Bcl-2/Bcl-xL Inhibition Increases the Efficacy of MEK Inhibition Alone and in Combination with PI3 Kinase Inhibition in Lung and Pancreatic Tumor Models

Although mitogen-activated protein (MAP)–extracellular signal-regulated kinase (ERK) kinase (MEK) inhibition is predicted to cause cell death by stabilization of the proapoptotic BH3-only protein BIM, the induction of apoptosis is often modest. To determine if addition of a Bcl-2 family inhibitor could increase the efficacy of a MEK inhibitor, we evaluated a panel of 53 non–small cell lung cancer and pancreatic cancer cell lines with the combination of navitoclax (ABT-263), a Bcl-2/Bcl-xL (BCL2/BCL2L1) antagonist, and a novel MAP kinase (MEK) inhibitor, G-963. The combination is synergistic in the majority of lines, with an enrichment of cell lines harboring KRAS mutations in the high synergy group. Cells exposed to G-963 arrest in G1 and a small fraction undergo apoptosis. The addition of navitoclax to G-963 does not alter the kinetics of cell-cycle arrest, but greatly increases the percentage of cells that undergo apoptosis. The G-963/navitoclax combination was more effective than either single agent in the KRAS mutant H2122 xenograft model; BIM stabilization and PARP cleavage were observed in tumors, consistent with the mechanism of action observed in cell culture. Addition of the phosphatidylinositol 3-kinase (PI3K, PIK3CA) inhibitor GDC-0941 to this treatment combination increases cell killing compared with double- or single-agent treatment. Taken together, these data suggest the efficacy of agents that target the MAPK and PI3K pathways can be improved by combination with a Bcl-2 family inhibitor. Mol Cancer Ther; 12(6); 853–64. ©2013 AACR.

[1]  S. Cook,et al.  The BH3 mimetic ABT-263 synergizes with the MEK1/2 inhibitor selumetinib/AZD6244 to promote BIM-dependent tumour cell death and inhibit acquired resistance. , 2013, The Biochemical journal.

[2]  Travis J Cohoon,et al.  Synthetic lethal interaction of combined BCL-XL and MEK inhibition promotes tumor regressions in KRAS mutant cancer models. , 2013, Cancer cell.

[3]  G. Giaccone,et al.  Phase II Study of Single-Agent Navitoclax (ABT-263) and Biomarker Correlates in Patients with Relapsed Small Cell Lung Cancer , 2012, Clinical Cancer Research.

[4]  N. Normanno,et al.  The RAS/RAF/MEK/ERK and the PI3K/AKT signalling pathways: role in cancer pathogenesis and implications for therapeutic approaches , 2012, Expert opinion on therapeutic targets.

[5]  T. Gilmer,et al.  Combinations of BRAF, MEK, and PI3K/mTOR Inhibitors Overcome Acquired Resistance to the BRAF Inhibitor GSK2118436 Dabrafenib, Mediated by NRAS or MEK Mutations , 2012, Molecular Cancer Therapeutics.

[6]  Gordon B Mills,et al.  Inhibition of PI3K/mTOR leads to adaptive resistance in matrix-attached cancer cells. , 2012, Cancer cell.

[7]  Hao Xiong,et al.  Substantial susceptibility of chronic lymphocytic leukemia to BCL2 inhibition: results of a phase I study of navitoclax in patients with relapsed or refractory disease. , 2012, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[8]  F. Peale,et al.  Navitoclax (ABT-263) Reduces Bcl-xL–Mediated Chemoresistance in Ovarian Cancer Models , 2012, Molecular Cancer Therapeutics.

[9]  B. Kholodenko,et al.  Cross-talk between mitogenic Ras/MAPK and survival PI3K/Akt pathways: a fine balance. , 2012, Biochemical Society transactions.

[10]  A. Tolcher,et al.  The Clinical Effect of the Dual-Targeting Strategy Involving PI3K/AKT/mTOR and RAS/MEK/ERK Pathways in Patients with Advanced Cancer , 2012, Clinical Cancer Research.

[11]  M. Konopleva,et al.  MEK inhibition enhances ABT-737-induced leukemia cell apoptosis via prevention of ERK-activated MCL-1 induction and modulation of MCL-1/BIM complex , 2012, Leukemia.

[12]  Mark Merchant,et al.  Intermittent administration of MEK inhibitor GDC-0973 plus PI3K inhibitor GDC-0941 triggers robust apoptosis and tumor growth inhibition. , 2012, Cancer research.

[13]  J. Holbrook,et al.  Comprehensive Predictive Biomarker Analysis for MEK Inhibitor GSK1120212 , 2011, Molecular Cancer Therapeutics.

[14]  E. Plise,et al.  Preclinical pharmacokinetics of the novel PI3K inhibitor GDC-0941 and prediction of its pharmacokinetics and efficacy in human , 2011 .

[15]  G. Tzivion,et al.  FoxO transcription factors; Regulation by AKT and 14-3-3 proteins. , 2011, Biochimica et biophysica acta.

[16]  Qiaojun He,et al.  GDC-0941 sensitizes breast cancer to ABT-737 in vitro and in vivo through promoting the degradation of Mcl-1. , 2011, Cancer letters.

[17]  C. Tse,et al.  The Bcl-2/Bcl-XL/Bcl-w Inhibitor, Navitoclax, Enhances the Activity of Chemotherapeutic Agents In Vitro and In Vivo , 2011, Molecular Cancer Therapeutics.

[18]  M. Deininger,et al.  Advances in the treatment of chronic myeloid leukemia , 2011, BMC medicine.

[19]  T. Mitchison,et al.  Navitoclax (ABT-263) accelerates apoptosis during drug-induced mitotic arrest by antagonizing Bcl-xL. , 2011, Cancer research.

[20]  Rodney J Hicks,et al.  In Vivo Activity of Combined PI3K/mTOR and MEK Inhibition in a KrasG12D;Pten Deletion Mouse Model of Ovarian Cancer , 2011, Molecular Cancer Therapeutics.

[21]  J. Engelman,et al.  Potential Therapeutic Strategies to Overcome Acquired Resistance to BRAF or MEK Inhibitors in BRAF Mutant Cancers , 2011, Oncotarget.

[22]  David B Solit,et al.  Resistance to MEK Inhibitors: Should We Co-Target Upstream? , 2011, Science Signaling.

[23]  C. Rudin,et al.  Phase I study of Navitoclax (ABT-263), a novel Bcl-2 family inhibitor, in patients with small-cell lung cancer and other solid tumors. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[24]  J. Zha,et al.  Navitoclax Enhances the Efficacy of Taxanes in Non–Small Cell Lung Cancer Models , 2011, Clinical Cancer Research.

[25]  E. Plise,et al.  Preclinical pharmacokinetics of the novel PI3K inhibitor GDC-0941 and prediction of its pharmacokinetics and efficacy in human. , 2011, Xenobiotica; the fate of foreign compounds in biological systems.

[26]  W. Wilson,et al.  Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity. , 2010, The Lancet. Oncology.

[27]  Aik Choon Tan,et al.  Identification of Predictive Markers of Response to the MEK1/2 Inhibitor Selumetinib (AZD6244) in K-ras–Mutated Colorectal Cancer , 2010, Molecular Cancer Therapeutics.

[28]  Deepak Sampath,et al.  Pharmacokinetic-Pharmacodynamic Modeling of Tumor Growth Inhibition and Biomarker Modulation by the Novel Phosphatidylinositol 3-Kinase Inhibitor GDC-0941 , 2010, Drug Metabolism and Disposition.

[29]  D. Elashoff,et al.  Identification of Common Predictive Markers of In vitro Response to the Mek Inhibitor Selumetinib (AZD6244; ARRY-142886) in Human Breast Cancer and Non–Small Cell Lung Cancer Cell Lines , 2010, Molecular Cancer Therapeutics.

[30]  B. Taylor,et al.  Transcriptional pathway signatures predict MEK addiction and response to selumetinib (AZD6244). , 2010, Cancer research.

[31]  Kwok-Kin Wong,et al.  Targeting the PI3K signaling pathway in cancer. , 2010, Current opinion in genetics & development.

[32]  Saul H. Rosenberg,et al.  The Bcl-2 inhibitor ABT-263 enhances the response of multiple chemotherapeutic regimens in hematologic tumors in vivo , 2010, Cancer Chemotherapy and Pharmacology.

[33]  C. Sander,et al.  V600EBRAF is associated with disabled feedback inhibition of RAF–MEK signaling and elevated transcriptional output of the pathway , 2009, Proceedings of the National Academy of Sciences.

[34]  A. Letai,et al.  Control of mitochondrial apoptosis by the Bcl-2 family , 2009, Journal of Cell Science.

[35]  Michael S. Cohen,et al.  betaTrCP- and Rsk1/2-mediated degradation of BimEL inhibits apoptosis. , 2009, Molecular cell.

[36]  Michael S. Cohen,et al.  TrCP-and Rsk 1 / 2-Mediated Degradation of BimEL Inhibits Apoptosis , 2009 .

[37]  A. Strasser,et al.  Treatment of B-RAF mutant human tumor cells with a MEK inhibitor requires Bim and is enhanced by a BH3 mimetic. , 2008, The Journal of clinical investigation.

[38]  Corey Nislow,et al.  Combination chemical genetics. , 2008, Nature chemical biology.

[39]  Gary Box,et al.  The identification of 2-(1H-indazol-4-yl)-6-(4-methanesulfonyl-piperazin-1-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidine (GDC-0941) as a potent, selective, orally bioavailable inhibitor of class I PI3 kinase for the treatment of cancer . , 2008, Journal of medicinal chemistry.

[40]  C. Tse,et al.  ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor. , 2008, Cancer research.

[41]  Alexei Degterev,et al.  Identification of RIP1 kinase as a specific cellular target of necrostatins. , 2008, Nature chemical biology.

[42]  G. Mills,et al.  ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation , 2008, Nature Cell Biology.

[43]  A. Brunet,et al.  FOXO transcription factors , 2007, Current Biology.

[44]  Miki Ebisuya,et al.  Continuous ERK Activation Downregulates Antiproliferative Genes throughout G1 Phase to Allow Cell-Cycle Progression , 2006, Current Biology.

[45]  Neal Rosen,et al.  The BAD protein integrates survival signaling by EGFR/MAPK and PI3K/Akt kinase pathways in PTEN-deficient tumor cells. , 2005, Cancer cell.

[46]  R. Craig,et al.  MCL1 is phosphorylated in the PEST region and stabilized upon ERK activation in viable cells, and at additional sites with cytotoxic okadaic acid or taxol , 2004, Oncogene.

[47]  V. Carey,et al.  Mixed-Effects Models in S and S-Plus , 2001 .

[48]  D. Bates,et al.  Mixed-Effects Models in S and S-PLUS , 2001 .

[49]  Andrius Kazlauskas,et al.  Growth-factor-dependent mitogenesis requires two distinct phases of signalling , 2001, Nature Cell Biology.

[50]  A. Brunet,et al.  Nuclear translocation of p42/p44 mitogen‐activated protein kinase is required for growth factor‐induced gene expression and cell cycle entry , 1999, The EMBO journal.