HRAS as a potential therapeutic target of salirasib RAS inhibitor in bladder cancer

  • Authors:
    • Satoshi Sugita
    • Hideki Enokida
    • Hirofumi Yoshino
    • Kazutaka Miyamoto
    • Masaya Yonemori
    • Takashi Sakaguchi
    • Yoichi Osako
    • Masayuki Nakagawa
  • View Affiliations

  • Published online on: June 11, 2018     https://doi.org/10.3892/ijo.2018.4435
  • Pages: 725-736
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The active form of the small GTPase RAS binds to downstream effectors to promote cell growth and proliferation. RAS signal enhancement contributes to tumorigenesis, invasion, and metastasis in various different cancers. HRAS proto-oncogene GTPase (HRAS), one of the RAS isoforms, was the first human oncogene for which mutations were reported in T24 bladder cancer (BC) cells in 1982, and HRAS mutation or upregulation has been reported in several cancers. According to data from The Cancer Genome Atlas, HRAS expression was significantly upregulated in clinical BC samples compared to healthy samples (P=0.0024). HRAS expression was also significantly upregulated in BC with HRAS mutation compared to patients without HRAS mutation (P<0.0001). The tumor suppressive effect of salirasib, a RAS inhibitor, has been reported in several cancer types, but only at relatively high concentrations. As such, RAS inhibitors have not been used for clinical applications. The aim of the current study was to investigate the therapeutic potential of targeting HRAS using salirasib and small interfering RNA (siRNA) and to characterize the mechanism by which HRAS functions using recently developed quantitative in vitro proteome-assisted multiple reaction monitoring for protein absolute quantification (iMPAQT), in BC cells. iMPAQT allows measurement of the absolute abundance of any human protein with the high quantitative accuracy. Salirasib and siRNA targeting of HRAS inhibited cell proliferation, migration and invasion in HRAS wild type and HRAS-mutated cell lines. Proteomic analyses revealed that several metabolic pathways, including the oxidative phosphorylation pathway and glycolysis, were significantly downregulated in salirasib-treated BC cells. However, the expression levels of hexokinase 2, phosphoglycerate kinase 1, pyruvate kinase, muscle (PKM)1, PKM2 and lactate dehydrogenase A, which are downstream of RAS and target genes of hypoxia inducible factor-1α, were not notably downregulated, which may explain the high concentration of salirasib required to inhibit cell viability. These findings provide insight into the mechanisms of salirasib, and suggest the need for novel therapeutic strategies to treat cancers such as BC.

Introduction

Bladder cancer (BC) was the 5th most commonly diagnosed cancer and the 8th most common cause of cancer-associated mortality among the 40 European Union countries in 2012. In that same year, 429,800 new cases of BC were diagnosed, and 165,000 patients succumbed to BC worldwide (1,2). The 5-year survival rate of patients with BC has improved by only a small percentage during the last 30 years according to the National Cancer Institute program Surveillance, Epidemiology and End Results (3). One factor in the lack of improvement in BC survival rates is the limited efficacy of cisplatin-based combination chemotherapy (4). Thus, innovative therapeutic strategies are required to improve BC outcomes.

RAS proteins are small molecular weight GTPases that couple extracellular signals to intracellular effector pathways. Mammalian cells encode three closely related RAS proteins, HRas proto-oncogene GTPase (HRAS), NRAS protooncogene GTPase (NRAS) and KRAS proto-oncogene GTPase (KRAS), which have critical roles in fundamental cellular processes, including proliferation, survival, differentiation, motility and transcription (5). The RAS pathway is one of the most commonly deregulated pathways in human cancer (6), and activating mutations in RAS genes occur in ~30% of all tumors (7). These mutations typically render RAS as constitutively GTP-bound, resulting in activation of downstream effector pathways regardless of extracellular stimulation (6). Substitution of glycine for valine at amino acid 12 (G12V) is one of the most frequently observed RAS mutations that interferes with GTPase-activating protein-mediated GTP hydrolysis, leading to excess amounts of active GTP-bound RAS. Notably, the type of mutated RAS gene (HRAS, KRAS or NRAS) varies depending on the tumor type; KRAS mutations are frequently detected in pancreatic carcinoma (80–90%) and colorectal carcinoma (30–60%), whereas HRAS mutations are frequent in BC (7–66%) and thyroid cancer (0–60%) (8).

RAS is considered 'undruggable' because the RAS protein lacks a druggable binding pocket (9,10). Additionally, development of specific and competitive nucleotide inhibitors is challenging, because RAS binds nucleotide ligands with high affinity (10). To overcome these challenges, the RAS antagonist salirasib, also termed farnesylthiosalicylate, was designed to competitively inhibit attachment of GTP-bound RAS to the plasma membrane, which in turn inactivates RAS signaling (11). Salirasib inhibits all RAS isoforms and inhibits the growth of RAS-driven cancer (11,12). Although applications of salirasib have been tested in several clinical trials for cancers other than BC (1316), it exhibited insufficient tumor suppressive effects in trials in which salirasib was the single agent. In addition, relatively high concentrations of salirasib were required to achieve sufficient tumor suppressive effects (17,18). Therefore, a more detailed examination of the effects of salirasib is required to understand the mechanism by which salirasib acts on RAS. Recently, Matsumoto et al (19) developed a targeted proteomics platform, in vitro proteome-assisted multiple reaction monitoring for protein absolute quantification (iMPAQT), that analyzes 18,000 human recombinant proteins to enable absolute protein quantification on a genome-wide scale (19) and overcome limitations in quantitative accuracy, reproducibility, and analysis speed associated with conventional analysis methods. Use of iMPAQT allows large-scale and accurate assessment of protein abundances that can influence cellular phenotypes.

In the current study, the therapeutic of potential of HRAS knockdown by salirasib or RNA interference was investigated in two BC cell lines (T24 cells with HRAS G12V mutation and BOY cells without HRAS mutation). Furthermore, newly developed quantitative proteome analysis of BC cells treated with salirasib was performed to elucidate the mechanisms underlying the actions of salirasib toward HRAS.

Materials and methods

Analysis in the BC cohort of The Cancer Genome Atlas (TCGA)

Sequencing data were available for 407 BC samples and 19 normal bladder epithelial samples in TCGA database (tcga-data.nci.nih. gov/tcga/). We used TCGA to analyze HRAS mRNA expression levels in normal and BC tissues and to evaluate differences in HRAS mRNA expression levels according to HRAS mutational status. RNA-Seq by Expectation Maximization software was used for gene expression quantification (20). Full sequencing information, somatic mutation information, and clinical information were acquired using UCSC Xena (xena.ucsc.ed/) and TCGA. The current study meets publication guidelines provided by TCGA (cancergenome.nih.gov/publications/publicationguidelines).

Cell culture and RNA extraction

Four human BC cell lines were used. T24, KK47 and UMUC cells, which were obtained from the American Type Culture Collection (Manassas, VA, USA), and BOY cells, which were established in our laboratory from a 66-year-old Asian male patient diagnosed with stage III BC with lung metastasis. These cell lines were maintained in the minimum essential Eagle's medium (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany), containing 10% fetal bovine serum (Equitech-Bio, Inc., Kerrville, TX, USA), 50 µg/ml streptomycin, and 50 U/ml penicillin in a humidified atmosphere of 95% air/5% CO2 at 37°C. Total RNA was isolated using Isogen (Nippon Gene Co., Ltd., Tokyo, Japan) according to the manufacturer's protocol. The integrity of the RNA was checked with an RNA 6000 Nano assay kit and a 2100 Bioanalyzer (Agilent Technologies, Inc., Santa Clara, CA, USA).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

A SYBR-Green qPCR-based array approach was used for RT-qPCR. RT was performed using the TaqMan High-Capacity cDNA Reverse Transcription Kit (cat. no. 4368814; Applied Biosystems; Thermo Fisher Scientific, Inc., Waltham, MA, USA) under the incubation conditions (25°C for 10 min, 37°C for 120 min and 85°C for 5 min) according to the manufacturer's instructions. The primer set for determination of mRNA expression levels was as follows: HRAS, forward, 5′-ATGACGGAATATAAGCTGGTGGT-3′ and reverse, 5′-GGCACGTCTCCCCATCAATG-3′; hypoxia inducible factor-1α (HIF-1α), forward, 5′-GAACGTCGAAAAGAAAA GTCTCG-3′ and reverse, 5′-CCTTATCAAGATGCGAACTC ACA-3′; glucuronidase β (GUSB), forward, 5′-CGTCCCACCTAGAATCTGCT-3′ and reverse, 5′-TTGCTCACAAAGGT CACAGG-3′. The experimental procedures followed the protocol recommended by the manufacturer. RT-qPCR was performed with 500 ng total RNA using the Power SYBR-Green Master Mix (cat. no. 4367659) with the 7300 Real-time PCR System (both from Applied Biosystems; Thermo Fisher Scientific, Inc.). Amplification specificity was monitored using the dissociation curve of the amplified product. All data values were normalized with respect to GUSB, and the ΔΔCq method was used to calculate the fold-change (21). Human Bladder Total RNA (cat. no. AM7990; Applied Biosystems; Thermo Fisher Scientific, Inc.) as control RNA derived from normal bladder tissue.

Salirasib treatment

For in vitro experiments, salirasib (CAS 162520-00-5; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) was solubilized in 0.1% DMSO. The salirasib/DMSO solution and control vehicle (0.1% DMSO) were prepared in Dulbecco's modified Eagle's medium at different concentrations, and each mixture was placed in cell culture plates (8×104/ml) so that the final salirasib concentrations were 1.6, 3.1, 6.3, 12.5, 25, 50, 100 and 200 µM, whereas the DMSO concentration was adjusted to 0.1%. Each cell culture plate was treated with salirasib or control vehicle for 24 h. For in vivo experiments, salirasib was solubilized with 0.5% ethanol. The salirasib/ethanol solution was alkalinized with 1 N NaOH and then diluted with phosphate buffered saline to yield a 4 mg/ml (pH 8.0) solution. This solution or control vehicle (0.5% ethanol) were intraperitoneally (i.p.) injected daily 100 µl per mouse.

Transfection with small interfering RNA (siRNA)

As described previously (22), T24 and BOY cells were transfected using Lipofectamine RNAiMAX transfection reagent and Opti-MEM (both from Thermo Fisher Scientific, Inc.) together with 10 nM HRAS siRNA (nos. Hs_HRAS_1174, Hs_HRAS_1177 and Hs_HRAS_1178; Sigma-Aldrich; Merck KGaA) or negative-control siRNA (no. D-001810-10; Thermo Fisher Scientific, Inc.) for loss-of-function experiments. The sequences of the siRNAs were as follows: Hs_HRAS_1174_s, 5′rGUrGrCrCUrGUUrGr GrArCrAUrCrCUrGTT; Hs_HRAS_1174_as, 5′rCrArGrGr AUrGUrCrCrArArCrArGrGrCrArCTT; Hs_HRAS_1177_s, 5′rGrArCrGUrGrCrCUrGUUrGrGrArCrAUrCTT; Hs_HRAS_1177_as, 5′rGrAUrGUrCrCrArArCrArGrGrCrArCr GUrCTT; Hs_HRAS_1178_s, 5′rGrGrGrCUUrCrCUrGUrGU rGUrGUUUTT; and Hs_HRAS_1178_as, 5′rArArArCrArCrAr CrArCrArGrGrArArGrCrCrCTT. Subsequent experiments were performed 72 h after siRNA transfection.

Cell proliferation, migration, and invasion assays

T24 and BOY cells were transfected with 10 nM siRNA by reverse transfection. Cells were seeded in 96-well plates with 3×103 cells/well for XTT assays. After 72 h, cell proliferation was determined using a Cell Proliferation Kit II (Roche Diagnostics GmbH, Mannheim, Germany) as described previously (22). Cell migration activity was evaluated with wound healing assays. Cells were plated in 6-well plates at 2×105 cells per well, and after 48 h of transfection the cell monolayer was scraped using a P-20 micropipette tip. The initial (0 h) and residual gap length 18 h after wounding were calculated from photomicrographs as previously described (22). Cell invasion assays were performed using modified Boyden chambers consisting of Matrigel-coated Transwell membrane filter inserts with 8-µM pores in 24 well tissue culture plates (BD Biosciences, San Jose, CA, USA). At 72 h after transfection, cells were plated in 24-well plates at 1×105 cells/well. Minimum essential Eagle's medium containing 10% fetal bovine serum (Equitech-Bio, Inc.) in the lower chamber served as the chemoattractant, as described previously (22). Medium Eagle fetal bovine serum and cells were prepared in the upper chamber and incubated for 24 h.

Western blot analysis

Cells were harvested 72 h after transfection, and lysates were prepared in radioimmunoprecipitation assay lysis buffer (Thermo Fisher Scientific, Inc.) containing protease inhibitor cocktail (Sigma-Aldrich; Merck KGaA). Proteins were quantified by Bradford method using BioPhotometer (Eppendorf, Hamburg, Germany). Proteins (50 µg) were separated by NuPAGE on 4–12% bistris gels (Invitrogen; Thermo Fisher Scientific, Inc.) and transferred to polyvinylidene difluoride membranes. Following blocking in Tris-buffered saline containing 0.1% Tween-20 (TBS-T) with 5% nonfat dry milk for 15 min at 25°C, membranes were washed four times in TBS-T and incubated with primary antibodies overnight at 4°C. Immunoblotting was performed with diluted rabbit polyclonal anti-HRAS antibodies (1:1,000; cat. no. GTX116041; GeneTex, Inc., Irvine, CA, USA), goat polyclonal anti-HIF-1α antibodies (1:1,000; cat. no. AF1935; R&D Systems, Inc., Minneapolis, MN, USA) and rabbit polyclonal anti-β-actin antibodies (1:1,000; cat. no. bs-0061R; BIOSS, Beijing, China) according to the manufacturer's instructions for each antigen. The secondary antibodies were peroxidase-labelled anti-rabbit IgG (1 h at 25°C; 1:5,000; cat. no. 7074S; Cell Signaling Technology, Inc., Danvers, MA, USA) and anti-goat IgG (1 h at 25°C; 1:5,000; cat. no. sc-2020; Santa Cruz Biotechnology, Inc.). Specific complexes were visualized with an enhanced chemiluminescence detection system (GE Healthcare Life Sciences, Little Chalfont, UK) as described previously (23).

Proteomic analysis

To comprehensively investigate metabolic changes in BC cells treated with salirasib, proteomic analysis was performed using iMPAQT (19). Proteins with downregulated expression were detected in salirasib-treated BC cells compared with untreated cells (fold change <0.5) and proteins that were common to both T24 and BOY were identified. The proteins were then categorized into Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways through GeneCodis analysis (genecodis.cnb.csic.es).

In vivo tumor xenograft model

To investigate in vivo effects of salirasib, a mixture containing 100 µl BOY cells (5×106) and 100 µl Matrigel Matrix (Corning Incorporated, Corning, NY, USA) was injected subcutaneously into one side flank of 9 female nude mice (BALB/c nu/nu; 8 weeks old; 16–19 g). The mice were randomly separated into salirasib-treated (n=5) and control (n=4) groups. Each breeding room was kept at a temperature of 23±1°C and a humidity of 40–70%. The light/dark cycle was set to 12 h. Food and water was placed to be accessible from each cage. From the day following tumor implantation, salirasib (0.4 mg/mouse, i.p., daily) and control vehicle (0.5% ethanol, 100 µl/mouse, i.p., daily) treatment were administered for 25 days. Tumor sizes were measured twice weekly and tumor volumes were calculated as follows: Tumor volume = [(long axis length in millimeters/2) × (short axis length/2)2 × π × 4]/3. All animal experiments were performed in accordance with institutional guidelines and were approved by the animal care review board of Kagoshima University (Kagoshima, Japan).

Statistical analysis

Data are presented as the mean ± standard deviation at least three independent experiments. The relationships between two groups were analyzed using Mann-Whitney U tests. The relationships between three or more variables and numerical values were analyzed using Bonferroni-adjusted Mann-Whitney U tests. All analyses were performed using Expert StatView software, version 5.0 (SAS Institute, Inc., Cary, NC, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Expression levels of HRAS in BC and BC cell lines

The expression levels of HRAS were evaluated using TCGA data from BC samples (n=407) and normal samples (n=19). HRAS expression levels were significantly upregulated in tumor tissues compared with those in normal bladder epithelia (tumor, 10.314±0.813; normal, 9.707±0.826; P=0.0024, Mann-Whitney U tests; Fig. 1A). Furthermore, HRAS expression was significantly upregulated in patients with BC with mutant HRAS compared with patients with wild-type HRAS (mutant HRAS, 11.277±0.805; wild-type HRAS, 10.267±0.788; P<0.0001, Mann-Whitney U tests) (Fig. 1B). HRAS mRNA expression was also significantly upregulated in BC cell lines compared to patients with normal bladder tissues (T24, 5.960±0.344, P<0.0001; BOY, 7.528±1.506, P<0.0001; KK47, 2.934±0.464, P=0.0176; UMUC, 3.561±0.854, P=0.0034; Bonferroni-adjusted Mann-Whitney U tests; Fig. 1C). Sequencing data from TCGA revealed that T24 cells had HRASG12V (the substitution of glycine by valine at codon 12 in HRAS) and BOY cells had wild-type HRAS (Fig. 1D).

Effects of HRAS knockdown on cell proliferation, migration, and invasion of BC cell lines

To investigate the functional role of HRAS in BC cells, loss-of-function studies were performed using T24 and BOY BC cells transfected with three si-HRAS constructs (si-HRAS-1, si-HRAS-2 and si-HRAS-3). RT-qPCR analysis and western blot analysis indicated that these siRNAs effectively downregulated HRAS mRNA and protein expression in both cell lines (Fig. 2A). XTT assays demonstrated that cell proliferation was inhibited in si-HRAS transfectants compared with mock or siRNA-control transfectants (T24, mock 1.0±0.047, control 1.0±0.015, si-HRAS-1 0.701±0.015, si-HRAS-2 0.615±0.011, si-HRAS-3 0.599±0.024; BOY, mock 1.0±0.024, control 0.996±0.087, si-HRAS-1 0.585±0.030, si-HRAS-2 0.499±0.020, si-HRAS-3 0.508±0.016; each P<0.0001, Bonferroni-adjusted Mann-Whitney test; Fig. 2B). Cell migration activity was also significantly inhibited in si-HRAS transfectants compared with mock or siRNA-control transfectants (T24, mock 1.0±0.111, control 1.013±0.117, si-HRAS-1 0.684±0.198, si-HRAS-2 0.583±0.309, si-HRAS-3 0.462±0.348, P=0.0019 and P<0.0001 vs. mock; BOY, mock 1.0±0.186, control 0.978±0.080, si-HRAS-1 0.158±0.105, si-HRAS-2 0.438±0.130, si-HRAS-3 0.448±0.186, each P<0.0001 vs. mock, Bonferroni-adjusted Mann-Whitney test; Fig. 2C), as was cell invasion activity in Matrigel assays (T24, mock 1.0±0.280, control 1.443±0.289, si-HRAS-1 0.543±0.113, si-HRAS-2 0.552±0.154, si-HRAS-3 0.353±0.074; BOY, mock 1.0±0.288, control 1.309±0.268, si-HRAS-1 0.117±0.070, si-HRAS-2 0.296±0.121, si-HRAS-3 0.142±0.086, each P<0.0001, Bonferroni-adjusted Mann-Whitney test; Fig. 2D).

Effects of salirasib on cell proliferation, migration, and invasion activities in BC cell lines

The effect of salirasib treatment was investigated in T24 and BOY cells. XTT assays demonstrated that ≥100 µM salirasib significantly reduced T24 and BOY viability compared with untreated cells (each P<0.0001, Bonferroni-adjusted Mann-Whitney test; Fig. 3A). Salirasib treatment significantly reduced T24 and BOY cell migration compared with the mock control (T24, mock 1.0±0.258, salirasib 0.498±0.326, P=0.0020; BOY, mock 1.0±0.470, salirasib 0.556±0.289, P= 0.0193, Mann-Whitney U tests; Fig. 3B) and invasion activity (T24, mock 1.0±0.336, salirasib 0.200±0.116; BOY, mock 1.0±0.477, salirasib 0.122±0.125, P=0.0009, Mann-Whitney U tests; Fig. 3C) relative to untreated cells.

Proteomic analysis in BC cells treated with salirasib

To comprehensively investigate metabolic changes in BC cells treated with salirasib, proteomic analysis of metabolism-associated genes was performed using iMPAQT. The results revealed 58 proteins with downregulated expression (fold change <0.5) in both T24 and BOY BC cells treated with salirasib compared to untreated cells (Table I). GeneCodis analysis to categorize the proteins into KEGG pathways demonstrated that these proteins were included in 50 pathways that were significantly enriched following salirasib treatment (listed in descending order of corrected P-values in Table II; Fig. 4). 'Oxidative phosphorylation', 'pyrimidine metabolism', 'glycolysis/gluconeogenesis', 'pentose phosphate pathway', 'cysteine and methionine metabolism', 'glutathione metabolism', and 'purine metabolism' were significantly downregulated pathways in BC cells treated with salirasib. However, target genes of the RAS effector HIF-1α, including hexokinase 2, phosphoglycerate kinase 1, pyruvate kinase, muscle (PKM)1, PKM2 and lactate dehydrogenase A, showed only modest downregulation (fold change >0.5 in T24 and BOY cells) (Table III) (2426). Furthermore, RT-qPCR analysis and western blot analysis indicated that expression of HIF-1α was not downregulated in salirasib-treated BC cells (Fig. 5) (5,27).

Table I

Metabolic changes in bladder cancer cells treated by salirasib.

Table I

Metabolic changes in bladder cancer cells treated by salirasib.

Gene symbolDescriptionExpression ratio (treated/untreated cells)
T24BOYMean of T24 and BOY
AHCYL1 Adenosylhomocysteinase-like 1NDNDND
AK3Adenylate kinase 3NDNDND
ALDH3A2Aldehyde dehydrogenase 3 family, member A2NDNDND
ALDH9A1Aldehyde dehydrogenase 9 family, member A1NDNDND
ASNSAsparagine synthetase (glutamine-hydrolyzing)NDNDND
ATP5LATP synthase, H+ transporting, mitochondrial Fo complex, subunit GNDNDND
ATP6V1E1Atpase, H+ transporting, lysosomal, V1 subunit E1NDNDND
ATP6V1G1Atpase, H+ transporting, lysosomal, V1 subunit G1NDNDND
DCXR Dicarbonyl/L-xylulose reductaseNDNDND
DERA Deoxyribose-phosphate aldolase (putative)NDNDND
DLDDihydrolipoamide dehydrogenaseNDNDND
G6PDGlucose-6-phosphate dehydrogenaseNDNDND
GBAGlucosidase β, acidNDNDND
GMPPAGDP-mannose pyrophosphorylase ANDNDND
GMPR2Guanosine monophosphate reductase 2NDNDND
GNPDA1 Glucosamine-6-phosphate deaminase 1NDNDND
HSD17B12Hydroxysteroid (17-β) dehydrogenase 12NDNDND
IDI1 Isopentenyl-diphosphate delta isomerase 1NDNDND
IVDIsovaleryl-coa dehydrogenaseNDNDND
MPSTMercaptopyruvate sulfurtransferaseNDNDND
MTMR1Myotubularin related protein 1NDNDND
NDUFS8NADH dehydrogenase (ubiquinone)NDNDND
Fe-S protein 8, 2 (NADH-coenzyme Q reductase)
NUDT9Nudix (nucleoside diphosphate linked moiety X)-type motif 9NDNDND
PAFAH1B3Platelet-activating factor acetylhydrolase 1b, catalytic subunit 3NDNDND
PANK4Pantothenate kinase 4NDNDND
PFKM Phosphofructokinase, muscleNDNDND
PGDPhosphogluconate dehydrogenaseNDNDND
PGM2Phosphoglucomutase 2NDNDND
PLOD3Procollagen-lysine, 2-oxoglutarate 5-dioxygenase 3NDNDND
PMVKPhosphomevalonate kinaseNDNDND
PRIM1Primase, DNA, polypeptide 1NDNDND
SCP2Sterol carrier protein 2NDNDND
UQCRB Ubiquinol-cytochrome c reductase binding proteinNDNDND
GAPDH Glyceraldehyde-3-phosphate dehydrogenase0.0680.1130.091
GOT1 Glutamic-oxaloacetic transaminase 1, soluble (aspartate aminotransferase 1)ND0.2420.121
BCAT1Branched chain amino-acid transaminase 1, cytosolic0.247ND0.123
AASDHPPT Aminoadipate-semialdehyde dehydrogenase-phosphopantetheinyl transferaseND0.2560.128
UAP1 UDP-N-acteylglucosamine pyrophosphorylase 10.1070.2120.16
PGM3Phosphoglucomutase 3ND0.3930.197
PAFAH1B2Platelet-activating factor acetylhydrolase 1b, catalytic subunit 2ND0.4050.203
BLVRBBiliverdin reductase B (flavin reductase (NADPH))ND0.4070.204
NDUFS1NADH dehydrogenase (ubiquinone) Fe-S protein 1, 7 (NADH-coenzyme Q reductase)ND0.4920.246
DLSTDihydrolipoamide S-succinyltransferase (E2 component of 2-oxo-glutarate complex)0.2930.2010.247
PHGDHPhosphoglycerate dehydrogenase0.2240.2840.254
HMOX1Heme oxygenase (decycling) 10.1650.3780.271
DUTDeoxyuridine triphosphatase0.2230.3690.296
AHCY Adenosylhomocysteinase0.2970.3150.306
ATP5A1ATP synthase, H+ transporting, mitochondrial F1 complex, α subunit 1, cardiac muscle0.2520.3630.308
SRMSpermidine synthase0.2970.4010.349
RRM1Ribonucleotide reductase M10.3470.360.353
ACAT1Acetyl-coa acetyltransferase 10.2710.4390.355
ISYNA1 Inositol-3-phosphate synthase 10.2340.480.357
NDUFA4NADH dehydrogenase (ubiquinone) 1α subcomplex, 40.2860.450.368
RRM2BRibonucleotide reductase M2 B (TP53 inducible)0.3730.4350.404
NUDT2Nudix (nucleoside diphosphate linked moiety X)-type motif 20.4430.3830.413
UQCRQ Ubiquinol-cytochrome c reductase, complex III subunit VII0.4140.4510.432
CMPK1Cytidine monophosphate (UMP-CMP) kinase 1, cytosolic0.4620.4430.452
ATP6V1B2Atpase, H+ transporting, lysosomal, V1 subunit B20.4980.4790.489

[i] ND, not detectable (set as 0).

Table II

Downregulated KEGG pathways in bladder cancer cells treated by salirasib.

Table II

Downregulated KEGG pathways in bladder cancer cells treated by salirasib.

KEGGIDAnnotationsNumber of genesCorrected P-valueGenes
00190Oxidative phosphorylation10 1.22×10−12ATP5A1, ATP6V1G1, NDUFS8, ATP5L, ATP6V1E1, NDUFA4, NDUFS1, UQCRB, ATP6V1B2, UQCRQ
00280Valine, leucine and isoleucine degradation6 3.79×10−9ALDH3A2, DLD, BCAT1, IVD, ALDH9A1, ACAT1
00240Pyrimidine metabolism7 4.84×10−9CMPK1, NUDT2, RRM2B, AK3, RRM1, PRIM1, DUT
00010 Glycolysis/gluconeogenesis6 1.29×10−8ALDH3A2, PGM2, DLD, ALDH9A1, GAPDH, PFKM
00030Pentose phosphate pathway5 1.43×10−8PGM2, DERA, G6PD, PGD, PFKM
00270Cysteine and methionine metabolism5 4.39×10−8MPST, AHCY, AHCYL1, SRM, GOT1
00480Glutathione metabolism5 8.35×10−8RRM2B, G6PD, RRM1, SRM, PGD
00230Purine metabolism7 8.57×10−8NUDT2, PGM2, GMPR2, RRM2B, RRM1, NUDT9, PRIM1
05010Alzheimer's disease7 9.31×10−8ATP5A1, NDUFS8, NDUFA4, NDUFS1, UQCRB, GAPDH, UQCRQ
00520Amino sugar and nucleotide sugar metabolism5 1.18×10−7PGM2, UAP1, GMPPA, PGM3, GNPDA1
00310Lysine degradation5 1.19×10−7ALDH3A2, ALDH9A1, PLOD3, DLST, ACAT1
05012Parkinson's disease6 4.55×10−7ATP5A1, NDUFS8, NDUFA4, NDUFS1, UQCRB, UQCRQ
05016Huntington's disease6 2.79×10−6ATP5A1, NDUFS8, NDUFA4, NDUFS1, UQCRB, UQCRQ
00620Pyruvate metabolism4 2.79×10−6ALDH3A2, DLD, ALDH9A1, ACAT1
00330Arginine and proline metabolism4 8.56×10−6ALDH3A2, ALDH9A1, SRM, GOT1
00900Terpenoid backbone biosynthesis3 8.57×10−6PMVK, IDI1, ACAT1
00770Pantothenate and CoA biosynthesis3 1.13×10−5BCAT1, AASDHPPT, PANK4
00410β-alanine metabolism3 3.82×10−5ALDH3A2, ALDH9A1, SRM
04966Collecting duct acid secretion3 3.82×10−5ATP6V1G1, ATP6V1E1, ATP6V1B2
00640Propanoate metabolism3 6.87×10−5ALDH3A2, ALDH9A1, ACAT1
00051Fructose and mannose metabolism3 9.37×10−5GMPPA, MTMR1, PFKM
00071Fatty acid metabolism30.000127ALDH3A2, ALDH9A1, ACAT1
00380Tryptophan metabolism30.000127ALDH3A2, ALDH9A1, ACAT1
05110Vibrio cholerae infection30.000264ATP6V1G1, ATP6V1E1, ATP6V1B2
05120Epithelial cell signaling in Helicobacter pylori infection30.000509ATP6V1G1, ATP6V1E1, ATP6V1B2
05323Rheumatoid arthritis30.000953ATP6V1G1, ATP6V1E1, ATP6V1B2
00053Ascorbate and aldarate metabolism20.002054ALDH3A2, ALDH9A1
00052Galactose metabolism20.002137PGM2, PFKM
00340Histidine metabolism20.002381ALDH3A2, ALDH9A1
00020Citrate cycle (TCA cycle)20.002464DLD, DLST
00040Pentose and glucuronate interconversions20.002546ALDH3A2, DCXR
00250Alanine, aspartate and glutamate metabolism20.002548ASNS, GOT1
00260Glycine, serine and threonine metabolism20.002548DLD, PHGDH
00565Ether lipid metabolism20.002791PAFAH1B3, PAFAH1B2
04145Phagosome30.002924ATP6V1G1, ATP6V1E1, ATP6V1B2
00860Porphyrin and chlorophyll metabolism20.004011HMOX1, BLVRB
00561Glycerolipid metabolism20.004873ALDH3A2, ALDH9A1
04260Cardiac muscle contraction20.011811UQCRB, UQCRQ
04146Peroxisome20.011811PMVK, SCP2
00400Phenylalanine, tyrosine and tryptophan biosynthesis10.013097GOT1
00072Synthesis and degradation of ketone bodies10.022922ACAT1
00290Valine, leucine and isoleucine biosynthesis10.024264BCAT1
04122Sulfur relay system10.024264MPST
00740Riboflavin metabolism10.026063BLVRB
00360Phenylalanine metabolism10.036111GOT1
00120Primary bile acid biosynthesis10.036111SCP2
00511Other glycan degradation10.03752GBA
00630Glyoxylate and dicarboxylate metabolism10.038867ACAT1
01040Biosynthesis of unsaturated fatty acids10.044309HSD17B12
00910Nitrogen metabolism10.04748ASNS

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes.

Table III

Effects of salirasib downstream of hypoxia inducible factor-1α.

Table III

Effects of salirasib downstream of hypoxia inducible factor-1α.

NameDescriptionT24 fold changeBOY fold changeMean of T24 and BOY
PKM2Pyruvate kinase, muscle 21.3691.1941.281
LDHALactate dehydrogenase A0.9181.0050.961
PKM1Pyruvate kinase, muscle 10.9001.0440.972
PGK1Phosphoglycerate kinase 10.8571.1921.025
HK2Hexokinase 20.7350.6970.716
ENO1Enolase 1, (α)0.4530.5870.520
GAPDH Glyceraldehyde-3-phosphate dehydrogenase0.0680.1130.091
Xenograft model study to investigate the in vivo effects of salirasib

To investigate the in vivo effects of salirasib, either salirasib or control vehicle was i.p. injected daily into BC xenograft mice from one day after tumor implantation. There was no difference in tumor growth between the salirasib-treated group (n=5, 545.9±187.4 mm3) and control group (n=4, 511.2±165.6 mm3) (Fig. 6) on day 27 after tumor implantation.

Discussion

HRAS was the first human oncogene reported in the T24 BC cell line in 1982 (28). Several reports have indicated that HRAS mutations critically influence tumorigenesis and development of BC (8,2933). Haliassos et al (29) detected HRAS codon 12 point mutations in 66% of BC specimens and the mutant HRAS allele in the urine of 47% of patients with BC, Pandith et al (33) reported that HRAS single nucleotide polymorphism increases BC risk, and rare allele is a predictive marker of advanced bladder tumors. However, RAS had been considered to be 'undruggable' because the RAS protein lacked a druggable binding pocket until salirasib was produced. Salirasib inhibits RAS-dependent cell growth by dislodging all isoforms of RAS from the plasma membrane (11,12). The anti-tumor efficacy of salirasib has been demonstrated in several cell lines and xenograft models (17,3436). Goldberg et al (34) demonstrated that salirasib induces pancreatic cancer cell death and tumor shrinkage in mice, and that salirasib was efficient and nontoxic for treatment of glioblastoma in a rat model (35). Charette et al (36) reported that salirasib inhibits the growth of hepatocarcinoma cell lines in vitro and in vivo through RAS and mTOR inhibition. Salirasib was evaluated as a single agent in two clinical trials; however, neither produced promising results in patients with KRAS mutation positive lung adenocarcinoma (13) or refractory hematologic malignancies (14). Even though these clinical trials demonstrated the relative safety of salirasib, diarrhea, nausea and fatigue were the most common toxicities, and there were no grade 4 or 5 drug-associated adverse events or dose-limiting toxicity. On the other hand, results of a combination study of salirasib with gemcitabine to treat pancreatic adenocarcinoma were sufficiently encouraging to warrant further investigation (15,16).

Although the efficacy of salirasib has been reported for several types of cancers, to the best of our knowledge, this is the first report concerning the effect of salirasib in BC. Two BC cell lines were used to evaluate the ability of salirasib to target HRAS. T24 carries the HRASG12V mutation (substitution of glycine by valine at codon 12 of HRAS) and sequencing data demonstrated that BOY cells have wild-type HRAS. siRNA-induced HRAS knockdown and salirasib inhibition of HRAS exerted tumor suppressive effects regardless of HRAS mutational status in vitro, which was consistent with several previously published results demonstrating a lack of correlation between RAS mutational status and response to RAS-targeting therapy (37,38). However, salirasib still required relatively high concentrations to achieve a tumor-suppressive effect in vitro, and exhibited no tumor-suppressive effects in vivo.

It had been reported that oncogenic RAS predominantly affects the metabolic reprogramming of cancer cells through the upregulation of HIF-1α, one of target genes of RAS (5). Although salirasib is known to competitively block intracellular signaling via the RAS cascade, there are no reports concerning comprehensive metabolomic analysis of salirasib mechanisms. In the current study, proteomic analysis was performed using iMPAQT to investigate metabolic changes in salirasib-treated BC cells. Pathway analysis using the proteomic data indicated that 50 pathways were significantly downregulated following salirasib treatment of BC cells, including 'Oxidative phosphorylation', 'Glycolysis/gluconeogenesis', and 'Pentose phosphate pathway'. However, proteomic analysis showed that the expression of proteins downstream of HIF-1α were not significantly downregulated. Furthermore, HIF-1α expression was not efficiently suppressed in salirasib-treated BC cells, although it was previously reported that salirasib suppressed HIF-1α expression in other type of cancer cells (27). Therefore, downregulation of RAS target genes in in vitro assays involving BC cell lines may require a high concentration of salirasib, and this need for high concentrations was responsible for the lack of tumor suppressive effects observed in the BC xenograft mouse model. In this study, whether factors downstream of HIF-1α were insufficiently downregulated in the tumors from animal experiments was not analyzed by iMPAQT, because iMPAQT is so sensitive that contamination of surrounding tissues adjacent to tumor tissue may make the interpretation of the results difficult. However, these analyses of micro-dissected in vivo samples will be performed in the future. Recently, a novel RAS inhibitor developed using an innovative approach was reported to inhibit tumor growth in animal models of RAS-dependent cancers at low concentrations (39). This novel RAS inhibitor was computationally designed to target multiple sites on RAS proteins, thus enabling sufficient affinity and selectivity for pharmacological RAS inhibition. This new inhibitor may provide successful targeting of RAS in the near future. Therefore, clinical trials with these inhibitors or next-generation RAS inhibitors are required to improve cancer treatment options in the near future.

In conclusion, the current study demonstrated that salirasib and siRNA-induced HRAS knockdown produced tumor suppressive effects regardless of HRAS mutational status in BC cell lines. However, high concentrations of salirasib were required to inhibit cell proliferation, migration and invasion activity in vitro, and the same high concentrations exhibited no tumor suppressive effects in vivo. Proteomic analysis revealed that several metabolic pathways were significantly downregulated in BC cells treated with salirasib. However, salirasib treatment of BC cells did not significantly affect expression of genes targeted by HIF-1α in BC cells. These findings provide novel information concerning the mechanism of salirasib effects, and suggest that novel therapeutics involving combination therapies of salirasib with other inhibitors, or the newly-identified novel RAS inhibitor, may be effective for treating BC and other types of cancer.

Acknowledgments

The authors wish to thank Ms. Mutsumi Miyazaki, (Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan) for laboratory assistance.

Funding

This study was supported by JSPS Grants-in-Aid for Scientific Research (grant nos. 16H05464, 17H04332 and 16K11015).

Availability of data and materials

The analyzed datasets generated during the study are available from the corresponding author on reasonable request.

Authors' contributions

HY conceived and designed the experiments. SS, HY, KM, MY, TS and YO performed the experiments. SS, HE, HY and KM performed the validation and formal analysis. SS and KM wrote the manuscript. HE, HY and MN interpreted experimental data for the work, and reviewed and revised the manuscript. HE, HY and MN acquired funding. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, Forman D and Bray F: Cancer incidence and mortality patterns in Europe: Estimates for 40 countries in 2012. Eur J Cancer. 49:1374–1403. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Abdollah F, Gandaglia G, Thuret R, Schmitges J, Tian Z, Jeldres C, Passoni NM, Briganti A, Shariat SF, Perrotte P, et al: Incidence, survival and mortality rates of stage-specific bladder cancer in United States: A trend analysis. Cancer Epidemiol. 37:219–225. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Advanced Bladder Cancer Meta-analysis Collaboration: Neoadjuvant chemotherapy in invasive bladder cancer: A systematic review and meta-analysis. Lancet. 361:1927–1934. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Pylayeva-Gupta Y, Grabocka E and Bar-Sagi D: RAS oncogenes: Weaving a tumorigenic web. Nat Rev Cancer. 11:761–774. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Karnoub AE and Weinberg RA: Ras oncogenes: Split personalities. Nat Rev Mol Cell Biol. 9:517–531. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K, Sougnez C, Greulich H, Muzny DM, Morgan MB, et al: Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 455:1069–1075. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Kiaris H and Spandidos D: Mutations of ras genes in human tumors (Review). Int J Oncol. 7:413–421. 1995.PubMed/NCBI

9 

Baines AT, Xu D and Der CJ: Inhibition of Ras for cancer treatment: The search continues. Future Med Chem. 3:1787–1808. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Cox AD, Fesik SW, Kimmelman AC, Luo J and Der CJ: Drugging the undruggable RAS: Mission possible? Nat Rev Drug Discov. 13:828–851. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Elad G, Paz A, Haklai R, Marciano D, Cox A and Kloog Y: Targeting of K-Ras 4B by S-trans, transfarnesyl thiosalicylic acid. Biochim Biophys Acta. 1452:228–242. 1999. View Article : Google Scholar : PubMed/NCBI

12 

Weisz B, Giehl K, Gana-Weisz M, Egozi Y, Ben-Baruch G, Marciano D, Gierschik P and Kloog Y: A new functional Ras antagonist inhibits human pancreatic tumor growth in nude mice. Oncogene. 18:2579–2588. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Riely GJ, Johnson ML, Medina C, Rizvi NA, Miller VA, Kris MG, Pietanza MC, Azzoli CG, Krug LM, Pao W, et al: A phase II trial of Salirasib in patients with lung adenocarcinomas with KRAS mutations. J Thorac Oncol. 6:1435–1437. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Badar T, Cortes JE, Ravandi F, O'Brien S, Verstovsek S, Garcia-Manero G, Kantarjian H and Borthakur G: Phase I study of S-trans, trans-farnesylthiosalicylic acid (salirasib), a novel oral RAS inhibitor in patients with refractory hematologic malignancies. Clin Lymphoma Myeloma Leuk. 15:433–438.e2. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Bustinza-Linares E, Kurzrock R and Tsimberidou AM: Salirasib in the treatment of pancreatic cancer. Future Oncol. 6:885–891. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Laheru D, Shah P, Rajeshkumar NV, McAllister F, Taylor G, Goldsweig H, Le DT, Donehower R, Jimeno A, Linden S, et al: Integrated preclinical and clinical development of S-trans, trans-Farnesylthiosalicylic Acid (FTS, Salirasib) in pancreatic cancer. Invest New Drugs. 30:2391–2399. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Makovski V, Haklai R and Kloog Y: Farnesylthiosalicylic acid (salirasib) inhibits Rheb in TSC2-null ELT3 cells: A potential treatment for lymphangioleiomyomatosis. Int J Cancer. 130:1420–1429. 2012. View Article : Google Scholar

18 

Zundelevich A, Elad-Sfadia G, Haklai R and Kloog Y: Suppression of lung cancer tumor growth in a nude mouse model by the Ras inhibitor salirasib (farnesylthiosalicylic acid). Mol Cancer Ther. 6:1765–1773. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Matsumoto M, Matsuzaki F, Oshikawa K, Goshima N, Mori M, Kawamura Y, Ogawa K, Fukuda E, Nakatsumi H, Natsume T, et al: A large-scale targeted proteomics assay resource based on an in vitro human proteome. Nat Methods. 14:251–258. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Li B and Dewey CN: RSEM: Accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 12:3232011. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Ichimi T, Enokida H, Okuno Y, Kunimoto R, Chiyomaru T, Kawamoto K, Kawahara K, Toki K, Kawakami K, Nishiyama K, et al: Identification of novel microRNA targets based on microRNA signatures in bladder cancer. Int J Cancer. 125:345–352. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Yoshino H, Chiyomaru T, Enokida H, Kawakami K, Tatarano S, Nishiyama K, Nohata N, Seki N and Nakagawa M: The tumoursuppressive function of miR-1 and miR-133a targeting TAGLN2 in bladder cancer. Br J Cancer. 104:808–818. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Masoud GN and Li W: HIF-1α pathway: Role, regulation and intervention for cancer therapy. Acta Pharm Sin B. 5:378–389. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Qing G, Skuli N, Mayes PA, Pawel B, Martinez D, Maris JM and Simon MC: Combinatorial regulation of neuroblastoma tumor progression by N-Myc and hypoxia inducible factor HIF-1alpha. Cancer Res. 70:10351–10361. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Hong SS, Lee H and Kim KW: HIF-1alpha: A valid therapeutic target for tumor therapy. Cancer Res Treat. 36:343–353. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Hameiri-Grossman M, Porat-Klein A, Yaniv I, Ash S, Cohen IJ, Kodman Y, Haklai R, Elad-Sfadia G, Kloog Y, Chepurko E, et al: The association between let-7, RAS and HIF-1α in Ewing Sarcoma tumor growth. Oncotarget. 6:33834–33848. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Shih C and Weinberg RA: Isolation of a transforming sequence from a human bladder carcinoma cell line. Cell. 29:161–169. 1982. View Article : Google Scholar : PubMed/NCBI

29 

Haliassos A, Liloglou T, Likourinas M, Doumas C, Ricci N and Spandidos D: H-ras oncogene mutations in the urine of patients with bladder-tumors - description of a noninvasive method for the detection of neoplasia. Int J Oncol. 1:731–734. 1992.PubMed/NCBI

30 

Zhang X and Zhang Y: Bladder cancer and genetic mutations. Cell Biochem Biophys. 73:65–69. 2015. View Article : Google Scholar

31 

Kompier LC, Lurkin I, van der Aa MN, van Rhijn BW, van der Kwast TH and Zwarthoff EC: FGFR3, HRAS, KRAS, NRAS and PIK3CA mutations in bladder cancer and their potential as biomarkers for surveillance and therapy. PLoS One. 5:e138212010. View Article : Google Scholar : PubMed/NCBI

32 

Beukers W, Hercegovac A and Zwarthoff EC: HRAS mutations in bladder cancer at an early age and the possible association with the Costello Syndrome. Eur J Hum Genet. 22:837–839. 2014. View Article : Google Scholar :

33 

Pandith AA, Shah ZA, Khan NP, Baba KM, Wani MS and Siddiqi MA: HRAS T81C polymorphism modulates risk of urinary bladder cancer and predicts advanced tumors in ethnic Kashmiri population. Urol Oncol. 31:487–492. 2013. View Article : Google Scholar

34 

Goldberg L, Israeli R and Kloog Y: FTS and 2-DG induce pancreatic cancer cell death and tumor shrinkage in mice. Cell Death Dis. 3:e2842012. View Article : Google Scholar : PubMed/NCBI

35 

Goldberg L, Ocherashvilli A, Daniels D, Last D, Cohen ZR, Tamar G, Kloog Y and Mardor Y: Salirasib (farnesyl thiosalicylic acid) for brain tumor treatment: A convection-enhanced drug delivery study in rats. Mol Cancer Ther. 7:3609–3616. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Charette N, De Saeger C, Lannoy V, Horsmans Y, Leclercq I and Stärkel P: Salirasib inhibits the growth of hepatocarcinoma cell lines in vitro and tumor growth in vivo through ras and mTOR inhibition. Mol Cancer. 9:2562010. View Article : Google Scholar : PubMed/NCBI

37 

Kurzrock R, Kantarjian HM, Blascovich MA, Bucher C, Verstovsek S, Wright JJ, Pilat SR, Cortes JE, Estey EH, Giles FJ, et al: Phase I study of alternate-week administration of tipifarnib in patients with myelodysplastic syndrome. Clin Cancer Res. 14:509–514. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Lancet JE, Gojo I, Gotlib J, Feldman EJ, Greer J, Liesveld JL, Bruzek LM, Morris L, Park Y, Adjei AA, et al: A phase 2 study of the farnesyltransferase inhibitor tipifarnib in poor-risk and elderly patients with previously untreated acute myelogenous leukemia. Blood. 109:1387–1394. 2007. View Article : Google Scholar

39 

Welsch ME, Kaplan A, Chambers JM, Stokes ME, Bos PH, Zask A, Zhang Y, Sanchez-Martin M, Badgley MA, Huang CS, et al: Multivalent small-molecule Pan-RAS inhibitors. Cell. 168:878–889.e29. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 53 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sugita S, Enokida H, Yoshino H, Miyamoto K, Yonemori M, Sakaguchi T, Osako Y and Nakagawa M: HRAS as a potential therapeutic target of salirasib RAS inhibitor in bladder cancer. Int J Oncol 53: 725-736, 2018
APA
Sugita, S., Enokida, H., Yoshino, H., Miyamoto, K., Yonemori, M., Sakaguchi, T. ... Nakagawa, M. (2018). HRAS as a potential therapeutic target of salirasib RAS inhibitor in bladder cancer. International Journal of Oncology, 53, 725-736. https://doi.org/10.3892/ijo.2018.4435
MLA
Sugita, S., Enokida, H., Yoshino, H., Miyamoto, K., Yonemori, M., Sakaguchi, T., Osako, Y., Nakagawa, M."HRAS as a potential therapeutic target of salirasib RAS inhibitor in bladder cancer". International Journal of Oncology 53.2 (2018): 725-736.
Chicago
Sugita, S., Enokida, H., Yoshino, H., Miyamoto, K., Yonemori, M., Sakaguchi, T., Osako, Y., Nakagawa, M."HRAS as a potential therapeutic target of salirasib RAS inhibitor in bladder cancer". International Journal of Oncology 53, no. 2 (2018): 725-736. https://doi.org/10.3892/ijo.2018.4435