Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling...

14
Small Molecule Therapeutics Targeting the MAPK Signaling Pathway in Cancer: Promising Preclinical Activity with the Novel Selective ERK1/2 Inhibitor BVD-523 (Ulixertinib) Ursula A. Germann 1 , Brinley F. Furey 1 , William Markland 1 , Russell R. Hoover 1 , Alex M. Aronov 1 , Jeffrey J. Roix 2 , Michael Hale 1 , Diane M. Boucher 1 , David A. Sorrell 3 , Gabriel Martinez-Botella 1 , Matthew Fitzgibbon 1 , Paul Shapiro 4 , Michael J. Wick 5 , Ramin Samadani 4 , Kathryn Meshaw 6 , Anna Groover 2 , Gary DeCrescenzo 2 , Mark Namchuk 1 , Caroline M. Emery 2 , Saurabh Saha 2 , and Dean J. Welsch 2 Abstract Aberrant activation of signaling through the RASRAFMEKERK (MAPK) pathway is implicated in numerous cancers, making it an attractive therapeutic target. Although BRAF and MEK-targeted combination therapy has demonstrated signi- cant benet beyond single-agent options, the majority of patients develop resistance and disease progression after approximately 12 months. Reactivation of ERK signaling is a common driver of resistance in this setting. Here we report the discovery of BVD-523 (ulixertinib), a novel, reversible, ATP-competitive ERK1/2 inhibitor with high potency and ERK1/2 selectivity. In vitro BVD-523 treatment resulted in reduced proliferation and enhanced caspase activity in sensitive cells. Interestingly, BVD-523 inhibited phosphorylation of tar- get substrates despite increased phosphorylation of ERK1/2. In in vivo xenograft studies, BVD-523 showed dose-dependent growth inhibition and tumor regression. BVD-523 yielded synergistic antiproliferative effects in a BRAF V600E -mutant mel- anoma cell line xenograft model when used in combination with BRAF inhibition. Antitumor activity was also demonstrat- ed in in vitro and in vivo models of acquired resistance to single- agent and combination BRAF/MEKtargeted therapy. On the basis of these promising results, these studies demonstrate BVD-523 holds promise as a treatment for ERK-dependent cancers, including those whose tumors have acquired resistance to other treatments targeting upstream nodes of the MAPK pathway. Assessment of BVD-523 in clinical trials is underway (NCT01781429, NCT02296242, and NCT02608229). Mol Cancer Ther; 16(11); 235163. Ó2017 AACR. Introduction Through aberrant activation of ERK signaling, genetic altera- tions in RAS or RAF family members result in rapid tumor growth, increased cell survival, and resistance to apoptosis. Activating mutations of RAS family members KRAS and NRAS are found in approximately 30% of all human cancers, with particularly high incidence in pancreatic and colorectal cancer (1, 2). Constitutively activating mutations in BRAF (codon for V600) have been observed primarily in melanoma (48%), papillary thyroid carcinoma (44%), colorectal cancer (15%), hairy cell leukemia (100%), and nonsmall cell lung cancer (4%; ref. 3). Although rare, cancers bearing genetic mutations that result in changes of the downstream components ERK (MAPK1 and MAPK3) and MEK (MAP2K1 and MAP2K2) have also been reported (4, 5). Furthermore, alterations known to activate the MAPK (RASRAFMEKERK) pathway are also common in the setting of resistance to targeted therapies, for example inhibitors of BRAF, MEK, and ALK (6). Thus, targeting the MAPK pathway terminal master kinases (ERK1/2) is a promising strategy for the treatment of a broad spectrum of tumors harboring pathway-activating alterations. Clinical precedent for targeting the MAPK pathway already exists; three MAPK pathway-targeting drugs have been approv- ed by the FDA for single-agent treatment of nonresectable or metastatic cutaneous melanoma with BRAF V600 mutations: the BRAF inhibitors vemurafenib and dabrafenib and the MEK inhibitor trametinib (7, 8). An additional MEK inhib- itor, cobimetinib, is approved in this indication as part of a 1 Vertex Pharmaceuticals Inc, Cambridge, Massachusetts. 2 BioMed Valley Dis- coveries, Inc., Kansas City, Missouri. 3 Horizon Discovery Ltd, Cambridge, United Kingdom. 4 University of Maryland School of Pharmacy, Baltimore, Maryland. 5 START, San Antonio, Texas. 6 Charles River Discovery Services, Morrisville, North Carolina. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). S. Saha and D.J. Welsch contributed equally to this article. Current address for U.A. Germann: OnKognos Scientic Consulting, Newton, Massachusetts and InnovaTID Pharmaceuticals Incorporated, Cambridge, Mas- sachusetts; current address for J.J. Roix, PhoreMost Ltd, Cambridge, United Kingdom; current address for M. Hale, Ra Pharmaceuticals, Cambridge, Massa- chusetts; current address for G. Martinez-Botella, Praxis Precision Medicines Incorporated, Cambridge, Massachusetts; current address for R. Samadani, MedImmune, Gaithersburg, Maryland; current address for M. Namchuk, Alkermes, Waltham, Massachusetts; and current address for S. Saha, Atlas Venture, Cambridge, Massachusetts. Corresponding Author: Dean J. Welsch, BioMed Valley Discoveries, 4435 Main Street, Suite 550, Kansas City, MO 64111. Phone: 816-389-8831; Fax: 816-960- 6976; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-17-0456 Ó2017 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org 2351 on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Transcript of Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling...

Page 1: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

Small Molecule Therapeutics

Targeting the MAPK Signaling Pathway in Cancer:Promising Preclinical Activity with the NovelSelectiveERK1/2 InhibitorBVD-523 (Ulixertinib)Ursula A. Germann1, Brinley F. Furey1,William Markland1, Russell R. Hoover1,Alex M. Aronov1, Jeffrey J. Roix2, Michael Hale1, Diane M. Boucher1,David A. Sorrell3, Gabriel Martinez-Botella1, Matthew Fitzgibbon1, Paul Shapiro4,Michael J.Wick5, Ramin Samadani4, Kathryn Meshaw6, Anna Groover2,Gary DeCrescenzo2, Mark Namchuk1, Caroline M. Emery2, Saurabh Saha2,and Dean J.Welsch2

Abstract

Aberrant activation of signaling through the RAS–RAF–MEK–ERK (MAPK) pathway is implicated in numerous cancers,making it an attractive therapeutic target. Although BRAF andMEK-targeted combination therapy has demonstrated signifi-cant benefit beyond single-agent options, the majority ofpatients develop resistance and disease progression afterapproximately 12 months. Reactivation of ERK signaling is acommon driver of resistance in this setting. Here we reportthe discovery of BVD-523 (ulixertinib), a novel, reversible,ATP-competitive ERK1/2 inhibitor with high potency andERK1/2 selectivity. In vitro BVD-523 treatment resulted inreduced proliferation and enhanced caspase activity in sensitivecells. Interestingly, BVD-523 inhibited phosphorylation of tar-get substrates despite increased phosphorylation of ERK1/2. In

in vivo xenograft studies, BVD-523 showed dose-dependentgrowth inhibition and tumor regression. BVD-523 yieldedsynergistic antiproliferative effects in a BRAFV600E-mutant mel-anoma cell line xenograft model when used in combinationwith BRAF inhibition. Antitumor activity was also demonstrat-ed in in vitro and in vivo models of acquired resistance to single-agent and combination BRAF/MEK–targeted therapy. On thebasis of these promising results, these studies demonstrateBVD-523 holds promise as a treatment for ERK-dependentcancers, including those whose tumors have acquired resistanceto other treatments targeting upstream nodes of the MAPKpathway. Assessment of BVD-523 in clinical trials is underway(NCT01781429, NCT02296242, and NCT02608229). Mol CancerTher; 16(11); 2351–63. �2017 AACR.

IntroductionThrough aberrant activation of ERK signaling, genetic altera-

tions in RAS or RAF family members result in rapid tumor

growth, increased cell survival, and resistance to apoptosis.Activating mutations of RAS family members KRAS and NRASare found in approximately 30% of all human cancers, withparticularly high incidence in pancreatic and colorectal cancer(1, 2). Constitutively activating mutations in BRAF (codon forV600) have been observed primarily in melanoma (�48%),papillary thyroid carcinoma (�44%), colorectal cancer(�15%), hairy cell leukemia (�100%), and non–small celllung cancer (�4%; ref. 3). Although rare, cancers bearinggenetic mutations that result in changes of the downstreamcomponents ERK (MAPK1 and MAPK3) and MEK (MAP2K1and MAP2K2) have also been reported (4, 5). Furthermore,alterations known to activate the MAPK (RAS–RAF–MEK–ERK)pathway are also common in the setting of resistance totargeted therapies, for example inhibitors of BRAF, MEK, andALK (6). Thus, targeting the MAPK pathway terminal masterkinases (ERK1/2) is a promising strategy for the treatment of abroad spectrum of tumors harboring pathway-activatingalterations.

Clinical precedent for targeting the MAPK pathway alreadyexists; three MAPK pathway-targeting drugs have been approv-ed by the FDA for single-agent treatment of nonresectable ormetastatic cutaneous melanoma with BRAFV600 mutations:the BRAF inhibitors vemurafenib and dabrafenib and theMEK inhibitor trametinib (7, 8). An additional MEK inhib-itor, cobimetinib, is approved in this indication as part of a

1Vertex Pharmaceuticals Inc, Cambridge, Massachusetts. 2BioMed Valley Dis-coveries, Inc., Kansas City, Missouri. 3Horizon Discovery Ltd, Cambridge, UnitedKingdom. 4University of Maryland School of Pharmacy, Baltimore, Maryland.5START, San Antonio, Texas. 6Charles River Discovery Services, Morrisville,North Carolina.

Note: Supplementary data for this article are available at Molecular CancerTherapeutics Online (http://mct.aacrjournals.org/).

S. Saha and D.J. Welsch contributed equally to this article.

Current address for U.A. Germann: OnKognos Scientific Consulting, Newton,Massachusetts and InnovaTID Pharmaceuticals Incorporated, Cambridge, Mas-sachusetts; current address for J.J. Roix, PhoreMost Ltd, Cambridge, UnitedKingdom; current address for M. Hale, Ra Pharmaceuticals, Cambridge, Massa-chusetts; current address for G. Martinez-Botella, Praxis Precision MedicinesIncorporated, Cambridge, Massachusetts; current address for R. Samadani,MedImmune, Gaithersburg, Maryland; current address for M. Namchuk,Alkermes, Waltham, Massachusetts; and current address for S. Saha, AtlasVenture, Cambridge, Massachusetts.

Corresponding Author: Dean J. Welsch, BioMed Valley Discoveries, 4435 MainStreet, Suite 550, Kansas City, MO 64111. Phone: 816-389-8831; Fax: 816-960-6976; E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-17-0456

�2017 American Association for Cancer Research.

MolecularCancerTherapeutics

www.aacrjournals.org 2351

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Page 2: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

combination regimen with vemurafenib; furthermore, thecombination of dabrafenib and trametinib is also approvedin this indication (9, 10). Approved BRAF inhibitors providethe advantage of selectivity for BRAFV600-mutant proteins, byvirtue of ability to signal as monomers, unlike wild-type RAFproteins which require dimerization to activate signaling,therefore providing a greater therapeutic index.

Importantly, the concomitant use of BRAF- plus MEK-targetedtherapies has demonstrated simultaneous targeting of differentnodes in the MAPK pathway can enhance the magnitude andduration of response (11–13).

Despite improvements in clinical outcomes seen with BRAF-/MEK-inhibitor combination therapies, durable benefit is lim-ited by the eventual development of acquired resistance andsubsequent disease progression, with median progression-freesurvival (PFS) ranging from approximately 9 to 11 months(11, 13–15). Genetic mechanisms of acquired resistance tosingle-agent BRAF inhibition have been intensely studied;some examples of identified resistance mechanisms includesplice variants of BRAF (16), BRAFV600E amplification (17),MEK mutations (18), NRAS mutations, and RTK activation(19, 20). Resistance mechanisms in the setting of BRAF/MEKinhibitor combination therapy are beginning to emerge andmirror that of BRAF single-agent resistance (21, 22). Thesegenetic events all share the ability to reactivate ERK signaling.Indeed, reactivated MAPK pathway signaling as measured byERK transcriptional targets is common in tumor biopsies fromBRAF inhibitor–resistant patients (23). Moreover, ERK1/2 reac-tivation has been observed in the absence of a genetic mech-anism of resistance (24, 25). The quest to achieve durableclinical benefit has led researchers to focus on evaluatingadditional agents that target the downstream MAPK compo-nents ERK1/2. Inhibiting ERK may provide important clinicalbenefit to patients with acquired resistance to BRAF/MEKinhibition. ERK family kinases have shown promise as thera-peutic targets in preclinical cancer models, including thoseresistant to BRAF or MEK inhibitors (26–28); however, thepotential for ERK inhibitors expands beyond acquired resis-tance in melanoma.

Targeting ERK1/2 is a rational cornerstone therapy in anytumor type harboring known drivers of MAPK, not only BRAF/MEK therapy–relapsed patients. As ERK1/2 reside downstreamin the pathway, they represent a particularly attractive treatmentstrategy within the MAPK cascade that may avoid upstreamresistance mechanisms. Here, we report the preclinical charac-terization of BVD-523 (ulixertinib) in models of MAPK path-way–dependent cancers, including drug-na€�ve and BRAF/MEKtherapy acquired–resistance models. Phase I clinical studies ofBVD-523 are ongoing (NCT01781429, NCT02296242, andNCT02608229).

Materials and MethodsCompounds

Compounds were sourced from the following vendors;SCH772984 (SelleckChem), pyrazolylpyrrole (Santa Cruz Bio-technology), Temozolomide (Temodar, Schering Corporation),dabrafenib (Chemietek), trametinib (LC Laboratories), vemura-fenib (Focus Synthesis LLC), and selumetinib (Chemietek).BVD-523 was synthesized as per method described in US patentnumber 7,354,939 B2.

Ki determination of ERK2The inhibitory activity of BVD-523 against ERK2 was deter-

mined using a radiometric assay, with final concentration of thecomponents being 100 mmol/L HEPES (pH 7.5), 10 mmol/LMgCl2, 1 mmol/L dithiothreitol (DTT), 0.12 nmol/L ERK2,10 mmol/L myelin basic protein (MBP), and 50 mmol/L33P-g-ATP. All reaction components, with the exception of ATPand MBP, were premixed and aliquoted (33 mL) into a 96-wellplate. A stock solution of compound in DMSO was used tomake up to 500-fold dilutions; a 1.5-mL aliquot of DMSO orinhibitor in DMSO was added to each well. The reaction wasinitiated by adding the substrates 33P-g-ATP and MBP (33 mL).After 20 minutes, the reaction was quenched with 20% (w/v)tricholoracetic acid (TCA; 55 mL) containing 4 mmol/L ATP,transferred to the GF/B filter plates, and washed 3 times with5% (w/v) TCA. Following the addition of Ultimate Goldscintillant (50 mL), the samples were counted in a PackardTopCount. From the activity versus concentration titrationcurve, the Ki value was determined by fitting the data to anequation for competitive tight binding inhibition kinetics usingPrism software, version 3.0.

IC50 determination of ERK2ERK2 activity was assayed by a standard coupled-enzyme assay.

The final concentrations were as follows: 0.1 mol/L HEPES (pH7.5), 10 mmol/L MgCl2, 1 mmol/L DTT, 2.5 mmol/L phospho-enolpyruvate, 200 mmol/L NADH, 50 mg/mL pyruvate kinase, 10mg/mL lactate dehydrogenase, 65 mmol/L ATP, and 800 mmol/Lpeptide (ATGPLSPGPFGRR). All of the reaction componentsexcept ATP were premixed with ERK and aliquoted into assay-plate wells. BVD-523 in DMSO was introduced into each well,keeping the concentration of DMSO per well constant. BVD-523concentrations spanned a 500-fold range for each titration. Theassay plate was incubated at 30�C for 10 minutes in the platereader compartment of the spectrophotometer (MolecularDevices) before initiating the reaction by adding ATP. The absor-bance change at 340 nmwas monitored as a function of time; theinitial slope corresponds to the rate of the reaction. The rate versusconcentration of the BVD-523 titration curve was fitted either toan equation for competitive tight-binding inhibition kinetics todetermine a value for Ki or to a 3-parameter fit to determine theIC50 using Prism software, version 3.0.

Cell linesThe cell lines RKO, SW48, A375, MIAPaCa-2, HCT116,

Colo205,HT-29, ZR-75-1, andAN3Cawere obtained fromATCC,confirmed mycoplasma negative, and authenticated by STR gen-otyping. Cell line G-361 was obtained from ECACC and con-firmed mycoplasma negative and authenticated by STR genotyp-ing. For each cell line, upon thawing, experiments were performedwhen required cell number was achieved to avoid long-termpassaging of cells.

Antitumor activity in A375 xenograftsXenografts were initiated with A375 cells maintained by serial

subcutaneous transplantation in female athymic nudemice. Eachtest mouse received an A375 tumor fragment (1mm3) implantedsubcutaneously in the right flank. Once tumors reached target size(80–120 mm3), animals were randomized into treatment andcontrol groups, and drug treatment was initiated. BVD-523 in 1%

Germann et al.

Mol Cancer Ther; 16(11) November 2017 Molecular Cancer Therapeutics2352

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Page 3: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

(w/v) carboxymethylcellulose (CMC) was administered orallytwice daily at doses of 5, 25, 50, 100, or 150 mg/kg.

The efficacy of BVD-523 in combination with dabrafenibwas evaluated in mice randomized into 9 groups of 15 and1 group (Group 10) of 10. Dabrafenib was administered orallyat 50 or 100 mg/kg once daily and BVD-523 was administeredorally at 50 or 100 mg/kg twice daily, alone and in combina-tion, until study end; vehicle-treated control groups were alsoincluded. Combination dosing was stopped on Day 20 tomonitor for tumor regrowth. Animals were monitored individ-ually and euthanized when each tumor reached an endpointvolume of 2,000 mm3 or on the final day (day 45), whichevercame first, and median time to endpoint (TTE) calculated. Thecombination was also evaluated in an upstaged A375 modelwhere larger tumors in the range 228–1,008 mm3 were evalu-ated. Here, mice were randomized into 1 group (Group 1) of14 and 4 groups (Groups 2–5) of 20. Dosing was initiated onday 1 with dabrafenib plus BVD-523 (25 mg/kg dabrafenib þ50 mg/kg BVD-523 or 50 mg/kg dabrafenib þ 100 mg/kg BVD-523), with each agent given orally twice daily until study end.The study included 50 mg/kg dabrafenib and 100 mg/kg BVD-523 monotherapy groups as well as a vehicle-treated controlgroup. Tumors were measured twice weekly. Combinationdosing was stopped on day 42 to monitor for tumor regrowththrough study end (day 60). Treatment outcome was deter-mined from % tumor growth delay (TGD), defined as thepercent increase in median TTE for treated versus control mice,with differences between groups analyzed via log-rank survivalanalysis. For tumor growth inhibition (TGI) analysis, % TGIvalues were calculated and reported for each treatment (T)group versus the control (C). Mice were also monitored forcomplete regression (CR) and partial response (PR). Animalswith a CR at the end of the study were additionally classified astumor free survivors (TFS). All in vivo studies were conducted inaccordance with generally recognized good laboratory practices,all procedures were in compliance with the Animal Welfare ActRegulations (9 CFR 3). All in vivo studies were reviewed andapproved by the appropriate Institutional Animal Care and UseCommittees (IACUC).

BVD-523 activity in Colo205 xenograftsHuman Colo205 cells were cultured in RPMI1640 supplemen-

ted with 10% (v/v) FBS, 100 U/mL penicillin, 100 mg/mL strep-tomycin (Invitrogen), and 2 mmol/L L-glutamine. Cells werecultured for <4 passages prior to implantation. Female athymicnude mice (19–23 g) were injected subcutaneously with 2 � 106

Colo205 cells into the right dorsal axillary region on day 0.Mice with an approximate tumor volume of 200 mm3 were

randomized unblinded into 6 experimental groups. Vehicle con-trol, 1% CMC (w/v), was prepared weekly. BVD-523 was sus-pended in 1% (w/v) CMC at the desired concentration andhomogenized on ice at 6,500 rpm for 50 minutes. BVD-523suspensions were prepared weekly and administered orally twicedaily at total daily doses of 50, 100, 150, and 200 mg/kg (n ¼ 12/group)onan8- or 16-hour dosing schedule for 13days. The vehiclecontrol (n¼ 12) was administered using the same dosing regimen.

Efficacy testing in a patient-derived tumor xenograftBVD-523 was evaluated in a patient-derived xenograft

(ST052C) representing melanoma from a BRAFV600E patient whohad become clinically refractory to vemurafenib. Immunocom-

promised mice between 5–8 weeks of age were housed onirradiated corncob bedding (Teklad) in individual HEPA venti-lated cages (Sealsafe Plus, Techniplast USA) on a 12-hour light–dark cycle at 70–74�F (21–23�C) and 40–60% humidity. Animalswere fed water ad libitum (reverse osmosis, 2 ppm Cl2) and anirradiated standard rodent diet (Teklad 2919) consisting of 19%protein, 9% fat, and 4% fiber. Tumor fragments were harvestedfrom host animals and implanted into immunodeficient mice.The study was initiated at amean tumor volume of approximately170 mm3, whereby the animals were randomized unblindedinto 4 groups, including a control [1% (v/v) CMC orally, twicedaily � 31] and 3 treatment groups [BVD-523 (100 mg/kg),dabrafenib (50 mg/kg), or BVD-523/dabrafenib (100/50 mg/kg),n ¼ 10/group]; All treatment drugs were administered orally on atwice daily � 31 schedule.

ResultsDiscovery and initial characterization of the novel ERK1/2inhibitor, BVD-523 (ulixertinib)

Following extensive optimization of leads originally identifiedusing a high-throughput, small-molecule screen (29), a noveladenosine triphosphate (ATP)-competitive ERK1/2 inhibitor,BVD-523 (ulixertinib) was identified (Fig. 1A). BVD-523 is apotent ERK inhibitor with a Ki of 0.04 � 0.02 nmol/L againstERK2. It was shown to be a reversible, competitive inhibitor ofATP, as the IC50 values for ERK2 inhibition increased linearly withincreasingATP concentration,with dependence of enzyme rate onBVD-523 concentration (Fig. 1B andC). The IC50 remained nearlyconstant for incubation times �10 minutes, suggesting rapidequilibrium and binding of BVD-523 with ERK2 (Fig. 1D).BVD-523 is also a tight-binding inhibitor of recombinant ERK1(30), exhibiting a Ki of <0.3 nmol/L.

Binding of BVD-523 to ERK2 was demonstrated using calori-metric studies and compared with data generated using the ERKinhibitors SCH772984 (26) and pyrazolylpyrrole (31). All com-pounds bound and stabilized inactive ERK2 with increasingconcentration, as indicated by positive DTm values (Fig. 1E). The10- to 15-degree change in DTm observed with BVD-523 andSCH772984 is consistent with compounds that have low nano-molar binding affinities (32). BVD-523 demonstrated a strongbinding affinity to both phosphorylated active ERK2 (pERK2) andinactive ERK2 (Fig. 1F), with a stronger affinity to pERK2 com-pared with inactive ERK2. BVD-523 did not interact with thenegative control protein p38a MAP kinase (Fig. 1F).

BVD-523 demonstrated excellent ERK1/2 kinase selectivitybased on biochemical counter-screens against 75 kinases, inaddition to ERK1 andERK2. The ATP concentrationswere approx-imately equal to the Km in all assays. Kinases inhibited to greaterthan 50% by 2 mmol/L BVD-523 were retested to generateKi values (or apparent Ki; Fig. 1G). Twelve of the 14 kinases hada Ki of <1 mmol/L. The selectivity of BVD-523 for ERK2 was>7,000-fold for all kinases tested except ERK1, which was inhib-ited with a Ki of <0.3 nmol/L (10-fold). Therefore, BVD-523 is ahighly potent and selective inhibitor of ERK1/2.

BVD-523 inhibits cellular proliferation and enhancescaspase-3/7 activity in vitro while demonstrating substrateinhibition despite increased ERK1/2 phorphorylation

The antiproliferative impact of BVD-523 treatment on sen-sitive cancer cell lines was characterized by FACS analysis on

ERK Inhibitor BVD-523 in Preclinical Cancer Models

www.aacrjournals.org Mol Cancer Ther; 16(11) November 2017 2353

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Page 4: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

BRAFV600E-mutant melanoma cell line, UACC-62, following24-hour treatment with BVD-523. BVD-523–treated cells werearrested in the G1 phase of the cell cycle in a concentration-dependent manner (Fig. 2A; Supplementary Fig. S1A). The per-

centage of cells arrested in G1 was also demonstrated to be timedependent, as shown by increasing proportion of KRASG12C-mutant pancreatic cell line MiaPaca-2 in G1 following 24 and48 hours treatment with BVD-523 (Supplementary Fig. S1B).

BVD-523161412Cl

NH

HN

AE

N

OH

CI

HO N

1086420

0.01 0.1 1 10

Compound (mmol/L)

DTm

(K)

SCH772984Pyrazolylpyrrole

ERK2154

3

2

1

00 1,000

4,000

3,000

2,000Rat

e

1,000

00 1 2 3

2,000 3,000 4,000

12

9

6

3

0

0.01 0.1 1 10BVD-523 (mmol/L)

KinaseCDK1/cyclinB 0.07a

0.09a

0.09a

0.65a

0.45a

0.24a

0.24a

0.45

11.1

0.00030.00004

0.32

1.3

0.36

CDK5/p35

ERK1ERK2GSK3bJNK2aJNK3P38gP38dROCKIROCKIIRSK3aApparent.<50% inhibition at 2 mmol/L:ABL, AKT3, AMPK,

SRC, SYK, TAK1, TIE2, ZAP70PKCg, PKCi, PKC8, PRAK, PRK2, cRAF, SGK,PAK2 PDGFRa, PDK1, PKA, PKCa, PKCb II,MSK1, MST2, NAK, NEK2, p38a, p38b, p70S6K,LCK, LYN, cMET, MKK4, MKK6, MKK7b, MLK2,IKKi, IRAK4, IRTK, ITK, JAK3, JNK1a 1, KDR,FES, FGFR3, FLT3, FYN, IGF1R, IKKa, IKKb,CHK1, CHK2, CK1, CK2, CSK, EGFR, EPHB4,AUR1, AUR2, AXL, BLK, CAMKII, CAMKIV,

CDK2/cyclinA

CDK6/cycinD3

Ki (mmol/L)

ATP (mmol/L)

BVD-523 (nmol/L)

Incubation time (min)

00

5

10

15

10 20 30 40 50 60 70

IC50

(nm

ol/L

)IC

50 (n

mol

/L)

DTm

(K)

pERK2p38a

B F

C

D

G

Figure 1.

Discovery and characterization ofnovel ERK1/2 inhibitor BVD-523(ulixertinib). A, Structure of BVD-523.B, Inhibitory potency of BVD-523 forERK2 as a function of ATPconcentration, C, Demonstration ofenzyme inhibition as a function ofinhibitor concentration in thepresence of 50 mmol/L ATP, and D,Measurement of inhibitory potency asa function of incubation time. Bindingof the ERK inhibitors BVD-523,SCH772984, and pyrazolylpyrrole toERK2 (E) and phosphorylated ERK2as measured by changes in enzymemelting temperatures (p38a includedas negative control; F). G, Potency ofBVD-523 against a panel of kinases.For B–G, representative data areshown.

Germann et al.

Mol Cancer Ther; 16(11) November 2017 Molecular Cancer Therapeutics2354

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Page 5: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

00

102030405060708090

100

BA

250 500 1,000

BVD-523 (nmol/L)

% C

ells

2,000 4,000

Cell phase% G2

% G1

% S

BVD-523 (nmol/L)

pERK

4 h 24 h0 30 150 20000 30 150 2000

pRSK

DUSP6

Actin

CyclinD1

pRB(S780)

BIM-EL

Prot

ein

abun

danc

e (%

cha

nge

from

DM

SO c

ontr

ol)

Cell line

pRSK1

CyclinD1

pERK1/2

A37

5

AN

3Ca

Col

o205

HC

T116

HT2

9

MIA

PaC

a2

Cell line

ZR-7

5-1

DLD

-1

G36

1

HT2

9

Mia

PaC

a2

A37

5

SK

-Mel

-5

LS41

1N

Col

o205

UA

CC

-62

WM

-266

-4

MK

N-4

5

BVD

-523

IC50

(mm

ol/L

)

0

−10

−90

−80

−70

−60

−50

−40

−30

−20

−10

0

0

100

200

300

400

500

600

700

800

900

−20

−30

−40

−50

−60

−70

−80

−90

−100

20.00

10.00

6.004.00

2.00

1.00

0.600.40

0.20

0.100.06

Compound

Mutation

Caspaseactivity

-++/-++++++++

BVD

BRAFKRAS-G13DKRAS-G12CKRAS-wtBRAF-wt

VxGDCSCH

DC

Figure 2.

BVD-523 inhibits cellular proliferation and enhances caspase-3/7 activity in vitro. A, Percentage of UACC-62 cell population in G2, S, or G1 phase of thecell cycle following incubation for 24 hours with increasing concentrations of BVD-523 (representative data shown). B, Cell line–dependent changes incaspase activity after 48 hours of exposure to BVD-523 (0, 0.082, 0.247, 0.741, 2.222, 6.667, 20 mmol/L) compared with the IC50 for cell viability(maximum change in caspase shown, <2-fold: �; 2-fold: þ/�; 2- 3- fold increase: þ; 3- 6- fold increase: þþþ; >6-fold increase þþþþþ; n ¼ 3). C, MAPKpathway and effector proteins are modulated by acute (4-hour) and prolonged (24-hour) BVD-523 treatment in BRAFV600E-mutant A375 cells(representative data shown). D, Levels of pERK1/2, pRSK, and Cyclin D1 measured in cell lines (A375, AN3Ca, Colo205, HCT116, HT29, and MIAPaca2) following24-hour treatment with ERK1/2 inhibitors BVD-523 (BVD), Vx11e (Vx), GDC-0994 (GDC), or SCH772984 (SCH; n ¼ 4 for each treatment group percell line). Data are average � SEM relative to DMSO control.

ERK Inhibitor BVD-523 in Preclinical Cancer Models

www.aacrjournals.org Mol Cancer Ther; 16(11) November 2017 2355

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Page 6: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

In addition, caspase-3/7 activity was analyzed as a measureof apoptosis in multiple human cancer cell lines (Fig. 2B). Acell-line–dependent increase in caspase-3/7 was observed fol-lowing treatment with BVD-523 for 48 hours. BVD-523 treat-ment resulted in pronounced caspase-3/7 induction (greaterthan 3-fold) in a subset of cell lines with sensitivity to BVD-523(<2 mmol/L IC50).

To further characterize the mechanism of action and effectson signaling elicited by BVD-523, the levels of various effectorand MAPK-related proteins were assessed in BVD-523–treatedBRAFV600E-mutant A375 melanoma cells (Fig. 2C). Phospho-ERK1/2 levels increased in a concentration-dependent mannerafter 4 and 24 hours of BVD-523 treatment. Despite prominentconcentration-dependent increases in pERK1/2 observed with2 mmol/L BVD-523 treatment, phosphorylation of the ERK1/2target RSK1/2 was reduced at both 4 and 24 hours, which isconsistent with sustained inhibition. Total protein levels ofDUSP6, transcription of which is a target of ERK1/2, were alsoattenuated at 4 and 24 hours. Following 24 hours of treatmentwith BVD-523, the apoptotic marker BIM-EL increased in adose-dependent manner, while cyclin D1 and pRB were atten-uated at 2 mmol/L. All effects are consistent with on-targetERK1/2 inhibition.

To examine the effects of BVD-523 on signaling relativeto other known ERK1/2 inhibitors (SCH772984, GDC-0994,and Vx-11e), a large-scale reverse-phase protein array (RPPA) of39 proteins was employed in a variety of cell lines withsensitivity to ERK inhibition (Supplementary Fig. S2). Celllines with common alterations in BRAF and RAS were assayed:BRAFV600E-mutant lines A375, Colo205, and HT29; KRASG12C-mutant cell line MIAPACa-2; KRASG13D-mutant cell lineHCT116; and AN3Ca with atypical HRASF82L mutation.Changes in protein levels are shown as a percentage changefrom dimethyl sulfoxide (DMSO)-treated control (Supplemen-tary Fig. S2). All ERK inhibitors elicited qualitatively simi-lar protein effects, with the exception of phosphorylation ofERK1/2 [pERK1/2 (ERK1/2 -T202, -Y204)]; SCH772984 inhib-ited pERK1/2 in all cell lines, while BVD-523, GDC-0994, andVx-11e markedly increased pERK1/2 (Fig. 2D). Phospho-p90RSK (pRSK1) and cyclin D1, which are proximal and distaltargets of pERK1/2, respectively, were similarly inhibited by allinhibitors tested regardless of the degree of ERK1/2 phosphor-ylation (Fig. 2D). These independent findings for BVD-523 areconsistent with studies showing that phosphorylation ofERK1/2 substrates RSK1/2 remained inhibited despite dramat-ically elevated pERK1/2 by Western blot analysis in A375 cells(Fig. 2C), in addition to protein-binding studies demonstratingBVD-523binding and stabilizationofpERK1/2 and inactive ERK1/2(Fig. 1E and F). Therefore, measuring increased pERK1/2 levelscould be considered as a pharmacodynamic biomarker for BVD-523, while quantifying inhibition of ERK1/2 targets such as pRSK orDUSP6 could serve this purpose more directly.

Additional protein changes are of note in this RPPA dataset(Supplementary Fig. S2). Decreased pS6-ribosomal protein isindicated to be another pharmacodynamic marker of ERK1/2inhibition, as evidenced in all cell lines with all inhibitors (Sup-plementary Fig. S3A). Furthermore, prominent induction of pAKTappears to be a cell line–dependent observation, where eachERK1/2 inhibitor induced pAKT in cell lines A375 and AN3CAcells (Supplementary Fig. S3A). Interestingly, the degree of inhi-bition of survival marker pBAD appears to differ between com-

pounds, with only modest inhibition of pBAD by GDC-0994compared with the other ERK1/2 inhibitors tested (Supplemen-tary Fig. S3B).

Next, we investigated how BVD-523 affects cellular localiza-tion of ERK1/2 and downstream target pRSK in a BRAFV600E-mutant RKO colorectal cell line (Supplementary Fig. S3C). Inresting cells, ERK1/2 localizes to the cytoplasm, and once stim-ulated, pERK1/2 migrates to target organelles, particularly thenucleus where transcriptional targets are activated (33). InDMSO-treated control cells, pERK1/2 is evident in both nuclearand cytoplasmic fractions, which is likely reflective of MAPKpathway activity due to the presence of BRAFV600E in this cell line.Treatment with BVD-523 resulted in elevated pERK1/2 in thenucleus and cytoplasm, as well as a modest increase in nucleartotal ERK1/2 compared with DMSO-treated cells, suggesting thatcompound-induced stabilization of pERK1/2 stimulates somenuclear translocation. Despite increased pERK1/2 in both com-partments, pRSK levels are lower in the cytoplasmic and nuclearcompartments compared with DMSO control. ComparatorMAPK signaling inhibitors (i.e., trametinib, SCH772984, dabra-fenib) inhibited phosphorylation of ERK1/2 and RSK, as reflectedby lower levels in the nuclear and cytoplasmic compartments.These data again suggest that BVD-523–associated increases inpERK1/2 are evident in both the cytoplasm and nucleus; how-ever, this does not translate to activation of target substrates. Thisis consistent with data presented in Fig. 2C and D.

BVD-523 demonstrates in vivo antitumor activity inBRAFV600E-mutant cancer cell line xenograft models

On the basis of our in vitro findings that BVD-523 reducedproliferation and induced apoptosis in a concentration-depen-dent manner, BVD-523 was administered by oral gavage todemonstrate its in vivo antitumor activity in models withMAPK/ERK pathway dependency. Xenograft models of melano-ma (cell line A375), and colorectal cancer (cell line Colo205),were utilized, both of which harbor a BRAFV600E mutation.

In A375 cell line xenografts, 5, 25, 50, and 100 mg/kg twicedaily doses of BVD-523 were compared following 18 days oftreatment. For all doses, no significant body weight changes wereobserved (Supplementary Fig. S4A). BVD-523 demonstrated sig-nificant dose-dependent antitumor activity starting at 50 mg/kgtwice daily (Fig. 3A). Doses of 50 and 100 mg/kg twice dailysignificantly attenuated tumor growth, with tumor growth inhi-bition (TGI) of 71% (P ¼ 0.004) and 99% (P < 0.001), respec-tively. Seven partial regressions (PR) were noted in the 100mg/kgtwice daily group; no regression responseswere noted in any othergroup.

BVD-523 demonstrated antitumor efficacy in a Colo205human colorectal cancer cell line xenograft model (Fig. 3B). Nosignificant body weight changes were observed (SupplementaryFig. S4B). BVD-523 again showed significant dose-dependenttumor volume regressions at doses of 50, 75, and 100 mg/kgtwice daily, yielding mean tumor regressions T/Ti (T ¼ end oftreatment, Ti ¼ treatment initiation) of �48.2%, �77.2%, and�92.3%, respectively (all P < 0.0001). Regression was notobserved at the lowest dose of BVD-523 (25 mg/kg twice daily);however, significant tumor growth inhibition, with a T/C (T ¼Treatment, C ¼ Control) of 25.2% (P < 0.0001), was observed.

In addition, BVD-523 resulted in dose-dependent antitumoractivity in KRASG12C-mutant pancreatic cell line xenograft model,MIAPaCa2, (Supplementary Fig. S4C). Here, animals were dosed

Germann et al.

Mol Cancer Ther; 16(11) November 2017 Molecular Cancer Therapeutics2356

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Page 7: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

with 10, 25, 50, 75, and 100 mg/kg twice daily BVD-523 for 15days. No significant body weight loss was observed in any treat-ment group (Supplementary Fig. S4D). At the highest doseadministered for BVD-523 (100 mg/kg twice daily), significanttumor growth inhibition compared with vehicle-treated controlswas observed, with a T/C of 5.3% on the last day of treatment (P <0.0001). As a negative control, xenografts ofmammary tumor cellline ZR-75-1were also tested for BVD-523 antitumor activity. Thismodel was not anticipated to respond to ERK inhibition, as it isnot known to harbor alterations in the MAPK/ERK pathway.Indeed, at each dose of BVD-523 tested (10, 30, 60, 100 mg/kgtwice daily), no antitumor activity was observed (SupplementaryFig. S4E), and no significant body weight loss was incurred(Supplementary Fig. S4F).

To establish the relationship between pharmacokineticsand pharmacodynamics, BVD-523 tumor concentrations werecompared with DUSP6 mRNA and pERK1/2 protein levelsover a 24-hour period following a single 100 mg/kg oraldose of BVD-523 (Fig. 3C and D). Phosphorylation ofERK1/2 was low in untreated tumors. Following treatmentwith BVD-523, ERK1/2 phosphorylation steadily increasedfrom 1 hour postdose to maximal levels at 8 hours postdose,then returned to predose levels by 24 hours. This increase inpERK1/2 correlated with BVD-523 drug tumor concentrations.The in vivo observation of increased pERK1/2 with BVD-523

treatment is consistent with earlier in vitro findings (Fig. 2Cand D). Conversely, DUSP6 expression decreased withincreasing BVD-523 tumor concentration (Fig. 3D). MaximalDUSP6 inhibition was observed 3 hours post-BVD-523 dose(�96% change from baseline), and similar to pERK, returnedto baseline by 24 hours.

Combination therapy with BVD-523 and a BRAF inhibitorprovides promising antitumor activity

Patients with BRAF-mutant cancer may acquire resistance tocombined BRAF/MEK therapy (21), warranting considerationof other combination approaches within the MAPK pathway.We assessed the antiproliferative effects of combining BVD-523with the BRAF inhibitors dabrafenib or vemurafenib in theBRAFV600E-mutant melanoma cell lines G-361 and A375. Asanticipated, single agents BVD-523, dabrafenib, and vemura-fenib were each active, and modest synergy was observed with aBVD-523 plus BRAF inhibitor combination (SupplementaryFig. S5A and S5B). This indicated that BVD-523 combinedwith BRAF inhibitors were at least additive and potentiallysynergistic in melanoma cell lines carrying a BRAFV600E muta-tion. Furthermore, generating acquired resistance in vitro fol-lowing continuous culturing of BRAFV600E-mutant cell line(A375) in BRAF inhibitor plus BVD-523 was challenging. Incontrast, generating resistance to dabrafenib alone occurred

3,500A B

3,000

2,5002,000

1,5001,000

5000

0 4

Vehicle

pERK IHC

BVD-523 (5 mg/kg, po, BID)BVD-523 (25 mg/kg, po, BID)

BVD-523 (50 mg/kg, po, BID)BVD-523 (100 mg/kg, po, BID)

28,000

24,500

21,000

17,50014,000

10,500

7,000

3,500

0

0 1 3 8

Time (hours)

BVD

-523

Tum

or c

once

ntra

tion

(ng/

mL)

DU

SP (%

Cha

nge

from

bas

elin

e)

pER

K1/

2 (%

Cha

nge

from

bas

elin

e)

16 24-100

-50

050

100150200250

300350400450

VehicleBVD-523 (25 mg/kg, po, BID)

BVD-523 (50 mg/kg, po, BID)BVD-523 (75 mg/kg, po, BID)BVD-523 (100 mg/kg, po, BID)

8 12Time (day)

16 20

1,200

Tum

or v

olum

e (m

m3 )

Tum

or v

olum

e (m

m3 )

1,000

800

600

400

200

00 4 862 12 14

Time (day)1610

C D

Figure 3.

In vivo BVD-523 antitumor activity.Effects of orally dosed BVD-523monotherapy on tumor volume inA375 (BVD-523 groups n ¼ 15,vehicle group n ¼ 10; A) andColo205 (n ¼ 12 per group; B) cellline xenografts. Mean tumor volume� SEM is plotted for each group. InA375, 50 mg/kg and 100 mg/kgBVD-523 twice daily are significantcompared with vehicle control (P ¼0.004 andP <0.001 respectively). InColo205, 50, 75, and 100 mg/kgBVD-523 twice-daily groups aresignificant compared with vehiclecontrol (all P < 0.0001). C, IHCimages for pERK in Colo205xenograft tumors at 0, 1, 3, 8, 16, and24 hours post-single 100 mg/kgdose of BVD-523.D,BVD-523 tumorconcentrations andpercent changesfrom baseline for pERK and DUSP6mRNA are plotted at 0, 1, 3, 8, 16,and 24 hours post-single dose100 mg/kg BVD-523(representative data shown).

ERK Inhibitor BVD-523 in Preclinical Cancer Models

www.aacrjournals.org Mol Cancer Ther; 16(11) November 2017 2357

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Page 8: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

relatively rapidly (Supplementary Fig. S5C). Even resistanceto combined dabrafenib and trametinib emerged before dab-rafenib plus BVD-523.

The benefit of combined BRAF and ERK inhibition may notbe fully realized in in vitro combination studies where con-centrations are not limited by tolerability. To understand thebenefit of the combination, efficacy was assessed in vivo uti-lizing xenografts of the BRAFV600E-mutant human melanomacell line A375. Because of the response of combined dabrafe-nib and BVD-523 treatment, dosing in the combination groupswas stopped on day 20 to monitor for tumor regrowth, andwas reinitiated on day 42 (Fig. 4A). Tumors were measuredtwice weekly until the study was terminated on day 45. Themedian time to endpoint (TTE) for controls was 9.2 days, andthe maximum possible tumor growth delay (TGD) of 35.8

days (end of study) was defined as 100%. Temozolomidetreatment resulted in a TGD of 1.3 days (4%) and no regres-sions. The 50- and 100-mg/kg dabrafenib monotherapiesproduced TGDs of 6.9 days (19%) and 19.3 days (54%),respectively, a significant survival benefit (P < 0.001), and 1PR in the 100 mg/kg group. The 100-mg/kg BVD-523 mono-therapy resulted in a TGD of 9.3 days (26%), a significantsurvival benefit (P < 0.001), and 2 durable complete responses.The combinations of dabrafenib with BVD-523 each producedthe maximum possible 100% TGD with statistically superioroverall survival compared with their corresponding mono-therapies (P < 0.001). The lowest dose combination produceda noteworthy 7/15 tumor-free survivors (TFS), and the 3 higherdosage combinations produced a total of 43/44 TFS, consistentwith curative or near-curative activity (Fig. 4B). In summary,

00

10

10

20

30

40

50

60

70

80

Per

cent

sur

viva

l

90

100

0 10

Groups 2–5dosing stopped

Groups 2–5dosing resumed

Time (day)3020 40 50

0

1,000

2,000

Tum

or v

olum

e (m

m3 )

3,000

A C

0 10

Time (day)

3020 40 50 60 701

10

100

1,000Tu

mor

vol

ume

(mm

3 )

10,000

G1: Vehicle: OD to end G1: Vehicle: po; BID to endG2: Dabrafenib, 50 mg/kg + BVD-523, 50 mg/kgG3: Dabrafenib, 50 mg/kg + BVD-523, 100 mg/kgG4: Dabrafenib, 100 mg/kg + BVD-523, 50 mg/kgG5: Dabrafenib, 100 mg/kg + BVD-523, 100 mg/kgG6: Dabrafenib, 50 mg/kg orally: QD to endG7: Dabrafenib, 100 mg/kg orally: QD to end

G3: Dabrafenib, 50 mg/kg orally: BID x 41G4: Dabrafenib, 25 mg/kg + BVD-523, 50 mg/kg

G5: Dabrafenib, 50 mg/kg + BVD-523, 100 mg/kgG8: BVD-523, 50 mg/kg orally: BID to endG9: BVD-523, 100 mg/kg orally: BID to end

G2: BVD-523, 100 mg/kg orally: BID x 41

G10: Temozolomide, 150 mg/kg orally: OD x 5

20 30

Time (day)40 50 0

010

10

20

30

40

50

60

70

80

Per

cent

sur

viva

l

90

100

20 30

Time (day)40 50 60 70

B D

Figure 4.

Effect of combined BVD-523 and BRAF inhibition. A375 BRAFV600E-mutant melanoma cell line xenograft model with a tumor start volume of 75–144mm3 (A andB),and in the same model with larger tumor volume (700–800 mm3) at the initiation of dosing (C and D). Mean � SEM tumor volumes (top) and Kaplan–Meier survival (bottom) are presented for each study (n ¼ 10 for each treatment group). In each experiment, combinations of dabrafenib with BVD-523 resultsin statistically superior overall survival compared with their corresponding controls (P < 0.001).

Germann et al.

Mol Cancer Ther; 16(11) November 2017 Molecular Cancer Therapeutics2358

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Page 9: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

the combination of dabrafenib with BVD-523 produced agreater number of TFS and superior efficacy to either singleagent.

On the basis of the activity of BVD-523 plus dabrafenib inA375 xenograft models with a starting tumor volume ofapproximately 75–144 mm3, a follow-up experiment was con-ducted to determine the efficacy of combination therapy in"upstaged" A375 xenografts (average tumor start volume, 700–800 mm3; Fig. 4C). The median TTE for controls was 6.2 days,establishing a maximum possible TGD of 53.8 days, which wasdefined as 100% TGD for the 60-day study. BVD-523 100 mg/kg monotherapy produced a negligible TGD (0.7 day, 1%) andno significant survival difference from controls (P > 0.05). Thedistribution of TTEs and 2 PRs suggested there might have beena subset of responders to treatment with BVD-523 alone.Dabrafenib 50-mg/kg monotherapy was efficacious, yieldinga TGD of 46.2 days (86%) and a significant survival benefitcompared with controls (P < 0.001). This group had 5 PRs and5 CRs, including 3 TFS, among the 11 evaluable mice (Fig. 4D).Both combinations of dabrafenib with BVD-523 produced themaximum 100% TGD and a significant survival benefit com-pared with controls (P < 0.001). Each combination produced100% regression responses among evaluable mice, thoughthere were distinctions in regression activity. The 25 mg/kgdabrafenib and 50 mg/kg BVD-523 combination had 2 PRs and8 CRs, with 6/10 TFS, whereas the 50-mg/kg dabrafenib and100-mg/kg BVD-523 combination had 11/11 TFS on day 60(Fig. 4D). Overall, these data support the rationale for frontlinecombination of BVD-523 with BRAF-targeted therapy inBRAFV600-mutant melanoma, and this is likely to extend toother tumor types harboring this alteration.

BVD-523 exhibits activity in in vitro models of BRAF andMEK inhibitor resistance

Emergence of resistance to BRAF and MEK inhibitors limitstheir clinical efficacy. Here, we sought to model and compare thedevelopment of resistance to BRAF (dabrafenib), MEK (trameti-nib), and ERK1/2 (BVD-523) inhibition in vitro. Over severalmonths, BRAFV600E-mutant A375 cells were cultured in progres-sively increasing concentrations of each inhibitor. Drug-resistantA375 cell lines were readily obtained following growth in highconcentrations of trametinib or dabrafenib, while developing celllines with resistance to BVD-523 proved challenging (Fig. 5A).Overall, these in vitro data suggest that at concentrations yieldingsimilar target inhibition, resistance to BVD-523 is delayed com-paredwith dabrafenib or trametinib, andmay translate to durableresponses in the clinic.

Reactivation and dependence on ERK1/2 signaling is a com-mon feature of acquired resistance to BRAF/MEK inhib-ition (26, 27); therefore, we evaluated the activity of BVD-523 in in vitro models of acquired resistance. First, a dabra-fenib and trametinib combination-resistant A375 populationwas obtained using the increased concentration method (asdescribed in detail in Supplementary Materials and Methodssection of this manuscript). The IC50-fold change from parentalA375 for dabrafenib, trametinib, and BVD-523 in the BRAF/MEK combination-resistant population is shown in Fig. 5B.BVD-523 IC50 was modestly shifted (2.5-fold), while dabrafe-nib and trametinib were more significantly shifted (8.5-foldand 13.5-fold, respectively; Fig. 5B). The cytotoxic agent pac-litaxel was tested as a control with only a modest shift in

potency observed. These data support the investigation ofBVD-523 in the setting of BRAF/MEK therapy resistance,although the mechanism of resistance in this cell populationremains to be characterized.

To further investigate the tractability of ERK1/2 inhibitionin a model with a known mechanism of BRAF inhibitorresistance, we used AAV-mediated gene targeting to generatea pair of RKO BRAFV600E-mutant cell lines isogenic for thepresence or absence of an engineered heterozygous knock-in ofMEK1Q56P-activating mutation. MEK1/2 mutations, includingMEK1Q56P, have been implicated in both single-agent BRAFand combination BRAF/MEK targeting therapy-acquired resis-tance in patients (18, 21, 34–36). Single-agent assays demon-strated that relative to the parental BRAFV600E::MEK1wt cells,the double-mutant BRAFV600E::MEK1Q56P cells displayed amarkedly reduced sensitivity to the BRAF inhibitors vemur-afenib and dabrafenib and the MEK inhibitor trametinib(Fig. 5C). In contrast, response to BVD-523 was essentiallyidentical in both the parental and MEKQ56P-mutant cells,indicating that BVD-523 is not susceptible to this mechanismof acquired resistance. These results were confirmed in 2independently derived double-mutant BRAFV600E::MEK1Q56P

cell line clones, thus validating that results were specificallyrelated to the presence of the MEK1Q56P mutation rather thanan unrelated clonal artifact. Similar results were also observedwith a second mechanistically distinct ERK1/2 inhibitor(SCH772984), supporting the expectation that these observa-tions are specifically related to mechanistic inhibition ofERK1/2 and not due to an off-target compound effect.

To further characterize the mechanistic effects of BVD-523 onMAPK pathway signaling in BRAFV600E::MEK1Q56P cell lines,protein levels were assessed by Western blot analysis (Fig. 5D).In the parental BRAFV600E RKO cells, a reduced level of pRSK1/2was observed following 4-hour treatment with BRAF (vemurafe-nib), MEK (trametinib), or ERK1/2 (BVD-523) inhibitors atpharmacologically active concentrations. In contrast, isogenicdouble-mutant BRAFV600E::MEK1Q56P cells did not exhibitreduced RSK phosphorylation following BRAF or MEK inhibitortreatment, while BVD-523 remained effective in inhibitingpRSK1/2 to a level comparable with parental RKO. Similarly,pRB is reduced, indicating G0–G1 arrest by 24 hours of BVD-523treatment in both parental RKOandBRAFV600E::MEK1Q56P.Nota-bly, as introduction of the MEK-Q56P mutation alone increasedthe level of pERK compared with the RKO parental cell line, BVD-523 treatment did not appear to significantly further boost pERKlevels in the engineered cells.

Acquired KRAS mutations are also known drivers of resis-tance to MAPK pathway inhibitors. To understand the sus-ceptibility of BVD-523 to this mechanism of resistance, anisogenic panel of clinically relevant KRAS mutations incolorectal cell line SW48 was utilized. Sensitivity to BVD-523 was compared with MEK inhibitors selumetinib (37)and trametinib (Fig. 5E and F). Sensitivity to paclitaxel wasunaltered (Supplementary Fig. S6). While several mutantKRAS alleles conferred robust to intermediate levels ofresistance to MEK inhibition, sensitivity to BVD-523 wasunaltered by the majority of alleles, and where a shift insensitivity was observed, it was not to the extent observedwith trametinib or selumetinib. Overall, these data suggestthat BVD-523 is more efficacious in this context than MEKinhibitors.

ERK Inhibitor BVD-523 in Preclinical Cancer Models

www.aacrjournals.org Mol Cancer Ther; 16(11) November 2017 2359

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Page 10: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

120

100

80

60

40

20

00 –9 –8 –7 –6 –5

Selumetinib [Log(mol/L)]

Viab

ility

(% c

ontr

ol)

120

100

80

60

40

20

00 –9 –8 –7 –6 –5

Trametinib [Log(mol/L)]

Viab

ility

(% c

ontr

ol)

120

100

80

60

40

20

00 –9 –8 –7 –6 –5

BVD-523 [Log(mol/L)]

+ KRAS (G12A)

+ KRAS (G12D)

+ KRAS (G12C)

+ KRAS (G12S)

ParentalKRAS (G12C/+)KRAS (G12A/+)KRAS (G12D/+)KRAS (G12S/+)KRAS (G12R/+)KRAS (G12V/+)KRAS (G13D/+)

+ KRAS (G12R)

+ KRAS (G13D)

+ KRAS (G12V)

Parental SW48

10.002.001.00

IC50 Foldchange

Selu

met

inib

Tram

etin

ibB

VD-5

23

24 h

4 h

4 h

Viab

ility

(% c

ontr

ol)

pERK

0

Dabrafenib(nmol/L)

Vemurafenib [Log(mol/L)]

00

20MEK1 Q56PRKO Parental

40

60

80

100

Viab

ility

(% c

ontr

ol)

120

-9 -8 -7 -6 -5 -4 00

20MEK1 Q56PRKO Parental

40

60

80

100

Viab

ility

(% c

ontr

ol)

120

-9 -8 -7 -6 -50

20MEK1 Q56P

RKO Parental

40

60

80

100

Viab

ility

(% c

ontr

ol)

120

-9-11 -10 -7 -6-8 00

20MEK1 Q56PRKO Parental

40

60

80

100

Viab

ility

(% c

ontr

ol)

120

-9 -8 -7 -6 -5

Dabrafenib [Log(mol/L)] Trametinib [Log(mol/L)]

Dabrafenib(nmol/L)

BVD-523(mmol/L)

BVD-523(mmol/L)

BVD-523 [Log(mol/L)]

Vemurafenib

00

200

400

600

Fold

cha

nge

IC50

(com

pare

d to

par

enta

l)

Fold change IC50 (compared to parental)

800

1,000A

C

D

E F

B

20 40 60 80 100Time (day)

Dabrafenib

Dabrafenib

Dabrafenib

0 2 4 6 8 10 12 14

Trametinib

Trametinib

Trametinib

BVD-523

BVD-523BVD-523

Paclitaxel

Trametinib(nmol/L)

Trametinib(nmol/L)

RKO Parental RKO (MEK1-Q56P)

1 3 10 30 100 0 1 3 10 30 100 0 0.5 2 0 0.5 20 1 3 10 30 100 0 1 3 10 30 100

pRSK

pRB (S780)

Figure 5.

BVD-523 demonstrates activity in models of resistance to BRAF/MEK inhibition. A, The emergence of resistance to BVD-523, dabrafenib, or trametinib inBRAFV600E A375 cells following exposure to increasing concentrations of drug is indicated. A strict set of "criteria" was applied (see Materials andMethods section) as to when the concentration of drug was increased. Each point on the plotted line represents a change of medium or cell split. B, In A375cells cultured to be resistant to combined BRAF (dabrafenib) þ MEK (trametinib), the fold change in IC50 compared with parental A375 of BVD-523,dabrafenib, trametinib, and paclitaxel was measured. C, Parental and BRAFV600E RKO cells engineered with endogenous heterozygous knock-in ofMEK1Q56P were incubated with BRAF (vemurafenib or dabrafenib), MEK (trametinib), or ERK (BVD-523) inhibitor and cell viability assessed as a functionof inhibitor concentration. Data presented are average � SEM of three independent titrations of each inhibitor and cell line. D, Western blot analyses ofpERK, pRSK, and pRB from cell lines treated in C. E and F, Knock-in of KRAS-mutant alleles into SW48 cell line significantly diminishes sensitivity to theMEK inhibitors trametinib and selumetinib, while comparative sensitivity to BVD-523 is retained (n ¼ 3 per titration; data are average � SEM).

Germann et al.

Mol Cancer Ther; 16(11) November 2017 Molecular Cancer Therapeutics2360

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Page 11: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

BVD-523 demonstrates in vivo activity in a BRAF inhibitor–resistant patient-derived melanoma xenograft model

To confirm and extend the antitumor effects of BVD-523observed in in vitro models of BRAF-/MEK-acquired resistance,we utilized a BRAF-resistant xenograft model derived from apatient with resistance to vemurafenib. BVD-523 was dosed byoral gavage at 100 mg/kg twice daily for 28 days, both aloneand in combination with dabrafenib at 50 mg/kg twice daily(Fig. 6). With all regimens, no significant change in bodyweight was observed (Supplementary Fig. S7). As expected,minimal antitumor activity was demonstrated for single-agentdabrafenib (22% TGI). BVD-523 activity was significant com-pared with vehicle control (P� 0.05), with a TGI of 78%. In thismodel, combining BVD-523 with dabrafenib resulted in a TGIof 76% (P � 0.05); therefore, further benefit was not gained forthe combination compared with single-agent BVD-523 in thisparticular model of BRAF-acquired resistance. Whole-exomesequencing was performed on both the patient biopsy and thePDX model. A number of alterations with unknown signifi-cance were identified in addition to the known driver mutationPIK3CA-H1047L. We speculate that the presence of this muta-tion may actually attenuate response to BVD-523, as stasisrather than regression was observed.

DiscussionBVD-523 is a potent, highly selective, reversible, small-mole-

cule ATP-competitive inhibitor of ERK1/2 with activity in in vitroand in vivo cancer models. In vitro, BVD-523 demonstrated potentinhibition against several human tumor cell lines, particularlythose harboring activating mutations in the MAPK signalingpathway, consistent with its mechanism of action. BVD-523elicited changes in downstream target and effector proteins,including inhibition of direct substrate of ERK1/2, pRSK, andtotal DUSP6 protein levels (transcriptional target of ERK1/2).These findings are in line with those of previous studies of otherERK1/2 inhibitors, which demonstrated effective suppression of

pRSK with ERK1/2 inhibition (26, 27). Interestingly, BVD-523treatment resulted in a marked increase in ERK1/2 phosphoryla-tion in vitro and in vivo. Similar to our findings, an increase inpERK1/2 has been reported with the ERK1/2 inhibitor Vx11e;conversely, pERK1/2 increases are not observed with SCH772984(26). Although differences in pERK1/2 levels were observedamong the various ERK1/2 inhibitors tested, downstream effec-tors (i.e., pRSK1 and total DUSP6) were similarly inhibited. Thesefindings suggest quantifying ERK1/2 target substrates, such aspRSK1, may serve as reliable pharmacodynamic biomarkers forBVD-523–mediated inhibitionof ERK1/2 activity. Thedifferentialeffect of BVD-523 and SCH772984 upon the levels of pERK isstriking. The significance of this to efficacy and tolerability in theclinic remains to be determined.

While BRAF (dabrafenib, vemurafenib) and MEK (trameti-nib, cobimetinib) inhibitors validate the MAPK pathway as atherapeutic target, particularly in patients with BRAFV600 muta-tions, the antitumor response is limited by the emergence ofacquired resistance and subsequent disease progression (38).Reactivation of the ERK1/2 pathway is one common conse-quence of acquired resistance mechanism. When introducedinto the BRAFV600E-mutant colorectal cell line RKO, MEKQ56P

conferred resistance to MEK and BRAF inhibition. In contrast,BVD-523 retained its potent inhibitory activity in the engi-neered MEKQ56P cell line, indicating that ERK1/2 inhibition iseffective in the setting of upstream activating alterations, whichcan arise in response to BRAF/MEK treatment. As further evi-dence of a role for BVD-523 in the context of acquired resis-tance, efficacy of BVD-523 was evident in a xenograft modelderived from a patient whose disease progressed on vemurafe-nib; the BRAF inhibitor dabrafenib was not effective in thismodel. These data support a role for targeting ERK1/2 in thesetting of BRAF/MEK resistance, and complement previouslypublished findings (26, 27).

To further characterize resistance to inhibitors of the MAPKpathway, the emergence of resistance to BVD-523 itself wasinvestigated. We found that single-agent treatment of cancer cellswith BVD-523 was durable, and it was more challenging todevelop resistance against BVD-523 than other agents targetingupstream MAPK signaling components (i.e., dabrafenib, trame-tinib). This may suggest that acquiring resistance to ERK1/2-targeting agents is harder to achieve than acquiring resistance toBRAForMEK therapy.However, in vitro studieswith other ERK1/2inhibitors have identified specific mutants in ERK1/2 that driveresistance (39, 40); these specific mutations have yet to beidentified in clinical samples from ERK1/2 inhibitor–relapsedpatients.

The potential clinical benefit of ERK1/2 inhibition with BVD-523 extends beyond the setting of BRAF/MEK therapy–resistantpatients. As ERK1/2 is a downstream master node within thisMAPK pathway, its inhibition is attractive in numerous cancersettings where tumor growth depends on MAPK signaling.Approximately 30% of all cancers harbor RAS mutations;therefore, targeting downstream ERK1/2 with BVD-523 is arational treatment approach for these cancers. Furthermore,results from a study by Hayes and colleagues indicate thatprolonged ERK1/2 inhibition in KRAS-mutant pancreatic can-cer is associated with senescent-like growth suppression (41).However, a combination approach may be required for max-imal and durable attenuation of MAPK signaling in thesetting of RAS mutations. For example, MEK inhibition in

Vehicle control2,500

2,000

1,500

1,000

500

00 8 16 24 32

Time (day)

Tum

or v

olum

e (m

m3 )

BVD-523

BVD-523/Dabrafenib

Dabrafenib

Figure 6.

BVD-523 in vivo activity in xenograft derived from a vemurafenib-relapsedpatient. Mean tumor volume (�SEM) is shown for BVD-523 100 mg/kgtwice daily (BID) alone, dabrafenib 50 mg/kg twice daily alone, andBVD-523 100 mg/kg twice daily plus dabrafenib 50 mg/kg twice daily.

ERK Inhibitor BVD-523 in Preclinical Cancer Models

www.aacrjournals.org Mol Cancer Ther; 16(11) November 2017 2361

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Page 12: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

KRAS-mutant colorectal cancer cell results in an adaptiveresponse of ErbB family activation, which dampens theresponse to MEK inhibition (42). Similar context-specific adap-tive responses may occur following ERK1/2 inhibition withBVD-523. The optimal treatment combinations for variousgenetic profiles and cancer histologies are the subject of ongo-ing research. In addition to BRAF and RAS mutations, otheralterations which drive MAPK are emerging. For example, novelRAF fusions and atypical (non-V600) BRAF mutations whichpromote RAF dimerization activate the MAPK pathway (43).BRAF inhibitors such as vemurafenib and dabrafenib whichinhibit BRAFV600-mutant monomer proteins have been shownto be inactive in atypical RAF alterations which drive MAPKsignaling in a dimerization-dependent manner (43). However,treatment with BVD-523 to target downstream ERK1/2 in thesetumors may be a novel approach to addressing this unmetmedical need; indeed patients harboring such atypical BRAFalterations are included in an ongoing clinical trial of BVD-523(NCT01781429).

In the setting of BRAFV600-mutant melanoma tumors, com-bined BRAF andMEK inhibition exemplifies how agents targetingdifferent nodes of the same pathway can improve treatmentresponse and duration. Our combination studies in BRAFV600E-mutant xenografts of human melanoma cell line A375 providesupport for combination therapy with BVD-523 and BRAF inhi-bitors. The combination demonstrated superior benefit relative tosingle-agent treatments, including results consistent with curativeresponses. The clinical efficacy and tolerability of combinedBRAF/BVD-523 therapy remains to be determined. It would notbeunreasonable to expect that aBRAF/BVD-523 combinationwillat least be comparable in efficacy to a targeted BRAF/MEK com-bination. Furthermore, the in vitro observation that acquiredresistance to BVD-523 is more challenging to achieve comparedwith other MAPK pathway inhibitors suggests that the BRAF/BVD-523 combination has the potential to provide a moredurable response.

It would be remiss to not highlight that significant progress hasalso been made using immunotherapy for the treatment ofmelanoma. The FDA has approved various immune checkpointinhibitors for the treatment of advancedmelanoma, including thecytotoxic T-lymphocyte antigen-4 targeted agent ipilimumab, andthe programmed cell death protein-1 (PD-1) inhibitors pembro-lizumab and nivolumab. Emerging data suggest MEK inhibitorsmay have benefit in combination with anti-PD-L1 therapy ate-zolizumab (44). Combining BVD-523 with such immunothera-pies is an attractive therapeutic option; further investigation iswarranted to explore dosing schedules and to assess whethersynergistic response can be achieved.

On the basis of the preclinical data, BVD-523 may holdpromise for treatment of patients with malignancies dependent

on MAPK signaling, including those whose tumors haveacquired resistance to other treatments. Phase I clinical studiesof BVD-523 are ongoing (NCT01781429, NCT02296242, andNCT02608229).

Disclosure of Potential Conflicts of InterestU.A. Germann has ownership interest (including patents) in Vertex

Pharmaceuticals Incorporated. J.J. Roix is an associate at BioMed ValleyDiscoveries. D.A. Sorrell has ownership interest (including patents) inHorizon Discovery. M. Fitzgibbon is a senior research scientist and hasownership interest (including patents) in Vertex Pharmaceuticals. Nopotential conflicts of interest were disclosed by the other authors.

Authors' ContributionsConception and design: U.A. Germann, W. Markland, R.R. Hoover,A.M. Aronov, J.J. Roix, M. Hale, R. Samadani, G. DeCrescenzo, S. Saha,D.J. WelschDevelopment of methodology: B.F. Furey, W. Markland, R.R. Hoover,J.J. Roix, D.A. Sorrell, R. Samadani, G. DeCrescenzo, M. Namchuk, S. Saha,D.J. WelschAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): B.F. Furey, W. Markland, R.R. Hoover, D.M. Boucher,D.A. Sorrell, M. Fitzgibbon, P. Shapiro, M.J. Wick, R. Samadani, K. Meshaw,C.M. Emery, S. Saha, D.J. WelschAnalysis and interpretation of data (e.g., statistical analysis, biostatis-tics, computational analysis): U.A. Germann, B.F. Furey, W. Markland,R.R. Hoover, J.J. Roix, D.M. Boucher, D.A. Sorrell, P. Shapiro, R. Sama-dani, A. Groover, G. DeCrescenzo, M. Namchuk, C.M. Emery, S. Saha,D.J. WelschWriting, review, and/or revision of themanuscript:U.A. Germann, B.F. Furey,W. Markland, A.M. Aronov, J.J. Roix, M. Hale, D.M. Boucher, D.A. Sorrell,G. Martinez-Botella, M.J. Wick, R. Samadani, A. Groover, G. DeCrescenzo,M. Namchuk, C.M. Emery, S. Saha, D.J. WelschAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): J.J. Roix,M. Fitzgibbon, A. Groover, C.M. Emery,S. Saha, D.J. WelschStudy supervision: U.A. Germann, J.J. Roix, D.A. Sorrell, M. Namchuk,C.M. Emery, S. Saha, D.J. WelschOther (his group while at Vertex designed and made the compound in thestudy and holds patents on the chemical matter): M. Hale

AcknowledgmentsThe authors thank Corinne Ramos and Nicholas Hoke for performing the

large-scale reverse phase protein array. They also thank SharonMaguire andPaulA. Russell for assay support and Amy Smith and Mark Stockdale for theconstruction of the MEK1Q56P cell line.

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received May 19, 2017; revised August 2, 2017; accepted August 23, 2017;published OnlineFirst September 22, 2017.

References1. Kanda M, Matthaei H, Wu J, Hong SM, Yu J, Borges M, et al. Presence of

somatic mutations in most early-stage pancreatic intraepithelial neo-plasia. Gastroenterology 2012;142:730–3.

2. Arrington AK, Heinrich EL, Lee W, Duldulao M, Patel S, Sanchez J, et al.Prognostic and predictive roles of KRAS mutation in colorectal cancer.Int J Mol Sci 2012;13:12153–68.

3. Hall RD, Kudchadkar RR. BRAF mutations: signaling, epidemiology, andclinical experience in multiple malignancies. Cancer Control 2014;21:221–30.

4. Ojesina AI, Lichtenstein L, Freeman SS, Pedamallu CS, Imaz-Rosshandler I,Pugh TJ, et al. Landscape of genomic alterations in cervical carcinomas.Nature 2014;506:371–5.

5. Arcila ME, Drilon A, Sylvester BE, Lovly CM, Borsu L, Reva B, et al.MAP2K1 (MEK1) mutations define a distinct subset of lungadenocarcinoma associated with smoking. Clin Cancer Res 2015;21:1935–43.

6. Groenendijk FH, Bernards R. Drug resistance to targeted therapies: deja vuall over again. Mol Oncol 2014;8:1067–83.

Germann et al.

Mol Cancer Ther; 16(11) November 2017 Molecular Cancer Therapeutics2362

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Page 13: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

7. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al.Improved survival with vemurafenib in melanoma with BRAF V600Emutation. N Engl J Med 2011;364:2507–16.

8. Hauschild A, Grob JJ, Demidov LV, Jouary T, Gutzmer R, MillwardM, et al.Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012;380:358–65.

9. Queirolo P, Picasso V, Spagnolo F. Combined BRAF and MEK inhibitionfor the treatment of BRAF-mutatedmetastatic melanoma. Cancer Treat Rev2015;41:519–26.

10. Massey PR, Prasad V, Figg WD, Fojo T. Multiplying therapies and reducingtoxicity in metastatic melanoma. Cancer Biol Ther 2015;16:1014–8.

11. Robert C, Karaszewska B, Schachter J, Rutkowski P, Mackiewicz A, Stroia-kovski D, et al. Improved overall survival in melanoma with combineddabrafenib and trametinib. N Engl J Med 2015;372:30–9.

12. LongGV, StroyakovskiyD,GogasH, Levchenko E, deBraud F, Larkin J, et al.Dabrafenib and trametinib versus dabrafenib and placebo for Val600BRAF-mutant melanoma: a multicentre, double-blind, phase 3 rando-mised controlled trial. Lancet 2015;386:444–51.

13. Larkin J, Ascierto PA, Dreno B, Atkinson V, Liszkay G, Maio M, et al.Combined vemurafenib and cobimetinib in BRAF-mutated melanoma.N Engl J Med 2014;371:1867–76.

14. LongGV, StroyakovskiyD,GogasH, Levchenko E, deBraud F, Larkin J, et al.Combined BRAF and MEK inhibition versus BRAF inhibition alone inmelanoma. N Engl J Med 2014;371:1877–88.

15. Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, et al.Combined BRAF and MEK inhibition in melanoma with BRAF V600mutations. N Engl J Med 2012;367:1694–703.

16. Poulikakos PI, Persaud Y, JanakiramanM, Kong X, Ng C,Moriceau G, et al.RAF inhibitor resistance is mediated by dimerization of aberrantly splicedBRAF(V600E). Nature 2011;480:387–90.

17. Corcoran RB, Dias-Santagata D, Bergethon K, Iafrate AJ, Settleman J,Engelman JA. BRAF gene amplification can promote acquired resistanceto MEK inhibitors in cancer cells harboring the BRAF V600E mutation.Sci Signal 2010;3:ra84.

18. Wagle N, Emery C, Berger MF, Davis MJ, Sawyer A, Pochanard P, et al.Dissecting therapeutic resistance to RAF inhibition inmelanoma by tumorgenomic profiling. J Clin Oncol 2011;29:3085–96.

19. Nazarian R, Shi H, Wang Q, Kong X, Koya RC, Lee H, et al. Melanomasacquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregula-tion. Nature 2010;468:973–7.

20. Shi H, Hugo W, Kong X, Hong A, Koya RC, Moriceau G, et al. Acquiredresistance and clonal evolution in melanoma during BRAF inhibitortherapy. Cancer Discov 2014;4:80–93.

21. WagleN, VanAllen EM, TreacyDJ, FrederickDT, Cooper ZA, Taylor-WeinerA, et al. MAP kinase pathway alterations in BRAF-mutant melanomapatients with acquired resistance to combined RAF/MEK inhibition.Cancer Discov 2014;4:61–8.

22. Long GV, Fung C, Menzies AM, Pupo GM, Carlino MS, Hyman J, et al.Increased MAPK reactivation in early resistance to dabrafenib/trameti-nib combination therapy of BRAF-mutant metastatic melanoma.Nat Commun 2014;5:5694.

23. Rizos H,Menzies AM, Pupo GM, CarlinoMS, Fung C, Hyman J, et al. BRAFinhibitor resistance mechanisms in metastatic melanoma: spectrum andclinical impact. Clin Cancer Res 2014;20:1965–77.

24. Moriceau G, Hugo W, Hong A, Shi H, Kong X, Yu CC, et al. Tunable-combinatorial mechanisms of acquired resistance limit the efficacy ofBRAF/MEK cotargeting but result in melanoma drug addiction. CancerCell 2015;27:240–56.

25. Johannessen CM, Johnson LA, Piccioni F, Townes A, Frederick DT, Dona-hue MK, et al. A melanocyte lineage program confers resistance to MAPkinase pathway inhibition. Nature 2013;504:138–42.

26. Morris EJ, Jha S, Restaino CR, Dayananth P, Zhu H, Cooper A, et al.Discovery of a novel ERK inhibitor with activity in models of acquiredresistance to BRAF and MEK inhibitors. Cancer Discov 2013;3:742–50.

27. Hatzivassiliou G, Liu B, O'Brien C, Spoerke JM, Hoeflich KP, Haverty PM,et al. ERK inhibition overcomes acquired resistance to MEK inhibitors.Mol Cancer Ther 2012;11:1143–54.

28. Carlino MS, Long GV, Kefford RF, Rizos H. Targeting oncogenic BRAFand aberrant MAPK activation in the treatment of cutaneous melanoma.Crit Rev Oncol Hematol 2015;96:385–98.

29. Aronov AM, Tang Q, Martinez-Botella G, Bemis GW, Cao J, Chen G, et al.Structure-guided design of potent and selective pyrimidylpyrrole inhibi-tors of extracellular signal-regulated kinase (ERK) using conformationalcontrol. J Med Chem 2009;52:6362–8.

30. Rudolph J, Xiao Y, Pardi A, Ahn NG. Slow inhibition and conformationselective properties of extracellular signal-regulated kinase 1 and 2 inhi-bitors. Biochemistry 2015;54:22–31.

31. Aronov AM, Baker C, Bemis GW, Cao J, Chen G, Ford PJ, et al. Flippedout: structure-guided design of selective pyrazolylpyrrole ERK inhibitors.J Med Chem 2007;50:1280–7.

32. FedorovO,Niesen FH, Knapp S. Kinase inhibitor selectivity profiling usingdifferential scanning fluorimetry. Methods Mol Biol 2012;795:109–18.

33. Wainstein E, Seger R. The dynamic subcellular localization of ERK:mechanisms of translocation and role in various organelles. Curr OpinCell Biol 2016;39:15–20.

34. Trunzer K, Pavlick AC, Schuchter L, Gonzalez R, McArthur GA, HutsonTE, et al. Pharmacodynamic effects and mechanisms of resistance tovemurafenib in patients with metastatic melanoma. J Clin Oncol 2013;31:1767–74.

35. Emery CM, Vijayendran KG, Zipser MC, Sawyer AM, Niu L, Kim JJ, et al.MEK1 mutations confer resistance to MEK and B-RAF inhibition.Proc Natl Acad Sci U S A 2009;106:20411–6.

36. JohnsonDB,Menzies AM, Zimmer L, Eroglu Z, Ye F, Zhao S, et al. AcquiredBRAF inhibitor resistance: a multicentermeta-analysis of the spectrum andfrequencies, clinical behaviour, and phenotypic associations of resistancemechanisms. Eur J Cancer 2015;51:2792–9.

37. Yeh TC, Marsh V, Bernat BA, Ballard J, Colwell H, Evans RJ, et al. Biologicalcharacterization of ARRY-142886 (AZD6244), a potent, highly selectivemitogen-activated protein kinase kinase 1/2 inhibitor. Clin Cancer Res2007;13:1576–83.

38. Corcoran RB, Settleman J, Engelman JA. Potential therapeutic strategies toovercome acquired resistance to BRAF or MEK inhibitors in BRAF mutantcancers. Oncotarget 2011;2:336–46.

39. Jha S, Morris EJ, Hruza A, Mansueto MS, Schroeder GK, Arbanas J, et al.Dissecting therapeutic resistance to ERK inhibition. Mol Cancer Ther2016;15:548–59.

40. Goetz EM, Ghandi M, Treacy DJ, Wagle N, Garraway LA. ERK mutationsconfer resistance to mitogen-activated protein kinase pathway inhibitors.Cancer Res 2014;74:7079–89.

41. Hayes TK, Neel NF, Hu C, Gautam P, Chenard M, Long B, et al. Long-termERK inhibition in KRAS-mutant pancreatic cancer is associated with MYCdegradation and senescence-like growth suppression. Cancer Cell 2016;29:75–89.

42. Sun C, Hobor S, Bertotti A, Zecchin D, Huang S, Galimi F, et al. Intrinsicresistance to MEK inhibition in KRAS mutant lung and colon cancerthrough transcriptional induction of ERBB3. Cell Rep 2014;7:86–93.

43. Yao Z, TorresNM, TaoA,Gao Y, Luo L, LiQ, et al. BRAFmutants evade ERK-dependent feedback by different mechanisms that determine their sensi-tivity to pharmacologic inhibition. Cancer Cell 2015;28:370–83.

44. Miller WH, Kim TM, Lee CB, Flaherty KT, Reddy S, Jamal R, et al. Atezo-lizumab (A) þ cobimetinib (C) in metastatic melanoma (mel): updatedsafety and clinical activity. J Clin Oncol 35:15s, 2017 (suppl; abstr 3057).

www.aacrjournals.org Mol Cancer Ther; 16(11) November 2017 2363

ERK Inhibitor BVD-523 in Preclinical Cancer Models

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456

Page 14: Targeting the MAPK Signaling Pathway in Cancer: Promising … · Targeting the MAPK Signaling Pathway in Cancer: ... potential for ERK inhibitors expands beyond acquired resis-tance

2017;16:2351-2363. Published OnlineFirst September 22, 2017.Mol Cancer Ther   Ursula A. Germann, Brinley F. Furey, William Markland, et al.   BVD-523 (Ulixertinib)Preclinical Activity with the Novel Selective ERK1/2 Inhibitor Targeting the MAPK Signaling Pathway in Cancer: Promising

  Updated version

  10.1158/1535-7163.MCT-17-0456doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2017/09/22/1535-7163.MCT-17-0456.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/16/11/2351.full#ref-list-1

This article cites 43 articles, 13 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/16/11/2351.full#related-urls

This article has been cited by 12 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://mct.aacrjournals.org/content/16/11/2351To request permission to re-use all or part of this article, use this link

on October 3, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0456