Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Sep 10.
Published in final edited form as: Leukemia. 2022 Mar 10;36(5):1313–1323. doi: 10.1038/s41375-022-01536-x

In Vivo Anti-Tumor Effect of PARP Inhibition in IDH1/2 Mutant MDS/AML Resistant to Targeted Inhibitors of Mutant IDH1/2

Rana Gbyli 1,*, Yuanbin Song 1,2,*,#, Wei Liu 1,*, Yimeng Gao 1,*, Giulia Biancon 1, Namrata S Chandhok 1,3, Xiaman Wang 1,4, Xiaoying Fu 1,5, Amisha Patel 1, Ranjini Sundaram 6, Toma Tebaldi 1, Padmavathi Mamillapalli 1, Amer M Zeidan 1, Richard A Flavell 7,8, Thomas Prebet 1, Ranjit S Bindra 6,9, Stephanie Halene 1,10,#
PMCID: PMC9103411  NIHMSID: NIHMS1782098  PMID: 35273342

Abstract

Treatment options for patients with relapsed/ refractory acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) are scarce. Recurring mutations, such as mutations in isocitrate dehydrogenase-1 and −2 (IDH1/2) are found in subsets of AML and MDS, are therapeutically targeted by mutant enzyme-specific small molecule inhibitors (IDHmi). IDH mutations induce diverse metabolic and epigenetic changes that drive malignant transformation. IDHmi alone are not curative and resistance commonly develops, underscoring the importance of alternate therapeutic options. We were first to report that IDH1/2 mutations induce a homologous recombination (HR) defect which confers sensitivity to poly (ADP)-ribose polymerase inhibitors (PARPi). Here, we show that the PARPi olaparib is effective against primary patient-derived IDH1/2-mutant AML/ MDS xeno-grafts (PDXs). Olaparib efficiently reduced overall engraftment and leukemia-initiating cell frequency as evident in serial transplantation assays in IDH1/2-mutant but not -wildtype AML/MDS PDXs. Importantly, we show that olaparib is effective in both IDHmi-naïve and -resistant AML PDXs, critical given the high relapse and refractoriness rates to IDHmi. Our pre-clinical studies provide a strong rationale for the translation of PARP inhibition to patients with IDH1/2-mutant AML/ MDS, providing an additional line of therapy for patients who do not respond to or relapse after targeted mutant IDH inhibition.

Introduction

The isocitrate dehydrogenase-1 and −2 (IDH1/2) genes encode key metabolic enzymes in the Krebs cycle that catalyze the reversible conversion of isocitrate to alpha-ketoglutarate (αKG) 1. IDH1/2 mutations are found in approximately 20% of AML and in 4 to 12% of MDS 2, 3. Somatic missense mutations in IDH1/2 occur within the catalytic sites at arginine residues R132 for IDH1 (the cytoplasmic isoform) and R140 or R172 for IDH2 (the mitochondrial isoform) 24. IDH1/2 enzymes function as homodimers, and when mutated, acquire the neomorphic function as heterodimers to catalyze the conversion of αKG to 2hydroxyglutarate (2HG) 1. 2HG is an oncometabolite that has been shown to promote leukemogenesis via the competitive inhibition of αKG-dependent dioxygenases involved in the regulation of DNA and histone methylation and other functions 58. Given their important role in tumor initiation and progression, mutant IDH1/2 enzymes represent an attractive therapeutic target leading to the development of selective small molecule inhibitors 9. Ivosidenib and enasidenib, inhibitors of mutant IDH1 and IDH2, respectively, have been approved in the treatment of adults with AML and MDS 10, 11, however, when given as single agents are not curative, with complete response (CR) rates and median overall survival (OS) ranging between 20–30% and ~8–12 months, respectively 12. IDHmi induce cell differentiation 2, 13 with persistence of the mutant IDH1/2 clones even in patients who achieve CR 14. Resistance to IDHmi is notable for either persistent or restored 2HG production 1518.

We previously reported that mutant IDH1/2-induced 2HG accumulation induces a HR defect that confers sensitivity to PARPi 19. Mechanistically, 2HG-induced inhibition of the lysine demethylase KDM4B results in aberrant hypermethylation of histone 3 lysine 9 (H3K9) at loci surrounding DNA breaks, masking the local H3K9 trimethylation signal essential for DNA double-strand break (DSB) repair via HR 20. Tip60 and ATM fail to localize to DSBs, which results in impaired end-resection and recruitment of downstream HR factors. We identified similar oncogenic mechanisms secondary to TCA cycle gene mutations in fumarate hydratase and succinate dehydrogenase deficient cancers, highlighting the tumor agnostic nature of this “oncometabolite-induced BRCAness” and its potential as an exploitable vulnerability across different cancers 21 including leukemia and MDS. 2HG-induced HR deficiency renders cancer cells unable to resolve DSBs and sensitive to additional inhibition of single strand break (SSB) repair by PARPi 9, 19. In vivo pre-clinical studies are necessary to bring novel concepts to the clinic and prove the validity of exploiting the 2HG-induced BRCAness defect with PARPi 19, 22, 23.

Here we show that mutant IDH1 and IDH2 confer sensitivity to PARPi in vivo not only in IDHmi naïve but also in resistant MDS/AML. Using genetically matched patient-derived IDH1/2 mutant and wildtype MDS/AML PDX models in cytokine humanized immunodeficient mice that recapitulate the mutational heterogeneity of AML/MDS 24, we demonstrate that the PARP inhibitor olaparib significantly reduces MDS/AML engraftment and leukemia-initiating cell (LIC) frequency in IDH1/2 mutant but not wildtype MDS/AML. This sensitivity is maintained in IDHmi resistant AML irrespective of co-occurring mutations. Our findings provide the rationale for the incorporation of PARPi into the treatment armamentarium for MDS/AML and the basis upon which to design combination therapies for relapsed/refractory and possibly treatment for naïve IDH1/2 mutant MDS/AML.

Materials and Methods

Primary patient samples

All patients’ PB and BM samples were obtained with donor’s written consent and accessed from the Yale hematology tissue bank. All human studies were approved by the Yale University Human Investigation Committee. Mononuclear cells were isolated using Ficoll-Paque separation (GE Healthcare, Cat#17–1440-03) and cryopreserved in fetal bovine serum (Gemini-Bioproducts, Cat#100–106) with 10% dimethyl sulfoxide.

Animal studies

All animal experiments were approved by and carried out in compliance with the Yale Institutional Animal Care and Use Committee protocols.

Primary murine AML model:

FLT3ITD/WT IDH2R140Q/WT mice were a gift from Dr. Ross Levine’s lab and have been previously described 25. Mx1Cre+ Flt3ITD/WT IDH2R140Q/WT mice were treated every other day with Polyinosinic: polycytidylic acid [poly (I:C)] (Calbiochem, Cat#528906) via intra-peritoneal (IP) injection at a dose of 20μg/g starting 4–6 weeks after birth. 6–9 months later, donor mice were euthanized and BM cells harvested from femurs and tibias, and lineage depleted using anti-mouse lineage cell detection cocktail antibody and BD IMag Streptavidin Particles Plus (BD Biosciences, Cat# 557812, RRID: AB_10050580).

MISTRG patient derived xenotransplantation (PDX) studies:

MISh/hTRG mice (CSF1h/h IL3/CSF2h/h SIRPAh/h THPOh/h Rag2−/− Il2rg−/−) were bred to MISm/mTRG mice (CSF1h/h IL3/CSF2h/h Sirpam/m THPOh/h Rag2−/− Il2rg−/−) to generate human cytokine homozygous and hSIRPA heterozygous mice (MISh/mTRG, labeled MISTRG throughout the study) 24, 26. Mice were maintained on continuous treatment with enrofloxacin in the drinking water (0.27 mg/mL, Baytril, Bayer Healthcare). MISTRG mice have been deposited at the Jackson laboratory. IDH1/2 mutant and wild type AML patient samples were transplanted into newborn (3–5 days) MISTRG mice after irradiation twice with 150 cGy (XRAD 320, PXI X-Ray Systems) 4 hours apart. AML patient PB or BM mononuclear cells were incubated with anti-human CD3 antibody (clone Okt3, BioXCell, Cat# BE0001–2, RRID: AB_1107632) at 5 μg/100 μl for 30min prior to injection. Cells were injected intra-hepatically in a volume of 20 μL with a 22-gauge Hamilton needle and Hamilton syringe (Hamilton, Reno). Mice were analyzed at time points indicated via flowcytometry as previously described 24. For secondary transplantation, cryopreserved BM cells from primary MISTRG recipient were thawed and depleted of murine cells via negative selection of murine CD45+ and Ter119+ cells by magnetic labeling with biotin-anti-mouse CD45 (clone 30-F11, Biolegend, Cat#103103, RRID: AB_312968) and biotin anti-mouse TER119 (clone TER-119, Biolegend, Cat#116204, RRID: AB_313705) and BD IMag Streptavidin Particles Plus (BD Biosciences, Cat# 557812, RRID: AB_10050580). Purified cells were incubated with anti-human CD3 antibody and injected intra-hepatically into MISTRG mice as described above. 8 to 12 weeks (AML) and 12 to 16 weeks (MDS) post transplantation, BM was aspirated from transplanted mice to quantitate human CD45+ engraftment levels by flow cytometry and to assign mice to treatment groups based on equal engraftment levels. For tertiary transplantation to determine LIC frequency, equal numbers of huCD45+ cells obtained from vehicle or drug treated mice were transplanted into newborn recipients, and analysis was performed at 16 weeks post transplantation.

Drug treatment

Olaparib (AZD2281) (Selleckchem, Cat#S1060) was dissolved in PBS containing 10% (w/v) 2-hydroxy-propyl-beta-cyclodextrin (Sigma) at a concentration of 20mg/ml. For in vivo studies, olaparib was administered IP at 100 mg/kg once daily. Enasidenib was purchased from LeadGen Labs, USA and dissolved in 0.5% methylcellulose and 0.2% Tween 80 in PBS at a concentration of 4 mg/ml and administered via oral gavage at 40 mg/kg once daily. 2-HG was measured in plasma before and after treatment with vehicle, enasidenib, or olaparib in triplicates with the D-2-Hydroxyglutarate (D2HG) assay kit (Biovision, Cat#K970–100, Sigma, Cat# MAK320–1KT) according to manufacturer’s protocol.

Flow cytometric analysis of human cell engraftment in MISTRG mice

Engraftment of human CD45+ cells and their subsets were determined by flow cytometry. In brief, cells were isolated from engrafted mice, blocked with human BD Fc block antibody (BD Biosciences, Cat#564219, RRID: AB_2728082) and mouse BD Fc block (2.4G2, BD Pharmingen, Cat#553141, RRID: AB_394656), and stained with combinations of antibodies purchased from Biolegend: Hematopoietic Stem and Progenitor Cell (HSPC) panel: APC/Cy7 mCD45 (30-F11, 1:300, Cat#103116, RRID: AB_312981), APC/Cy7 mTer119 (Ter-119, 1:300, Cat#116223, RRID: AB_2137788), BV510 hCD45 (HI30, 1:100, Cat#304036, RRID: AB_2561940), BV421 huCD38 (HIT2, 1:100, Cat#303526, RRID: AB_10983072), PE huCD34 (561, 1:100, Cat#343606, RRID:AB_1732008), PE/Cy7 huCD10 (HI10a, 1:100, Cat#312214, RRID: AB_2146548). Human cell engraftment panel: APC/Cy7 mCD45 (30-F11, 1:300), APC/Cy7 mTer119 (Ter-119, 1:300), BV510 hCD45 (HI30, 1:100), FITC huCD3 (OKT3, 1:100, Cat#317306, RRID: AB_571907), PE/Cy7 huCD19 (HIB19, 1:100, Cat#302216, RRID: AB_314246), APC huCD33 (WM53, 1:100, Cat#983902, RRID: AB_2810824), PE huCD34 (561, 1:100). Data were acquired with FACSDiva on an LSR Fortessa (BD Biosciences) equipped with 5 lasers and analyzed with FlowJo V10 software.

Histologic analysis

Tissues were fixed in BBC Biochemical B-PLUS FIX Fixative solution (Thermo Fisher Scientific, Cat#NC9496646) and embedded in paraffin. Femurs were decalcified with Formic Acid Bone Decalcifier (Decal Chemical, NY, USA). Paraffin blocks were sectioned at 4 μm and stained with hematoxylin and eosin (H&E) and antigen-specific antibodies by the Yale Clinical Pathology and Yale Pathology Tissue Services. Antibodies: human CD45, Leucocyte Common Antigen, (clone PD7/26+2B11, Dako, Cat#GA75161–2). Images were acquired using a Nikon Eclipse 80i microscope.

DNA cloning and cell culture

Site-directed mutagenesis to introduce the IDH2Q316E and IDH2I319M mutations was performed using the QuikChange II Site-Directed Mutagenesis Kit (Agilent Technologies, Cat#200521) with primers (IDH2Q316E Forward: CTATGACGGAGATGTGGAGTCAGACATCCTGGC; IDH2Q316E Reverse: GATACTGCCTCTACACCTCAGTCTGTAGGACCG; IDH2I319M Forward: AGATGTGCAGTCAGACATGCTGGCCCAGG; IDH2I319M Reverse: CCTGGGCCAGCATGTCTGACTGCACATCT). Wild type (WT) and mutant IDH2 were cloned into the LeGO-iC plasmid, a gift from Boris Fehse (Addgene plasmid, #27462) and verified by Sanger sequencing. Lentiviral particles were produced by co-transfection with transgene, VSVG and psPAX2 (gifts from Tannishtha Reya (Addgene plasmid # 14888 and #12260) in HEK293T cells (ATCC Cat# CRL-3216) using polyethylenimine transfection reagent (Polysciences Cat# 23966–2) followed by spin-concentration (Beckman, 27,000xg, 90min). Lineage depleted murine Flt3ITDIDH2R140Q BM cells were pre-cultured with 8 μM Cyclosporine H (Sigma, SML1575) for 4 hours and then infected with viral particles via spinfection (1000xg at 25C for 1 hour) in StemSpan™ medium (Stem Cell Technologies) with addition of 8 μg/ml polybrene (Sigma) and recombinant murine cytokines (20 ng/ml Interleukin-3 (mIL-3, Cat#300–324P), 100 ng/ml Stem Cell Factor (mSCF, Cat#300–348P), 50 ng/ml Thrombopoietin (mTPO, Cat# 300–351P), 100 ng/mL Flt-3 Ligand (mFLT-3L, Cat#300–306P)) and 10% Penicillin-Streptomycin (Sigma). All cytokines were purchased from Gemini-Bioproducts. Cells were sorted for mCherry fluorescence 72 hours after transduction on a FACS Aria (BD Biosciences). Sorted cells were plated in methocult (StemCell Technologies Cat# M3434) in the presence of DMSO, enasidenib (50nM), or olaparib (1.25μM) in 35mm dishes in triplicates at a density of 1.2 × 104 cells per ml. Colonies were counted after 7 days and serially re-plated and re-counted.

Targeted next generation sequencing

Genomic DNA (gDNA) was extracted from BM (diagnosis, Y1597) or PB (relapse, Y2260) mononuclear cells using QIAGEN DNeasy Blood & Tissue kit according to manufacturer’s instructions. Concentration and purity of the extracted gDNA were assessed using NanoDrop 1000 spectrophotometer (Thermo Scientific). During library preparation, IDT’s xGen Acute Myeloid Leukemia Cancer Panel v1.0 (Integrated DNA Technologies) was used for the targeted enrichment of 264 genes associated with AML (11731 probes, 1.19 Mb of target regions from 6235 exons). Hybridization capture of DNA libraries and sequencing (Illumina HiSeq, paired-end 100bp) were performed at the Yale Center for Genome Analysis (YCGA). Raw FASTQ files were subjected to quality control through FastQC v0.11.5 27. Sequencing reads were then aligned to the human genome (GRCh38.p10) with Burrows-Wheeler Aligner (BWA-MEM, v0.7.15) using default parameters 28. Duplicate reads were removed with Picard v2.9.0 (http://broadinstitute.github.io/picard/). The number of total aligned reads before and after duplicate removal was obtained using SAMtools 29 v1.5. Median coverage at each position in each enriched targeted region was calculated using BEDTools v2.27.1 30. Single-nucleotide variants (SNVs) and indels were detected with Genome Analysis Toolkit (GATK) v4.0.6.0 (https://github.com/broadinstitute/gatk) running Mutect2 31 in tumor-only mode after base quality recalibration. Variants were annotated using ANNOVAR (2019Oct24) 32 and high-confidence variants were filtered as follows: exclusion of false positives (technical artifacts, germline variants and sequencing errors) through GATK4 filtering; exclusion of variants with synonymous or unknown effect; selection of deleterious variants (FATHMM-MKL prediction score) 33; selection of variants annotated in COSMIC v90 34. This last filter was not applied to IDH2 variants to deeply explore regions potentially affected by PARPi treatment. All reported variants were visually inspected with the Integrative Genomics Viewer (IGV) 35.

Statistical analysis

Data was analyzed using Prism 8 (GraphPad Software, La Jolla, CA) with the use of paired Ratio t-test and unpaired Mann-Whitney U test as indicated. P value was considered significant at values less than 0.05, n.s. not significant, * statistically significant (p<0.05), ** statistically significant (p<0.01), *** statistically significant (p<0.001), **** statistically significant (p<0.0001).

Data availability

All relevant data are available from the authors upon reasonable request.

Results

Enhanced PARPi sensitivity in IDH2-mutant AML in vivo

Our earlier work on the oncometabolite 2HG suggested that the HR defect and resultant PARPi sensitivity would be tumor agnostic 1921.

To determine whether PARPi would be efficacious in primary human MDS/AML within the context of their clonal complexity and variable co-mutational patterns, we generated a panel of IDH1/2-mutant and matched wildtype MDS and serially transplantable AML PDXs in cytokine humanized immunodeficient MISTRG mice (Table 1 and Supplementary Fig. S1A). MISTRG mice express physiologic levels of human cytokines (M-CSFh/h, IL-3/GM-CSFh/h and hTPOh/h) on the RAG2−/− γc−/− background with additional humanization of the CD47 receptor hSIRPα 26, 36; they support the engraftment, maintenance, and differentiation of AML and other myeloid malignancies 24, 37. 12–16 weeks post transplantation we confirmed primary engraftment levels of human CD45+ (huCD45+) AML in MISTRG mice (Supplementary Fig. S1B, C) and generated secondary recipients for drug administration as described previously 24. Eight to twelve weeks after secondary transplantation, MISTRG mice engrafted with newly diagnosed (Y566, Y577) or relapsed/refractory (Y747) IDH2R140Q human AML were assigned to either vehicle or olaparib based on similar engraftment levels determined by BM aspiration and dosed intraperitoneally with vehicle or olaparib (100mg/kg) 6 days a week for 3 weeks. While in vehicle treated mice leukemia burden (Fig. 1AC) and 2HG levels in plasma (Fig. 1D) rose sharply, especially for Y577 and Y747, engraftment and 2HG levels in olaparib treated mice remained stable or fell compared to pre-treatment levels (Fig. 1AD and Supplementary figure S1DF). Representative histologic assessment of huCD45+ engraftment confirmed the olaparib effect on BM leukemic burden (Fig. 1E). Increased γH2AX+ huCD45 cells in bone marrow confirmed that olaparib induced DNA damage in IDH mutant cells (Fig. 1F). To assure that olaparib toxicity against human AML was specific to IDH mutant AML, we engrafted MISTRG mice with IDH wildtype AML and treated mice as before with vehicle or olaparib (100mg/kg) 6 days a week for 3 weeks. In MISTRG mice engrafted with IDH wildtype AML (Y689, Y406, and Y456) olaparib administration showed no benefit when compared to vehicle regardless of mutational composition, with significantly increased PB and BM tumor burden compared to pre-treatment bone marrow aspiration levels (Supplementary Fig. S2AC).

Table 1. Clinical characterization of the selected IDH2R140Q and IDHWT AML/MDS patients.

Patients 1–7 were IDH2R140QAML; patients 8–10 were IDH1/2WT; patients 11–12 were IDH1R132K, and patients 13–14 were IDH2R140QMDS. N/A, not applicable.

Patient ID Sample type Age Gender Diagnosis Blast% Karyotype IDH mutation status IDH mutation VAF Co-occuring mutations

Y566 PB 61 M AML arising from chronic myelomonocytic leukemia PB:7, BM:35 47,XY,add(5)(q34),add(6)(q23),+8,−17, der(22)t(1;22)(q21;p11.2)[cp7]/46, XY[8] IDH2R140Q 42.3 NRAS, IDH2, JAK2, SRSF2
Y577 PB 26 F New diagnosis AML with no prior chemotherapy PB:68, BM:90 NK IDH2R140Q 38.4 NPM1
Y747 BM 62 M Relapsed refractory AML (progressed from MDS) PB: 28, BM:59 46,XY,der(15)t(1;15)(q12;p12)[7]/46,idem, del(20)(q11.2q13.3)[8] IDH2R140Q 47 SRSF2, TET2
Y1597 BM 77 M New diagnosis AML PB:15, BM:48 46,XY,del(8)(q23.2q24.13)[8]/46, XY[7] IDH2R140Q 50.1 SRSF2, JAK2, ASXL1
Y2260 PB 78 M Relapsed AML after azacitidine+enasidenib PB:13 46,XY,del(8)(q23.2q24.13)[8]/46, XY[7] IDH2R140Q NA SRSF2
Y1550 BM 75 M Relapsed AML PB:3, BM:23 NK IDH2R140Q NA SRSF2, RUNX1
Y1840 PB 75 M Relapsed AML after enasidenib PB:25 NK IDH2R140Q NA SRSF2, RUNX1
Y689 BM 63 M New diagnosis AML, progressed from MDS after failed azacitidine therapy PB:12, BM:13 NK WT SRSF2, ASXL1, CBL, NRAS, KRAS
Y406 PB 59 M New diagnosis AML PB:15, BM:40 46,XY,inv(3)(q21q26),del(5)(q14q34), del(13)(q13q21)[15] WT RUNX1
Y456 BM 46 F AML PB:5, BM:36 NK WT NPM1, WT1, FLT3ITD, FLT3D835
Y276 BM 39 F New diagnosis AML on Hydrea PB:92, BM:77 NK IDH1R132K NA NPM1, FLT3D835
Y493 PB 59 F Relapsed/Refractory AML from MDS- on clinical trial with IDH1 inhibitor with new anemia PB:6, BM:30 NK IDH1R132K NA
Y1952 BM 86 F MDS-EB1 BM:8 NK IDH2R140Q 47.3 SRSF2, ASXL1, STAG2
Y1365 BM 58 M MDS-EB2 PB:3, BM:9 NK IDH2R140Q 39 DNMT3A

N/A, not applicable

Fig 1. Olaparib targets IDH2-mutant AML blasts and leukemia initiating cells in vivo.

Fig 1.

A-E, Secondary MISTRG recipients engrafted with BM cells collected from primary MISTRG mice, transplanted with three different IDH2R140Q AML patient samples. Secondary mice were treated with either vehicle or olaparib (100mg/Kg) 6 days a week for 21 days. A-C, Pre- and post-treatment comparison of human CD45+ engraftment levels in BM of secondary treated mice. A, Y566 (vehicle, n=4, olaparib, n= 4); B, Y577 (vehicle n= 12, olaparib n= 8); C, Y747 (vehicle n= 4, olaparib n= 4). Individual mice are represented by matching pre-post symbols; P values were determined by ratio paired t test for pre-post treatment within groups and by Mann-Whitney test for comparison between treatment groups; n.s. not significant, *p < 0.05, ****p < 0.0001). D, Plasma concentrations of D-2-HG in pre-versus post administration of vehicle or olaparib. Individual mice are represented by symbols with mean ± S.E.M.; P values were determined by Mann–Whitney test; n.s. not significant, *p < 0.05, **p < 0.01. E, Immunohistochemistry (IHC) staining for hCD45 of BM of vehicle (top panel) and olaparib (lower panel) treated mice (scale bars 100 μm, original magnification 10x). F, Comparison of γH2AX+ huCD45 cells in bone marrow of secondary mice of Y577 treated with vehicle and olaparib (vehicle n= 8, olaparib n= 6). G-I, Secondary (2°) and tertiary (3°) transplantation of IDH2R140Q AML (Y577). G, Comparison of huCD45+linhuCD34+CD38 population in BM of vehicle (n=8) vs olaparib (n=6) treated mice. Quantification of huCD45+ engraftment levels (H) and huCD45+linhuCD34+CD38 population (I) in the BM of tertiary mice engrafted with equal numbers of huCD45+ BM cells (5×105) of vehicle (n=9) vs. olaparib (n=8) treated mice. J-L, Secondary and tertiary transplantation of IDH2R140Q AML (Y747). J, Comparison of huCD45+linhuCD34+CD38 population in the BM of vehicle (n=4) vs olaparib (n=4) treated mice. Quantification of huCD45+ engraftment levels (K) and huCD45+linhuCD34+CD38 population (L) in the BM of tertiary mice engrafted with equal numbers of huCD45+ BM cells (5×105) of vehicle (n=6) vs. olaparib (n=7) post-treatment mice. Individual mice are represented by symbols with means ± S.E.M.; symbols for corresponding 2° and 3° recipient mice are color coded; statistics represent Mann–Whitney test; n.s. not significant, *p < 0.05, **p < 0.01.

The IDHm inhibitors enasidenib and ivosidenib are known to induce differentiation of leukemic blasts, and we have previously been able to recapitulate the differentiation effect in IDH2 mutant MDS in MISTRG mice 24. Unlike IDHmi, olaparib exhibits direct toxicity on IDH mutant cells secondary to the 2HG-induced BRCAness and dependence on PARP-mediated DNA repair9, 38, 39. Consistent with this mechanism, olaparib did not alter lineage representation (Supplementary Fig. S3A) but significantly reduced the population of huCD45+LinCD34+CD38 cells, previously shown to be enriched for LICs 40, 41 (Fig. 1G and Supplementary Fig. S3B). To validate that functional LIC were targeted by PARPi, we transplanted equal numbers of huCD45+ BM cells from vehicle or olaparib treated mice into tertiary recipient mice (Supplementary Fig. S3C). 16 weeks after transplantation of olaparib versus vehicle treated BM, we quantitated engraftment levels in recipient mice as readout of olaparib toxicity against LIC in treated donor mice. Overall huCD45+ AML engraftment levels and LICs were significantly lower in the recipients of olaparib- compared to vehicle-treated BM cells (Fig. 1H and Supplementary Fig. S3D, E and Fig. 1I, respectively), suggesting that olaparib exerted direct toxicity on LICs. These findings held true in mice engrafted with a IDH2R140Q-mutant AML (Y747) relapsed after chemotherapy. Similarly, huCD45+LinCD34+CD38 LICs were significantly reduced in BM of olaparib compared to vehicle treated mice (Fig. 1J) again translating into reduced leukemia burden in tertiary recipients (Fig. 1K, L and Supplementary Fig. S3FH).

In summary, our data confirm the effectiveness of the PARPi olaparib in vivo specifically against IDH2R140Q, but not IDHWT AML, regardless of co-mutational pattern with toxicity against LICs.

Olaparib-induced toxicity is preserved in IDHmi-resistant IDH2-mutant AML

IDHmi rarely effect cure and ultimately patients relapse with restoration of 2HG levels suggesting an oncometabolite dependence of IDH mutant AML 1518. We therefore sought to test whether PARPi will be effective in enasidenib-resistant IDH2-mutant AML. We first resorted to a syngeneic mouse model of AML relying on co-mutation of Flt3 with an internal tandem duplication (ITD) (Flt3ITD) and of IDH2R140Q. Polyinosinic: polycytidylic acid [poly (I:C)] induced activation of Mx1 promoter-driven Cre recombinase (M) results in heterozygous expression of IDH2R140Q (I2) in the background of constitutive Flt3ITD (F) expression and mice develop a transplantable (MFI2) leukemia as previously described 25. To test olaparib in vitro in colony forming assays we established that olaparib at 1.25μM was non-toxic to IDH wildtype cells while effectively reducing colony formation in IDH2R140Q mutant cells (Supplementary Fig. S4A). We generated enasidenib-resistant murine AML by transduction of MFI2 AML with lentiviral vectors expressing IDH2 carrying second site mutations Q316E or I319M, previously reported to confer enasidenib resistance in patients 18. Lentivirally transduced MFI2 AML expressing IDH2I319M, IDH2Q316E or IDH2WT were then subjected to serial colony formation in vitro in the presence of vehicle, enasidenib (50nM), or olaparib (1.25μM). Enasidenib inhibited serial replating capacity of cells expressing IDH2R140/WT at the third replating (CFU3) as previously shown, while the Q316E and I319M mutations conferred resistance to enasidenib but not to olaparib (Fig. 2A). These data suggest that restoration of 2HG may be sufficient to confer sensitivity to PARPi. While second site mutations have been identified only in a minority of patients 18, restoration of aberrant 2HG production is found in a significant subset of IDHmi resistant patients, suggesting that PARP inhibition may be more widely applicable to mutant IDH inhibitor-resistant AML 14, 16.

Fig 2. Enasidenib resistant AML is sensitive to olaparib administration both in vitro and in vivo.

Fig 2.

A, Colony forming unit assay (CFU) re-plating of hematopoietic stem and progenitor cells (HSPCs) from FLT3ITDIDH2R140Q linage depleted primary murine cells expressing IDH2 WT or Q316E or I319M in trans and cultured in methocult containing vehicle, AG-221 at 50nM, or olaparib at 1.25μM. Data are mean ± SEM. for triplicate (CFU1/2) culture. P values were determined by Mann-Whitney; *p < 0.05, **p < 0.01, ***p<0.001. This is a representation of 2 independent experiments. B-C, Clinical and laboratory features for a AML patient at the time of IDH2R140Q AML diagnosis (Y1597) (B) and after relapse on azacytidine + enasidenib treatment (Y2260) (C), including blood absolute neutrophil count (ANC) and white blood cell count (WBC), and blast percentage in PB and BM. D, Human CD45+ engraftment levels in the BM of secondary mice of (Y1597) treated with vehicle (n=4), enasidenib (n=4), or olaparib (n=5); enasidenib (40mg/kg) was administered via oral gavage and olaparib (100mg/kg) intraperitoneally for 6 days per week for 3 weeks. E, Human CD45+ engraftment levels in the BM of secondary mice of (Y2260) treated with vehicle (n=5), enasidenib (n=5), or olaparib (n=5). Individual mice are represented by matching pre-post symbols; P values were determined by ratio paired t test for pre-post treatment within groups and by Mann-Whitney test for comparison between different treatment groups; n.s. not significant, *p < 0.05, ***p<0.001. F-G, Comparison of huCD45+linhuCD34+CD38 population in BM of treated mice of Y1597 (F) and of Y2260 (G).

We therefore investigated the olaparib response in enasidenib-resistant IDH2R140Q AML from 2 patients treated at our institution. The first patient presented with newly diagnosed, intermediate-risk AML with inversion 8 (inv (8)), with an elevated WBC of 42×103/μl with 48% blasts in BM and 10% blasts in PB (Fig. 2B, C). Targeted next generation sequencing (NGS) of bone marrow cells at time of diagnosis revealed an IDH2R140Q mutation along with mutations in SRSF2, JAK2, and ASXL1 (Table 1). The patient received azacitidine and enasidenib, which induced a significant reduction in WBC count and blast percentage (Fig. 2B, C). After almost 12 months of continuous treatment with enasidenib, the patient relapsed with rapid rise in the WBC with leukemic blast predominance (Fig. 2B, C), which preceded the patient’s death. Resequencing of the mutant AML did not identify the mechanism of acquired resistance neither in the clinical targeted panel (Table 1) nor using a comprehensive AML targeted panel (Supplementary Table S1). We transplanted MISTRG mice with BM and PB mononuclear cells banked at the time of diagnosis (Y1597) and at time of relapse on enasidenib (Y2260) (Table 1 and Fig. 2B, C). Both diagnosis and relapsed AML engrafted MISTRG mice had elevated 2HG levels (Supplementary Fig. S4B, C). Secondary recipient mice were treated with vehicle, olaparib or enasidenib (40mg/kg via oral gavage) for 6 days per week for 3 weeks. While both enasidenib and olaparib effectively reduced engraftment and plasma 2HG levels in mice engrafted with the new AML diagnosis sample (Y1597), enasidenib failed to limit proliferation of the relapsed AML (Y2260) with significant rise in disease burden during the treatment course (Fig. 2D, E and Supplementary Fig. S4BH). Olaparib on the other hand significantly inhibited leukemic progression compared to enasidenib (Fig. 2D, E and Supplementary Fig. S4CH). Moreover, olaparib significantly reduced LinCD45+CD34+CD38 LICs in both diagnosis (enasidenib-naïve) and enasidenib-resistant AML compared to vehicle and enasidenib, confirming effectiveness of olaparib against LICs (Fig. 2F, G).

Persistent sensitivity to olaparib despite acquisition of resistance to IDHmi was also evident in a second patient with relapsed AML (Fig. 3A, B) with IDH2R140Q and RUNX1 mutations (Table 1). While the patient’s AML was sensitive to both enasidenib and olaparib prior to treatment with enasidenib (Y1550, Fig. 3C), after progression of disease on enasidenib, olaparib but not enasidenib effectively limited leukemic cell expansion in MISTRG (Y1840, Fig. 3D). Both enasidenib and olaparib significantly inhibited LinCD45+CD34+CD38 LICs in the enasidenib-naïve AML engrafted mice (Y1550, Fig. 3E) while only olaparib significantly reduced LICs in the enasidenib-resistant AML engrafted mice (Y1840, Fig. 3F).

Fig 3. Olaparib remains effective in a second enasidenib-resistant IDH2-mutant AML PDX.

Fig 3.

A-B, Clinical and laboratory features for a relapsed IDH2R140Q AML patient prior to treatment with enasidenib (Y1550) (A) and after relapse on enasidenib treatment (Y1840) (B), including blood absolute neutrophil count (ANC) and white blood cell count (WBC), and blast percentage in PB and BM. C, Human CD45+ engraftment levels in the PB and BM of secondary mice of (Y1550) treated with vehicle (n=3), enasidenib (n=4), or olaparib (n=3). D, Human CD45+ engraftment levels in the PB and BM of secondary mice of (Y1840) treated with vehicle (n=3), enasidenib (n=3), or olaparib (n=3). E-F, Comparison of huCD45+linhuCD34+CD38 population in BM of treated mice of Y1550 (E) and of Y1840 (F). Individual mice are represented by matching pre-post symbols; P values were determined by ratio paired t test for pre-post treatment within groups and by Mann-Whitney test for comparison between different treatment groups; n.s. not significant, *p < 0.05, ***p<0.001.

Collectively, these data show that PARP inhibition is effective in IDHmi resistant AML and may offer a novel treatment approach for patients with relapsed AML.

PARP sensitivity is IDH isoform- and disease-agnostic

AML and MDS carry mutations at two hotspot sites in IDH2 and in IDH1and different mutations may have in part distinct oncogenic mechanisms 42. We therefore tested whether susceptibility to PARP inhibition was indeed independent of the mutant IDH isoform and whether PARPi could be effective in patients with MDS. We transplanted MISTRG mice with one newly diagnosed (Y276) and one relapsed/refractory secondary (Y493) IDH1R132K -mutant AML sample (Table 1 and Supplementary Fig. S5A). Olaparib effectively treated both IDH1R132K -mutant AMLs compared to vehicle (Fig. 4A, B) and despite prior exposure of Y493 to an IDH1 inhibitor (Fig. 4C, D). Olaparib administration reduced the huCD45+linCD34+CD38 LIC population compared to vehicle treatment in both AMLs (Supplementary Fig. S5B, C) as shown for IDH2 mutant AML. Overall, these results demonstrated that olaparib monotherapy was also active against IDH1-mutant AML and thus isoform- agnostic.

Fig 4. PARP inhibition is also effective in IDH1-mutant AML and IDH1/2-mutant MDS.

Fig 4.

A-B, Assessment of human CD45+ chimerism levels in vehicle vs olaparib treated secondary MISTRG mice engrafted with BM cells collected from primary mice transplanted with IDH1R132K AML. A, Y276 (vehicle, n=4, olaparib, n= 4); B, Y493 (vehicle n= 2, olaparib n= 3). Primary patient BM CD34+ HSPCs were pre-incubated with anti-CD3 antibody (OKT3) and injected intra-hepatically into newborn MISTRG mice conditioned twice with 150cGy. C-D, Clinical and laboratory data for a relapsed/refractory IDH1R132K AML (Y493) while on ivosidenib trial treatment, including ANC, WBC, (C) and blast percentage (D) in PB. E-F, Chimerism detection of huCD45+ percentages in BM preceded treating the mice with either vehicle, enasidenib, or olaparib. Mice were analyzed 21 days after drug administration. E, Comparison of overall human CD45+ engraftment in BM of mice engrafted with IDH2R140Q MDS-EB1 (Y1952) treated with vehicle (n=3), enasidenib (n=3), or olaparib (n=4). F, Comparison of overall human CD45+ engraftment in BM of mice engrafted with IDH2R140Q MDS-EB2 (Y1365) treated with vehicle (n=4), enasidenib (n=5), or olaparib (n=6). Individual mice are represented by matching pre-post symbols; P values were determined by ratio paired t test for pre-post treatment within groups and by Mann-Whitney test for comparison between treatment groups; n.s. not significant, *p < 0.05, **p<0.01.

Lastly, to determine whether PARP inhibition is a relevant therapy for IDH-mutant MDS, we transplanted MISTRG mice with CD34+ cells from two patients with IDH2 mutant MDS, namely IDH2R140Q-mutant MDS-excess blast (EB) 1 (Y1952) and EB2 (Y1365) (Table 1). Primary engrafted mice were assigned to treatment groups based on pre-treatment marrow engraftment and treated with vehicle, enasidenib or olaparib for 21 days. Enasidenib and Olaparib showed efficacy in both MDS samples with reduction in huCD45+ engraftment in both BM and PB (Fig. 4EF and Supplementary Fig. S5DE) and with significant reduction in the huCD45+LinCD38CD34+ LIC populations (Supplementary Fig. S5FG).

Collectively, our data showed the effectiveness of the synthetic lethality-mediated approach as a novel alternative therapeutic strategy in IDH2-mutant MDS.

Discussion

There is significant need for improvement for patients with relapsed/ refractory AML and MDS despite the availability of IDH inhibitors, which have modest and transient effectiveness. Both primary and acquired resistance mechanisms to IDH inhibitors have been identified 1518. Primary resistance has been highly associated with the presence of NRAS, KRAS and MAPK pathway mutations, and increased incidence of relapse has been linked to higher mutational burden 12, 14, 43. In addition, adaptive resistance develops through the acquisition of second site mutations or via isoform switching or yet unknown mechanisms, but almost invariably aberrant 2HG levels are restored 18. Post-resistance treatments are thus critical to prolong patients’ lives.

Recent advances in the understanding of metabolic vulnerabilities induced by IDH mutations have led to the identification of an alternative strategy to leverage the concept of synthetic lethality associated with the impaired HR conferred by 2HG 44. We here validated the efficacy of PARPi exploiting the 2HG-induced BRCAness defect in primary AML and MDS samples in vivo building upon our and others’ prior in vitro studies and our previously reported highly efficient MDS/AML PDX model 19, 22, 24.

Cytokine humanized MISTRG mice express physiological levels of human cytokines instead of their murine counterparts and have overcome many challenges in modeling myeloid malignancies 36, 45. Accordingly, our group previously reported MISTRG as an advanced and highly efficient model of MDS that faithfully replicates the intra- and inter-disease heterogeneity, the dysplastic morphology, and the mutational profile and clonal complexity 24. We leveraged the MISTRG model to test whether PARP inhibition would be effective against a variety of IDH mutant AML, AML resistant to IDHmi, and against IDH mutant MDS. Olaparib treatment demonstrated antileukemic efficacy irrespective of co-occuring mutations as long as cells carried IDH1/2 mutations, while olaparib was ineffective against IDH1/2 wildtype AML. In addition, olaparib demonstrated toxicity against phenotypic and functional LICs, a rare subpopulation of leukemic cells that possess stem cell properties, including leukemia initiation and self-renewal, as evident by engraftment in immunodeficient mice as shown here, and drug resistance 46, 47.

Our data also demonstrates that PARP inhibition could represent a new choice for patients with primary refractoriness to IDH inhibitors or for those who relapse on IDH1/2 inhibitor treatment irrespective of tumor type 19. The PRIME clinical trial (NCI10264) is currently testing the overall response of IDH1/2-mutant relapsed/refractory AML and MDS to PARP inhibitor monotherapy, which further underlines the clinical relevance of the results presented here. Future studies combining PARP inhibitors with other DNA damaging agents or agents with complementary activity will be important to leverage this mechanism against MDS/AML as monotherapy rarely results in cure.

Collectively, our data provide critical insights which support the pursuit of PARP inhibition as a promising therapeutic tool for IDH mutated MDS/AML patients.

Supplementary Material

Supplemental material

Acknowledgements

We thank Yale Pathology Tissue Services especially A. Brooks for research histology services. We thank Yale Animal Resources Center, especially P. Ranney, J. Fonck for animal care. We thank Yale Flow Cytometry Core, especially Lesley Divine, Diane Trotta and Chao Wang for flow cytometry analysis. We thank our patients for donating blood and bone marrow to research. This study was supported in part by the NIH/NIDDK R01DK102792 (to S.H.), The Frederick A. Deluca Foundation and the Vera and Joseph Dresner Foundation (to S.H.), the Howard Hughes Medical Institute (to R.A.F), General Program of the National Natural Science Foundation of China (Grant No.82170137, to Y.S.), and the Leukemia and Lymphoma Society and the NIH/NCI R01CA266604 (to S.H. and R.S.B.). Y.S. was supported by the General Program of the National Natural Science Foundation of China (Grant No.82170137). X.F. was supported by the Young Scientists Fund of the National Natural Science Foundation of China (Grant No. 81801588). Y.G. was supported by the American Society of Hematology Scholar Award. This study was supported by the Animal Modeling Core, Pilot and Feasibility Program of the Yale Cooperative Center of Excellence in Hematology (NIDDK U54DK106857).

Disclosure of conflicts of interest

Zeidan: A.M.Z. received research funding from Celgene/BMS, Abbvie, Astex, Pfizer, Medimmune/AstraZeneca, Boehringer-Ingelheim, Trovagene/Cardiff Oncology, Incyte, Takeda, Novartis, Aprea, Amgen, and ADC Therapeutics. A.M.Z participated in advisory boards, had a consultancy with, and/or received honoraria from AbbVie, Otsuka, Pfizer, Celgene/BMS, Jazz, Incyte, Agios, Boehringer-Ingelheim, Novartis, Acceleron, Astellas, Daiichi Sankyo, Cardinal Health, Taiho, Seattle Genetics, BeyondSpring, Trovagene/Cardiff Oncology, Takeda, Ionis, Amgen, Tyme, Aprea, Kura, Janssen, Gilead, and Epizyme. A.M.Z received travel support for meetings from Pfizer, Novartis, and Trovagene.

Flavell: Zai labs: Consultancy; GSK: Consultancy.

Prebet: Boehringer Ingelheim: Research Funding; Novartis: Honoraria; Pfizer: Honoraria; Agios: Consultancy, Research Funding; Jazz Pharmaceuticals: Consultancy, Honoraria, Research Funding; Genentech: Consultancy; Tetraphase: Consultancy; Bristol-Myers Squibb: Honoraria, Research Funding.

Bindra: Cybrexa Therapeutics: Consultancy, Equity Ownership. Athena Therapeutics: Equity Ownership.

Halene: Forma Therapeutics: Consultancy

References

  • 1.Showalter MR, Hatakeyama J, Cajka T, VanderVorst K, Carraway KL, Fiehn O, et al. Replication Study: The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Elife 2017. Jun 27; 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Winer ES, Stone RM. Novel therapy in Acute myeloid leukemia (AML): moving toward targeted approaches. Ther Adv Hematol 2019; 10: 2040620719860645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Clark O, Yen K, Mellinghoff IK. Molecular Pathways: Isocitrate Dehydrogenase Mutations in Cancer. Clin Cancer Res 2016. Apr 15; 22(8): 1837–1842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.DiNardo CD, Jabbour E, Ravandi F, Takahashi K, Daver N, Routbort M, et al. IDH1 and IDH2 mutations in myelodysplastic syndromes and role in disease progression. Leukemia 2016. Apr; 30(4): 980–984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Losman JA, Kaelin WG Jr., What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer. Genes Dev 2013. Apr 15; 27(8): 836–852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Rakheja D, Medeiros LJ, Bevan S, Chen W. The emerging role of d-2-hydroxyglutarate as an oncometabolite in hematolymphoid and central nervous system neoplasms. Front Oncol 2013; 3: 169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer Cell 2011. Jan 18; 19(1): 17–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Molenaar RJ, Radivoyevitch T, Maciejewski JP, van Noorden CJ, Bleeker FE. The driver and passenger effects of isocitrate dehydrogenase 1 and 2 mutations in oncogenesis and survival prolongation. Biochim Biophys Acta 2014. Dec; 1846(2): 326–341. [DOI] [PubMed] [Google Scholar]
  • 9.Wang Y, Wild AT, Turcan S, Wu WH, Sigel C, Klimstra DS, et al. Targeting therapeutic vulnerabilities with PARP inhibition and radiation in IDH-mutant gliomas and cholangiocarcinomas. Sci Adv 2020. Apr; 6(17): eaaz3221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Norsworthy KJ, Luo L, Hsu V, Gudi R, Dorff SE, Przepiorka D, et al. FDA Approval Summary: Ivosidenib for Relapsed or Refractory Acute Myeloid Leukemia with an Isocitrate Dehydrogenase-1 Mutation. Clin Cancer Res 2019. Jun 1; 25(11): 3205–3209. [DOI] [PubMed] [Google Scholar]
  • 11.Kim ES. Enasidenib: First Global Approval. Drugs 2017. Oct; 77(15): 1705–1711. [DOI] [PubMed] [Google Scholar]
  • 12.Liu X, Gong Y. Isocitrate dehydrogenase inhibitors in acute myeloid leukemia. Biomark Res 2019; 7: 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Upadhyay VA, Brunner AM, Fathi AT. Isocitrate dehydrogenase (IDH) inhibition as treatment of myeloid malignancies: Progress and future directions. Pharmacol Ther 2017. Sep; 177: 123–128. [DOI] [PubMed] [Google Scholar]
  • 14.Amatangelo MD, Quek L, Shih A, Stein EM, Roshal M, David MD, et al. Enasidenib induces acute myeloid leukemia cell differentiation to promote clinical response. Blood 2017; 130(6): 732–741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Choe S, Wang H, DiNardo CD, Stein EM, de Botton S, Roboz GJ, et al. Molecular mechanisms mediating relapse following ivosidenib monotherapy in IDH1-mutant relapsed or refractory AML. Blood Adv 2020. May 12; 4(9): 1894–1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Quek L, David MD, Kennedy A, Metzner M, Amatangelo M, Shih A, et al. Clonal heterogeneity of acute myeloid leukemia treated with the IDH2 inhibitor enasidenib. Nat Med 2018. Aug; 24(8): 1167–1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Harding JJ, Lowery MA, Shih AH, Schvartzman JM, Hou S, Famulare C, et al. Isoform Switching as a Mechanism of Acquired Resistance to Mutant Isocitrate Dehydrogenase Inhibition. Cancer Discov 2018. Dec; 8(12): 1540–1547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Intlekofer AM, Shih AH, Wang B, Nazir A, Rustenburg AS, Albanese SK, et al. Acquired resistance to IDH inhibition through trans or cis dimer-interface mutations. Nature 2018. Jul; 559(7712): 125–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Sulkowski PL, Corso CD, Robinson ND, Scanlon SE, Purshouse KR, Bai H, et al. 2-Hydroxyglutarate produced by neomorphic IDH mutations suppresses homologous recombination and induces PARP inhibitor sensitivity. Sci Transl Med 2017. Feb 1; 9(375). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sulkowski PL, Oeck S, Dow J, Economos NG, Mirfakhraie L, Liu Y, et al. Oncometabolites suppress DNA repair by disrupting local chromatin signalling. Nature 2020. Jun; 582(7813): 586–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Sulkowski PL, Sundaram RK, Oeck S, Corso CD, Liu Y, Noorbakhsh S, et al. Krebs-cycle-deficient hereditary cancer syndromes are defined by defects in homologous-recombination DNA repair. Nat Genet 2018. Aug; 50(8): 1086–1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Molenaar RJ, Radivoyevitch T, Nagata Y, Khurshed M, Przychodzen B, Makishima H, et al. IDH1/2 Mutations Sensitize Acute Myeloid Leukemia to PARP Inhibition and This Is Reversed by IDH1/2-Mutant Inhibitors. Clin Cancer Res 2018. Apr 1; 24(7): 1705–1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Philip B, Yu DX, Silvis MR, Shin CH, Robinson JP, Robinson GL, et al. Mutant IDH1 Promotes Glioma Formation In Vivo. Cell Rep 2018. May 1; 23(5): 1553–1564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Song Y, Rongvaux A, Taylor A, Jiang T, Tebaldi T, Balasubramanian K, et al. A highly efficient and faithful MDS patient-derived xenotransplantation model for pre-clinical studies. Nat Commun 2019. Jan 21; 10(1): 366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Shih AH, Meydan C, Shank K, Garrett-Bakelman FE, Ward PS, Intlekofer AM, et al. Combination Targeted Therapy to Disrupt Aberrant Oncogenic Signaling and Reverse Epigenetic Dysfunction in IDH2- and TET2-Mutant Acute Myeloid Leukemia. Cancer Discov 2017. May; 7(5): 494–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Rongvaux A, Willinger T, Martinek J, Strowig T, Gearty SV, Teichmann LL, et al. Development and function of human innate immune cells in a humanized mouse model. Nat Biotechnol 2014. Apr; 32(4): 364–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wingett SW, Andrews S. FastQ Screen: A tool for multi-genome mapping and quality control. F1000Res 2018; 7: 1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Li H, Durbin R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics 2010. Mar 1; 26(5): 589–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 2009. Aug 15; 25(16): 2078–2079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 2010. Mar 15; 26(6): 841–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Benjamin D, Sato T, Cibulskis K, Getz G, Stewart C, Lichtenstein L. Calling Somatic SNVs and Indels with Mutect2. bioRxiv 2019: 861054. [Google Scholar]
  • 32.Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res 2010. Sep; 38(16): e164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Shihab HA, Gough J, Cooper DN, Stenson PD, Barker GL, Edwards KJ, et al. Predicting the functional, molecular, and phenotypic consequences of amino acid substitutions using hidden Markov models. Hum Mutat 2013. Jan; 34(1): 57–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N, et al. COSMIC: the Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res 2019. Jan 8; 47(D1): D941–D947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Robinson JT, Thorvaldsdottir H, Wenger AM, Zehir A, Mesirov JP. Variant Review with the Integrative Genomics Viewer. Cancer Res 2017. Nov 1; 77(21): e31–e34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Saito Y, Ellegast JM, Rafiei A, Song Y, Kull D, Heikenwalder M, et al. Peripheral blood CD34(+) cells efficiently engraft human cytokine knock-in mice. Blood 2016. Oct 6; 128(14): 1829–1833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ellegast JM, Rauch PJ, Kovtonyuk LV, Muller R, Wagner U, Saito Y, et al. inv(16) and NPM1mut AMLs engraft human cytokine knock-in mice. Blood 2016. Oct 27; 128(17): 2130–2134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lu Y, Kwintkiewicz J, Liu Y, Tech K, Frady LN, Su YT, et al. Chemosensitivity of IDH1-Mutated Gliomas Due to an Impairment in PARP1-Mediated DNA Repair. Cancer Res 2017. Apr 1; 77(7): 1709–1718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Tateishi K, Higuchi F, Miller JJ, Koerner MVA, Lelic N, Shankar GM, et al. The Alkylating Chemotherapeutic Temozolomide Induces Metabolic Stress in IDH1-Mutant Cancers and Potentiates NAD(+) Depletion-Mediated Cytotoxicity. Cancer Res 2017. Aug 1; 77(15): 4102–4115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994. Feb 17; 367(6464): 645–648. [DOI] [PubMed] [Google Scholar]
  • 41.Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 1997. Jul; 3(7): 730–737. [DOI] [PubMed] [Google Scholar]
  • 42.Wang HY, Tang K, Liang TY, Zhang WZ, Li JY, Wang W, et al. The comparison of clinical and biological characteristics between IDH1 and IDH2 mutations in gliomas. J Exp Clin Cancer Res 2016. May 31; 35: 86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Stein EM, DiNardo CD, Fathi AT, Pollyea DA, Stone RM, Altman JK, et al. Molecular remission and response patterns in patients with mutant-IDH2 acute myeloid leukemia treated with enasidenib. Blood 2019. Feb 14; 133(7): 676–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Madala HR, Punganuru SR, Arutla V, Misra S, Thomas TJ, Srivenugopal KS. Beyond Brooding on Oncometabolic Havoc in IDH-Mutant Gliomas and AML: Current and Future Therapeutic Strategies. Cancers (Basel) 2018. Feb 11; 10(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Rongvaux A, Takizawa H, Strowig T, Willinger T, Eynon EE, Flavell RA, et al. Human hemato-lymphoid system mice: current use and future potential for medicine. Annu Rev Immunol 2013; 31: 635–674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Dick JE. Stem cell concepts renew cancer research. Blood 2008. Dec 15; 112(13): 4793–4807. [DOI] [PubMed] [Google Scholar]
  • 47.Saito Y, Uchida N, Tanaka S, Suzuki N, Tomizawa-Murasawa M, Sone A, et al. Induction of cell cycle entry eliminates human leukemia stem cells in a mouse model of AML. Nat Biotechnol 2010. Mar; 28(3): 275–280. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental material

Data Availability Statement

All relevant data are available from the authors upon reasonable request.

RESOURCES