Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma

Gideon Bollag, Peter Hirth, James Tsai, Jiazhong Zhang, Prabha N Ibrahim, Hanna Cho, Wayne Spevak, Chao Zhang, Ying Zhang, Gaston Habets, Elizabeth A Burton, Bernice Wong, Garson Tsang, Brian L West, Ben Powell, Rafe Shellooe, Adhirai Marimuthu, Hoa Nguyen, Kam Y J Zhang, Dean R Artis, Joseph Schlessinger, Fei Su, Brian Higgins, Raman Iyer, Kurt D'Andrea, Astrid Koehler, Michael Stumm, Paul S Lin, Richard J Lee, Joseph Grippo, Igor Puzanov, Kevin B Kim, Antoni Ribas, Grant A McArthur, Jeffrey A Sosman, Paul B Chapman, Keith T Flaherty, Xiaowei Xu, Katherine L Nathanson, Keith Nolop, Gideon Bollag, Peter Hirth, James Tsai, Jiazhong Zhang, Prabha N Ibrahim, Hanna Cho, Wayne Spevak, Chao Zhang, Ying Zhang, Gaston Habets, Elizabeth A Burton, Bernice Wong, Garson Tsang, Brian L West, Ben Powell, Rafe Shellooe, Adhirai Marimuthu, Hoa Nguyen, Kam Y J Zhang, Dean R Artis, Joseph Schlessinger, Fei Su, Brian Higgins, Raman Iyer, Kurt D'Andrea, Astrid Koehler, Michael Stumm, Paul S Lin, Richard J Lee, Joseph Grippo, Igor Puzanov, Kevin B Kim, Antoni Ribas, Grant A McArthur, Jeffrey A Sosman, Paul B Chapman, Keith T Flaherty, Xiaowei Xu, Katherine L Nathanson, Keith Nolop

Abstract

B-RAF is the most frequently mutated protein kinase in human cancers. The finding that oncogenic mutations in BRAF are common in melanoma, followed by the demonstration that these tumours are dependent on the RAF/MEK/ERK pathway, offered hope that inhibition of B-RAF kinase activity could benefit melanoma patients. Herein, we describe the structure-guided discovery of PLX4032 (RG7204), a potent inhibitor of oncogenic B-RAF kinase activity. Preclinical experiments demonstrated that PLX4032 selectively blocked the RAF/MEK/ERK pathway in BRAF mutant cells and caused regression of BRAF mutant xenografts. Toxicology studies confirmed a wide safety margin consistent with the high degree of selectivity, enabling Phase 1 clinical trials using a crystalline formulation of PLX4032 (ref. 5). In a subset of melanoma patients, pathway inhibition was monitored in paired biopsy specimens collected before treatment initiation and following two weeks of treatment. This analysis revealed substantial inhibition of ERK phosphorylation, yet clinical evaluation did not show tumour regressions. At higher drug exposures afforded by a new amorphous drug formulation, greater than 80% inhibition of ERK phosphorylation in the tumours of patients correlated with clinical response. Indeed, the Phase 1 clinical data revealed a remarkably high 81% response rate in metastatic melanoma patients treated at an oral dose of 960 mg twice daily. These data demonstrate that BRAF-mutant melanomas are highly dependent on B-RAF kinase activity.

Figures

Fig 1. Three-dimensional structure of PLX4032 binding…
Fig 1. Three-dimensional structure of PLX4032 binding to B-RAFV600E
A: The chemical structure of PLX4032. B: Structure highlights the interactions of azaindole with the kinase hinge and the sulfonamide with the DFG loop, with F595 rendered in balls and other key protein residues shown as sticks. C: The structure of the asymmetric dimer of B-RAFV600E is shown with the PLX4032-protomer bound to PLX4032 colored yellow (consistent with panel B). The surface outline of the other protomer (blue) is shown lightly shaded. Highlighted residues are R509 to reflect its role in anchoring the dimer and F595 to show that both protomers are in the DFG-in state. The αc-helix shown in magenta is overlaid on the PLX4032-bound protomer to show its typical configuration in an unoccupied protomer; the binding of PLX4032 causes a shift of the αc-helix as noted by the arrow. D. Magnified view of the salt bridge between Lys-507 and Glu-600 that helps prevent compound binding to the apo protomer.
Fig 2. Effect of PLX4032 on COLO205…
Fig 2. Effect of PLX4032 on COLO205 xenograft tumor growth
Tumor volume measurements of mice treated by oral gavage with the indicated doses of PLX4032 or vehicle (n=10 for all groups, error bars indicate standard error) are shown. A. Administration in conventional formulation occurred daily. Exposures measured on day 7 are shown. At the 6, 20 and 20 BID doses, 1/10, 1/10, and 8/10 animals achieved CR, respectively. B. Administration in the MBP formulation occurred twice daily. At the 25 mg/kg BID dose (blue), 7/10 animals achieved CR and 3/10 animals achieved PR; at the 75 mg/kg BID dose (red), all animals achieved CR.
Fig 3. Semi-quantitative immunohistochemistry in paired tumor…
Fig 3. Semi-quantitative immunohistochemistry in paired tumor biopsies
Matched baseline and day 15 tumor samples are at the same magnification; the measurement bar is 70 μm. A. Representative IHC for Ki67 and pERK staining is shown for patient 12. B. Representative IHC for Ki67, pERK and H&E staining is shown for patient 42. The arrow indicates tumor breakdown with macrophages engulfing the released melanin in the day 15 sample. C. Summary graph showing correlation of reduction in cytoplasmic pERK with tumor responses (data from Supplementary Table 2). D. Summary graph indicating weak correlation of reduction in nuclear pERK with tumor responses.
Fig 4. Representative PET scans for patients…
Fig 4. Representative PET scans for patients taken pre-dose and following 2 weeks of dosing with PLX4032
Each of these image pairs demonstrates significant reduction in FDG uptake following PLX4032 treatment. Note that tumor regressions were later documented for each of these patients: best responses were 70% for patient 45, 70% for patient 59, 68% for patient 61 and 37% for patient 69.

References

    1. Greenman C, et al. Patterns of somatic mutation in human cancer genomes. Nature. 2007;446:153–158.
    1. Davies H, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–954.
    1. Solit DB, et al. BRAF mutation predicts sensitivity to MEK inhibition. Nature. 2006;439:358–362.
    1. Yang H, et al. RG7204 (PLX4032), a selective BRAFV600E inhibitor, displays potent antitumor activity in preclinical melanoma models. Cancer Res. 2010;70:5518–5527.
    1. Flaherty K, et al. Inhibition of Mutated, Activated BRAF in Metastatic Melanoma. N Engl J Med. 2010;363:809–819.
    1. Tsai J, et al. Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity. Proc Natl Acad Sci U S A. 2008;105:3041–3046.
    1. Hatzivassiliou G, et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature. 2010
    1. Koch WH. Technology platforms for pharmacogenomic diagnostic assays. Nature reviews. 2004;3:749–761.
    1. Rubinstein JC, et al. Incidence of the V600K mutation among melanoma patients with BRAF mutations, and potential therapeutic response to the specific BRAF inhibitor PLX4032. J Transl Med. 2010;8:67.
    1. Esteve-Puig R, Canals F, Colome N, Merlino G, Recio JA. Uncoupling of the LKB1-AMPKalpha energy sensor pathway by growth factors and oncogenic BRAF. PloS one. 2009;4:e4771.
    1. Zheng B, et al. Oncogenic B-RAF negatively regulates the tumor suppressor LKB1 to promote melanoma cell proliferation. Mol Cell. 2009;33:237–247.
    1. Søndergaard JN, et al. Differential Sensitivity of Melanoma Cell Lines with BRAF V600E Mutation to the Specific B-Raf Inhibitor PLX4032. J Transl Med. 2010;8:39.
    1. Arnault JP, et al. Keratoacanthomas and squamous cell carcinomas in patients receiving sorafenib. J Clin Oncol. 2009;27:e59–61.
    1. Dubauskas Z, et al. Cutaneous squamous cell carcinoma and inflammation of actinic keratoses associated with sorafenib. Clin Genitourin Cancer. 2009;7:20–23.
    1. Kong HH, et al. Keratoacanthomas associated with sorafenib therapy. J Am Acad Dermatol. 2007;56:171–172.
    1. Schwartz GK, et al. A phase I study of XL281, a selective oral RAF kinase inhibitor, in patients (Pts) with advanced solid tumors. J Clin Oncol. 2009;27:15s.
    1. Kefford R, et al. Phase I/II study of GSK2118436, a selective inhibitor of oncogenic mutant BRAF kinase, in patients with metastatic melanoma and other solid tumors. J Clin Oncol. 2010;28:8503.
    1. Heidorn SJ, et al. Kinase-Dead BRAF and Oncogenic RAS Cooperate to Drive Tumor Progression through CRAF. Cell. 2010;140:209–221.
    1. Poulikakos P, Zhang C, Bollag G, Shokat K, Rosen N. RAF inhibitors transactivate RAF dimers and ERK signaling in cells with wild-type BRAF. Nature. 2010;464:427–430.
    1. Courtois-Cox S, et al. A negative feedback signaling network underlies oncogene-induced senescence. Cancer Cell. 2006;10:459–472.
    1. Dougherty MK, et al. Regulation of Raf-1 by direct feedback phosphorylation. Mol Cell. 2005;17:215–224.
    1. Hall-Jackson CA, et al. Paradoxical activation of Raf by a novel Raf inhibitor. Chem Biol. 1999;6:559–568.
    1. Rajakulendran T, Sahmi M, Lefrancois M, Sicheri F, Therrien M. A dimerization-dependent mechanism drives RAF catalytic activation. Nature. 2009;461:542–545.
    1. Pratilas CA, et al. (V600E)BRAF is associated with disabled feedback inhibition of RAF-MEK signaling and elevated transcriptional output of the pathway. Proc Natl Acad Sci U S A. 2009;106:4519–4524.
    1. Kolch W. Coordinating ERK/MAPK signalling through scaffolds and inhibitors. Nat Rev Mol Cell Biol. 2005;6:827–837.
    1. Pollock PM, et al. High frequency of BRAF mutations in nevi. Nat Genet. 2003;33:19–20.
    1. Michaloglou C, et al. BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature. 2005;436:720–724.
    1. Korn EL, et al. Meta-analysis of phase II cooperative group trials in metastatic stage IV melanoma to determine progression-free and overall survival benchmarks for future phase II trials. J Clin Oncol. 2008;26:527–534.

Source: PubMed

3
구독하다