Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Experimental Therapeutics, Molecular Targets, and Chemical Biology

Flavopiridol Targets c-KIT Transcription and Induces Apoptosis in Gastrointestinal Stromal Tumor Cells

Elliot B. Sambol, Grazia Ambrosini, Rula C. Geha, Peter T. Kennealey, Penelope DeCarolis, Rachael O'Connor, Yuhsin V. Wu, Monica Motwani, Jin-Hong Chen, Gary K. Schwartz and Samuel Singer
Elliot B. Sambol
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Grazia Ambrosini
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rula C. Geha
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Peter T. Kennealey
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Penelope DeCarolis
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rachael O'Connor
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yuhsin V. Wu
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Monica Motwani
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jin-Hong Chen
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Gary K. Schwartz
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Samuel Singer
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/0008-5472.CAN-05-2933 Published June 2006
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Gastrointestinal stromal tumors (GIST) are characterized by activating mutations in the c-KIT gene which confers ligand-independent activation of the KIT receptor. Imatinib mesylate has been shown to effectively block constitutively active KIT and delay tumor growth. However, resistance to imatinib mesylate is emerging as a major clinical problem and novel therapies are needed. We report that treatment of GIST cells with the transcriptional inhibitor flavopiridol, initially down-regulates the antiapoptotic proteins bcl-2, mcl-1, and X-linked inhibitor of apoptosis protein which occurs as early as 4 hours after exposure. This is followed at 24 hours by the transcriptional suppression of KIT resulting in poly(ADP-ribose) polymerase cleavage and apoptosis. To separate the apoptotic effect of KIT suppression relative to the down-regulation of antiapoptotic proteins, we used small interfering RNA–directed knockdown of KIT. Results show that focused suppression of KIT alone is sufficient to induce apoptosis in GIST cells, but not to the same extent as flavopiridol. In contrast, imatinib mesylate, which inhibits KIT kinase activity but does not suppress total KIT expression, fails to cause apoptosis. We also show that flavopiridol suppresses KIT mRNA expression through positive transcriptional elongation factor inhibition and decreases KIT promoter activity. This causes a global decrease in the level of functionally mature KIT at the cell surface, resulting in a decrease in autophosphorylation at tyrosine residues 703 and 721, which characterizes activated KIT. Our results indicate that targeting KIT expression and these antiapoptotic proteins with flavopiridol represents a novel means to disrupt GIST cell dependence on KIT signaling and collectively renders these cells sensitive to apoptosis. (Cancer Res 2006; 66(11): 5858-66)

  • flavopiridol
  • GIST
  • KIT
  • P-TEFb

Introduction

The c-KIT gene product is a 145 kDa transmembrane protein belonging to the type III receptor tyrosine kinase family. It is an integral cellular component required for normal hematopoiesis, melanogenesis, gametogenesis, and the growth and differentiation of mast cells and the interstitial cells of Cajal ( 1). Mutations in the c-KIT gene resulting in its constitutive activation have been implicated in various cancers including germ cell tumors ( 2), mastocytosis ( 3), small cell lung cancers ( 4), breast cancers ( 5), and gastrointestinal stromal tumors (GIST; ref. 6). The KIT receptor has become a model for targeted therapeutics with the advent of imatinib mesylate (Gleevec, Novartis Pharma AG, Basel, Switzerland). Imatinib mesylate is a tyrosine kinase inhibitor that preferentially blocks the constitutively active mutated forms of the KIT ( 7), bcr-abl ( 8), and platelet-derived growth factor receptors ( 9). KIT inhibition by imatinib mesylate abrogates receptor dimerization and resultant autophosphorylation suppressing downstream activity. In KIT-dependent GIST cell lines, imatinib mesylate has been shown to be highly active as an antiproliferative agent ( 10, 11) and is now used clinically for the treatment of advanced GIST.

Despite the effectiveness of imatinib mesylate in ∼80% of patients with metastatic GIST ( 12), resistance is emerging as a major clinical problem. Some 20% of GIST patients show overt disease progression within 3 months of exposure indicating primary resistance to imatinib mesylate. In addition, ∼50% of patients who initially respond to imatinib mesylate, relapse after 2 years ( 13). The mechanism for this acquired resistance is postulated to occur because of: (a) secondary mutations, (b) amplification of KIT, and (c) alternative signaling kinases driving cell growth ( 14). For these reasons, it has become essential to identify novel therapeutics for GIST that, although targeting the KIT receptor, work through different pathways.

One potential agent that has gained interest is flavopiridol, a semisynthetic flavone, and one of the few cyclin-dependent kinase inhibitors that has been extensively studied in multiple phase I and II clinical trials ( 15). It primarily functions by blocking CDK activity by competitively interfering with the docking of ATP in the adenine-rich binding pockets of these and other enzymes ( 16). Despite having a high affinity for the CDKs, flavopiridol is not specific to only these enzymes. It has also been shown to inhibit the src family of kinases ( 17), as well as inhibit phosphorylation of survivin ( 18). In addition, by suppressing CDK9-cyclin T function ( 19), also known as the positive transcriptional elongation factor (P-TEFb), flavopiridol becomes a global transcriptional repressor of numerous genes ( 20). Studies have shown that flavopiridol as a single agent has antiproliferative activity in non–small cell lung, breast, prostate, glioma, and head and neck cancer cell lines ( 21). To date, however, it has not been studied in GIST, nor has it been shown to modulate the KIT pathway.

Considering the requirement for constitutive KIT signaling in GIST, we questioned whether flavopiridol could be an effective alternative to imatinib mesylate by inducing transcriptional suppression of the KIT receptor. We hypothesized that the cellular loss of total KIT protein would result in a reduction in active autophosphorylated KIT leading to subsequent downstream silencing and eventually apoptosis. Furthermore, the global transcriptional effect induced by flavopiridol may further enhance GIST cell apoptosis by altering the milieu of antiapoptotic signals. Using the GIST cell line, GIST882 ( 10), we show that flavopiridol blocks KIT promoter activity and inhibits RNA polymerase II (RNA pol II) activation, thereby reducing transcribable mRNA. This causes a time-dependent cellular depletion in mcl-1, bcl-2, X-linked inhibitor of apoptosis protein (XIAP), and total KIT protein, with a reduction in KIT surface expression that potently induces an apoptotic response in these cells that is superior to imatinib mesylate. We show that targeting KIT and these antiapoptotic proteins at the transcriptional level could be more effective than direct receptor inhibition.

Materials and Methods

Cell lines and drug preparation. GIST882 cells (kindly provided by Dr. J. Fletcher, Department of Pathology, Harvard Medical School, Boston, MA) harbor a homozygous missense mutation in c-KIT exon 13 (K642E) confirmed by standard DNA sequencing ( 10). GIST882 was maintained in standard RPMI 1640 supplemented with 2% glutamate and either 15% heat-inactivated fetal bovine serum (FBS) or 10% CELLect human serum (Fisher, Fair Lawn, NJ). HCT 116 cells (American Type Culture Collection, Rockville, MD) were maintained in RPMI supplemented with 10% FBS. All cells were cultured in media supplemented with 1% penicillin/streptomycin (Invitrogen, Carlsbad, CA) and kept at 37°C and humidified in 5% CO2. Flavopiridol (National Cancer Institute, Bethesda, MD) and imatinib mesylate (Novartis Pharma) stock solutions were prepared in water. 5,6-Dichloro-1β-d-ribofuranosylbenzimidazole (DRB; Sigma, St. Louis, MO) was resuspended in DMSO.

Proliferation assays. To evaluate the antiproliferative effect of drug exposure on GIST cells, DNA content was estimated using the CyQuant Cell Proliferation Kit (Molecular Probes, Eugene, OR) as previously described ( 22). In brief, cells were plated in 96-well plates at a density of 2 × 104 cells per well and allowed to adhere overnight at 37°C. After treatment, cells were incubated with CyQuant dye and lysed. This dye exhibits strong fluorescence when bound to cellular nucleic acids, and as such, the fluorescent signal is proportional to the DNA content. Plates were read using the Spectramax M2 fluorescence microplate reader (Molecular Devices, Sunnyvale, CA) at 480/520 nm excitation/emission.

Assessment of apoptosis. Evaluation of apoptosis was carried out using the Guava Nexin Kit (Guava Technologies, Hayward, CA). Briefly, cells were treated with varying concentrations of flavopiridol or imatinib mesylate for 12 to 48 hours in 10 cm culture plates. Both floating and adherent cells were collected and stained with Annexin V and 7-AAD. The Annexin V–positive population was counted using the Guava PCA (Guava Technologies) and data analyzed using FloJo software (Tree Star, Ashland, OR). Both the early and late apoptotic cells were summed to determine total apoptosis. Caspase-3 activity was evaluated using the EnzChek caspase-3 assay kit 1 (Molecular Probes), according to the manufacturer's instructions. Fluorescence (excitation/emission, ∼342/441) was measured using the Spectramax reader (Molecular Devices), and was normalized to the total number of cells per condition.

Western blot. After treatment, cells were washed in cold PBS and lysed in a buffer containing 50 mmol/L HEPES-KOH (pH 7.5), 150 mmol/L NaCl, 1 mmol/L each EDTA, 2.5 mmol/L EGTA, 1 mmol/L DTT, 0.1% Tween 20, 10% glycerol, 10 mmol/L β-glycerophosphate, 1 mmol/L NaF, 0.1 mmol/L Na3VO4, 0.2 mmol/L phenylmethylsulfonyl fluoride, and complete mini-protease inhibitor tabs (Roche, Nutley, NJ). Cell lysates containing 10 to 40 μg of protein were resolved by SDS-PAGE, transferred onto Immuno-Blot polyvinylidene difluoride membranes (Bio-Rad, Hercules, CA) and probed with antibodies including KIT (Dako Cytomation, Carpinteria, CA), phospho-KITY721, phospho-KITY703 (Zymed Laboratories, San Francisco, CA), phospho-ERKY204, total ERK 1/2, bcl-2 (Santa Cruz Biotechnology, Santa Cruz, CA), poly(ADP-ribose) polymerase (PARP), mcl-1, XIAP (BD PharMingen, San Diego, CA), phospho-STAT3Y705, phospho-STAT1Y701, total STAT3 (Cell Signaling, Beverly, MA), total STAT1 (Transduction Laboratories, Lexington, KY), total RNA pol II and phosphorylated RNA pol II at Ser2 on the carboxyl-terminal domain (CTD; Covance Research Products, Berkeley, CA) and α-tubulin (Calbiochem, San Diego, CA). Detection was done with enhanced chemiluminescence reagents (Amersham Biosciences, England).

Quantitative real-time reverse transcription PCR. RNA isolation was done using RNeasy Mini Kit (Qiagen, Valencia, CA) from ∼20 million cells. Reverse transcription was done with 1.5 μg of RNA in a 100 μL reaction using random hexamer priming and TaqMan reverse transcription reagents (Applied Biosystems, Foster City, CA) on a Thermo Hybaid thermocycler (Thermo Hybaid, Waltham, MA). All quantitative real-time reverse transcription PCR (qPCR) assays were done on the ABI Prism 7900HT Sequence Detection System and analyzed using SDS version 2.1 software (Applied Biosystems). TaqMan Gene Expression Assays (Applied Biosystems), which include gene-specific probe and primer sets were used according to the manufacturer's protocol to detect both c-KIT (Hs00174029_m1) and 18s rRNA (Hs99999901_s1). The relative expression of KIT was calculated by normalizing KIT expression to 18s rRNA.

Plasmid construction. A genomic DNA fragment of the human c-KIT promoter ( 23) containing 720 bp of the 5′-flanking region was prepared by PCR amplification of human genomic DNA (Clontech, Mountain View, CA). The following primers were used: 5′-CTGGTACCGTCTCCACACAAGTCCCGGC-3′ (forward) and 5′-AATCAAGCTTGCTCTGGTCCACGTTCCAGCTC-3′ (reverse) containing 5′-overhang restriction sites KpnI and HindIII, respectively (underlined). A standard PCR reaction was conducted for 30 cycles using a PTC-200 Peltier Thermal Cycler (MJ Research, Watertown, MA). The purified fragment was subcloned upstream of the firefly luciferase reporter gene in a pGL2-Basic vector (Promega, Madison, WI). The sequence of the resulting construct (pGL2-KIT) was verified by automated DNA sequencing.

Luciferase reporter assay. Transient transfections on HCT 116 cells were done in six-well plates using Fugene 6 transfection reagent (Roche). Plasmid DNA consisting of the promoter construct, pGL2-KIT (1 μg/well) and pRL-TK, a Renilla luciferase vector (0.05 μg/well) was then added to the diluted transfection reagent in a 3:1 reagent/DNA ratio. Twenty-four hours after transfection, cells were treated with drug for an additional 24 hours, then harvested in 200 μL of 1× passive lysis buffer (Dual-Luciferase Reporter Assay, Promega). Both firefly and Renilla luciferase activity was analyzed using a TD 20/20 Luminometer (Turner Designs, Sunnyvale, CA). pGL-2-KIT luciferase activity was normalized to Renilla luciferase to control for variations in transfection efficiency.

Analysis of KIT surface expression. After drug treatment, both floating and adherent cells were collected for fluorescence-activated cell sorting (FACS) analysis. Approximately 3 × 105 cells were stained with phycoerythrin-conjugated KIT monoclonal antibody (BD PharMingen). To control for nonspecificity, cells were similarly stained with a phycoerythrin-conjugated isotype IgG1 monoclonal antibody. Following staining, surface expression was determined using a FACSCalibur flow cytometer (BD Immunocytometry Systems, San Jose, CA) and analyzed using FloJo software.

Small interfering RNA experiments. c-KIT and control small interfering RNAs (siRNA; 800 ng/mL) were obtained from the SureSilencing human c-KIT siRNA kit (SuperArray Biosciences, Frederick, MD) and used according to the recommendations of the manufacturer. Subconfluent GIST882 cells in six-well plates were transiently transfected with the siRNAs in OptiMEM I medium (Invitrogen) using OligofectAMINE transfection reagent (Invitrogen). After 4 hours of incubation, the medium was changed to a complete culture medium. Apoptotic effects were evaluated 72 hours after transfection using Annexin V/7-AAD as described above.

Statistical analysis. Student's t test was used to determine significant differences between two means (P < 0.05).

Results

Flavopiridol has potent antiproliferative effects in GIST882 cells. In agreement with previous studies ( 10), GIST cell proliferation, as measured by nucleic acid content, was inhibited by imatinib mesylate exposure in a dose- and time-dependent fashion, with up to 60% inhibition by day 5 ( Fig. 1A ). The maximal effect was achieved at 500 nmol/L with no substantial difference even at 20 times (10 μmol/L) that dose. This plateau of activity is important because it has been shown that imatinib mesylate binds α1 acid glycoprotein in human plasma, which decreases the level of unbound active imatinib mesylate by ∼90% ( 24). By testing a considerably higher dose (10 μmol/L) in RPMI which has lower acid glycoprotein levels compared with human plasma, we failed to increase cytotoxicity. Therefore, we used 1 μmol/L for all subsequent experimentations. With flavopiridol exposure, GIST proliferation relative to control cells was markedly reduced, achieving >80% inhibition by day 5 at 500 nmol/L ( Fig. 1B). These viability assays reveal that both drugs are capable of suppressing GIST cell growth. However, in comparison, 300 nmol/L of flavopiridol decreased viability by 77 ± 0.8% compared with 63 ± 2.7% for 1 μmol/L imatinib mesylate at 5 days (P < 0.001). The combination of flavopiridol and imatinib mesylate was essentially equivalent to flavopiridol alone (data not shown).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Dose- and time-dependent cytotoxicity profile of GIST882 cells following exposure to flavopiridol or imatinib. A and B, exponentially growing GIST882 cells were exposed to varying concentrations of either imatinib mesylate (A) or flavopiridol (B) in RPMI supplemented with 10% FBS, and cell proliferation was determined at 1 to 5 days. Experiments were done in 96-well plates using the CyQuant cell proliferation kit according to the manufacturer's instructions. Points, mean fluorescence of six replicate wells relative to untreated controls; bars, ±SD; bottom left, concentrations used. The experiments were repeated on three separate occasions with similar results.

Flavopiridol mediates its antiproliferative effect through a potent induction of apoptosis in GIST cells that is superior to imatinib mesylate. To further characterize the antiproliferative effect of flavopiridol in GIST cells, we evaluated the apoptotic response by Annexin V labeling. As shown in Fig. 2A , 12 hours of exposure was insufficient to induce any measurable increase in apoptosis above the baseline (<10%) across all drug conditions tested. At 24 and 48 hours, 150 nmol/L of flavopiridol and 1.0 μmol/L of imatinib mesylate still failed to cause a significant apoptotic response with essentially no increase in Annexin V labeling above the baseline. With 300 nmol/L of flavopiridol, however, there was a profound increase in the level of apoptosis with ∼42% and 58% Annexin V staining at 24 and 48 hours, respectively. This represents a 3.0- and 4.5-fold higher level of apoptosis induction following 300 nmol/L of flavopiridol exposure compared with 1.0 μmol/L of imatinib mesylate (P < 0.005). In addition, the induction of apoptosis did not seem to appreciably increase with imatinib mesylate concentrations up to 10 μmol/L (data not shown). The effect of flavopiridol was dependent on caspase-3 activity ( 25), as evidenced by the significant increase in cleavage product measured using a fluorescence-based assay ( Fig. 2B). Flavopiridol treatment at 300 nmol/L resulted in a 4.4- and 3.9-fold higher level of activity at 24 and 48 hours, respectively, compared with imatinib mesylate (P < 0.001). However, there was no significant increase in caspase activity above the baseline at 12 hours with 300 nmol/L of flavopiridol, in concert with the lack of early Annexin V labeling.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Flavopiridol-mediated apoptosis is superior to imatinib mesylate and is dose- and time-dependent. GIST882 cells were treated with either flavopiridol or imatinib mesylate and harvested for apoptosis assays. A, 12, 24, and 48-hour treated cells were stained with Annexin V/7-AAD and analyzed using FACS. Bottom left quadrant, accumulation of viable (double-negative) cells in the four-quadrant gate; bottom right quadrant, early apoptotic cells, staining with Annexin V only; top right quadrant, double-positive staining cells undergoing late apoptosis and early necrosis. The numbers represent the percentage of the cell population in each of the respective gates. The sum of the Annexin V–positive quadrants provides the total percentage of apoptotic cells. The experiments were done at least thrice with similar findings and these dot plots are representative of one of these experiments. B, caspase-3 activity was determined after 12, 24, and 48 hours of drug exposure using the EnzCheck Caspase-3 Assay Kit as described in Materials and Methods. Columns, mean of relative caspase-3 activity calculated by normalizing fluorescence to the total number of cells per sample; bars, ±SD. Experiments were repeated and done in triplicate. *, P < 0.005 versus all other conditions. C, GIST882 cells were cultured and treated for 24 hours in RPMI containing 10% human serum. Annexin V/7-AAD and FACS analysis was done as described previously.

Considering that flavopiridol, like imatinib mesylate, is also highly bound to plasma proteins ( 26), we sought to test the effects of human plasma on the capacity of flavopiridol to induce apoptosis. As such, Annexin V labeling done in cells cultured in 10% human serum exhibited an almost complete loss of apoptotic effect at 300 nmol/L ( Fig. 2C). However, by increasing the flavopiridol concentration to 3.0 μmol/L, a clinically achievable dose, 65% of the cells underwent apoptosis. Thus, under conditions which recapitulate those achieved in human plasma, we can still induce high levels of apoptosis with flavopiridol in GIST cells.

Flavopiridol-mediated down-regulation of KIT induces apoptosis. Based on the apoptotic response observed with flavopiridol exposure, we sought to examine the molecular changes associated with this effect. The ability of flavopiridol to down-regulate antiapoptotic proteins such as XIAP, mcl-1, and bcl-2, which occurs in other systems ( 27), was verified in the GIST model ( Fig. 3A ). As evidenced by Western blot analysis, these three antiapoptotic proteins were almost completely suppressed after 24 hours of exposure to 300 nmol/L of flavopiridol. Concomitant with this loss was PARP cleavage; the 85 kDa cleaved product of PARP is identified only in the 300 nmol/L flavopiridol-treated cells following 24 hours of exposure. Flavopiridol at 150 nmol/L and imatinib mesylate at 1.0 μmol/L did not significantly change the expression of XIAP, mcl-1, or bcl-2, and PARP cleavage did not occur, which is consistent with the lack of apoptosis observed under these drug conditions.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Flavopiridol-mediated effects on antiapoptotic proteins, PARP cleavage, and KIT, and the induction of apoptosis by siRNA knockdown of KIT. A, Western blot of 24-hour drug-treated cellular extracts. The cleavage product of PARP is identified as an 85 kDa band. Membranes were probed for α-tubulin to confirm equal protein loading. These blots were done on three independent occasions with similar results. B, GIST882 cells were cultured in the presence of 300 nmol/L flavopiridol for various time intervals and cellular lysates were subjected to Western blot analysis. Blots were probed for KIT, antiapoptotic proteins, PARP cleavage, and tubulin. C, GIST882 cells were transfected with siRNA oligos directed at KIT as well as a control siRNA. Western blot analysis shows the efficiency of knockdown after 72 hours with tubulin loading control. D, apoptosis was determined at 72 hours after siRNA transfection using Annexin V/7-AAD labeling as previously described. Top left quadrant, the numbers represent the percentage of cells in each gate with the exception of the viable cell population (bottom left quadrant). These experiments were repeated thrice and are representative of one experiment.

In examining the time dependence for down-regulation of the antiapoptotic milieu ( Fig. 3B), 300 nmol/L of flavopiridol caused an early reduction in bcl-2, mcl-1, and XIAP. Compared with untreated cells, bcl-2 and mcl-1 were reduced as early as 4 hours after exposure, and by 24 hours, were undetectable. XIAP down-regulation began by 12 hours and also progressively decreased to undetectable levels. KIT protein, however, remained relatively stable until 24 hours, at which point, we observed significant reductions compared with earlier time points. However, PARP cleavage and apoptosis only occurred following 24 hours of flavopiridol exposure, concomitant with KIT down-regulation.

The observation that KIT down-regulation occurred in conjunction with apoptosis, whereas the early decrease in XIAP, mcl-1, and bcl-2 was not associated with an observable apoptotic response, suggested that KIT down-regulation may in itself be required for cell death. In order to separate the effect of KIT suppression from that of mcl-1, bcl-2, and XIAP in these cells, we used a focused siRNA-based approach to substantially disrupt KIT protein levels after a 72-hour transfection in GIST cells. As shown in Fig. 3C, compared with the control siRNA, KIT siRNA reduced KIT protein expression by ∼80%, demonstrating a high efficiency of knockdown. Staining with Annexin V ( Fig. 3D) revealed substantial apoptosis in siRNA-treated cells (29 ± 2.3%) compared with the control siRNA (9.7 ± 0.53%; P < 0.01). This data supports the notion that a reduction in KIT protein, by itself, induces an apoptotic response in GIST cells. However, this still remains less than that observed with flavopiridol, suggesting that the maximal induction of apoptosis with flavopiridol reflects a collective suppression of both KIT, as well as the antiapoptotic proteins.

Suppression of total KIT by flavopiridol causes a loss in autophosphorylated active KIT. The constitutive activity of mutant KIT is identified by autophosphorylation of the receptor at the cell surface. Considering that apoptosis induction by flavopiridol occurs concomitant with KIT down-regulation, we sought to examine the changes in KIT autophosphorylation following flavopiridol treatment. Phospho-KIT at Y721 is the association site for phosphoinositide-3-kinase, which is one of the first proteins to be phosphorylated in the KIT cascade ( 28). Phospho-KIT at Y703 has been shown to be the binding partner for the adapter protein GRB2 which activates the ras pathway leading to cell growth ( 29). As shown in Fig. 4A , GIST882 cells in drug-free medium have constitutively active KIT with no change in total KIT protein over time. Flavopiridol (150 nmol/L) had no effect on inhibiting KIT autophosphorylation at either Y721 or Y703, and there were only minor reductions in total KIT protein expression observed over time. As anticipated, 1.0 μmol/L of imatinib mesylate completely abolished KIT phosphorylation at both residues within 2 hours of exposure to the drug. This is a result of direct binding of imatinib mesylate into the juxtamembrane pocket of the receptor with subsequent inhibition of kinase activity ( 30). There was no change in total KIT across time points. In contrast, with 300 nmol/L of flavopiridol, there was no immediate effect on the suppression of KIT autophosphorylation at 2 or 4 hours. However, after 24 hours of flavopiridol exposure, phospho-KIT Y721 was almost completely abolished and Y703 was drastically reduced. Thus, the concomitant decrease in total KIT protein levels observed at 24 to 48 hours depletes the pool of available receptor to undergo autophosphorylation.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Flavopiridol causes a time-dependent reduction in KIT autophosphorylation through a loss of total KIT protein. A, protein lysates from GIST882 cells treated with flavopiridol (150 and 300 nmol/L) or imatinib mesylate (1.0 μmol/L) for 2, 4, 24, and 48 hours were resolved by SDS-PAGE. Autophosphorylation of the KIT receptor was studied by probing with an antibody specific for the phosphorylated KIT protein at tyrosine 721 (P-KIT Y721) and tyrosine 703 (P-KIT Y703). Total KIT levels were also evaluated in the same lysates revealing both mature (upper band, 145 kDa) and immature (lower band, 125 kDa) KIT forms (arrows). B, FACS analysis of KIT surface expression in GIST882 cells after 24 hours of drug treatment. Histograms, the percentage of cells staining positively for the KIT surface marker using a monoclonal antibody to KIT, conjugated to phycoerythrin (KIT-PE). A, untreated (shaded); B, flavopiridol (150 nmol/L); C, imatinib mesylate (1.0 μmol/L); D, flavopiridol (300 nmol/L). Background, nonspecific KIT staining was determined using a matched IgG1 isotype control antibody (E, shaded). These results are representative of three independent experiments.

Based on the observed loss in total KIT protein with 300 nmol/L of flavopiridol exposure, we elected to determine the extent to which mature surface receptor expression is reduced. This is important because it is the mature form of the receptor that propagates constitutive survival signals. FACS analysis of KIT surface expression was done on GIST882 cells after 24 hours of drug exposure ( Fig. 4B). The untreated population stained uniformly for the KIT receptor (shaded histogram, A). Exposure to 300 nmol/L of flavopiridol (solid line, D), however, resulted in a drastic reduction in KIT surface expression by ∼75% compared with the untreated cells as well as the other drug conditions.

Abrogation of KIT autophosphorylation by flavopiridol reduces activation of downstream signals. The ability of flavopiridol to suppress GIST cell growth in a KIT-dependent manner would require a reduction in the activity of downstream signaling molecules including Ras/Erk, phosphoinositide-3-kinase, mitogen-activated protein kinase, and STAT pathways ( 31, 32). We therefore evaluated the expression and activity of several of these signaling proteins over a time course by probing for the phosphorylated active forms by Western blotting ( Fig. 5A ). We compared the differences in expression after treatment with 300 nmol/L of flavopiridol and 1.0 μmol/L of imatinib mesylate. The data reveals that phosphorylated active STAT1 (p-STAT1 Y701), STAT3 (p-STAT3 Y705), and ERK (p-ERK Y204) were all reduced in both treatment groups. However, with imatinib mesylate treatment, there was an earlier loss of these activated forms which parallels the early abolishment of KIT autophosphorylation ( Fig. 4A). All activated proteins were significantly reduced as early as 4 hours after imatinib mesylate exposure and were undetectable by 24 hours. With flavopiridol treatment, the decrease occurred later. To confirm that the down-regulation of these downstream mediators was not related to generalized inhibition by flavopiridol, we evaluated the amount of total protein. As shown in Fig. 5B, total protein for ERK1/2, STAT1, and STAT3 went unchanged in the flavopiridol and imatinib mesylate treatment groups relative to untreated cells at 24 hours. This suggests that the loss of the phosphorylated active downstream mediators was likely related to reduced constitutive KIT signaling.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

Time-dependent effects on the downstream mediators of the KIT cascade after exposure to imatinib mesylate and flavopiridol. A, KIT pathway activation in GIST882 cells was evaluated after treatment with either flavopiridol or imatinib mesylate for various time intervals. Western blot analysis with phospho-specific antibodies to ERK (p-ERKY204), STAT3 (p-STAT3Y705), and STAT1 (p-STAT1Y701) was done. B, expression of total protein levels (ERK1/2, STAT1, STAT3) after 24 hours of treatment, were analyzed via Western blotting. Equal loading was confirmed with tubulin. These blots are representative of multiple independent experiments.

Flavopiridol inhibits RNA pol II phosphorylation and blocks KIT promoter activity causing a reduction in KIT transcription. Based on our findings of reduced expression of total KIT protein, we sought to explore the mechanism responsible. As a general transcriptional inhibitor which suppresses elongation control by RNA pol II ( 19), we hypothesized that flavopiridol activity functioned through potent reductions in KIT mRNA. As shown in Fig. 6A , GIST882 cells were treated with either flavopiridol or imatinib mesylate for 4, 6, 12, and 24 hours, whereas KIT mRNA levels were determined by qPCR. Flavopiridol at 300 nmol/L induced significant reductions in KIT mRNA relative to the untreated cells with a 68% and 81% down-regulation at 12 and 24 hours, respectively (P < 0.01). The initial decrease in KIT mRNA was observed at 12 hours, whereas reductions in KIT protein were not observed until 24 hours ( Fig. 3B). This time lag can be explained by the prolonged half-life of KIT mRNA, which was shown to be ∼10 to 12 hours, especially in cells with high levels of KIT protein ( 33).

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

KIT transcription is reduced by flavopiridol-mediated inhibition of RNA pol II phosphorylation and down-regulation of KIT promoter activity. A, qPCR analysis of GIST882 cells treated for various time intervals with either flavopiridol (150 and 300 nmol/L) or imatinib mesylate (1.0 μmol/L). Columns, mean of relative KIT expression determined by normalizing the level of KIT mRNA to the 18s rRNA gene; bars, ±SD; *, P < 0.01 versus untreated and imatinib mesylate groups at 12 and 24 hours. Means were obtained from three independently performed experiments done in triplicate. B, GIST882 cells were treated with 300 nmol/L flavopiridol for varying time intervals. Whole cell lysates were subjected to immunoblotting probing for RNA pol II phosphorylated at Ser2 on the CTD (CTD Ser2) as well as total RNA pol II and tubulin. C, KIT promoter activity in GIST 882cells. The KIT promoter was cloned upstream of a luciferase reporter gene in a pGL2-basic vector (pGL2-KIT) as described in Materials and Methods. After transient transfection in HCT116 cells, treatment with flavopiridol or imatinib mesylate was initiated for 24 hours and lysates were harvested for luciferase activity. Columns, mean of the relative luciferase activity in arbitrary units of light for each condition, by normalizing pGL2-KIT activity to Renilla luciferase (pRL-TK) activity; bars, ±SD; **, P < 0.05 versus untreated. D, GIST 882 cells were treated for increasing time intervals with 50 μmol/L DRB. KIT activation, total KIT, bcl-2, and mcl-1 were analyzed via Western blotting. E, Annexin V/7-AAD labeling of GIST882 cells after 24 hours of exposure to DRB (50 μmol/L). The percentage of cells is noted in the respective gates as in the previous experiments. These assays were repeated thrice with similar results.

We then showed that 300 nmol/L of flavopiridol was capable of inhibiting phosphorylation of the CTD at Ser2 on RNA pol II. This site is preferentially phosphorylated by the CDK9/cyclin T complex (P-TEFb) and is required for effective elongation control of transcription ( 34). Western blotting in Fig. 6B shows a significant reduction in the level of phospho-CTD (Ser2) RNA pol II as early as 4 hours after exposure to 300 nmol/L of flavopiridol, and by 24 hours, the protein is undetectable. In contrast, 1.0 μmol/L of imatinib mesylate and 150 nmol/L of flavopiridol did not have an appreciable effect on phospho-CTD RNA pol II compared with the untreated control (data not shown). Total RNA pol II did not significantly change and remains constant across all sampling time points. This data indicates that flavopiridol-mediated inhibition of transcription happens early and is dependent on direct inhibition of RNA pol II activation rather than loss of RNA pol II protein.

We further examined this transcriptional effect of flavopiridol on KIT by assessing promoter activity. We used HCT116 colon cancer cells because of an inability to adequately transfect the reporter vector into the nucleus of the GIST cell line despite optimization of the transfection conditions. Treatment of vector-transfected cells for 24 hours produced significant reductions in KIT promoter activity with 300 nmol/L of flavopiridol ( Fig. 6C). There was an 11.5-fold decrease in luciferase activity at 300 nmol/L compared with untreated cells (P < 0.02) after normalization to the Renilla vector. The promoter effect also seemed to be dependent on a threshold concentration of 300 nmol/L because half the dose resulted in only a modest decrease in promoter activity, 1.5-fold lower than the untreated cells (P = 0.10). Imatinib mesylate, which binds directly to the KIT receptor, did not have an appreciable effect on the promoter activity in these cells. These results support the observation that the suppression of KIT expression by flavopiridol occurs at the transcriptional level, likely through P-TEFb.

Finally, considering that this is the first time flavopiridol has been shown to transcriptionally suppress KIT, we wanted to confirm our findings with another established inhibitor of P-TEFb. The compound DRB is a nucleoside analogue that also potently inhibits P-TEFb, thus, inhibiting CTD phosphorylation ( 35). As shown in Fig. 6D, following exposure of GIST cells to 50 μmol/L of DRB for up to 24 hours, both KIT autophosphorylation sites at Y703 and Y721 were almost completely suppressed. Total KIT protein expression was significantly down-regulated to a similar level as that observed with flavopiridol after 24 hours. In addition, examination of mcl-1 and bcl-2 showed an early reduction by at least 4 hours that preceded KIT suppression. Annexin V labeling ( Fig. 6E) revealed that DRB exposure for 24 hours was required for significant apoptosis induction compared with control (30 ± 1.6% versus 8 ± 0.72%; P < 0.01). Again, earlier sampling did not show measurable apoptosis (data not shown). Thus, with DRB, in transcriptionally down-regulated KIT, we observed an apoptotic response in these cells that approximates the effect mediated by flavopiridol.

Discussion

In the present study, we have shown for the first time that flavopiridol inhibits receptor autophosphorylation by depleting KIT through transcriptional suppression. In doing so, we have taken an alternative approach to blocking KIT dependency. The loss of critical levels of KIT protein, especially at the cell surface, produced an absolute decrease in phosphorylated KIT. This, in turn, inhibited the downstream events that would normally propagate unchecked proliferation signals in GIST cells that have constitutively active KIT. Although flavopiridol initially down-regulated the antiapoptotic proteins bcl-2, mcl-1, and XIAP, demonstrable PARP cleavage and apoptosis was only induced when KIT protein levels were significantly reduced. Exposure of GIST882 cells to siRNA directed at KIT further indicates that suppression of KIT alone results in apoptosis. Whereas KIT siRNA resulted in ∼30% apoptosis, 48 hours of exposure to flavopiridol resulted in almost 60% apoptosis. This increased induction is likely related to the shift in the antiapoptotic milieu to include mcl-1, bcl-2, and XIAP, but still requires KIT suppression. Other factors may also contribute to lethality, including the inhibition of other CDK's, as well as nuclear factor κB inhibition ( 36). The role these other factors play remains the subject of future investigations.

The concept of using transcriptional suppression to block receptor tyrosine kinase signaling has been explored in other studies. Nahta and colleagues showed that the transcriptional suppression of epidermal growth factor receptor by flavopiridol, independent of its effect on the cell cycle, caused apoptotic synergy with trastuzumab in breast cancer cells ( 37). They confirmed their findings by substituting flavopiridol with DRB, a known transcriptional suppressor and P-TEFb inhibitor. We similarly treated our cells with DRB and recapitulated the same time-dependent reduction in KIT autophosphorylation and overall KIT expression observed with flavopiridol, whereas inducing an apoptotic response. Our data, showing inhibition of RNA pol II phosphorylation by flavopiridol, coupled with the results after DRB exposure, indicate for the first time that KIT expression is regulated, at least in part, by P-TEFb. As P-TEFb has the capacity to be recruited to gene promoters to enhance transcription, the observed reduction in luciferase activity with flavopiridol exposure also implicates P-TEFb in c-KIT promoter control ( 38, 39).

An interesting finding of this study was that imatinib mesylate at 1.0 μmol/L, despite completely inhibiting KIT activation at both Y703 and Y721, produced minimal apoptosis over a 48-hour period. The antiproliferative effects of imatinib mesylate were mediated more by a G1 cell cycle arrest (data not shown). We are not the first to show that imatinib mesylate fails to induce apoptosis despite blocking the KIT receptor. In neuroblastoma cells, Beppu et al. has shown that imatinib mesylate concentrations of 0.5 to 1.0 μmol/L were capable of inhibiting KIT autophosphorylation but could not induce apoptosis after 48 hours ( 40). Other investigators have shown that imatinib mesylate can induce apoptosis in GIST cells, but only with protracted drug exposure of 4 to 7 days, despite showing a reduction in KIT phosphorylation as early as 90 minutes after imatinib mesylate treatment ( 10). In addition, clinically, a paucity of apoptosis has also been observed with imatinib mesylate therapy, and to date, there have been only rare complete responses in GIST patients. Collectively, this data would indicate that GIST882 cells are not completely dependent on KIT signaling for cell survival. Although we show that targeted disruption of KIT with siRNA results in cell death, the enhanced induction of apoptosis by flavopiridol indicates a co-dependency on antiapoptotic proteins including XIAP, mcl-1, and bcl-2. In addition, by transcriptionally silencing KIT expression with either siRNA or flavopiridol, we seem to induce a more complete interruption of this signaling cascade, leading to cell death that is not achievable with direct receptor inhibition by imatinib mesylate alone.

In evaluating the dose dependency of flavopiridol activity, 150 nmol/L was relatively ineffective in the GIST882 cells, having minimal antiproliferative and apoptotic effects, and failing to induce significant reductions in KIT mRNA or protein. Other cancer cell lines have been shown to be inhibited at these lower concentrations of flavopiridol, mostly through CDK modulation. The threshold dependency of 300 nmol/L in these cells, however, confirms the likelihood that transcriptional suppression of KIT is required for activity. Lam et al. did a microarray analysis on flavopiridol-treated lymphoma cells and found that gene expression profiles were unaffected by concentrations <300 nmol/L ( 41). In relating the gene expression changes to drug activity, the authors similarly found no viability changes in their cell lines at concentrations <250 nmol/L. This suggests that higher concentrations of flavopiridol are required to produce the transcriptional suppression that we observed.

The emergence of imatinib mesylate resistance in both GIST as well as CML patient populations has prompted the search for other molecularly targeted therapeutics. SU11248, a tyrosine kinase inhibitor with KIT-inhibitory properties has had some success in patients with imatinib mesylate–refractory GIST, and is currently undergoing clinical trials ( 42). There are other tyrosine kinase inhibitors that have also been evaluated preclinically and shown to inhibit KIT kinase activity including PKC412 ( 43) and PP1 ( 44); however, the direct receptor binding properties of many of these tyrosine kinase inhibitors, including imatinib mesylate, likely contributes to the mutational capacity of the gene. Structural studies have proven that imatinib mesylate binds in the juxtamembrane region and other mutations in the gene, specifically in the kinase region, render imatinib mesylate ineffective ( 30). Other human malignancies such as human mastocytosis are characterized by KIT kinase domain mutations (codon 816) that are unequivocally resistant to inhibition by imatinib mesylate ( 45). The capacity of flavopiridol to transcriptionally down-regulate the KIT gene and induce apoptosis suggests that a mutation, unless located in the promoter region, would not be a factor in the activity of this drug.

Clinically, peak flavopiridol levels of 3.0 μmol/L have been reported ( 46). With 90% protein binding, this would indicate that peak plasma levels of 300 nmol/L could be achieved. With 3.0 μmol/L of flavopiridol, in the presence of human plasma proteins, we show that flavopiridol can still induce significant degrees of apoptosis in the GIST cells. In addition, recent pharmacologic models have indicated that these levels can be maintained for several hours if the drug is first administered as an i.v. bolus followed by a 4-hour infusion. This approach has been clinically applied to the treatment of chemotherapy-refractory CLL resulting in dramatic clinical responses in the setting of tumor lysis syndrome ( 47). Longer durations of exposure beyond 4 hours are under clinical investigation. Thus, flavopiridol could represent a new direction for the treatment of imatinib mesylate–refractory GIST. Rather than targeting mutated sites on the KIT receptor, flavopiridol can induce a selective suppression of KIT expression by inhibiting transcription at the KIT promoter through P-TEFb. This, in essence, would encompass all forms of imatinib mesylate resistance. Although imatinib mesylate should continue to be used as a first line agent in the treatment of GIST, our improved understanding of flavopiridol pharmacology and transcriptional suppression has solidified a new niche for this drug. We envision flavopiridol as a strategic alternative to the current receptor-targeted approaches in the treatment of imatinib mesylate–refractory GIST tumors and perhaps other KIT-dependent malignancies.

Acknowledgments

Grant support: The Soft Tissue Sarcoma Program Project PO1 CA047179. Support for this project was also provided in part by the Kristin Ann Carr Fund. E.B. Sambol was supported by NIH T32 training grant no. CA09501.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

We thank the laboratory of Dr. Bhuvanesh Singh for technical support, Maureen Thyne and Dr. Jonanthan Fletcher for assistance with the GIST cell line, Dr. Annette Duensing for helpful discussion, and Dr. Murray Brennan for his guidance and support of the MSKCC surgical research fellowship.

Footnotes

  • Note: This data was presented as a poster at the 96th Annual Meeting of the AACR, April 2005.

  • Received August 17, 2005.
  • Revision received March 15, 2006.
  • Accepted March 23, 2006.
  • ©2006 American Association for Cancer Research.

References

  1. ↵
    Ashman LK. The biology of stem cell factor and its receptor C-kit. Int J Biochem Cell Biol 1999; 31: 1037–51.
    OpenUrlCrossRefPubMed
  2. ↵
    Tian Q, Frierson HF, Jr., Krystal GW, Moskaluk CA. Activating c-kit gene mutations in human germ cell tumors. Am J Pathol 1999; 154: 1643–7.
    OpenUrlCrossRefPubMed
  3. ↵
    Furitsu T, Tsujimura T, Tono T, et al. Identification of mutations in the coding sequence of the proto-oncogene c-kit in a human mast cell leukemia cell line causing ligand-independent activation of c-kit product. J Clin Invest 1993; 92: 1736–44.
    OpenUrlCrossRefPubMed
  4. ↵
    Krystal GW, Hines SJ, Organ CP. Autocrine growth of small cell lung cancer mediated by coexpression of c-kit and stem cell factor. Cancer Res 1996; 56: 370–6.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    Zhang W, Stoica G, Tasca SI, Kelly KA, Meininger CJ. Modulation of tumor angiogenesis by stem cell factor. Cancer Res 2000; 60: 6757–62.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Hirota S, Isozaki K, Moriyama Y, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science 1998; 279: 577–80.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Heinrich MC, Griffith DJ, Druker BJ, Wait CL, Ott KA, Zigler AJ. Inhibition of c-kit receptor tyrosine kinase activity by STI 571, a selective tyrosine kinase inhibitor. Blood 2000; 96: 925–32.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat Med 1996; 2: 561–6.
    OpenUrlCrossRefPubMed
  9. ↵
    Buchdunger E, Cioffi CL, Law N, et al. Abl protein-tyrosine kinase inhibitor STI571 inhibits in vitro signal transduction mediated by c-kit and platelet-derived growth factor receptors. J Pharmacol Exp Ther 2000; 295: 139–45.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Tuveson DA, Willis NA, Jacks T, et al. STI571 inactivation of the gastrointestinal stromal tumor c-KIT oncoprotein: biological and clinical implications. Oncogene 2001; 20: 5054–8.
    OpenUrlCrossRefPubMed
  11. ↵
    Duensing A, Medeiros F, McConarty B, et al. Mechanisms of oncogenic KIT signal transduction in primary gastrointestinal stromal tumors (GISTs). Oncogene 2004; 23: 3999–4006.
    OpenUrlCrossRefPubMed
  12. ↵
    Demetri GD, von Mehren M, Blanke CD, et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med 2002; 347: 472–80.
    OpenUrlCrossRefPubMed
  13. ↵
    Verweij J, Casali PG, Zalcberg J, et al. Progression-free survival in gastrointestinal stromal tumours with high-dose imatinib: randomised trial. Lancet 2004; 364: 1127–34.
    OpenUrlCrossRefPubMed
  14. ↵
    Corless CL, Fletcher JA, Heinrich MC. Biology of gastrointestinal stromal tumors. J Clin Oncol 2004; 22: 3813–25.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Shapiro GI. Preclinical and clinical development of the cyclin-dependent kinase inhibitor flavopiridol. Clin Cancer Res 2004; 10: 4270–5s.
    OpenUrlCrossRef
  16. ↵
    De Azevedo WF, Jr., Mueller-Dieckmann HJ, Schulze-Gahmen U, Worland PJ, Sausville E, Kim SH. Structural basis for specificity and potency of a flavonoid inhibitor of human CDK2, a cell cycle kinase. Proc Natl Acad Sci U S A 1996; 93: 2735–40.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Dai Y, Rahmani M, Pei XY, Dent P, Grant S. Bortezomib and flavopiridol interact synergistically to induce apoptosis in chronic myeloid leukemia cells resistant to imatinib mesylate through both Bcr/Abl-dependent and -independent mechanisms. Blood 2004; 104: 509–18.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Wall NR, O'Connor DS, Plescia J, Pommier Y, Altieri DC. Suppression of survivin phosphorylation on Thr34 by flavopiridol enhances tumor cell apoptosis. Cancer Res 2003; 63: 230–5.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Chao SH, Price DH. Flavopiridol inactivates P-TEFb and blocks most RNA polymerase II transcription in vivo. J Biol Chem 2001; 276: 31793–9.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Lu X, Burgan WE, Cerra MA, et al. Transcriptional signature of flavopiridol-induced tumor cell death. Mol Cancer Ther 2004; 3: 861–72.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Sedlacek HH. Mechanisms of action of flavopiridol. Crit Rev Oncol Hematol 2001; 38: 139–70.
    OpenUrlCrossRefPubMed
  22. ↵
    Huang R, Lin Y, Wang CC, et al. Connexin 43 suppresses human glioblastoma cell growth by down-regulation of monocyte chemotactic protein 1, as discovered using protein array technology. Cancer Res 2002; 62: 2806–12.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Yamamoto K, Tojo A, Aoki N, Shibuya M. Characterization of the promoter region of the human c-kit proto-oncogene. Jpn J Cancer Res 1993; 84: 1136–44.
    OpenUrlCrossRef
  24. ↵
    Gambacorti-Passerini C, Zucchetti M, Russo D, et al. α1 acid glycoprotein binds to imatinib (STI571) and substantially alters its pharmacokinetics in chronic myeloid leukemia patients. Clin Cancer Res 2003; 9: 625–32.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Byrd JC, Shinn C, Waselenko JK, et al. Flavopiridol induces apoptosis in chronic lymphocytic leukemia cells via activation of caspase-3 without evidence of bcl-2 modulation or dependence on functional p53. Blood 1998; 92: 3804–16.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Rudek MA, Bauer KS, Jr., Lush RM III, et al. Clinical pharmacology of flavopiridol following a 72-hour continuous infusion. Ann Pharmacother 2003; 37: 1369–74.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Kitada S, Zapata JM, Andreeff M, Reed JC. Protein kinase inhibitors flavopiridol and 7-hydroxy-staurosporine down-regulate antiapoptosis proteins in B-cell chronic lymphocytic leukemia. Blood 2000; 96: 393–7.
    OpenUrl
  28. ↵
    Yarden Y, Kuang WJ, Yang-Feng T, et al. Human proto-oncogene c-kit: a new cell surface receptor tyrosine kinase for an unidentified ligand. EMBO J 1987; 6: 3341–51.
    OpenUrlPubMed
  29. ↵
    Thommes K, Lennartsson J, Carlberg M, Ronnstrand L. Identification of Tyr-703 and Tyr-936 as the primary association sites for Grb2 and Grb7 in the c-Kit/stem cell factor receptor. Biochem J 1999; 341: 211–6.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Mol CD, Dougan DR, Schneider TR, et al. Structural basis for the autoinhibition and STI-571 inhibition of c-Kit tyrosine kinase. J Biol Chem 2004; 279: 31655–63.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Kapur R, Chandra S, Cooper R, McCarthy J, Williams DA. Role of p38 and ERK MAP kinase in proliferation of erythroid progenitors in response to stimulation by soluble and membrane isoforms of stem cell factor. Blood 2002; 100: 1287–93.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Casteran N, De Sepulveda P, Beslu N, et al. Signal transduction by several KIT juxtamembrane domain mutations. Oncogene 2003; 22: 4710–22.
    OpenUrlCrossRefPubMed
  33. ↵
    Ogawa K, Takeda Y, Tashima M, et al. High expression of c-kit in K562YO cells due to the prolonged half-life of its mRNA: the effects of modification with serine/threonine kinase signals. Blood 1995; 85: 1496–503.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Sims RJ III, Belotserkovskaya R, Reinberg D. Elongation by RNA polymerase II: the short and long of it. Genes Dev 2004; 18: 2437–68.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    Marshall NF, Peng J, Xie Z, Price DH. Control of RNA polymerase II elongation potential by a novel carboxyl-terminal domain kinase. J Biol Chem 1996; 271: 27176–83.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Takada Y, Aggarwal BB. Flavopiridol inhibits NF-κB activation induced by various carcinogens and inflammatory agents through inhibition of IκBα kinase and p65 phosphorylation: abrogation of cyclin D1, cyclooxygenase-2, and matrix metalloprotease-9. J Biol Chem 2004; 279: 4750–9.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Nahta R, Trent S, Yang C, Schmidt EV. Epidermal growth factor receptor expression is a candidate target of the synergistic combination of trastuzumab and flavopiridol in breast cancer. Cancer Res 2003; 63: 3626–31.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Barboric M, Nissen RM, Kanazawa S, Jabrane-Ferrat N, Peterlin BM. NF-[κ]B binds P-TEFb to stimulate transcriptional elongation by RNA polymerase II. Mol Cell 2001; 8: 327–37.
    OpenUrlCrossRefPubMed
  39. ↵
    Lis JT, Mason P, Peng J, Price DH, Werner J. P-TEFb kinase recruitment and function at heat shock loci. Genes Dev 2000; 14: 792–803.
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Beppu K, Jaboine J, Merchant MS, Mackall CL, Thiele CJ. Effect of imatinib mesylate on neuroblastoma tumorigenesis and vascular endothelial growth factor expression. J Natl Cancer Inst 2004; 96: 46–55.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Lam LT, Pickeral OK, Peng AC, et al. Genomic-scale measurement of mRNA turnover and the mechanisms of action of the anti-cancer drug flavopiridol. Genome Biol.com 2001; 2: Research0041. Epub 2001 Sept 13.
    OpenUrl
  42. ↵
    Maki RG, Fletcher JA, Heinrich MC, et al. Results from a continuation trial of SU11248 in patients with imatinib (IM)-resistant gastrointestinal stromal tumors (GIST) [abstract #9011]. Proc Am Soc Clin Oncol 2005; 23: 818S.
    OpenUrl
  43. ↵
    Growney JD, Clark JJ, Adelsperger J, et al. Activation mutations of human c-KIT resistant to imatinib are sensitive to the tyrosine kinase inhibitor PKC412. Blood 2005; 106: 721–4.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    Tatton L, Morley GM, Chopra R, Khwaja A. The Src-selective kinase inhibitor PP1 also inhibits Kit and Bcr-Abl tyrosine kinases. J Biol Chem 2003; 278: 4847–53.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    Ma Y, Zeng S, Metcalfe DD, et al. The c-KIT mutation causing human mastocytosis is resistant to STI571 and other KIT kinase inhibitors; kinases with enzymatic site mutations show different inhibitor sensitivity profiles than wild-type kinases and those with regulatory-type mutations. Blood 2002; 99: 1741–4.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Shah MA, Kortmansky J, Motwani M, et al. A phase I clinical trial of the sequential combination of irinotecan followed by flavopiridol. Clin Cancer Res 2005; 11: 3836–45.
    OpenUrlAbstract/FREE Full Text
  47. ↵
    Byrd JC, Peterson BL, Gabrilove J, et al. Treatment of relapsed chronic lymphocytic leukemia by 72-hour continuous infusion or 1-hour bolus infusion of flavopiridol: results from Cancer and Leukemia Group B study 19805. Clin Cancer Res 2005; 11: 4176–81.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top
Cancer Research: 66 (11)
June 2006
Volume 66, Issue 11
  • Table of Contents
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Flavopiridol Targets c-KIT Transcription and Induces Apoptosis in Gastrointestinal Stromal Tumor Cells
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Flavopiridol Targets c-KIT Transcription and Induces Apoptosis in Gastrointestinal Stromal Tumor Cells
Elliot B. Sambol, Grazia Ambrosini, Rula C. Geha, Peter T. Kennealey, Penelope DeCarolis, Rachael O'Connor, Yuhsin V. Wu, Monica Motwani, Jin-Hong Chen, Gary K. Schwartz and Samuel Singer
Cancer Res June 1 2006 (66) (11) 5858-5866; DOI: 10.1158/0008-5472.CAN-05-2933

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Flavopiridol Targets c-KIT Transcription and Induces Apoptosis in Gastrointestinal Stromal Tumor Cells
Elliot B. Sambol, Grazia Ambrosini, Rula C. Geha, Peter T. Kennealey, Penelope DeCarolis, Rachael O'Connor, Yuhsin V. Wu, Monica Motwani, Jin-Hong Chen, Gary K. Schwartz and Samuel Singer
Cancer Res June 1 2006 (66) (11) 5858-5866; DOI: 10.1158/0008-5472.CAN-05-2933
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • MBP-1 Inhibits Breast Cancer Metastasis
  • Decitabine Sensitivity in Testicular Cancer
  • Drug-Induced Regulation of FA/BRCA Gene Expression
Show more Experimental Therapeutics, Molecular Targets, and Chemical Biology
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement