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

  • Register
  • Log in
  • Log out
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Focus on Computer Resources
    • 75th Anniversary
    • Meeting Abstracts
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • OnlineFirst
    • Editors' Picks
    • Citations
    • Author/Keyword
  • News
    • Cancer Discovery News
  • AACR Publications
    • 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
  • Log out

Search

  • Advanced search
Cancer Research
Cancer Research

Advanced Search

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Focus on Computer Resources
    • 75th Anniversary
    • Meeting Abstracts
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • OnlineFirst
    • Editors' Picks
    • Citations
    • Author/Keyword
  • News
    • Cancer Discovery News
Priority Reports

Demonstration of a Genetic Therapeutic Index for Tumors Expressing Oncogenic BRAF by the Kinase Inhibitor SB-590885

Alastair J. King, Denis R. Patrick, Roberta S. Batorsky, Maureen L. Ho, Hieu T. Do, Shu Yun Zhang, Rakesh Kumar, David W. Rusnak, Andrew K. Takle, David M. Wilson, Erin Hugger, Lifu Wang, Florian Karreth, Julie C. Lougheed, Jae Lee, David Chau, Thomas J. Stout, Earl W. May, Cynthia M. Rominger, Michael D. Schaber, Lusong Luo, Ami S. Lakdawala, Jerry L. Adams, Rooja G. Contractor, Keiran S.M. Smalley, Meenhard Herlyn, Michael M. Morrissey, David A. Tuveson and Pearl S. Huang
Alastair J. King
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Denis R. Patrick
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Roberta S. Batorsky
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Maureen L. Ho
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hieu T. Do
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shu Yun Zhang
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rakesh Kumar
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
David W. Rusnak
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Andrew K. Takle
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
David M. Wilson
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Erin Hugger
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lifu Wang
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Florian Karreth
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Julie C. Lougheed
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jae Lee
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
David Chau
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Thomas J. Stout
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Earl W. May
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Cynthia M. Rominger
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michael D. Schaber
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lusong Luo
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ami S. Lakdawala
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jerry L. Adams
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rooja G. Contractor
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Keiran S.M. Smalley
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Meenhard Herlyn
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michael M. Morrissey
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
David A. Tuveson
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Pearl S. Huang
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/0008-5472.CAN-06-2554 Published December 2006
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Oncogenic BRAF alleles are both necessary and sufficient for cellular transformation, suggesting that chemical inhibition of the activated mutant protein kinase may reverse the tumor phenotype. Here, we report the characterization of SB-590885, a novel triarylimidazole that selectively inhibits Raf kinases with more potency towards B-Raf than c-Raf. Crystallographic analysis revealed that SB-590885 stabilizes the oncogenic B-Raf kinase domain in an active configuration, which is distinct from the previously reported mechanism of action of the multi-kinase inhibitor, BAY43-9006. Malignant cells expressing oncogenic B-Raf show selective inhibition of mitogen-activated protein kinase activation, proliferation, transformation, and tumorigenicity when exposed to SB-590885, whereas other cancer cell lines and normal cells display variable sensitivities or resistance to similar treatment. These studies support the validation of oncogenic B-Raf as a target for cancer therapy and provide the first evidence of a correlation between the expression of oncogenic BRAF alleles and a positive response to a selective B-Raf inhibitor. (Cancer Res 2006; 66(23): 11100-5)

  • B-Raf
  • c-Raf
  • MEK
  • kinase
  • inhibitor
  • melanoma
  • cancer

Introduction

The success of novel cancer therapies depends on the identification of functional targets that play an essential role in tumor growth and survival. One strategy for identifying such targets is to consider products of genes that have been amplified or have acquired activating mutations in a tumor genotype. This strategy is attractive if a causal link can be established between these events (gene amplification, mutation) and the disease state, and also if the “gain of function” that is acquired in the tumor cells can be antagonized by traditional small molecule inhibitors.

The activation and amplification of growth factor signaling pathways are commonly observed in many cancers. The clinical utility of therapies targeted to the Erb family growth factor receptors (e.g., trastuzumab, gefitinib, erlotinib, and lapatinib) and Bcr-Abl (e.g., imatinib), shows the dependence of the cancer phenotype on the enzymatic activity of these oncogenes (reviewed in ref. 1). Recently, a subset of patients with lung tumors expressing mutant, activated forms of ErbB1, were shown to be the most responsive to targeted kinase inhibition by gefitinib (reviewed in ref. 2). This correlation, between the presence of an activating mutation in an oncogene and a positive response to an inhibitor, suggests that responding populations to a targeted therapy can be identified based on the genetic signature of the target in tumors.

The requirement for growth factor signaling in tumor cells can be replaced by activating mutations in downstream effectors of the signaling pathway. In particular, the KRAS gene is frequently mutated to activated forms in pancreatic cancer, and activated forms of the BRAF gene have been identified in a number of neoplasms, including melanoma, ovarian, colorectal, thyroid, cholangiocarcinoma, and lung adenocarcinoma (reviewed in ref. 3). Activating mutations in the regulatory domain of B-Raf, most frequently, V600E, bypass the need for growth factor stimulation and create a constitutively activated kinase that has a 500-fold higher kinase activity, relative to the basal activity of wild-type protein ( 4). Expression of the activated BRAF gene has been shown to be both sufficient and necessary to maintain tumorigenesis in experimental settings. Introduction of activated BRAFV600E into immortalized melanocytes is sufficient to impart a transformed phenotype ( 5) and ablation of BRAFV600E mRNA by RNA interference induces growth inhibition and cell death in human tumor cell lines that express activated B-Raf ( 6, 7). Although it is clear that knock-down of B-Raf protein by RNA interference can impede the survival of tumor cells, these experiments do not address the utility of an enzyme inhibitor for this kinase in tumor cells.

A potent and specific inhibitor of the B-Raf kinase, SB-590885, was evaluated as a candidate therapeutic agent in cell culture models and used here to address the significance of an activating BRAF mutation in cancer. Importantly, SB-590885 showed potent and selective growth inhibition of tumor cells harboring a mutant BRAF allele, both in human tumor cell lines and primary tumor samples, validating oncogenic B-Raf as an attractive therapeutic target in human malignancies.

Materials and Methods

Compounds and B-Raf/SB-590885 complex crystallization. SB-590885 and CI-1040 were synthesized ( 8, 9) and characterized to ensure purity. Human B-Raf kinase domain (residues 445–723) was expressed in Sf-9 insect cells using a modified pAcGP67 baculovirus DNA transfer vector (BD PharMingen, San Diego, CA). B-Raf produced from this vector contained an NH2-terminal hexahistidine tag and TEV protease cleavage site (MLLGSH6GENLYFQGS) and is referred to as N-His6-B-Raf. Production of N-His6-B-Raf in the soluble fraction was increased by coexpression with human Cdc37. After recovery over Ni-NTA resin (Qiagen, Valencia, CA), N-His6-B-Raf was exchanged into 20 mmol/L Bis-Tris Propane (pH 7.0), 10% glycerol, 5 mmol/L 2-mercaptoethanol for cleavage with recombinant TEV protease. Cleaved B-Raf was then purified away from cleavage products over Ni-NTA. B-Raf [2 mg/mL in 20 mmol/L Bis-Tris Propane (pH 7.0), 15% glycerol, and 1 mmol/L DTT] was incubated with 0.5 mmol/L of SB-590885 for 30 minutes at room temperature, before mixing 1 μL of this solution with 1 μL of a reservoir solution of 8% PEG 4000, 0.1 mol/L of Tris (pH 8.5), and 0.4 mol/L of LiCl containing apo B-Raf micro-seeds. Complex crystals were grown in sitting drops at 18°C and transferred to a cryoprotectant solution, consisting of reservoir solution supplemented with 4% PEG 4000 and 35% ethylene glycol, before being flash-cooled by immersion in liquid nitrogen. Crystals belonged to the space group P41212 (a = 95.638 Å, c = 159.222 Å) and contained two complexes in the asymmetric unit. X-ray diffraction data were collected from a single crystal at beamline 9-1 of the Stanford Synchrotron Radiation Laboratory. Resultant images were processed with MOSFLM ( 10) and scaled with SCALA (Collaborative Computational Project, 1994). A molecular replacement solution was found with MOLREP ( 11) using a previously solved structure of B-Raf bound to another inhibitor as a search model. The model was subsequently refined in REFMAC ( 12) using rigid body refinement and maximum likelihood procedures. Fourier syntheses with coefficients of Fo-Fc and 2Fo-Fc yielded clearly interpretable electron density for the ligand, however, portions of the protein were adjusted by iterative cycles of manual rebuilding in QUANTA (Accelrys, Inc., San Diego, CA) and refinement in REFMAC before the ligand was placed into the Fo-Fc map using QUANTA/X-Ligand. The complex was subsequently refined using maximum likelihood procedures in REFMAC. Water molecules were built and manually inspected in QUANTA.

Cell lines. Human cell lines were obtained from the American Type Culture Collection (Manassas, VA), the Coriell Institute for Medical Research (Camden, NJ), and provided by M. Herlyn. BRAF mutations were determined by cDNA sequencing of the BRAF (GenBank accession no. M95712) mRNA isolated from growing cells using SuperScript III first-strand synthesis reverse transcription-PCR (Invitrogen, Carlsbad, CA) and HotStar Taq DNA polymerase (Qiagen). The primers used for amplification can be provided upon request. Amplicons were purified using Qiagen QIAquick PCR purification kits, sequenced with BigDye Terminator chemistry (Applied Biosystems, Foster City, CA). Newborn human melanocytes were prepared as described in http://www.wistar.org/herlyn/resource_culture_isolation.htm.

Biochemical and cellular assays. Western blot analysis was done as described using a LI-COR Odyssey IR imager ( 13). For proliferation assays, cells were treated with compounds in 0.1% DMSO and incubated for 72 hours at 37°C, 5% CO2. Viable cells were quantified using CellTiter-Glo reagent (Promega, Madison, WI) and luminescence detection on a Victor 2V plate reader (Perkin-Elmer, Turku, Finland). Cells were prepared for cell cycle analysis on a Becton Dickinson FACScan, according to the manufacturer's instructions. Data was acquired and analyzed using CellQuest v3.3 software. Anchorage-independent growth assays were done as described elsewhere ( 6), with inhibitors or DMSO vehicle included in the agar layer. Cultures were re-fed with media and inhibitor or DMSO every 5 to 7 days for a total of 28 days. Colonies were visualized and photographed by conventional light microscopy and quantified by counting on a grid in triplicate.

Spheroid preparation and growth assay. Human melanoma cells were isolated from metastatic lesions and grown to form spheroids, as previously described ( 14). Spheroid cultures were treated with compounds or DMSO and incubated for 72 hours. Metabolic activity was measured by conversion of the tetrazolium salt, WST-1 (Roche Diagnostics, Indianapolis, IN), in cultures for 4 hours at 37°C. Lack of metabolic activity in spheroid cultures was determined using the “Live/Dead cell kit” (Molecular Probes, Invitrogen, Carlsbad, CA), which does not distinguish between senescent cells and cells undergoing early apoptosis. Spheroids were photographed under inverted fluorescence microscopy using FITC/TRITC channels.

A375P xenograft model. The pharmacokinetic properties and safety of SB-590885, following i.p. injection, were determined and 50 mg/kg daily injections were found to give therapeutic levels with minimal body weight changes. Tumors were initiated in 8- to 12-week-old female nude mice by s.c. injection of 5 × 106 A375P cells in Matrigel suspension, and 3 weeks after tumor induction when the tumors had reached a volume of 150 to 250 mm3, mice were randomized into groups of eight prior to treatment. Animals were treated with vehicle [2% N,N-dimethylacetamide, 2% Cremophor EL, and 96% acidified water (pH ∼4–5)], or vehicle containing 50 mg/kg of SB-590885 daily for 21 days. A cohort of mice treated with SB-590885 were then observed an additional 14 days following cessation of treatment. Tumor volume was measured for 55 days by calipers twice weekly. All in vivo procedures were carried out in accordance with protocols approved by the GSK Institutional Animal Care and Use Committee.

Results

SB-590885 is a novel inhibitor of B-Raf that stabilizes the open conformation of the enzyme. Chemical inhibitors of B-Raf were designed as described previously ( 8). Using a coupled assay in which MEK activity was dependent on B-Raf activity, the triarylimidazole, SB-590885 ( Fig. 1A ), was shown to potently inhibit the oncogenic B-Raf protein kinase with a Ki app of 0.16 ± 0.03 nmol/L. 12 Interestingly, selectivity towards B-Raf was shown as SB-590885 inhibited c-Raf with a Ki app of 1.72 ± 0.65 nmol/L. These results showed that SB-590885 was a more potent inhibitor than the previously described Raf/VEGFR kinase inhibitor BAY 43-9006 (Ki app = 38 nmol/L for mutant B-Raf, 6 nmol/L for c-Raf) and exhibited more selectivity towards B-Raf inhibition ( 15). SB-590885 was further profiled against a panel of 48 other human kinases, comprising representatives from all of the main branches of the human kinome ( 16). Strikingly, this compound displayed remarkable specificity for B-Raf inhibition, with significant activity measured only against c-Raf and minimal off-target activity ( Table 1 ). Therefore, SB-590885 is a novel, low molecular weight compound that potently and selectively inhibits mutant B-Raf kinase activity.

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

SB-590885 is a potent and selective inhibitor of B-Raf kinase. A, SB-590885 is a small molecule kinase inhibitor of the triarylimidazole class. B, the crystal structure of SB-590885 bound to B-RafV600E reveals competition for the ATP-binding pocket and interaction with nearby residues. Dashed lines, hydrogen bonds and electrostatic interactions. C, view of the packing between B-Raf and SB-590885. Light blue, F583 and W531 side chain surfaces; beige, protein surfaces. The ligand is shown with a transparent surface.

View this table:
  • View inline
  • View popup
Table 1.

Kinase inhibition selectivity profile for SB-590995

The structural basis for B-Raf enzyme inhibition by SB-590885 was investigated by preparing co-crystals of the B-RafV600E kinase domain and SB-590885, with X-ray diffraction patterns resolved at a resolution of 2.9 Å. Analysis determined that SB-590885 occupies the ATP-binding pocket and binds to an active conformation of B-Raf ( Fig. 1B and C). The origin of the selectivity of SB-590885 for B-Raf seems to be due to interactions with several B-Raf amino acids, as well as the presence of the indane-oxime. In particular, a phenylalanine residue (F583) in the COOH-terminal lobe forms favorable π-stacking interactions with the imidazole and pyridine rings of SB-590885. Furthermore, a tryptophan residue (W531) is located above the opposing face of the pyridine, also forming favorable π-stacking interactions. Additionally, the indane-oxime forms a strong interaction with the salt bridge of B-Raf as well as a hydrogen bond with a backbone amide in the activation loop, thus likely contributing to the selectivity and potency of SB-590885. The stabilization of an active conformation of B-Raf by SB-590885 is in contrast to the report that BAY 43-9006 binds to the inactive conformation of B-Raf ( 4), demonstrating an alternative means to inhibit B-Raf kinase.

SB-590885 potently inhibits ERK phosphorylation and proliferation in tumor cells expressing B-RafV600E. The effects of B-Raf kinase inhibition on biochemical and cellular characteristics were determined by incubating cultures of normal and malignant human cell lines with SB-590885 ( Fig. 2 ). Mitogen-activated protein kinase pathway inhibition was most apparent in established colorectal carcinoma (Colo205 and HT29) and melanoma cell lines (A375P, SKMEL28, and MALME-3M) that expressed B-RafV600E following brief incubation with SB-590885 ( Fig. 2A and B; data not shown), whereas normal cells (HFF, HMEC, and PREC) and malignant cells not expressing mutant B-Raf (HT-1080, HCT-116, and SKMEL2) showed intermediate sensitivity or resistance. Furthermore, prolonged incubation with SB-590885 preferentially decreased the proliferation of cells expressing oncogenic B-RafV600E ( Fig. 2B). As previously reported ( 17), the MEK inhibitor, CI-1040, also potently inhibited ERK signaling and proliferation in cells expressing BRAF mutations. However, in contrast to SB-590885, CI-1040 showed significant inhibition of ERK signaling and cellular proliferation in normal and malignant cells that do not express mutant B-Raf ( Fig. 2A and B). Cell cycle analysis revealed that a G1 phase cell cycle block was the predominant mechanism of decreased proliferation, with only minimal induction of apoptosis and a cytostatic growth arrest apparent in adherent cell cultures ( Fig. 2D; data not shown). A panel of normal melanocytes (newborn human melanocytes), early stage melanoma cells (RGP line WM-35), and primary melanoma cells (WM-NCI and WM-3434) also showed decreased phosphorylated ERK levels following treatment with CI-1040; and, as expected, only those cells harboring oncogenic B-RafV600E showed decreased ERK signaling following SB-590885 treatment ( Fig. 2C). Interestingly, both normal melanocytes and primary melanoma cells that express wild-type B-Raf (WM-NCI) paradoxically showed increased phosphorylated ERK levels following brief incubation with SB-590885 ( Fig. 2C). Importantly, the increased phosphorylated ERK levels in normal melanocytes did not correlate with any appreciable changes in the cell cycle profile, with partial G1 arrest noted at high concentrations of SB-590885 ( Fig. 2D). Therefore, SB-590885 may represent an improved therapeutic approach for the treatment of tumors expressing oncogenic B-Raf, as it spares more normal cells compared with MEK inhibition, and it is the first B-Raf kinase inhibitor described with selectivity towards tumor cells expressing oncogenic B-Raf.

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

SB-590885 preferentially inhibits ERK phosphorylation and proliferation in tumor cells expressing B-RafV600E. A, SB-590885 and CI-1040 were compared for their ability to inhibit ERK phosphorylation in normal and malignant cell lines at single 2 μmol/L concentrations following 60-minute incubations by Western blot analysis (D, DMSO; B, SB-590885; M, CI-1040). B, ERK phosphorylation and cellular proliferation are selectively inhibited in cells expressing oncogenic B-Raf. SB-590885 or CI-1040 was used at a concentration range of 1 nmol/L to 20 μmol/L, and a tabular summary of EC50 values determined from calculated ratios of phospho-ERK1/2 to total ERK1/2 and cellular proliferation is given. Data are representative of two or more runs and are shown with the BRAF and RAS status of each cell line for reference. C, effects of SB-590885 and CI-1040 on ERK phosphorylation in normal and malignant melanocytes at single 2 μmol/L concentrations following 60 minutes of incubation by Western blot analysis (D, DMSO; B, SB-590885; M, CI-1040). D, SB-590885 inhibits cell growth in normal melanocytes and adherent tumor cells bearing B-RafV600E predominantly by induction of a G1 arrest. The effect of incubation with 1 and 10 μmol/L of SB-590885 for 24 hours was examined in normal human melanocytes, A375P and Colo205 tumor cells. Staurosporine was used at 5 μmol/L for 6 hours as a positive control for the induction of apoptotic cell death (data not shown). Sub-G1 (solid columns), G1 phase (open columns), S phase (hatched columns), and G2-M phase (speckled columns); representative of two independent experiments.

SB-590885 inhibits cellular transformation and tumorigenesis. The promising results obtained with SB-590885 treatment of adherent cell cultures suggested that SB-590885 may have therapeutic relevance in three-dimensional preclinical model systems. Accordingly, the potential antineoplastic effects of SB-590885 were directly examined using several in vitro and in vivo assays of cellular transformation. Consistent with the previously reported requirement for B-RafV600E protein in melanoma cells ( 6, 7), inhibition of B-Raf kinase with SB-590885 decreased anchorage-independent growth of established melanoma cell lines in a BRAF mutant–selective manner over a dose range reflecting the potency of SB-590885 in two-dimensional culture ( Figs. 2B and 3A and B ). This result was extended to primary human melanoma cell spheroids to determine the effects of B-Raf inhibition on melanoma cell invasion and metabolic activity. As expected, when treated with SB-590885, spheroids comprised of melanoma cells expressing mutant B-Raf (WM-3434) showed decreased matrix invasion and metabolic activity as compared with spheroids comprised of HT-1080 fibrosarcoma cells or melanoma cells expressing wild-type B-Raf (WM-NCI; Fig. 3C), over a dose range reflecting the potency of SB-590885 in two-dimensional culture (data not shown). Finally, SB-590885 decreased tumorigenesis in murine xenografts established from mutant B-Raf-expressing A375P melanoma cells ( Fig. 3D). Although the effect on tumor growth was modest, it is consistent with the average degree of target inhibition observed in treated tumor tissues (∼50–80% reduction in pERK levels; data not shown). The regrowth of tumors after treatment cessation is also consistent with work in cell culture, as tissue analysis showed no overt evidence of increased apoptosis (data not shown). Therefore, SB-590885 showed important characteristics of a targeted therapeutic by decreasing the transformed and tumorigenic properties of malignant cells expressing oncogenic B-Raf.

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

Prolonged exposure to SB-590885 inhibits cellular transformation and proliferation in three-dimensional culture. A, SB-590885 inhibits anchorage-independent tumor colony growth selectively in tumor cell lines expressing B-RafV600E (light microscopy, 4×). B, anchorage-independent growth was quantified by counting individual colonies on a grid in triplicate and normalizing to vehicle controls (±SE) for each cell line. C, spheroid cultures of HT-1080, WM-NCI, and WM-3434 were treated with a range of SB-590885 for 72 hours, and assessed for invasiveness by light microscopy (top) and metabolic activity by using the “Live/Dead cell assay” as indicated by the green and red fluorescent images, respectively. D, decreased tumorigenicity of A375P cells following treatment with SB-590885. Points, tumor growth for cohorts of animals treated with vehicle alone (▪) or with SB-590885 (X); bars, ±SE. Mice were treated for 21 days, and following this, the mice treated with SB-590885 were observed for an additional 14 days.

Discussion

The generation of specific agents that neutralize required oncogenic signaling pathways in tumors is a major thrust of cancer drug discovery, and here, we describe the characteristics of the first specific B-Raf inhibitor, SB-590885. Both the potency and specificity of SB-590885 show attractive properties for further development of this and related compounds as therapeutics in cancer medicine.

SB-590885 inhibits B-Raf kinase enzymatic activity ∼100-fold more potently than the Raf/VEGFR inhibitor, BAY 43-9006. The structural basis of B-Raf inhibition by SB-590885 is due to stabilization of the active conformation of B-Raf by SB-590885, in contrast to stabilization of the inactive conformation of B-Raf by BAY 43-9006 ( 4). This alternative mechanism for kinase inhibition offers an additional method to target oncogenic B-Raf in patients with melanoma and the potential for overcoming generated resistance to inhibitors which bind to the inactive conformation of B-Raf, as has been shown for other oncogene-dependent malignancies ( 18).

Preferential inhibition by SB-590885 of biochemical signaling, proliferation, survival, and transformation was noted for human tumor cell lines expressing oncogenic B-Raf. The correlation between B-RafV600E expression and the inhibition of proliferation induced by SB-590885 was striking. Two tumor cell lines harboring mutant KRAS (HCT-116) or NRAS (SKMEL2) alleles exhibited moderate sensitivity to SB-590885 and the MEK inhibitor CI-1040, consistent with prior work ( 17). These data suggest a primary role for B-Raf as a downstream effector of Ras in these tumor cell lines. Importantly, little to no activity was observed for SB-590885 in three normal human cell types. The mechanism for the paradoxical increase in phosphorylated ERK in normal melanocytes and melanoma cells expressing wild-type B-Raf following treatment with SB-590885 ( Fig. 2C) is currently unknown, but may reflect alterations in biochemical feedback inhibition loops ( 19, 20). Nonetheless, this change in the phosphorylation state for ERK did not correlate with increased cell proliferation in either case. In contrast, the MEK inhibitor, CI-1040, inhibited ERK phosphorylation and cellular proliferation in a wide array of malignant and normal cells in the panel tested. The lack of selectivity between the different tumor cell types and primary cells following treatment with CI-1040 is consistent with the fact that MEK is an effector of multiple signaling pathways in mammalian cells ( 9, 19), and predicts that direct B-Raf inhibition will be advantageous in the treatment of patients suffering from oncogenic BRAF-driven tumors. Therefore, by exhibiting this “genetic therapeutic index” towards mutant B-Raf-expressing malignant cells, SB-590885 represents an additional compound that should be considered in the clinical development of small molecule therapeutics targeting oncogenic B-Raf.

Acknowledgments

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 regret our inability to include many primary references due to space constraints. We gratefully acknowledge the valuable scientific discussions with our colleagues at GlaxoSmithKline, especially the members of Oncology NERD for stimulating discussion on drug discovery; the support of the University of Pennsylvania-GlaxoSmithKline ADDI (L. Wang, D.A. Tuveson), Wistar NCI-Specialized Programs of Research Excellence (M. Herlyn, K. Smalley, D.A. Tuveson), and the Samuel Waxman Cancer Research Foundation (D.A. Tuveson). D.A. Tuveson is a Rita Allen Foundation Scholar. We also thank Bonnie Orr and Gary Stelman for support in the mouse pharmacokinetic analysis.

Footnotes

  • ↵12 E. May, C. Rominger, M. Schaber, and P. Huang, unpublished data.

  • Received July 11, 2006.
  • Revision received October 4, 2006.
  • Accepted October 10, 2006.
  • ©2006 American Association for Cancer Research.

References

  1. ↵
    Zhang Z, Li M, Rayburn ER, Hill DL, Zhang R, Wang H. Oncogenes as novel targets for cancer therapy (part IV): regulators of the cell cycle and apoptosis. Am J Pharmacogenomics 2005; 5: 397–407.
    OpenUrlCrossRefPubMed
  2. ↵
    von Eyben FE. Epidermal growth factor receptor inhibition and non-small cell lung cancer. Crit Rev Clin Lab Sci 2006; 43: 291–323.
    OpenUrlCrossRefPubMed
  3. ↵
    Wooster R, Futreal AP, Stratton MR. Sequencing analysis of BRAF mutations in human cancers. Methods Enzymol 2005; 407: 218–24.
    OpenUrlPubMed
  4. ↵
    Wan PT, Garnett MJ, Roe SM, et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell 2004; 116: 855–67.
    OpenUrlCrossRefPubMed
  5. ↵
    Karasarides M, Chiloeches A, Hayward R, et al. B-RAF is a therapeutic target in melanoma. Oncogene 2004; 23: 6292–8.
    OpenUrlCrossRefPubMed
  6. ↵
    Hingorani SR, Jacobetz MA, Robertson GP, Herlyn M, Tuveson DA. Suppression of BRAF(V599E) in human melanoma abrogates transformation. Cancer Res 2003; 63: 5198–202.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Sumimoto H, Miyagishi M, Miyoshi H, et al. Inhibition of growth and invasive ability of melanoma by inactivation of mutated BRAF with lentivirus-mediated RNA interference. Oncogene 2004; 23: 6031–9.
    OpenUrlCrossRefPubMed
  8. ↵
    Takle AK, Brown MJ, Davies S, et al. The identification of potent and selective imidazole-based inhibitors of B-Raf kinase. Bioorg Med Chem Lett 2006; 16: 378–81.
    OpenUrlCrossRefPubMed
  9. ↵
    Sebolt-Leopold JS, Dudley DT, Herrera R, et al. Blockade of the MAP kinase pathway suppresses growth of colon tumors in vivo. Nat Med 1999; 5: 810–6.
    OpenUrlCrossRefPubMed
  10. ↵
    Leslie AG. The integration of macromolecular diffraction data. Acta Crystallogr D Biol Crystallogr 2006; 62: 48–57.
    OpenUrlCrossRefPubMed
  11. ↵
    Vagin A, Teplyakov A. An approach to multi-copy search in molecular replacement. Acta Crystallogr D Biol Crystallogr 2000; 56: 1622–4.
    OpenUrlCrossRefPubMed
  12. ↵
    Murshudov GN, Vagin AA, Dodson EJ. Refinement of macromolecular structures by the maximum-likelihood method. Acta Crystallogr D Biol Crystallogr 1997; 53: 240–5.
    OpenUrlCrossRefPubMed
  13. ↵
    Chen H, Kovar J, Sissons S, et al. A cell-based immunocytochemical assay for monitoring kinase signaling pathways and drug efficacy. Anal Biochem 2005; 338: 136–42.
    OpenUrlCrossRefPubMed
  14. ↵
    Smalley KS, Haass NK, Brafford PA, Lioni M, Flaherty KT, Herlyn M. Multiple signaling pathways must be targeted to overcome drug resistance in cell lines derived from melanoma metastases. Mol Cancer Ther 2006; 5: 1136–44.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    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.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science 2002; 298: 1912–34.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Solit DB, Garraway LA, Pratilas CA, et al. BRAF mutation predicts sensitivity to MEK inhibition. Nature 2006; 439: 358–62.
    OpenUrlCrossRefPubMed
  18. ↵
    Burgess MR, Skaggs BJ, Shah NP, Lee FY, Sawyers CL. Comparative analysis of two clinically active BCR-ABL kinase inhibitors reveals the role of conformation-specific binding in resistance. Proc Natl Acad Sci U S A 2005; 102: 3395–400.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Hagemann C, Blank JL. The ups and downs of MEK kinase interactions. Cell Signal 2001; 13: 863–75.
    OpenUrlCrossRefPubMed
  20. ↵
    Ueki K, Matsuda S, Tobe K, et al. Feedback regulation of mitogen-activated protein kinase kinase kinase activity of c-Raf-1 by insulin and phorbol ester stimulation. J Biol Chem 1994; 269: 15756–61.
    OpenUrlAbstract/FREE Full Text
View Abstract
PreviousNext
Back to top
Cancer Research: 66 (23)
December 2006
Volume 66, Issue 23
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by Author

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.
Demonstration of a Genetic Therapeutic Index for Tumors Expressing Oncogenic BRAF by the Kinase Inhibitor SB-590885
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
Citation Tools
Demonstration of a Genetic Therapeutic Index for Tumors Expressing Oncogenic BRAF by the Kinase Inhibitor SB-590885
Alastair J. King, Denis R. Patrick, Roberta S. Batorsky, Maureen L. Ho, Hieu T. Do, Shu Yun Zhang, Rakesh Kumar, David W. Rusnak, Andrew K. Takle, David M. Wilson, Erin Hugger, Lifu Wang, Florian Karreth, Julie C. Lougheed, Jae Lee, David Chau, Thomas J. Stout, Earl W. May, Cynthia M. Rominger, Michael D. Schaber, Lusong Luo, Ami S. Lakdawala, Jerry L. Adams, Rooja G. Contractor, Keiran S.M. Smalley, Meenhard Herlyn, Michael M. Morrissey, David A. Tuveson and Pearl S. Huang
Cancer Res December 1 2006 (66) (23) 11100-11105; DOI: 10.1158/0008-5472.CAN-06-2554

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Demonstration of a Genetic Therapeutic Index for Tumors Expressing Oncogenic BRAF by the Kinase Inhibitor SB-590885
Alastair J. King, Denis R. Patrick, Roberta S. Batorsky, Maureen L. Ho, Hieu T. Do, Shu Yun Zhang, Rakesh Kumar, David W. Rusnak, Andrew K. Takle, David M. Wilson, Erin Hugger, Lifu Wang, Florian Karreth, Julie C. Lougheed, Jae Lee, David Chau, Thomas J. Stout, Earl W. May, Cynthia M. Rominger, Michael D. Schaber, Lusong Luo, Ami S. Lakdawala, Jerry L. Adams, Rooja G. Contractor, Keiran S.M. Smalley, Meenhard Herlyn, Michael M. Morrissey, David A. Tuveson and Pearl S. Huang
Cancer Res December 1 2006 (66) (23) 11100-11105; DOI: 10.1158/0008-5472.CAN-06-2554
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

  • RAGE/PR3 Interaction Mediates Bone Metastasis
  • MCPyV Small T Ag Initiates Mouse Merkel Cell Carcinoma
  • Colon Cancer Stemness Is Regulated through LUST
Show more Priority Reports
  • Home
  • Alerts
  • Feedback
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

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

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians
  • Reviewers

About Cancer Research

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

Copyright © 2018 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