Open Access

Exploring the anticancer potential of hydrogen sulfide and BAY‑876 on clear cell renal cell carcinoma cells: Uncovering novel mutations in VHL and KDR genes among ccRCC patients

  • Authors:
    • Peshraw Salih Hamadamin
    • Kalthum Asaf Maulood
  • View Affiliations

  • Published online on: January 18, 2024     https://doi.org/10.3892/mco.2024.2719
  • Article Number: 21
  • Copyright: © Hamadamin 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

The aim of the present study was to determine the cytotoxic effect of BAY‑876 and NaSH alone or in combination with sunitinib against the 786‑O cell line (renal adenocarcinoma). The IC50 of sunitinib, BAY‑876 and NaSH were estimated. Cells were cultured in a 96‑well plate and then different concentration of each drug alone was exposed for different incubation time; afterwards, cell cytotoxicity was measured using Cell Counting Kit‑8 kit. The IC50 for each drug was used in next experiment to determine the influence of drug combinations. Furthermore, to observe the effect of mutations of few driver genes in development of clear cell renal cell carcinoma (ccRCC), direct sanger sequencing was used to find single nucleotide polymorphisms in exon 1 and exon 13 of tumor suppressor gene Von Hippel Lindau (VHL) and kinase insert domain receptor (KDR) genes respectively in ccRCC formalin fixed paraffin embedded block samples. The results revealed that the IC50 for sunitinib (after 72 h), BAY‑876 (after 96 h) and NaSH (after 48 h) was 5.26, 53.56 and 692 µM respectively. The cytotoxic effect of sunitinib and BAY‑876, sunitinib and NaSH combinations after 24‑ and 48‑h incubation respectively was significantly higher (P<0.05) compared with the control group as well as to sunitinib group alone. These results proved that each of BAY‑876 and NaSH have anticancer effect; thus, they could be used in future for ccRCC treatment purpose. Furthermore, direct sequencing results demonstrated unrecorded mutations of VHL and KDR genes is 43.7 and 31.5% of cases respectively. These findings confirmed the leading role of VHL gene in development of ccRCC and the crucial role of KDR gene in angiogenesis and drug resistance.

Introduction

Renal cancer accounts for 5% of all cancer and ranks as the sixth most prevalent type of cancer in male patients. In female patients, it contributes to 3% of malignancies and stands as the tenth most popular form of tumor (1,2). Renal cell carcinoma (RCC) accounts for >90% of kidney tumors (2,3). Depending on population surveys, a 5-year survival rate was observed in 70% of patients diagnosed with regional cancer. However, for those with metastatic RCC, this rate drops down to 13% (4). Clear cell RCC (ccRCC) is the most common histological subtype of RCC, accounting for 75% of cases, which develops in the epithelium of kidney tubules and can metastasize to different organs (5).

The inactivation of the tumor suppressor gene Von Hippel Lindau (VHL) has a key role in the pathogenesis of clear cell carcinoma (6). It was demonstrated that >90% of patients with ccRCC experience a loss of VHL heterozygosity and inactivating mutations of VHL are detected in 50-65% of instances (7,8). Reducing the level of VHL proteins directly promotes an increase in the expression of both hypoxia-inducible factors (HIFs) including HIF-1A and HIF-2A (9). These factors activate various target genes in the tumor microenvironment which are stimulating tumor cell survival, cell proliferation, angiogenesis, metabolism of sugar and tumor metastasis (10-12). Targeting HIFs or downstream effector molecules in the VHL/HIF pathway (such as VEGFs) has been used for three decades in the treatment of disease outcomes in advanced patients with ccRCC (13).

Sunitinib is one of the earliest approved multi-targeted tyrosine kinase inhibitors (TKIs) that have been extensively used as first-line treatment for metastatic ccRCC (14). TKIs act by inhibiting the kinase activity of various receptors and are still used in therapy (15). Notably, despite its demonstrated efficacy in treating advanced RCC, a substantial proportion of patients develop primary resistance or acquired resistance to sunitinib within 6-15 months of treatment (16). BAY-876 was previously identified as a new-generation inhibitor of glucose transporter 1 (GLUT1) in ovarian cancer (17); however, since its discovery, there have been no studies investigating the effect of BAY-876 as GLUTI in ccRCC.

Hydrogen sulfide (H2S) is a gaseous transmitter found in mammalian tissues; along with nitric oxide (NO) and carbon monoxide (CO), it has a variety of biological functions (18,19). Other studies demonstrated that H2S gas has a dual effect on cancerous diseases, acting as both a pro- and antitumor agent (20,21). Sodium hydrosulfide (NaSH) salt is a solid analog of H2S gas that provides immediate and direct access to biologically significant forms of sulfide. NaSH has been used extensively as an exogenous delivery method for H2S to evaluate its therapeutic potential (22). To the best of the authors' knowledge, the effects of NaSH and BAY-876 on RCC remain unclear and a significant number of patients with ccRCC have demonstrated a lack of response, recurrence, or resistance to sunitinib. For these reasons, the present study aimed to investigate the effects of NaSH and BAY-876 alone and in combination with sunitinib on ccRCC cells. The evaluation of genotypic variations and polymorphisms in the VHL and kinase insert domain receptor (KDR) genes in patients with ccRCC was another aim of the present study.

Materials and methods

Cell line

The human metastatic ccRCC cell line (786-O) was obtained from the American Type Culture Collection (cat. no. CRL-1932). The cells were cultured in RPMI-1640 medium (Thermo Fisher Scientific, Inc.; cat. no. 31800089) which was supplemented with 10% fetal bovine serum (FBS) and 1% of antibiotic (consisting of 10,000-unit penicillin, 10 mg streptomycin and 25 µg amphotericin; Sigma Aldrich; Merck KGaA; cat. no. A5955). Cells were split every 2-3 days to maintain their density and maintained at 37˚C in a humidified incubator with 5% CO2 and 95% air (RS Biotech Galaxy Model R+). Following an adaptation step of three passages, the experiments were conducted within passages 11 to 16 of 786-O cells (23).

Determining IC50

To determine the IC50 of sunitinib (MedChemExpress; cat. no. HY-10255A), BAY-876 (MedChemExpress; cat. no. HY-100017) and NaSH on 786-O viability, cells were seeded in a 96-well culture plate (4x103 cell/well) and incubated at 37˚C in a humidified incubator with 5% CO2. After 24 h, the cells were treated with various concentrations of sunitinib (range, 1-100 µM), BAY-876 (range, 10-1,000 µM) and NaSH (range, 10-103 µM) in quadruplicate for each concentration. The plates were incubated for 24, 48 and 72 h at 37˚C with 5% CO2. For BAY-876 another plate was incubated for 96 h. After each incubation interval, the culture media of the plate wells were changed with new media and then the cell cytotoxicity was measured by using Cell Counting Kit-8 (CCK-8; MedChemExpress; cat. no. HY-K0301) (24) according to the manufacturer's instruction. Briefly, 10 µM of the CCK-8 reagent was added to each well, incubated for 2-4 h at 37˚C and then the absorbance was measured at 540 nm by using an ELIZA reader (BioTek Instruments, Inc.). Each experiment was carried out three times.

Drug combinations

The IC50 of sunitinib after 72 h (5.26x10-6 M), BAY-876 after 96 h (53.56x10-6 M) and NaSH after 72 h (692x10-6 M) were used to determine the cytotoxicity of the combination of drugs against 786-O cells. Briefly, 4x103 cells/well were seeded in 96 well plates and incubated at 37˚C with 5% CO2. After 24 h incubation, the cells were exposed to the IC50 of each drug alone and a combination of sunitinib with BAY-876, sunitinib with NaSH and BAY-876 with NaSH. Following incubation for 24 and 48 h at 37˚C and 5% CO2, cell cytotoxicity was measured by using the CCK-8 kit.

Statistical analysis

Data analysis was conducted via GraphPad Prism 9 Software (Dotmatics). Non-linear regression was employed to establish the IC50 of medications, while ordinary one-way ANOVA with Tukey's multiple comparisons test was performed to assess the statistically significant differences in the anticancer activity between various agents and untreated cells. The data were reported as the mean value ± standard error (SE) of the difference. P<0.05 was considered to indicate a statistically significant difference.

Formalin-fixed paraffin-embedded tissues of patients with ccRCC

A total of 30 formalin-fixed paraffin-embedded tissues (FFPE) of patients with ccRCC were collected form PAR and Rzgary teaching hospitals (Erbil, Iraq) between July 2022 and May 2023. After histological confirmation of ccRCC through hematoxylin and eosin (H&E) staining, the blocks were collected for extraction of DNA. The approval (approval no. 45/90) of the present study was granted by the Human Ethics Committee of the College of Education of Salahaddin University-Erbil (Erbil, Iraq). The study was conducted according to the criteria set by The Declaration of Helsinki (October 2013) and signed informed consent forms were obtained from the participants approving the use of their tissues for investigation. DNA samples were extracted from the FFPE tissues. Clinicopathological characteristics of patients (including sex and age distribution) are presented in Table I.

Table I

Clinicopathological characteristics of patients with clear cell renal cell carcinoma.

Table I

Clinicopathological characteristics of patients with clear cell renal cell carcinoma.

Clinicopathological characteristicsPercentage (%)
Sex 
     Male44.40
     Female56.60
Age, years 
     >5050
     51-6020
     61-7020
     <7110
Degree of differentiation 
     Well-differentiated tumor43.33
     Moderately differentiated tumor50
     Poorly differentiated6.66
TNM staging 
     Stage I 
          pT1a N0 Mx50
          pT1b N0 Mx26.60
     Stage II 
          pT2 N0 Mx3.33
     Stage III 
          pT1b N1 Mx6.33
          pT3a Nx Mx3.66
          pT3a N0 Mx6.33
          pT3 N1 Mx3.33
Stage assessment of ccRCC

The TNM cancer staging system provides significant predictors of prognosis in RCC. The present study used the eighth edition (TNM8) of the American Joint Committee on Cancer (AJCC) and the Union for International Cancer Control (UICC) for ccRCC tumor staging (25,26). In TNM, the T refers to the size and regional extension of the primary tumor, the N refers to the tumor involvement in the regional lymph nodes and the M refers to metastasis or spread of the tumor to other parts of the body away from the original location (27).

DNA extraction from FFPE tissues

Genomic DNA extraction from FFPE was performed using the QIAamp DNA FFPE Tissue kit (Qiagen GmbH; cat. no. 56404) according to the manufacturer's protocol. Briefly, 5-µm thick ribbon sections were created from the paraffin blocks, followed by deparaffination by xylene. The cells were lysed with lysis buffer and proteinase k. The DNA bound to the membrane and the contaminants were flowing through the membrane. Finally, pure DNA was eluted from the membrane with an elution buffer. The eluted DNA was quantified based on the absorbance (A) measured at 260 nm (A260) and purity was estimated based on the A260/A280 ratio using a Nanodrop (Thermo Fisher Scientific, Inc.). An elution buffer was used as a blank for the Nanodrop instrument. The assessment of DNA purity relied on an ideal A260/A280 ratio of 1.8(28).

Genotype determination

The current study examined variants of each VHL and KDR gene. Based on the results of a previous study, exon 1 of VHL (29) and exon 13 of KDR genes were chosen for the current study. Initially, the purified DNA underwent an independent process of amplification. For this purpose, a pre-prepared master mix (including dNTP, MgCl2, Taq DNA polymerase and PCR reaction buffer; Promega Corporation) and the following primer pairs were used: VHL forward, 5'-GTCTGGATCGCGGAGGGAAT-3' and reverse, 5'-GGACTGCGATTGCAGAAGAT-3'; KDR forward, 5'-CGTGTCTTTGTGGTGCACTG-3' and reverse, 5'-CTAGGACTGTGAGCTGCCTG-3'. The thermocycling conditions used for PCR were: Initial denaturation at 95˚C for 5 min; 35 cycles of denaturation at 95˚C for 30 sec, annealing at 61˚C for 30 sec and extension at 72˚C for 1 min; final elongation at 72˚C for 6 min and the samples were maintained at 4˚C.

The PCR products were separated by 1.5% agarose gel electrophoresis. DNA ladder of 100 base pairs (GeneDireX, Inc.; cat. no. DM003-R500) was used for band composition as shown in Fig. S1. Before pouring it into the tray, the gel was mixed with Safe gel stain Dye (ADD BIO) and then it was visualized using the UV Transilluminator UST-20M-8K (Biostep GmbH). Following the PCR protocol, the products were delivered to the cytogenetic laboratory-Zheen International Hospital-Erbil-Iraq for sequencing performed on the Automatic 3130 Genetic Analyzer (Applied Biosystems; Thermo Fisher Scientific, Inc.) using the same forward primers for each exon in the genes. The sequencing data were deposited into the GenBank database (https://www.ncbi.nlm.nih.gov/genbank/; accession no. OR294044 and OR339875). Analysis and interpretation of the Sanger sequencing data were performed using the Mutation Surveyor software package 5.1 (SoftGenetics, LLC) by comparing the sequencing results with reference genes in public databases, including ClinVar, dbSNP, gnomAD and COSMIC (30).

Database's gene mutation retrieval and gene interactions

Genome aggregation database (gnomAD) version 3.1.2 (https://gnomad.broadinstitute.org) was used for mutation retrieval of selected genes. This population referencing database currently is one of the most powerful tools that provides information about genetic variation and gene interpretation found in the human population. This resource is developed by worldwide investigators who collect and integrate both genome and exome data from massive sequencing programs. The database nowadays is widely used in clinical genetics and genomic research (31).

All recorded gene mutations of chosen genes (VHL and KDR) in RCC were retrieved from both the Mutagene database (https://www.ncbi.nlm.nih.gov/research/mutagene) and Catalog of Somatic Mutation of Cancer (COSMIC v98, released 23-MAY-23; https://cancer.sanger.ac.uk/cosmic). The Mutagene database contains 441 RRC samples with 33,063 gene mutations. Based on Bscore thresholds, Mutagene is used to determine the cancer driver, potential cancer driver and passenger mutations of selected genes. COSMIC, which is readily available to researchers globally, is a thorough database encompassing somatic mutations in cancer. It provides extensive information about mutation location, frequency and impact, along with clinical and pathological data. By utilizing this database, researchers can identify prevalent mutations and other genetic changes that potentially play a role in the development or advancement of cancer. Moreover, these findings may offer potential targets for therapeutic interventions (32).

In addition, the interaction of VHL and KDR genes in RCC is predicted through the GeneMANIA anticipation tool (https://genemania.org/). Based on genetic connection, physical protein-protein interaction and co-expression pattern, this public online database can provide gene interaction. By examining the anticipated associations among these two genes, valuable insights into their participation in cellular processes and disease pathways can be gained. This, in turn, could pave the way for the identification of fresh therapeutic targets or diagnostic biomarkers. GeneMANIA through anticipation of gene interaction will lead to determining the function of genes, uncovering gene interaction in specific cellular assays (33) and identifying potential gene candidates for further study.

Results

Anticancer activity of sunitinib, BAY-876 and NaSH

To determine the in vitro cytotoxic effect of sunitinib, BAY-876 and NaSH, dose-dependent viability studies were performed to find the appropriate concentration of each drug that can eliminate 50% (IC50) of 786-O cells by using the CCK-8 kit. The IC50s for sunitinib were found to be 9.85x106, 9.1x10-6 and 5.26x10-6 M at 24, 48 and 72 h, respectively (Fig. 1A), with no significant difference among the measured timepoints. The IC50 for BAY-876 was reached only after 4 days of treatment and it was 53.56x10-6 M (Fig. 1B). Furthermore, the present study also revealed that the IC50 for NaSH as an exogenous donor of H2S after 24 and 48 h of treatment were equal to 726x10-6 and 692x10-6 M, respectively (Fig. 1C), with no significant difference between the two timepoints.

In addition, the cytotoxic effect against 786-O cells of several drug combinations was assessed. Sunitinib IC50 after 3 days, BAY-IC50 after 4 days and NaSH after 2 days were used alone and in combination for 24 (Fig. 2) and 48 h (Fig. 3). The results after 24 h demonstrated that there were no significant differences in cytotoxicity between sunitinib and BAY-876 treatments alone compared with the control group; however, their combination demonstrated statistically different cytotoxicity (P<0.001) compared with the control group and each drug used individually.

Regarding the cytotoxic effect of NaSH, NaSH alone and in combination with either sunitinib or BAY-876 revealed a statistically significant increase (P<0.001) in cytotoxicity compared with the control group. The cytotoxicity of NaSH in combination with BAY-876 was significantly higher (P<0.05) compared with NaSH alone. However, there was no significant difference between NaSH and sunitinib co-treatment and NaSH.

The outcomes of the drug combinations at 24 and 48 h were comparable and the only exception was that there was a statistically significant difference (P<0.05) between the NaSH and sunitinib co-treatment and NaSH alone in reducing cell viability.

Patient characteristics

In the current study, out of 30 patients with ccRCC, 17 were male (56.6%) and 13 were female (44.40%). Regarding the patient age, 50 (50%) were under 50 years, 6 (20%) were between 51 and 60 years, 6 (20%) were between 61 and 70 years, and 3 (10%) were >71 years old. The degree of cell differentiation varied among patients with ccRCC. A large number of patients were grouped as medium-differentiated (50%) and well-differentiated carcinomas (43.33%), while poorly-differentiated carcinomas accounted only for 6.66% (Table I).

Mutation analysis

Single nucleotide polymorphism (SNP) was recorded in 47.7% of the VHL gene and 31.5% of the KDR gene (data not shown). In exon 1 of the VHL gene of ccRCC, 36 SNP mutations were identified, including 35 substitution mutations (A>AT, C>CT, A>AG, G>GA, T>TC, C>CA, A>T) and one deletion mutation (delA). Both heterozygous (n=35) and homozygous (n=1) variant mutations were identified as demonstrated in Fig. 4. In total, 35 variant mutations were previously not recorded in external databases and 30 of them had their sequences of amino acids changed (Table II). The single homozygous substitution variant (946A>T) accounts for the greatest percentage (40%) among all other variants; however, among heterozygous variants, two heterozygous variants (778A>AT and 793A>AT) constitute the second greatest percentage (33.30%) among other mutation of VHL gene.

Table II

Variants identified in VHL and KDR genes in ccRCC patients analyzed with mutation DNA variant analysis.

Table II

Variants identified in VHL and KDR genes in ccRCC patients analyzed with mutation DNA variant analysis.

 Chromosome positionMutationMutation enotype Heterozygous/HomozygousVariantsVariant percentage (%)Amino acid changeExternal database
VHL gene
13:10183764SubstitutionA>THomozygous946A>T40.00 Asparagine/IsoleucineNot found
23:10183623SubstitutionA>ATHeterozygous805A>AT30.00Glutamic acid/ValineNot found
33:10183626SubstitutionA>ATHeterozygous808A>AT30.00Glutamic acid/ValineNot found
43:10183638SubstitutionA>ATHeterozygous820A>AT30.00Glutamic acid/ValineNot found
53:10183641SubstitutionA>ATHeterozygous823A>AT30.00Glutamic acid/ValineNot found
63:10183656SubstitutionA>ATHeterozygous838A>AT30.00Glutamic acid/MethionineNot found
73:10183671SubstitutionA>ATHeterozygous853A>AT30.00Glutamic acid/ValineNot found
83:10183673SubstitutionC>CAHeterozygous855C>CA10.00 Lucine/MethionineNot found
93:10183686SubstitutionA>ATHeterozygous868A>AT30.00Glutamic acid/ValineNot found
103:10183730SubstitutionA>ATHeterozygous912A>AT18.20 Asparagine/TyrosineNot found
113:10183570SubstitutionA>ATHeterozygous752A>AT25noneNot found
123:10183578SubstitutionA>ATHeterozygous760A>AT25Glutamic acid/ValineNot found
133:10183581SubstitutionA>ATHeterozygous763A>AT25Glutamic acid/ValineNot found
143:10183582SubstitutionG>GAHeterozygous764G>GA25Glutamic acid/ValineNot found
153:10183596SubstitutionA>ATHeterozygous778A>AT33.30Glutamic acid/ValineNot found
163:10183597SubstitutionA>ATHeterozygous793A>AT33.30Glutamic acid/ValineNot found
173:10183612SubstitutionA>AGHeterozygous794A>AG22.20Glutamic acid/ValineNot found
183:10183635SubstitutionC>CTHeterozygous817C>CT30.00Alanine/ValineNot found
193:10183655SubstitutionG>GAHeterozygous837G>GA10Glutamic acid/MethionineNot found
203:10183611SubstitutionA>ATHeterozygous793A>AT33.30Glutamic acid/ValineNot found
213:10183614SubstitutionA>ATHeterozygous796A>AT11.10Aspartic acid/ValineNot found
223:10183764DeletionA 946delA25noneNot found
233:10183635SubstitutionC>CTHeterozygous817C>CT30.00Aspartic acid/ValineNot found
243:10183579SubstitutionG>GAHeterozygous761G>GA12.50Glutamic acid/ValineNot found
253:10183608SubstitutionA>ATHeterozygous790A>AT11.10Glutamic acid/ValineNot found
263:10183624SubstitutionG>GAHeterozygous806G>GA10.00Glutamic acid/ValineNot found
273:10183740SubstitutionA>ATHeterozygous922A>AT16.70Glutamic acid/ValineNot found
283:10183672SubstitutionA>AGHeterozygous854A>AG9.10Glutamic acid/ValineNot found
293:10183762SubstitutionC>CTHeterozygous944C>CT10noneNot found
303:10183763SubstitutionA>AGHeterozygous945A>AG12.50 Isoleucine/ValineNot found
313:10183691SubstitutionA>ATHeterozygous873A>AT9.10 Methionine/LeucineNot found
323:10183753SubstitutionC>CTHeterozygous935C>CT8.30noneNot found
333:10183759SubstitutionC>CTHeterozygous941C>CT9.10noneNot found
343:10183756SubstitutionC>CTHeterozygous938C>CT8.30noneNot found
353:10183719SubstitutionT>TCHeterozygous901T>TC9.10 Leucine/ProlineNot found
363:10183764SubstitutionA>AGHeterozygous946A>AG12.50 Asparagine/serinefound
KDR gene
14:55971029SubstitutionC>CGHeterozygous21234C>CG28.00 Proline/AlanineNot found
24:55970820SubstitutionC>CTHeterozygous21443C>CT8.30NoneNot found
34:55970985SubstitutionG>GTHeterozygous21278G>GT16.70 Lysin/AsparagineNot found
44:55970981SubstitutionT>TCHeterozygous21282T>TC12.50 Leucine/ProlineNot found
54:55970980SubstitutionT>TCHeterozygous21283T>TC12.50 Leucine/ProlineNot found
64:55970991SubstitutionT>TCHeterozygous21272T>TC16.70NoneNot found
74:55970969SubstitutionT>TGHeterozygous21294T>TG20 Tryptophane/GlycineNot found
84:55970819SubstitutionA>ACHeterozygous21444A>AC8.30 Threonine/ProlineNot found
94:55971034SubstitutionC>CTHeterozygous21229C>CT25.30 Proline/LeucineNot found
104:55971000SubstitutionC>CTHeterozygous21263C>CT25.10NoneNot found
114:55970978SubstitutionG>GCHeterozygous21285G>GC12.50Aspartic acid/HistidineNot found
124:55970970SubstitutionT>TCHeterozygous21293T>TC10.00NoneNot found
134:55970996SubstitutionC>CGHeterozygous21267C>CG25.00Proline/Aspartic acidNot found
144:55970995SubstitutionC>CAHeterozygous21268C>CA25.00Proline/Aspartic acidNot found
154:55970892SubstitutionG>GAHeterozygous21371G>GA7.10NoneNot found
164:55970925SubstitutionT>TGHeterozygous21338T>TG7.10Aspartic acid/HistidineNot found

Regarding to KDR gene, 16 heterozygous substitution variant mutations (C>CG, C>CT, G>GT, T>TC, A>AC, G>GC, C>CA, G>GA) were recorded in exon 13; all of them were not recorded previously in external databases. A total of 11 of these mutations are associated with changes in the sequence of amino acids. Furthermore, the heterozygous variant (21234C>CG) constitutes the largest percentage (28%) among other variants (Table II).

Database mutations and gene interactions

The GnomAD tool was used to retrieve the entire recorded mutations of VHL and KDR genes in all types of cancer. VHL had 386 mutations, while KDR had 1,595 mutations as presented in Table SI.

Additional analysis of retrieved VHL and KDR gene mutations in RCC was performed using Metagene (all genome-wide studies in ICGC) and COSMIC databases as demonstrated in Tables SII and SIII. The number of mutations in VHL was 125 and 1,569 in the Metagene and COSMIC databases, respectively; however, the number of mutations of the KDR gene was 229 and 27 in the same two databases, respectively. In the Mutagen database the most common type of mutation in the VHL gene of patients with RCC was missense mutation followed by nonsense mutation. Out of 125 mutations, 66 were passengers followed by 34 drivers and 25 potential driver mutations. Regarding the KDR gene, the most common type of mutation was missense followed by silent mutation, and their status in cancer participation was mostly passenger (n=179) followed by potential driver (n=34) and driver (n=16) mutations. In the COSMIC database, the frameshift deletion (n=530) was the most common type of mutation of VHL in patients with ccRCC followed by missense-substitution (n=339), frameshift insertion (n=219) and nonsense substitution (n=62). However, missense substitutions (n=20) were common types of mutations of the KDR gene in patients with ccRCC followed by frameshift deletions (n=3), nonsense substitution (n=2) and substitution-coding silent (n=2).

Moreover, data from the GeneMANIA predicting tool revealed that 13 genes (RBX1, VHLL, CUL2, VEGFA, EPAS1, ELOC, PAX1, ELOB, HIF1, JADE1, EGLN1, EGLN2 and EGLN3) and 12 genes (CDH5, PDCD6, VEGFA, VEGFC, VEGFD, EPAS1, GREM1, HIF1, JADE1, NRP1, EGLN3 and SH2D2A) are associated with VHL and KDR genes, respectively, mainly through physical interaction then co-expression, prediction, colocalization, genetic interaction, pathway and minimum level by sharing protein domain (Fig. 5 and Table SIV). A total of five genes (VEGFA, EPAS1, HIF1, JADE1 and EGLN3) are shared between VHL and KDR genes, and this is considered a close relation between these two genes. Furthermore, the KDR gene showed genetic interaction with most of the genes having a role in angiogenesis (Fig. 6).

Discussion

In the present study, the sunitinib IC50 after 24 and 48 h was ~9x10-6 M, whereas after 72 h it dropped to 5.26x10-6M. These results are very similar to previous results demonstrating the cytotoxicity of sunitinib against RCC cell lines. Sato et al (34) identified that the IC50 value of sunitinib against 786-O cells (3,000/well) was 5 µM after a 48-h incubation. This disparity in the results might be attributed to the initial cell count used in the experiment as the current study involved plating 4,000 cells in each well of a 96-well plate, which could have affected the observed IC50 value due to variations in cell density. Another study reported that sunitinib IC50 was 3.99 µM against Caki-1 cells after 48 h (35). In this case, the discrepancy with the present results might be attributed to the differences in the cell line.

In the current study, ccRCC 786-O cells were exposed for 24, 48, 72, and 96 h to several concentrations of BAY-876. The results demonstrated that BAY-876 can reduce 786-O cells in a dose-dependent manner and the IC50 was 53.56x10-6 M. As RCC specimens demonstrated a remarkably significant rise in the expression levels of GLUT1 compared with the corresponding normal kidney tissue (36,37), the present finding proves that reducing cell glucose intake by inhibiting GLUT1 through BAY-876 treatment, could represent a new strategy to suppress the growth of ccRCC. Moreover, the present result is consistent with a recent study (38) showing that BAY-876 has an inhibitory effect against bladder urothelial carcinoma (BLCA) cell lines and tumor inhibition effect in xenograft models. It was proposed that CLUTI inhibition through BAY-876 could be an effective approach to inhibiting the growth of BLCA tumors. Furthermore, another study (17) revealed that BAY-876 is a strong blocker of GLUT1 activity, mechanisms of glycolysis and ovarian cancer cell growth in ovarian cancer cell lines, as well as cell line-derived xerographs and ovarian cancer patient-derived xenografts.

The present results identified that the combination of BAY-876 and sunitinib has a significant cytotoxic effect on 786-O cells at shorter incubation times (after 24 and 48 h) compared with each drug alone, which had no cybotactic effect at these intervals. This combination may eliminate the cell by reducing the sugar intake by blocking GLUT1 transporters as well as VEGF, PDGF and c-Kit receptors (39). The multi-shunt elimination of 786-O cells during drug combination exposure may be attributable to a reduction in the duration of cell death when compared with each drug administered separately. This could be a promising advancement in the treatment of RCC.

RCC presents a contradictory aspect of the action of endogenous H2S. Numerous studies demonstrated an increase in H2S levels or H2S-producing enzymes in RCC cell lines, xenograft models and ccRCC tissues. These findings suggested a correlation between H2S and RCC growth and progression (40,41). On the contrary, another study argued that the expression of H2S-producing enzymes in ccRCC was reduced (42). The findings of the present study indicated that the use of NaSH alone resulted in a significant increase in cell death compared with the cells that were not treated. This finding aligns with the results of a previous study which demonstrated that NaSH has a significant impact on reducing cell viability in three different breast cancer cell lines (MCF-10, MCF-7 and MDA-MB-23) (43). The aforementioned study also demonstrated that exogenous H2S can suppress the progression of RCC by inhibiting the P13K/AKT pathway as well as by inducing apoptosis.

The cytotoxic activity of both sunitinib and BAY-876 increased when combined with NaSH. Overall, the combination of NaSH and BAY-876 could represent a novel approach to treating resistant metastatic ccRCC. If they are used in treatment, they may reduce the serious health impact and adverse reaction of sunitinib and other TKIs.

The clinicopathological characteristics of the patients in the present study revealed a slightly higher incidence rate of ccRCC in male patients (56.6%) compared with female patients (44.4%). However, a recent study demonstrated a significantly higher incidence rate of ccRCC in male patients (80%) compared with female patients (20%) (44). In addition, the TNM staging revealed that the incidence rates for stages I, II and III were 76.66, 3.33 and 20%, respectively.

Inactivation of the gene encoding the tumor suppressor VHL mutations is the most prevalent in ccRCC (45,46). This present study revealed that the incidence of intragenic VHL mutation was 43.7%, which is marginally lower than the percentage observed in a study conducted in Japan, which was 51% (47). This may be because only exon 1 was sequenced in the present research, whereas the entire gene was sequenced in the study by Kondo et al (47). In addition, the present study identified 36 missense SNP mutations in exon 1 of the VHL gene. This funding is consistent with the findings of a recent study which demonstrated that all mutations in VHL are missense mutations in ccRCC (48), and with COSMIC databases, which demonstrated that missense mutation is superior to other types of mutations in RCC. Based on this number of point mutations in VHL, mostly with an amino acid change (30 out of 36), the present study suggested that mutations in this gene could be the primary cause of ccRCC and confirmed that TKIs such as sunitinib may be one of the best options for inhibiting the growth of this type of kidney cancer, as they inhibit angiogenesis by inhibiting the downstream pathways of VHL/HIF.

VEGFR-2 serves a crucial role as a primary mediator in tumor angiogenesis and is regarded as a significant target for therapeutic interventions to counteract angiogenesis. Several anti-angiogenic medications, including ramucirumab, sunitinib, axitinib and sorafenib, have been developed to target this pathway (49). A previous study revealed that the rs1870377 A>T genetic polymorphism of VEGFR-2 is associated with the prognosis of gastric cancer (50). The present study demonstrated that there were 16 heterozygous missense variants in exon 13 of KDR, which is consistent with data from both the COSMIC and mutagen databases. This may explain the function of KDR gene mutation in the development of ccRCC and resistance to numerous anti-angiogenic drugs. KDR mutation testing is crucial for guiding the choice of therapy and improving patient outcomes.

Moreover, the present study found that BAY-876 and NaSH could improve sunitinib's anticancer efficacy in ccRCC cell lines, suggesting a novel treatment approach. It also found that VHL mutation is the main cause of ccRCC development and KDR mutation is necessary for angiogenesis. Testing these mutations might help choose the right therapy and understand the illness process. To the best of the authors' knowledge, the present study is the first to identify a significant number of VHL and KDR gene mutations in patients with ccRCC. By interpreting these gene mutations and relating their detection with relevant mutation-related datasets, the molecular mechanisms that initiate and progress ccRCC were elucidated. The present investigation is important in the context of RCC research and lays the groundwork for future studies. However, additional research is needed to confirm the current outcomes on other cell lines, also to determine the cellular and physiological effects of these agents. Functional studies could investigate the molecular effects of these mutations, their effects on clinical outcomes in larger groups, their predictive value for treatment response and genomic analysis to identify more mutations and variations. Notably, the significance of tracking response rate and cancer regression rate over time is recognized as a crucial avenue for future research and it is intended by the authors to contemplate integrating this data into subsequent research endeavors or offering a more comprehensive analysis in subsequent studies.

Supplementary Material

Agarose gel electrophoresis of PCR product for tumor suppressor gene Von Hippel Lindau and kinase insert domain receptor genes. (A) Represent amplification of exon I of VHL gene. (B) Represent the amplification segment of axon 13 of KRD gene. Lane 1: Ladder. Lane 2-16 demonstrate PCR products of ccRCC patients. The bands for both VHL and KRD fragments for all patients were located within the expected fragment size (VHL=260 bp and KDR=290 bp).
Summary of mutations in the VHL and KDR genes in all types of cancer retrieved from the gnomAD database.
Summary of mutations in the VHL and KDR genes in renal cell carcinoma retrieved from the Mutagene database.
Summary of mutations in the VHL and KDR genes in ccRCC retrieved from the COSMIC database.
The interaction between VHL ad KDR gene and their related genes retrieved from GeneMania database

Acknowledgements

The authors would like to thank Dr Abbas Salihi from Salahaddin University in Erbil, Salahaddin Universality Research Center as well as the Histopathology department and laboratory staff members at Nanakaly Hospital, Rizgary Hospital, and Par Hospital in Erbil, Iraq, for providing samples.

Funding

Funding: No funding was received.

Availability of data and materials

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

Authors' contributions

PSH conceived and designed the experiments, performed the experiments, analyzed the data, prepared figures and/or tables, authored or reviewed drafts of the article. KAM conceived and designed the experiments, performed the experiments, authored, or reviewed drafts of the article. PSH and KAM confirm the authenticity of all the raw data. Both authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved (approval no. 45/90) by the Human Research Ethics Committee of Affiliated Salahaddin University (Erbil, Iraq). Written informed consent forms were obtained from the participants approving the use of their tissues for investigation.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bahadoram S, Davoodi M, Hassanzadeh S, Bahadoram M, Barahman M and Mafakher L: Renal cell carcinoma: An overview of the epidemiology, diagnosis, and treatment. G Ital Nefrol. 39:32–47. 2022.PubMed/NCBI

2 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2021. CA Cancer J Clin. 71:7–33. 2021.PubMed/NCBI View Article : Google Scholar

3 

Monjaras-Avila CU, Lorenzo-Leal AC, Luque-Badillo AC, D'Costa N, Chavez-Muñoz C and Bach H: The tumor immune microenvironment in clear cell renal cell carcinoma. Int J Mol Sci. 24(7946)2023.PubMed/NCBI View Article : Google Scholar

4 

Kase AM, George DJ and Ramalingam S: Clear cell renal cell carcinoma: From biology to treatment. Cancers (Basel). 15(665)2023.PubMed/NCBI View Article : Google Scholar

5 

Padala SA and Barsouk A, Thandra KC, Saginala K, Mohammed A, Vakiti A, Rawla P and Barsouk A: Epidemiology of renal cell carcinoma. World J Oncol. 11:79–87. 2020.PubMed/NCBI View Article : Google Scholar

6 

Jonasch E, Walker CL and Rathmell WK: Clear cell renal cell carcinoma ontogeny and mechanisms of lethality. Nat Rev Nephrol. 17:245–261. 2021.PubMed/NCBI View Article : Google Scholar

7 

Nickerson ML, Jaeger E, Shi Y, Durocher JA, Mahurkar S, Zaridze D, Matveev V, Janout V, Kollarova H, Bencko V, et al: Improved identification of von Hippel-Lindau gene alterations in clear cell renal tumors. Clin Cancer Res. 14:4726–4734. 2008.PubMed/NCBI View Article : Google Scholar

8 

Dizman N, Philip EJ and Pal SK: Genomic profiling in renal cell carcinoma. Nat Rev Nephrol. 16:435–451. 2020.PubMed/NCBI View Article : Google Scholar

9 

Williamson SR: Clear cell papillary renal cell carcinoma: An update after 15 years. Pathology. 53:109–119. 2021.PubMed/NCBI View Article : Google Scholar

10 

Sharma R, Kadife E, Myers M, Kannourakis G, Prithviraj P and Ahmed N: Determinants of resistance to VEGF-TKI and immune checkpoint inhibitors in metastatic renal cell carcinoma. J Exp Clin Cancer Res. 40(186)2021.PubMed/NCBI View Article : Google Scholar

11 

Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, Heng DY, Larkin J and Ficarra V: Renal cell carcinoma. Nat Rev Dis Primers. 3(17009)2017.PubMed/NCBI View Article : Google Scholar

12 

Makino T, Kadomoto S, Izumi K and Mizokami A: Epidemiology and prevention of renal cell carcinoma. Cancers (Basel). 14(4059)2022.PubMed/NCBI View Article : Google Scholar

13 

Choi WSW, Boland J and Lin J: Hypoxia-inducible factor-2α as a novel target in renal cell carcinoma. J Kidney Cancer VHL. 8:1–7. 2021.PubMed/NCBI View Article : Google Scholar

14 

Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, et al: Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 356:115–124. 2007.PubMed/NCBI View Article : Google Scholar

15 

Jin J, Xie Y, Zhang JS, Wang JQ, Dai SJ, He WF, Li SY, Ashby CR Jr, Chen ZS and He Q: Sunitinib resistance in renal cell carcinoma: From molecular mechanisms to predictive biomarkers. Drug Resist Updat. 67(100929)2023.PubMed/NCBI View Article : Google Scholar

16 

Rini BI, Tamaskar I, Shaheen P, Salas R, Garcia J, Wood L, Reddy S, Dreicer R and Bukowski RM: Hypothyroidism in patients with metastatic renal cell carcinoma treated with sunitinib. J Natl Cancer Inst. 99:81–83. 2007.PubMed/NCBI View Article : Google Scholar

17 

Ma Y, Wang W, Idowu MO, Oh U, Wang XY, Temkin SM and Fang X: Ovarian cancer relies on glucose transporter 1 to fuel glycolysis and growth: Anti-tumor activity of BAY-876. Cancers (Basel). 11(33)2018.PubMed/NCBI View Article : Google Scholar

18 

Mantle D and Yang G: Hydrogen sulfide and metal interaction: The pathophysiological implications. Mol Cell Biochem. 477:2235–2248. 2022.PubMed/NCBI View Article : Google Scholar

19 

Zaorska E, Tomasova L, Koszelewski D, Ostaszewski R and Ufnal M: hydrogen sulfide in pharmacotherapy, beyond the hydrogen sulfide-donors. Biomolecules. 10(323)2020.PubMed/NCBI View Article : Google Scholar

20 

Khattak S, Rauf MA, Khan NH, Zhang QQ, Chen HJ, Muhammad P, Ansari MA, Alomary MN, Jahangir M, Zhang CY, et al: Hydrogen sulfide biology and its role in cancer. Molecules. 27(3389)2022.PubMed/NCBI View Article : Google Scholar

21 

Cao X, Ding L, Xie ZZ, Yang Y, Whiteman M, Moore PK and Bian JS: A review of hydrogen sulfide synthesis, metabolism, and measurement: Is modulation of hydrogen sulfide a novel therapeutic for cancer? Antioxid Redox Signal. 31:1–38. 2019.PubMed/NCBI View Article : Google Scholar

22 

Powell CR, Dillon KM and Matson JB: A review of hydrogen sulfide (H(2)S) donors: Chemistry and potential therapeutic applications. Biochem Pharmacol. 149:110–123. 2018.PubMed/NCBI View Article : Google Scholar

23 

Chen G, Zhang Y and Wu X: 786-0 renal cancer cell line-derived exosomes promote 786-0 cell migration and invasion in vitro. Oncol Lett. 7:1576–1580. 2014.PubMed/NCBI View Article : Google Scholar

24 

Gu L, Sang Y, Nan X, Zheng Y, Liu F, Meng L, Sang M and Shan B: circCYP24A1 facilitates esophageal squamous cell carcinoma progression through binding PKM2 to regulate NF-κB-induced CCL5 secretion. Mol Cancer. 21(217)2022.PubMed/NCBI View Article : Google Scholar

25 

Warren AY and Harrison D: WHO/ISUP classification, grading and pathological staging of renal cell carcinoma: Standards and controversies. World J Urol. 36:1913–1926. 2018.PubMed/NCBI View Article : Google Scholar

26 

Elkassem AA, Allen BC, Sharbidre KG, Rais-Bahrami S and Smith AD: Update on the role of imaging in clinical staging and restaging of renal cell carcinoma based on the AJCC 8th edition, from the AJR special series on cancer staging. AJR Am J Roentgenol. 217:541–555. 2021.PubMed/NCBI View Article : Google Scholar

27 

Delahunt B, Eble JN, Samaratunga H, Thunders M, Yaxley JW and Egevad L: Staging of renal cell carcinoma: Current progress and potential advances. Pathology. 53:120–128. 2021.PubMed/NCBI View Article : Google Scholar

28 

Abid MN, Qadir FA and Salihi A: Association between the serum concentrations and mutational status of IL-8, IL-27 and VEGF and the expression levels of the hERG potassium channel gene in patients with colorectal cancer. Oncol Lett. 22(665)2021.PubMed/NCBI View Article : Google Scholar

29 

Alves MR, Carneiro FC, Lavorato-Rocha AM, da Costa WH, da Cunha IW, de Cássio Zequi S, Guimaraes GC, Soares FA, Carraro DM and Rocha RM: Mutational status of VHL gene and its clinical importance in renal clear cell carcinoma. Virchows Arch. 465:321–330. 2014.PubMed/NCBI View Article : Google Scholar

30 

Housein Z, Kareem TS and Salihi A: In vitro anticancer activity of hydrogen sulfide and nitric oxide alongside nickel nanoparticle and novel mutations in their genes in CRC patients. Sci Rep. 11(2536)2021.PubMed/NCBI View Article : Google Scholar

31 

Gudmundsson S, Singer-Berk M, Watts NA, Phu W, Goodrich JK and Solomonson M: Genome Aggregation Database Consortium. Rehm HL, MacArthur DG and O'Donnell-Luria A: Variant interpretation using population databases: Lessons from gnomAD. Hum Mutat. 43:1012–1030. 2022.PubMed/NCBI View Article : Google Scholar

32 

Sondka Z, Bamford S, Cole CG, Ward SA, Dunham I and Forbes SA: The COSMIC cancer gene census: Describing genetic dysfunction across all human cancers. Nat Rev Cancer. 18:696–705. 2018.PubMed/NCBI View Article : Google Scholar

33 

Franz M, Rodriguez H, Lopes C, Zuberi K, Montojo J, Bader GD and Morris Q: GeneMANIA update 2018. Nucleic Acids Res. 46:W60–W64. 2018.PubMed/NCBI View Article : Google Scholar

34 

Sato H, Siddig S, Uzu M, Suzuki S, Nomura Y, Kashiba T, Gushimiyagi K, Sekine Y, Uehara T, Arano Y, et al: Elacridar enhances the cytotoxic effects of sunitinib and prevents multidrug resistance in renal carcinoma cells. Eur J Pharmacol. 746:258–266. 2015.PubMed/NCBI View Article : Google Scholar

35 

Amaro F, Pisoeiro C, Valente MJ, Bastos ML, de Pinho PG, Carvalho M and Pinto J: Sunitinib versus pazopanib dilemma in renal cell carcinoma: New Insights into the in vitro metabolic impact, efficacy, and safety. Int J Mol Sci. 23(9898)2022.PubMed/NCBI View Article : Google Scholar

36 

Singer K, Kastenberger M, Gottfried E, Hammerschmied CG, Büttner M, Aigner M, Seliger B, Walter B, Schlösser H, Hartmann A, et al: Warburg phenotype in renal cell carcinoma: high expression of glucose-transporter 1 (GLUT-1) correlates with low CD8(+) T-cell infiltration in the tumor. Int J Cancer. 128:2085–2095. 2011.PubMed/NCBI View Article : Google Scholar

37 

Abdolahi M, Alam M, Ghaffarpasand A, Nouri F, Badkoobeh A, Golkar M, Abassi K and Torbati P: Assessment of the expression of GLUT1 in renal cell carcinoma and its various subtypes. Open Access Maced J Med Sci. 10:2581–2585. 2022.

38 

Wang X, He H, Rui W, Zhang N, Zhu Y and Xie X: TRIM38 triggers the uniquitination and degradation of glucose transporter type 1 (GLUT1) to restrict tumor progression in bladder cancer. J Transl Med. 19(508)2021.PubMed/NCBI View Article : Google Scholar

39 

Kollmannsberger C, Soulieres D, Wong R, Scalera A, Gaspo R and Bjarnason G: Sunitinib therapy for metastatic renal cell carcinoma: recommendations for management of side effects. Can Urol Assoc J. 1:S41–S54. 2007.PubMed/NCBI View Article : Google Scholar

40 

Sonke E, Verrydt M, Postenka CO, Pardhan S, Willie CJ, Mazzola CR, Hammers MD, Pluth MD, Lobb I, Power N, et al: Inhibition of endogenous hydrogen sulfide production in clear-cell renal cell carcinoma cell lines and xenografts restricts their growth, survival and angiogenic potential. Nitric Oxide. 49:26–39. 2015.PubMed/NCBI View Article : Google Scholar

41 

Shackelford RE, Abdulsattar J, Wei EX, Cotelingam J, Coppola D and Herrera GA: Increased nicotinamide phosphoribosyltransferase and cystathionine-β-synthase in renal oncocytomas, renal urothelial carcinoma, and renal clear cell carcinoma. Anticancer Res. 37:3423–3427. 2017.PubMed/NCBI View Article : Google Scholar

42 

Breza J Jr, Soltysova A, Hudecova S, Penesova A, Szadvari I, Babula P, Chovancova B, Lencesova L, Pos O, Breza J, et al: Endogenous H(2)S producing enzymes are involved in apoptosis induction in clear cell renal cell carcinoma. BMC Cancer. 18(591)2018.PubMed/NCBI View Article : Google Scholar

43 

Dong Q, Yang B, Han JG, Zhang MM, Liu W, Zhang X, Yu HL, Liu ZG, Zhang SH, Li T, et al: A novel hydrogen sulfide-releasing donor, HA-ADT, suppresses the growth of human breast cancer cells through inhibiting the PI3K/AKT/mTOR and Ras/Raf/MEK/ERK signaling pathways. Cancer Lett. 455:60–72. 2019.PubMed/NCBI View Article : Google Scholar

44 

Sekar H, Krishnamoorthy S, Kumaresan N, Chandrasekaran D, Ramaswamy P, Sundaram S and Raj N: Clinicopathological comparison of VHL expression as a prognostic tumor marker in renal cell carcinoma: A single center experience. Niger J Clin Pract. 24:614–620. 2021.PubMed/NCBI View Article : Google Scholar

45 

Audenet F, Yates DR, Cancel-Tassin G, Cussenot O and Rouprêt M: Genetic pathways involved in carcinogenesis of clear cell renal cell carcinoma: Genomics towards personalized medicine. BJU Int. 109:1864–1870. 2012.PubMed/NCBI View Article : Google Scholar

46 

Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME, Wykoff CC, Pugh CW, Maher ER and Ratcliffe PJ: The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 399:271–275. 1999.PubMed/NCBI View Article : Google Scholar

47 

Kondo K, Yao M, Yoshida M, Kishida T, Shuin T, Miura T, Moriyama M, Kobayashi K, Sakai N, Kaneko S, et al: Comprehensive mutational analysis of the VHL gene in sporadic renal cell carcinoma: Relationship to clinicopathological parameters. Genes Chromosomes Cancer. 34:58–68. 2002.PubMed/NCBI View Article : Google Scholar

48 

Lin PH, Huang CY, Yu KJ, Kan HC, Liu CY, Chuang CK, Lu YC, Chang YH, Shao IH and Pang ST: Genomic characterization of clear cell renal cell carcinoma using targeted gene sequencing. Oncol Lett. 21(169)2021.PubMed/NCBI View Article : Google Scholar

49 

Shah AA, Kamal MA and Akhtar S: Tumor angiogenesis and VEGFR-2: Mechanism, pathways and current biological therapeutic interventions. Curr Drug Metab. 22:50–59. 2021.PubMed/NCBI View Article : Google Scholar

50 

Zhu X, Wang Y, Xue W, Wang R, Wang L, Zhu ML and Zheng L: The VEGFR-2 protein and the VEGFR-2 rs1870377 A>T genetic polymorphism are prognostic factors for gastric cancer. Cancer Biol Ther. 20:497–504. 2019.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

March-2024
Volume 20 Issue 3

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hamadamin PS and Hamadamin PS: Exploring the anticancer potential of hydrogen sulfide and BAY‑876 on clear cell renal cell carcinoma cells: Uncovering novel mutations in VHL and KDR genes among ccRCC patients. Mol Clin Oncol 20: 21, 2024
APA
Hamadamin, P.S., & Hamadamin, P.S. (2024). Exploring the anticancer potential of hydrogen sulfide and BAY‑876 on clear cell renal cell carcinoma cells: Uncovering novel mutations in VHL and KDR genes among ccRCC patients. Molecular and Clinical Oncology, 20, 21. https://doi.org/10.3892/mco.2024.2719
MLA
Hamadamin, P. S., Maulood, K. A."Exploring the anticancer potential of hydrogen sulfide and BAY‑876 on clear cell renal cell carcinoma cells: Uncovering novel mutations in VHL and KDR genes among ccRCC patients". Molecular and Clinical Oncology 20.3 (2024): 21.
Chicago
Hamadamin, P. S., Maulood, K. A."Exploring the anticancer potential of hydrogen sulfide and BAY‑876 on clear cell renal cell carcinoma cells: Uncovering novel mutations in VHL and KDR genes among ccRCC patients". Molecular and Clinical Oncology 20, no. 3 (2024): 21. https://doi.org/10.3892/mco.2024.2719