Although it is well established that cyclin-dependent kinases phosphorylate and inactivate Rb, the Raf-1 kinase physically interacts with Rb and initiates the phosphorylation cascade early in the cell cycle. We have identified an orally active small molecule, Rb/Raf-1 disruptor 251 (RRD-251), that potently and selectively disrupts the Rb/Raf-1 but not Rb/E2F, Rb/prohibitin, Rb/cyclin E, and Rb/HDAC binding. The selective inhibition of Rb/Raf-1 binding suppressed the ability of Rb to recruit Raf-1 to proliferative promoters and inhibited E2F1-dependent transcriptional activity. RRD-251 inhibited anchorage-dependent and anchorage-independent growth of human cancer cells and knockdown of Rb with short hairpin RNA or forced expression of E2F1 rescued cells from RRD-251–mediated growth arrest. P.o. treatment of mice resulted in significant tumor growth suppression only in tumors with functional Rb, and this was accompanied by inhibition of angiogenesis, inhibition of proliferation, decreased phosphorylated Rb levels, and inhibition of Rb/Raf-1 but not Rb/E2F1 binding in vivo. Thus, selective targeting of Rb/Raf-1 interaction seems to be a promising approach for developing novel chemotherapeutic agents. [Cancer Res 2008;68(10):3810–8]

The retinoblastoma tumor suppressor protein, Rb, is a vital regulator of the mammalian cell cycle, and its inactivation facilitates S-phase entry (1, 2). Rb is inactivated through multiple waves of phosphorylation during cell cycle progression, mediated by kinases associated with D and E type cyclins in the G1 phase (3, 4). Rb is inactivated in most cancers, either by mutation or deletion of the gene, interaction with viral oncoproteins, or alterations in the levels and activity of upstream regulators of Rb function (58). Rb controls the G1-S boundary by repressing the transcriptional activity of the E2F family of transcription factors, especially E2F1, E2F2, and E2F3 (9). Many genes necessary for DNA synthesis and cell cycle progression, such as cyclins A and E, cdc2, thymidylate synthase (TS), dihydrofolate reductase, ORC1, and DNA polymerase α require E2F for their expression (1013). Whereas cyclins and cyclin-dependent kinases phosphorylate Rb in mid to late G1 phase releasing transcriptionally active E2F (1416), Raf-1 kinase binds and phosphorylates Rb early in the G1 phase (17). Disruption of this Rb/Raf-1 interaction by an eight–amino acid peptide (corresponding to Raf-1 residues 10–18) prevented Rb phosphorylation even late in the G1 phase, suggesting that the binding of Raf-1 is necessary for the eventual inactivation of Rb (18). Furthermore, the level of Rb/Raf-1 interaction was elevated in non–small cell lung cancer tissue compared with adjacent normal tissue (19), suggesting that this interaction might contribute to oncogenesis. These observations suggested that disruption of the Rb/Raf-1 interaction might have anticancer effects and raised the possibility that small molecules that can disrupt the Rb/Raf-1 interaction might be useful as anticancer drugs. Here, we report a potent and selective small molecule disruptor of Rb/Raf-1 interaction that significantly inhibits angiogenesis and tumor growth in vivo in an Rb-dependent manner.

Cell culture and transfection. The human myelomonocytic leukemia cell line U937 was cultured in RPMI (Mediatech) containing 10% fetal bovine serum (FBS; Mediatech). U2-OS, Saos-2, PANC1, CAPAN2, A375, SK-MEL-5, SK-MEL-28, and MDA-MB-231 cell lines were cultured in DMEM (Mediatech) containing 10% FBS. A549 cells and A549 short hairpin RNA (shRNA) Rb cell lines were maintained in Ham F-12K supplemented with 10% FBS; media for shRNA cells lines contained 0.5 μg/mL puromycin. shRNA cell lines were generated by stably transfecting A549 cells with two different shRNA constructs that specifically target Rb obtained from a shRNAmir library from Open Biosystems. H1650, PC-9, LNCap, and Aspc1 cell lines were cultured in RPMI (Life Technologies) containing 10% FBS. Human aortic endothelial cells (HAEC; Clonetics) were cultured in endothelial growth medium, supplemented with 5% FBS. U251MG and U87MG glioma cells were maintained in DMEM supplemented with nonessential amino acids, 50 mmol/L β-mercaptoethanol, and 10% FBS. The adenovirus constructs Ad-green fluorescent protein (GFP) and Ad-E2F1 were obtained from W.D. Cress. Ad-cyclin D was kindly provided by I. Cozar-Castellano.

In vitro library screening assays. ELISA 96-well plates (Nunc) were coated with 1 μg/mL of glutathione S-transferase (GST) Raf-1 (1–149 amino acids) overnight at 4°C. Subsequently, the plates were blocked and GST-Rb at 20 μg/mL was rotated at room temperature for 30 min in the presence or absence of the compounds at 20 μmol/L. GST-Rb with or without compounds was then added to the plate and incubated for 90 min at 37°C. The amount of Rb bound to Raf-1 was detected by Rb polyclonal antibody (Santa Cruz) 1:1,000 incubated for 60 min at 37°C. Donkey anti-rabbit-IgG-HRP (1:10,000) was added to the plate and incubated at 37°C for 60 min. The color was developed with orthophenylenediamine peroxidase substrate tablets (Sigma); the reaction was terminated with 3 mol/L H2SO4 and absorbance read at 490 nm. To determine disruption of Rb to E2F1, Phb, or HDAC1, the above protocol was used with the exception of coating GST-Rb on the ELISA plate and adding the drugs in the presence or absence of GST E2F1, Phb, or HDAC1. These interactions were detected with E2F1 monoclonal antibody (Santa Cruz; 1:2,000 dilution), prohibitin monoclonal antibody (NeoMarkers; 1:1,000 dilution), and HDAC1 polyclonal antibody (Santa Cruz; 1:1,000). For disruption of Mek-Raf-1 binding ELISAs, Raf-1 (1 μg/mL) was coated on the plate and GST-Mek (20 μg/mL) was incubated in the presence or absence of compounds for 30 min at room temperature. Mek1 polyclonal antibody (Cell Signaling) was used at 1:1,000 to detect the binding of Raf-1 to Mek1. The IC50 concentrations for the Rb/Raf-1 inhibitors were determined by plotting with Origin 7.5 software.

Lysate preparation, immunoprecipitation, and Western blotting. Cell lysates were prepared by NP40 lysis, as described earlier (18). Tumor lysates were prepared with T-Per tissue lysis buffer (Pierce) and a Fisher PowerGen 125 dounce homogenizer. Physical interaction between proteins in vivo was analyzed by immunoprecipitation–Western blot analyses with 200 μg of lysate and 1 μg of the indicated antibody (18). Polyclonal E2F1, B-Raf, ASK1, and cyclins D and E were obtained from Santa Cruz Biotechnology. Monoclonal Rb and Raf-1 were obtained from BD Transduction Laboratories, and polyclonal antibodies to phosphorylated Rb (807, 811) and Mek1/2 were from Cell Signaling.

Chloramphenicol acetyltransferase assays. A549 cells were transfected by calcium phosphate and treated with drug for 24 h. Assays for chloramphenicol acetyltransferase (CAT) and β-galactosidase were performed using standard protocols (17).

Chromatin immunoprecipitation assay. A549 cells were serum-starved and restimulated with serum for 2 or 16 h in the presence or absence of Rb/Raf-1 disruptor 251 (RRD-251) at 20 μmol/L, and chromatin immunoprecipitation (ChIP) lysates were prepared (18). Immunoprecipitations were conducted using antibodies against E2F1, Rb, Raf-1, Brg1, HP1, and HDAC1, and the association with specific promoters was detected by PCR. Rabbit anti-mouse secondary antibody was used as the control for all reactions. The sequences of the PCR primers used in the PCRs were as follows: Cdc6 promoter forward primer, 5′-GGCCTCACAGCGACTCTAAGA-3′ and Cdc6 promoter reverse primer, 5′-CTCGGACTCACCACAAGC-3′; TS promoter forward primer, 5′-TGG CGC ACG CTC TCT AGA GC-3′ and TS promoter reverse primer, 5′-GAC GGA GGC AGG CCA AGT G-3′. The cdc25A and c-fos primers are described in ref. 18.

Real-time PCR. A549 cells were rendered quiescent by serum starvation and subsequently stimulated with serum in the presence or absence of RRD-251. Unstimulated serum-starved cells were used as control. Total RNA was isolated by an RNeasy miniprep kit from QIAGEN following the manufacturer's protocol. One microgram of RNA was DNase treated using RQ1 DNase (Promega), followed by first-strand cDNA synthesis using the iScript cDNA synthesis kit (Bio-Rad). A fraction (1/20) of the final cDNA reaction volume was used in each PCR (20). Primer sequences are as follows: 5′-CTG CCA GCT GTA CCA GAG AT-3′ (TS forward primer), 5′-ATG TGC ATC TCC CAA AGT GT-3′ (TS reverse primer), 5′-CCC CAT GAT TGT GTT GGT AT-3′ (Cdc6 forward primer), 5′-TTC AAC AGC TGT GGC TTA CA-3′ (Cdc6 reverse primer), 5′-CTC AAC ACG GGA AAC CTC AC-3′ (18S forward primer), and 5′-AAA TCG CTC CAC CAA CTA AGA A-3′ (18S reverse primer). Real-time PCR was performed on a Bio-Rad iCycler.

In vitro kinase assay. The kinase reaction for Raf-1 was carried out with 100 ng of Raf-1 (Upstate Biotechnologies), 0.5 μg of MEK1 (Upstate) as the substrate, 10 μmol/L ATP, 10 μCi of [γ-32P]ATP in the kinase assay buffer in the presence or absence of the drugs at 30°C for 30 min. BAY-43-9006 (1 μmol/L) was used as a control, and RRD-251 (20 μmol/L) was used. Cyclins D and E kinase assays are described in ref. 18.

Proliferation assays. Bromodeoxyuridine (BrdUrd) labeling kits were obtained from Roche Biochemicals. Cells were plated in poly-d-lysine–coated chamber slides at a density of 10,000 cells per well and serum starved for 24 h. Cells were then stimulated with serum in the presence or absence of the indicated drugs for 18 h. S-phase cells were visualized by microscopy and quantitated by counting three fields of 100 in quadruplicate. For adenovirus experiments, A549 cells were serum starved for 48 h and subsequently infected with adenovirus in the presence or absence of RRD-251 for 36 h. Ad-GFP [6 × 106 plaque-forming units (pfu)/μL], Ad-E2F1 (6 × 106 pfu/μL), and Ad-cyclin D (7 × 108 pfu/μL) were added at a multiplicity of infection of 150 particles per cell.

Soft agar colony formation assay. Assays were done in triplicate in 12-well plates (Corning). After allowing the bottom layer of agar (0.6%) to solidify at room temperature, the top layer of agar (0.3%) was mixed with 5,000 cells per well, and the indicated drug was added. Drugs were added twice weekly in complete media to the agar wells. Colonies were quantified by staining with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (1 mg/mL) for 1 h at 37°C.

Matrigel assays. Matrigel (Collaborative Biomedical Products) was used to promote the differentiation of human umbilical vascular endothelial cells (HUVEC) into capillary tube–like structures (18). A total of 100 μL of thawed Matrigel was added to 96-well tissue culture plates, followed by incubation at 37°C for 60 min to allow polymerization. Subsequently, 1 × 104 HUVECs were seeded on the gels in EGM medium supplemented with 5% FBS in the presence or absence of 20 μmol/L concentrations of the indicated compounds, followed by incubation for 24 h at 37°C. Capillary tube formation was assessed using a Leica DMIL phase contrast microscope.

Ex vivo rat aorta ring angiogenesis assays. Forty-eight well tissue culture plates were coated with 200 μL of Matrigel and allowed to polymerize for 1 h at 37°C. Thoracic aorta was excised from 8-wk-old to 10-wk-old male Sprague-Dawley rats (250–300 g; ref. 21). After removing fibroadipose tissue, aortas were rinsed several times with EGM-2 (Clonetics), sectioned into 1-mm rings, and placed on the Matrigel-coated wells. The rings were covered with an additional 200 μL of Matrigel and allowed to polymerize. The rings were cultured in EGM-2 media in the presence or absence of 20 μmol/L of RRD-251. The media and drug were supplemented twice a week for 1 wk. The aortic rings were photographed on day 7 using a Leica phase contrast microscope. Quantitation of microvessel growth was done using Image Pro Plus (v.6.0) software, and values are reported as microvessel area.

In vivo Matrigel plug angiogenesis assays.In vivo Matrigel plug assays were carried out as previously described (22). Cooled liquid Matrigel (300 μL) was injected s.c. into both flanks of nude mice. One group of mice received the vehicle, and the second group received RRD-251 50 MPK daily by i.p. injection. At 7 d post-Matrigel injection, the mice were injected with 100 μL of 100 MPK FITC-Dextran (Sigma) through the tail vein. At 30 min later, the mice were euthanized and the Matrigel plugs were removed and fixed in buffered formalin. Samples were viewed with a Leica DMI6000 inverted microscope, TCS SP5 confocal scanner, and a 20×/0.7NA Plan Apochromat objective (Leica Microsystems). An Argon 488 laser line was applied to excite the samples, and tunable filters were used to minimize background fluorescence. Image sections at 2.0 μm were captured with photomultiplier detectors three-dimensional projections prepared with the LAS AF software version 1.6.0 build 1016 (Leica Microsystems). Quantification of intensity and angiogenesis was performed using Image Pro Plus 6.2 (Media Cybernetics, Inc.). Average intensity per pixel is plotted as percentage angiogenesis in each image (n = 12). Each image is representative of areas of vessel formation throughout entire Matrigel plug. After confocal imaging, samples were paraffin blocked and stained with H&E. H&E images display 1/4 of the Matrigel plug.

Animal studies. A549 or H1650 cells were harvested and resuspended in PBS and implanted s.c. into the right and left flanks (10 × 106 cells per flank) of 8-wk-old female athymic nude mice (Charles River) as described (18, 23). When tumors reached ∼100 to 200 mm3, animals were dosed i.p. or p.o. by gavage with 0.1 mL of the drug or the vehicle daily. Tumor volumes were determined by measuring the length (l) and the width (w) and calculating the volume (V = lw2 / 2). Statistical significance between control and treated animals were evaluated using Student's t test.

Immunohistochemistry staining. Tumors were fixed in 10% neutral-buffered formalin before processing into paraffin blocks. Paraffin sections (5-μm thick) were rehydrated into PBS and processed using the following protocols. Sections were rinsed in dH2O and then subjected to microwave “antigen retrieval” in 0.01 mol/L sodium citrate (pH 6.0). Sections were cooled, rinsed thrice in dH2O and twice in PBS, and incubated in 5% normal goat serum. Sections were incubated in primary antibody in 5% normal goat serum and rinsed thrice in PBS. For color development, slides were treated with ABC kit from Vector Laboratories, rinsed in dH2O, and developed with 3,3′-diaminobenzidine. Then, sections were lightly counterstained in hematoxylin, dehydrated, cleared, and cover-slipped. Tissue sections were stained with H&E using standard histologic techniques. Tissue sections were immunostained using Ki-67, CD31 phosphorylated Rb, and CD31 antibodies (BD Biosciences) using the avidin-biotin peroxidase complex technique. Mouse monoclonal antibody was used at 1:50 dilution after microwave antigen retrieval (four cycles of 5 min each on high in 0.1 mol/L citrate buffer). Stained slides were scanned on an Ariol SL-50 Automatic Scanning System, and whole tumor sections were quantitated using Image Pro Plus (v.5.1.0) software.

Statistical analysis. Statistical analysis was performed using one-tailed Student's t test. Values were considered significant when the P value was <0.01.

Identification of Rb/Raf-1 disruptor RRD-251. An ELISA was used to identify compounds that could inhibit the binding of GST-Rb to GST-Raf-1. Screening of the National Cancer Institute (NCI) diversity library of 1,981 compounds identified two compounds, NSC-35400 and NSC-35950, which inhibited Rb/Raf-1 interaction 100% and 95%, respectively, at 20 μmol/L concentration. NSC-35400 and NSC-35950 each contained a benzyl-isothiourea derivative and a phenyl-based counter ion (Fig. 1A); to establish whether the benzyl-isothiourea derivative is the active component, we synthesized RRD-251 (Fig. 1A), which contains chloride as the counter ion. ELISA analysis showed that NSC-35400 disrupted the Rb/Raf-1 interaction with an IC50 of 81 ± 4 nmol/L, NSC-35950 with an IC50 of 283 ± 46 nmol/L, and RRD-251 with an IC50 of 77 ± 3.6 nmol/L (Fig. 1B), suggesting that benzylisothiouronium pharmacophore disrupts the Rb/Raf-1 interaction. ELISAs showed that these disruptors were highly selective for Rb/Raf-1 interaction over Rb/E2F1, Rb/HDAC1, Rb/prohibitin (Fig. 1C), and Raf-1/Mek (Fig. 1D) associations at a concentration of 20 μmol/L. Their selectivity in living cells was examined by immunoprecipitation–Western blot analysis. A549 cells were serum starved for 48 hours and subsequently serum stimulated for 2 hours in the presence or absence of 20 μmol/L of NSC-35400, NSC-35950, and RRD-251; Raf-1 peptide conjugated to penetratin (18) was used as a positive control, and a Raf-1 scrambled peptide was used as a negative control. It was found that the compounds inhibited the binding of Raf-1 to Rb (Fig. 2A), whereas the binding of Rb to E2F1 was not affected. To further confirm the selectivity of RRD-251, cyclin E was immunoprecipitated from lysates of quiescent cells or those serum stimulated for 8 hours in the presence or absence of RRD-251; Western blotting of the immunoprecipitates showed that RRD-251 did not inhibit the binding of Rb to cyclin E (Fig. 2B). A similar experiment was done on lysates from cells that were serum stimulated for 2 hours; RRD-251 did not inhibit the binding of B-Raf to Rb (Fig. 2C). Similarly, the binding of Raf-1 to Mek1/2 was not affected by RRD-251 (Fig. 2D). Examination of lysates from cells serum stimulated for 2 hours in the presence of RRD-251 showed a reduction in Rb phosphorylation, as seen by Western blotting (Fig. 2C,, row 5). Interestingly, in vitro kinase assays showed that RRD-251 did not affect the kinase activities associated with cyclin D (Fig. 3A,, left) or cyclin E (Fig. 3A,, middle) or Raf-1 (Fig. 3A , right). These results suggest that the reduction in Rb phosphorylation in cells treated with RRD-251 is due to a disruption of the association of Raf-1 with Rb and that Raf-1 has to physically interact with Rb to inactivate it.

Figure 1.

Identification of highly selective Rb/Raf-1 inhibitors. A, compounds identified in the NCI diversity set that showed the highest inhibition of Rb/Raf-1 by ELISA. Highest scoring compounds NSC-35400 and NSC-35950 are both benzyl isothiourea derivatives. RRD-251 was synthesized to determine activity based on isothiourea structure. B, NSC35400, NSC35950, and RRD-251 disrupt the Rb/Raf-1 interaction with high potency. IC50 values (81, 283, and 77 nmol/L, respectively) were determined using ELISA. C and D, Rb/Raf-1 inhibitors at 20 μmol/L concentration do not inhibit other binding partners to (C) Rb (E2F1, prohibitin, and HDAC1) and (D) Raf-1 (Mek).

Figure 1.

Identification of highly selective Rb/Raf-1 inhibitors. A, compounds identified in the NCI diversity set that showed the highest inhibition of Rb/Raf-1 by ELISA. Highest scoring compounds NSC-35400 and NSC-35950 are both benzyl isothiourea derivatives. RRD-251 was synthesized to determine activity based on isothiourea structure. B, NSC35400, NSC35950, and RRD-251 disrupt the Rb/Raf-1 interaction with high potency. IC50 values (81, 283, and 77 nmol/L, respectively) were determined using ELISA. C and D, Rb/Raf-1 inhibitors at 20 μmol/L concentration do not inhibit other binding partners to (C) Rb (E2F1, prohibitin, and HDAC1) and (D) Raf-1 (Mek).

Close modal
Figure 2.

RRD-251 is selective for Rb/Raf-1 interaction in vivo. A, serum-stimulated binding of Raf-1 to Rb is inhibited by Rb/Raf-1 disruptors, as well as a Raf-1 peptide conjugated to penetratin; the drugs do not inhibit the binding of E2F1 to Rb. Specificity of the disruption was assessed by immunoprecipitation–Western blots. B, RRD-251 does not inhibit Rb/cyclin E interaction in cells serum-stimulated for 8 h. C, RRD-251 does not disrupt the Rb/B-Raf binding in immunoprecipitation–Western blots. D, treatment of cells with RRD-251 for 5 min in the presence of serum does not affect the binding of MEK1/2 to Raf-1.

Figure 2.

RRD-251 is selective for Rb/Raf-1 interaction in vivo. A, serum-stimulated binding of Raf-1 to Rb is inhibited by Rb/Raf-1 disruptors, as well as a Raf-1 peptide conjugated to penetratin; the drugs do not inhibit the binding of E2F1 to Rb. Specificity of the disruption was assessed by immunoprecipitation–Western blots. B, RRD-251 does not inhibit Rb/cyclin E interaction in cells serum-stimulated for 8 h. C, RRD-251 does not disrupt the Rb/B-Raf binding in immunoprecipitation–Western blots. D, treatment of cells with RRD-251 for 5 min in the presence of serum does not affect the binding of MEK1/2 to Raf-1.

Close modal
Figure 3.

RRD-251 does not inhibit kinase activity. A, left, RRD-251 does not inhibit cyclin D kinase activity in in vitro kinase assays. Middle, RRD-251 does not inhibit cyclin E kinase activity in in vitro kinase assays. Right, RRD-251 treatment does not inhibit Raf-1 kinase activity in in vitro kinase assays; BAY-43-9006 was used as a control. B, RRD-251 inhibits E2F1-mediated E2CAT transcription in CAT reporter assays. C, RRD-251 inhibits TS gene expression in real-time PCR experiments. D, ChIP assays show that Brg1, not Raf-1, is present on quiescent A549 cdc6, cdc25A, and TS promoters. Upon serum stimulation, Brg1 is dissociated from both promoters, correlating with Raf-1 binding. Serum stimulation in the presence of RRD-251 causes the dissociation of Raf-1 and retention of Brg1 on E2F1 responsive promoters. Serum stimulation for 16 h causes dissociation of Rb, Raf-1, HP1, Brg1, and HDAC1 from the promoters. An irrelevant antibody was used as a control for immunoprecipitations; c-fos promoter was used as a negative control.

Figure 3.

RRD-251 does not inhibit kinase activity. A, left, RRD-251 does not inhibit cyclin D kinase activity in in vitro kinase assays. Middle, RRD-251 does not inhibit cyclin E kinase activity in in vitro kinase assays. Right, RRD-251 treatment does not inhibit Raf-1 kinase activity in in vitro kinase assays; BAY-43-9006 was used as a control. B, RRD-251 inhibits E2F1-mediated E2CAT transcription in CAT reporter assays. C, RRD-251 inhibits TS gene expression in real-time PCR experiments. D, ChIP assays show that Brg1, not Raf-1, is present on quiescent A549 cdc6, cdc25A, and TS promoters. Upon serum stimulation, Brg1 is dissociated from both promoters, correlating with Raf-1 binding. Serum stimulation in the presence of RRD-251 causes the dissociation of Raf-1 and retention of Brg1 on E2F1 responsive promoters. Serum stimulation for 16 h causes dissociation of Rb, Raf-1, HP1, Brg1, and HDAC1 from the promoters. An irrelevant antibody was used as a control for immunoprecipitations; c-fos promoter was used as a negative control.

Close modal

RRD-251 inhibits E2F transcriptional activity. We next reasoned that if the disruption of the Rb/Raf-1 binding has functional consequences on cellular physiology, then RRD-251 should affect the transcriptional activity of E2F1. To examine this, transient transfection experiments were done in control A549 cells, as well as A549 cells stably expressing two different shRNA constructs (sh6 and sh8) targeting Rb; these A549 cells had significantly less Rb protein compared with parental A549 cells (Supplementary Fig. S1A). Transfection of E2F1 induced the expression of an E2CAT reporter; treatment of the transfected cells with RRD-251 repressed E2F1-mediated transcription in a dose-dependent manner in wild-type (wt) A549 cells but not in the A549 cells lacking Rb (Fig. 3B); this suggests that the presence of Rb is necessary for RRD-251 to function. ChIP analysis of the transfected E2 promoter revealed a reduction in the amount of Raf-1 on the promoter with increasing doses of RRD-251 (Supplementary Fig. S1B). We had reported that Raf-1 can be detected on proliferative promoters upon serum stimulation, and these results indicate that RRD-251 probably affects E2F-mediated transcription by dissociating Raf-1 from the promoters. The effect of RRD-251 on the expression of two endogenous E2F-regulated proliferative promoters was next examined. A549 cells were serum starved for 72 hours and serum stimulated for 24 hours in the presence or absence of RRD-251 and the level of TS gene expression was assessed by real-time PCR. Inhibition of the Rb/Raf-1 interaction led to the silencing of the TS promoter (Fig. 3C); similar results were obtained on the cdc6 gene (Supplementary Fig. S1C). We had reported that the binding of Raf-1 to Rb resulted in the dissociation of the corepressor Brg-1 from E2F-responsive proliferative promoters (18); ChIP assays were carried out to examine whether RRD-251 affects this process. It was found that the association of Raf-1 to the above promoters upon serum stimulation was disrupted by pretreatment of cells with RRD-251 (Fig. 3D). Furthermore, dissociation of the corepressor Brg-1 from these promoters was also inhibited by RRD-251. This suggests that RRD-251 can modulate the transcriptional regulatory functions of Rb by modulating its phosphorylation status and affecting its interaction with chromatin remodeling proteins like Brg-1. The association of E2F1, HDAC1, and HP1 with these promoters was not affected by RRD-251, as seen by ChIP assays (Fig. 3D).

Inhibition of proliferation by RRD-251 is dependent on Rb status. Given the ability of RRD-251 to inhibit Rb phosphorylation and repress E2F transcriptional activity, it was examined if it could inhibit cell proliferation and whether such an inhibition required a functional Rb gene. RRD-251 (20 μmol/L) was effective at inhibiting serum-induced S-phase entry in parental A549 cells but had no effect on cells stably expressing sh6 and sh8, which lacked Rb (Fig. 4A). We next tested RRD-251 on cancer cell lines that carried natural mutations or deletion of Rb. Proliferation of Saos-2 osteosarcoma cells that have a loss of Rb (24) was not affected by RRD-251, whereas the U2-OS osteosarcoma cells carrying wt Rb were arrested (Fig. 4A). RRD-251 was unable to inhibit proliferation in the Rb mutant DU145 prostate cancer cells, yet could inhibit 50% of S-phase cells in PC3 cells (wt Rb; Fig. 4A). RRD-251 (20 μmol/L) did not inhibit proliferation of lung cancer cell lines H596 and H2172, both of which harbor mutations in Rb, yet treatment of H1650 and H1299 (wt Rb) with RRD-251 inhibited proliferation by 90% and 70%, respectively (Fig. 4A). It was next examined whether RRD-251 could inhibit the proliferation of cells that have mutations in the signaling pathways that regulate Rb function rather than in the Rb gene itself. RRD-251 could inhibit S-phase entry by 50% to 65% in pancreatic cancer cell lines Aspc1, PANC1, and CAPAN2 that harbor a nonfunctional p16INK4a gene (ref. 25; Fig. 4B). RRD-251 also inhibited S-phase entry of glioblastoma cell lines U87MG and U251MG, both of which are null for p16 and PTEN (26). The metastatic human breast cancer cell line MDA-MB-231 harbors a K-Ras mutation and overexpresses epidermal growth factor receptor (EGFR; ref. 27); 20 μmol/L RRD-251 inhibited its proliferation by 56% (Fig. 4B). The A375 melanoma cell line harbors the V600E B-Raf mutation (28), and RRD-251 inhibited S-phase entry by 58%. Prostate cell lines LNCaP and PC3 both contain mutations in K-Ras and PTEN genes (29), and RRD-251 inhibited proliferation at 86% and 35%, respectively (Fig. 4B). These results indicate that disruption of Rb/Raf-1 interaction could inhibit the proliferation of cell lines harboring a wide array of mutations in upstream signaling molecules, as long as Rb itself is not altered at the genetic level. Experiments were also carried out to examine the effect of RRD-251 in suppressing the adherence-independent growth of cells in soft agar; it was found that RRD-251 could significantly inhibit the growth of A549, H1650, and SK-MEL-5, SK-MEL-28, and PANC1 cells in soft agar (Fig. 4C).

Figure 4.

RRD-251 inhibits S-phase entry dependent on Rb status. A, BrdUrd incorporation assay showing that 20 μmol/L of RRD-251 does not inhibit the proliferation of A549 cells overexpressing shRNA constructs to Rb, but arrests wt A549 cells and a nonhomologous control shRNA. RRD-251 also does not inhibit S-phase entry in cancer cell lines that contain mutant Rb. B, BrdUrd incorporation assays showing the growth arrest mediated by RRD-251 in a variety of tumor cell lines harboring various mutations. RRD-251 could effectively arrest cells with mutations in EGFR, p16, PTEN, K-Ras, and p53, but not Rb. C, RRD-251 inhibits adherence-independent growth of several cell lines in soft agar. D, overexpression of E2F1 was sufficient to overcome cell cycle arrest mediated by RRD-251, whereas cyclin D overexpression had only a partial effect.

Figure 4.

RRD-251 inhibits S-phase entry dependent on Rb status. A, BrdUrd incorporation assay showing that 20 μmol/L of RRD-251 does not inhibit the proliferation of A549 cells overexpressing shRNA constructs to Rb, but arrests wt A549 cells and a nonhomologous control shRNA. RRD-251 also does not inhibit S-phase entry in cancer cell lines that contain mutant Rb. B, BrdUrd incorporation assays showing the growth arrest mediated by RRD-251 in a variety of tumor cell lines harboring various mutations. RRD-251 could effectively arrest cells with mutations in EGFR, p16, PTEN, K-Ras, and p53, but not Rb. C, RRD-251 inhibits adherence-independent growth of several cell lines in soft agar. D, overexpression of E2F1 was sufficient to overcome cell cycle arrest mediated by RRD-251, whereas cyclin D overexpression had only a partial effect.

Close modal

We next reasoned that, if RRD-251 targets selectively the Rb/Raf-1 interaction, the forced expression of a downstream target of Rb, such as E2F1, but not of the upstream regulator cyclin D, would rescue the antiproliferative effects of RRD-251. To this end, A549 cells were serum starved for 48 hours and infected with Ad-E2F1 or Ad-cyclin D in the presence or absence of 20 μmol/L of RRD-251 for 36 hours. Ad-GFP infected cells were used as a control. BrdUrd incorporation assays showed that ectopic expression of E2F1 efficiently overcame the antiproliferative activity of RRD-251, whereas overexpression of cyclin D had only a partial effect (Fig. 4D), showing that the growth inhibition by RRD-251 occurs at the level of Rb and its downstream target, E2F1, can rescue the growth suppression.

RRD-251 inhibits angiogenesis in vitro and in vivo. Raf-1 kinase plays a role in facilitating angiogenesis (30, 31), and it has been suggested that Raf-1–mediated inactivation of Rb is involved in the process (18). To examine whether angiogenic tubule formation could be inhibited by RRD-251, HUVECs were grown in Matrigel in the presence or absence of 20 μmol/L RRD-251; RRD-251 significantly inhibited the angiogenic tubule formation (Fig. 5A). These results were confirmed in an ex vivo experiment using rat aortic rings. As shown in Fig. 5B, 20 μmol/L RRD-251 was able to inhibit angiogenic sprouting from rat aortic rings grown in Matrigel for 7 days. Quantitation of vessel area showed a significant reduction in angiogenesis (P = 0.000007; Supplementary Fig. S2A). We next examined whether RRD-251 could inhibit angiogenesis in Matrigel plugs in vivo (22). Aythmic nude mice were injected with cold Matrigel in both flanks. Mice were given either vehicle or RRD-251 50 MPK body weight by i.p. injection daily for 1 week. Mice were injected with 100 MPK FITC-Dextran via the tail vein before euthanasia, and Matrigel plugs were fixed in formalin. Angiogenesis in the entire plugs were assessed by confocal imaging. FITC images showed growth of angiogenic tubules in plugs from mice that received vehicle; in contrast, there was a remarkable inhibition of angiogenic vessel formation in the Matrigel plugs from mice treated with RRD-251 (Fig. 5C). Quantitation of vessel intensity plotted as relative angiogenesis per image shows significant inhibition; P = 0.0004 (n = 12; Supplementary Fig. S2B). Further examination of the Matrigel plugs by H&E staining showed a complete inhibition of cells migrating into the Matrigel for vessel formation (Fig. 5D). It was also examined if RRD-251 could inhibit vascular endothelial growth factor (VEGF) levels in cell culture. A549 cells treated with RRD-251 for 24 hours displayed decreased VEGF levels compared with control (Supplementary Fig. S2C). Furthermore, treatment with RRD-251 in the presence of VEGF for 12 hours could prevent Rb phosphorylation in HAEC cells (Supplementary Fig. S2D).

Figure 5.

RRD-251 inhibits angiogenesis in vitro and in vivo. A, RRD-251 inhibits HUVEC angiogenic tubule formation in Matrigel. B, RRD-251 inhibits angiogenesis in a rat aorta Matrigel model. C, confocal FITC images of Matrigel plugs from nude mice treated with vehicle or RRD-251 at 50 MPK daily for 1 wk. D, H&E staining of Matrigel plugs from nude mice treated with vehicle or RRD-251 at 50 MPK. H&E images display 1/4 of Matrigel plug.

Figure 5.

RRD-251 inhibits angiogenesis in vitro and in vivo. A, RRD-251 inhibits HUVEC angiogenic tubule formation in Matrigel. B, RRD-251 inhibits angiogenesis in a rat aorta Matrigel model. C, confocal FITC images of Matrigel plugs from nude mice treated with vehicle or RRD-251 at 50 MPK daily for 1 wk. D, H&E staining of Matrigel plugs from nude mice treated with vehicle or RRD-251 at 50 MPK. H&E images display 1/4 of Matrigel plug.

Close modal

Antitumor activity of RRD-251. Given the ability of RRD-251 to inhibit cell proliferation, adherence-independent growth, and angiogenesis, we examined whether RRD-251 could inhibit tumor growth in vivo in nude mouse xenograft models. Athymic nude mice were implanted s.c. with 1 × 107 A549 cells bilaterally, and the tumors were allowed to reach 200 mm3 in size before p.o. or i.p. administration of RRD-251 or vehicle (18, 23). Tumors from vehicle-treated mice grew to an average size of 1,040 ± 128 mm3; in contrast, tumors in mice treated with RRD-251 did not grow and even regressed slightly (50 MPK i.p. 145 ± 20 mm3, 150 MPK p.o. 148 ± 32 mm3; Fig. 6A,, left). P.o. dose response experiments were carried out on A549 xenografts, which resulted in RRD-251 at 100 MPK and 150 MPK completely inhibiting tumor growth (Fig. 6A,, middle). Tumors from vehicle-treated mice reached an average size of 996 ± 180 mm3; in contrast, tumors in mice treated with RRD-251 (p.o.) responded in a dose-dependent manner. Complete inhibition was seen in 100 MPK p.o. at 293 ± 44 mm3 and 150 MPK p.o. at 237 ± 67 mm3 (Fig. 6A,, middle). Similar results were observed with H1650 xenograft tumors; RRD-251 inhibited tumor growth significantly [2,185 ± 326 mm3 in vehicle-treated animals compared with 557 ± 76 mm3 in RRD-251 (50 MPK i.p.)–treated animals; Fig. 6A , right].

Figure 6.

I.p. and p.o. administration of RRD-251 inhibits human tumor growth in nude mice. A, left, A549 cells xenotransplanted bilaterally into the flanks of athymic nude mice were allowed to grow for 14 d until tumor volume reached 200 mm3; daily administration of RRD-251 at 50 MPK i.p. and 150 MPK p.o. completely inhibited tumor growth. Middle, dose response of RRD-251 given by oral gavage at 100 MPK and 150 MPK could completely inhibit tumor growth. Right, RRD-251 inhibited H1650 xenograft tumor growth in nude mice. B, immunohistochemical staining of tumors from mice treated with RRD-251. Tumors were stained with Ki-67 for proliferation, pRb for cell cycle, and CD31 for angiogenesis. C, both doses of RRD-251 inhibit the Rb/Raf-1 interaction in tumor lysates without inhibiting Rb/E2F1 interaction, as seen by immunoprecipitation–Western blots. D, left and right, inhibition of tumor growth is dependent on a functional Rb protein. A549-sh6 and A549-sh8 cells were implanted into the flanks of nude mice. RRD-251 was unable to inhibit tumor growth in tumors lacking Rb protein.

Figure 6.

I.p. and p.o. administration of RRD-251 inhibits human tumor growth in nude mice. A, left, A549 cells xenotransplanted bilaterally into the flanks of athymic nude mice were allowed to grow for 14 d until tumor volume reached 200 mm3; daily administration of RRD-251 at 50 MPK i.p. and 150 MPK p.o. completely inhibited tumor growth. Middle, dose response of RRD-251 given by oral gavage at 100 MPK and 150 MPK could completely inhibit tumor growth. Right, RRD-251 inhibited H1650 xenograft tumor growth in nude mice. B, immunohistochemical staining of tumors from mice treated with RRD-251. Tumors were stained with Ki-67 for proliferation, pRb for cell cycle, and CD31 for angiogenesis. C, both doses of RRD-251 inhibit the Rb/Raf-1 interaction in tumor lysates without inhibiting Rb/E2F1 interaction, as seen by immunoprecipitation–Western blots. D, left and right, inhibition of tumor growth is dependent on a functional Rb protein. A549-sh6 and A549-sh8 cells were implanted into the flanks of nude mice. RRD-251 was unable to inhibit tumor growth in tumors lacking Rb protein.

Close modal

Tumors were harvested at the end of the treatment and analyzed by immunohistochemical staining with H&E and antibodies to Ki-67, phosphorylated Rb (807, 811), and CD-31. A significant inhibition of proliferation was observed in tumors from RRD-251–treated animals, as seen by Ki-67 staining (Fig. 6B); phosphorylation of Rb was also reduced as seen by staining with an antibody to phosphorylated Rb (Fig. 6B). Tumors also showed a reduction in microvasculature, as seen by CD31 staining (Fig. 6B). Quantitation of Ki-67 staining, phosphorylated Rb staining, and CD31 staining is shown in Supplementary Fig. S3A–C. To assess whether RRD-251 affected its target, tumors lysates were prepared from vehicle-treated and RRD-251–treated mice and Rb/Raf-1 interaction assessed by immunoprecipitation–Western blots. RRD-251 treatment had led to a reduction in Rb/Raf-1, but not Rb/E2F1 interaction in tumor xenografts (Fig. 6C).

Tumor growth inhibition by RRD-251 is Rb-dependent. Because RRD-251 did not inhibit the proliferation of A549 cells lacking Rb in vitro, experiments were done to assess whether tumors generated from these cells can respond to RRD-251 in vivo. An experiment as in Fig. 6A was carried out on nude mice carrying tumors from A549 cells stably expressing shRNAs for Rb (sh6 and sh8). Interestingly, these tumors did not respond to RRD-251 and continued to grow at the rate of the vehicle treated tumors (Fig. 6D , left and right). To examine whether the sh6 and sh8 tumors maintained down-regulation of Rb, lysates were made from the sh6 and sh8 tumors at the end of the experiment and a Western blot was done for Rb. It was found that these tumors lacked Rb, further confirming that RRD-251 specifically targets the Rb/Raf-1 protein interaction to inhibit cell proliferation and tumor growth (Supplementary Fig. S3D).

The Ras/Raf/Mek/MAPK cascade is a proliferative pathway induced by a wide array of growth factors, is activated in many human tumors (3234), and is an attractive target for the development of anticancer drugs (30, 31, 35, 36). Raf-1 kinase itself has been targeted for cancer therapy, and two clinical attempts have been made to inhibit Raf-1 activity in patients (3739). It has been shown that signaling events involving the MAPK cascade do not proceed in a linear fashion and instead they have substrates outside the cascade (17, 40, 41). In this context, the Rb protein seems to be a cellular target of the Raf-1 kinase outside the MAPK cascade. In addition, Rb/Raf-1 binding was elevated in tumor compared with adjacent normal controls (19), suggesting that Raf-1–mediated inactivation of Rb might contribute to oncogenesis. Whereas it is established that Rb gene itself is mutated in cancers like retinoblastoma, osteosarcoma, and small cell lung carcinoma, majority of tumors harbor mutations in the upstream regulators of Rb function (7, 8). These include genes like Ras, PTEN, p16INK4, as well as receptor tyrosine kinases (4244). Our results show that the disruption of the Rb/Raf-1 interaction can be fruitfully used to inhibit the proliferation of cells harboring such mutations in the Rb regulatory pathway. Thus, we believe that these molecules have the potential to target a wide variety of human cancers.

Whereas inhibitors of cell proliferation, DNA-damaging agents, and microtubule disruptors have widely been used as anticancer agents, developments in the past decade have shown that targeting angiogenesis is also an effective way of combating tumor growth (45). In this context, our results show that RRD-251 cannot only inhibit cell proliferation, but can also inhibit neoangiogenesis in vitro and in vivo. Given the published reports that Raf-1 kinase contributes to angiogenesis and that VEGF can induce Rb phosphorylation, it is likely that RRD-251 is inhibiting angiogenesis by affecting these molecules (31, 46). The ability of RRD-251 to inhibit both cell proliferation and angiogenesis might be acting in a two-pronged manner to inhibit the growth of tumors in vivo; as can be imagined, these are desirable features in anticancer drugs.

Whereas it has been difficult to generate small molecule inhibitors of protein-protein interactions that are clinically active (39), recent success in disrupting the hdm2-p53 (47) interaction shows that this is a viable strategy to develop novel anticancer drugs. Identification of RRD-251 as a cell-permeable, p.o. available, and highly selective inhibitor of the Rb/Raf-1 interaction is an example of the practicality of targeting protein-protein interaction for cancer therapy. Although we find that RRD-251 inhibits Rb/Raf-1 in vitro at nanomolar concentrations in an in vitro ELISA assay, higher concentrations are needed to inhibit cell proliferation, as well as growth of cells in soft agar; this finding is similar to what has been observed with other anticancer drugs, such as BAY-43-9006, R547, and Iressa (4850). At the same time, our in vivo studies show that concentrations can be achieved in vivo where RRD-251 has a significant therapeutic benefit.

The finding that RRD-251 is effective at inhibiting the proliferation of cells harboring a wide variety of mutations in signaling cascades that inactivate Rb, but does not affect cells carrying mutated Rb or no Rb, shows the specificity of this agent. Rb protein has been reported to interact with ∼100 proteins in the cell; it can be imagined that small molecules that can maintain the tumor suppressor functions of Rb by disrupting its physical interaction with other proteins would be a fruitful avenue to develop novel anticancer drugs.

No potential conflicts of interest were disclosed.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

P. Dasgupta and A. Carie contributed equally to this work.

Current address for P. Dasgupta: Department of Pharmacology, Physiology and Toxicology, Marshall University, BBSC, 1 John Marshall Drive, Huntington, WV 25755. Current address for M. Carless: Southwest Foundation for Biomedical Research, P.O. Box 760549, San Antonio, TX 78245-0549.

Grant support: National Cancer Institute grants CA63136 and CA118210. R. Kinkade is a recipient of a predoctoral award from American Heart Association.

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.

1
Sherr CJ. The ins and outs of RB: coupling gene expression to the cell cycle clock.
Trends Cell Biol
1994
;
4
:
15
–8.
2
Weinberg RA. The retinoblastoma protein and cell cycle control.
Cell
1995
;
81
:
323
–30.
3
Reddy GP. Cell cycle: regulatory events in G1-S transition of mammalian cells.
J Cell Biochem
1994
;
54
:
379
–86.
4
Reed SI. Control of the G1-S transition.
Cancer Surv
1997
;
29
:
7
–23.
5
Nevins JR. Cell cycle targets of the DNA tumor viruses.
Curr Opin Genet Dev
1994
;
4
:
130
–4.
6
Nevins JR. The Rb/E2F pathway and cancer.
Hum Mol Genet
2001
;
10
:
699
–703.
7
Friend SH, Bernards R, Rogelj S, et al. A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma.
Nature
1986
;
323
:
643
–6.
8
Harbour JW, Lai SL, Whang-Peng J, Gazdar AF, Minna JD, Kaye FJ. Abnormalities in structure and expression of the human retinoblastoma gene in SCLC.
Science
1988
;
241
:
353
–7.
9
Cobrinik D. Pocket proteins and cell cycle control.
Oncogene
2005
;
24
:
2796
–809.
10
Nevins JR. E2F: a link between the Rb tumor suppressor protein and viral oncoproteins.
Science
1992
;
258
:
424
–9.
11
DeGregori J. The genetics of the E2F family of transcription factors: shared functions and unique roles.
Biochim Biophys Acta
2002
;
1602
:
131
–50.
12
Dimova DK, Dyson NJ. The E2F transcriptional network: old acquaintances with new faces.
Oncogene
2005
;
24
:
2810
–26.
13
Trimarchi JM, Lees JA. Sibling rivalry in the E2F family.
Nat Rev Mol Cell Biol
2002
;
3
:
11
–20.
14
Harbour JW, Luo RX, Dei Santi A, Postigo AA, Dean DC. Cdk phosphorylation triggers sequential intramolecular interactions that progressively block Rb functions as cells move through G1.
Cell
1999
;
98
:
859
–69.
15
Knudsen ES, Wang JY. Differential regulation of retinoblastoma protein function by specific Cdk phosphorylation sites.
J Biol Chem
1996
;
271
:
8313
–20.
16
Knudsen ES, Wang JY. Dual mechanisms for the inhibition of E2F binding to RB by cyclin-dependent kinase-mediated RB phosphorylation.
Mol Cell Biol
1997
;
17
:
5771
–83.
17
Wang S, Ghosh RN, Chellappan SP. Raf-1 physically interacts with Rb and regulates its function: a link between mitogenic signaling and cell cycle regulation.
Mol Cell Biol
1998
;
18
:
7487
–98.
18
Dasgupta P, Sun J, Wang S, et al. Disruption of the Rb-Raf-1 interaction inhibits tumor growth and angiogenesis.
Mol Cell Biol
2004
;
24
:
9527
–41.
19
Dasgupta P, Rastogi S, Pillai S, et al. Nicotine induces cell proliferation by β-arrestin-mediated activation of Src and Rb-Raf-1 pathways.
J Clin Invest
2006
;
116
:
2208
–17.
20
Rastogi S, Joshi B, Dasgupta P, Morris M, Wright K, Chellappan S. Prohibitin facilitates cellular senescence by recruiting specific corepressors to inhibit E2F target genes.
Mol Cell Biol
2006
;
26
:
4161
–71.
21
Nicosia RF, Zhu WH, Fogel E, Howson KM, Aplin AC. A new ex vivo model to study venous angiogenesis and arterio-venous anastomosis formation.
J Vasc Res
2005
;
42
:
111
–9.
22
Akhtar N, Dickerson EB, Auerbach R. The sponge/Matrigel angiogenesis assay.
Angiogenesis
2002
;
5
:
75
–80.
23
Carie AE, Sebti SM. A chemical biology approach identifies a β-2 adrenergic receptor agonist that causes human tumor regression by blocking the Raf-1/Mek-1/Erk1/2 pathway.
Oncogene
2007
;
26
:
3777
–88.
24
Wang S, Nath N, Minden A, Chellappan S. Regulation of Rb and E2F by signal transduction cascades: divergent effects of JNK1 and p38 kinases.
EMBO J
1999
;
18
:
1559
–70.
25
Gerdes B, Bartsch DK, Ramaswamy A, et al. Multiple primary tumors as an indicator for p16INK4a germline mutations in pancreatic cancer patients?
Pancreas
2000
;
21
:
369
–75.
26
Weller M, Rieger J, Grimmel C, et al. Predicting chemoresistance in human malignant glioma cells: the role of molecular genetic analyses.
Int J Cancer
1998
;
79
:
640
–4.
27
Price JT, Tiganis T, Agarwal A, Djakiew D, Thompson EW. Epidermal growth factor promotes MDA-MB-231 breast cancer cell migration through a phosphatidylinositol 3′-kinase and phospholipase C-dependent mechanism.
Cancer Res
1999
;
59
:
5475
–8.
28
Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer.
Nature
2002
;
417
:
949
–54.
29
Tanaka M, Rosser CJ, Grossman HB. PTEN gene therapy induces growth inhibition and increases efficacy of chemotherapy in prostate cancer.
Cancer Detect Prev
2005
;
29
:
170
–4.
30
Alavi A, Hood JD, Frausto R, Stupack DG, Cheresh DA. Role of Raf in vascular protection from distinct apoptotic stimuli.
Science
2003
;
301
:
94
–6.
31
Hood JD, Cheresh DA. Targeted delivery of mutant Raf kinase to neovessels causes tumor regression.
Cold Spring Harb Symp Quant Biol
2002
;
67
:
285
–91.
32
Beeram M, Patnaik A, Rowinsky EK. Raf: a strategic target for therapeutic development against cancer.
J Clin Oncol
2005
;
23
:
6771
–90.
33
Kolch W. Meaningful relationships: the regulation of the Ras/Raf/MEK/ERK pathway by protein interactions.
Biochem J
2000
;
351
Pt 2:
289
–305.
34
Kolch W, Kotwaliwale A, Vass K, Janosch P. The role of Raf kinases in malignant transformation.
Expert Rev Mol Med
2002
;
2002
:
1
–18.
35
Kinkade R, Dasgupta P, Chellappan S. The ABC's of targeting Raf: novel approaches to cancer therapy.
Curr Cancer Ther Rev
2006
;
2
:
305
–14.
36
Sridhar SS, Hedley D, Siu LL. Raf kinase as a target for anticancer therapeutics.
Mol Cancer Ther
2005
;
4
:
677
–85.
37
Rudin CM, Holmlund J, Fleming GF, et al. Phase I Trial of ISIS 5132, an antisense oligonucleotide inhibitor of c-raf-1, administered by 24-hour weekly infusion to patients with advanced cancer.
Clin Cancer Res
2001
;
7
:
1214
–20.
38
Wilhelm S, Carter C, Lynch M, et al. Discovery and development of sorafenib: a multikinase inhibitor for treating cancer.
Nat Rev Drug Discov
2006
;
5
:
835
–44.
39
Arkin MR, Wells JA. Small-molecule inhibitors of protein-protein interactions: progressing towards the dream.
Nat Rev Drug Discov
2004
;
3
:
301
–17.
40
Ehrenreiter K, Piazzolla D, Velamoor V, et al. Raf-1 regulates Rho signaling and cell migration.
J Cell Biol
2005
;
168
:
955
–64.
41
Lee KY, Ladha MH, McMahon C, Ewen ME. The retinoblastoma protein is linked to the activation of Ras.
Mol Cell Biol
1999
;
19
:
7724
–32.
42
Bai F, Pei XH, Pandolfi PP, Xiong Y. p18 Ink4c and Pten constrain a positive regulatory loop between cell growth and cell cycle control.
Mol Cell Biol
2006
;
26
:
4564
–76.
43
Braden WA, Lenihan JM, Lan Z, et al. Distinct action of the retinoblastoma pathway on the DNA replication machinery defines specific roles for cyclin-dependent kinase complexes in prereplication complex assembly and S-phase progression.
Mol Cell Biol
2006
;
26
:
7667
–81.
44
Peeper DS, Upton TM, Ladha MH, et al. Ras signalling linked to the cell-cycle machinery by the retinoblastoma protein.
Nature
1997
;
386
:
177
–81.
45
Hood JD, Cheresh DA. Building a better Trap.
Proc Natl Acad Sci U S A
2003
;
100
:
8624
–5.
46
Suzuma K, Takahara N, Suzuma I, et al. Characterization of protein kinase C β isoform's action on retinoblastoma protein phosphorylation, vascular endothelial growth factor-induced endothelial cell proliferation, and retinal neovascularization.
Proc Natl Acad Sci U S A
2002
;
99
:
721
–6.
47
Issaeva N, Bozko P, Enge M, et al. Small molecule RITA binds to p53, blocks p53-HDM-2 interaction and activates p53 function in tumors.
Nat Med
2004
;
10
:
1321
–8.
48
DePinto W, Chu XJ, Yin X, et al. In vitro and in vivo activity of R547: a potent and selective cyclin-dependent kinase inhibitor currently in phase I clinical trials.
Mol Cancer Ther
2006
;
5
:
2644
–58.
49
Moasser MM, Basso A, Averbuch SD, Rosen N. The tyrosine kinase inhibitor ZD1839 (“Iressa”) inhibits HER2-driven signaling and suppresses the growth of HER2-overexpressing tumor cells.
Cancer Res
2001
;
61
:
7184
–8.
50
Wilhelm SM, Carter C, Tang L, et al. BAY 43–9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis.
Cancer Res
2004
;
64
:
7099
–109.

Supplementary data