2, where VEGFR2 is the primary mediator of such VEGF-A biological functions as embryogenesis and hematopoiesis (43,44). According to the present study, expression of VEGFR2, the known negative prognostic factor for many solid tumors, (4547) is not important in MDS. It was hypothesized that VEGF-A-dependent signaling may be preferentially realized through another receptor for VEGF-A, VEGFR1, as VEGFR1 expression is higher in the peripheral blood samples of patients with MDS compared with VEGFR2 expression. Previous data on VEGF and VEGFR expression levels in BM samples of patients with MDS have shown increased expression of VEGFR1, but not VEGFR2 (48). Only VEGFR1 expression in the present study had prognostic impact for patients with MDS. The elevated VEGF-A/VEGFR1 expression in patients with MDS with BM blast levels >5% and patients with BM blast levels <5% with worse survival (group 1) indicated that the progression of the disease was accompanied by VEGF-A/VEGFR1 activation. ROC analysis showed that VEGFR1 expression rather than VEGF-A expression could serve as a potential prognostic marker in MDS with low and intermediate-1 risk.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The data used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

NKa, GD, NKo, AS and AK contributed to study conception and design. Clinical material preparation and data collection were performed by GD. RT-qPCR analysis was performed by NKa, NKo and AS. AK and NKa analyzed gene expression data and wrote the manuscript. GD, NKa, NKo, AS and AK confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved (approval no. 6/2016) by the ethics commission of the A.S. Loginov Moscow Clinical Scientific Center (Moscow, Russia). All patients and volunteers provided written informed consent to participate in the present study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Papaemmanuil E, Gerstung M, Malcovati L, Tauro S, Gundem G, Van Loo P, Yoon CJ, Ellis P, Wedge DC, Pellagatti A, et al: Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood. 122:3616–3627. 36992013. View Article : Google Scholar : PubMed/NCBI

2 

Odenike O, Anastasi J and Le Beau MM: Myelodysplastic Syndromes. Clin Lab Med. 31:763–784. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Foucar K: Myelodysplastic/myeloproliferative neoplasms. Am J Clin Pathol. 132:281–289. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Dayyani F, Conley AP, Strom SS, Stevenson W, Cortes JE, Borthakur G, Faderl S, O'Brien S, Pierce S, Kantarjian H and Garcia-Manero G: Cause of death in patients with lower-risk myelodysplastic syndrome. Cancer. 116:2174–2179. 2010.PubMed/NCBI

5 

Garcia-Manero G: Myelodysplastic syndromes: 2014 Update on diagnosis, risk-stratification, and management. Am J Hematol. 89:97–108. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Price DJ, Miralem T, Jiang S, Steinberg R and Avraham H: Role of vascular endothelial growth factor in the stimulation of cellular invasion and signaling of breast cancer cells. Cell Growth Differ. 12:129–135. 2001.PubMed/NCBI

7 

Podar K and Anderson KC: The pathophysiologic role of VEGF in hematologic malignancies: Therapeutic implications. Blood. 105:1383–1395. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Lee TH, Seng S, Sekine M, Hinton C, Fu Y, Avraham HK and Avraham S: Vascular endothelial growth factor mediates intracrine survival in human breast carcinoma cells through internally expressed VEGFR1/FLT1. PLoS Med. 4:e1862007. View Article : Google Scholar : PubMed/NCBI

9 

Adams RH and Alitalo K: Molecular regulation of angiogenesis and lymphangiogenesis. Nat Rev Mol Cell Biol. 8:464–478. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Veikkola T, Karkkainen M, Claesson-Welsh L and Alitalo K: Regulation of angiogenesis via vascular endothelial growth factor receptors. Cancer Res. 60:203–212. 2000.PubMed/NCBI

11 

Vincent L, Jin DK, Karajannis MA, Shido K, Hooper AT, Rashbaum WK, Pytowski B, Wu Y, Hicklin DJ, Zhu Z, et al: Fetal stromal-dependent paracrine and intracrine vascular endothelial growth factor-a/vascular endothelial growth factor receptor-1 signaling promotes proliferation and motility of human primary myeloma cells. Cancer Res. 65:3185–3192. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Wang ES, Teruya-Feldstein J, Wu Y, Zhu Z, Hicklin DJ and Moore MA: Targeting autocrine and paracrine VEGF receptor pathways inhibits human lymphoma xenografts in vivo. Blood. 104:2893–2902. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Boiocchi L, Vener C, Savi F, Bonoldi E, Moro A, Fracchiolla NS, Iurlo A, Deliliers GL, Coggi G, Bosari S and Gianelli U: Increased expression of vascular endothelial growth factor receptor 1 correlates with VEGF and microvessel density in Philadelphia chromosomenegative myeloproliferative neoplasms. J Clin Pathol. 64:226–231. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Bellamy WT, Richter L, Sirjani D, Roxas C, Glinsmann-Gibson B, Frutiger Y, Grogan TM and List AF: Vascular endothelial cell growth factor is an autocrine promoter of abnormal localized immature myeloid precursors and leukemia progenitor formation in myelodysplastic syndromes. Blood. 97:1427–1434. 2001. View Article : Google Scholar : PubMed/NCBI

15 

Wimazal F, Krauth MT, Vales A, Böhm A, Agis H, Sonneck K, Aichberger KJ, Mayerhofer M, Simonitsch-Klupp I, Müllauer L, et al: Immunohistochemical detection of vascular endothelial growth factor (VEGF) in the bone marrow in patients with myelodysplastic syndromes: Correlation between VEGF expression and the FAB category. Leuk Lymphoma. 47:451–460. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Aguayo A, Kantarjian HM, Estey EH, Giles FJ, Verstovsek S, Manshouri T, Gidel C, O'Brien S, Keating MJ and Albitar M: Plasma vascular endothelial growth factor levels have prognostic significance in patients with acute myeloid leukemia but not in patients with myelodysplastic syndromes. Cancer. 95:1923–1930. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Verstovsek S, Estey E, Manshouri T, Giles FJ, Cortes J, Beran M, Rogers A, Keating M, Kantarjian H and Albitar M: Clinical relevance of vascular endothelial growth factor receptors 1 and 2 in acute myeloid leukaemia and myelodysplastic syndrome. Br J Haematol. 118:151–156. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Su JL, Yen CJ, Chen PS, Chuang SE, Hong CC, Kuo IH, Chen HY, Hung MC and Kuo ML: The role of the VEGF-C/VEGFR-3 axis in cancer progression. Br J Cancer. 96:541–545. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H and Thiele J: WHO Classification of Tumors of Haematopoietic and Lymphoid Tissues. Revised 4th edition. IARC; Lyon: 2017

20 

Greenberg PL, Tuechler H, Schanz J, Sanz G, Garcia-Manero G, Solé F, Bennett JM, Bowen D, Fenaux P, Dreyfus F, et al: Revised international prognostic scoring system for myelodysplastic syndromes. Blood. 120:2454–2465. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Shaffer LG, Slovak ML and Campbell LJ: ISCN 2009: An international system for human cytogenetic nomenclature (2009). Basel, Switzerland: S. Karger; 2009

22 

Kalitin NN and Buravtsova IV: Transcription factor RARa expression correlates with VEGFR3-dependent signaling system components expression in multiple myeloma. Klinicheskaya Onkogematologiya. 8:31–36. 2015.(In Russian).

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Dor Y, Porat R and Keshet E: Vascular endothelial growth factor and vascular adjustments to perturbations in oxygen homeostasis. Am J Physiol Cell Physiol. 280:C1367–C1374. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G, Sanz M, Vallespi T, Hamblin T, Oscier D, et al: International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 89:2079–2088. 1997. View Article : Google Scholar : PubMed/NCBI

26 

Wimazal F, Sperr WR, Kundi M, Meidlinger P, Fonatsch C, Jordan JH, Thalhammer-Scherrer R, Schwarzinger I, Geissler K, Lechner K and Valent P: Prognostic value of lactate dehydrogenase activity in myelodysplastic syndromes. Leuk Res. 25:287–294. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Naqvi K, Jabbour E, Bueso-Ramos C, Pierce S, Borthakur G, Estrov Z, Ravandi F, Faderl S, Kantarjian H and Garcia-Manero G: Implications of discrepancy in morphologic diagnosis of myelodysplastic syndrome between referral and tertiary care centers. Blood. 118:4690–4693. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Mufti GJ, Bennett JM, Goasguen J, Bain BJ, Baumann I, Brunning R, Cazzola M, Fenaux P, Germing U, Hellström-Lindberg E, et al: Diagnosis and classification of myelodysplastic syndrome: International working group on morphology of myelodysplastic syndrome (IWGM-MDS) consensus proposals for the definition and enumeration of myeloblasts and ring sideroblasts. Haematologica. 93:1712–1717. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Montalban-Bravo G and Garcia-Manero G: Myelodysplastic syndromes: 2018 Update on diagnosis, risk-stratification and management. Am J Hematol. 93:129–147. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Ohta Y, Endo Y, Tanaka M, Shimizu J, Oda M, Hayashi Y, Watanabe Y and Sasaki T: Significance of vascular endothelial growth factor messenger RNA expression in primary lung cancer. Clin Cancer Res. 2:1411–1416. 1996.PubMed/NCBI

31 

Fontanini G, Bigini D, Vignati S, Basolo F, Mussi A, Lucchi M, Chine S, Angeletti CA, Harris AL and Bevilacqua G: Microvessel count predicts metastatic disease and survival in non-small cell lung cancer. J Pathol. 177:57–63. 1995. View Article : Google Scholar : PubMed/NCBI

32 

Meitar D, Crawford SE, Rademaker AW and Cohn SL: Tumor angiogenesis correlates with metastatic disease, N-myc amplification, and poor outcome in human neuroblastoma. J Clin Oncol. 14:405–414. 1996. View Article : Google Scholar : PubMed/NCBI

33 

O'Brien T, Cranston D, Fuggle S, Bicknell R and Harris AL: Different angiogenic pathways characterize superficial and invasive bladder cancer. Cancer Res. 55:510–513. 1995.PubMed/NCBI

34 

Takahashi Y, Kitadai Y, Bucana CD, Cleary KR and Ellis LM: Expression of vascular endothelial growth factor and its receptor, KDR, correlates with vascularity, metastasis, and proliferation of human colon cancer. Cancer Res. 55:3964–3968. 1995.PubMed/NCBI

35 

Anan K, Morisaki T, Katano M, Ikubo A, Kitsuki H, Uchiyama A, Kuroki S, Tanaka M and Torisu M: Vascular endothelial growth factor and platelet-derived growth factor are potential angiogenic and metastatic factors in human breast cancer. Surgery. 119:333–339. 1996. View Article : Google Scholar : PubMed/NCBI

36 

Pruneri G, Bertolini F, Soligo D, Carboni N, Cortelezzi A, Ferrucci PF, Buffa R, Lambertenghi-Deliliers G and Pezzella F: Angiogenesis in myelodysplastic syndromes. Br J Cancer. 81:1398–1401. 1999. View Article : Google Scholar : PubMed/NCBI

37 

Legros L, Slama B, Karsenti JM, Vey N, Natarajan-Amé S, Watel E, Richard B, Bouabdallah K, Mannone L, Benchetrit M, et al: Treatment of myelodysplastic syndromes with excess of blasts by bevacizumab is well tolerated and is associated with a decrease of VEGF plasma level. Ann Hematol. 91:39–46. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Giles FJ, Stopeck AT, Silverman LR, Lancet JE, Cooper MA, Hannah AL, Cherrington JM, O'Farrell AM, Yuen HA, Louie SG, et al: SU5416, a small molecule tyrosine kinase receptor inhibitor, has biologic activity in patients with refractory acute myeloid leukemia or myelodysplastic syndromes. Blood. 102:795–801. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Giles FJ, Bellamy WT, Estrov Z, O'Brien SM, Verstovsek S, Ravandi F, Beran M, Bycott P, Pithavala Y, Steinfeldt H, et al: The anti-angiogenesis agent, AG-013736, has minimal activity in elderly patients with poor prognosis acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Leuk Res. 30:801–811. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Gupta P, Mulkey F, Hasserjian RP, Sanford BL, Vij R, Hurd DD, Odenike OM, Bloomfield CD, Owzar K, Stone RM, et al: A phase II study of the oral VEGF receptor tyrosine kinase inhibitor vatalanib (PTK787/ZK222584) in myelodysplastic syndrome: Cancer and leukemia group B study 10105 (alliance). Invest New Drugs. 31:1311–1320. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Kirschbaum MH, Frankel P, Synold TW, Zain J, Claxton D, Tuscano J, Newman EM, Gandara DR and Lara PN Jr: A phase II study of vascular endothelial growth factor trap (Aflibercept, NSC 724770) in patients with myelodysplastic syndrome: A California cancer consortium study. Br J Haematol. 180:445–448. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Cheng CL, Hou HA, Jhuang JY, Lin CW, Chen CY, Tang JL, Chou WC, Tseng MH, Yao M, Huang SY, et al: High bone marrow angiopoietin-1 expression is an independent poor prognostic factor for survival in patients with myelodysplastic syndromes. Br J Cancer. 105:975–982. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Shalaby F, Rossant J, Yamaguchi T, Gertsenstein M, Wu XF, Breitman ML and Schuh AC: Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature. 376:62–66. 1995. View Article : Google Scholar : PubMed/NCBI

44 

Eichmann A, Corbel C, Nataf V, Vaigot P, Bréant C and Le Douarin NM: Ligand-dependent development of the endothelial and hemopoietic lineages from embryonic mesodermal cells expressing vascular endothelial growth factor receptor 2. Proc Natl Acad Sci USA. 94:5141–5146. 1997. View Article : Google Scholar : PubMed/NCBI

45 

Carrillo de Santa PE, Arias FC, Caso Peláez E, Muñoz Molina GM, Sánchez Hernández I, Muguruza Trueba I, Moreno Balsalobre R, Sacristán López S, Gómez Pinillos A and del Val Toledo Lobo M: Prognostic significance of the expression of vascular endothelial growth factors A, B, C, and D and their receptors R1, R2, and R3 in patients with non-small cell lung cancer. Cancer. 115:1701–1712. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Ock CY, Nam AR, Bang JH, Kim TY, Lee KH, Han SW, Im SA, Kim TY, Bang YJ and Oh DY: The distinct signatures of VEGF and soluble VEGFR2 increase prognostic implication in gastric cancer. Am J Cancer Res. 5:3376–3388. 2015.PubMed/NCBI

47 

Dhakal HP, Naume B, Synnestvedt M, Borgen E, Kaaresen R, Schlichting E, Wiedswang G, Bassarova A, Holm R, Giercksky KE and Nesland JM: Expression of vascular endothelial growth factor and vascular endothelial growth factor receptors 1 and 2 in invasive breast carcinoma: Prognostic significance and relationship with markers for aggressiveness. Histopathology. 61:350–364. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Kalitin NN, Dudina GA, Semochkin SV and Karamysheva AF: Analysis of VEGF-A/VEGFR1/VEGFR2 genes expression analyses in patients with myelodysplastic syndromes. Ther Arch. 89:39–44. 2017.(In Russian). View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2023
Volume 25 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kalitin N, Dudina G, Kostritsa N, Sivirinova A and Karamysheva A: Evaluation of <em>VEGF</em> and <em>VEGFR</em> gene expression as prognostic markers in low and intermediate‑1 risk patients with myelodysplastic syndromes. Oncol Lett 25: 95, 2023
APA
Kalitin, N., Dudina, G., Kostritsa, N., Sivirinova, A., & Karamysheva, A. (2023). Evaluation of <em>VEGF</em> and <em>VEGFR</em> gene expression as prognostic markers in low and intermediate‑1 risk patients with myelodysplastic syndromes. Oncology Letters, 25, 95. https://doi.org/10.3892/ol.2023.13681
MLA
Kalitin, N., Dudina, G., Kostritsa, N., Sivirinova, A., Karamysheva, A."Evaluation of <em>VEGF</em> and <em>VEGFR</em> gene expression as prognostic markers in low and intermediate‑1 risk patients with myelodysplastic syndromes". Oncology Letters 25.3 (2023): 95.
Chicago
Kalitin, N., Dudina, G., Kostritsa, N., Sivirinova, A., Karamysheva, A."Evaluation of <em>VEGF</em> and <em>VEGFR</em> gene expression as prognostic markers in low and intermediate‑1 risk patients with myelodysplastic syndromes". Oncology Letters 25, no. 3 (2023): 95. https://doi.org/10.3892/ol.2023.13681