Top Banner
Small Molecule Therapeutics A Novel RAF Kinase Inhibitor with DFG-OutBinding Mode: High Efcacy in BRAF-Mutant Tumor Xenograft Models in the Absence of Normal Tissue Hyperproliferation Irene C. Waizenegger 1 , Anke Baum 1 , Steffen Steurer 2 , Heinz Stadtmuller 2 , Gerd Bader 2 , Otmar Schaaf 3 , Pilar Garin-Chesa 1 , Andreas Schlattl 1 , Norbert Schweifer 1 , Christian Haslinger 1 , Florian Colbatzky 4 , Sien Mousa 4 , Arno Kalkuhl 4 , Norbert Kraut 1 , and Gunther R. Adolf 1 Abstract BI 882370 is a highly potent and selective RAF inhibitor that binds to the DFG-out (inactive) conformation of the BRAF kinase. The compound inhibited proliferation of human BRAFmutant melanoma cells with 100 higher potency (110 nmol/L) than vemurafenib, whereas wild-type cells were not affected at 1,000 nmol/L. BI 882370 administered orally was efcacious in multi- ple mouse models of BRAF-mutant melanomas and colorectal carcinomas, and at 25 mg/kg twice daily showed superior efcacy compared with vemurafenib, dabrafenib, or trametinib (dosed to provide exposures reached in patients). To model drug resistance, A375 melanomabearing mice were initially treated with vemur- afenib; all tumors responded with regression, but the majority subsequently resumed growth. Trametinib did not show any efcacy in this progressing population. BI 882370 induced tumor regression; however, resistance developed within 3 weeks. BI 882370 in combination with trametinib resulted in more pronounced regressions, and resistance was not observed during 5 weeks of second-line therapy. Importantly, mice treated with BI 882370 did not show any body weight loss or clinical signs of intolerability, and no pathologic changes were observed in several major organs investigated, including skin. Furthermore, a pilot study in rats (up to 60 mg/kg daily for 2 weeks) indicated lack of toxicity in terms of clinical chemistry, hematology, pathology, and toxicogenomics. Our results indicate the feasibility of developing novel compounds that provide an improved therapeutic window compared with rst-generation BRAF inhibitors, resulting in more pronounced and long-lasting pathway suppression and thus improved efcacy. Mol Cancer Ther; 15(3); 35465. Ó2016 AACR. Introduction In multiple normal tissues, the MAPK signaling pathway transmits extracellular signals via tyrosine kinase receptors, RAS, RAF, MEK, and ERK proteins to regulate diverse cellular functions, including survival and proliferation. Hyperactiva- tion of this pathway due to increased transducer expression (based on focal gene amplication) or higher intrinsic activity (due to gain-of-function point mutations, deletions, or chro- mosomal rearrangements) contributes to the pathogenesis of a wide range of solid tumors as well as hematologic malignan- cies, with mutations in RAS GTPases as the most frequent drivers of malignant proliferation. Genetic alterations of mem- bers of the RAF family of serine/threonine kinases are likewise involved in multiple types of cancers. Whereas mutations in ARAF and CRAF are very rare, BRAF mutations are found with widely varying frequency across multiple cancer types, ranging from about 5% in colorectal and nonsmall cell lung carcino- ma (NSCLC) to 50% in malignant melanoma and close to 100% in hairy cell leukemia. Among a variety of BRAF altera- tions identied to date, point mutations affecting amino acid position 600 are by far the most frequent ones (V600E or V600K; melanoma, >90%); the oncogenic potential of these and the dependence of BRAF-mutant melanomas on the activ- ity of the mutant protein was clearly demonstrated in a variety of in vitro and animal disease models. Subsequently, multiple drug discovery programs to synthesize potent and drug-like BRAF inhibitors were initiated, and the rst compound, vemur- afenib, was approved for treatment of melanoma patients in 2011, only nine years after the initial report on BRAF mutations in cancer (1, 2). During clinical development of vemurafenib and, subsequent- ly, of further BRAF inhibitors such as dabrafenib, two main issues limiting the utility of this class of compounds became apparent. First, treatment resulted in the progression of skin lesions 1 Department of Pharmacology and Translational Research, Boehrin- ger Ingelheim RCV GmbH & Co KG, Vienna, Austria. 2 Department of Medicinal Chemistry, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria. 3 Department of Discovery ADME, Boehringer Ingel- heim RCV GmbH & Co KG, Vienna, Austria. 4 Department of Non- clinical Drug Safety, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Current address for S. Mousa: Janssen Pharmaceutica NV, Belgium. Prior presentation: The authors declare that some of the results were presented at the AACR meeting in 2013. Note added in proof: The crystal structure of the dabrafenibBRAF kinase complex was recently published by Zhang and colleagues in ref. 42. Corresponding Author: Irene C. Waizenegger, Boehringer Ingelheim RCV GmbH & Co KG, Dr. Boehringer-Gasse 5-11, Vienna 1121, Austria. Phone: 43-1-80105-2787; Fax: 43-1-80105-32787; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-15-0617 Ó2016 American Association for Cancer Research. Molecular Cancer Therapeutics Mol Cancer Ther; 15(3) March 2016 354 Downloaded from http://aacrjournals.org/mct/article-pdf/15/3/354/1850899/354.pdf by guest on 01 October 2022
12

Binding Mode: High Efficacy in BRAF-Mutant Tumor

May 12, 2023

Download

Documents

Khang Minh
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Binding Mode: High Efficacy in BRAF-Mutant Tumor

Small Molecule Therapeutics

A Novel RAF Kinase Inhibitor with DFG-Out–Binding Mode: High Efficacy in BRAF-MutantTumorXenograftModels in theAbsenceofNormalTissue HyperproliferationIrene C.Waizenegger1, Anke Baum1, Steffen Steurer2, Heinz Stadtm€uller2, Gerd Bader2,Otmar Schaaf3, Pilar Garin-Chesa1, Andreas Schlattl1, Norbert Schweifer1,Christian Haslinger1, Florian Colbatzky4, Sien Mousa4, Arno Kalkuhl4, Norbert Kraut1, andG€unther R. Adolf1

Abstract

BI 882370 is a highly potent and selective RAF inhibitor thatbinds to theDFG-out (inactive) conformation of the BRAF kinase.The compound inhibited proliferation of human BRAF–mutantmelanoma cells with 100� higher potency (1–10 nmol/L) thanvemurafenib, whereas wild-type cells were not affected at 1,000nmol/L. BI 882370 administered orally was efficacious in multi-ple mouse models of BRAF-mutant melanomas and colorectalcarcinomas, and at 25 mg/kg twice daily showed superior efficacycompared with vemurafenib, dabrafenib, or trametinib (dosed toprovide exposures reached in patients). Tomodel drug resistance,A375 melanoma–bearing mice were initially treated with vemur-afenib; all tumors responded with regression, but the majoritysubsequently resumed growth. Trametinib did not show anyefficacy in this progressing population. BI 882370 induced tumor

regression; however, resistance developed within 3 weeks.BI 882370 in combination with trametinib resulted in morepronounced regressions, and resistance was not observed during5 weeks of second-line therapy. Importantly, mice treated withBI 882370 did not show any body weight loss or clinical signs ofintolerability, and no pathologic changeswere observed in severalmajor organs investigated, including skin. Furthermore, a pilotstudy in rats (up to 60 mg/kg daily for 2 weeks) indicated lack oftoxicity in termsof clinical chemistry, hematology, pathology, andtoxicogenomics. Our results indicate the feasibility of developingnovel compounds that provide an improved therapeutic windowcomparedwith first-generation BRAF inhibitors, resulting inmorepronounced and long-lasting pathway suppression and thusimproved efficacy. Mol Cancer Ther; 15(3); 354–65. �2016 AACR.

IntroductionIn multiple normal tissues, the MAPK signaling pathway

transmits extracellular signals via tyrosine kinase receptors,RAS, RAF, MEK, and ERK proteins to regulate diverse cellularfunctions, including survival and proliferation. Hyperactiva-tion of this pathway due to increased transducer expression(based on focal gene amplification) or higher intrinsic activity

(due to gain-of-function point mutations, deletions, or chro-mosomal rearrangements) contributes to the pathogenesis of awide range of solid tumors as well as hematologic malignan-cies, with mutations in RAS GTPases as the most frequentdrivers of malignant proliferation. Genetic alterations of mem-bers of the RAF family of serine/threonine kinases are likewiseinvolved in multiple types of cancers. Whereas mutations inARAF and CRAF are very rare, BRAF mutations are found withwidely varying frequency across multiple cancer types, rangingfrom about 5% in colorectal and non–small cell lung carcino-ma (NSCLC) to 50% in malignant melanoma and close to100% in hairy cell leukemia. Among a variety of BRAF altera-tions identified to date, point mutations affecting amino acidposition 600 are by far the most frequent ones (V600E orV600K; melanoma, >90%); the oncogenic potential of theseand the dependence of BRAF-mutant melanomas on the activ-ity of the mutant protein was clearly demonstrated in a varietyof in vitro and animal disease models. Subsequently, multipledrug discovery programs to synthesize potent and drug-likeBRAF inhibitors were initiated, and the first compound, vemur-afenib, was approved for treatment of melanoma patients in2011, only nine years after the initial report on BRAF mutationsin cancer (1, 2).

During clinical development of vemurafenib and, subsequent-ly, of further BRAF inhibitors such as dabrafenib, twomain issueslimiting the utility of this class of compounds became apparent.First, treatment resulted in the progression of skin lesions

1Department of Pharmacology and Translational Research, Boehrin-ger Ingelheim RCV GmbH & Co KG, Vienna, Austria. 2Department ofMedicinal Chemistry, Boehringer Ingelheim RCV GmbH & Co KG,Vienna, Austria. 3Department of Discovery ADME, Boehringer Ingel-heim RCV GmbH & Co KG, Vienna, Austria. 4Department of Non-clinical Drug Safety, Boehringer Ingelheim Pharma GmbH & Co. KG,Biberach an der Riss, Germany.

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

Current address for S. Mousa: Janssen Pharmaceutica NV, Belgium.

Prior presentation: The authors declare that some of the results were presentedat the AACR meeting in 2013.

Note added in proof: The crystal structure of the dabrafenib–BRAF kinasecomplex was recently published by Zhang and colleagues in ref. 42.

CorrespondingAuthor: IreneC.Waizenegger,Boehringer IngelheimRCVGmbH&Co KG, Dr. Boehringer-Gasse 5-11, Vienna 1121, Austria. Phone: 43-1-80105-2787;Fax: 43-1-80105-32787; E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-15-0617

�2016 American Association for Cancer Research.

MolecularCancerTherapeutics

Mol Cancer Ther; 15(3) March 2016354

Dow

nloaded from http://aacrjournals.org/m

ct/article-pdf/15/3/354/1850899/354.pdf by guest on 01 October 2022

Page 2: Binding Mode: High Efficacy in BRAF-Mutant Tumor

with preexisting RAS mutations, in particular of squamous cellcarcinomas and keratoacanthomas, in a subset of patients; mech-anistically, these adverse events involve a paradoxical, inhibitor-induced activation ofwild-type (WT) B/CRAF signaling, increasedpathway output, and thus increased proliferation. Clinically,these skin malignancies are not of major concern, as the numberof lesions per patient is usually small, and the tumors can beremoved surgically (3). Initial concerns that BRAF inhibitorsmight also promote progression of other RAS-mutant cancershave not been substantiated to date, although a few individualcases have been reported (4). More importantly, the benefitprovided by BRAF inhibitors to melanoma patients was foundto be quite limited in duration. Following initial, often dra-matic regression of lesions, secondary resistance develops in amajority of patients over the course of a few months oftreatment. More recent clinical trials have shown that simul-taneous treatment with a BRAF (dabrafenib) and a MEK inhib-itor (trametinib) provides superior outcomes compared withsingle-agent therapy in terms of higher response rates andprolonged progression-free and overall survival, and theBRAF–MEK inhibitor combination is now the standard of carefor BRAFV600-mutant melanoma (5, 6). Nevertheless, theimprovement achieved by combination therapy appears mod-est, and a high medical need for effective treatment of relapsingpatients remains. Recently, Girotti and colleagues reported onnovel RAF inhibitors that showed efficacy in preclinical mela-noma models resistant to the vemurafenib analogue PLX4720(7). Interestingly, these compounds, like BI 882370, bind to theinactive conformation of BRAF; in addition to RAF kinases,however, they also inhibit SRC family kinases with similarpotency. Although SRC inhibition may well contribute toefficacy, the therapeutic window that can be achieved bycombined RAF/SRC inhibition remains to be determined.

In the course of a discovery program aiming for the identifi-cation of improved BRAF inhibitors, we noted that various com-pounds from the same chemical class displayed widely varyingtherapeutic windows in mice. Whereas some compounds, eventhough reasonably potent and selective BRAF inhibitors in in vitroexperiments and efficacious in tumor xenograft models, inducedhyperproliferation of various stratified and transitional epitheliain mice, in accordance with previous reports (8–10), othersachieved similar efficacy in the absence of detectable histologicchanges. Although some of these differences may be due to off-target effects, we reasoned that different modes of drug–targetinteraction may play a fundamental role in determining thetherapeutic index and thus the extent and duration of tumorresponses. We here present the chemical structure, target bindingmode, and preclinical pharmacologic profile of a highly potentand selective RAF inhibitor that displays favorable characteristicsin models of BRAF-mutant cancers.

Materials and MethodsCompounds

Compounds were synthesized and/or formulated according topublished procedures: BI 882370 (¼ I-42; ref. 11); afatinib (BIBW2992; ref. 12); vemurafenib (example P-0956; refs. 13, 14); dab-rafenib (example 58a; ref. 15); trametinib (example 4-1; ref. 16);and GDC-0879 (example 110; ref. 17). In addition, vemurafenib(Zelboraf), cetuximab (Erbitux), and irinotecan (Campto) werepurchased from Roche, Merck, and Pfizer, respectively.

Structural researchA histidine-tagged mutant form of the human BRAF kinase

domain (18) expressed in E. coli was purified by affinity chroma-tography onNi Sepharose, ion exchange chromatography and gelfiltration. Crystals were grown by the vapor diffusion method.Datasetswere collected at 100Kon theX60SAbeamline at SLS andprocessed with XDS (19). Structures were solved by molecularreplacement using a published BRAF structure and refined byiterative cycles of model building using COOT (20) and refine-ment by REFMAC (21). The structures were deposited at RCSB[accession codes 5CSX (BI 882370) and 5CSW (dabrafenib)]. Allcrystallographic work was done by Proteros Biostructures GmbH(more details are available in the Supplementary Materials andMethods).

Kinase assaysKD values were determined by Proteros Biostructures GmbH,

following published methods (22).IC50 values for RAF kinases were determined by Invitrogen in

a RAF–MEK–ERK cascade assay measuring the activity of ERKon a fluoro-probe (Z0-LYTE technology). Selectivity was deter-mined on the Invitrogen kinase panel at a concentration of1 mmol/L.

Cell lines and cellular assaysCell lines were cultured according to the manufacturer's

instruction and authenticated by short tandem repeat (STR)analysis at Boehringer Ingelheim on the dates indicated. A101(obtained, October 9, 2006; STR, September 15, 2015), A375(obtained, July 7, 2006; STR, April 14, 2012), C32 (obtained,January 1, 2007; STR, April 28, 2014), COLO 205 (obtained,February 21, 2003; STR, May 4, 2011), G-361 (obtained,October 16, 2006; STR, September 9, 2015), HCT-116(obtained, June 17, 2011; STR, April 17, 2012), HT-29(obtained, January 24, 2012; STR, February 28, 2013), LS411N(obtained, August 24, 2006; STR, June 16, 2015), NCI-H1395(obtained, July 20, 2011; STR, March 15, 2012), NCI-H1666(obtained, December 13, 2004; STR, September 15, 2015),NCI-H1755 (obtained, July 20, 2011; STR, March 15, 2012),and SK-MEL-28 (obtained, October 15, 2007; STR, December11, 2014) were from the ATCC. CAL-12T (obtained, August 21,2006; STR, September 15, 2015) were from the DSMZ, andHCC-364 (obtained, June 22, 2013; STR, August 12, 2013)were from University of Texas Southwestern Medical Center(Dallas, TX). BRO cell line was a kind gift from Prof. Piet Borst(Amsterdam Free University, Amsterdam, the Netherlands) in1995; STR data for BRO cells are from September 9, 2014, nopublished reference STR available.

Proliferation assays. Cell lines were analyzed by alamarBlue(AbD Serotec), except HT-29 and COLO 205, which were testedby 3H-thymidine incorporation assay. In brief, cells were seed-ed into 96-well plates, and the next day, "time zero" plates wereread, or serially diluted compounds were added to test plates(final DMSO concentration 1%; compounds were usually test-ed in duplicates). After 3 days of compound treatment, theantiproliferative effect was determined by measuring the met-abolic activity of remaining cells using alamarBlue. GI50 orEC50 values were calculated by nonlinear regression curve fit(sigmoidal dose response with variable hill slope). To deter-mine the capacity of cells to incorporate 3H-thymidine, 0.1 mCi/

A Novel RAF Inhibitor with DFG-Out–Binding Mode

www.aacrjournals.org Mol Cancer Ther; 15(3) March 2016 355

Dow

nloaded from http://aacrjournals.org/m

ct/article-pdf/15/3/354/1850899/354.pdf by guest on 01 October 2022

Page 3: Binding Mode: High Efficacy in BRAF-Mutant Tumor

well 3H-thymidine (Amersham TRK296) was added after 3 daysof compound treatment. After additional 16 hours of incuba-tion, plates were frozen (�20�C) to lyse cells. Cell lysates wereprepared and harvested on MultiScreen Harvest Plates FC(Merck Millipore, #MAHFC1H60); 25 mL MicroScint/well wasadded to dried filter plates, and the plates were measured with aWallac TriLux 1450 MicroBeta Counter. EC50 values werecalculated as described previously. In assays performed atEurofins Panlabs, cells were incubated for 3 days, fixed, andstained with DAPI nuclear dye. Cell counts were determined byfluorescence imaging, and EC50 values were determined bycomparing cultures treated for 3 days and cultures fixed at thebeginning of the incubation period.

Phospho-ERK assay.A375 (10,000 cells/well) or SK-MEL-28 (7,500cells/well) were plated in 96-well flat bottom Costar plates(#3598), and the next day, cells were treated with compounds for2 hours, fixed with 4% formaldehyde, permeabilized and washedseveral times with 0.1% Triton X-100/PBS (20 minutes), blockedwith 5% nonfat dry milk in TBST, and incubated with anti–phospho-ERK1/2 (Sigma #M8159; 1:500) overnight. After severalwashing steps with 0.1% Tween-20/PBS, cells were stained withHRPO-coupled rabbit anti-mouse IgG (Dako, #P0161; 1:1,000; 1hour), washed, and stained with TMB substrate (Bender MedSys-tems, #BMS406); the reactionwas stopped after 5 to 30minutes by1 mol/L phosphoric acid. The plates were measured at 450 nmusing aWallacVictor 2. The EC50was calculated asdescribed above.

Figure 1.Structure of BI 882370, dabrafenib, and their co-crystalswith BRAFWTkinase. A, chemical structure of BI 882370 (propane-1-sulfonic acid (3-{5-[(1-ethyl-piperidin-4-yl)-methyl-amino]-3-pyrimidin-5-yl-pyrrolo[3,2-b]pyridin-1-yl}-2,4-difluoro-phenyl)-amide, molecular weight 570 g/mol), and schematic representationof its binding mode. B, chemical structure of dabrafenib and schematic representation of its binding mode. C, enlarged view of the co-crystal of BI 882370with BRAF showing the ATP pocket of the kinase. BI 882370 is shown in green, BRAF in light blue, Phe595 from the DFG motif in magenta, and Phe583 in yellow.D, enlarged view of the co-crystal of dabrafenib with BRAF, showing the ATP pocket of the kinase. Dabrafenib is shown in green, BRAF kinase in light blue,Phe595 from the DFG motif in magenta, and Phe583 in yellow.

Waizenegger et al.

Mol Cancer Ther; 15(3) March 2016 Molecular Cancer Therapeutics356

Dow

nloaded from http://aacrjournals.org/m

ct/article-pdf/15/3/354/1850899/354.pdf by guest on 01 October 2022

Page 4: Binding Mode: High Efficacy in BRAF-Mutant Tumor

To address the reversibility of phospho-ERK modulation cellswere treated with compounds for 2 hours, washed 5 times withmedium, and incubated further until they were processed asdescribed above.

Immunoblots. Lysates were prepared from cells treated for 2 or24 h. Harvested cells were lysed in 20 mmol/L Tris pH 7.7, 100mmol/LNaCl, 20mmol/L b-glycerophosphate, 5mmol/LMgCl2,1mmol/LNaF, 0.1%Triton X-100, 5%glycerol plus freshly added1 mmol/L DTT, 0.1 mmol/L okadaic acid, 1 mmol/L sodiumvanadate, and cOmplete Mini Protease Inhibitor Tablets (Roche,#11836170001). For SDS-PAGE, equal amounts of protein/lanewere loaded. After gel transfer onto PDV membrane, immuno-blots were performed using the following antibodies: phospho-MEK 1/2 (Cell Signaling Technology #9121; 1:1,000), MEK1 (BDBiosciences #610121; 1:10,000), phospho-ERK (Sigma #8159;1:30,000), ERK1/2 (Upstate Biotechnology #06-182; 1:5,000),cyclinD1/2 (Upstate Biotechnology #05-362; 1:5,000), Kip1/p27(BD Biosciences #610241; 1:5,000), and a-tubulin (Sigma#T6199; 1:4,000). Secondary antibodies were from DAKO(#P0448 or #P0447). ECL detection was performed.

Coimmunoprecipitation.Cells were treated for 1 hour with variouscompounds prior to harvesting. NP40 lysates and CRAF immu-noprecipitates were prepared according to ref. 23 using rabbitanti-CRAF (Santa Cruz Biotechnology, sc-227) and analyzed byimmunoblot using mouse anti-BRAF (Santa Cruz Biotechnology,sc-5284).

Animal studiesAll experiments were carried out under ethics committee–

approved protocols and in compliance with federal guidelinesfor the humane treatment and care of laboratory animals.

Compound formulations. BI 882370 was dissolved in 0.5%Natro-sol (pH 3, citric acid). Vemurafenib in amorphous form wasstabilized by coprecipitationwith hydroxypropylmethyl celluloseacetate succinate (HPMC-AS). The polymer–vemurafenib powder(30% drug load) was suspended in 2% Klucel LF hydroxypro-pylcellulose (pH 3–3.5, HCl). For one of the experiments (Fig. 4),Zelboraf tablets were crushed and milled, and the resultingpowderwas suspended in0.5%Natrosol (pH3,HCl).Dabrafenibwas suspended in 0.5% HPMC-AS and 0.02% Tween 80 (pH 8,NaOH). Trametinib was administered as a suspension in 1%Natrosol/5% DMSO.

Mouse xenograft studies. A375, COLO 205, or G-361 (5� 106), orHT-29 (1.5�106) cellswere injected subcutaneously into 7-week-old female BomTac:NMRI-Foxn1nu mice (Taconic). Mice wererandomized when tumors were well established. All compoundswere administered intragastrically (10 mL/kg). Tumor diameterswere measured 3 times a week, and volumes were calculatedaccording to the formula "tumor volume¼ length� diameter2�p/6".Mice were inspected daily for clinical signs, and bodyweightwas determined 3 times weekly. Median TGI values were calcu-lated as follows:

TGI ¼ 100 � {1�[(treatedfinal day � treatedday1)/(controlfinal day � controlday1)]}

The P values obtained from the Mann–Whitney U test wereadjusted using the Bonferroni–Holm correction. For graphical

representation of tumor growth kinetics, data were plotted asaverage with SEM.

On the last dayof experiments, blood sampleswere takenunderisoflurane anesthesia to determine drug plasma concentrations.

For pharmacokinetic/pharmacodynamic analyses, blood sam-ples and tumors were either snap frozen to determine drugconcentrations or formalin-fixed and paraffin-embedded for IHCevaluation.

IHCImmunohistochemical analysis was carried out in formalin-

fixed paraffin-embedded tumor samples from xenograft models.The indirect immunoperoxidase assay Envision System (Dako#4003) was used with 3,3’-diaminobenzidine solution (SigmaAldrich, # D5905) as a substrate for the immunoreaction and thestaining was performed according to the manufacturer’s instruc-tion. Sections were counterstained with hemotoxylin. The rabbitmonoclonocal antibody against phospho-ERK1/2 (Cell Signaling,#4376) and an antibody against Cyclin D1 (Biocare medical,#CP236A) were used to monitor pathway modulation undertreatment.

For more details, see Supplementary Materials and Methodssection.

Table 1. RAF isoform inhibition and kinase selectivity

A. Vemurafenib Dabrafenib BI 882370Binding mode DFG-in DFG-in DFG-out

Kinase IC50 (nmol/L)BRAFV600E 72 1 0.4BRAFWT 117 2 0.8CRAF 33 0.7 0.6

Kinase KD (nmol/L)BRAFV600E 240 3 4BRAFWT 560 4 6CRAF 220 16 3

Selectivity (number of kinases inhibited by >50% at 1,000 nmol/L/totalnumber of kinases)

6/243 24/238 15/238NOTE: The IC50 values and KD for BI 882370, vemurafenib, and dabrafenib weredetermined on BRAFV600E mutant, BRAF WT, and CRAF kinase. Selectivity wasdetermined by testing the compounds on a large panel of kinases at a singleconcentration (1,000 nmol/L); inhibition by >50% compared with controls wasscored as positive.

B. Kinase selectivity of BI 882370

KinaseIC50

(nmol/L)BRAFV600E 0.4CSF1R 39LCK 49SRMS 133FGR 226PTK6 316YES1 383RET 405KIT 415SRC N1 432BTK 447SRC 485LYN B 715LYN A 750FRK 979PDGFR A 1,220

NOTE: IC50 values for all kinases that scored positive in the primary screen ofBI 882370.

A Novel RAF Inhibitor with DFG-Out–Binding Mode

www.aacrjournals.org Mol Cancer Ther; 15(3) March 2016 357

Dow

nloaded from http://aacrjournals.org/m

ct/article-pdf/15/3/354/1850899/354.pdf by guest on 01 October 2022

Page 5: Binding Mode: High Efficacy in BRAF-Mutant Tumor

ResultsChemical structure and binding mode

BI 882370 (Fig. 1) is a compound resulting from an extensivelead optimization program to identify highly potent and selectiveRAF inhibitors. The target-bindingmodewas investigatedbyX-raycrystallography using amodified formof the BRAF kinase domainwith improved solubility (18). The structure of the approved RAFinhibitor dabrafenib in complexwith the sameproteinwas solvedin parallel; resolutions of 2.51 and 2.66 Å, respectively, were

achieved (Fig. 1 and Supplementary Fig. S1; SupplementaryTables S1 and S2).

Dabrafenib was found to bind to the hinge region in thecleft between the N- and C-lobes of the kinase, i.e., to the ATPbinding site, in the "DFG-in" (active) conformation ofthe enzyme. The compound shows a classical 2-point hinge-binding interaction via its aminopyridyl moiety: the sulfon-amide oxygen forms H-bonds to the backbone NH of Phe595and the side chain of Lys594. The nitrogen atom of the sul-fonamide moiety is within H-bonding distance (3.3 Å) to themain chain NH of Asp594. This suggests that the nitrogen is ina deprotonated state, stabilizing the DFG-in conformation ofthe kinase.

In contrast to dabrafenib, BI 882370 binds to the ATPbinding site of the kinase positioned in the "DFG-out" con-formation, considered to represent the enzymatically inactivestate of the protein. N20 of the pyrimidyl moiety forms aclassical H-bond to the hinge region; a nonclassical H-bond viathe acidified hydrogen of C21 contributes to binding. The twosulfonamide oxygen atoms form H-bonds to the side chainof Lys601 and the backbone NH of Asp594, respectively.This binding mode stabilizes the DFG-out conformation byplacing Phe595 of the DFG motif just below the pyrrolopyr-idine moiety of BI 882370, enabling an aromatic T-stackinginteraction that may contribute to the high potency of thisinhibitor.

Potency and selectivity in biochemical assaysIn enzymatic assays using a cascade RAF–MEK–ERK setup,

BI 882370 potently inhibited the oncogenic BRAFV600E-mutantas well as theWT BRAF andCRAF kinases, with similar IC50 values

BRAFV600E BRAF WT BRO-mutant A375

GDC-0879 BI 882370

p-MEK1/2

MEK1

p-ERK1/2

ERK1/2

D1/D2Cyclin

Kip1/p27

0.3

1 3DMSO

nmol/L300.1

10 100

100

300

1,00

0

DMSO

30 3,00

0

10.3

30.1

10

BA

nmol/L

α-Tubulin (2 h)

α-Tubulin (24 h)

nmol/LWB: ARAF

WB: BRAF

IP: C

RAF

WB: CRAF

DMSO

100

1,00

010

,000

DMSO

100

1,00

010

,000

10

C

Figure 2.Biomarker modulation and RAFheterodimerization. BRAFV600E-mutant A375melanoma cells (A) or BRAF WT, NRAS-mutantBRO melanoma cells (B) were treated withincreasing concentrations of BI 882370 or with1% DMSO as vehicle control. After 2 hours ofincubation [samples for detection of p-MEK1/2,MEK1, p-ERK1/2, ERK1/2, and a-tubulin (loadingcontrol for 2-hour study)] or 24 hours ofincubation [cyclinD1/D2, Kip1/p27, anda-tubulin(loading control for 24-hour study)] cells wereharvested and analyzed by immunoblotting.C, to analyze compound-induced dimerization,BRO melanoma cells were treated withBI 882370, GDC-0879 (positive control), or 1%DMSO (vehicle control) for 1 hour. Proteinextracts were prepared and CRAFimmunoprecipitates (IP) were analyzed byimmunoblotting with A, B, or CRAF-specificantibodies. WB, Western blots.

Table 2. Pharmacodynamic biomarker modulation and antiproliferative effecton cancer cell lines

Vemurafenib Dabrafenib BI 882370

PD biomarker EC50 (nmol/L)p-ERK (A375) 59 5 0.5p-ERK (SK-MEL-28) 71 5 0.7

Proliferation EC50 (nmol/L)A375V600E 110 4 0.9SK-MEL-28V600E 264 7 1G-361V600V/E 1,272 39 6A101DV600V/E�

216 Not done 2.6A375V600E

�83 Not done 1.4

BROWT >20,000 7,581 5,960COLO 205V600V/E

��227 9 3

HT-29V600V/E��

196 7 2LS411NV600V/E 177 9 3HCT-116WT >20,000 >20,000 5,768

NOTE: EC50 values were determined on BRAF-mutant and WT melanoma(A101D, A375, SK-MEL-28, G-361, and BRO) and colorectal cancer cell lines(COLO 205, HT-29, LS411N, and HCT-116) using cell-based ELISA technology forthe detection of p-ERK and alamarBlue, DAPI-labelled nuclei (�), or 3H-thymi-dine incorporation tests (��) for determining antiproliferative activity.

Waizenegger et al.

Mol Cancer Ther; 15(3) March 2016 Molecular Cancer Therapeutics358

Dow

nloaded from http://aacrjournals.org/m

ct/article-pdf/15/3/354/1850899/354.pdf by guest on 01 October 2022

Page 6: Binding Mode: High Efficacy in BRAF-Mutant Tumor

(0.4, 0.8, and 0.6 nmol/L, respectively; Table 1A). Reporterdisplacement assays resulted in KD values of 4, 6, and 3 nmol/L, respectively. Dabrafenib showed similar potency in enzymaticas well as competitive binding assays, whereas vemurafenib wasabout 100-fold less potent.

The selectivity of BI 882370 was determined at a concen-tration of 1,000 nmol/L on a panel of 253 kinases. The activityof 15 enzymes was inhibited by 50% or more; IC50 valueswere determined for all of them (Table 1B). The most sensitivekinase CSF1R was inhibited with an IC50 value of 39 nmol/L,

i.e., with about 100-fold lower potency compared withBRAFV600E inhibition.

Pathway inhibition and antiproliferative activityHigh potency of BI 882370 was observed in cellular assays. In

human A375 melanoma cells carrying a homozygous BRAFV600E

mutation, treatment for 2 hours resulted in a reduction of phos-pho-MEK1/2 and phospho-ERK1/2 signals as seen in immuno-blots, with EC50 values estimated as 0.3 nmol/L and completesuppression observed at 3 nmol/L (Fig. 2A). Treatment for 24

0

200

400

600

800

1,000

1,200

1,400

0 5 10 15 20Av

erag

e tu

mor

volu

me

(mm

3 )

Day

Vehicle control

BI 882370 (25 mg/kg bid)

Vemurafenib (120 mg/kg qd)

Dabrafenib (60 mg/kg qd)

A

0

200

400

600

800

1,000

0 5 10 15 20

Aver

age

tum

or v

olum

e (m

m3 )

Day

Vehicle control

Vemurafenib (120 mg/kg qd)

BI 882370 (25 mg/kg bid)

BI 882370 (12.5 mg/kg bid)

B

0

200

400

600

800

1,000

0 20 40 60

Aver

age

tum

or v

olum

e (m

m3 )

Day

Vehicle control

Trame�nib (0.25 mg/kg bid) + Dabrafenib (50 mg/kg qd)

BI 882370 (25 mg/kg bid) + Trame�nib (0.25 mg/kg bid)

BI 882370 (25 mg/kg bid)

Vemurafenib (120 mg/kg qd)

Trame�nib (0.25 mg/kg bid)

Dabrafenib (50 mg/kg qd)

Treatment stopped

C

Figure 3.First-line therapy of human melanomaxenografts in nudemice. A, mice bearingA375 tumors (BRAFV600E homozygous)were treated with BI 882370 at 25 mg/kg twice daily (bid), with vemurafenib at120 mg/kg once daily (qd), or withdabrafenib at 60 mg/kg once daily.Adjusted P values: BI 882370 vs.vemurafenib, P ¼ 0.0012 and vs.dabrafenib, P ¼ 0.0012. B, micebearing G-361 tumors (BRAFV600V/E

heterozygous) were treated withBI 882370 at 12.5 or 25mg/kg twice dailyor with vemurafenib at 120 mg/kg oncedaily. Adjusted P values: BI 88237025 mg/kg twice daily vs. vemurafenib,P ¼ 0.0012; BI 882370 12.5 mg/kg twicedaily vs. vemurafenib, P ¼ 0.0070).C, mice bearing G-361 tumors(BRAFV600V/E heterozygous) weretreated with BI 882370 at 25 mg/kgtwice daily, vemurafenib at 120 mg/kgonce daily, dabrafenib at 50mg/kg oncedaily, trametinib at 0.25 mg/kg twicedaily, or a combination of trametinibwith either BI 882370 or dabrafenib. Thegroups receiving BI 882370 and thecombination groups were treated for41 days and thereafter observed untilday 60 without further treatment. Onday 23, adjusted P values wereBI 882370 vs. vemurafenib, P ¼ 0.0018,vs. trametinib, P ¼ 0.0018, and vs.dabrafenib, P ¼ 0.0018.

A Novel RAF Inhibitor with DFG-Out–Binding Mode

www.aacrjournals.org Mol Cancer Ther; 15(3) March 2016 359

Dow

nloaded from http://aacrjournals.org/m

ct/article-pdf/15/3/354/1850899/354.pdf by guest on 01 October 2022

Page 7: Binding Mode: High Efficacy in BRAF-Mutant Tumor

A

B

C

D

0

200

400

600

800

1,000

1,200

1,400

1,600

–40 –30 –20 –10 403020100

Aver

age

tum

or v

olum

e (m

m3 )

Day

Trame�nib (0.25 mg/kg bid)+ BI 882370 (25 mg/kg bid)

BI 882370 (25 mg/kg bid)

Vemurafenib (120 mg/kg qd)

Trame�nib (0.25 mg/kg bid)

Vemurafenib (120 mg/kg qd)

Randomiza�on

Nadir of median

Day 39: VemurafenibDay 5: VemurafenibControl

–100

100

300

500

700

900

1,100

Mice with tumor volumes between 200–500 mm 3 used for second-line treatment

tumorsIndividual a�er days36

% C

hang

e fr

om b

asel

ine

–100

–80

–60

–40

–20

0tumorsIndividual a�er days11

% C

hang

e fr

om b

asel

ine

Waizenegger et al.

Mol Cancer Ther; 15(3) March 2016 Molecular Cancer Therapeutics360

Dow

nloaded from http://aacrjournals.org/m

ct/article-pdf/15/3/354/1850899/354.pdf by guest on 01 October 2022

Page 8: Binding Mode: High Efficacy in BRAF-Mutant Tumor

hours suppressed cyclin D1/D2 expression, whereas Kip1/p27was induced at concentrations of 1 nmol/L or higher, indicating acell-cycle arrest in theG1phase. Thepotencyof the compoundwasalso determined in cell-based ELISAs for ERK phosphorylation(Table 2). BI 882370 showed EC50 values of 0.5 and 0.7 nmol/L inA375 and SK-MEL-28 (BRAFV600E) melanoma cells, respectively.Dabrafenibwas 5- to 10-fold less active, in spite of similar potencyin biochemical tests (see above); vemurafenibwas about 100-foldless potent than BI 882370.

Compound-induced RAF heterodimerization in BRAF WTcells is considered to be the initiating event for paradoxicalpathway activation, resulting in hyperproliferation of normalas well as RAS-mutant cells (23–25). For BI 882370, copreci-pitation assays did not provide evidence of heterodimer for-mation in the NRAS-mutant melanoma cell line BRO at phar-macologically relevant concentrations; even at high concentra-tions (1–10 mmol/L), dimer formation was barely detectable(Fig. 2C). In contrast, the tool compound GDC-0879 (25)clearly induced formation of both CRAF/ARAF as well asCRAF/BRAF dimers. Nevertheless, BI 882370 induced phos-phorylation of MEK1/2 and enhanced phosphorylation ofERK1/2 in BRO cells at concentrations between 3 and 300nmol/L; importantly, however, expression of cyclins D1/D2 orKip1/p27 was not affected (Fig. 2B).

To simulate transient compound exposure in vivo, the timecourse of pathway modulation after short-term in vitro treatmentwas analyzed using the A375 model. Cells were treated for 2hours, washed extensively, incubated in medium without inhi-bitors, and harvested 1, 2, 3, 4, 6, or 24 hours thereafter todetermine ERK phosphorylation by cell-based ELISA. Treatmentwith BI 882370 resulted in long-lasting suppression, with EC50

values 24 hours after washout still in the range of 10 to 20nmol/L.In contrast, ERK phosphorylation in cells pretreated with vemur-afenib returned to control values within 1 hour of drug removal,even when the pretreatment concentration was as high as10 mmol/L. In cells pretreated with dabrafenib, pathway suppres-sion was observed for up to 4 hours after washout (EC50, 5–15nmol/L), whereas phospho-ERK levels returned to baseline after 6hours [pretreatment concentrations up to 1,400or 10,000nmol/L(vemurafenib); Supplementary Fig. S2].

In an extended panel of BRAF-mutant human melanomaand colorectal cancer cell lines, BI 882370 showed EC50 valuesin the range of 1 to 10 nmol/L (Table 2); in contrast, proliferationof BRAF WT cells was only inhibited at concentrations > 1,000nmol/L. In accordancewith data on pathway inhibition presentedabove, dabrafenib was about 5-fold and vemurafenib about 100-fold less potent than BI 882370. In additional experiments todetermine the drug sensitivity of BRAF-mutant NSCLC, theBRAFV600E cell line HCC-364 responded with an EC50 of 7nmol/L, whereas none of the cell lines with BRAFG366 orBRAFG469 mutation were sensitive (EC50 > 1,000 nmol/L: NCI-H1395, NCI-H1666, NCI-H1755, and CAL-127).

Efficacy in melanoma modelsIn initial dose-finding and scheduling experiments, we deter-

mined that BI 882370 showed higher efficacy when oral doseswere given twice daily rather than once daily (same total dailydose; Supplementary Table S3, COLO 205 model). Treatment ofmice bearing established A375 melanomas (50–100 mm3) withBI 882370 at doses of 6.25 or 12.5 mg/kg twice daily resulted inhigh efficacy; on day 18, when the control group had to besacrificed for ethical reasons (median tumor volume, 1,000mm3), the median TGI was higher than 100%, and partialregressions were observed in subsets of tumors. At 25 mg/kgtwice daily, all tumors showed either partial (3/7) or complete(4/7) regression. For comparison, vemurafenib (120 mg/kg oncedaily) and dabrafenib (60 mg/kg once daily) resulted in TGIvalues of 89% (one partial regression) and 97% (2 partial regres-sions), respectively (Fig. 3A; Supplementary Table S3; see belowfor pharmacokinetic considerations). BI 882370 was well toler-ated at all dose levels, as judged by body weight gain andclinical signs. 25 mg/kg administered twice daily was thereforeused as the standard treatment dose/schedule for subsequentefficacy studies; an MTD has not been defined. Pharmacokineticexperiments indicated that under these conditions, drug exposure(AUC0–24h) at the endof a treatment period of 2 to 3weeks rangedfrom 12,000 to 25,000 nmol.h/L.

In additional experiments in the A375 model, we comparedcompound concentrations in tumors and plasma after a singleoral dose of BI 882370 (50 mg/kg), dabrafenib (75 mg/kg), orvemurafenib (120 mg/kg). For BI 882370, tumor concentra-tions were higher than the plasma concentrations at each timepoint investigated, in contrast to dabrafenib or vemurafenib(Supplementary Fig. S3). Following treatment with BI 882370,the phospho-ERK signal in tumor cells as determined by IHCwas strongly or partially suppressed for at least 16 hours in spiteof undetectable drug plasma concentrations at 16 hours (Sup-plementary Fig. S4).

G-361melanoma cells with heterozygous BRAFV600E mutationare less sensitive to BRAF inhibitors than A375 cells in vitro(Table 2) and when grown as xenograft tumors may thusserve as a more discriminative pharmacologic model. In the firstexperiment (Fig. 3B; Supplementary Table S3), BI 882370 at 12.5mg/kg twice daily resulted in a median TGI of 98% with 2 of 7tumors showing partial regression; at 25 mg/kg twice daily, alltumors regressed. Vemurafenib, dosed at 120 mg/kg daily toachieve exposures that correspond to those observed in patients(AUC0–24h approximately 1,700,000 nmol.h/L for 960 mg twicedaily; ref. 26), was less efficacious; after initial stabilization, tumorsresumed growth after around 8 days of treatment. In an indepen-dent experiment (Fig. 3C; Supplementary Table S3), vemurafenibagain showed only moderate efficacy. Similarly, dabrafenib at50 mg/kg once daily (AUC0–24h 23,000 nmol.h/L compared withAUC0-t8,400nmol.h/L inpatientsupon repeatdosingwith150mgtwice daily; ref. 27) had only moderate impact on tumor growth.

Figure 4.Second-line therapy of vemurafenib-resistant A375 tumors. A375 tumor-bearing mice were treated with vemurafenib at 120 mg/kg once daily (qd). A, all tumorsshowed regression, and a nadir of the median was reached on day 11 (individual tumor volumes as percent change from baseline/prior treatment are shown).B, on day 36, the majority of tumors had resumed growth (individual tumor volumes as percent change from baseline/prior treatment are shown); thered circle indicates tumorswith complete regression. Green bars indicate tumors ofmice selected for second-line treatment. C,MAPKpathway activity as determinedby phospho-ERK status in representative tumor samples. D, on day 39, a subset of tumors (tumor volumes between 200–500 mm3) was randomized forsecond-line treatment with vemurafenib (120 mg/kg once daily), trametinib [0.25 mg/kg twice daily (bid)], BI 882370 (25 mg/kg twice daily) or a combinationof BI 882370 and trametinib.

A Novel RAF Inhibitor with DFG-Out–Binding Mode

www.aacrjournals.org Mol Cancer Ther; 15(3) March 2016 361

Dow

nloaded from http://aacrjournals.org/m

ct/article-pdf/15/3/354/1850899/354.pdf by guest on 01 October 2022

Page 9: Binding Mode: High Efficacy in BRAF-Mutant Tumor

The MEK inhibitor trametinib at the MTD was only moderatelyefficacious in delaying tumor growth (0.25 mg/kg twice daily;AUC0–24h ¼ 520 nmol.h/L, compared with 600 nmol.h/L uponrepeat dosing with 2 mg once daily in patients; ref. 28). A com-binationof dabrafenib and trametinib showedmarkedly improvedefficacy, stabilizing the tumor volume throughout the 42-daytreatment period (1/7 tumors in regression); when treatment wasdiscontinued, however, tumors started to regrowwithin a few days.BI 882370 dosed at 25mg/kg twice daily showed high efficacy as asingleagent, as all tumorspartially regressedunder treatment; upondiscontinuation, tumor regrowth was markedly delayed. Interest-ingly, the combination of BI 882370 with trametinib did notfurther improve efficacy; in particular, no complete regression wasobserved.

Additional experiments were performed using the heterozy-gous BRAFV600V/E A101D melanoma model. Both, BI 882370(25 mg/kg twice daily) and trametinib (0.25 mg/kg twice daily)were highly efficacious, achieving partial regression of alltumors (n ¼ 7/group); vemurafenib (120 mg/kg once daily)was less efficacious (TGI¼ 80%, no regressions; SupplementaryTable S3).

Acquired resistance and second-line treatmentIn the experiments described above,we hadobserved that A375

xenografts were sensitive to vemurafenib but rapidly acquiredresistance. Tomodel second-line therapy,we treated a large cohortof animals bearing A375melanomas with vemurafenib (120mg/kg daily). As expected, all tumors initially responded, although todifferent extent (day 11, regression by 10%–80%; Fig. 4A).Subsequently, a large proportion of tumors resumed growth,whereas a minority continued to regress, and on day 39, aremarkably broad spectrum of responses ranging from completeregression to rapid growth was observed (Fig. 4B). A subset ofmice with progressing tumors (200 to 500 mm3, median 280mm3)was then randomized into four groups (Fig. 4D). Tumors inmice treated with vemurafenib continued to grow rapidly, andtrametinib as a single agent didnot provide anybenefit. Treatmentwith BI 882370 at 25 mg/kg twice daily resulted in initial tumorregression; a nadir was reached after about 2weeks, but all tumorseventually resumed growth. When animals were treated with acombination of BI 882370 and trametinib, regression wasmore pronounced, and no regrowth was observed during theentire 5-week treatment period.

In a first step towards a mechanistic insight into acquiredvemurafenib resistance, we investigated phospho-ERK expressionin tumors on day 5 and 39 of treatment. We observed a reductionof the signal onday5 (Fig. 4C),whereas ERKphosphorylationwasno longer suppressed on day 39.

Several genetic alterations leading to acquired resistance to RAFinhibitors have been described, including NRAS amplification ormutation, BRAF or CRAF gene amplification, aberrant splicing ofBRAF, MEK1 mutations and upregulation MAP3K8/COT expres-sion (29–33). To investigate whether any of these mechanismsmay be operating in vemurafenib-resistant A375 tumors, weobtained samples prior to treatment (n ¼ 4) as well as on day39 of vemurafenib treatment (n ¼ 5) and analyzed the entiretranscriptome by RNA sequencing [20,297 expressed genes; thedata are accessible through GEO series accession numberGSE74729 (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc¼GSE74729)].No single-nucleotide sequence alterationswereobserved. Expression levels of BRAF were unchanged, and no

BRAF splice variants were detected in resistant tumors (data notshown); however, expression levels of several genes were consis-tently altered in all resistant samples: expression of genes encod-ing SPRY2, SPRY4, DUSP6, CCND1, PIK3R3, FGFR1, EPHA4,MCL1, and IGF1R was downregulated, whereas expression ofPDGFC, VEGFC, ABCB9, and KITLG was increased (Supplemen-tary Table S4).

Efficacy in colorectal cancer modelsIn the BRAFV600V/E-mutant COLO 205 model, previously

reported to be sensitive to BRAF inhibition, treatment withvemurafenib at 120 mg/kg once daily for 2 weeks resulted inpartial regression of 2 of 7 tumors (median TGI ¼ 100%; Sup-plementary Table S3). BI 882370 at 25mg/kg twice daily inducedshrinkage of all tumors; 4 of these regressed completely. From thisexperiment, multiple organs and tissues of 5 animals were pre-served and examined histologically; no evidence for BI 882370–induced hyperplasia or any other pathologic alterations wasseen in skin, heart, lung, bladder, stomach, liver, kidney, andbladder (Supplementary Table S5). In an independent COLO205experiment, BI 882370 showed high efficacy at doses down to6.25 mg/kg twice daily (Supplementary Fig. S5).

The HT-29 model (BRAFV600V/E) was reported to be only mod-erately sensitive to BRAF inhibition (34). We confirmed this obser-vation using vemurafenib at 120mg/kg once daily, achieving a TGIof 60%. BI 882370 at 12.5mg/kg twice daily resulted in 73% TGI; aminor improvement was noted when the dose was increased to 25mg/kg twice daily (TGI¼ 85%). Further doubling of the dose to 50mg/kg twice daily resulted in an increase of exposure from 25,000to 62,000 nmol.h/L, but no increase in efficacy (TGI ¼ 88%). Ofnote, BI 882370 was well tolerated even at this high dose, admin-istered over a 3-week period (Supplementary Fig. S6A). Uponhistopathologic examination (haematoxylin/eosin staining),tumors in controlmice exhibited a poorlydifferentiatedphenotypein contrast to well-differentiated adenocarcinomas observed aftertreatment with BI 882370 (data not shown).

Previous studies have demonstrated that partial resistance toBRAF inhibition in BRAF-mutant colorectal cancer is mediated byEGFR signaling (35). We have confirmed that the EGFR-specificantibody cetuximab as well as the ErbB family kinase inhibitorafatinib, although not significantly efficacious as single agents,synergize with BI 882370, achieving tumor growth control andevenpartial regressions in 5 and6of 7mice per group, respectively(Supplementary Fig. S6B; Supplementary Table S3). Similarly,trametinib showedonlymoderate efficacy as a single agent (TGI¼82%, no regression) but markedly improved the efficacy ofBI 882370 (TGI ¼ 100%, 2/7 regressions). Finally, treatment ofthe animals with a combination of BI 882370, trametinib, andcetuximab resulted in regression of all tumors (7/7); however,complete regressions were not observed even with this triple drugcombination (Supplementary Table S3; this table also includesadditional results of other combination therapy regimens).

Exploratory evaluation of safety and tolerability in ratsAs noted above, a first study evaluating multiple organs of

tumor-bearingmice treated for 2weekswith BI 882370 at a highlyefficacious dose level had not provided any evidence of pertinentpathologic changes. As a next step, an exploratory toxicologystudy was performed in male rats (Supplementary Table S6).Doses of 10, 30, and 60 mg/kg were administered once daily for

Waizenegger et al.

Mol Cancer Ther; 15(3) March 2016 Molecular Cancer Therapeutics362

Dow

nloaded from http://aacrjournals.org/m

ct/article-pdf/15/3/354/1850899/354.pdf by guest on 01 October 2022

Page 10: Binding Mode: High Efficacy in BRAF-Mutant Tumor

2 weeks (5 animals per group). Pharmacokinetic analysis at theend of the study showed that the exposures achieved in the high-dose group exceeded those required for high efficacy in xenograftmodels (60 mg/kg; 43,000 nmol.h/L). Clinical chemistry andhematology investigations (days 3 and14), histopathologic exam-ination as well as toxicogenomic analysis of liver (days 1 and 14)and skin (day 14) did not reveal any toxicologically relevantchanges.

Discussion

Approval of BRAF inhibitors for use in patients withBRAFV600-mutant melanomas has provided an importantimprovement in treatment outcomes over the alkylating agentdacarbazine, the previous standard of melanoma therapy. Inmany patients, these targeted agents achieve rapid disease control,with response rates of approximately 50%. However, completeresponses are rarely observed, and moreover, median progres-sion-free survival is less than one year even in patients treatedwitha RAF/MEK inhibitor combination (6), with biomarker studiesindicating that the MAPK pathway is active in progressing tumorsin spite of continuous treatment. Dose escalation, an obviousmeasure to intensify pathway suppression, is not an option due totolerability limitations. In recent years, a large number of pre-clinical studies as well as analyses of patient-derived tumor tissuehave attempted to shed light on the mechanisms of resistance.Surprisingly, secondary point mutations in the target protein, afrequent cause of resistance to other kinase inhibitors, were notobserved; instead, an unexpectedly broad spectrum of geneticalterations leading to reactivation of the MAPK pathway has beenuncovered. More recently, it was shown that treatment of mela-noma cells with BRAF inhibitors induces secretion of solublefactors that enhance the survival of drug-sensitive cells andaccelerate the expansion of drug-resistant clones (36). Finally,experiments in mouse models have revealed a role of the tumorstroma in drug resistance, as melanoma-associated fibroblasts areactivated by BRAF inhibitors to promote matrix remodeling,which in turn results in elevated integrin signaling in malignantcells and increased ERKactivity independent of BRAF (37). In viewof this plethora of potential resistance mechanisms, it appearsquestionable whether any single-targeted agent, or in fact anycombination of targeted agents, may be able to overcome resis-tance or at least delay relapse to a clinically meaningful extentbeyond that achieved by the approved dabrafenib–trametinibcombination. We reasoned that further improvement of clinicaloutcomesmaybe realized bydesigning a novel RAF inhibitorwithimproved drug properties, such as high potency and selectivity tominimize off-target activity, favorable effects on target conforma-tion with respect to undesired B/CRAF dimer formation, and longresidence time on the target as well as favorable tissue distribu-tion, all converging on an improved therapeutic window thatenables stronger suppression of target activity for extended per-iods of time. The results we present here indicate that this strategymay be feasible. On the basis of preliminary experiments, weselected BI 882370 for in-depth profiling, a compound that bindsto BRAF (and presumably also to A/CRAF) in the inactive con-formation termed "DFG-out" with reference to the position of theactivation loop of the kinase. DFG-out binders, or type II inhi-bitors, in distinction to type I inhibitors that bind to the "DFG-in"active conformation, are considered to potentially achieve ahigher degree of potency and selectivity and likely to possess

greater cellular potency and slower dissociation rates than theirtype I counterparts (38). BI 882370 indeed showed higher cellularpotency than approved BRAF inhibitors, high selectivity in a largekinase panel, long duration of action, and favorable tissue dis-tributionwith consequent long-term suppression of target activityin melanoma xenografts. In several xenograft models of BRAF-mutant cancers, BI 882370 showed superior efficacy in compar-ison with established BRAF and MEK inhibitors dosed to achieveexposures observed in patients. Of particular interest are theresults of our attempt to model second-line treatment of A375BRAFV600E melanomas after the failure of first-line vemurafenib.In the majority of vemurafenib-treated animals, we noted thecharacteristic pattern of initial regression and subsequent pro-gression observed in patients. RNA sequencing of resistant tumorsshowed no evidence of point mutations, BRAF overexpression, ortruncation, whereas the expression of several genes was consis-tently up or downregulated. These results indicate that epigeneticmechanisms are underlying drug resistance in this model, inaccordance with recent findings in a subset of clinical melanomasamples (39). Progressing tumors were insensitive to treatmentwith a MEK inhibitor, again reflecting the clinical situation (40).In contrast, treatment of progressing tumors with BI 882370resulted in tumor regression; however, resistance developed with-in about 2 to 3 weeks. Treatment with BI 882370 in combinationwith trametinib resulted in more pronounced regression, and noregrowth was observed within 5 weeks.

BI 882370 at doses of 25 mg/kg twice daily was well toleratedby mice for periods of at least several weeks; in one experiment,dose escalation to 50 mg/kg twice daily with concomitant higherexposurewas likewisewell tolerated. AMTDhas not beendefined.As a first step towards a more refined evaluation of safety andtolerability of BI 882370, tumor-bearing mice treated for 2 weekswere submitted to exploratory examination of multiple organs;neither epithelial hyperplasia nor any other pathologic changeswere observed. In addition, exploratory toxicologic examinationof male rats treated with doses up to 60 mg/kg once daily for 2weeks, resulting in exposures in excess of those providing superiorefficacy in xenograft models, did not result in any adverse clinicalobservations or any relevant findings in terms of clinical chem-istry, hematology, pathology, or toxicogenomics.

Taken together, our results indicate that it may indeed bepossible to develop second-generation RAF inhibitors that exhibita superior therapeutic window compared with approved com-pounds. BI 882370, or agents with similar mode of target inter-action and physicochemical properties, may provide higherresponse rates and longer duration of response and may besuperior combination partners for MEK inhibitors as well as fornovel agents suggested by recent preclinical studies, includinginhibitors of FAK (37), of the PI3K pathway (41) as well as, forBRAF-mutant colon carcinomas,with inhibitors of the EGFR (35).

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: I.C. Waizenegger, A. Baum, S. Steurer, H. Stadtm€uller,O. Schaaf, P. Garin-Chesa, F. Colbatzky, S. Mousa, N. Kraut, G.R. AdolfDevelopment of methodology: I.C. Waizenegger, O. Schaaf, P. Garin-Chesa,N. Schweifer, F. ColbatzkyAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): I.C.Waizenegger, A. Baum, O. Schaaf, P. Garin-Chesa,F. Colbatzky, S. Mousa, A. Kalkuhl

A Novel RAF Inhibitor with DFG-Out–Binding Mode

www.aacrjournals.org Mol Cancer Ther; 15(3) March 2016 363

Dow

nloaded from http://aacrjournals.org/m

ct/article-pdf/15/3/354/1850899/354.pdf by guest on 01 October 2022

Page 11: Binding Mode: High Efficacy in BRAF-Mutant Tumor

Analysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): I.C.Waizenegger, A. Baum, S. Steurer, H. Stadtm€uller,G. Bader, O. Schaaf, P. Garin-Chesa, A. Schlattl, N. Schweifer, C. Haslinger,F. Colbatzky, A. Kalkuhl, G.R. AdolfWriting, review, and/or revision of themanuscript: I.C.Waizenegger, A. Baum,G. Bader, O. Schaaf, P. Garin-Chesa, A. Schlattl, F. Colbatzky, S. Mousa,A. Kalkuhl, N. Kraut, G.R. AdolfAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): G. Bader, P. Garin-Chesa, N. Schweifer,F. ColbatzkyStudy supervision: I.C. Waizenegger, P. Garin-Chesa, F. Colbatzky, S. Mousa,N. Kraut, G.R. Adolf

AcknowledgmentsThe authors acknowledge the contributions of the entire BI BRAF research

and development team. In particular, the authors thank Horst Ahorn, RosaBaumgartinger, Karin Bichler, Karin Bosch, RalphKnoll, KarenK€ohler, Franziska

Popp, Christina Puri, Nina Rodi, Regina Ruzicka, Christian Salamon, MichaelaStreicher, Alexander Wlachos, and Susanne Wollner-Gaida for their excellent,dedicated contributions to experiments described in this article and ThomasBogenrieder for discussion on medical perspectives.

Grant SupportThis work was financially supported by Boehringer Ingelheim.The costs of publication of this article were defrayed in part by the

payment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received July 23, 2015; revised November 13, 2015; accepted December 30,2015; published OnlineFirst February 25, 2016.

References1. Wimmer R, Baccarini M. Partner exchange: protein-protein interactions in

the Raf pathway. Trends Biochem Sci 2010;35:660–8.2. Samatar AA, Poulikakos PI. Targeting RAS-ERK signalling in cancer: pro-

mises and challenges. Nat Rev Drug Discov 2014;13:928–42.3. Welsh SJ, Corrie PG. Management of BRAF and MEK inhibitor toxicities in

patients with metastatic melanoma. Ther Adv Med Oncol 2015;7:122–36.4. Gibney GT, Messina JL, Fedorenko IV, Sondak VK, Smalley KS. Paradoxical

oncogenesis–the long-term effects of BRAF inhibition in melanoma. NatRev Clin Oncol 2013;10:390–9.

5. 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.

6. LongGV, StroyakovskiyD,GogasH, LevchenkoE, de Braud 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.

7. Girotti MR, Lopes F, Preece N, Niculescu-Duvaz D, Zambon A, Davies L,et al. Paradox-breaking RAF inhibitors that also target SRC are effective indrug-resistant BRAF mutant melanoma. Cancer Cell 2015;27:85–96.

8. Torti VR, Wojciechowicz D, Hu W, John-Baptiste A, Evering W, Troche G,et al. Epithelial tissue hyperplasia induced by the RAF inhibitor PF-04880594 is attenuated by a clinically well-tolerated dose of the MEKinhibitor PD-0325901. Mol Cancer Ther 2012;11:2274–83.

9. Wisler JA, Afshari C, FieldenM, ZimmermannC, Taylor S, Carnahan J, et al.Raf inhibition causes extensive multiple tissue hyperplasia and urinarybladder neoplasia in the rat. Toxicol Pathol 2011;39:809–22.

10. King AJ, Arnone MR, Bleam MR, Moss KG, Yang J, Fedorowicz KE, et al.Dabrafenib; preclinical characterization, increased efficacy when com-bined with trametinib, while BRAF/MEK tool combination reduced skinlesions. PLoS One 2013;8:e67583.

11. Stadtmueller H, inventor; Boehringer Ingelheim International GmbH,assignee. New azyindolylphenyl sulfonamides as serine/threonine kinaseinhibitors). WO2012/104388 A1. 2012.

12. Himmelsbach F, Langkopf E, Blech S, Jung B, Baum ESF, inventors;Boehringer Ingelheim Pharma KG, assignee. Quinazoline derivatives,medicaments containing said compounds, their utilization and methodfor the production thereof. WO2002/50043 A1. 2002.

13. Ibrahim PN, Artis DR, Bremer R, Mamo S, Nespi M, Zhang C, et al.,inventors; Plexxikon, Inc., assignee. Pyrrolo(2,3-B) pyridine derivatives asprotein kinase inhibitors. WO2007/002325 A1. 2007.

14. Desai D, Diodone R, Go Z, Ibrahim PN, Iyer RM,Mair H-J, et al., inventors;F. Hoffmann-La Roche AG; Plexxikon, Inc, assignees. Compositions anduses thereof. WO2010/114928 A2. 2010.

15. Adams JL, Dickerson SH, Johnson NW, Kuntz K, Petrov K, Ralph JM, et al.,inventors; Smithkline Beecham Coorporation, assignee. Benzene sulfon-amide thiazole and oxazole compounds. WO2009/137391 A2. 2009.

16. Sakai T, Kawasaki H, AbeH,HayakawaK, Iida T, Kikuchi S, et al., inventors;Japan Tobacco INC., assignee. 5-Amino-2,4,7-Trioxo-2,4,7,8-Tetrahydro-2H-Pyrido-2,3D-Pyrimidine derivate and related compounds for the treat-ment of cancer. WO2005/121142 A1. 2005.

17. Laird E, Lyssikatos JP, Welch M, Grina J, Hansen J, Newhouse B, et al.,inventors; Genentech, INC.; Array Biopharma, Inc., assignees. Raf inhibitorcompounds and methods. WO2006/084015 A2. 2006.

18. Tsai J, Lee JT, Wang W, Zhang J, Cho H, Mamo S, et al. Discovery of aselective inhibitor of oncogenic B-Raf kinase with potent antimelanomaactivity. Proc Natl Acad Sci U S A 2008;105:3041–6.

19. Kabsch W. XDS. Biol Crystallogr Online 2010;66:125–32.20. Emsley P, Lohkamp B, Scott WG, Cowtan K. Features and development of

Coot. Acta Crystallogr D Biol Crystallogr 2010;66:486–501.21. Murshudov GN, Vagin AA, Dodson EJ. Refinement of macromolecular

structures by the maximum-likelihood method. Acta Crystallogr D BiolCrystallogr 1997;53:240–55.

22. Neumann L, von KK, Ullmann D. HTS reporter displacement assay forfragment screening and fragment evolution toward leads with optimizedbinding kinetics, binding selectivity, and thermodynamic signature. Meth-ods Enzymol 2011;493:299–320.

23. Heidorn SJ, Milagre C,Whittaker S, Nourry A, Niculescu-Duvas I, DhomenN, et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumorprogression through CRAF. Cell 2010;140:209–21.

24. Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF inhibitorstransactivate RAF dimers and ERK signalling in cells with wild-type BRAF.Nature 2010;464:427–30.

25. Hatzivassiliou G, Song K, Yen I, Brandhuber BJ, Anderson DJ, Alvarado R,et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathwayand enhance growth. Nature 2010;464:431–5.

26. Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, et al.Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl JMed 2010;363:809–19.

27. Wang J, Earp J, Grimstein C, Mehrotra N, Orbach RC, Pacanowski M, et al.Clinical pharmacology and biopharmaceutics review 202806/S-000 Part01; 2013. Available from: https://www.pharmapendium.com/fda.do?query¼%28202806Orig1s000%29&includeSynonyms¼true&drugName-¼DabrafenibþMesylate&document¼b85ba5032af11ea8797fd933e8495-6e8.

28. Leong R, Yu J, Shord S, Mehrotra N, Orbach RC, Pacanowski M, et al.Clinical pharmcology and biopharmaceutics review 204114/S-000 part01; 2013. Available from: https://www.pharmapendium.com/fda.do?query¼%28204114Orig1s000%29&includeSynonyms¼true&drug-Name¼TrametinibþDimethylþSulfoxide&document¼036279dde5f64-e7c0505e441d38a0646.

29. Johannessen CM, Boehm JS, Kim SY, Thomas SR, Wardwell L, Johnson LA,et al. COT drives resistance to RAF inhibition throughMAP kinase pathwayreactivation. Nature 2010;468:968–72.

30. 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.

31. Villanueva J, Vultur A, Lee JT, Somasundaram R, Fukunaga-Kalabis M,Cipolla AK, et al. Acquired resistance to BRAF inhibitorsmediated by a RAFkinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer Cell 2010;18:683–95.

Waizenegger et al.

Mol Cancer Ther; 15(3) March 2016 Molecular Cancer Therapeutics364

Dow

nloaded from http://aacrjournals.org/m

ct/article-pdf/15/3/354/1850899/354.pdf by guest on 01 October 2022

Page 12: Binding Mode: High Efficacy in BRAF-Mutant Tumor

32. 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.

33. 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.

34. Yang H, Higgins B, Kolinsky K, Packman K, Bradley WD, Lee RJ, et al.Antitumor activity of BRAF inhibitor vemurafenib in preclinicalmodels of BRAF-mutant colorectal cancer. Cancer Res 2012;72:779–89.

35. Prahallad A, Sun C, Huang S, Di NF, Salazar R, Zecchin D, et al. Unre-sponsiveness of colon cancer to BRAF(V600E) inhibition through feedbackactivation of EGFR. Nature 2012;483:100–3.

36. Obenauf AC, Zou Y, Ji AL, Vanharanta S, Shu W, Shi H, et al. Therapy-induced tumour secretomes promote resistance and tumour progression.Nature 2015;520:368–72.

37. Hirata E, Girotti MR, Viros A, Hooper S, Spencer-Dene B, Matsuda M, et al.Intravital imaging reveals how BRAF inhibition generates drug-tolerant

microenvironments with high integrin beta1/FAK signaling. Cancer Cell2015;27:574–88.

38. Liu Y, Gray NS. Rational design of inhibitors that bind to inactive kinaseconformations. Nat Chem Biol 2006;2:358–64.

39. Hugo W, Shi H, Sun L, Piva M, Song C, Kong X, et al. Non-genomic andimmune evolution of melanoma acquiring MAPKi resistance. Cell2015;162:1271–85.

40. KimKB, KeffordR, Pavlick AC, Infante JR, Ribas A, Sosman JA, et al. Phase IIstudy of the MEK1/MEK2 inhibitor Trametinib in patients with metastaticBRAF-mutant cutaneous melanoma previously treated with or without aBRAF inhibitor. J Clin Oncol 2013;31:482–9.

41. Coffee EM, Faber AC, Roper J, Sinnamon MJ, Goel G, Keung L, et al.Concomitant BRAF and PI3K/mTOR blockade is required for effectivetreatment of BRAF(V600E) colorectal cancer. Clin Cancer Res 2013;19:2688–98.

42. Zhang C, Spevak W, Zhang Y, Burton EA, Ma Y, Habets G, et al. RAFinhibitors that evade paradoxical MAPK pathway activation. Nature2015;526:583–6.

www.aacrjournals.org Mol Cancer Ther; 15(3) March 2016 365

A Novel RAF Inhibitor with DFG-Out–Binding Mode

Dow

nloaded from http://aacrjournals.org/m

ct/article-pdf/15/3/354/1850899/354.pdf by guest on 01 October 2022