15
Small Molecule Therapeutics Targeting of Hematologic Malignancies with PTC299, A Novel Potent Inhibitor of Dihydroorotate Dehydrogenase with Favorable Pharmaceutical Properties Liangxian Cao 1 , Marla Weetall 1 , Christopher Trotta 1 , Katherine Cintron 1 , Jiyuan Ma 1 , Min Jung Kim 1 , Bansri Furia 1 , Charles Romfo 1 , Jason D. Graci 1 , Wencheng Li 1 , Joshua Du 1 , Josephine Sheedy 1 , Jean Hedrick 1 , Nicole Risher 1 , Shirley Yeh 1 , Hongyan Qi 1 , Tamil Arasu 1 , Seongwoo Hwang 1 , William Lennox 1 , Ronald Kong 1 , Janet Petruska 1 ,Young-Choon Moon 1 , John Babiak 1 , Thomas W. Davis 1 , Allan Jacobson 2 , Neil G. Almstead 1 , Art Branstrom 1 , Joseph M. Colacino 1 , and Stuart W. Peltz 1 Abstract PTC299 was identied as an inhibitor of VEGFA mRNA translation in a phenotypic screen and evaluated in the clinic for treatment of solid tumors. To guide precision cancer treatment, we performed extensive biological char- acterization of the activity of PTC299 and demonstrated that inhibition of VEGF production and cell proliferation by PTC299 is linked to a decrease in uridine nucleotides by targeting dihydroorotate dehydrogenase (DHODH), a rate-limiting enzyme for de novo pyrimidine nucleotide synthesis. Unlike previously reported DHODH inhibitors that were identied using in vitro enzyme assays, PTC299 is a more potent inhibitor of DHODH in isolated mito- chondria suggesting that mitochondrial membrane lipid engagement in the DHODH conformation in situ is required for its optimal activity. PTC299 has broad and potent activity against hematologic cancer cells in preclin- ical models, reecting a reduced pyrimidine nucleotide salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased levels of dihydroorotate, the substrate of DHODH, indi- cating target engagement in patients. PTC299 has advan- tages over previously reported DHODH inhibitors, includ- ing greater potency, good oral bioavailability, and lack of off-target kinase inhibition and myelosuppression, and thus may be useful for the targeted treatment of hemato- logic malignancies. Introduction De novo pyrimidine nucleotide biosynthesis is activated in proliferating cancer cells in response to an increased demand for synthesis of DNA, RNA, and phospholipids by activation of oncogenic pathways such as Myc, PI3K, PTEN, and mTOR (15). Dihydroorotate dehydrogenase (DHODH), located on the surface of the inner mitochondrial membrane, catalyzes the fourth enzymatic step, the ubiquinone-mediated oxidation of dihydroorotate to orotate in the de novo pyrimidine synthesis pathway (6). DHODH is a validated therapeutic target for the treatment of autoimmune diseases such as rheumatoid arthritis (7, 8). Inhibition of DHODH was shown in preclinical studies to prevent growth of many types of cancers (913). Moreover, it was demonstrated that inhibition of DHODH induces differentiation of acute myeloid leukemia (AML) cells (1416). Pharmacologic inhibition of de novo pyrimidine synthesis sensitizes triple-nega- tive breast cancer cells to genotoxic chemotherapy agents and reduces chemotherapy resistance (2). DHODH inhibitors such as teriunomide, brequinar, and vidoudimus have been consid- ered for use in oncology (1720), but their use may be limited due to a narrow therapeutic window as a result of lesser potency and/ or off-target activities such as their inhibition of kinases (21, 22). There is great interest in the discovery and development of new DHODH inhibitors for therapeutic uses (23, 24). These efforts have mainly relied on in vitro enzyme assays to dene and optimize activity, in which the puried enzyme conformation may differ from that found in the cells due to lack of membrane lipids in the assay (25). Recently, it was demonstrated that DHODH attaches to the inner mitochondrial membrane by binding charged phospholipids which stabilize the exible sub- strate- and drug-binding site, suggesting that the design and 1 PTC Therapeutics, Inc., South Plaineld, New Jersey. 2 Department of Microbi- ology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Current address for T. Arasu: US Food and Drug Administration, 629 Cranbury Road, 2nd Floor, East Brunswick, NJ 08816; and current address for T.W. Davis: PMV Pharmaceuticals Inc., Cedar Brook Corporate Center, 8 Clarke Drive, Cranbury Township, NJ 08512. Corresponding Author: Liangxian Cao, PTC Therapeutics, Inc., 100 Corporate CT, South Plaineld, NJ 07080. Phone: 908-912-9132; Fax: 908-222-0567; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-18-0863 Ó2018 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org 3 on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

Small Molecule Therapeutics

Targeting of Hematologic Malignancies withPTC299, A Novel Potent Inhibitor ofDihydroorotate Dehydrogenase with FavorablePharmaceutical PropertiesLiangxian Cao1, Marla Weetall1, Christopher Trotta1, Katherine Cintron1, Jiyuan Ma1,Min Jung Kim1, Bansri Furia1, Charles Romfo1, Jason D. Graci1,Wencheng Li1, Joshua Du1,Josephine Sheedy1, Jean Hedrick1, Nicole Risher1, Shirley Yeh1, Hongyan Qi1, Tamil Arasu1,SeongwooHwang1,William Lennox1, Ronald Kong1, Janet Petruska1,Young-ChoonMoon1,John Babiak1, Thomas W. Davis1, Allan Jacobson2, Neil G. Almstead1, Art Branstrom1,Joseph M. Colacino1, and Stuart W. Peltz1

Abstract

PTC299 was identified as an inhibitor of VEGFA mRNAtranslation in a phenotypic screen and evaluated in theclinic for treatment of solid tumors. To guide precisioncancer treatment, we performed extensive biological char-acterization of the activity of PTC299 and demonstratedthat inhibition of VEGF production and cell proliferationby PTC299 is linked to a decrease in uridine nucleotidesby targeting dihydroorotate dehydrogenase (DHODH), arate-limiting enzyme for de novo pyrimidine nucleotidesynthesis. Unlike previously reported DHODH inhibitorsthat were identified using in vitro enzyme assays, PTC299is a more potent inhibitor of DHODH in isolated mito-chondria suggesting that mitochondrial membrane lipid

engagement in the DHODH conformation in situ isrequired for its optimal activity. PTC299 has broad andpotent activity against hematologic cancer cells in preclin-ical models, reflecting a reduced pyrimidine nucleotidesalvage pathway in leukemia cells. Archived serum samplesfrom patients treated with PTC299 demonstrated increasedlevels of dihydroorotate, the substrate of DHODH, indi-cating target engagement in patients. PTC299 has advan-tages over previously reported DHODH inhibitors, includ-ing greater potency, good oral bioavailability, and lack ofoff-target kinase inhibition and myelosuppression, andthus may be useful for the targeted treatment of hemato-logic malignancies.

IntroductionDe novo pyrimidine nucleotide biosynthesis is activated in

proliferating cancer cells in response to an increased demand forsynthesis of DNA, RNA, and phospholipids by activation ofoncogenic pathways such as Myc, PI3K, PTEN, and mTOR (1–5). Dihydroorotate dehydrogenase (DHODH), located on thesurface of the inner mitochondrial membrane, catalyzes the

fourth enzymatic step, the ubiquinone-mediated oxidation ofdihydroorotate to orotate in the de novo pyrimidine synthesispathway (6). DHODH is a validated therapeutic target for thetreatment of autoimmune diseases such as rheumatoid arthritis(7, 8). Inhibition of DHODH was shown in preclinical studies toprevent growth of many types of cancers (9–13). Moreover, it wasdemonstrated that inhibition of DHODH induces differentiationof acute myeloid leukemia (AML) cells (14–16). Pharmacologicinhibition of de novo pyrimidine synthesis sensitizes triple-nega-tive breast cancer cells to genotoxic chemotherapy agents andreduces chemotherapy resistance (2). DHODH inhibitors such asteriflunomide, brequinar, and vidofludimus have been consid-ered for use in oncology (17–20), but their usemaybe limiteddueto a narrow therapeutic window as a result of lesser potency and/or off-target activities such as their inhibition of kinases (21, 22).

There is great interest in the discovery and development of newDHODH inhibitors for therapeutic uses (23, 24). These effortshave mainly relied on in vitro enzyme assays to define andoptimize activity, in which the purified enzyme conformationmay differ from that found in the cells due to lack of membranelipids in the assay (25). Recently, it was demonstrated thatDHODH attaches to the inner mitochondrial membrane bybinding charged phospholipids which stabilize the flexible sub-strate- and drug-binding site, suggesting that the design and

1PTC Therapeutics, Inc., South Plainfield, New Jersey. 2Department of Microbi-ology and Physiological Systems, University of Massachusetts Medical School,Worcester, Massachusetts.

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

Current address for T. Arasu: US Food and Drug Administration, 629 CranburyRoad, 2nd Floor, East Brunswick, NJ 08816; and current address for T.W. Davis:PMV Pharmaceuticals Inc., Cedar Brook Corporate Center, 8 Clarke Drive,Cranbury Township, NJ 08512.

Corresponding Author: Liangxian Cao, PTC Therapeutics, Inc., 100 CorporateCT, South Plainfield, NJ 07080. Phone: 908-912-9132; Fax: 908-222-0567;E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-18-0863

�2018 American Association for Cancer Research.

MolecularCancerTherapeutics

www.aacrjournals.org 3

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 2: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

development of novel inhibitors that additionally engage in lipidinteractions may represent a promising approach to targetingDHODH in vivo (26).

PTC299 was identified as an inhibitor of the translation ofVEGFA mRNA using PTC Therapeutics' proprietary GEMS tech-nology phenotypic screening platform (27). PTC299 was inclinical trials for the treatment of solid tumors (28, 29) and iscurrently being reevaluated for use in hematologic cancers. Toguide safe and effective precision cancer treatment, we performedextensive biochemical characterization of PTC299 and demon-strated that it interacts with and inhibits DHODH, a rate-limitingenzyme in the de novo biosynthesis of pyrimidine nucleotides (5).Inhibition of VEGFA production by PTC299 is a downstreameffect of inhibiting de novo pyrimidine synthesis as it can becompletely rescued by exogenously added uridine but not byother nucleosides. Collectively, PTC299 represents a novel class ofpotent DHODH inhibitors, and our data demonstrate the poten-tial of PTC299 as a treatment option to address unmet needs incancers, such as leukemia and lymphoma, that have lower levelsof uridine salvage activity and thus rely on the de novobiosynthesisof pyrimidine nucleotides for survival and rapid proliferation.

Materials and MethodsGeneral methods

DNA constructs were generated using standard procedures asreported previously (27). VEGFA 50- and 30-untranslated regions(UTR) were amplified from human genomic DNA (Clontech),while the VEGFA165 open reading frame from RNA derived fromHeLa cells incubated under hypoxia (1% oxygen) was amplifiedby RT-PCR. DNAs generated with PCR were confirmed by DNAsequencing.

PTC299, PTC-371, GSK983, and vidofludimus (4SC-101) weresynthesized at PTC Therapeutics; for details, see the Supplemen-tary Materials and Methods in the Supplementary Data File.Brequinar (SML0113) and teriflunomide (SML0936) were pur-chased from Sigma, whereas pyrazofurin-MP (P61-B01A) waspurchased from TriLink Biotechnologies.

Cell cultureTumor cell lines, except for MOLM-13 which were obtained

from Dr. Kensuke Kojima in July 2015 (Saga University, Japan)andHuh-7whichwere obtained in February 2001 from the StuartPeltz laboratory at University of Medicine and Dentistry of NewJersey (nowRutgers University, New Jersey), were purchased fromthe ATCC and were maintained in DMEM (adherent cells) orRPMI-1640 (suspension cells) supplemented with 10% FBS,penicillin (50 IU/mL), and streptomycin (50 mg/mL). Culturemedia and supplement agents were purchased from Gibco BRL,Invitrogen. Cells bought from the ATCC were expanded andfrozen at early passages. Cells were typically used for studieswithin 2 to 5 weeks after thawed from frozen stocks. Cells thatwere cultured for more than 2 months (about 20 passages) werediscarded. Patient-derived AML cells, purchased from AllCells(January 2017), Inc. and Champions Oncology, Inc. (April2017), were cultured in StemSpan medium (Stemcell Technolo-gies). Cell lines in culture were routinely verified once per monthto be mycoplasma free assayed by using the MycoAlert Myco-plasma Detection Kit (Cat#: LT07-318, Lonza). The 240 cell linesused for screening of PTC299 activity were authenticated withshort tandem repeat profiling andmycoplasma tested by the CRO

(CrownBioscience); otherwise we did not perform any additionalcell line authentication inhouse. The information on the source ofcells and when they were obtained can be found in Supplemen-tary Table S1.

Stable cell lines were generated as reported previously (27).Briefly, plasmid DNA was transfected into HT1080 cells usingFugene-6 transfection reagent (Roche Diagnostics GmbH), fol-lowed by selection with 200 mg/mL hygromycin for luciferase-stable cell lines or 200 mg/mLZeocin (Invitrogen) for V5 tag stablecells in the culture medium.

Western blot analysisWestern blot was performed as reported previously (27). Anti-

body (C1) specific for VEGFA was purchased from Santa CruzBiotechnologies (1:200 dilution); antibody specific for humanb-actin was purchased fromAbcam (1:10,000 dilution); antibodyspecific for V5 tag was purchased from Invitrogen (1:5,000dilution). Antibody specific for DHODH was purchased fromProteintech (Cat#: 14877-1-AP, 1:500 dilution); and antibodyspecific for human prohibitin was purchased from ThermoScientific (Cat#: PA5-12274 and PA5-14133, 1:1,000 dilution).Immunodetection was done using the corresponding secondaryantibodies conjugatedwith infrared dyes or horseradish peroxide.The expression levels of proteins were detected with Odyssey(LI-COR) or using enhanced chemiluminescence (Pierce).

Determination of ELISA EC50 and cytotoxicity CC50 valuesAll ELISAs were performed using commercially available ELISA

kits (R&D Systems) according to the manufacturer's instructions.The screen of 240 cell lines was performed at Crown Bioscience.Cellswere treatedwithPTC299at a seriesofdoses for 72hours, andthe inhibitionof cell proliferationwasdeterminedusinga standardassay, CellTiter-Glo Luminescent Cell Viability Assay (Promega),that measures total cellular adenosine triphosphate (ATP) con-centrations as an indicator of cell viability. Data generated fromELISA or cytotoxicity studies were plotted with Prism software. Asigmoidal dose responsewith a variable slope regression curvewasgenerated for each compound. Maximal and minimal inhibitionswere set at 100% and 0%, respectively, and CC50 values werecalculated after curve fitting using the Prism software.

35S-labeling and immunoprecipitationHT1080 cells harboring either the VEGFA 50-UTR-VEGFA165-V5

construct or the control VEGFA165-V5were treated for 8 hourswithvehicle alone (0.5% DMSO) or vehicle with 100 nmol/L PTC299,and then incubated with 35S-labeled methionine and cysteine forapproximately 4 hours in the presence or absence of the proteintransporter inhibitor brefeldin A or the proteasome inhibitorMG-132. Equal amounts of supernatant or the cell extracts wereimmunoprecipitated with the anti-V5 antibody (Invitrogen).The immunoprecipitated products were resolved by 4% to 20%gradient SDS-PAGE and exposed to Kodak Biomax MR film.

Human tumor xenograft studiesAll studies involving animalswereperformed in accordancewith

guidelines promulgated by the American Association for Accredi-tation of Laboratory Animal Care with the oversight of the animaluseandcare committees atRutgersUniversity. Subcutaneous tumorxenograft studies were performed as reported previously (27). Forsystemic leukemia lethality model, male NOD-SCID mice wereinoculated with MOLT-4 human ALL tumor cells (1 � 107 cells in

Cao et al.

Mol Cancer Ther; 18(1) January 2019 Molecular Cancer Therapeutics4

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 3: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

200 mL of PBS) by i.v. injection. Eight days after tumor inoculation,mice were randomized into two groups (10 mice per groups)and treated with compounds as indicated in the results. The micewereobserveduntilmoribund, atwhich time theywere euthanized.At 2- and 4-week postinoculation, whole bloodwas obtained from5micepergroupbyretro-orbitalbleeding, stainedwithanantibodyagainst human CD45, and the number of CD45-positive cells wasdetermined by FACS analysis.

ImmunohistochemistryTumor samples were cut into pieces of less than 3 mm in

thickness and placed in a zinc fixative for 24 hours. Paraffinsections were prepared by AmericanHistoLab, Inc. using standardprocedures. Mouse endothelial cells were stained with rat anti-mouse CD31 monoclonal antibody MEC13.3 (BD Pharmingen)and detected with a rabbit anti-rat IgG HRP detection kit accord-ing to the manufacturer's instructions (BD Pharmingen).

Transcriptome profiling with microarraysPTC299-resistant and parent HT1080 cells were seeded in 6-

well plates (5 � 105 cells/well) and incubated overnight. Fourreplicates of cells for each cell typewere performed. Total RNAwaspurified with RNeasy Mini Kit (Qiagen) according to the manu-facturer's instructions. cRNA was prepared according to the stan-dard Affymetrix protocol. cRNA was hybridized at 45�C for 16hours on a GeneChip Human Genome U133 plus 2.0 Array.GeneChips were analyzed using the Affymetrix scanner. Dataanalysis was performed using Transcriptome Analysis ConsoleSoftware (Thermo Fisher Scientific). The MAS5 normalizationmethod was used. ANOVA was used for statistical evaluation.

Quantification of pyrimidine nucleotides in PTC299-treatedcells

Cells in log phase growth were cultured in 10 cm dishes (4 �106 cells/dish) with glutamine-free DMEM containing 1 g/Lglucose, 10% FBS, penicillin (50 IU/mL), streptomycin (50 mg/mL), and 1 mmol/L 15N-glutamine (Sigma, Cat#: 490024). Afterculturing for the indicated time in the presence of compounds orvehicle control (0.5% DMSO), the cells were washed once with 5mL cold PBS, harvested, and lysed in 0.5 mL cold distilled H2Oplus 1 mL �80�C methanol using a plastic cell scraper. The celllysates were then centrifuged at 10,000 x g for 15 minutes at 4�C,and the supernatant was collected for LC-MS/MS analysis of the15N-labeled de novo pyrimidine nucleotide metabolites.

LC-MS/MS detection of metabolitesQuantification of 15N-labeled de novo–synthesized pyrimidine

nucleotides was carried out on an Accela pump and a PALautosampler coupled to aTSQQuantumUltramass spectrometer.The mass spectrometer was equipped with a heated electrosprayionization source operated in negative-ion mode (Thermo FisherScientific). The ion spray voltage was set at 2500 V, capillarytemperature at 350�C, vaporizer temperature at 300�C, sheath gaspressure at 50 units, and auxiliary gas pressure at 5 units. AThermo Fisher Scientific Hypercab column (3 mm, 50� 2.1 mm)was used and maintained at 50�C for metabolite separation.Mobile phase A was 10 mmol/L NH4HCO3 in water, pH 9.4,and mobile phase B was 10 mmol/L NH4HCO3 in ACN-water(9:1). The flow rate was set at 0.50 mL/min. Ion transitionsmonitored were at m/z 156.0 ! m/z 112.0 for 15N-orotic acid,

m/z 158.0!m/z 114.0 for 15N-dihydrorotic acid, andm/z 324.0! m/z 79.0 for 15N-monophosphate uridine (UMP).

Conjugation of PTC299 or PTC-371 to Sephorose-6B beadsPTC299 or its inactive enantiomer PTC-371 was conjugated to

the epoxy-activated Sepharose 6B according to the manufacturer'sinstructions (GE healthcare; Cat. # 17-0480-01). Briefly, 1 g ofepoxy-activated Sepharose 6B was stirred in water (20 mL) for 2hours at room temperature and then filtered. The wet Sepharose 6Bwas diluted with pH 11 phosphate/NaOH buffer (5 mL). To asolution of 1 mmol/L 4-chlorophenyl (S)-6-chloro-1-(4-hydroxy-phenyl)-1,3,4,9-tetrahydro-2H-pyrido[3,4-b] indole-2-carboxylateinDMF(12mL)at40�Cwasadded the Sepharose6B inphosphate/NaOH buffer. The reactionmixture was stirred further for 24 hoursat 40�C. The Sepharose 6B-PTC299 product was filtered andwashed with DMF (100 mL), water (100 mL), pH 4 buffer (100mL), pH 11buffer (100mL), water (100mL), and 0.5mol/LNaCl/water. Sepharose 6B-PTC-371 was prepared with 4-chlorophenyl(R)-6-chloro-1-(4-hydroxyphenyl)-1,3,4,9-tetrahydro-2H-pyrido-[3,4-b] indole-2-carboxylate using above reaction procedures.

Pull-down of proteins that are specifically bound to PTC299-conjugated beads

Cells in log phase growth were washed with cold PBS once andthen collected by trypsin digestion. After centrifugation at 1,200rpm for 5 minutes, the cells were resuspended in lysis buffer [PBSþ 0.5% Triton X100 þ 1x proteinase inhibitors (Halt, Roche;Cat#: 78440)] and incubated on ice for 10 minutes; supernatantswere collected after passing cells through a 22-gauge needle 10times and centrifuging at 13,000 rpm for 15 minutes to removecell debris. Subsequently, 1 mL of cell lysate solution (2 mg/mLprotein) was added to each reaction tube containing 25 mL(packed volume) PTC299- or the R-enantiomer control (PTC-371) beads and incubated for 2 hours at 4�Cwith gentle agitationon a rotator; after three washes with 1 mL washing Buffer, 5 min/each, the protein bound with beads was eluted with indicatedbuffer as shown in the Results section. Proteins in the elutedsamples were verified by Western blot analysis.

Mitochondria isolated from cultured K562 cells as source ofDHODH for in vitro enzyme inhibition studies

Mitochondria were isolated fromK562 cells in log phase growthusing a Dounce homogenizer as reported (30). The isolated mito-chondria in the supernatant were pelleted at 12,000 g for 15minutes and resuspended in 1 mL 1x MS homogenization bufferfor analysis of downstream DHODH activity as described below.

In vitro DHODH activity assaysThe chromogen reduction assay was carried out as previously

reported (31).DHODHactivitywasdetermined in thepresence ofcompounds or vehicle control using the standard colorimetricDHODH continuous assay inwhich the oxidation of dihydrooro-tic acid (DHO) and the subsequent coupled reduction of ubiqui-none is measured by monitoring the reduction of 2,6-dichlor-ophenolindophenol (DCIP) on a BioTek Power Wave XS2. Thevelocity for each reaction was derived using the GEN5 software.

Docking analysisSoftware Sybyl X 2.1.1 was used to prepare ligand structures.

Ligands used in docking procedures were imported as SDF 3Dformat which were processed using Pipeline Pilot. Protein crystal

PTC299 Is a Novel Potent DHODH Inhibitor

www.aacrjournals.org Mol Cancer Ther; 18(1) January 2019 5

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 4: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

structures were downloaded from the Protein Data Bank (PDB;http://www.rcsb.org/). Protein structure preparations were per-formed using the standard protocol of Sybyl X 2.1.1, and theresulting structures were saved as SYBYL ''.mol2'' files. The pocketsprotocols were generated using standard fully automated Surflex-Dock procedures (32). Sybyl provides a docking score for eachdocked conformation, which includes two parts: an affinity score(-logKd) and a crash score (also pKd units). The crash score is thedegree of inappropriate penetration into the protein by the ligandas well as the degree of internal self-clashing that the ligand isexperiencing. The top docking scores for each ligandwere used forcomparison.

TheDockingModewas set as Surflex_DockGeomX(SFXC). Inputoptions were set to allow 3D Coordinate Generation, if necessary.Protein movement was set to allow for the flexibility of proteinatomswhose van derWaals surface distances from ligand atoms are< 4 Å and for the adaptation of the active site conformation to thedocked ligand. The covalent force field weighting of the ligand wasset to 1.0, whereas the covalent force field weighting of the proteinwas set to0.6 to allowheavy atoms tomove. Themaximumnumberof poses was set to 10 to optimize and rescore each ligand after adocking run that included protein movement. Additional StartingConformations perMoleculewere set to 6 (GeomX). Themaximumnumber of conformations per fragment to be submitted to thedocking procedure was set to 20. The maximum number of posesper ligand was set to 20. The minimum root-mean-square distancebetween final poses was set to 0.50. The Sybyl version of theAMBER7 FF99 Force Field was used for energy calculations (33).

Statistical analysisData are the mean � SD or SEM as indicated for quantitative

experiments. For statistical analysis, P values were derived usingunpaired Student t tests for any study with only two groupspresented. Otherwise, comparisons of groups were performed onlog-transformed data using a one-way ANOVA test. All analyseswere made using GraphPad Prism Software.

Data availabilityAll data generated or analyzed during this study are included in

this article and its Supplementary Information Files. Additionalinformation is available from the corresponding author uponrequest.

ResultsPTC299 was identified from a phenotypic screen and chemicaloptimization as an inhibitor of VEGFA protein synthesis

Using the contextual regulation of the production of VEGFA inthe tumor cell imparted by critical elements in the 50- and 30-UTRsof the VEGFAmRNA, we initiated a drug discovery program usingPTC Therapeutics' proprietary GEMS technology phenotypicscreening platform (27). Subsequently, structure–activity–rela-tionship (SAR) studies led to the identification of PTC299 (Fig.1A) as a clinical development candidate for cancer therapy.PTC299 is the pharmacologically active S-enantiomer of 4-chlor-ophenyl (1S)-6-chloro-1-(4-methoxyphenyl)-1,3,4,9-tetrahydro-2H-beta-carboline-2-carboxylate [synthesis of PTC299 (5) andintermediates 1–4 shown in Supplementary Fig. S1A]. Similar tothe compounds identified in a high-throughput screen as previ-ously described (27), biological characterization suggested thatPTC299 inhibited VEGFA-50UTR–mediated mRNA translationrather than transcription (Supplementary Fig. S1B–S1D).

In a dose-dependent and stereo-selective manner, PTC299inhibited hypoxia-induced VEGFA protein production in HeLacells with an EC50 of 1.64� 0.83 nmol/L (Fig. 1B). Similar activitywas also observed in HT1080 cells which constitutively producehigh levels of VEGFA (Supplementary Fig. S1E). The effect ofPTC299 is not limited to secreted isoforms of the protein as thelevels of high molecular weight matrix-bound variants ofVEGFA189/206 were similarly decreased (Supplementary Fig.S1F). PTC-371, the R-enantiomer of PTC299, was inactive ininhibiting VEGFA synthesis.

To verify the role of the VEGFA 50-UTR for PTC299 activity, a V5epitope–tagged plasmid containing the VEGFA165 open readingframe was constructed (Fig. 1C, top plot). PTC299 dose-depen-dently inhibited VEGF 50-UTR–mediated production of theVEGFA165-V5 protein as determined by Western blot analysis(Fig. 1C) and by ELISA (Supplementary Fig. S1G). Pulse-chaseexperiments using 35S-radiolabledmethionine and cysteine dem-onstrated that PTC299 decreased the levels of newly synthesizedVEGFA protein (Fig. 1D) while not decreasing total proteinsynthesis (Supplementary Fig. S1H). This effect could not bereversed by treatment with the protein transport blocker brefeldinA (BFA) or with the proteasome inhibitor MG132 (Supplemen-tary Fig. S1I and S1J). These results suggest that PTC299 treatmentresults in the inhibition of VEGFA translation rather than theinhibition of VEGFA secretion or promotion of VEGFA proteindegradation and support the conclusion that the VEGFA 50-UTR isrequired for the suppression of VEGFA mRNA translation byPTC299.

In amouse tumor xenograft model, PTC299 dose-dependentlyinhibited the growth of HT1080 fibrosarcoma (Fig. 1E), withoutovert toxicity or significant bodyweight changes at any dose whencompared with treatment with the vehicle alone (SupplementaryFig. S1K). In parallel, treatment ofmice with PTC299 resulted in adose-dependent reduction in the concentrations of intratumorand circulating plasma human VEGFA relative to those in controlmice (Fig. 1F) but had no significant impact on the levels of othercytokines such as PDGF and FGF-2 in tumors as assessed in aseparate study (Supplementary Fig. S1L). At the suboptimal doseof 0.3 mg/kg, treatment of mice with PTC299 resulted in partialreductions in the levels of both tumor and plasmahumanVEGFA,indicating that inhibition of intratumor human VEGFA correlateswith tumor growth control.

When stained with anti-murine CD31 antibody, the diametersof tumor vessels were visibly reduced, and their distribution wasmore uniform in tumors from PTC299-treated mice than invehicle-treated mice (Fig. 1G), consistent with results in preclin-ical models using other VEGF-targeted therapies (34, 35).

Inhibition of VEGFA production by PTC299 is a downstreameffect of inhibiting de novo pyrimidine synthesis

To gain insight into the mechanism of action of PTC299, wegenerated drug-resistant cells by culturing HT1080 cells in esca-lating concentrations (1 nmol/L to 1,000 nmol/L) of PTC299,doubling the PTC299 concentration weekly. PTC299-resistantHT1080 cells proliferate at a rate similar to that of wild-typeHT1080 cells (Supplementary Fig. S2A), but the IC50 for theinhibition of VEGFA production by PTC299 in resistant cells isshifted dramatically, from low nmol/L to single-digit mmol/Lvalues (Supplementary Fig. S2B). Transcriptome profiling withmicroarrays to compare the gene expression in PTC299-resistantand in parent HT1080 cells demonstrated that the gene with the

Cao et al.

Mol Cancer Ther; 18(1) January 2019 Molecular Cancer Therapeutics6

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 5: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

Figure 1.

Structure, biological activity, and stereoselectivity of PTC299. A, Chemical structure of PTC299, the 1S configuration, and the inactive R enantiomer, PTC-371. B,PTC299 inhibits hypoxia-induced VEGFA expression in HeLa cells. Results are expressed as the percent inhibition of hypoxia-induced VEGFA productionrelative to vehicle-treated controls. Hypoxia (1% oxygen)-induced VEGFA production is measured by subtracting the VEGFA produced under normoxic conditions(21% oxygen) from the levels of VEGFA produced under hypoxia. Data are mean � SD from a representative study (n ¼ 3). C, The VEGFA 50-UTR is required forPTC299 inhibition of a V5-tagged VEGFA165 protein production. HT1080 cells were stably transfected with the constructs shown in the diagrams on the topof the graph. The culture supernatant and cell lysates were collected for Western blot after treatment with PTC299 or 0.5% DMSO control for 36 hours. D, PTC299selectively inhibits production of pulse 35S-labeled VEGFA. The VEGFA-V5 stable cell lines were treated with PTC299 (100 nmol/L) for 8 hours and thenpulsed/chased with 35S-radiolabled methionine and cysteine for 4 hours prior to immunoprecipitation with V5 antibody. E, PTC299 dose-dependently inhibitsHT1080 tumor growth in mice. Values represent the average � SEM of 10 mice per group. Compound or vehicle was orally administered b.i.d. as indicated inthe graph. �� ,P<0.001, one-wayANOVA test. F,PTC299 reduces intratumor and serum levels of humanVEGFA. Values represent the average�SEMof 10mice. One-way ANOVA test compared with vehicle; � , P < 0.05 and �� , P < 0.001. G, PTC299 inhibits tumor angiogenesis, and tumor sections were stained withanti-CD31 antibody.

PTC299 Is a Novel Potent DHODH Inhibitor

www.aacrjournals.org Mol Cancer Ther; 18(1) January 2019 7

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 6: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

Cao et al.

Mol Cancer Ther; 18(1) January 2019 Molecular Cancer Therapeutics8

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 7: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

greatest increase in expression in PTC299-resistant cells is cytidinedeaminase (CDA), whereas the gene with greatest decrease isEukaryotic Translation Initiation Factor 1A, Y-Linked (EIF1AY)[Fig. 2A, themicroarray data can be found in Supplementary ExcelFile S1 or downloaded from GEO database (Accession number:GSE121266)]. These changes in expression were verified by quan-titative PCR as shown in Supplementary Fig. S2C. Because CDA isan enzyme in the uridine salvage pathway, we evaluated the effectof PTC299 treatment on intracellular nucleotide levels. PTC299selectively decreased the total levels of the deoxypyrimidinenucleotides dCTP and dTTP, but not those of the deoxypurinenucleotides dATP and dGTP (Fig. 2B). Metabolic labeling studiesusing 15N-glutamine demonstrated that PTC299 inhibited de novopyrimidine synthesis, whereas its inactive R-enantiomer PTC-371had no effect (Fig. 2C). Further studies demonstrated that inhi-bitionwas dose-dependent andmeasurable after only 30minutesof treatment (Supplementary Fig. S3A and S3B). The inhibitionprofile of PTC299 in these assays is similar to that of the knownDHODH inhibitor brequinar rather than that of the UMPsynthase (UMPS) inhibitor pyrazofurin-monophosphate(refs. 18, 36; Supplementary Table S2). As shown in Fig. 2D,treatment with pyrazofurin-monophosphate resulted in an over40-fold increase of 15N-orotate, the substrate of UMPS. However,cotreatment with PTC299 blocked the pyrazofurin-MP–inducedincrease of 15N-orotate, consistent with inhibition of DHODHthat acts upstream of UMPS.

Weperformednucleoside rescue studies anddemonstrated thatthe PTC299-mediated inhibition of VEGFA protein synthesis wasdose-dependently blocked by the addition of uridine, but not bythe addition of adenosine, cytidine, or guanosine, to the culturemedium (Fig. 2E and F). As a consequence of DHODH inhibitionand subsequent pyrimidine nucleotide depletion, PTC299 alsoinduced an S phase cell-cycle arrest in HT1080 cells (Fig. 2G, 2ndplot). This cell-cycle arrest was prevented by the addition of thepyrimidine nucleosides uridine or cytidine (Fig. 2G, bottom twoplots), but not the purine nucleosides adenosine or guanosine (L.Cao; unpublished data). These data suggest that inhibition ofVEGFA translation and cell-cycle arrest by PTC299 are two sep-arate downstream effects of inhibiting de novo pyrimidine nucle-otide synthesis (Fig. 2H).

PTC299 interact with and inhibits the activity of DHODHExtensive SAR studies demonstrated that a methoxy group on

PTC299 can tolerate modification and is not critical for thebiological activity of PTC299 (Y.C. Moon; unpublished data).

By utilizing this methoxy group, we linked either PTC299, or itsinactive R-enantiomer PTC-371, to epoxy-activated Sepharose 6B(Fig. 3A). In pilot studies, we focused on those highly enrichedproteins in PTC299-conjugated bead pull-down samples by cut-ting out the most enriched protein bands for the proteomicstudies and demonstrated that these proteins are Prohibitin-1/2and voltage-dependent anion channel (VDAC). After demons-trating that PTC299 inhibits de novo pyrimidine synthesis andhas an activity profile similar to that of the DHODH inhibitorbrequinar (Fig. 2; Supplementary Table S2), we performedanother pull-down study and demonstrated that PTC299 beadsenriched not only for Prohibitin-1/2 but also for DHODH in thepull-down samples (Fig. 3B). To determine if binding to theseproteins was PTC299 specific, we first performed "compete-on"studies and demonstrated that preincubation with 100 mmol/LPTC299 selectively blocked the binding of DHODH to thePTC299-conjugated beads while it had no effect on the bindingof Prohibitin-1/2 (Fig. 3C). The inactive enantiomer PTC-371 hadno effect on the binding of either DHODH or Prohibitin-1/2.Similar results were observedwhen isolatedmitochondrial lysateswere used as the protein source (Supplementary Fig. S3C). Wenext demonstrated that both free PTC299 and brequinar effec-tively blocked the binding of DHODH to the PTC299-conjugatedbeads while having no effect on prohibitin binding (Fig. 3D),suggesting that PTC299 and brequinar bind to the same site inDHODH. Subsequently, we performed a "competing off" studyand demonstrated both PTC299 and brequinar effectively elutedout the DHODH from the PTC299-conjugated beads while theinactive isomer PTC-371 did not (Fig. 3E), further supporting thenotion that PTC299 and brequinar bind to the same pocket inDHODH. Proteomic study of those elution samples verified thatone of the most selectively enriched proteins was DHODH(Supplementary Table S3). Because PTC299 is species selectiveand does not have activity inmouse and rat cell lines tested in vitro(Supplementary Table S4), we performed a side-by-side pull-down study with human and rat cell lysates. As shown in Fig.3F, PTC299-conjugated beads enriched for both humanDHODHand Prohibitin-1/2. However, these beads only bound to ratprohibitin rather than rat DHODH when compared with theblankbeads or the PTC-371–conjugated beads. These data suggestthat PTC299 specifically binds to humanDHODH, and that otherenriched proteins such as Prohibitin-1/2 are PTC299-bead selec-tive rather than PTC299 specific.

Further studies demonstrated PTC299 inhibited the activity ofrecombinant DHODHusing in vitro enzyme assays, though not as

Figure 2.Inhibition of VEGFA translation by PTC299 is coupled with inhibition of de novo pyrimidine synthesis. A, Transcriptome profiling of PTC299-resistant HT1080 cellsusingmicroarrays. Highlighted on the left volcano plot are those geneswith greater than 2-fold changes in expressionwith a FDR less than 0.05; the heatmaps of theexpression of these genes are shown on the right. B, PTC299 selectively decreases pyrimidine nucleotides in HT1080 cells. HT1080 cells were treated withPTC299 (100 nmol/L) or 0.5% DMSO control for 8 hours. Data are representative of two independent studies. C, PTC299 inhibits de novo pyrimidine nucleotidesynthesis. HT1080 cells in log phase growthwere treatedwith compounds (100 nmol/L) or vehicle control (0.5%DMSO) in the presence of 1mmol/L 15N-glutamine for8 hours. 15N-labeled CTP, UTP, and UMP were quantified with LC/MS. Data are representative of two independent studies. D, PTC299 acts upstream of UMPS.Cells were treated with 1 mmol/L PTC299 or MP-pyrazofurin or combination of the two for 8 hours. Peak area ratio of orotic acid (normalized to an internal control)for each treatment was determined, and average � SD was shown in the picture (n ¼ 2). MP-pyrazofurin is a UMPS inhibitor. E, Inhibition of VEGFA production inHT1080 cells by PTC299 is completely blocked by addition of exogenous uridine into the cell culture. Cells were treated with PTC299 (100 nmol/L) or vehicle (0.5%DMSO) in the presence of 100 mmol/L various nucleosides (indicated in the figure) for 48 hours. Data are mean � SD, n ¼ 2. F, Inhibition of VEGFA production inHT1080 cells by PTC299 is dose-dependently blocked by addition of exogenous uridine. Assays were done in triplicate, and data represent average inhibitionagainst DMSO control after VEGFA levels normalized to viable cell number measured by Celltiter-Glo. Data represent the mean � SD, n ¼ 2. G, Cell-cycle arrestin S-phase induced by PTC299 is completely blocked by addition of exogenous uridine or cytidine into the cell culture. HT1080 cells were treated with PTC299 (100nmol/L) or vehicle (0.5% DMSO) in the presence of 100 mmol/L of the indicated nucleosides for 24 hours. H, A scheme showing de novo and salvage pathways ofpyrimidine nucleotide synthesis. CAD, Carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase; UCK, uridine cytidine kinase.

PTC299 Is a Novel Potent DHODH Inhibitor

www.aacrjournals.org Mol Cancer Ther; 18(1) January 2019 9

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 8: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

Figure 3.

PTC299binds to and inhibits the activity of DHODH.A,Schematic drawingof PTC299, the inactive isomer linked to the agarose beads and the outline of chemical pull-down study. B, PTC299 beads bind to DHODH and Prohibitin-1/2. Pull-down samples from Blank-, PTC-371–, and PTC299-conjugated beads were resolvedon4%–20%SDS-PAGE. Left plot shows the silver staining,whereas the right plot showsWesterns blottedwith antibodies against humanDHODH, PHB1 (Prohibitin-1),PHB2 (Prohibitin-2), and VDAC respectively. C, PTC299 selectively inhibits DHODH binding to the PTC299 beads. HT1080 cell lysates were preincubated with 100mmol/L free PTC299 or PTC-371 as indicated in the figure for 2 hours at 4�C prior to the pull-down study described in the methods. D, Western blot analysisdemonstrated that free PTC299 and brequinar competes DHODH binding to the PTC299 beads. The experiment was done in the sameway as for Fig. 3C. E,Westernblot analysis demonstrated that free PTC299 selectively elutes DHODH from the PTC299 beads. F, PTC299-conjugated beads selective bind to human rather than ratDHODH. G, PTC299 inhibits DHODH activity in isolated mitochondria, data are representative of three independent mitochondria preparations, and orotateproduction was quantified by LC-MS/MS analysis of the end product after 30 minutes of enzyme reaction. H, Computer modeling using crystal structures of thehuman DHODH protein. Both the brequinar analog C44 (Purple) and PTC299 (cyan) bind deep in the pocket, but the inactive enantiomer PTC-371 (orange) does not,which is reflected by the docking scores on the right. The cofactor flavin mononucleotide (FMN) and the product orotate are shown in yellow and blue,respectively. Right plot shows a localized view that PTC299 (cyan) binds to the pocket of human DHODH crystal at 4IGH. Amino acids within 5 angstroms of PTC299are shown as a ribbon. Amino acid residues within 3 angstroms were labeled (hydrophobic, red, charged, purple; polar, blue). Relevant side chains that make upthe pocket are displayed explicitly.

Cao et al.

Mol Cancer Ther; 18(1) January 2019 Molecular Cancer Therapeutics10

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 9: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

potently as do brequinar or teriflunomide (Supplementary Fig.S3D). However, when purified mitochondria were used as thesource of DHODH, PTC299 inhibited DHODH activity morepotently than did teriflunomide and with potency similar to thatof brequinar (Fig. 3G; Supplementary Fig. S3E). Along with theresult that uridine can completely block the inhibition of VEGFAproduction and cell-cycle arrest by PTC299, these data indicatethat DHODH is the primary target responsible for the observedactivities of PTC299.

Computer modeling shows that PTC299 binds to the same sitein DHODH as does brequinar and with similar strength

Docking studies on available human DHODH protein (PDBID: 4IGH, http://www.rcsb.org/) revealed that PTC299 and thepotent brequinar analog C44 bind to the same site with similarstrength (Fig. 3H), consistent with the pull-down results thatdemonstrated brequinar competes with DHODH binding toPTC299 beads. The Sybyl Surflex docking score, calculated basedon the binding affinity and crash score (37), is 9.55 for C44 versus9.27 for PTC299. The interaction of PTC299 with the bindingpocket is mainly a "hand-in-glove" type nonpolar hydrophobicinteraction. Unlike with C44, there are no H-bond interactionsbetweenPTC299 andArg136 inDHODH.As anticipated from the

pull-down studies, the inactive enantiomer, PTC-371, bound lessefficiently in the pocket with a final docking score of 7.05,probably because of the unique nonsymmetrical shape of thecavity. Although PTC299 is a less potent inhibitor of rhDHODHin vitro, it more potently inhibits DHODH from isolated mito-chondria (Fig. 3G; Supplementary Fig. S3E). Because PTC299 islipophilic, its entry into the binding pocket of the enzyme is likelyfacilitated when the enzyme is embedded in the mitochondrialmembrane with engagement of membrane phospholipids. Instudies using purified recombinant protein, there is nomembraneor lipid layer thatmayhinder the entry of PTC299 into thebindingpocket. Efforts to cocrystallize PTC299 and purified recombinantDHODH have not been successful to date (Nonato MC, Uni-versidade de S~ao Paulo, Brazil; personal communication), pos-sibly because of the requirement for the lipid engagementwith theenzyme (26). We are currently working to optimize crystallo-graphic conditions to determine the structure of lipid-engagedDHODH interaction with PTC299.

PTC299 inhibits DHODH activity in patients withneurofibromatosis type 2

Comprehensive preclinical IND-enabling safety pharmacolo-gy, toxicology, and genotoxicity studies (SupplementaryMaterials

Figure 4.

PTC299 inhibits DHODH activity andreduces elevated VEGFA levels inpatients with NF2. A, Reduction inmean levels of serum VEGFA after 8weeks of PTC299 treatment. Data arerepresented as mean � SEM, n ¼ 11.B, VEGFA reduction for individualpatients over the course of the PTC299treatment. C, Serum levels of DHO arecorrelated with PTC299 concentrationin patient-002. D, Serum levels of DHOare inversely correlated with serumVEGFA levels in patient-002.E, PTC299 increases serum DHO in 3additional patients whose serumsamples were still available at the timeof analysis. BLQ: below the level ofquantification.

PTC299 Is a Novel Potent DHODH Inhibitor

www.aacrjournals.org Mol Cancer Ther; 18(1) January 2019 11

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 10: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

and Methods in the Supplementary Data File) demonstrated thatPTC299 is safe and well tolerated in preclinical studies (Supple-mentary Table S5). Subsequently, several clinical trials in healthyvolunteers and in patients with solid tumors were conducted toassess PTC299 (28, 29), including a prospective phase IIa study in2009–2010 in patients with neurofibromatosis type 2 (NF2), anautosomal-dominant tumor-suppressor syndrome characterizedby bilateral vestibular Schwannomas. Eleven patients wereenrolled including 4 males and 7 females with a median age of25 (range, 19–44) at the time of enrollment. The demographicinformation on these patients is summarized in SupplementaryTable S6. After 8 weeks of PTC299 treatment, levels of serumVEGFA were significantly decreased (Fig. 4A, P < 0.004, pairedStudent t test). Baseline levels in healthy volunteers were reportedto be approximately 140 pg/mL (38), indicating that serumVEGFA levels are elevated in NF2 patients and were reduced bytreatment with PTC299 to levels found in healthy volunteers.Circulating VEGFA was decreased in 11 of 11 patients as shownin Fig. 4B.

We next examined archived serum samples from some patientsin the NF2 clinical study for DHO levels, hypothesizing thatinhibition of DHODH would result in higher levels of thissubstrate as seen in patients with genetic deficiency in theDHODH enzyme (39). As shown in Fig. 4C and D, serum DHOlevels in Patient 010-002 who had been on study for 64 weekswere initially below the lower limit of quantification. After treat-ment with PTC299, plasma concentrations of PTC299 and serumlevels of DHO were significantly increased (Fig. 4C), whereasserum VEGFA protein levels were continuously decreased (Fig.4D). For another 3 patients whose sera were also available at thetime the analyses were performed, DHO levels were also belowthe lower limit of quantification prior to treatment but weresignificantly increased with PTC299 treatment (Fig. 4E). Theincreased levels of DHO in these samples indicate that PTC299inhibited DHODH in the previous clinical studies.

PTC299 demonstrates broad and potent inhibition ofhematologic cancer cell proliferation

PTC299 inhibits de novo pyrimidine nucleotide synthesis in allcancer cells analyzedwhether the cells are responsive to inhibitionof VEGFA production or not (Supplementary Fig. S4). Becausepyrimidine nucleotides can be synthesized by the de novo or thesalvage pathway (40), some cancer cells may be insensitive toPTC299 due to increased pyrimidine nucleotide salvage activity.Thus, we determined the IC50 (concentration to inhibit cellproliferation by 50%) of PTC299 against a panel of 240 tumorcell lines. We classified 68 cells lines as responders and 172 cellslines as nonresponders based on an arbitrary IC50 cutoff of 1mmol/L. Based on these results, sensitivity to PTC299 wasobserved across different tumor types (Supplementary Fig.S5A). When grouped as hematopoietic versus solid tumor cells,57% (32/56) of hematopoietic lines were sensitive to PTC299,whereas only 18%(33/184) in the solid tumor lineswere sensitiveto PTC299.

Gene expression of pyrimidine nucleotide synthesis enzymeswas analyzed using published microarray data in the Cancer CellLine Encyclopedia (CCLE) database (41). Of 240 cell lines tested,232were found in theCCLEdatabase including 54 hematopoieticand 178 solid tumor cell lines. Two salvage enzymes, CDA anduridine phosphorylase 1 (UPP1) that convert cytidine and uracil,respectively, to uridine, were significantly less expressed (3.2-fold

for CDA and 4.6-fold for UPP1) in the hematopoietic tumor linesthan in the solid tumor lines (Fig. 5A; Supplementary Table S7).We then performed LC-MS/MS analysis of UMP pools fromsalvage and de novo pathways in a panel of 8 tumor cell linesand demonstrated that PTC299-sensitive cells in general have lesssalvage UMP production relative to de novo UMP productionwhen compared with the insensitive tumor cells (Fig. 5B). Thesedata suggest that cells with reduced uridine salvage activity aremore sensitive to PTC299.

We then compared the potency of PTC299 in leukemia cellswith that of several well-known DHODH inhibitors (17–19).PTC299 was the most potent inhibitor with an IC50 of about 1nmol/L, over 10- to 1000-fold more potent than brequinar,vidofludimus, or teriflunomide (Fig. 5C), consistent with itssuperior potency in the inhibition of VEGFA production in tumorcells (Supplementary Fig. S6).We also tested PTC299 against AMLpatient-derived cells. As shown in Fig. 5D, PTC299 inhibited theproliferation of all five AML patient-derived cell lines tested withIC50 values ranging from 2 to 60 nmol/L. In addition, we haveshown that PTC299 induced differentiation in AML MOLM-13cells (Supplementary Fig. S5B), consistent with the activity ofvarious DHODH inhibitors as recently reported (14).

After confirming in vitro activity in leukemia cells, we assessedthe efficacy of PTC299 in two mouse models of leukemia. Asshown in Fig. 1E, the maximal effect in control of tumor growthand VEGFA production was achieved at a dose of 3 mg/kg b.i.d.Later studies found that qd dosing 10 mg/kg was equivalentlyeffective (M. Weetall; unpublished data) and thus this experi-mentally more feasible dosing regimen was used in the leukemiaanimal models described below.

In the first model, MOLT-4 human acute lymphoblastic leu-kemia (ALL) cells were injected i.v. into NOD-SCID mice whichwere then treated with PTC299 or with doxorubicin at the indi-cated doses. Doxorubicin was tested at the highest dose toleratedby NOD-SCIDmice. As shown in Fig. 5E, vehicle-dosed mice hada median survival time (MST) of 46 days and did not survivebeyond 71 days. Doxorubicin did not prolong survival time. TheMST for themice dosedwith PTC299was 136 days (P < 0.05, one-way ANOVA, multiple comparisons vs. vehicle). The number ofcirculatingMOLT-4 cells was significantly reduced inmice treatedwith PTC299 but not in mice treated with doxorubicin (Supple-mentary Fig. S5C).

In a second study, MOLM-13 AML cells were injected into theflank of nudemice to generate a solid tumor. Cytarabine (AraC) isa deoxycytidine analog used as a standard chemotherapeuticagent to treat AML (42). As shown in Fig. 5F, PTC299 treatmentalone resulted in a significant delay of tumor growth whencompared with vehicle or AraC treatment; the median time toreach a tumor volume of 1,000 mm3 was 23 days for PTC299versus 7 and 13 days for vehicle- and AraC-treated mice, respec-tively. The combination of PTC299 with AraC further delayedtumor growth for 39 days. The increased activity of the combi-nation of AraC with PTC299 may reflect a more efficient incor-poration of AraC into DNA due to a reduction in the levels ofcompeting endogenous pyrimidine nucleotides.

DiscussionTumor cells require the upregulation and integration of mul-

tiple biosynthetic processes including synthesis of nucleotidesand proteins to drive uncontrolled cell proliferation (43). Here,

Cao et al.

Mol Cancer Ther; 18(1) January 2019 Molecular Cancer Therapeutics12

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 11: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

Figure 5.

PTC299 demonstrates broad activity andpotent inhibition of leukemia cell proliferation in vitro and in animalmodels.A,Box plot of log2 expression level (microarray)for CDA andUPP1, two pyrimidine salvage enzymes for converting cytidine and uracil, respectively, to uridine. The box represents the lower and upper quartile of thedata. The white line in the box represents the median value. The whiskers represent the extension of data up to 1.5x of interquartile range (IQR) below thelower quartile or above the upper quartile. P values are calculated with Software R (https://www.r-project.org/, version 3.3.1) based on t test comparinghematopoietic tumors (n ¼ 54) and solid tumors (n ¼ 178); B, PTC299-sensitive cells in general have less salvage UMP production. Four sensitive (HeLa, HT1080,A549, and K562) and four insensitive lines (U87MG, MCF7, PC3, and Huh7) were cultured in the presence of 15N-Glutamine for 8 hours, then the unlabeled and15N-labeled UMP in the cell lysates were measured by LC-MS/MS analysis. Data are mean � SEM, n ¼ 4, P value from t test. C, PTC299 inhibits the proliferation ofMOLM13 AML cells more potently than do other known DHODH inhibitors. Cells were treated with indicated compounds for 72 hours, and cell proliferationwasmeasured usingCelltiter Glo (Promega). % inhibitionwas calculated against the vehicle (0.5%DMSO) treatment. Data aremean�SD, n¼ 2.D,PTC299 is active inall fiveAML patient-derived cells in vitro. Patient-derivedAML cellswere treatedwith PTC299 for 72 hours, and then cell proliferationwas assessed using Celltiter Glo(Promega). Assays were done in duplicate (AML-002, 004, and 005) or triplicate (AML-001 and 003). E, PTC299 prolongs survival of mice bearing human ALLafter i.v. inoculation of NOD/SCID mice with MOLT-4 cells (10 mice/group). Treatment with PTC299 (10 mg/kg, qd), or with doxorubicin (0.5 mg/kg or 1 mg/kg i.p.once per week), was initiated 7 days after tumor inoculation; F, PTC299 inhibits MOLM-13 AML xenograft tumor growth in nude mice. Values shown representthe mean of 8 mice per group, and error bars show the SEM.

PTC299 Is a Novel Potent DHODH Inhibitor

www.aacrjournals.org Mol Cancer Ther; 18(1) January 2019 13

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 12: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

we demonstrate that decreased translation of VEGFA mRNA byPTC299 is linked to the inhibition of de novo pyrimidine nucle-otide synthesis as exogenously added uridine, but not othernucleosides, can completely block the inhibition of VEGFA pro-duction by PTC299.Unlike uridine, cytidine only rescued the cell-cycle arrest in S-phase but not the inhibition of VEGFA produc-tion. This discrepancy may be due to the lower levels of CDAexpression (Fig. 2A; Supplementary Fig. S2C), and cytidine couldnot be efficiently converted to uridine in the HT1080 cells.Moreover, exogenously added cytidine in cells expressing lowlevels of CDA could be quickly converted to CTP/dCTP which inturn might further inhibit the CDA and other salvage enzymes(44, 45).

Extensive biological characterization demonstrated thatDHODH is the primary target of PTC299 as all the observedactivities of PTC299 can be completely rescued by addition ofexogenous uridine rather than other nucleosides. The proteomicstudy showed other potential targets may exist, and the selectivityand significance of those proteins identified in the proteomicstudy warrant further investigation.

Similar to PTC299, otherDHODH inhibitors such as brequinaralso inhibit VEGFA production albeit less potently (Supplemen-tary Fig. S6B and S6C). The link between the inhibition of VEGFAmRNA translation and de novo pyrimidine synthesis was unan-ticipated but may be critical for cancer cells to synchronize rapidproliferation and angiogenesis for tumor progression. Inhibitionof VEGFA mRNA translation by PTC299 is mediated by the 50-UTR of the mRNA prompting multiple hypotheses for this link:(1) Depletion of uridine nucleotide pools may affect certaintranslation initiation factor(s) required for the noncanonicalprotein synthesis of stress-regulated mRNAs such as the VEGFAmRNA. We observed that eIF1AY gene expression decreased over90% in PTC299-resistant HT1080 cells, when compared withwild-type HT1080 cells (Fig. 2A; Supplementary Fig. S2). eIF1Acontrols translation start codon recognition and has beenreported to affect IRES-mediated translation (46). (2) Low levelsof uridine nucleotides may also affect the mRNA substrate direct-ly. For example, depletion of pyrimidine nucleotides in cancercells can cause a starvation-like stress response which may lead tochanges in pseudouridylation state of mRNA (47). Reduction ofdyskerin expression, a component of pseudouridine synthase, hasbeen shown to selectively increase VEGFA-IRES–mediated trans-lation (48). As uridines in RNAs are substrates for dyskerin,depletion of uridine nucleotides by PTC299 may reduce freesubstrate competition and thus activate dyskerin to inhibit VEGFAmRNA translation. (3) A recent study has demonstrated that theactivity of mTOR, which controls synthesis of proteins andnucleotides (5), can be regulated by nucleotide pools (49). Thus,it is also possible that inhibition of the translation of regulatedmRNAs, such as the VEGFAmRNA, is mediated via disrupting theproduction of pyrimidine nucleotides causing imbalances innucleotide pools. Elucidating the molecular mechanisms linkinginhibition of DHODH and VEGFA mRNA translation requiresfurther investigation.

Two inhibitors of humanDHODHhave been approved for usein autoimmune diseases, leflunomide for rheumatoid arthritis,and its active metabolite, teriflunomide for multiple sclerosis(50). Brequinar, another potent inhibitor of human DHODH,was tested in the 1990s in clinical trials for solid tumors but wasassociated with myeloid suppression and had a limited thera-peutic window (18, 22). These DHODH inhibitors are associated

with side effects believed tobedue to their off-target activities suchas inhibition of kinases (21, 22). Recently, the inhibition ofDHODH by GSK983, which is structurally related to PTC299,was reported (51). However, we determined that this compoundhas a considerably lower bioavailability in mice than doesPTC299 (Supplementary Fig. S7).

PTC299 represents a novel class of DHODH inhibitors withfavorable biochemical and pharmacologic properties that maydistinguish it from previously published DHODH inhibitors: (1)Unlike the well-known DHODH inhibitors teriflunomide andbrequinar, PTC299 did not show off-target effects including anyovert effects on 205 kinases and 62 Novascreen pharmacologictargets tested (Supplementary Table S5) and did not result inmyeloid suppression in patients with solid tumors (28, 29); (2)PTC299 has been extensively tested and well-tolerated in preclin-ical studies. PTC299 did not inhibit the proliferation of normalprimary cells including hematopoietic stem cells at concentra-tions 1,000-fold greater than its IC50 in tumor cells (Supplemen-tary Table S5); (3) DHODH inhibitors were identified by othersusing in vitro enzymatic assay in which the purified enzymeconformation may differ from that found in the cells due to lackof lipids in the assay (25, 26). The potent activity of PTC299 inmitochondrial preparations is likely due to the presence of thelipid bilayer when the enzyme is in themitochondrial membranethat may facilitate the entry and/or docking of the lipophilicPTC299 into the binding pocket of the DHODH enzyme. Thisis consistent with the recent reports that interactions with thesmall lipophilic pocket in DHODH are critical for potent inhi-bition of its enzymatic activity (23, 26); (4) PTC299-resistantHT1080 cells are not cross-resistant toDHODH inhibitors such asbrequinar or teriflunomide (Supplementary Fig. S6), indicatingthat the mode of action for PTC299 is different from that of otherDHODHinhibitors even though they target the samedocking site;(5) It was demonstrated that inhibition of DHODH activates p53expression resulting in inhibition of cancer cell proliferation andapoptosis (11). However, we analyzed the p53 expression in theCCLE database and found no correlation of p53 expression withthe activity of PTC299 (IC50) in those cell lines. Dr. KensukeKojima (Kyoto University, Japan) tested PTC299 in a panel ofleukemia cell lines and also found that PTC299 activity does notcorrelate with the expression of p53 (personal communication);and finally, (6) a docking model shows that PTC299 bindsDHODH in the same pocket and with similar strength as doesbrequinar analog C44. However, the interaction of PTC299 withthe binding pocket is mainly a "hand-in-glove," nonpolar lipo-philic interaction (Fig. 3H). These findings demonstrate thatPTC299 is structurally and functionally different from previouslypublished DHODH inhibitors, thus it may be a useful new toolfor preclinical and clinical investigation of targeting DHODH fortherapeutic uses in treatment of cancer and immune diseases.

The elucidation of the mechanism of action of PTC299 willfacilitate the optimization of biomarker identification, patientselection, and design of combination therapies with other drug(s)for clinical development. Here, our data together point to thepotential clinical utility of PTC299 for the treatment of cancerssuch as leukemia and lymphoma which in general have lowerpyrimidine salvage activity and thus depend on de novo pyrimi-dine synthesis for uncontrolled proliferation. PTC299 was placedon clinical hold following the occurrence of severe hepatotoxicityin 2 patients of 279 individuals administered with PTC299 (28,29). Based on recent preclinical assessments demonstrating

Cao et al.

Mol Cancer Ther; 18(1) January 2019 Molecular Cancer Therapeutics14

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 13: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

antitumor activity in AMLmodels at low doses of PTC299, we arereevaluating the potential use of PTC299 in leukemia and lym-phoma, anticipating that efficacious concentrationof PTC299willbe lower than those achieved in previous clinical studies in solidtumors.

ConclusionPTC299 is a novel potent inhibitor of DHODH that is differ-

entiated from and has advantages over other DHODH inhibitors.Studies demonstrated the target engagement of PTC299 inpatients, thus supporting its potential for treatment of hemato-logic malignancies which rely on de novo pyrimidine nucleotidesynthesis for survival and proliferation.

Disclosure of Potential Conflicts of InterestThe authors are or were employed by PTC Therapeutic and have received

salary compensation for time, effort, and hold or held financial interest in thecompany. Travel expenses related to attendance at scientific conferences topresent datawere reimbursedbyPTCTherapeutics, Inc. All patents related to thework presented here are held by PTC Therapeutics, Inc.

Authors' ContributionsConception and design: L. Cao, M. Weetall, C. Trotta, C. Romfo, T. Arasu, S.Hwang, R. Kong, T.W. Davis, J.M. Colacino, S.W. PeltzDevelopment of methodology: L. Cao, M. Weetall, J. Ma, M.J. Kim, B. Furia, C.Romfo, T. Arasu, S. Hwang, W. Lennox, R. Kong, S.W. PeltzAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): L. Cao, M. Weetall, J. Ma, M.J. Kim, B. Furia,

C. Romfo, J.D. Graci, J. Sheedy, J. Hedrick, N. Risher, S. Yeh, H. Qi, T. Arasu,R. Kong, A. Jacobson, J.M. Colacino, S.W. PeltzAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): L. Cao, M. Weetall, C. Trotta, J. Ma, C. Romfo,J.D. Graci, W. Li, J. Du, S. Yeh, H. Qi, T. Arasu, R. Kong, J. Petruska, A. Jacobson,A. Branstrom, J.M. Colacino, S.W. PeltzWriting, review, and/or revision of the manuscript: L. Cao, M. Weetall,C. Trotta, J. Ma, M.J. Kim, B. Furia, C. Romfo, J.D. Graci, W. Li, J. Du, J. Petruska,J. Babiak, N.G. Almstead, A. Branstrom, J.M. Colacino, S.W. PeltzAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): L.Cao,M.Weetall, C. Romfo,H.Qi, A. Jacobson,N.G. Almstead, A. Branstrom, J.M. ColacinoStudy supervision: L. Cao,M.Weetall, J. Sheedy, R. Kong, Y.-C.Moon, J. Babiak,N.G. Almstead, J.M. Colacino, S.W. PeltzOther (performed laboratory experiments): K. CintronOther (medicinal chemist): H. QiOther (originally designed and synthesized PTC299 molecule): T. Arasu

AcknowledgmentsWe thank all the members of the Discovery group at PTC for the thoughtful

discussion and constructive advises throughout the PTC299 program.We thankJana Narasimhan for a critical reading and discussion of this article. Thisprogram was partially supported by funding from NCI to J.M. Colacino andL. Cao (2 R44 CA108330-02).

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

ReceivedAugust 1, 2018; revised August 22, 2018; acceptedOctober 17, 2018;published first October 23, 2018.

References1. Martinez-Outschoorn UE, Peiris-Pages M, Pestell RG, Sotgia F, Lisanti MP.

Cancer metabolism: a therapeutic perspective. Nat Rev Clin Oncol2017;14:113.

2. Brown KK, Spinelli JB, Asara JM, Toker A. Adaptive reprogramming of denovo pyrimidine synthesis is a metabolic vulnerability in triple-negativebreast cancer. Cancer Discov 2017;7:391–9.

3. McMahon SB. Control of nucleotide biosynthesis by theMYConcoprotein.Cell Cycle 2008;7:2275–6.

4. Mathur D, Stratikopoulos E, Ozturk S, Steinbach N, Pegno S, SchoenfeldS, et al. PTEN regulates glutamine flux to pyrimidine synthesis andsensitivity to dihydroorotate dehydrogenase inhibition. Cancer Discov2017;7:380–90.

5. Robitaille AM, Christen S, Shimobayashi M, Cornu M, Fava LL, Moes S,et al. Quantitative phosphoproteomics reveal mTORC1 activates de novopyrimidine synthesis. Science 2013;339:1320–3.

6. Rawls J, Knecht W, Diekert K, Lill R, Loffler M. Requirements for themitochondrial import and localization of dihydroorotate dehydrogenase.Eur J Biochem 2000;267:2079–87.

7. Vyas VK, Ghate M. Recent developments in the medicinal chemistry andtherapeutic potential of dihydroorotate dehydrogenase (DHODH) inhi-bitors. Mini Rev Med Chem 2011;11:1039–55.

8. Hoffmann HH, Kunz A, Simon VA, Palese P, Shaw ML. Broad-spectrumantiviral that interferes with de novo pyrimidine biosynthesis. Proc NatlAcad Sci U S A 2011;108:5777–82.

9. KoundinyaM, Sudhalter J, Courjaud A, Lionne B, Touyer G, Bonnet L, et al.Dependence on the pyrimidine biosynthetic enzyme DHODH is a syn-thetic lethal vulnerability in mutant KRAS-driven cancers. Cell Chem Biol2018;25:705–717.e11.

10. Mohamad Fairus AK, Choudhary B, Hosahalli S, Kavitha N, Shatrah O.Dihydroorotate dehydrogenase (DHODH) inhibitors affect ATP deple-tion, endogenous ROS and mediate S-phase arrest in breast cancer cells.Biochimie 2017;135:154–63.

11. Ladds MJGW, van Leeuwen IMM, Drummond CJ, Chu S, Healy AR,Popova G, et al. A DHODH inhibitor increases p53 synthesis andenhances tumor cell killing by p53 degradation blockage. Nat Commun2018;9:1107.

12. Dorasamy MS, Choudhary B, Nellore K, Subramanya H, Wong PF.Dihydroorotate dehydrogenase inhibitors target c-Myc and arrest mel-anoma, myeloma and lymphoma cells at S-phase. J Cancer 2017;8:3086–98.

13. Zhang X, Yang J, Chen M, Li L, Huan F, Li A, et al. Metabolomics profilesdelineate uridine deficiency contributes to mitochondria-mediated apo-ptosis induced by celastrol in human acute promyelocytic leukemia cells.Oncotarget 2016;7:46557–72.

14. Sykes DB, Kfoury YS, Mercier FE, Wawer MJ, Law JM, Haynes MK, et al.Inhibition of dihydroorotate dehydrogenase overcomes differentiationblockade in acute myeloid leukemia. Cell 2016;167:171–86.

15. Lewis TA, Sykes DB, Law JM, Munoz B, Rustiguel JK, Nonato MC, et al.Development of ML390: a human DHODH inhibitor that induces differ-entiation in acute myeloid leukemia. ACSMed Chem Lett 2016;7:1112–7.

16. Wu D, Wang W, Chen W, Lian F, Lang L, Huang Y, et al. Pharmacologicinhibition of dihydroorotate dehydrogenase induces apoptosis anddifferentiation in acute myeloid leukemia cells. Haematologica 2018;103:1472–83.

17. Baumann P,Mandl-Weber S, Volkl A, AdamC, Bumeder I, Oduncu F, et al.Dihydroorotate dehydrogenase inhibitor A771726 (leflunomide) inducesapoptosis and diminishes proliferation of multiple myeloma cells. MolCancer Ther 2009;8:366–75.

18. Cody R, Stewart D, DeForni M, Moore M, Dallaire B, Azarnia N, et al.Multicenter phase II study of brequinar sodium in patients with advancedbreast cancer. Am J Clin Oncol 1993;16:526–8.

19. Fitzpatrick LR, Small JS, Doblhofer R, Ammendola A. Vidofludimusinhibits colonic interleukin-17 and improves hapten-induced colitis inrats by a unique dual mode of action. J Pharmacol Exp Ther 2012;342:850–60.

20. Arteaga CL, Brown TD, Kuhn JG, Shen HS, O'Rourke TJ, Beougher K, et al.Phase I clinical and pharmacokinetic trial of Brequinar sodium (DuP 785;NSC 368390). Cancer Res 1989;49:4648–53.

21. Xu X, Williams JW, Gong H, Finnegan A, Chong AS. Two activities of theimmunosuppressive metabolite of leflunomide, A77 1726. Inhibition ofpyrimidine nucleotide synthesis and protein tyrosine phosphorylation.Biochem Pharmacol 1996;52:527–34.

PTC299 Is a Novel Potent DHODH Inhibitor

www.aacrjournals.org Mol Cancer Ther; 18(1) January 2019 15

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 14: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

22. Xu X, Williams JW, Shen J, Gong H, Yin DP, Blinder L, et al. Invitro and in vivo mechanisms of action of the antiproliferative andimmunosuppressive agent, brequinar sodium. J Immunol 1998;160:846–53.

23. Sainas S, Pippione AC, Giorgis M, Lupino E, Goyal P, Ramondetti C,et al. Design, synthesis, biological evaluation and X-ray structuralstudies of potent human dihydroorotate dehydrogenase inhibitorsbased on hydroxylated azole scaffolds. Eur J Med Chem 2017;129:287–302.

24. Lucas-Hourani M, Munier-Lehmann H, El MF, Malmquist NA, Harpon J,Coutant EP, et al. Original 2-(3-Alkoxy-1H-pyrazol-1-yl)azines inhibitorsof human dihydroorotate dehydrogenase (DHODH). J Med Chem2015;58:5579–98.

25. Basso LGM, Mendes LFS, Costa-Filho AJ. The two sides of a lipid-proteinstory. Biophys Rev 2016;8:179–91.

26. Costeira-Paulo J, Gault J, Popova G, Ladds MJGW, van Leeuwen IMM, SarrM, et al. Lipids shape the electron acceptor-binding site of the peripheralmembrane protein dihydroorotate dehydrogenase. Cell Chem Biol2018;25:309–17.

27. Cao L, Weetall M, Bombard J, Qi H, Arasu T, Lennox W, et al. Discoveryof novel small molecule inhibitors of expression in tumor cells using acell-based high throughput screening platform. PLoS One 2016;11:e0168366.

28. Weetall M, Davis T, Elfring G, Northcutt V, Cao L, Moon YC, et al. Phase 1study of safety, tolerability, and pharmacokinetics of PTC299, an inhibitorof stress-regulated protein translation. Clin Pharmacol Drug Dev 2016;5:296–305.

29. Packer RJ, Rood BR, Turner DC, Stewart CF, FisherM, SmithC, et al. Phase Iandpharmacokinetic trial of PTC299 inpediatric patientswith refractoryorrecurrent central nervous system tumors: a PBTC study. J Neurooncol2015;121:217–24.

30. Clayton DA, Shadel GS. Isolation of mitochondria from cells and tissues.Cold Spring Harb Protoc 2014;2014:db.

31. Chen SF, Perrella FW, Behrens DL, Papp LM. Inhibition of dihydroor-otate dehydrogenase activity by brequinar sodium. Cancer Res 1992;52:3521–7.

32. Ruppert J, Welch W, Jain AN. Automatic identification and representa-tion of protein binding sites for molecular docking. Protein Sci 1997;6:524–33.

33. Cheatham TE III, Cieplak P, Kollman PA. Amodified version of the Cornellet al. force field with improved sugar pucker phases and helical repeat. JBiomol Struct Dyn 1999;16:845–62.

34. Prewett M, Huber J, Li Y, Santiago A, O'Connor W, King K, et al. Anti-vascular endothelial growth factor receptor (fetal liver kinase 1) mono-clonal antibody inhibits tumor angiogenesis and growth of several mouseand human tumors. Cancer Res 1999;59:5209–18.

35. Lichtenbeld HC, Ferarra N, Jain RK, Munn LL. Effect of local anti-VEGFantibody treatment on tumor microvessel permeability. Microvasc Res1999;57:357–62.

36. Ohnuma T, Roboz J, Shapiro ML, Holland JF. Pharmacological andbiochemical effects of pyrazofurin in humans. Cancer Res 1977;37:2043–9.

37. Jain AN. Effects of protein conformation in docking: improved poseprediction through protein pocket adaptation. J Comput Aided Mol Des2009;23:355–74.

38. Di RF, Azzaro MP, Palumbo GA, Bagnato S, Stagno F, Giustolisi GM, et al.Elevated vascular endothelial growth factor (VEGF) serum levels in idio-pathic myelofibrosis. Leukemia 2001;15:976–80.

39. Duley JA, HenmanMG, Carpenter KH, BamshadMJ, Marshall GA, Ooi CY,et al. Elevated plasma dihydroorotate in Miller syndrome: Biochemical,diagnostic and clinical implications, and treatment with uridine. MolGenet Metab 2016;119:83–90.

40. Lane AN, Fan TW. Regulation of mammalian nucleotide metabolism andbiosynthesis. Nucleic Acids Res 2015;43:2466–85.

41. Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S,et al. The Cancer Cell Line Encyclopedia enables predictive modelling ofanticancer drug sensitivity. Nature 2012;483:603–7.

42. Wang X, Harrison JS, Studzinski GP. Enhancement of arabinocytosine(AraC) toxicity to AML cells by a differentiation agent combination.J Steroid Biochem Mol Biol 2016;164:72–8.

43. Cunningham JT, Moreno MV, Lodi A, Ronen SM, Ruggero D. Protein andnucleotide biosynthesis are coupled by a single rate-limiting enzyme,PRPS2, to drive cancer. Cell 2014;157:1088–103.

44. Ipata PL, Falcone G, Serra MC. Some regulatory properties of cytidinedeaminase and uridine phosphorylase of Bacillus cereus. FEBS Lett 1970;10:67–70.

45. Gemble S, Ahuja A, Buhagiar-Labarchede G, Onclercq-Delic R, Dairou J,Biard DS, et al. Pyrimidine pool disequilibrium induced by a cytidinedeaminase deficiency inhibits PARP-1 activity, leading to the under rep-lication of DNA. PLoS Genet 2015;11:e1005384.

46. Jaafar ZA, Oguro A, Nakamura Y, Kieft JS. Translation initiation by thehepatitis C virus IRES requires eIF1A and ribosomal complex remodeling.Elife 2016;5.

47. Wu G, Radwan MK, Xiao M, Adachi H, Fan J, Yu YT. The TOR signalingpathway regulates starvation-induced pseudouridylation of yeast U2snRNA. RNA 2016;22:1146–52.

48. Rocchi L, Pacilli A, Sethi R, Penzo M, Schneider RJ, Trere D, et al. Dyskerindepletion increases VEGF mRNA internal ribosome entry site-mediatedtranslation. Nucleic Acids Res 2013;41:8308–18.

49. EmmanuelN, Ragunathan S, ShanQ,Wang F, GiannakouA,HuserN, et al.Purine nucleotide availability regulatesmTORC1 activity through the RhebGTPase. Cell Rep 2017;19:2665–80.

50. Schiff MH, Strand V, Oed C, Loew-Friedrich I. Leflunomide: efficacy andsafety in clinical trials for the treatment of rheumatoid arthritis. DrugsToday (Barc) 2000;36:383–94.

51. Deans RM, Morgens DW, Okesli A, Pillay S, Horlbeck MA, Kampmann M,et al. Parallel shRNA and CRISPR-Cas9 screens enable antiviral drug targetidentification. Nat Chem Biol 2016;12:361–6.

Mol Cancer Ther; 18(1) January 2019 Molecular Cancer Therapeutics16

Cao et al.

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863

Page 15: Targeting of Hematologic Malignancies with PTC299, A Novel ... · salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased

2019;18:3-16. Published OnlineFirst October 23, 2018.Mol Cancer Ther   Liangxian Cao, Marla Weetall, Christopher Trotta, et al.   Pharmaceutical PropertiesPotent Inhibitor of Dihydroorotate Dehydrogenase with Favorable Targeting of Hematologic Malignancies with PTC299, A Novel

  Updated version

  10.1158/1535-7163.MCT-18-0863doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2018/10/23/1535-7163.MCT-18-0863.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/18/1/3.full#ref-list-1

This article cites 49 articles, 12 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/18/1/3.full#related-urls

This article has been cited by 1 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/18/1/3To request permission to re-use all or part of this article, use this link

on August 11, 2020. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863