Summary
Purpose We investigated the combination of the MEK inhibitor, cobimetinib, and the pan-PI3K inhibitor, pictilisib, in an open-label, phase Ib study. Experimental Design Patients with advanced solid tumors were enrolled in 3 dose escalation schedules: (1) both agents once-daily for 21-days-on 7-days-off (“21/7”); (2) intermittent cobimetinib and 21/7 pictilisib (“intermittent”); or (3) both agents once-daily for 7-days-on 7-days-off (“7/7”). Starting doses for the 21/7, intermittent, and 7/7 schedules were 20/80, 100/130, and 40/130 mg of cobimetinib/pictilisib, respectively. Nine indication-specific expansion cohorts interrogated the recommended phase II dose and schedule. Results Of 178 enrollees (dose escalation: n = 98), 177 patients were dosed. The maximum tolerated doses for cobimetinib/pictilisib (mg) were 40/100, 125/180, and not reached, for the 21/7, intermittent, and 7/7 schedules, respectively. Six dose-limiting toxicities included grade 3 (G3) elevated lipase, G4 elevated creatine phosphokinase, and G3 events including fatigue concurrent with a serious adverse event (SAE) of diarrhea, decreased appetite, and SAEs of hypersensitivity and dehydration. Common drug-related adverse events included nausea, fatigue, vomiting, decreased appetite, dysgeusia, rash, and stomatitis. Pharmacokinetic parameters of the drugs used in combination were unaltered compared to monotherapy exposures. Confirmed partial responses were observed in patients with BRAF-mutant melanoma (n = 1) and KRAS-mutant endometrioid adenocarcinoma (n = 1). Eighteen patients remained on study ≥6 months. Biomarker data established successful blockade of MAP kinase (MAPK) and PI3K pathways. The metabolic response rate documented by FDG-PET was similar to that observed with cobimetinib monotherapy. Conclusions Cobimetinib and pictilisib combination therapy in patients with solid tumors had limited tolerability and efficacy.





Similar content being viewed by others
Explore related subjects
Discover the latest articles and news from researchers in related subjects, suggested using machine learning.References
McCubrey JA, Steelman LS, Kempf CR, Chappell WH, Abrams SL, Stivala F, Malaponte G, Nicoletti F, Libra M, Bäsecke J, Maksimovic-Ivanic D, Mijatovic S, Montalto G, Cervello M, Cocco L, Martelli AM (2011) Therapeutic resistance resulting from mutations in Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR signaling pathways. Journal of Cellular Physiology 226(11):2762–2781. https://doi.org/10.1002/jcp.22647
McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Montalto G, Cervello M, Nicoletti F, Fagone P, Malaponte G, Mazzarino MC, Candido S, Libra M, Basecke J, Mijatovic S, Maksimovic-Ivanic D, Milella M, Tafuri A, Cocco L, Evangelisti C, Chiarini F, Martelli AM (2012) Mutations and deregulation of Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR cascades which alter therapy response. Oncotarget 3(9):954–987. https://doi.org/10.18632/oncotarget.652
Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix-Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JW, Leung SY, Yuen ST, Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR, Futreal PA (2002) Mutations of the BRAF gene in human cancer. Nature 417(6892):949–954. https://doi.org/10.1038/nature00766
Saldanha G, Potter L, Daforno P, Pringle JH (2006) Cutaneous melanoma subtypes show different BRAF and NRAS mutation frequencies. Clin Cancer Res 12(15):4499–4505. https://doi.org/10.1158/1078-0432.ccr-05-2447
Hatzivassiliou G, Haling JR, Chen H, Song K, Price S, Heald R, Hewitt JF, Zak M, Peck A, Orr C, Merchant M, Hoeflich KP, Chan J, Luoh SM, Anderson DJ, Ludlam MJ, Wiesmann C, Ultsch M, Friedman LS, Malek S, Belvin M (2013) Mechanism of MEK inhibition determines efficacy in mutant KRAS- versus BRAF-driven cancers. Nature 501(7466):232–236. https://doi.org/10.1038/nature12441
Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, Yan H, Gazdar A, Powell SM, Riggins GJ, Willson JK, Markowitz S, Kinzler KW, Vogelstein B, Velculescu VE (2004) High frequency of mutations of the PIK3CA gene in human cancers. Science 304(5670):554. https://doi.org/10.1126/science.1096502
Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, Puc J, Miliaresis C, Rodgers L, McCombie R, Bigner SH, Giovanella BC, Ittmann M, Tycko B, Hibshoosh H, Wigler MH, Parsons R (1997) PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 275(5308):1943–1947
Aoki M, Batista O, Bellacosa A, Tsichlis P, Vogt PK (1998) The akt kinase: molecular determinants of oncogenicity. Proc Natl Acad Sci U S A 95(25):14950–14,955
Wee S, Jagani Z, Xiang KX, Loo A, Dorsch M, Yao Y-M, Sellers WR, Lengauer C, Stegmeier F (2009) PI3K Pathway Activation Mediates Resistance to MEK Inhibitors in KRAS Mutant Cancers. Cancer Res 69(10):4286–4293. https://doi.org/10.1158/0008-5472.can-08-4765
Turke AB, Song Y, Costa C, Cook R, Arteaga CL, Asara JM, Engelman JA (2012) MEK Inhibition Leads to PI3K/AKT Activation by Relieving a Negative Feedback on ERBB Receptors. Cancer Res 72(13):3228–3237. https://doi.org/10.1158/0008-5472.can-11-3747
Mohan S, Vander Broek R, Shah S, Eytan DF, Pierce ML, Carlson SG, Coupar JF, Zhang J, Cheng H, Chen Z, Van Waes C (2015) MEK Inhibitor PD-0325901 Overcomes Resistance to PI3K/mTOR Inhibitor PF-5212384 and Potentiates Antitumor Effects in Human Head and Neck Squamous Cell Carcinoma. Clin Cancer Res 21(17):3946–3956. https://doi.org/10.1158/1078-0432.ccr-14-3377
Wong H, Vernillet L, Peterson A, Ware JA, Lee L, Martini JF, Yu P, Li C, Del Rosario G, Choo EF, Hoeflich KP, Shi Y, Aftab BT, Aoyama R, Lam ST, Belvin M, Prescott J (2012) Bridging the gap between preclinical and clinical studies using pharmacokinetic-pharmacodynamic modeling: an analysis of GDC-0973, a MEK inhibitor. Clin Cancer Res 18(11):3090–3099. https://doi.org/10.1158/1078-0432.ccr-12-0445
Deng Y, Musib L, Choo E, Chapple M, Burke S, Johnson J, Eppler S, Dean B (2014) Determination of cobimetinib in human plasma using protein precipitation extraction and high-performance liquid chromatography coupled to mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 972:117–123. https://doi.org/10.1016/j.jchromb.2014.09.034
Salphati L, Wong H, Belvin M, Bradford D, Edgar KA, Prior WW, Sampath D, Wallin JJ (2010) Pharmacokinetic-pharmacodynamic modeling of tumor growth inhibition and biomarker modulation by the novel phosphatidylinositol 3-kinase inhibitor GDC-0941. Drug Metab Dispos 38(9):1436–1442. https://doi.org/10.1124/dmd.110.032912
Raynaud FI, Eccles SA, Patel S, Alix S, Box G, Chuckowree I, Folkes A, Gowan S, De Haven Brandon A, Di Stefano F, Hayes A, Henley AT, Lensun L, Pergl-Wilson G, Robson A, Saghir N, Zhyvoloup A, McDonald E, Sheldrake P, Shuttleworth S, Valenti M, Wan NC, Clarke PA, Workman P (2009) Biological properties of potent inhibitors of class I phosphatidylinositide 3-kinases: from PI-103 through PI-540, PI-620 to the oral agent GDC-0941. Mol Cancer Ther 8(7):1725–1738. https://doi.org/10.1158/1535-7163.mct-08-1200
Hoeflich KP, Merchant M, Orr C, Chan J, Den Otter D, Berry L, Kasman I, Koeppen H, Rice K, Yang NY, Engst S, Johnston S, Friedman LS, Belvin M (2012) Intermittent administration of MEK inhibitor GDC-0973 plus PI3K inhibitor GDC-0941 triggers robust apoptosis and tumor growth inhibition. Cancer Res 72(1):210–219. https://doi.org/10.1158/0008-5472.can-11-1515
Choo EF, Ng CM, Berry L, Belvin M, Lewin-Koh N, Merchant M, Salphati L (2013) PK-PD modeling of combination efficacy effect from administration of the MEK inhibitor GDC-0973 and PI3K inhibitor GDC-0941 in A2058 xenografts. Cancer Chemother Pharmacol 71(1):133–143. https://doi.org/10.1007/s00280-012-1988-6
Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, Verweij J, Van Glabbeke M, van Oosterom AT, Christian MC, Gwyther SG (2000) New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 92(3):205–216
Spoerke JM, O’Brien C, Huw L, Koeppen H, Fridlyand J, Brachmann RK, Haverty PM, Pandita A, Mohan S, Sampath D, Friedman LS, Ross L, Hampton GM, Amler LC, Shames DS, Lackner MR (2012) Phosphoinositide 3-kinase (PI3K) pathway alterations are associated with histologic subtypes and are predictive of sensitivity to PI3K inhibitors in lung cancer preclinical models. Clin Cancer Res 18(24):6771–6783. https://doi.org/10.1158/1078-0432.ccr-12-2347
Young H, Baum R, Cremerius U, Herholz K, Hoekstra O, Lammertsma AA, Pruim J, Price P (1999) Measurement of clinical and subclinical tumour response using [18F]-fluorodeoxyglucose and positron emission tomography: review and 1999 EORTC recommendations. European Organization for Research and Treatment of Cancer (EORTC) PET Study Group. Eur J Cancer 35(13):1773–1782
Rosen LS, LoRusso P, Ma WW, Goldman JW, Weise A, Colevas AD, Adjei A, Yazji S, Shen A, Johnston S, Hsieh HJ, Chan IT, Sikic BI (2016) A first-in-human phase I study to evaluate the MEK1/2 inhibitor, cobimetinib, administered daily in patients with advanced solid tumors. Invest New Drugs 34(5):604–613. https://doi.org/10.1007/s10637-016-0374-3
Sarker D, Ang JE, Baird R, Kristeleit R, Shah K, Moreno V, Clarke PA, Raynaud FI, Levy G, Ware JA, Mazina K, Lin R, Wu J, Fredrickson J, Spoerke JM, Lackner MR, Yan Y, Friedman LS, Kaye SB, Derynck MK, Workman P, de Bono JS (2015) First-in-Human Phase I Study of Pictilisib (GDC-0941), a Potent Pan-Class I Phosphatidylinositol-3-Kinase (PI3K) Inhibitor, in Patients with Advanced Solid Tumors. Clin Cancer Res 21(1):77–86. https://doi.org/10.1158/1078-0432.ccr-14-0947
Tolcher AW, Peng W, Calvo E (2018) Rational Approaches for Combination Therapy Strategies Targeting the MAP Kinase Pathway in Solid Tumors. Mol Cancer Ther 17(1):3–16. https://doi.org/10.1158/1535-7163.mct-17-0349
Jokinen E, Koivunen JP (2015) MEK and PI3K inhibition in solid tumors: rationale and evidence to date. Ther Adv Med Oncol 7(3):170–180. https://doi.org/10.1177/1758834015571111
Engelman JA, Chen L, Tan X, Crosby K, Guimaraes AR, Upadhyay R, Maira M, McNamara K, Perera SA, Song Y, Chirieac LR, Kaur R, Lightbown A, Simendinger J, Li T, Padera RF, Garcia-Echeverria C, Weissleder R, Mahmood U, Cantley LC, Wong KK (2008) Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med 14(12):1351–1356. https://doi.org/10.1038/nm.1890
Sos ML, Fischer S, Ullrich R, Peifer M, Heuckmann JM, Koker M, Heynck S, Stuckrath I, Weiss J, Fischer F, Michel K, Goel A, Regales L, Politi KA, Perera S, Getlik M, Heukamp LC, Ansen S, Zander T, Beroukhim R, Kashkar H, Shokat KM, Sellers WR, Rauh D, Orr C, Hoeflich KP, Friedman L, Wong KK, Pao W, Thomas RK (2009) Identifying genotype-dependent efficacy of single and combined PI3K- and MAPK-pathway inhibition in cancer. Proc Natl Acad Sci U S A 106(43):18351–18,356. https://doi.org/10.1073/pnas.0907325106
Grilley-Olson JE, Bedard PL, Fasolo A, Cornfeld M, Cartee L, Razak AR, Stayner LA, Wu Y, Greenwood R, Singh R, Lee CB, Bendell J, Burris HA, Del Conte G, Sessa C, Infante JR (2016) A phase Ib dose-escalation study of the MEK inhibitor trametinib in combination with the PI3K/mTOR inhibitor GSK2126458 in patients with advanced solid tumors. Invest New Drugs 34(6):740–749. https://doi.org/10.1007/s10637-016-0377-0
Bedard PL, Tabernero J, Janku F, Wainberg ZA, Paz-Ares L, Vansteenkiste J, Van Cutsem E, Perez-Garcia J, Stathis A, Britten CD, Le N, Carter K, Demanse D, Csonka D, Peters M, Zubel A, Nauwelaerts H, Sessa C (2015) A phase Ib dose-escalation study of the oral pan-PI3K inhibitor buparlisib (BKM120) in combination with the oral MEK1/2 inhibitor trametinib (GSK1120212) in patients with selected advanced solid tumors. Clin Cancer Res 21(4):730–738. https://doi.org/10.1158/1078-0432.ccr-14-1814
Shimizu T, Tolcher AW, Papadopoulos KP, Beeram M, Rasco DW, Smith LS, Gunn S, Smetzer L, Mays TA, Kaiser B, Wick MJ, Alvarez C, Cavazos A, Mangold GL, Patnaik A (2012) The clinical effect of the dual-targeting strategy involving PI3K/AKT/mTOR and RAS/MEK/ERK pathways in patients with advanced cancer. Clin Cancer Res 18(8):2316–2325. https://doi.org/10.1158/1078-0432.ccr-11-2381
Wainberg ZA, Alsina M, Soares HP, Brana I, Britten CD, Del Conte G, Ezeh P, Houk B, Kern KA, Leong S, Pathan N, Pierce KJ, Siu LL, Vermette J, Tabernero J (2017) A Multi-Arm Phase I Study of the PI3K/mTOR Inhibitors PF-04691502 and Gedatolisib (PF-05212384) plus Irinotecan or the MEK Inhibitor PD-0325901 in Advanced Cancer. Target Oncol 12(6):775–785. https://doi.org/10.1007/s11523-017-0530-5
Jokinen E, Koivunen JP (2015) Bcl-xl and Mcl-1 are the major determinants of the apoptotic response to dual PI3K and MEK blockage. Int J Oncol 47(3):1103–1110. https://doi.org/10.3892/ijo.2015.3071
Corcoran RB, Cheng KA, Hata AN, Faber AC, Ebi H, Coffee EM, Greninger P, Brown RD, Godfrey JT, Cohoon TJ, Song Y, Lifshits E, Hung KE, Shioda T, Dias-Santagata D, Singh A, Settleman J, Benes CH, Mino-Kenudson M, Wong KK, Engelman JA (2013) Synthetic lethal interaction of combined BCL-XL and MEK inhibition promotes tumor regressions in KRAS mutant cancer models. Cancer Cell 23(1):121–128. https://doi.org/10.1016/j.ccr.2012.11.007
Ramanathan RK, Hoff DDV, Eskens F, Blumenschein GR, Richards DA, Renshaw FG, Rajagopalan P, Kelly A, Pena CE, Mross KB (2014) A phase 1b trial of PI3K inhibitor copanlisib (BAY 80–6946) combined with the allosteric-MEK inhibitor refametinib (BAY 86–9766) in patients with advanced cancer. Journal of Clinical Oncology 32(15_suppl):2588–2588. https://doi.org/10.1200/jco.2014.32.15_suppl.2588
Juric D, Soria J-C, Sharma S, Banerji U, Azaro A, Desai J, Ringeisen F, Kaag A, Radhakrishnan R, Hourcade-Potelleret F, Maacke H, Ahnert J (2014) A phase 1b dose-escalation study of BYL719 plus binimetinib (MEK162) in patients with selected advanced solid tumors. Journal of Clinical Oncology 32(15_suppl):9051–9051. https://doi.org/10.1200/jco.2014.32.15_suppl.9051
Acknowledgements
The authors thank the patients and their families. All authors participated in manuscript writing and approved the final version of the manuscript. Editing and writing assistance was provided by A. Daisy Goodrich (Genentech, Inc.) and funded by Genentech, Inc.
Funding
The work was supported by Genentech, Inc., South San Francisco, CA, USA.
Author information
Authors and Affiliations
Corresponding author
Ethics declarations
Conflict of interest
Geoffrey I. Shapiro declares that he was on the advisory board at Almac, Angiex, Astex, Bayer, Bicycle Therapeutics, Cybrexa Therapeutics, Daiichi Sankyo, Fusion Pharmaceuticals, G1 Therapeutics, Ipsen, Lilly, Merck/EMD Serono, Pfizer, Roche, and Sierra Oncology; funding for an investigator-initiated clinical trial from Array and Pfizer; sponsored research agreements from Lilly, EMD Serono, Merck, and Sierra Oncology; support to the Dana-Farber Cancer Institute for this study from Genentech; travel from Bayer, Bicycle Therapeutics, G1 Therapeutics, Lilly, Pfizer, and Sierra Oncology; patent 9,872,874 issued for “Dosage regimen for sapacitabine and seliciclib” and provisional patent application 62/538,319 for “Compositions and methods for predicting response and resistance to CDK4/6 inhibition.” Patricia LoRusso declares that she was on the advisory board at Alexion, Ariad, GenMab, Glenmark, Menarini, Novartis, Genentech, CytomX, Omniox, Ignyta, and Takeda; data safety monitoring board at Agios, Halozyme, and FivePrime; imCORE alliance at Roche/Genentech; and consultant at SOTIO. Eunice Kwak declares that she is an employee of Novartis and former employee of Massachusetts General Hospital Cancer Center where this study was conducted. Susan Pandya declares that she is an employee of Agios and former employee of Beth Israel Deaconess Medical Center where this study was conducted. Charles M. Rudin declares that he was a consultant for AbbVie, Amgen, Ascentage, Astra Zeneca, Bicycle, BMS, Celgene, Daiichi Sankyo, Genentech/Roche, Ipsen, Loxo, Pharmamar, and scientific advisory board at Elucida and Harpoon. Carla Kurkjian declares that she has no conflict of interest. James M. Cleary declares research funding from Merck and Tesaro; consulting at Bristol Myers Squib; and travel funding from Bristol Myers Squib, Agios, and Roche. Mary Jo Pilat declares that she has no conflict of interest. Suzanne Jones declares that her institution received payment for services related to clinical trial from AbbVie, Acerta Pharma, Agios, Amgen, ARMO Biosciences, Array Biopharma, AstraZeneca, Blueprint Medicine, Boston Biomedical, Bristol Myers Squibb, CALGB, Celgene, Daiichi Sankyo, Eisai, EMD Serono, Genentech, Gilead Sciences, Imclone, Incyte, Ipsen Biopharma, Leap Therapeutics, Lilly, Macrogenics, MedImmune, Merck, Merrimack, Merus NV, Millennium, Novartis, Novocure, OncoMed Pharmaceuticals, Pfizer, Rgenix, Roche, Taiho Oncology, and Takeda Pharmaceuticals; and institution received payment for consulting services performed by Dr. Jones from Vertex, Novartis, Teva, Onyx, Clovis, and Janssen. Alex de Crespigny, Jill Fredrickson, Luna Musib, Yibing Yan, and Matthew Wongchenko declare that they are employees of Genentech, Inc., and are stockholders of Roche. Hsin-Ju Hsieh declares that she is an employee at MedImmune; former employment at Genentech, Inc., and stockholder of Roche. Mary R. Gates and Iris T. Chan declare that they are employees of Genentech, Inc. and are stockholders of Roche. Johanna Bendell declares that her institution received payment for services related to clinical trial from AbbVie, Acerta Pharma, Agios, Amgen, ARMO Biosciences, Array Biopharma, AstraZeneca, Blueprint Medicine, Boston Biomedical, Bristol Myers Squibb, CALGB, Celgene, Daiichi Sankyo, Eisai, EMD Serono, Genentech, Gilead Sciences, Imclone, Incyte, Ipsen Biopharma, Leap Therapeutics, Lilly, Macrogenics, MedImmune, Merck, Merrimack, Merus NV, Millennium, Novartis, Novocure, OncoMed Pharmaceuticals, Pfizer, Rgenix, Roche, Taiho Oncology, and Takeda Pharmaceuticals; institution received payment for consulting services performed by Dr. Bendell from Amgen, Arrys Therapeutics, BeiGene, Bristol Myers Squibb, Celgene, Cerulean, Continuum Clinical, Daiichi Sankyo, Evelo Biosciences, Five Prime Therapeutics, Forma Therapeutics, Genentech, Janssen, MedImmune, Merck, Merrimack, Moderna Therapeutics, Roche, Seattle Genetics, Taiho Oncology, Tanabe Research Laboratories, Tolero, and Translational Drug Development; Dr. Bendell received non-financial support including travel reimbursement and medical editing support from Genentech; and Dr. Bendell also received in-kind support for food and beverage from Genentech.
Ethical approval
This article contains studies with human participants performed by the authors. All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.
Informed consent
Informed consent was obtained from all individual participants included in the study.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
ESM 1
(PDF 1094 kb)
Rights and permissions
About this article
Cite this article
Shapiro, G.I., LoRusso, P., Kwak, E. et al. Phase Ib study of the MEK inhibitor cobimetinib (GDC-0973) in combination with the PI3K inhibitor pictilisib (GDC-0941) in patients with advanced solid tumors. Invest New Drugs 38, 419–432 (2020). https://doi.org/10.1007/s10637-019-00776-6
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1007/s10637-019-00776-6