Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Journal of Pharmacology and Experimental Therapeutics
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • My Cart
Journal of Pharmacology and Experimental Therapeutics

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Visit jpet on Facebook
  • Follow jpet on Twitter
  • Follow jpet on LinkedIn
Research ArticleCellular and Molecular

Poly(ADP-Ribose) Polymerase Inhibitors for Arsenic Trioxide–Resistant Acute Promyelocytic Leukemia: Synergistic In Vitro Antitumor Effects with Hypomethylating Agents or High-Dose Vitamin C

Manuela Giansanti, Antonio De Gabrieli, Salvatore Pasquale Prete, Tiziana Ottone, Maria Domenica Divona, Terry Karimi, Fabio Ciccarone, Maria Teresa Voso, Grazia Graziani and Isabella Faraoni
Journal of Pharmacology and Experimental Therapeutics June 2021, 377 (3) 385-397; DOI: https://doi.org/10.1124/jpet.121.000537
Manuela Giansanti
Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy (M.G., A.D.G., S.P.P., T.K., G.G., I.F.); Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, Rome, Italy (M.G.); Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy (T.O., M.D., M.T.V.); Unit of Neuro-Oncohematology, Santa Lucia Foundation-IRCCS, Rome, Italy (T.O., M.T.V.); and IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy (F.C.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Antonio De Gabrieli
Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy (M.G., A.D.G., S.P.P., T.K., G.G., I.F.); Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, Rome, Italy (M.G.); Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy (T.O., M.D., M.T.V.); Unit of Neuro-Oncohematology, Santa Lucia Foundation-IRCCS, Rome, Italy (T.O., M.T.V.); and IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy (F.C.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Salvatore Pasquale Prete
Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy (M.G., A.D.G., S.P.P., T.K., G.G., I.F.); Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, Rome, Italy (M.G.); Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy (T.O., M.D., M.T.V.); Unit of Neuro-Oncohematology, Santa Lucia Foundation-IRCCS, Rome, Italy (T.O., M.T.V.); and IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy (F.C.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Tiziana Ottone
Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy (M.G., A.D.G., S.P.P., T.K., G.G., I.F.); Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, Rome, Italy (M.G.); Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy (T.O., M.D., M.T.V.); Unit of Neuro-Oncohematology, Santa Lucia Foundation-IRCCS, Rome, Italy (T.O., M.T.V.); and IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy (F.C.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Maria Domenica Divona
Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy (M.G., A.D.G., S.P.P., T.K., G.G., I.F.); Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, Rome, Italy (M.G.); Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy (T.O., M.D., M.T.V.); Unit of Neuro-Oncohematology, Santa Lucia Foundation-IRCCS, Rome, Italy (T.O., M.T.V.); and IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy (F.C.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Terry Karimi
Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy (M.G., A.D.G., S.P.P., T.K., G.G., I.F.); Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, Rome, Italy (M.G.); Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy (T.O., M.D., M.T.V.); Unit of Neuro-Oncohematology, Santa Lucia Foundation-IRCCS, Rome, Italy (T.O., M.T.V.); and IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy (F.C.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Fabio Ciccarone
Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy (M.G., A.D.G., S.P.P., T.K., G.G., I.F.); Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, Rome, Italy (M.G.); Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy (T.O., M.D., M.T.V.); Unit of Neuro-Oncohematology, Santa Lucia Foundation-IRCCS, Rome, Italy (T.O., M.T.V.); and IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy (F.C.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Maria Teresa Voso
Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy (M.G., A.D.G., S.P.P., T.K., G.G., I.F.); Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, Rome, Italy (M.G.); Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy (T.O., M.D., M.T.V.); Unit of Neuro-Oncohematology, Santa Lucia Foundation-IRCCS, Rome, Italy (T.O., M.T.V.); and IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy (F.C.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Grazia Graziani
Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy (M.G., A.D.G., S.P.P., T.K., G.G., I.F.); Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, Rome, Italy (M.G.); Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy (T.O., M.D., M.T.V.); Unit of Neuro-Oncohematology, Santa Lucia Foundation-IRCCS, Rome, Italy (T.O., M.T.V.); and IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy (F.C.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Grazia Graziani
Isabella Faraoni
Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy (M.G., A.D.G., S.P.P., T.K., G.G., I.F.); Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, Rome, Italy (M.G.); Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy (T.O., M.D., M.T.V.); Unit of Neuro-Oncohematology, Santa Lucia Foundation-IRCCS, Rome, Italy (T.O., M.T.V.); and IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy (F.C.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Isabella Faraoni
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Abstract

Arsenic trioxide (ATO) is an anticancer agent used for the treatment ofacute promyelocytic leukemia (APL). However, 5%–10% of patients fail to respond or experience disease relapse. Based on poly(ADP-ribose) polymerase (PARP) 1 involvement in the processing of DNA demethylation, here we have tested the in vitro susceptibility of ATO-resistant clones (derived from the human APL cell line NB4) to PARP inhibitors (PARPi) in combination with hypomethylating agents (azacitidine and decitabine) or high-dose vitamin C (ascorbate), which induces 5-hydroxymethylcytosine (5hmC)-mediated DNA demethylation. ATO-sensitive and -resistant APL cell clones were generated and initially analyzed for their susceptibility to five clinically used PARPi (olaparib, niraparib, rucaparib, veliparib, and talazoparib). The obtained PARPi IC50 values were far below (olaparib and niraparib), within the range (talazoparib), or above (rucaparib and veliparib) the Cmax reported in patients, likely as a result of differences in the mechanisms of their cytotoxic activity. ATO-resistant APL cells were also susceptible to clinically relevant concentrations of azacitidine and decitabine and to high-dose ascorbate. Interestingly, the combination of these agents with olaparib, niraparib, or talazoparib resulted in synergistic antitumor activity. In combination with ascorbate, PARPi increased the ascorbate-mediated induction of 5hmC, which likely resulted in stalled DNA repair and cytotoxicity. Talazoparib was the most effective PARPi in synergizing with ascorbate, in accordance with its marked ability to trap PARP1 at damaged DNA. These findings suggest that ATO and PARPi have nonoverlapping resistance mechanisms and support further investigation on PARPi combination with hypomethylating agents or high-dose ascorbate for relapsed/ATO-refractory APL, especially in frail patients.

SIGNIFICANCE STATEMENT This study found that poly(ADP-ribose) inhibitors (PARPi) show activity as single agents against human acute promyelocytic leukemia cells resistant to arsenic trioxide at clinically relevant concentrations. Furthermore, PARPi enhance the in vitro efficacy of azacitidine, decitabine, and high-dose vitamin C, all agents that alter DNA methylation. In combination with vitamin C, PARPi increase the levels of 5-hydroxymethylcytosine, likely as a result of altered processing of the oxidized intermediates associated with DNA demethylation.

Introduction

Acute promyelocytic leukemia (APL) is a subtype of acute myeloid leukemia (AML) with aggressive clinical presentation that is characterized by the reciprocal balanced translocation t(15;17), involving the promyelocytic leukemia (PML) and retinoic acid receptor α (RARA) genes. The PML/RARA fusion protein blocks myeloid differentiation at the promyelocyte stage and induces aberrant self-renewal of APL cells with disruption of normal hematopoiesis. PML-RARA acts as a transcriptional repressor of RARA target genes, deregulating the homeostatic control of development, expansion, and maturation/differentiation of hematopoietic stem cells. Moreover, PML-RARA interferes with the normal formation of PML-nuclear bodies, leading to impaired stress response, decreased DNA damage repair, and reduced cell propensity to undergo senescence and apoptosis (Gurnari et al., 2019).

For several years, treatment of newly diagnosed APL has been centered on the use of all-trans retinoic acid (ATRA) in combination with an anthracycline, resulting in long-term remission rates above 80% (Sanz et al., 2009). For low-risk APL, ATRA plus arsenic trioxide (ATO) is the standard of care in the frontline setting (Sanz et al., 2019), with long-term response rates exceeding 90% (Lo-Coco et al., 2013; Cicconi et al., 2020). The current National Comprehensive Cancer Network treatment guidelines for APL have also included ATO in the frontline therapy of high-risk APL patients without cardiac dysfunction, in combination with ATRA and an anthracycline or ATRA and the antibody-drug conjugate gemtuzumab ozogamicin (i.e., an anti-CD33 monoclonal antibody conjugated with the DNA-damaging agent calicheamycin). Moreover, ATO is used for relapsed/refractory APL with or without ATRA, gemtuzumab ozogamicin, or an anthracycline, depending on remission duration and the chemotherapeutic agents used for first-line therapy (https://www.nccn.org/professionals/physician_gls/pdf/aml.pdf).

In APL blasts, ATO binds to the PML portion of the hybrid oncoprotein and stimulates its sumoylation, polyubiquitination, and proteasomal degradation. This process is followed by the restoration of PML-nuclear bodies and induction of apoptosis in APL cells (Noguera et al., 2019). ATO also possesses additional mechanisms, including generation of reactive oxygen species (ROS) (Miller et al., 2002). Despite the excellent results obtained with ATRA/ATO therapy, 5%–10% of patients develop relapsed/refractory disease (Sanz et al., 2019), and in patients not eligible for allogeneic hematopoietic cell transplantation or who fail to respond to second-line agents, enrollment in a clinical trial is encouraged.

The best-characterized molecular mechanism involved in ATO resistance is represented by missense somatic mutations within the B2 ATO-binding domain of PML gene (40% of ATO-resistant APL cases), which prevent ATO binding and impede degradation of PML/RARA oncoprotein (Goto et al., 2011; Zhu et al., 2014; Madan et al., 2016). The most common PML-A216V/T amino acidic mutation can also be found in the unrearranged PML allele (Iaccarino et al., 2016). Other PML-unrelated mechanisms may contribute to ATO resistance, such as cellular metabolic adaptation, dysregulation of redox signaling, presence of the X-RARA oncoprotein instead of PML-RARA, and mutations in other genes (Alex et al., 2014; Balasundaram et al., 2016; Iaccarino et al., 2019; Noguera et al., 2019).

In the search of potential therapeutic approaches for APL relapsed/refractory to ATO, we have generated an in vitro model of APL human sublines with acquired resistance to ATO and focused our attention on poly(ADP-ribose) polymerase inhibitors (PARPi) based on preclinical evidence of their activity against myeloid malignancies (Faraoni et al., 2015, 2018; Esposito et al., 2015; Nieborowska-Skorska et al., 2017; Zhao and So, 2017; Kohl et al., 2019). These agents belong to a new class of orally administered anticancer drugs that mainly act by dampening the activity of PARP1, a nuclear enzyme required for sensing and repairing DNA damage. Five PARPi have recently been approved for advanced/recurrent ovarian, breast, pancreatic, or prostate cancers with defective homologous recombination due to mutated BRCA1/2 genes or other genetic/epigenetic alterations leading to reduced repair of DNA double-strand breaks (Faraoni and Graziani, 2018). Moreover, these and other PARPi are currently under clinical investigation as monotherapy and in combination with targeted agents or chemotherapy for several types of cancers, including hematological malignancies (www.clinicaltrials.gov).

Our previous studies revealed that PARPi exerted cytotoxic effects in primary cultures of AML blasts and leukemia cell lines. Among the different AML cell lines tested, the promyelocytic cell line NB4 was the most sensitive to the PARPi olaparib (Faraoni et al., 2015). In addition, studies in murine and human AML grafts revealed that PML/RARA translocation-driven leukemia was extremely sensitive to olaparib and veliparib (Esposito et al., 2015).

In the present study, we have compared the antitumor activity of different PARPi (olaparib, niraparib, rucaparib, talazoparib, and veliparib) in APL cells rendered resistant to ATO as monotherapy and combined with agents endowed with antileukemic activity and whose mechanism of action involves a DNA damage response with PARP1 intervention. In particular, PARPi have been tested in combination with the DNA hypomethylating agents azacitidine and decitabine or with high-dose vitamin C (hereafter referred to as ascorbate), which has been shown to promote 5-hydroxymethylcytosine (5hmC)-mediated DNA demethylation by enhancing the activity of ten-eleven translocation (TET) enzymes (Minor et al., 2013). Results indicated that olaparib, niraparib, and talazoparib in combination with the aforementioned DNA demethylating agents exerted synergistic antiproliferative effects against APL cells, including those resistant to ATO. The increased DNA damage observed in APL cells exposed to PARPi plus ascorbate was associated with a significant increase in the levels of 5hmC, likely as a consequence of altered processing of the oxidized intermediates associated with DNA demethylation.

Materials and Methods

Generation of NB4 Clones and Cell Culture Conditions

The promyelocytic leukemia cell line NB4 (American Type Culture Collection, Manassas, VA) was cultured in RPMI 1640 medium (Sigma-Aldrich, St. Louis, MO) supplemented with 2 mM l-glutamine (EuroClone, Pero, Milan, Italy), 1% penicillin/streptomycin (EuroClone), and 20% fetal bovine serum at 37°C in a humidified CO2 incubator. Four different clones (CL1, CL2, CL3, and CL4) were produced by limiting dilution from the NB4 cell line at early passages from the initial stock purchased from American Type Culture Collection (Fig. 1). To generate ATO-resistant cells, cell clones were exposed to increasing concentrations of ATO (0.1–1 μM) for about 1 year, and the corresponding ATO-resistant clones were named CL1-R, CL2-R, CL3-R, and CL4-R. At this time, to preserve the resistant phenotype, cells were frozen in several aliquots. ATO-resistant clones were maintained in culture with 1 µM ATO; experiments were performed after at least two passages from the last ATO treatment. The parental NB4 cell line and its ATO-sensitive and -resistant clones were authenticated by testing the expression of the fusion PML/RARA gene (see below).

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

ATO-resistant clones maintain APL phenotype. (A) Flowchart of the generation of ATO-sensitive and -resistant NB4 clones. Four different clones were isolated by limiting dilution from the bulk NB4 cell line (CL1, CL2, CL3, CL4). Each clone was independently exposed to increasing concentrations of ATO (0.1–1 μM) to generate the corresponding four ATO-resistant clones (CL1-R, CL2-R, CL3-R, CL4-R). (B) Expression of the PML/RARA transcript evaluated by RT-PCR analysis in ATO-sensitive and -resistant clones and the NB4 cell line. The ABL proto-oncogene 1, non-receptor tyrosine kinase (ABL1) was used as a housekeeping gene. (C) Proliferation rate of ATO-sensitive and -resistant clones analyzed by cell count and trypan blue exclusion assay (triplicate counts) at 24, 48, and 72 hours. Each plot shows cell growth of the ATO-sensitive clone and its ATO-resistant counterpart at the indicated times. Values are means ± S.D. of three independent experiments.

Drug Treatment and Survival Assay

The ATO (As2O3; Sigma-Aldrich) stock solution was prepared by dissolving the drug in 1 N NaOH and diluting it in PBS (EuroClone) at a final concentration of 2 mM. The stock solutions of PARPi (2 mM olaparib, talazoparib, and veliparib; 20 mM niraparib and rucaparib; Selleck Chemicals, Houston, TX) were prepared by dissolving the powder of each compound in DMSO (Sigma-Aldrich), followed by dilution in RPMI 1640 medium. Decitabine (Cayman Chemical, Ann Arbor, MI) and azacitidine (Sigma-Aldrich) were dissolved in PBS (2 mM) and DMSO (20 mM), respectively. Ascorbate (l-ascorbate; Sigma-Aldrich) was diluted in RPMI 1640 medium at 250 mM concentration. Drug aliquots were stored at −80°C, and for each experiment, a new aliquot was thawed and used. In all experiments, the DMSO final concentration in the culture medium was always <0.01% (v/v).

For cell treatment, drugs were added at the beginning of each experiment and left in culture medium until cell harvesting. NB4 parental cell line and its clones were treated with the PARPi olaparib (1.25–20 µM), niraparib (1.25–10 µM), talazoparib (12.5–100 nM), rucaparib (1.25–10 µM), and veliparib (5–20 µM) as single agents or in combination with azacitidine (1.25–1 µM), decitabine (25–500 nM), or ascorbate (0.25–1 mM). Drug concentrations tested always included the plasma peak concentration (Cmax) values reached in patients with cancer.

For survival assays, cells were analyzed by the MTS viability test (Promega, Madison, WI), according to the manufacturer’s instructions, or by trypan blue dye exclusion count. The drug concentration capable of inhibiting 50% of cell growth (IC50) compared with the untreated control was extrapolated from the dose-response curves by using linear regression (GraphPad Prism 5 software; GraphPad Inc., San Diego, CA). The dose-effect curves were analyzed by the median-effect method of Chou and Talalay with the CompuSyn software (ComboSyn Inc., Paramus, NJ). The combination index (CI) indicates a quantitative measure of the drug combination effects in terms of synergistic (CI < 1), additive (CI = 1), or antagonistic effect (CI > 1).

Apoptosis was evaluated by flow cytometry analysis of the sub-G1 fraction after cell fixation in ethanol, treatment with 10 μg/ml RNase A (Sigma-Aldrich), and staining with 100 μg/ml propidium iodide (PI) (Sigma-Aldrich) for 20 minutes at 37°C in the dark. Samples (5 × 104 cells) were acquired on a BD FACSCalibur flow cytometer and evaluated using CellQuest Software (BD Biosciences, San Jose, CA).

Molecular Analysis of PML/RARA

Total RNA was isolated by Trizol reagent (Invitrogen, Thermo Fisher Scientific, Waltham, MA). In total, 1 µg of RNA was reverse-transcribed with random hexamer primers and amplified (reagents from Life Technologies, Thermo Fisher Scientific) with the GeneAmp PCR System 9700 (Applied Biosystems, Foster City, CA). RT-PCR for PML/RARA detection was carried out using standard protocols (van Dongen et al., 1999).

For sequencing the region of PML gene corresponding to the B2 ATO-binding domain, the PML/RARA fusion transcript was amplified by polymerase chain reaction and analyzed by Sanger sequencing as reported elsewhere (Iaccarino et al., 2016).

Immunoblot Analysis of Apoptosis and DNA Damage Markers

Total cellular proteins were extracted using a buffer containing 50 mM Tris-HCl (pH 7.5), 5 mM EDTA, 5 mM EGTA, 150 mM NaCl, 1% Nonidet P-40, 1 mM Na orthovanadate, 20 mM β-glycerophosphate, 1 mM AEBSF (Sigma-Aldrich), and protease inhibitor cocktail (Thermo Fisher Scientific). Protein aliquots were loaded onto SDS polyacrylamide gel electrophoresis and blotted to nitrocellulose membranes. Filters were incubated with the following antibodies: anti-PARP1 (1:1000, C2-10; Trevigen, Gaithersburg, MD), anti–caspase 8 (1:500, 12F5; Enzo Life Sciences, NY), anti–cleaved caspase 8 (1:400, Asp374; Cell Signaling Technology, Danvers, MA), anti–caspase 3 (1:1000, D3R6Y; Cell Signaling Technology), anti–cleaved caspase 3 (1:1000, D175; Cell Signaling Technology), anti-γH2AX (1:1000, JBW301; Millipore, Burlington, MA), anti–glyceraldehyde 3-phosphate dehydrogenase (GAPDH) (1:1000, 14C10; Cell Signaling Technology), and anti–β-actin (1:2000, A2066; Sigma-Aldrich). Horseradish peroxidase–conjugated IgGs were used as secondary antibodies (1:5000, anti-mouse A4416; Sigma-Aldrich; anti-rabbit NA934; GE Healthcare, Chicago, IL). The autoradiograms were subjected to densitometric analysis using the ImageJ 1.45s software (Schneider et al., 2012), and results were normalized against GAPDH or β-actin.

Determination of Intracellular ROS

Total intracellular ROS were evaluated by the 2,7-dichlorodihydrofluorescein diacetate (CM-H2DCFDA; Invitrogen, Thermo Fisher Scientific) reagent that is deacetylated by nonspecific esterases and oxidized in 2,7-dichlorodihydrofluorescein (DCF) fluorescent compound by hydroxyl and peroxyl radicals or other intracellular ROS in the cells. Cytosolic superoxide anion production was detected by dihydroethidium (DHE; Invitrogen, Thermo Fisher Scientific) reagent. DHE is oxidized by superoxide anion in 2-hydroxyethidium, which becomes fluorescent after intercalation into DNA. For ROS analysis, cells were harvested after 4 hours of treatment with graded concentrations of ascorbate. Cells (5 × 105) were incubated with 5 µM of CM-H2DCFDA or DHE at 37°C for 30 minutes in 5% fetal bovine serum in PBS and analyzed by the BD FACSCalibur flow cytometer.

Analysis of 5hmC by Dot Blot Assay

Cells were treated with ascorbate or PARPi (i.e., olaparib, niraparib, or talazoparib), as single agents or in combination, for 24 hours, and then genomic DNA was extracted with DNeasy Blood and Tissue Kit (Qiagen, Hilden, Germany). Dot blots were performed as previously published (Ciccarone et al., 2018). Briefly, DNA was denatured in 0.4 N NaOH, 10 mM EDTA, at 95°C for 10 minutes and neutralized with an equal volume of cold 4 M ammonium acetate (pH 7.0). Starting from 2 µg of denatured DNA, 2-fold dilutions of each sample were spotted on the nylon membrane Hybond-N+ (GE Healthcare) in an assembled Bio-Dot apparatus (Bio-Rad Laboratories, Hercules, CA). Each well was washed with 0.4 N NaOH, 10 mM EDTA, and then 2× saline-sodium citrate buffer. After baking at 80°C for 15 minutes, air-dried membranes were blocked in 5% skimmed milk in Tris-buffered saline/Tween 20and incubated with anti-5hmC antibody (39769; Active Motif, Carlsbad, CA) and anti-rabbit horseradish peroxidase–conjugated secondary antibody. Dot-blot signals were revealed by chemiluminescence. Equal spotting of total DNA onto the membrane was checked by staining the same blotted filter with 0.02% methylene blue in 0.3 M sodium acetate (pH 5.2).

Statistical Analysis

Statistical analysis was performed by using the GraphPad Prism 5 software, and data were reported as means ± S.D. Statistical analysis of the differences in IC50 values between two groups was performed by unpaired Student’s t test. For multiple comparisons, unpaired one-way ANOVA analysis followed by least significant difference (LSD) test was used. All statistical tests were two-sided. Differences were considered statistically significant when P < 0.05.

Results

Generation of ATO-Resistant Clones

The NB4 cell line was originally derived from the bone marrow of a 23-year-old woman with APL (French-American-British classification system M3) (Lanotte et al., 1991). From this cell line, we generated four clones by limiting dilution (CL1, CL2, CL3, and CL4), and these were subsequently exposed to increasing concentrations of ATO (0.1–1 µM) within a time frame of 1 year to generate the corresponding ATO-resistant clones (CL1-R, CL2-R, CL3-R, and CL4-R) (flowchart in Fig. 1A). Sensitive and resistant clones were characterized for PML/RARA expression by RT-PCR. All NB4 clones, including the ATO-resistant ones, maintained the expression of the fusion PML/RARA gene (Fig. 1B). Sequencing of the PML allelic region corresponding to the B2 ATO-binding domain indicated that all ATO-resistant clones lacked the PML-A216V/T mutation or other mutations in the PML B2 domain (data not shown).

Since the proliferation rate might affect tumor cell response to ATO, we analyzed the growth pattern of the APL clones by cell count and found no significant differences between the ATO-sensitive clones and their corresponding ATO-resistant counterparts (Fig. 1C).

The in vitro susceptibility to ATO antiproliferative effects of the parental and ATO-selected clones or of the NB4 bulk cell line was analyzed by MTS assay after 3 days of treatment with graded drug concentrations. The ATO IC50 in the resistant clones (2.6–4.5 µM range) was 5- to 9-fold higher than in the corresponding sensitive clones (0.5–0.9 μM range) (Fig. 2A). Analysis of apoptosis after treatment with 0.5 and 1 μM ATO revealed the induction of cell death in ATO-sensitive clones in a concentration-dependent manner, whereas no or marginal apoptotic effects (at the higher concentration tested) were observed in ATO-resistant cells (Fig. 2B). Consistent with the results of flow cytometry, showing apoptotic induction in ATO-sensitive cells, immunoblot analysis demonstrated that this effect was associated with cleavage of PARP1, caspase 3, and caspases 8, the latter indicating activation of the apoptotic extrinsic pathway. Moreover, ATO induced DNA damage only in ATO-sensitive clones, as evidenced by the high expression levels of histone 2AX phosphorylation at serine 139 (γH2AX) (Fig. 2C).

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Differential sensitivity of ATO-sensitive and -resistant APL clones to the antiproliferative, apoptotic, and DNA-damaging effects of ATO. (A) Analysis of APL clones’ susceptibility to the antiproliferative effects of ATO. All NB4-derived cell clones were treated with ATO (0–2 µM), and after 3 days, proliferation was evaluated by the MTS assay. The mean IC50 values ± S.D., calculated from at least three independent experiments, are reported. Statistical analysis was assessed by unpaired t test: *P < 0.05; **P < 0.01. (B) Apoptosis analysis. Induction of apoptosis was evaluated by PI staining and flow cytometry of untreated cells (white bars) or cells treated with 0.5 µM (gray bars) and 1 µM (black bars) ATO at 48 hours after drug exposure. The results of three independent experiments are expressed as mean percentages ± S.D. of PI-positive cells in ATO-sensitive (left panel) and -resistant (right panel) clones. Statistical analysis was performed by unpaired one-way ANOVA (*P < 0.05; **P < 0.01; ***P < 0.001). (C) Western blot analysis of proteins associated with the apoptotic pathway (PARP1, caspase 8 and 3) and DNA damage (γH2AX). GAPDH was used as a housekeeping gene.

ATO-Resistant Clones Are Sensitive to PARPi

Recent preclinical studies have shown that PARPi exert cytotoxic effects against myeloid malignancies (Faraoni, et al., 2019a and b; Fritz et al., 2021). To investigate the potential activity of different PARPi in APL cells, ATO-sensitive and -resistant NB4 clones as well as the bulk cell line were exposed to increasing concentrations of olaparib, niraparib, talazoparib, rucaparib, and the investigational PARPi veliparib. Cell growth was analyzed by MTS assay on day 6 after a single exposure to the PARPi. In fact, based on our previous studies, the antiproliferative activity of PARPi in myeloid tumor cells requires prolonged drug exposure (Faraoni et al., 2015, 2018, 2019a). The drug concentrations tested in the experiments always included the plasma Cmax reported in patients with cancer during phase 1 clinical trials (Cmax ranges were represented by dotted lines in Fig. 3) (Fong et al., 2009; Kummar et al., 2009; Sandhu et al., 2013; Mateo et al., 2016; de Bono et al., 2017; Kristeleit et al., 2017; Nishikawa et al., 2017; Shapiro et al., 2019). All clones and the NB4 cell line were sensitive to olaparib with IC50 values lower (2.9–12.1 µM) than the reference Cmax values (range 16–22 µM). Moreover, the ATO-resistant CL1-R and CL4-R clones showed significantly lower susceptibility to olaparib compared with their ATO-sensitive counterparts (Fig. 3A). All clone pairs presented comparable susceptibility to niraparib with IC50 values (0.7–2.0 µM) below or within the Cmax range (1.2–4.4 µM) (Fig. 3B). In the case of talazoparib, the IC50 values of most clones (five of eight clones; 16.0–56.7 nM) were below or within the Cmax (30–60 nM), and no significant differences were observed between ATO-sensitive and -resistant clones, except for the CL3/CL3-R couple, with CL3-R being significantly more sensitive to talazoparib than CL3 (Fig. 3C). Regarding rucaparib and veliparib, in almost all cases (seven of eight clones for rucaparib and eight of eight clones for veliparib), the obtained IC50 values were above the Cmax values (0.6–9.5 µM and 2.6–13.5 µM, respectively) (Fig. 3, D and E). Similar to what was observed with talazoparib, CL3-R was also more susceptible to these PARPi compared with its parental CL3 clone. Overall, these results indicated that ATO-resistant NB4 clones were still responsive to clinically achievable concentrations of the PARPi olaparib, niraparib, and in most cases, talazoparib.

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Susceptibility of ATO-sensitive and -resistant APL clones to the antiproliferative effects of different PARPi. NB4 cell clones were treated with increasing concentrations of the indicated PARPi, as described in Materials and Methods. After 6 days, proliferation was assessed by MTS assay, and the mean IC50 values were calculated. A) olaparib; B) niraparib; C) talazoparib; D) rucaparib; E) veliparib. The dotted line in each histogram represents the Cmax range for each PARPi reported in clinical studies. Values are means ± S.D. of three independent experiments. Statistical analysis was calculated by unpaired t test (*P < 0.05; **P < 0.01).

PARPi in Combination with Hypomethylating Agents Induce Synergistic Growth-Inhibitory Effects in ATO-Sensitive and -Resistant APL Clones

Based on our previous report on the synergistic cytotoxic effects induced by the PARPi olaparib in AML and MDS cells when tested in combination with hypomethylating agents (Faraoni, et al., 2019a), we have investigated the activity of azacitidine and decitabine against ATO-sensitive and -resistant APL cells, as single agents or in association with the PARPi. As shown in Fig. 4, A and B, in all cell clones, azacitidine and decitabine IC50 values, calculated 3 days after drug exposure, were within the range of clinically relevant concentrations previously reported for these drugs (Cashen et al., 2008; Keating, 2012). Interestingly, the growth of CL1-R and CL4-R clones was significantly more inhibited compared with their corresponding ATO-sensitive counterparts. Conversely, in other cases, no differences in azacitidine or decitabine IC50 values were observed between ATO-sensitive and -resistant clones, except for the CL2 clone, which showed higher sensitivity to decitabine compared with its ATO-resistant counterpart (Fig. 4, A and B). These findings suggested that there was no direct correlation between the susceptibility profile to ATO of APL clones and the response to both hypomethylating agents.

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Antiproliferative effects of hypomethylating agents as single agents or combined with PARPi in ATO-sensitive and -resistant APL cells. Susceptibility of NB4 clones to (A) azacitidine (AZA) or (B) decitabine (DAC) as single agents. NB4 cell clones were treated with azacitidine (0.125–2 µM) and decitabine (0.125–1 µM), followed by MTS assay. Drug IC50 values were evaluated after 3 days of treatment. Statistical analysis of the results from three independent experiments was performed by unpaired t test (*P < 0.05). (C) Antiproliferative effects of azacitidine or decitabine with a fixed concentration of the PARPi olaparib, niraparib, or talazoparib. ATO-sensitive (CL2) and -resistant (CL2-R) clones were treated with the indicated concentrations of olaparib (OLA, 2.5 μM), niraparib (NIR, 1.25 µM), or talazoparib (TAL, 25 nM) in combination with increasing concentrations of azacitidine (AZA, 0.125–1 µM) or decitabine (DAC, 31.25–500 nM). After 3 days, proliferation was evaluated by cell count in triplicate. Data are represented as surviving fraction of PARPi/azacitidine (left panel) or PARPi/decitabine (right panel) combined treatments in ATO-sensitive and -resistant CL2 clones. Values are means ± S.D. of three independent experiments. (D) PARPi/azacitidine (left panel) or PARPi/decitabine (right panel) combined effects were analyzed by CompuSyn software. Each Fa-CI plot (or Chou-Talalay plot) indicates the CI as a function of the fraction affected (Fa). CI < 1, synergistic (values below the dotted line); CI = 1, additive; CI > 1, antagonist.

To assess the activity of hypomethylating agents in combination with PARPi, CL2 and CL2-R cells were exposed to increasing concentrations of azacitidine (0.125–1 µM) or decitabine (0.031–0.5 µM) together with a fixed concentration of the PARPi that, based on our analysis, were the most active when tested as single agents (i.e., olaparib, niraparib, and talazoparib). The clone 2/2R couple was chosen because no significant differences were observed in the susceptibility to the PARPi tested between the ATO-sensitive and -resistant cells (Fig. 3). Analysis of the inhibitory effects on cell proliferation exerted by the drug combination was performed after 3 days of treatment because of the more rapid antiproliferative effects of the cytidine analogs compared with PARPi. Results indicated that the drug combination induced a greater inhibition of cell growth compared with the single hypomethylating agents in both ATO-sensitive and ATO-resistant clones (Fig. 4C). As assessed by the CompuSyn method (Chou, 2010), synergistic effects were observed regardless of the type of PARPi associated with azacitidine or decitabine, both in ATO-sensitive and -resistant clones (Fig. 4C), with CI values largely below 1 (dotted line of the Fa-CI plots) (Fig. 4D). The strong synergism observed with the combination of these drugs in APL cells is in agreement with previous data obtained with AML cell lines (Orta et al., 2014; Muvarak et al., 2016; Faraoni et al., 2019b) and primary cultures of MDS samples (Faraoni et al., 2019a).

Ascorbate Induces Synergistic Antiproliferative Effects in Combination with the PARPi Niraparib and Talazoparib

Recent in vitro and in vivo preclinical evidence indicates that high-dose ascorbate (i.e., mM concentrations) has cytotoxic activity against AML and APL cells (Mastrangelo et al., 2015; Cimmino et al., 2017; Noguera et al., 2017). In this study, NB4 clones were treated with graded concentrations of ascorbate (0.125–2 mM), and cell proliferation was assessed by cell count after 3 days of culture (Fig. 5A), a time point commonly used for evaluating ascorbate antiproliferative activity in other cellular models (Cimmino et al., 2017; Noguera et al., 2017). The obtained ascorbate IC50 values ranged from 0.6 to 1.8 mM (Fig. 5B), values comprising the range of plasma concentrations detected in cancer patients treated with high doses of this agent (Hoffer et al., 2008; Ngo et al., 2019). Interestingly, no significant differences were observed between ATO-sensitive and -resistant cells, indicating that there is no crossresistance between ascorbate and ATO.

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Antiproliferative effects of ascorbate as a single agent or combined with PARPi in ATO-sensitive and -resistant APL cells. (A and B) Susceptibility of ATO-sensitive and -resistant clones to ascorbate as a single agent. NB4 clones were treated with increasing concentrations of ascorbate (0.25–2 mM), and after 3 days, proliferation was evaluated by cell count. The surviving fraction (A) and IC50 values (B) were calculated for the parental NB4 cell line and ATO-sensitive clones (left panel) and ATO-resistant clones (right panel) (A). Results are the mean values ± S.D. of three independent experiments. (C) Antiproliferative effects of ascorbate in combination with a fixed concentration of the PARPi olaparib, niraparib, and talazoparib. ATO-sensitive and -resistant clones were treated with olaparib (OLA, 2.5 μM), niraparib (NIR, 1.25 µM), or talazoparib (TAL, 50 nM) in combination with ascorbate (ASC, 0.25–1 mM), and after 3 days, cells viability was evaluated by cell count. Data are represented as surviving fraction of ATO-sensitive (upper panel) and ATO-resistant (lower panel) clones after PARPi/ascorbate combined treatment. Mean values ± S.D. of three independent experiments are represented. (D) Combined treatment effects were analyzed by CompuSyn software as indicated in the legend of Fig. 4.

Since ascorbate has been reported to induce a TET2-mediated increase of 5hmC levels, whose processing requires base excision repair (BER) and PARP1 intervention (Pastor et al., 2013; Cimmino et al., 2017), we investigated the effect of its combination with PARPi on APL cells. Fixed PARPi concentrations were combined with graded concentrations of ascorbate. The antiproliferative effects were assessed after 3 days of treatment by cell count, and results were reported as proliferation curves in Fig. 5C (top panels, ATO-sensitive clones; bottom panels, ATO-resistant clones). The ascorbate combination with olaparib exerted synergistic effects only in three out of the eight clones (i.e., CL2, CL2-R, CL3-R), whereas its combination with niraparib resulted in additive effects in CL1 and CL1-R clones and synergistic effects in all the other clones. Notably, the ascorbate/talazoparib combination was highly effective in all ATO-sensitive and -resistant clones, with extremely low CI values (Fig. 5D), indicating this PARPi as the best candidate to be combined with ascorbate.

PARPi in Combination with Ascorbate Increase DNA Damage and 5-Hydroxymethylcytosine Levels

To investigate whether cotreatment with PARPi and ascorbate might enhance DNA damage, we first evaluated γH2AX expression by immunoblot analysis in the two ATO-resistant CL2-R and CL3-R clones, in which the combination was synergistic with all three PARPi tested (i.e., olaparib, niraparib, and talazoparib). As single agents, ascorbate (1 mM) and the PARPi induced low or moderate levels of γH2AX (Fig. 6A). Conversely, the ascorbate/PARPi combination significantly increased γH2AX expression compared with both single agents, indicating a significantly higher level of unrepaired DNA damage. This effect was more evident with 1 mM ascorbate: in CL2-R cells, it was observed with all three PARPi, whereas in CL3-R cells, it was observed mainly with talazoparib (Fig. 6A).

Fig. 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 6.

Combined treatment with PARPi/ascorbate increases DNA damage and 5hmC levels. Two ATO-resistant clones (CL2-R, CL3-R), in which the PARPi/ascorbate combination was synergistic in terms of antiproliferative effects, were treated with ascorbate (ASC, 0.5 and 1 mM) in combination with olaparib (OLA, 2.5 µM), niraparib (NIR, 1.25 µM), or talazoparib (TAL 50 nM). After 24 hours, cells were collected and analyzed for γH2AX as a marker of DNA damage, ROS production, and 5hmC levels. (A) γH2AX immunoblot analysis. Histograms represent the results of densitometric analysis of γH2AX normalized with β-actin and are the means ± S.D. of three independent experiments. Only the statistical significance of cotreatments compared with single treatments is reported. (B) Total Intracellular ROS and cytosolic superoxide anion were quantified by DCF and DHE fluorescence, respectively, in CL2-R and CL3-R cells after treatment with increasing concentrations of ascorbate. (C) DNA dot blots for 5hmC in CL2-R cells untreated (CTRL) or treated with 1 mM ascorbate, 50 nM talazoparib, and a combination of the two drugs. The same dot blot was stained with methylene blue as loading control. The graph shows the densitometric analysis of 5hmC normalized for methylene blue. (D) DNA dot blots for 5hmC in CL2-R and CL2-R clones untreated (CTRL) or treated with 1 mM ascorbate, 2.5 µM olaparib, 1.25 µM niraparib, 50 nM talazoparib, and PARPi/ascorbate combination. #This DNA sample was slightly overloaded. (E) DNA dot blots for 5hmC in the CL1-R clone, in which the PARPi/ascorbate combination was not synergistic in terms of antiproliferative effects, treated with 1 mM ascorbate, 2.5 µM olaparib, and the drug combination. The mean values ± S.D. of each histogram were obtained from three different experiments. Statistical analysis was evaluated by unpaired one-way ANOVA (*P < 0.05; **P < 0.01; ***P < 0.001).

High-dose ascorbate was previously reported to generate ROS (Chen et al., 2007) that contribute to DNA damage (Kim et al., 2018). However, in our cellular model, ascorbate concentrations below 2 mM did not increase either DCF or DHE fluorescence, tested as indicators of total intracellular ROS and cytosolic superoxide anion, respectively (Fig. 6B). Having excluded induction of DNA damage by oxidative stress, we evaluated whether the increase of γH2AX expression observed after APL cell exposure to ascorbate in combination with PARPi could be related to altered processing of 5hmC. In fact, ascorbate is a cofactor of TET enzymes, which catalyze the oxidation of 5-methylcytosine to 5hmC and other intermediates that are processed by BER during active DNA demethylation. Thus, when cells are treated with ascorbate, the rate of initial oxidation of 5-methylcytosine increases with consequent augmented formation of 5hmC (Gillberg et al., 2018). Since inhibition of PARP1 was suggested to block the BER-mediated processing of the oxidized intermediates associated with DNA demethylation (Cimmino et al., 2017), we verified whether the increase of DNA damage detected when PARPi were added to ascorbate was due to ineffective BER-mediated processing of 5hmC with a consequent rise of its levels. Indeed, in CL2-R cells, 1 mM ascorbate induced higher 5hmC levels compared with untreated or PARPi-treated cells (Fig. 6C). More interestingly, the addition of 50 nM talazoparib significantly increased 5hmC accumulation compared with ascorbate alone (Fig. 6C). Similar results were also obtained when ascorbate was combined with olaparib or niraparib and in another ATO-resistant clone (i.e., CL3-R), in which the drug combination resulted in synergistic antiproliferative effects. Conversely, combined treatment with ascorbate and olaparib of clone CL1-R did not further increase 5hmC levels compared with ascorbate alone (Fig. 6D). For this clone, the olaparib concentration tested (i.e., 2.5 µM) was markedly below the IC50 and did not result in synergistic antiproliferative effects with ascorbate (Fig. 5C). These results suggest that the synergistic cytotoxic effects observed when ascorbate was associated with PARPi are at least in part due to the blockade of BER activity during the demethylation process with consequent accumulation of 5mhC.

Discussion

In the present study, we demonstrated for the first time that PARPi increase the in vitro antiproliferative activity of hypomethylating agents and ascorbate against APL cells, including those resistant to ATO.

PARPi efficacy was first demonstrated in patients with ovarian cancer harboring germline or somatic deleterious mutations of BRCA1/2 genes, but a significant clinical benefit was also reported in the absence of BRCA1/2 mutations (Ledermann et al., 2014; Friedlander et al., 2018). In fact, genetic alterations affecting other genes involved in the repair of DNA double-strand breaks may render cancer cells more sensitive to PARPi compared with normal cells (Faraoni and Graziani, 2018). In APL cells, the presence of PML/RARA has been reported to alter the repair of DNA single- and double-strand breaks (Alcalay et al., 2003), sensitizing them to PARPi (Esposito et al., 2015). On this basis, in an attempt to identify alternative therapies for patients with APL refractory to ATO, we have generated an APL cellular model represented by clones sensitive or resistant to this arsenic derivative and tested their susceptibility to PARPi.

Among the different PARPi tested, olaparib, niraparib, and talazoparib, but not rucaparib and veliparib, induced antiproliferative and cytotoxic effects in both ATO-sensitive and -resistant APL cells at clinically relevant concentrations. Moreover, our data suggest the lack of crossresistance between ATO and PARPi. Indeed, crossresistance to ATO and conventional chemotherapeutic agents is uncommon, since ATO is not a substrate of the p-glycoprotein encoded by multidrug resistance protein 1 gene or other members of the ATP-binding cassette family of transporters, such as multidrug resistance associated protein1 or breakpoint cluster region pseudogene 1 (Takeshita et al., 2003; Sertel et al., 2012). However, repeated exposure of APL cells to ATO has been reported to induce expression of the p-glycoprotein (Takeshita et al., 2003). Regarding PARPi, although the most frequent resistance mechanism is the emergence of secondary mutations restoring BRCA1/2 function, for olaparib or other inhibitors (e.g., rucaparib, talazoparib), which are substrates of the p-glycoprotein or other efflux pumps, low intratumoral drug concentrations might also contribute to treatment failure in tumors overexpressing multidrug resistance protein 1(Lawlor et al., 2014). However, in our cellular model, the pattern of response of APL clones did not suggest the occurrence of common resistance mechanisms between the different PARPi tested.

The distinct susceptibility profile of APL clones to each PARPi may be attributed to the different mechanisms involved in the cytotoxic activity of the individual agents (i.e., catalytic inhibition vs. PARP1 trapping at DNA breaks). Indeed, although their inhibitory effects on PARP1 catalytic activity are not largely different (with IC50 values in the nanomolar range), PARPi markedly differ in the trapping ability, with talazoparib and veliparib being the most and least potent, respectively (Murai et al., 2012, 2014). Moreover, PARPi trapping potency correlated with cytotoxicity in tumor cells (Murai et al., 2012; Murai and Pommier, 2015). Consistent with their trapping efficiency, talazoparib, niraparib, and olaparib were, in this order, the most effective PARPi as a single agent in inhibiting cell proliferation of the APL clones.

ATO-sensitive and -resistant APL cells were also susceptible to clinically relevant concentrations of the hypomethylating agents azacitidine and decitabine, showing a chemosensitivity profile that did not parallel that of ATO. These agents are inhibitors of DNA methyltransferase and are used in clinical practice, particularly in elderly patients with AML who are ineligible for intensive chemotherapy and in intermediate/high-risk MDS. More recently, oral formulations of both drugs have been FDA-approved as maintenance therapy of patients with AML who achieve first complete remission but are not able to complete intensive induction chemotherapy. Both drugs are cytidine analogs that cause DNA damage as a consequence of their random incorporation into DNA (azacitidine also in RNA); covalent complex formation with DNA (cytosine-5)-methyltransferase 1, leading to its trapping onto DNA (Patel et al., 2010; Maes et al., 2014); and loss of methylated cytosines with a widespread change in gene expression (Santi et al., 1984). The synergistic effects observed in APL cells treated with hypomethylating agents plus PARPi (i.e., talazoparib, niraparib, and olaparib) are consistent with those previously reported in other experimental models, including AML, MDS, and solid tumors (e.g., ovarian cancer and non–small-cell lung cancer) (Muvarak et al., 2016; Zhao and So, 2017; Pulliam et al., 2018; Faraoni et al., 2019a; Abbotts et al., 2019). These effects are likely the result of increased DNA damage, as we and others have previously reported in several tumor models (Muvarak et al., 2016; Zhao and So, 2017; Faraoni et al., 2019a; Abbotts et al., 2019). The mechanisms underlying the observed synergistic activity include altered processing by BER and PARP1 of the aberrantly incorporated cytidine analog and trapped DNA (cytosine-5)-methyltransferase 1 (Orta et al., 2014), induction of a BRCAness (i.e., similar to hereditary BRCA-mutated tumors) phenotype by downregulating the expression of DNA repair enzymes (Abbotts et al., 2019), accumulation of ROS with consequent DNA damage that triggers PARP1 activation and becomes deleterious in the presence of PARPi (Pulliam et al., 2018), and increased drug retention at the DNA damage sites (Muvarak et al., 2016). It is reasonable to hypothesize that similar PARPi and hypomethylating agent interactions may also occur in APL cells.

ATO-sensitive and -resistant APL cells showed comparable susceptibility to millimolar concentrations of ascorbate. At physiologic concentrations, ascorbate acts as an antioxidant and cofactor of metabolic enzymes; conversely, when pharmacological doses are administered intravenously (resulting in plasma concentrations in the millimolar range), ascorbate behaves as a pro-oxidant, favoring the formation of large amounts of ROS (Mastrangelo et al., 2015; Chen et al., 2007; Kim et al., 2018; Gillberg et al., 2018). Treatment with ascorbate of patients with AML resulted in clinical benefit, especially in the presence of loss-of-function mutations of TET2 (Zhao et al., 2018; Das et al., 2019), which are frequently detected in AML and result in altered DNA demethylation (Abdel-Wahab et al., 2009; Delhommeau et al., 2009). Moreover, several clinical studies are testing high-dose ascorbate, as single agents or in combination with chemotherapeutic agents, in a variety of tumors, including AML and APL (www.clinicaltrials.gov). In our APL experimental model, ascorbate inhibited cell proliferation at concentrations devoid of pro-oxidant effects but capable of inducing DNA damage, as indicated by H2AX phosphorylation, which was likely the result of increased 5hmC formation (Cimmino et al., 2017; Agathocleous et al., 2017; Wu and Zhang, 2017).

In APL cells, the combination of ascorbate with PARPi resulted in a significant increase of cytotoxicity, DNA damage, and 5hmC levels, which is likely due to ineffective BER-mediated processing of the oxidized intermediates associated with DNA demethylation. Indeed, 5hmC is detected as DNA damage and triggers the intervention of BER and PARP1. Thus, in the presence of PARPi, the ascorbate-mediated activation of TET2 and increased generation of 5hmC in DNA may result in stalled repair and greater cytotoxicity (Kharat et al., 2020). In this context, talazoparib more potently synergized with ascorbate as compared with olaparib and niraparib, in accordance with its higher ability to trap PARP1 on DNA.

A potential drawback associated with ascorbate treatment relies on its complex pharmacokinetics and potential heterogeneous distribution in tumor and normal tissues (Giansanti et al., 2021). Although ascorbate millimolar concentrations can be achieved in plasma after intravenous injection, these high concentrations might not be easily reached at the tumor site, especially in the case of APL involving the central nervous system. In fact, only administration of its oxidized form, dehydroascorbate (DHA), may generate pharmacological levels of vitamin C in the brain, since DHA more readily crosses the blood-brain barrier via the glucose transporter GLUT1 (Spoelstra-de Man et al., 2018). However, high-dose DHA cannot always be considered a valid alternative to ascorbate, since DHA antitumor activity depends on its conversion to ascorbate by glutathione and glutathione transferases, and tumor cells might have different reducing ability and not always efficiently accumulate ascorbate (Ferrada et al., 2019). Moreover, modulation of TET activity, likely required for the observed synergism with PARPi, is mediated by ascorbate and not by vitamin C oxidized forms (Minor et al., 2013; Dickson et al., 2013; Yin et al., 2013; Guan et al., 2020).

A limitation of our study is represented by the use of different clones deriving from one cell line only (i.e., NB4). However, it should be noted that few human APL cell lines are presently available for in vitro studies. Furthermore, bone marrow samples collected from patients with APL resistant to ATO for establishing primary cultures are not readily available. Thus, preclinical in vivo studies in murine APL models might further validate our data. Moreover, our cellular model of ATO-resistant cells, lacking mutations in the PML B2 domain, did not allow evaluating the activity of the pharmacological treatment against APL cells harboring PML-A216V/T mutations. Thus, additional studies are required to evaluate drug treatment in specific genetic contexts or more complex resistance phenotypes (e.g., double resistance to both ATRA and ATO).

The antileukemic activity of PARPi may also be increased by their combination with agents used for APL treatment, such as anthracyclines and gemtuzumab ozogamicin. In fact, both agents are able to induce DNA damage, the repair of which can be hampered by inhibiting PARP1 activity (Yamauchi et al., 2014; Portwood et al., 2019). Previous reports have also suggested a potential role of PARPi in reducing the risk of cardiotoxicity associated with the use of anthracyclines based on the involvement of PARP1 overactivation in cardiomyocyte damage induced by these chemotherapeutic agents (Pacher et al., 2002; Ali et al., 2011). However, the protective effect of PARPi on anthracycline-induced cardiotoxicity is still debated (Damiani et al., 2018).

The favorable safety profile of PARPi, decitabine, azacitidine, and ascorbate encourages further investigation on their therapeutic potential as components of combination regimens for relapsed/ATO-refractory APL, especially in the case of frail patients who cannot tolerate the proarrhythmic effects of ATO or the adverse effects of more aggressive therapies.

Authorship Contributions

Participated in research design: Prete, Ciccarone, Voso, Graziani, Faraoni.

Conducted experiments: Giansanti, De Gabrieli, Prete, Ottone, Divona, Karimi, Ciccarone.

Performed data analysis: Giansanti, Ottone, Karimi.

Wrote or contributed to the writing of the manuscript: Voso, Graziani, Faraoni.

Footnotes

    • Received January 28, 2021.
    • Accepted March 29, 2021.
  • ↵1 G.G. and I.F. contributed equally to this work as co-last authors.

  • This work was supported by the Associazione Italiana per la Ricerca sul Cancro (AIRC) [IG 2017-ID. 20353 project-PI Grazia Graziani; AIRC 5 × 1000 call “Metastatic disease: the key unmet need in oncology to MYNERVA project; #21267 (MYeloid NEoplasms Research Venture AIRC) and PRIN Grant No. 2017WXR7ZT to M.T.V.].

  • The authors declare no financial conflict of interest.

  • This work was previously presented as two congress abstracts: Giansanti M, Faraoni I, Prete SP, Karimi T, Divona M, Ottone T, Consalvo MI, Voso MT, and Graziani G. XVI Congresso Nazionale SIES (Società Italiana di Ematologia Sperimentale). Postponed until a later date. Giansanti M, Faraoni I, Prete SP, Karimi T, Divona M, Ottone T, Consalvo MI, Voso MT, Graziani G (2020) Poly(ADP-ribose) polymerase inhibitors synergize with ascorbate and hypomethylating agents in arsenic trioxide-resistant promyelocytic leukaemia cells. Abstract Book, Proceedings of the XVI Congress of the Italian Society of Experimental Hematology (SIES); 2020 Oct 15–17; Napoli, Italy. Haematologica 105:S65, abstract n. C079. Available online: https://haematologica.org/article/view/haematol.2020.s2; and Giansanti M, Faraoni I, Prete SP, Karimi T, Divona M, Ottone T, Ciccarone F, Voso MT, and Graziani G. XIII SIF Seminar on Pharmacology for PhD Students, Fellows, Post Doc and Specialist Trainees, The 40th Congress of the Italian Pharmacological Society (SIF), Digital Edition; 2021 Mar 8–13. Giansanti M, Faraoni I, Prete SP, Karimi T, Divona M, Ottone T, Ciccarone F, Voso MT, and Graziani G (2021) Role of poly(ADP-ribose) polymerase inhibitors in combination with ascorbate and hypomethylating agents for acute promyelocytic leukaemia resistant to arsenic trioxide. Pharmadvances 3:174–175 DOI: 10.36118/pharmadvances.03.2021.01.

  • https://doi.org/10.1124/jpet.121.000537.

Abbreviations

AML
acute myeloid leukemia
APL
acute promyelocytic leukemia
ATO
arsenic trioxide
ATRA
all-trans retinoic acid
BER
base excision repair
CI
combination index
DCF
2,7-dichlorodihydrofluorescein
DHA
dehydroascorbate
DHE
dihydroethidium
Fa
fraction affected
GAPDH
glyceraldehyde 3-phosphate dehydrogenase
5hmC
5-hydroxymethylcytosine
MDS
myelodysplastic syndrome
PARP
poly(ADP-ribose) polymerase
PARPi
PARP inhibitors
PI
propidium iodide
PML
promyelocytic leukemia
RARA
retinoic acid receptor α
ROS
reactive oxygen species
TET
ten-eleven translocation
  • Copyright © 2021 by The American Society for Pharmacology and Experimental Therapeutics

References

  1. ↵
    1. Abbotts R
    2. Topper MJ
    3. Biondi C
    4. Fontaine D
    5. Goswami R
    6. Stojanovic L
    7. Choi EY
    8. McLaughlin L
    9. Kogan AA
    10. Xia L, et al.
    (2019) DNA methyltransferase inhibitors induce a BRCAness phenotype that sensitizes NSCLC to PARP inhibitor and ionizing radiation. Proc Natl Acad Sci U S A 116:22609–22618.
    OpenUrlCrossRefPubMed
  2. ↵
    1. Abdel-Wahab O
    2. Mullally A
    3. Hedvat C
    4. Garcia-Manero G
    5. Patel J
    6. Wadleigh M
    7. Malinge S
    8. Yao J
    9. Kilpivaara O
    10. Bhat R, et al.
    (2009) Genetic characterization of TET1, TET2, and TET3 alterations in myeloid malignancies. Blood 114:144–147.
    OpenUrlCrossRefPubMed
  3. ↵
    1. Agathocleous M
    2. Meacham CE
    3. Burgess RJ
    4. Piskounova E
    5. Zhao Z
    6. Crane GM
    7. Cowin BL
    8. Bruner E
    9. Murphy MM
    10. Chen W, et al.
    (2017) Ascorbate regulates haematopoietic stem cell function and leukaemogenesis. Nature 549:476–481.
    OpenUrlCrossRefPubMed
  4. ↵
    1. Alcalay M
    2. Meani N
    3. Gelmetti V
    4. Fantozzi A
    5. Fagioli M
    6. Orleth A
    7. Riganelli D
    8. Sebastiani C
    9. Cappelli E
    10. Casciari C, et al.
    (2003) Acute myeloid leukemia fusion proteins deregulate genes involved in stem cell maintenance and DNA repair. J Clin Invest 112:1751–1761.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Alex AA
    2. Chendamarai E
    3. Ganesan S
    4. Balasundaram N
    5. Palani HK
    6. David S
    7. Mathews V
    (2014) Arsenic trioxide resistance: more to it than mutations in PML-RARα. Blood 124:3605.
    OpenUrlCrossRefPubMed
  6. ↵
    1. Ali M
    2. Kamjoo M
    3. Thomas HD
    4. Kyle S
    5. Pavlovska I
    6. Babur M
    7. Telfer BA
    8. Curtin NJ
    9. Williams KJ
    (2011) The clinically active PARP inhibitor AG014699 ameliorates cardiotoxicity but does not enhance the efficacy of doxorubicin, despite improving tumor perfusion and radiation response in mice. Mol Cancer Ther 10:2320–2329. DOI: 10.1158/1535-7163.MCT-11-035.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Balasundaram N
    2. Ganesan S
    3. Palani HK
    4. Alex AA
    5. David S
    6. Korula A
    7. George B
    8. Chomienne C
    9. Balasubramanian P
    10. Mathews V
    (2016) Metabolic rewiring drives resistance to arsenic trioxide in acute promyelocytic leukemia. Blood 128:3956. DOI: 10.1182/blood.v128.22.3956.3956.
    OpenUrl
  8. ↵
    1. Cashen AF
    2. Shah AK
    3. Todt L
    4. Fisher N
    5. DiPersio J
    (2008) Pharmacokinetics of decitabine administered as a 3-h infusion to patients with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Cancer Chemother Pharmacol 61:759–766.
    OpenUrlCrossRefPubMed
  9. ↵
    1. Chen Q
    2. Espey MG
    3. Sun AY
    4. Lee J-H
    5. Krishna MC
    6. Shacter E
    7. Choyke PL
    8. Pooput C
    9. Kirk KL
    10. Buettner GR, et al.
    (2007) Ascorbate in pharmacologic concentrations selectively generates ascorbate radical and hydrogen peroxide in extracellular fluid in vivo. Proc Natl Acad Sci U S A 104:8749–8754.
    OpenUrlCrossRefPubMed
  10. ↵
    1. Chou T-C
    (2010) Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res 70:440–446.
    OpenUrlCrossRefPubMed
    1. Ciccarone F
    2. Castelli S
    3. Ioannilli L
    4. Ciriolo MR
    (2019) High dietary fat intake affects DNA methylation/hydroxymethylation in mouse heart: epigenetic hints for obesity-related cardiac dysfunction. Mol Nutr Food Res 63:e1800970.
    OpenUrlCrossRefPubMed
  11. ↵
    1. Cicconi L
    2. Platzbecker U
    3. Avvisati G
    4. Paoloni F
    5. Thiede C
    6. Vignetti M
    7. Fazi P
    8. Ferrara F
    9. Divona M
    10. Albano F, et al.
    (2020) Long-term results of all-trans retinoic acid and arsenic trioxide in non-high-risk acute promyelocytic leukemia: update of the APL0406 Italian-German randomized trial. Leukemia 34:914–918.
    OpenUrlCrossRefPubMed
  12. ↵
    1. Cimmino L
    2. Dolgalev I
    3. Wang Y
    4. Yoshimi A
    5. Martin GH
    6. Wang J
    7. Ng V
    8. Xia B
    9. Witkowski MT
    10. Mitchell-Flack M, et al.
    (2017) Restoration of TET2 function blocks aberrant self-renewal and leukemia progression. Cell 170:1079–1095.e20.
    OpenUrl
  13. ↵
    1. Damiani RM
    2. Moura DJ
    3. Viau CM
    4. Brito V
    5. Morás AM
    6. Henriques JAP
    7. Saffi J
    (2018) Influence of PARP-1 inhibition in the cardiotoxicity of the topoisomerase 2 inhibitors doxorubicin and mitoxantrone. Toxicol In Vitro 52:203–213.
    OpenUrlCrossRefPubMed
  14. ↵
    1. Das AB
    2. Kakadia PM
    3. Wojcik D
    4. Pemberton L
    5. Browett PJ
    6. Bohlander SK
    7. Vissers MCM
    (2019) Clinical remission following ascorbate treatment in a case of acute myeloid leukemia with mutations in TET2 and WT1. Blood Cancer J 9:82.
    OpenUrlCrossRefPubMed
  15. ↵
    1. de Bono J
    2. Ramanathan RK
    3. Mina L
    4. Chugh R
    5. Glaspy J
    6. Rafii S
    7. Kaye S
    8. Sachdev J
    9. Heymach J
    10. Smith DC, et al.
    (2017) Phase I, dose-escalation, two-part trial of the PARP inhibitor talazoparib in patients with advanced germline BRCA1/2 mutations and selected sporadic cancers. Cancer Discov 7:620–629.
    OpenUrlCrossRefPubMed
  16. ↵
    1. Delhommeau F
    2. Dupont S
    3. Della Valle V
    4. James C
    5. Trannoy S
    6. Massé A
    7. Kosmider O
    8. Le Couedic J-P
    9. Robert F
    10. Alberdi A, et al.
    (2009) Mutation in TET2 in myeloid cancers. N Engl J Med 360:2289–2301.
    OpenUrlCrossRefPubMed
  17. ↵
    1. Dickson KM
    2. Gustafson CB
    3. Young JI
    4. Züchner S
    5. Wang G
    (2013) Ascorbate-induced generation of 5-hydroxymethylcytosine is unaffected by varying levels of iron and 2-oxoglutarate. Biochem Biophys Res Commun 439:522–527.
    OpenUrl
  18. ↵
    1. Esposito MT
    2. Zhao L
    3. Fung TK
    4. Rane JK
    5. Wilson A
    6. Martin N
    7. Gil J
    8. Leung AY
    9. Ashworth A
    10. So CW
    (2015) Synthetic lethal targeting of oncogenic transcription factors in acute leukemia by PARP inhibitors. Nat Med 21:1481–1490.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Faraoni I
    2. Aloisio F
    3. De Gabrieli A
    4. Consalvo MI
    5. Lavorgna S
    6. Voso MT
    7. Lo-Coco F
    8. Graziani G
    (2018) The poly(ADP-ribose) polymerase inhibitor olaparib induces up-regulation of death receptors in primary acute myeloid leukemia blasts by NF-κB activation. Cancer Lett 423:127–138.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    1. Faraoni I
    2. Compagnone M
    3. Lavorgna S
    4. Angelini DF
    5. Cencioni MT
    6. Piras E
    7. Panetta P
    8. Ottone T
    9. Dolci S
    10. Venditti A, et al.
    (2015) BRCA1, PARP1 and γH2AX in acute myeloid leukemia: role as biomarkers of response to the PARP inhibitor olaparib. Biochim Biophys Acta 1852:462–472.
    OpenUrlAbstract
    1. Faraoni I
    2. Consalvo MI
    3. Aloisio F
    4. Fabiani E
    5. Giansanti M
    6. Di Cristino F
    7. Falconi G
    8. Tentori L
    9. Di Veroli A
    10. Curzi P, et al.
    (2019a) Cytotoxicity and differentiating effect of the poly(ADP-ribose) polymerase inhibitor olaparib in myelodysplastic syndromes. Cancers (Basel) 11:1373.
    OpenUrlPubMed
    1. Faraoni I
    2. Giansanti M
    3. Voso MT
    4. Lo-Coco F
    5. Graziani G
    (2019b) Targeting ADP-ribosylation by PARP inhibitors in acute myeloid leukaemia and related disorders. Biochem Pharmacol 167:133–148.
    OpenUrl
  21. ↵
    1. Faraoni I
    2. Graziani G
    (2018) Role of BRCA mutations in cancer treatment with poly(ADP-ribose) polymerase (PARP) inhibitors. Cancers (Basel) 10:487.
    OpenUrlCrossRefPubMed
  22. ↵
    1. Ferrada L
    2. Salazar K
    3. Nualart F
    (2019) Metabolic control by dehydroascorbic acid: questions and controversies in cancer cells. J Cell Physiol 234:19331–19338.
    OpenUrlCrossRefPubMed
  23. ↵
    1. Fong PC
    2. Boss DS
    3. Yap TA
    4. Tutt A
    5. Wu P
    6. Mergui-Roelvink M
    7. Mortimer P
    8. Swaisland H
    9. Lau A
    10. O’Connor MJ, et al.
    (2009) Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med 361:123–134.
    OpenUrlAbstract
  24. ↵
    1. Friedlander M
    2. Matulonis U
    3. Gourley C
    4. du Bois A
    5. Vergote I
    6. Rustin G
    7. Scott C
    8. Meier W
    9. Shapira-Frommer R
    10. Safra T, et al.
    (2018) Long-term efficacy, tolerability and overall survival in patients with platinum-sensitive, recurrent high-grade serous ovarian cancer treated with maintenance olaparib capsules following response to chemotherapy. Br J Cancer 119:1075–1085.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    1. Fritz C
    2. Portwood SM
    3. Przespolewski A
    4. Wang ES
    (2021) PARP goes the weasel! Emerging role of PARP inhibitors in acute leukemias. Blood Rev 45:100696.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Giansanti M
    2. Karimi T
    3. Faraoni I
    4. Graziani G
    (2021) High-dose vitamin C: preclinical evidence for tailoring treatment in cancer patients. Cancers (Basel) 13:1428.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    1. Gillberg L
    2. Ørskov AD
    3. Liu M
    4. Harsløf LBS
    5. Jones PA
    6. Grønbæk K
    (2018) Vitamin C - a new player in regulation of the cancer epigenome. Semin Cancer Biol 51:59–67.
    OpenUrlCrossRefPubMed
  28. ↵
    1. Goto E
    2. Tomita A
    3. Hayakawa F
    4. Atsumi A
    5. Kiyoi H
    6. Naoe T
    (2011) Missense mutations in PML-RARA are critical for the lack of responsiveness to arsenic trioxide treatment. Blood 118:1600–1609.
    OpenUrlCrossRefPubMed
  29. ↵
    1. Guan Y
    2. Greenberg EF
    3. Hasipek M
    4. Chen S
    5. Liu X
    6. Kerr CM
    7. Gackowski D
    8. Zarakowska E
    9. Radivoyevitch T
    10. Gu X, et al.
    (2020) Context dependent effects of ascorbic acid treatment in TET2 mutant myeloid neoplasia. Commun Biol 3:493.
    OpenUrlCrossRefPubMed
  30. ↵
    1. Gurnari C
    2. De Bellis E
    3. Divona M
    4. Ottone T
    5. Lavorgna S
    6. Voso MT
    (2019) When poisons cure: the case of arsenic in acute promyelocytic leukemia. Chemotherapy 64:238–247.
    OpenUrlCrossRefPubMed
  31. ↵
    1. Hoffer LJ
    2. Levine M
    3. Assouline S
    4. Melnychuk D
    5. Padayatty SJ
    6. Rosadiuk K
    7. Rousseau C
    8. Robitaille L
    9. Miller WH Jr..
    (2008) Phase I clinical trial of i.v. ascorbic acid in advanced malignancy. Ann Oncol 19:1969–1974.
    OpenUrlCrossRef
  32. ↵
    1. Iaccarino L
    2. Ottone T
    3. Alfonso V
    4. Cicconi L
    5. Divona M
    6. Lavorgna S
    7. Travaglini S
    8. Ferrantini A
    9. Falconi G
    10. Baer C, et al.
    (2019) Mutational landscape of patients with acute promyelocytic leukemia at diagnosis and relapse. Am J Hematol 94:1091–1097.
    OpenUrl
  33. ↵
    1. Iaccarino L
    2. Ottone T
    3. Divona M
    4. Cicconi L
    5. Cairoli R
    6. Voso MT
    7. Lo-Coco F
    (2016) Mutations affecting both the rearranged and the unrearranged PML alleles in refractory acute promyelocytic leukaemia. Br J Haematol 172:909–913.
    OpenUrlCrossRefPubMed
  34. ↵
    1. Keating GM
    (2012) Azacitidine: a review of its use in the management of myelodysplastic syndromes/acute myeloid leukaemia. Drugs 72:1111–1136.
    OpenUrlPubMed
  35. ↵
    1. Kharat SS
    2. Ding X
    3. Swaminathan D
    4. Suresh A
    5. Singh M
    6. Sengodan SK
    7. Burkett S
    8. Marks H
    9. Pamala C
    10. He Y, et al.
    (2020) Degradation of 5hmC-marked stalled replication forks by APE1 causes genomic instability. Sci Signal 13:eaba8091.
    OpenUrlCrossRefPubMed
  36. ↵
    1. Kim T-J
    2. Byun J-S
    3. Kwon HS
    4. Kim D-Y
    (2018) Cellular toxicity driven by high-dose vitamin C on normal and cancer stem cells. Biochem Biophys Res Commun 497:347–353.
    OpenUrlAbstract
  37. ↵
    1. Kohl V
    2. Flach J
    3. Naumann N
    4. Brendel S
    5. Kleiner H
    6. Weiss C
    7. Seifarth W
    8. Nowak D
    9. Hofmann WK
    10. Fabarius A, et al.
    (2019) Antileukemic efficacy in vitro of talazoparib and APE1 inhibitor III combined with decitabine in myeloid malignancies. Cancers (Basel) 11:1493.
    OpenUrlCrossRefPubMed
  38. ↵
    1. Kristeleit R
    2. Shapiro GI
    3. Burris HA
    4. Oza AM
    5. LoRusso P
    6. Patel MR
    7. Domchek SM
    8. Balmaña J
    9. Drew Y
    10. Chen LM, et al.
    (2017) A phase I-II study of the oral PARP inhibitor rucaparib in patients with germline BRCA1/2-mutated ovarian carcinoma or other solid tumors. Clin Cancer Res 23:4095–4106.
    OpenUrl
  39. ↵
    1. Kummar S
    2. Kinders R
    3. Gutierrez ME
    4. Rubinstein L
    5. Parchment RE
    6. Phillips LR
    7. Ji J
    8. Monks A
    9. Low JA
    10. Chen A, et al.
    (2009) Phase 0 clinical trial of the poly (ADP-ribose) polymerase inhibitor ABT-888 in patients with advanced malignancies. J Clin Oncol 27:2705–2711.
    OpenUrlCrossRefPubMed
  40. ↵
    1. Lanotte M
    2. Martin-Thouvenin V
    3. Najman S
    4. Balerini P
    5. Valensi F
    6. Berger R
    (1991) NB4, a maturation inducible cell line with t(15;17) marker isolated from a human acute promyelocytic leukemia (M3). Blood 77:1080–1086.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Lawlor D
    2. Martin P
    3. Busschots S
    4. Thery J
    5. O’Leary JJ
    6. Hennessy BT
    7. Stordal B
    (2014) PARP inhibitors as P-glyoprotein substrates. J Pharm Sci 103:1913–1920.
    OpenUrlCrossRefPubMed
  42. ↵
    1. Ledermann J
    2. Harter P
    3. Gourley C
    4. Friedlander M
    5. Vergote I
    6. Rustin G
    7. Scott CL
    8. Meier W
    9. Shapira-Frommer R
    10. Safra T, et al.
    (2014) Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol 15:852–861.
    OpenUrl
  43. ↵
    1. Lo-Coco F
    2. Avvisati G
    3. Vignetti M
    4. Thiede C
    5. Orlando SM
    6. Iacobelli S
    7. Ferrara F
    8. Fazi P
    9. Cicconi L
    10. Di Bona E, et al
    ; Gruppo Italiano Malattie Ematologiche dell’Adulto; German-Austrian Acute Myeloid Leukemia Study Group; Study Alliance Leukemia (2013) Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N Engl J Med 369:111–121.
    OpenUrl
  44. ↵
    1. Madan V
    2. Shyamsunder P
    3. Han L
    4. Mayakonda A
    5. Nagata Y
    6. Sundaresan J
    7. Kanojia D
    8. Yoshida K
    9. Ganesan S
    10. Hattori N, et al.
    (2016) Comprehensive mutational analysis of primary and relapse acute promyelocytic leukemia. Leukemia 30:1672–1681.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    1. Maes K
    2. De Smedt E
    3. Lemaire M
    4. De Raeve H
    5. Menu E
    6. Van Valckenborgh E
    7. McClue S
    8. Vanderkerken K
    9. De Bruyne E
    (2014) The role of DNA damage and repair in decitabine-mediated apoptosis in multiple myeloma. Oncotarget 5:3115–3129.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    1. Mastrangelo D
    2. Massai L
    3. Lo Coco F
    4. Noguera NI
    5. Borgia L
    6. Fioritoni G
    7. Berardi A
    8. Iacone A
    9. Muscettola M
    10. Pelosi E, et al.
    (2015) Cytotoxic effects of high concentrations of sodium ascorbate on human myeloid cell lines. Ann Hematol 94:1807–1816.
    OpenUrl
  47. ↵
    1. Mateo J
    2. Moreno V
    3. Gupta A
    4. Kaye SB
    5. Dean E
    6. Middleton MR
    7. Friedlander M
    8. Gourley C
    9. Plummer R
    10. Rustin G, et al.
    (2016) An adaptive study to determine the optimal dose of the tablet formulation of the PARP inhibitor olaparib. Target Oncol 11:401–415.
    OpenUrl
  48. ↵
    1. Miller WH Jr.
    2. Schipper HM
    3. Lee JS
    4. Singer J
    5. Waxman S
    (2002) Mechanisms of action of arsenic trioxide. Cancer Res 62:3893–3903.
    OpenUrl
  49. ↵
    1. Minor EA
    2. Court BL
    3. Young JI
    4. Wang G
    (2013) Ascorbate induces ten-eleven translocation (Tet) methylcytosine dioxygenase-mediated generation of 5-hydroxymethylcytosine. J Biol Chem 288:13669–13674.
    OpenUrlCrossRef
  50. ↵
    1. Murai J
    2. Huang S-YN
    3. Renaud A
    4. Zhang Y
    5. Ji J
    6. Takeda S
    7. Morris J
    8. Teicher B
    9. Doroshow JH
    10. Pommier Y
    (2014) Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib. Mol Cancer Ther 13:433–443.
    OpenUrl
  51. ↵
    1. Murai J
    2. Huang SY
    3. Das BB
    4. Renaud A
    5. Zhang Y
    6. Doroshow JH
    7. Ji J
    8. Takeda S
    9. Pommier Y
    (2012) Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res 72:5588–5599.
    OpenUrl
  52. ↵
    1. Curtin NJ
    2. Sharma RA
    1. Murai J
    2. Pommier Y
    (2015) Classification of PARP inhibitors based on PARP trapping and catalytic inhibition, and rationale for combinations with topoisomerase I inhibitors and alkylating agents, in PARP Inhibitors for Cancer Therapy (Curtin NJ Sharma RA eds) pp 261–274, Humana Press, Cham, Switzerland.
  53. ↵
    1. Muvarak NE
    2. Chowdhury K
    3. Xia L
    4. Robert C
    5. Choi EY
    6. Cai Y
    7. Bellani M
    8. Zou Y
    9. Singh ZN
    10. Duong VH, et al.
    (2016) Enhancing the cytotoxic effects of PARP inhibitors with DNA demethylating agents - a potential therapy for cancer. Cancer Cell 30:637–650.
    OpenUrlCrossRef
  54. ↵
    1. Ngo B
    2. Van Riper JM
    3. Cantley LC
    4. Yun J
    (2019) Targeting cancer vulnerabilities with high-dose vitamin C. Nat Rev Cancer 19:271–282.
    OpenUrl
  55. ↵
    1. Nieborowska-Skorska M
    2. Maifrede S
    3. Dasgupta Y
    4. Sullivan K
    5. Flis S
    6. Le BV
    7. Solecka M
    8. Belyaeva EA
    9. Kubovcakova L
    10. Nawrocki M, et al.
    (2017) Ruxolitinib-induced defects in DNA repair cause sensitivity to PARP inhibitors in myeloproliferative neoplasms. Blood 130:2848–2859.
    OpenUrlAbstract/FREE Full Text
  56. ↵
    1. Nishikawa T
    2. Matsumoto K
    3. Tamura K
    4. Yoshida H
    5. Imai Y
    6. Miyasaka A
    7. Onoe T
    8. Yamaguchi S
    9. Shimizu C
    10. Yonemori K, et al.
    (2017) Phase 1 dose-escalation study of single-agent veliparib in Japanese patients with advanced solid tumors. Cancer Sci 108:1834–1842.
    OpenUrl
  57. ↵
    1. Noguera NI
    2. Catalano G
    3. Banella C
    4. Divona M
    5. Faraoni I
    6. Ottone T
    7. Arcese W
    8. Voso MT
    (2019) Acute promyelocytic leukemia: update on the mechanisms of leukemogenesis, resistance and on innovative treatment strategies. Cancers (Basel) 11:1591.
    OpenUrl
  58. ↵
    1. Noguera NI
    2. Pelosi E
    3. Angelini DF
    4. Piredda ML
    5. Guerrera G
    6. Piras E
    7. Battistini L
    8. Massai L
    9. Berardi A
    10. Catalano G, et al.
    (2017) High-dose ascorbate and arsenic trioxide selectively kill acute myeloid leukemia and acute promyelocytic leukemia blasts in vitro. Oncotarget 8:32550–32565.
    OpenUrl
  59. ↵
    1. Orta ML
    2. Höglund A
    3. Calderón-Montaño JM
    4. Domínguez I
    5. Burgos-Morón E
    6. Visnes T
    7. Pastor N
    8. Ström C
    9. López-lázaro M
    10. Helleday T
    (2014) The PARP inhibitor Olaparib disrupts base excision repair of 5-aza-2′-deoxycytidine lesions. Nucleic Acids Res 42:9108–9120.
    OpenUrlCrossRefPubMed
  60. ↵
    1. Pacher P
    2. Liaudet L
    3. Bai P
    4. Virag L
    5. Mabley JG
    6. Haskó G
    7. Szabó C
    (2002) Activation of poly(ADP-ribose) polymerase contributes to development of doxorubicin-induced heart failure. J Pharmacol Exp Ther 300:862–867.
    OpenUrlAbstract/FREE Full Text
  61. ↵
    1. Pastor WA
    2. Aravind L
    3. Rao A
    (2013) TETonic shift: biological roles of TET proteins in DNA demethylation and transcription. Nat Rev Mol Cell Biol 14:341–356.
    OpenUrlCrossRefPubMed
  62. ↵
    1. Patel K
    2. Dickson J
    3. Din S
    4. Macleod K
    5. Jodrell D
    6. Ramsahoye B
    (2010) Targeting of 5-aza-2′-deoxycytidine residues by chromatin-associated DNMT1 induces proteasomal degradation of the free enzyme. Nucleic Acids Res 38:4313–4324.
    OpenUrlCrossRefPubMed
  63. ↵
    1. Portwood SM
    2. Cantella MC
    3. Cronin TL
    4. Wang ES
    (2019) Addition of the PARP inhibitor, talazoparib, to gemtuzumab ozogamicin significantly enhances anti-leukemic activity in human CD33+ acute myeloid leukemia. Blood 134 (Suppl 1):1371.
    OpenUrl
  64. ↵
    1. Pulliam N
    2. Fang F
    3. Ozes AR
    4. Tang J
    5. Adewuyi A
    6. Keer H
    7. Lyons J
    8. Baylin SB
    9. Matei D
    10. Nakshatri H, et al.
    (2018) An effective epigenetic-PARP inhibitor combination therapy for breast and ovarian cancers independent of BRCA mutations. Clin Cancer Res 24:3163–3175.
    OpenUrlAbstract/FREE Full Text
  65. ↵
    1. Sandhu SK
    2. Schelman WR
    3. Wilding G
    4. Moreno V
    5. Baird RD
    6. Miranda S
    7. Hylands L
    8. Riisnaes R
    9. Forster M
    10. Omlin A, et al.
    (2013) The poly(ADP-ribose) polymerase inhibitor niraparib (MK4827) in BRCA mutation carriers and patients with sporadic cancer: a phase 1 dose-escalation trial. Lancet Oncol 14:882–892.
    OpenUrlCrossRefPubMed
  66. ↵
    1. Santi DV
    2. Norment A
    3. Garrett CE
    (1984) Covalent bond formation between a DNA-cytosine methyltransferase and DNA containing 5-azacytosine. Proc Natl Acad Sci USA 81:6993–6997.
    OpenUrlAbstract/FREE Full Text
  67. ↵
    1. Sanz MA
    2. Fenaux P
    3. Tallman MS
    4. Estey EH
    5. Löwenberg B
    6. Naoe T
    7. Lengfelder E
    8. Döhner H
    9. Burnett AK
    10. Chen S-J, et al.
    (2019) Management of acute promyelocytic leukemia: updated recommendations from an expert panel of the European LeukemiaNet. Blood 133:1630–1643.
    OpenUrlAbstract/FREE Full Text
  68. ↵
    1. Sanz MA
    2. Grimwade D
    3. Tallman MS
    4. Lowenberg B
    5. Fenaux P
    6. Estey EH
    7. Naoe T
    8. Lengfelder E
    9. Büchner T
    10. Döhner H, et al.
    (2009) Management of acute promyelocytic leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet. Blood 113:1875–1891.
    OpenUrlAbstract/FREE Full Text
  69. ↵
    1. Schneider CA
    2. Rasband WS
    3. Eliceiri KW
    (2012) NIH Image to Image: 25 years of image analysis. Nature Methods 9:671–675.
    OpenUrl
  70. ↵
    1. Sertel S
    2. Tome M
    3. Briehl MM
    4. Bauer J
    5. Hock K
    6. Plinkert PK
    7. Efferth T
    (2012) Factors determining sensitivity and resistance of tumor cells to arsenic trioxide. PLoS One 7:e35584.
    OpenUrlPubMed
  71. ↵
    1. Shapiro GI
    2. Kristeleit RS
    3. Burris HA
    4. LoRusso P
    5. Patel MR
    6. Drew Y
    7. Giordano H
    8. Maloney L
    9. Watkins S
    10. Goble S, et al.
    (2019) Pharmacokinetic study of rucaparib in patients with advanced solid tumors. Clin Pharmacol Drug Dev 8:107–118.
    OpenUrl
  72. ↵
    1. Spoelstra-de Man AME
    2. Elbers PWG
    3. Oudemans-van Straaten HM
    (2018) Making sense of early high-dose intravenous vitamin C in ischemia/reperfusion injury. Crit Care 22:70.
    OpenUrl
  73. ↵
    1. Takeshita A
    2. Shinjo K
    3. Naito K
    4. Matsui H
    5. Shigeno K
    6. Nakamura S
    7. Horii T
    8. Maekawa M
    9. Kitamura K
    10. Naoe T, et al.
    (2003) P-glycoprotein (P-gp) and multidrug resistance-associated protein 1 (MRP1) are induced by arsenic trioxide (As(2)O(3)), but are not the main mechanism of As(2)O(3)-resistance in acute promyelocytic leukemia cells. Leukemia 17:648–650.
    OpenUrlPubMed
  74. ↵
    1. van Dongen JJ
    2. Macintyre EA
    3. Gabert JA
    4. Delabesse E
    5. Rossi V
    6. Saglio G
    7. Gottardi E
    8. Rambaldi A
    9. Dotti G
    10. Griesinger F, et al.
    (1999) Standardized RT-PCR analysis of fusion gene transcripts from chromosome aberrations in acute leukemia for detection of minimal residual disease. Report of the BIOMED-1 Concerted Action: investigation of minimal residual disease in acute leukemia. Leukemia 13:1901–1928.
    OpenUrlCrossRefPubMed
  75. ↵
    1. Wu X
    2. Zhang Y
    (2017) TET-mediated active DNA demethylation: mechanism, function and beyond. Nat Rev Genet 18:517–534.
    OpenUrlCrossRefPubMed
  76. ↵
    1. Yamauchi T
    2. Uzui K
    3. Nishi R
    4. Shigemi H
    5. Ueda T
    (2014) Gemtuzumab ozogamicin and olaparib exert synergistic cytotoxicity in CD33-positive HL-60 myeloid leukemia cells. Anticancer Res 34:5487–5494.
    OpenUrlAbstract/FREE Full Text
  77. ↵
    1. Yin R
    2. Mao S-Q
    3. Zhao B
    4. Chong Z
    5. Yang Y
    6. Zhao C
    7. Zhang D
    8. Huang H
    9. Gao J
    10. Li Z, et al.
    (2013) Ascorbic acid enhances Tet-mediated 5-methylcytosine oxidation and promotes DNA demethylation in mammals. J Am Chem Soc 135:10396–10403.
    OpenUrlCrossRefPubMed
  78. ↵
    1. Zhao H
    2. Zhu H
    3. Huang J
    4. Zhu Y
    5. Hong M
    6. Zhu H
    7. Zhang J
    8. Li S
    9. Yang L
    10. Lian Y, et al.
    (2018) The synergy of Vitamin C with decitabine activates TET2 in leukemic cells and significantly improves overall survival in elderly patients with acute myeloid leukemia. Leuk Res 66:1–7.
    OpenUrlCrossRef
  79. ↵
    1. Zhao L
    2. So CWE
    (2017) PARPi potentiates with current conventional therapy in MLL leukemia. Cell Cycle 16:1861–1869.
    OpenUrl
  80. ↵
    1. Zhu H-H
    2. Qin Y-Z
    3. Huang X-J
    (2014) Resistance to arsenic therapy in acute promyelocytic leukemia. N Engl J Med 370:1864–1866.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Journal of Pharmacology and Experimental Therapeutics: 377 (3)
Journal of Pharmacology and Experimental Therapeutics
Vol. 377, Issue 3
1 Jun 2021
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Journal of Pharmacology and Experimental Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Poly(ADP-Ribose) Polymerase Inhibitors for Arsenic Trioxide–Resistant Acute Promyelocytic Leukemia: Synergistic In Vitro Antitumor Effects with Hypomethylating Agents or High-Dose Vitamin C
(Your Name) has forwarded a page to you from Journal of Pharmacology and Experimental Therapeutics
(Your Name) thought you would be interested in this article in Journal of Pharmacology and Experimental Therapeutics.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Research ArticleCellular and Molecular

PARPi with Decitabine, Azacitidine, or Vitamin C for APL

Manuela Giansanti, Antonio De Gabrieli, Salvatore Pasquale Prete, Tiziana Ottone, Maria Domenica Divona, Terry Karimi, Fabio Ciccarone, Maria Teresa Voso, Grazia Graziani and Isabella Faraoni
Journal of Pharmacology and Experimental Therapeutics June 1, 2021, 377 (3) 385-397; DOI: https://doi.org/10.1124/jpet.121.000537

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Research ArticleCellular and Molecular

PARPi with Decitabine, Azacitidine, or Vitamin C for APL

Manuela Giansanti, Antonio De Gabrieli, Salvatore Pasquale Prete, Tiziana Ottone, Maria Domenica Divona, Terry Karimi, Fabio Ciccarone, Maria Teresa Voso, Grazia Graziani and Isabella Faraoni
Journal of Pharmacology and Experimental Therapeutics June 1, 2021, 377 (3) 385-397; DOI: https://doi.org/10.1124/jpet.121.000537
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Authorship Contributions
    • Footnotes
    • Abbreviations
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • CsA Downregulates Selenop Expression via a STAT3-FoxO1 Axis
  • Anisodamine Ameliorates Acute Lung Injury
  • ACE2 Inhibits LPS-Caused Lung Fibrosis
Show more Cellular and Molecular

Similar Articles

Advertisement
  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About JPET
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Drug Metabolism and Disposition
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-0103 (Online)

Copyright © 2022 by the American Society for Pharmacology and Experimental Therapeutics