Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 May;36(5):1313-1323.
doi: 10.1038/s41375-022-01536-x. Epub 2022 Mar 10.

In vivo anti-tumor effect of PARP inhibition in IDH1/2 mutant MDS/AML resistant to targeted inhibitors of mutant IDH1/2

Affiliations

In vivo anti-tumor effect of PARP inhibition in IDH1/2 mutant MDS/AML resistant to targeted inhibitors of mutant IDH1/2

Rana Gbyli et al. Leukemia. 2022 May.

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.

PubMed Disclaimer

Figures

Fig 1.
Fig 1.. Olaparib targets IDH2-mutant AML blasts and leukemia initiating cells in vivo.
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.
Fig 2.
Fig 2.. Enasidenib resistant AML is sensitive to olaparib administration both in vitro and in vivo.
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).
Fig 3.
Fig 3.. Olaparib remains effective in a second enasidenib-resistant IDH2-mutant AML PDX.
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.
Fig 4.
Fig 4.. PARP inhibition is also effective in IDH1-mutant AML and IDH1/2-mutant MDS.
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.

Similar articles

Cited by

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. - PMC - PubMed
    1. Winer ES, Stone RM. Novel therapy in Acute myeloid leukemia (AML): moving toward targeted approaches. Ther Adv Hematol 2019; 10: 2040620719860645. - PMC - PubMed
    1. Clark O, Yen K, Mellinghoff IK. Molecular Pathways: Isocitrate Dehydrogenase Mutations in Cancer. Clin Cancer Res 2016. Apr 15; 22(8): 1837–1842. - PMC - PubMed
    1. 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. - PMC - PubMed
    1. 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. - PMC - PubMed

Publication types

MeSH terms

Substances