scholarly journals DNA methyltransferase inhibitors induce a BRCAness phenotype that sensitizes NSCLC to PARP inhibitor and ionizing radiation

2019 ◽  
Vol 116 (45) ◽  
pp. 22609-22618 ◽  
Author(s):  
Rachel Abbotts ◽  
Michael J. Topper ◽  
Christopher Biondi ◽  
Daniel Fontaine ◽  
Reena Goswami ◽  
...  

A minority of cancers have breast cancer gene (BRCA) mutations that confer sensitivity to poly (ADP-ribose) polymerase (PARP) inhibitors (PARPis), but the role for PARPis in BRCA-proficient cancers is not well established. This suggests the need for novel combination therapies to expand the use of these drugs. Recent reports that low doses of DNA methyltransferase inhibitors (DNMTis) plus PARPis enhance PARPi efficacy in BRCA-proficient AML subtypes, breast, and ovarian cancer open up the possibility that this strategy may apply to other sporadic cancers. We identify a key mechanistic aspect of this combination therapy in nonsmall cell lung cancer (NSCLC): that the DNMTi component creates a BRCAness phenotype through downregulating expression of key homologous recombination and nonhomologous end-joining (NHEJ) genes. Importantly, from a translational perspective, the above changes in DNA repair processes allow our combinatorial PARPi and DNMTi therapy to robustly sensitize NSCLC cells to ionizing radiation in vitro and in vivo. Our combinatorial approach introduces a biomarker strategy and a potential therapy paradigm for treating BRCA-proficient cancers like NSCLC.

2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi71-vi72 ◽  
Author(s):  
Shaofang Wu ◽  
Feng Gao ◽  
Dimpy Koul ◽  
Alfred Yung

Abstract Temozolomide (TMZ) is the standard chemotherapy for malignant gliomas (MG), and resistance to this drug is mediated by the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT). Epigenetic silencing of the MGMT gene by promoter methylation in about 40% patients is associated with loss of MGMT expression that compromises DNA repair leading to favorable response to TMZ therapy. Understanding the mechanism of MGMT mediated repair and modulating MGMT activity will enhance TMZ activity in MGMT unmethylated MG. Here we report a novel mode of regulation of MGMT protein activity by Poly-ADP-ribose polymerase (PARP). We found that PAPR physically interacts with MGMT and PARylates MGMT in response to TMZ treatment. We further showed PARylation of MGMT by PAPR is required for MGMT binding to DNA and to remove O6-methylguanine adducts in damaged DNA induced by TMZ. All 4 PARP inhibitors (trapping and non-trapping) tested (Talazoparib, Pamiparib, Veliparib, Olaparib) can inhibit PARP-MGMT binding, PARylation of MGMT, binding to DNA and subsequent removal of O6 - lesions in damaged DNA. We showed combination of PARP inhibitor with TMZ potentiated TMZ cytotoxicity in both MGMT methylated and unmethylated Glioma stem cell lines, but more profoundly in unmethylated group in vitro and in vivo. PARP inhibition acted as a double-edged sword in MGMT unmethylated MG: blocking BER/SSBR pathway to repair TMZ induced N7-MetG and O3-MetA, and more importantly, suppressing PARP-mediated PARylation of MGMT and thus silencing MGMT activity to repair O6-MetG, resulting in augmented cytotoxicity. This is the first study to show that PARylation of MGMT by PARP is required for repairing TMZ-induced O6-methylguanine adducts, and inhibition of MGMT PARylation abolishes MGMT function and renders sensitization to TMZ treatment. This finding provides a rationale for combining TMZ/CCNU and PARP inhibitors in MGMT unmethylated MG patients to enhance the benefit of adjuvant chemotherapy.


NAR Cancer ◽  
2021 ◽  
Vol 3 (2) ◽  
Author(s):  
Amrita Sule ◽  
Jinny Van Doorn ◽  
Ranjini K Sundaram ◽  
Sachita Ganesa ◽  
Juan C Vasquez ◽  
...  

Abstract Mutations in the isocitrate dehydrogenase-1 and -2 (IDH1/2) genes were first identified in glioma and acute myeloid leukemia (AML), and subsequently found in multiple other tumor types. These neomorphic mutations convert the normal product of enzyme, α-ketoglutarate (αKG), to the oncometabolite 2-hydroxyglutarate (2HG). Our group recently demonstrated that 2HG suppresses the high-fidelity homologous recombination (HR) DNA repair pathway, resulting in a state referred to as ‘BRCAness’, which confers exquisite sensitivity to poly(ADP-ribose) polymerase (PARP) inhibitors. In this study, we sought to elucidate sensitivity of IDH1/2-mutant cells to DNA damage response (DDR) inhibitors and, whether combination therapies could enhance described synthetic lethal interactions. Here, we report that ATR (ataxia telangiectasia and Rad3-related protein kinase) inhibitors are active against IDH1/2-mutant cells, and that this activity is further potentiated in combination with PARP inhibitors. We demonstrate this interaction across multiple cell line models with engineered and endogenous IDH1/2 mutations, with robust anti-tumor activity in vitro and in vivo. Mechanistically, we found ATR and PARP inhibitor treatment induces premature mitotic entry, which is significantly elevated in the setting of IDH1/2-mutations. These data highlight the potential efficacy of targeting HR defects in IDH1/2-mutant cancers and support the development of this combination in future clinical trials.


2020 ◽  
Vol 22 (5) ◽  
pp. 1226-1234
Author(s):  
Florian Guibbal ◽  
Samantha L. Hopkins ◽  
Anna Pacelli ◽  
Patrick G. Isenegger ◽  
Michael Mosley ◽  
...  

Abstract Background Poly (ADP-ribose) polymerase (PARP) inhibitors are extensively studied and used as anti-cancer drugs, as single agents or in combination with other therapies. Most radiotracers developed to date have been chosen on the basis of strong PARP1–3 affinity. Herein, we propose to study AZD2461, a PARP inhibitor with lower affinity towards PARP3, and to investigate its potential for PARP targeting in vivo. Methods Using the Cu-mediated 18F-fluorodeboronation of a carefully designed radiolabelling precursor, we accessed the 18F-labelled isotopologue of the PARP inhibitor AZD2461. Cell uptake of [18F]AZD2461 in vitro was assessed in a range of pancreatic cell lines (PSN-1, PANC-1, CFPAC-1 and AsPC-1) to assess PARP expression and in vivo in xenograft-bearing mice. Blocking experiments were performed with both olaparib and AZD2461. Results [18F]AZD2461 was efficiently radiolabelled via both manual and automated procedures (9 % ± 3 % and 3 % ± 1 % activity yields non-decay corrected). [18F]AZD2461 was taken up in vivo in PARP1-expressing tumours, and the highest uptake was observed for PSN-1 cells (7.34 ± 1.16 %ID/g). In vitro blocking experiments showed a lesser ability of olaparib to reduce [18F]AZD2461 binding, indicating a difference in selectivity between olaparib and AZD2461. Conclusion Taken together, we show the importance of screening the PARP selectivity profile of radiolabelled PARP inhibitors for use as PET imaging agents.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1371-1371
Author(s):  
Scott M. Portwood ◽  
Marianna C Cantella ◽  
Tara L. Cronin ◽  
Eunice S. Wang

Background CD33 (Siglec3) is a cell surface transmembrane receptor that is rapidly internalized and highly expressed on AML blasts but is absent on normal hematopoietic stem cells. Gemtuzumab ozogamicin (GO), a humanized anti-CD33 antibody conjugated to a DNA strand scission inducing agent (calicheamicin) was recently FDA approved for the treatment of newly-diagnosed or relapsed/refractory CD33-positive acute myeloid leukemia (AML). GO has been shown to exert clinical activity in leukemia patients. Poly (ADP-ribose) polymerase (PARP) inhibitors prevent the repair of single stranded DNA breaks by blocking the nicotinamide adenine dinucleotide (NAD) catalytic domain of the PARP protein and by preventing the dissociation of PARP from the DNA (PARP trapping). Talazoparib is unique among clinical PARP inhibitors in displaying 10,000-fold increased PARP trapping as compared to other agents . We hypothesized that combination therapy using GO and Talazoparib would result in synergistic anti-leukemic effects on human CD33+ AML cells due to the ability of the PARP inhibitor to enhance levels of DNA damage induced by GO therapy. Materials and Methods Human AML cell lines were characterized for CD33 expression using flow cytometry after staining with antibody-linked fluorescent QuantiBrite Beads. Cells were continuously exposed to varying doses of GO (10pM - 100mM) and PARP inhibitors (1nM - 100mM) for 96h alone and in combination. Cell viability was measured immediately following treatment using a WST colorimetric assay. Treatment-induced apoptosis (annexin/PI) and DNA damage (H2AX) were quantified by flow cytometric assays. Synergy reports were generated using Compusyn software. In vivo efficacy was assessed in NSG mice systematically engrafted with luciferase labeled human CD33+ AML cells following tail vein injection. Animals were treated with varying doses of vehicle, GO (1 and 50ug/kg 1x/week for 3 weeks), or talazoparib (0.1 and 0.33mg/kg 5 days/week) either alone or in combination. Treatment effects on leukemia burden, toxicity, and survival were determined by weekly whole animal bioluminescent imaging, total animal weights, and time to morbidity. Results Human AML cell lines (HEL, HL60) express high expression levels of CD33 molecules/cell (43,645 and 31,286 respectively) relative to negative controls. Continuous exposure to single agent GO and Talazoparib for 96h resulted in a dose dependent inhibition of human AML cell growth (HEL, HL60) . IC values for GO were 0.01 - 6.6μg/ml and for Talazoparib were 0.8-0.95μM. Combination in vitro therapy with GO (0.005 - 1μg/ml) and Talazoparib (fixed dose 100nM) resulted in synergistic anti-leukemic effects (p<0.01) significantly improving upon monotherapy. Software analyses yielded a combination Index (CI) <1 consistent with synergistic anti-leukemic effects. Combination GO and Talazoparib therapy also significantly enhanced AML cell apoptosis (p=0.0111) and levels of DNA damage (phosphorylated H2AX) (p=0.0054) over single agent activity. Evaluation of PARP trapping by western blot analysis is ongoing. In vivo administration of GO (1-50μg/kg) and Talazoparib (0.1-0.33mg/kg) in NSG mice with systemic engraftment of luciferase tagged human CD33+ AML cells was generally well tolerated with no significant weight loss or early morbidity. Single agent GO and Talazoparib therapy decreased systemic AML burden in a dose dependent manner and prolonged overall survival over vehicle treated mice (P<0.05). Concomitant GO (1μg/kg) and Talazoparib (0.33μg/kg) treatment was similarly well tolerated with no notable weight loss or toxicities. Combination GO and Talazoparib therapy significantly prolonged overall survival of leukemia xenografted mice over vehicle (p=0.0018) and single agent therapy with the same doses of GO (p=0.0018) and Talazoparib (p=0.0499), respectively). Conclusions In summary, our results demonstrate that the addition of the PARP inhibitor, Talazoparib, to the CD33 antibody drug conjugate, GO, results in potent in vitro and in vivo anti-tumor activity in human CD33+ AML preclinical models. Further studies investigating this novel combinatorial approach in AML are currently ongoing. Due to GO's FDA approval for CD33+ AML in 2018, this data strongly supports future clinical investigation using PARP inhibitors as a novel class of agents for combination therapy to significantly enhance the efficacy of ADCs. Figure 1 Disclosures Wang: Amgen: Other: Advisory role; Agios: Other: Advisory role; Stemline: Other: Advisory role, Speakers Bureau; Daiichi: Other: Advisory role; Abbvie: Other: Advisory role; Kite: Other: Advisory role; Jazz: Other: Advisory role; Astellas: Other: Advisory role, Speakers Bureau; celyad: Other: Advisory role; Pfizer: Other: Advisory role, Speakers Bureau.


2021 ◽  
Author(s):  
Gregory D. Bowden ◽  
Sophie Stotz ◽  
Johannes Kinzler ◽  
Christian Geibel ◽  
Michael Laemmerhofer ◽  
...  

PARP inhibitors are proven chemotherapeutics and serve as lead structures for the development of PARP-targeted in vivo imaging probes. Given the clinical potential of PARP imaging for the detection and stratification of various cancers, the development of novel PARP imaging probes with improved pharmacologi-cal profiles over established PARP imaging agents is warranted. Here, we present a novel 18F-labeled PARP radiotracer based on the clinically superior PARP inhibitor talazoparib. An automated radiosynthesis of [18F]talazoparib (RCY: 13 ± 3.4 %; n = 4; molar radioactivity 52 – 176 GBq/μmol) was achieved using a “Design of Experiments” (DoE) optimized copper-mediated radiofluorination reaction. The chiral product was isolated from the reaction mixture using 2D reversed-phase/chiral radio-HPLC (>99% ee). (8S, 9R)-[18F]Talazoparib demonstrated PARP binding in HCC1937 cells in vitro and showed an excellent tumor-to-blood ratio in xeno-graft-bearing mice (10.2 ± 1.5). Despite expected uptake into muscle, bone, and abdominal tissue, a favorable pharmacological profile in terms of excretion, blood half-life, and target engagement was observed in the pilot in vivo study. This synthesis of [18F]talazoparib exemplifies how a DoE based tracer development pipeline can enable the radiosyntheses of clinically relevant but synthetically challenging radiolabeled compounds of high interest to the imaging community.


2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Kentaro Nakamura ◽  
Seido Takae ◽  
Eriko Shiraishi ◽  
Kiemi Shinya ◽  
Arby Jane Igualada ◽  
...  

Abstract The use of poly (ADP-ribose) polymerase (PARP) inhibitors is expected to increase, but their effect on fertility is still unclear. The aim of this study was to investigate the effect of PARP inhibitors on ovarian function. In an in vitro study, cultures of ovaries and granulosa cells (GCs) exposed to the PARP inhibitor olaparib were evaluated by real-time RT-PCR, histological study, and hormone assays. In an in vivo study, mice were administered olaparib orally and evaluated via in vitro fertilization (IVF), follicle count, immunohistochemical staining, and real-time RT-PCR. In vitro, the gene expression of GC markers decreased in the olaparib-treated group. Olaparib also negatively affected estradiol production and the expression of GC markers in cultured GCs, with abnormal morphology of GCs observed in the treated group. The follicle number indicated depletion of follicles due to atretic changes in the treatment group, both in vitro and in vivo. Also, olaparib reduced the number of retrieved oocytes and the fertilization rate of IVF, but they recovered after 3 weeks of cessation. Our results indicate that olaparib is toxic to ovaries.


Oncogene ◽  
2021 ◽  
Author(s):  
Stefano Di Giulio ◽  
Valeria Colicchia ◽  
Fabio Pastorino ◽  
Flaminia Pedretti ◽  
Francesca Fabretti ◽  
...  

AbstractMYCN drives aggressive behavior and refractoriness to chemotherapy, in several tumors. Since MYCN inactivation in clinical settings is not achievable, alternative vulnerabilities of MYCN-driven tumors need to be explored to identify more effective and less toxic therapies. We previously demonstrated that PARP inhibitors enhance MYCN-induced replication stress and promote mitotic catastrophe, counteracted by CHK1. Here, we showed that PARP and CHK1 inhibitors synergized to induce death in neuroblastoma cells and in primary cultures of SHH-dependent medulloblastoma, their combination being more effective in MYCN amplified and MYCN overexpressing cells compared to MYCN non-amplified cells. Although the MYCN amplified IMR-32 cell line carrying the p.Val2716Ala ATM mutation showed the highest sensitivity to the drug combination, this was not related to ATM status, as indicated by CRISPR/Cas9-based correction of the mutation. Suboptimal doses of the CHK1 inhibitor MK-8776 plus the PARP inhibitor olaparib led to a MYCN-dependent accumulation of DNA damage and cell death in vitro and significantly reduced the growth of four in vivo models of MYCN-driven tumors, without major toxicities. Our data highlight the combination of PARP and CHK1 inhibitors as a new potential chemo-free strategy to treat MYCN-driven tumors, which might be promptly translated into clinical trials.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Diana Zatreanu ◽  
Helen M. R. Robinson ◽  
Omar Alkhatib ◽  
Marie Boursier ◽  
Harry Finch ◽  
...  

AbstractTo identify approaches to target DNA repair vulnerabilities in cancer, we discovered nanomolar potent, selective, low molecular weight (MW), allosteric inhibitors of the polymerase function of DNA polymerase Polθ, including ART558. ART558 inhibits the major Polθ-mediated DNA repair process, Theta-Mediated End Joining, without targeting Non-Homologous End Joining. In addition, ART558 elicits DNA damage and synthetic lethality in BRCA1- or BRCA2-mutant tumour cells and enhances the effects of a PARP inhibitor. Genetic perturbation screening revealed that defects in the 53BP1/Shieldin complex, which cause PARP inhibitor resistance, result in in vitro and in vivo sensitivity to small molecule Polθ polymerase inhibitors. Mechanistically, ART558 increases biomarkers of single-stranded DNA and synthetic lethality in 53BP1-defective cells whilst the inhibition of DNA nucleases that promote end-resection reversed these effects, implicating these in the synthetic lethal mechanism-of-action. Taken together, these observations describe a drug class that elicits BRCA-gene synthetic lethality and PARP inhibitor synergy, as well as targeting a biomarker-defined mechanism of PARPi-resistance.


2021 ◽  
Author(s):  
Gregory D. Bowden ◽  
Sophie Stotz ◽  
Johannes Kinzler ◽  
Christian Geibel ◽  
Michael Laemmerhofer ◽  
...  

PARP inhibitors are proven chemotherapeutics and serve as lead structures for the development of PARP-targeted in vivo imaging probes. Given the clinical potential of PARP imaging for the detection and stratification of various cancers, the development of novel PARP imaging probes with improved pharmacologi-cal profiles over established PARP imaging agents is warranted. Here, we present a novel 18F-labeled PARP radiotracer based on the clinically superior PARP inhibitor talazoparib. An automated radiosynthesis of [18F]talazoparib (RCY: 13 ± 3.4 %; n = 4; molar radioactivity 52 – 176 GBq/μmol) was achieved using a “Design of Experiments” (DoE) optimized copper-mediated radiofluorination reaction. The chiral product was isolated from the reaction mixture using 2D reversed-phase/chiral radio-HPLC (>99% ee). (8S, 9R)-[18F]Talazoparib demonstrated PARP binding in HCC1937 cells in vitro and showed an excellent tumor-to-blood ratio in xeno-graft-bearing mice (10.2 ± 1.5). Despite expected uptake into muscle, bone, and abdominal tissue, a favorable pharmacological profile in terms of excretion, blood half-life, and target engagement was observed in the pilot in vivo study. This synthesis of [18F]talazoparib exemplifies how a DoE based tracer development pipeline can enable the radiosyntheses of clinically relevant but synthetically challenging radiolabeled compounds of high interest to the imaging community.


Sign in / Sign up

Export Citation Format

Share Document