IDH1-mutant gliomas are dependent upon the canonical coenzyme NAD+ for survival. It is known that PARP activation consumes NAD+ during base excision repair (BER) of chemotherapy-induced DNA damage. We therefore hypothesized that a strategy combining NAD+ biosynthesis inhibitors with the alkylating chemotherapeutic agent temozolomide could potentiate NAD+ depletion–mediated cytotoxicity in mutant IDH1 cancer cells. To investigate the impact of temozolomide on NAD+ metabolism, patient-derived xenografts and engineered mutant IDH1-expressing cell lines were exposed to temozolomide, in vitro and in vivo, both alone and in combination with nicotinamide phosphoribosyltransferase (NAMPT) inhibitors, which block NAD+ biosynthesis. The acute time period (<3 hours) after temozolomide treatment displayed a burst of NAD+ consumption driven by PARP activation. In IDH1-mutant–expressing cells, this consumption reduced further the abnormally lowered basal steady-state levels of NAD+, introducing a window of hypervulnerability to NAD+ biosynthesis inhibitors. This effect was selective for IDH1-mutant cells and independent of methylguanine methyltransferase or mismatch repair status, which are known rate-limiting mediators of adjuvant temozolomide genotoxic sensitivity. Combined temozolomide and NAMPT inhibition in an in vivo IDH1-mutant cancer model exhibited enhanced efficacy compared with each agent alone. Thus, we find IDH1-mutant cancers have distinct metabolic stress responses to chemotherapy-induced DNA damage and that combination regimens targeting nonredundant NAD+ pathways yield potent anticancer efficacy in vivo. Such targeting of convergent metabolic pathways in genetically selected cancers could minimize treatment toxicity and improve durability of response to therapy. Cancer Res; 77(15); 4102–15. ©2017 AACR.

Somatic mutations in the isocitrate dehydrogenase genes IDH1/2 define a class of adult diffuse gliomas with a distinct etiology and natural history (1–6). Molecular correlative analyses of international randomized trial cohorts have suggested that patients with IDH-mutant glioma, including both those with and without chromosome 1p/19q codeletion, gain a survival benefit from treatment with DNA-alkylating chemotherapy (7, 8). As a result of this emerging evidence, chemotherapy is now frequently integrated into the treatment regimen of these patients, even though they typically present with lower grade histology when compared with IDH wild-type gliomas. The oral alkylating agent temozolomide is commonly utilized by clinicians for this treatment, due to its tolerability in the adjuvant setting.

Unfortunately, the vast majority of these cancers still recur after adjuvant or salvage temozolomide treatment. The activities of the O-6 methylguanine DNA methyltransferase (MGMT) repair enzyme (9) and the mismatch repair (MMR) pathway (10) are critical mechanistic determinants of temozolomide-induced cancer cell cytotoxicity (11) and subsequent evasion and resistance to therapy (12–14). Salvage therapeutic strategies for post-temozolomide glioma recurrences are complicated by acquired mutations inactivating the MMR pathway, with the resulting alkylator-induced hypermutation driving a treatment-resistant malignant phenotype (15, 16). Improved chemotherapeutic strategies are needed to secure durable clinical responses in patients with IDH-mutant gliomas.

In addition to gliomas, mutations in IDH1 are found in a diverse spectrum of histopathologic tumor types, including leukemia, chondrosarcoma, cholangiocarcinoma, and a minor fraction of melanomas and breast cancers. Across each of these cancer types, IDH1 mutation is typically found in different background genetic contexts. As a common feature, however, mutant IDH1 drives widespread metabolic alterations in cancer cells (17). These include the production of 2-hydroxyglutarate (2HG; ref. 18), modulation of HIF1α (19), pyruvate dehydrogenase (20), and lactate dehydrogenase (21), as well as altered citric acid cycle flux (22), and depleted steady-state pools of several canonical metabolites, including glutathione (23) and NAD+ (24). This altered baseline metabolism results in the exposure of distinct enzymatic targets, including glutaminase (25) and the NAD+ biosynthetic enzyme nicotinamide phosphoribosyltransferase (NAMPT; ref. 24), to selective inhibition with small molecules, resulting in genotype-specific metabolic vulnerabilities in IDH-mutant cancer cells.

We hypothesized that study of the metabolic consequences of temozolomide exposure in IDH1-mutant cancers could uncover novel opportunities for therapeutic targeting. Despite the important role of O6-methylguanine adducts in mediating adjuvant temozolomide sensitivity, the majority (>80%) of temozolomide-induced DNA lesions are actually N3-methyladenine and N7-methylguanine adducts. These lesions are rapidly processed by the base excision repair (BER) machinery (26), as opposed to the O6-methylguanine–dependent MGMT and MMR systems. Importantly, the dynamic capacity of BER does not become saturated with these lesions (27), which is why they are not rate-limiting determinants of cytotoxicity in adjuvant temozolomide-treated cancers. Their abundance nevertheless does induce a significant stress response, through PARP, which polymerizes NAD+ into poly(ADP-ribose) (PAR) as the molecular repair signal activating recruitment of downstream BER proteins. Recognizing this activated PARP pathway, alongside the sirtuin (SIRT) pathway, is a primary mediator of NAD+ consumption in cells (28), we assessed whether chemotherapeutic targeting of these nonredundant NAD+ pathways could be exploited in IDH1-mutant cancer cells.

In experiments we describe here, we observed a burst of NAD+ consumption associated with PARP activation during the initial time period immediately following temozolomide treatment. In IDH1-mutant cancer cells, this consumption resulted in a transient but critical reduction of the already abnormally lowered basal steady-state levels of NAD+, introducing a window of hypervulnerability to NAD+ biosynthesis inhibitors. This finding provided a rationale for the therapeutic combination of temozolomide and NAMPT inhibitors, which resulted in improved efficacy when compared with their administration as single agents in an in vivo IDH1-mutant cancer model.

Creation of glioma tumorsphere lines

Under IRB-approved protocols, the patient-derived glioma lines used in this study (MGG18, MGG23, MGG85, MGG91, MGG119, MGG152, and MGG171) were obtained from 2008 to 2014 and were cultured in serum-free neural stem cell medium as described previously (29–31). BT142 (IDH1R132H-mutant anaplastic oligoastrocytoma) line was obtained from ATCC in 2014 and was not further authenticated. UACC257 line (IDH1/2 wild-type melanoma), HT1080 (IDH1R132C), and U87 (IDH1 wild type) lines were authenticated in 2017 by comparison of STR profiles to the ATCC public dataset. They were cryopreserved at passage number 3 or less prior to use for in vitro experiments. Normal human astrocytes (NHA) were obtained from ScienCell in 2014 and cultured in Astrocyte Medium (ScienCell) and were not further authenticated. All standard cell line media were supplemented with 10% FBS and penicillin–streptomycin–amphotericin B.

IDH1 genotyping and MGMT promoter methylation analysis

IDH1 genomic DNA PCR products (Platinum Taq polymerase) spanning coding exons were Sanger sequenced (Beckman Coulter Genomics). To assess MGMT promoter methylation status, methylation-specific PCR on genomic and bisulfite-modified DNA (Qiagen DNeasy Blood & Tissue Kit and EpiTect Bisulfite Kit) was performed in a two-step approach as described previously (32).

IDH1-R132H cell line generation

To generate IDH1R132H overexpressing lines, UACC257 cells were transduced with IDH1R132H lentivirus (ViraPower HiPerform T-Rex Gateway Expression System, Invitrogen), with the details of generation in Supplementary Methods. MGG18-IDH1-R132H cells have been described previously (24). After incubation with tetracycline (1 μg/mL) for 2 months, cells were used for experiments.

shRNA and control shRNA cell line generation

To knockdown MSH6, MGG152 or HT1080 cells were infected with lentivirus containing human MSH6 shRNA (#1, TRCN0000286578, Sigma Aldrich, #2 V3LHS_318784, GE Dharmacon), with the details of generation described in the Supplementary Methods. To knock down NAPRT1, UACC257 cells were infected with GIPZ Lentiviral Human NAPRT shRNA (V3LHS_359032, GE Dharmacon). GIPZ nonsilencing lentiviral shRNA control (RHS4348, GE Dharmacon) was used as a matched control.

Cell viability, cytotoxicity, PARP activity, and apoptosis analyses

To assess cell viability, under treatment conditions, CellTiter-Glo (Promega) assays were performed at the indicated time points, and the IC50 values were determined. PARP activity was quantified by HT Colorimetric PARP/Apoptosis Assay Kit (Trevigen) according to the manufacturer's recommendations. To evaluate caspase-3/7 activities, 7,000 to 8,000 cells were treated with DMSO or NAMPT inhibitors (12.5 nmol/L) and/or temozolomide (200 μmol/L), or Z-VAD-FMK (50 μmol/L) for 6 or 24 hours and were tested by Caspase-Glo 3/7 Assay (Promega) according to the manufacturer's recommendations. Details are described in Supplementary Methods.

NAD+ quantitation

To evaluate qualitative values of NAD+, NADH, NADP+, and NADPH, NAD/NADH-Glo Assay and NADP/NADPH-Glo Assay (Promega) were used according to the manufacturer's recommendations. To measure NAD+ quantitatively, NAD+/NADH Quantification Colorimetric Kit (BioVision Incorporated) was used according to the manufacturer's recommendations. Protein concentrations were measured and used for normalization. NADt (NADH and NAD) and NADH signals were measured at OD 450 nm (BIOTEK). NAD+ concentrations were calculated by subtracting NADH from NADt. Data were expressed as pmol/1 × 106 cells or pmol/mg protein.

Animal studies

All mouse experiments were approved by the Institutional Animal Care and Use Committee at Massachusetts General Hospital (Boston, MA). HT1080 cells (2.5 × 106) were subcutaneously implanted into the right flank of 7- to 10-week-old female SCID mice. When maximum tumor diameter reached 5 mm, cohorts were randomized to vehicle (oral gavage, n = 6), FK866 (15 mg/kg, i.p, n = 6), temozolomide (50 mg/kg, oral gavage, n = 6), or FK866 (15 mg/kg, i.p) plus temozolomide (50 mg/kg, oral gavage, n = 6). Distilled water with 20% Captisol (Cydex) and 5% dextrose (Sigma-Aldrich) was used as vehicle control. Treatment was given four times a week for 2 weeks. Tumor diameters were measured three times a week using a digital caliper. Calculated volume (mm3) = length (mm) × width (mm)2 × 0.5. Tumor volume was normalized at day 0 and expressed as percent change.

IHC

Tumor tissue sections were incubated with anti-Ki-67 (1:125, Wako), LC3A/B (1:100, Cell Signaling Technology), cleaved-PARP (1:100, Cell Signaling Technology), or cleaved-caspase-3 (1:100, Cell Signaling Technology) primary antibody.

Statistical analyses

All of the experiments were performed in three replicates per condition. For parametric analyses, two-tailed Student t tests or one-way ANOVA were used, and for analysis of frequencies of nominal data, two-tailed Fisher exact test was used, without Bonferroni correction. Data were expressed as mean ± SE. P values less than 0.05 were considered statistically significant.

Temozolomide induces NAD+ consumption via PARP activation

The PARP-activated BER pathway consumes NAD+ during repair of the DNA lesions induced by temozolomide exposure. Because of decreased NAD+ biosynthesis, IDH1-mutant cancer cells contain a lower baseline NAD+ level compared with IDH wild-type cancer cells. We therefore investigated the effect of temozolomide on NAD+ levels in mutant IDH1 cells. We initially tested the cytotoxic and PARP-inducing effect of temozolomide in a panel of endogenous IDH1-mutant and wild-type glioma tumorspheres (Fig. 1A; Supplementary Fig. S1A; Supplementary Table S1).

Figure 1.

Temozolomide (TMZ)-induced PARP activation deregulates NAD+ metabolism. A, CellTiter-Glo cell viability assay after 6-day temozolomide treatment of GBM tumorsphere lines, HT1080, and NHA. Left, IDH1-mutant lines; right, IDH1/2 wild-type lines with temozolomide at indicated doses compared with DMSO control. B, Relative cell viability of MGG152 cells treated with 200 μmol/L of temozolomide compared with DMSO control at the indicated time. C, Left, PARP activity after temozolomide treatment (200 μmol/L) at indicated time; right, Western blot analysis of PAR expression in temozolomide (200 μmol/L)-treated MGG152 cells at different time points. Actin, loading control. D, Left, PARP activity after 1-hour treatment with temozolomide at indicated dose; right, Western blot analysis of PAR expression in MGG152 cells with temozolomide at indicated dose for 1 hour. Actin, loading control. E, Relative NAD+ levels in MGG152 cells with temozolomide treatment at indicated dose for 3 hours. *, P < 0.05 for difference between DMSO versus temozolomide. F, PARP activity (left) and PAR expression (right, Western blot analysis) in MGG152 cells treated with DMSO, temozolomide (200 μmol/L), olaparib (5 μmol/L), or temozolomide plus olaparib for 1 hour. *, P < 0.05 for difference from DMSO. Actin, loading control. G, Relative NAD+ levels in MGG152 cells with olaparib (5 μmol/L), temozolomide (200 μmol/L), or temozolomide plus olaparib for 3 hours. *, P < 0.05 for difference between DMSO versus temozolomide. **, P < 0.05 for difference between temozolomide and temozolomide plus olaparib. Scale bars, SE.

Figure 1.

Temozolomide (TMZ)-induced PARP activation deregulates NAD+ metabolism. A, CellTiter-Glo cell viability assay after 6-day temozolomide treatment of GBM tumorsphere lines, HT1080, and NHA. Left, IDH1-mutant lines; right, IDH1/2 wild-type lines with temozolomide at indicated doses compared with DMSO control. B, Relative cell viability of MGG152 cells treated with 200 μmol/L of temozolomide compared with DMSO control at the indicated time. C, Left, PARP activity after temozolomide treatment (200 μmol/L) at indicated time; right, Western blot analysis of PAR expression in temozolomide (200 μmol/L)-treated MGG152 cells at different time points. Actin, loading control. D, Left, PARP activity after 1-hour treatment with temozolomide at indicated dose; right, Western blot analysis of PAR expression in MGG152 cells with temozolomide at indicated dose for 1 hour. Actin, loading control. E, Relative NAD+ levels in MGG152 cells with temozolomide treatment at indicated dose for 3 hours. *, P < 0.05 for difference between DMSO versus temozolomide. F, PARP activity (left) and PAR expression (right, Western blot analysis) in MGG152 cells treated with DMSO, temozolomide (200 μmol/L), olaparib (5 μmol/L), or temozolomide plus olaparib for 1 hour. *, P < 0.05 for difference from DMSO. Actin, loading control. G, Relative NAD+ levels in MGG152 cells with olaparib (5 μmol/L), temozolomide (200 μmol/L), or temozolomide plus olaparib for 3 hours. *, P < 0.05 for difference between DMSO versus temozolomide. **, P < 0.05 for difference between temozolomide and temozolomide plus olaparib. Scale bars, SE.

Close modal

Overall, sensitivity to temozolomide monotherapy varied across the panel, irrespective of IDH1 mutation or MGMT promoter methylation status (Supplementary Table S1). In MGG152 cells (IDH1 mutant, MGMT promoter methylated), temozolomide cytotoxicity was observed after day 4 (Fig. 1B). We observed PARP activation after temozolomide treatment, resulting in a marked induction of PAR in IDH1-mutant cancers in a time- and dose-dependent manner (Fig. 1C and D; Supplementary Fig. S1B). Notably, PARylation was induced rapidly, within 1 hour, and significantly decreased thereafter, consistent with its role in BER signaling response. Accordingly, temozolomide decreased NAD+ levels in a dose-dependent manner in multiple IDH1-mutant cell lines (Fig.1E; Supplementary Fig. S1C).

To investigate whether this effect was PARP specific, we repeated these experiments with the addition of the PARP inhibitor olaparib. Olaparib significantly suppressed PARP activation and PAR expression observed with temozolomide treatment (Fig. 1F). Olaparib also reversed the NAD+ reduction induced by temozolomide (Fig. 1G; Supplementary Fig. S1D). Thus, temozolomide promotes acute NAD+ consumption via PARP activation in IDH-mutant cancer cells.

NAMPT inhibitors and temozolomide have additive cytotoxicity in endogenous IDH1-mutant cancer cells

As noted above, IDH1-mutant cancer cells contain a lower baseline NAD+ level compared with IDH wild-type cancer cells, rendering IDH1-mutant cells selectively sensitive to NAD+ biosynthesis blockade by NAMPT inhibition (24). We hypothesized that temozolomide-induced NAD+ reduction could therefore potentiate the metabolic cytotoxicity of NAMPT inhibitors in IDH-mutant cells. To investigate this further, we first tested the temozolomide sensitivity of several endogenous IDH1-mutant cancer cell lines. Exposure to temozolomide alone for 24 to 36 hours had no or marginal impact on viability of IDH1-mutant cells, consistent with the effective BER of DNA damage during this time interval.

However, the same exposure to temozolomide significantly augmented the acute cytotoxic effect of the NAMPT inhibitors FK866 and GMX1778 (Fig. 2A and B; Supplementary Fig. S2A). Testing a range of NAMPT inhibitor doses revealed that low dose NAMPT inhibitor (2.5 nmol/L) combined with temozolomide had more potent cytotoxicity at this early time point (24 hours) than high dose (250 nmol/L) NAMPT inhibitor monotherapy in IDH1-mutant cancer cells (Fig. 2C). Importantly, this metabolic combination effect seen in IDH1-mutant cancer cells was distinct from, and not dependent upon the rate-limiting genotoxic damage of single-agent temozolomide itself, nor its well-established modifier, MGMT promoter methylation status (Supplementary Table S1). In contrast, we did not detect any effect on cell viability with temozolomide, NAMPT inhibitors, and their combination in several IDH1 wild-type glioma tumorsphere cells at 24 hours (Fig. 2D; Supplementary Fig. S2B). At 72 hours, temozolomide combination with a higher dose range of NAMPT inhibitors showed some cytotoxic effects in endogenous IDH1 wild-type glioma tumorsphere lines (Supplementary Fig. S2C and S2D). In addition, combination treatment was not detectably toxic to NHA (Fig. 2E). Thus, combined treatment with NAMPT inhibitors and temozolomide had potent and additive effects on acute cytotoxicity, in a manner that had greater selectivity for IDH1-mutant cells.

Figure 2.

Cytotoxic effect of temozolomide (TMZ) and NAMPT inhibitors in IDH1-mutant cancers. A, CellTiter-Glo cell viability assay after 24-hour treatment of IDH1-mutant cancer lines with no inhibitor (white bars), FK866 (gray bars), or GMX1778 (black bars) and indicated doses of temozolomide. *, P < 0.05 for difference between FK866 and FK866 plus temozolomide. **, P < 0.05 for difference between GMX1778 and GMX1778 plus temozolomide. B, CellTiter-Glo cell viability assay of IDH1-mutant cancer lines. Top, treatment with temozolomide (200 μmol/L; white bars), FK866 (12.5 nmol/L; gray bars), or FK866 plus temozolomide (black bars) at indicated time point. *, P < 0.05 for difference between DMSO and FK866. **, P < 0.05 for difference between FK866 and FK866 plus temozolomide. Bottom, treatment with temozolomide (200 μmol/L; white bars), GMX1778 (12.5 nmol/L; gray bars), or GMX1778 plus temozolomide (black bars) at indicated time point. *, P < 0.05 for difference between DMSO and GMX1778. **, P < 0.05 for difference between GMX1778 and GMX1778 plus temozolomide. Data were expressed as % DMSO. C and D, Relative cell viability of IDH1-mutant cancer lines (C) and IDH1 wild-type lines (D) after 24-hour exposure to DMSO (blue lines) and temozolomide (200 μmol/L; red lines) with/without FK866 (top) or GMX1778 (bottom) at indicated concentrations. E, Relative cell viability of NHA treated with DMSO (white bars) or temozolomide (200 μmol/L; black bars) and/or NAMPT inhibitors at 72 hours. Scale bars, SE.

Figure 2.

Cytotoxic effect of temozolomide (TMZ) and NAMPT inhibitors in IDH1-mutant cancers. A, CellTiter-Glo cell viability assay after 24-hour treatment of IDH1-mutant cancer lines with no inhibitor (white bars), FK866 (gray bars), or GMX1778 (black bars) and indicated doses of temozolomide. *, P < 0.05 for difference between FK866 and FK866 plus temozolomide. **, P < 0.05 for difference between GMX1778 and GMX1778 plus temozolomide. B, CellTiter-Glo cell viability assay of IDH1-mutant cancer lines. Top, treatment with temozolomide (200 μmol/L; white bars), FK866 (12.5 nmol/L; gray bars), or FK866 plus temozolomide (black bars) at indicated time point. *, P < 0.05 for difference between DMSO and FK866. **, P < 0.05 for difference between FK866 and FK866 plus temozolomide. Bottom, treatment with temozolomide (200 μmol/L; white bars), GMX1778 (12.5 nmol/L; gray bars), or GMX1778 plus temozolomide (black bars) at indicated time point. *, P < 0.05 for difference between DMSO and GMX1778. **, P < 0.05 for difference between GMX1778 and GMX1778 plus temozolomide. Data were expressed as % DMSO. C and D, Relative cell viability of IDH1-mutant cancer lines (C) and IDH1 wild-type lines (D) after 24-hour exposure to DMSO (blue lines) and temozolomide (200 μmol/L; red lines) with/without FK866 (top) or GMX1778 (bottom) at indicated concentrations. E, Relative cell viability of NHA treated with DMSO (white bars) or temozolomide (200 μmol/L; black bars) and/or NAMPT inhibitors at 72 hours. Scale bars, SE.

Close modal

Temozolomide and NAMPT inhibitor combination induced additive NAD+ depletion in endogenous IDH1-mutant cancers

To further explore the mechanism of this genotype-selective combination effect, we tested whether the combination of NAMPT inhibitor and temozolomide, each of which reduces intracellular NAD+ levels as single agents, would have an additive effect on NAD+ levels in IDH1 mutant cancer cells. We found that combined NAMPT inhibition and temozolomide exposure significantly decreased NAD+ and NADH levels compared with single-agent treatment, and the effect was dose dependent (Fig. 3A; Supplementary Fig. S3A). We also found that NADP+ and NADPH levels were only moderately decreased by the combination treatment (Supplementary Fig. S3A). Measurement of NAD+ levels at different time points revealed that, in contrast to the sustained NAD+ reduction induced by NAMPT inhibitors, the temozolomide-induced decrease in NAD+ was transient and recovered to baseline at 6 to 12 hours posttreatment (Fig. 3B; Supplementary Fig. S3A and S3B). Interestingly, preincubation with temozolomide for 36 hours prior to addition of NAMPT inhibitor attenuated combination benefit (Supplementary Fig. S3C), underscoring the role of the transient temozolomide-induced NAD+ decrease in the additive effect on NAD+ levels and efficacy. Nevertheless, the combination of NAMPT inhibitors and temozolomide progressively decreased NAD+ levels over time when given concurrently (Fig. 3B).

Figure 3.

NAD+ depletion with temozolomide (TMZ) and NAMPT inhibitors in endogenous IDH1-mutant cancers. A, Relative NAD+ levels in IDH1-mutant lines with no inhibitor (white bars), FK866 (12.5 nmol/L; gray bars), or GMX1778 (12.5 nmol/L; black bars) with/without temozolomide for 6 hours. P < 0.05 for difference between DMSO and temozolomide in no inhibitor–treated cells (*), FK866-treated cells (**), and GMX1778-treated cells (***). B, Relative NAD+ levels in IDH1-mutant lines treated with FK866 (12.5 nmol/L; red lines), GMX1778 (12.5 nmol/L; orange lines), temozolomide (200 μmol/L; blue lines), FK866 plus temozolomide (black lines), and GMX1778 plus temozolomide (green lines) at indicated time point. DMSO-treated cells were used as control. C, Relative NAD+ levels in IDH1-mutant lines treated with no inhibitor (white bars), FK866 (12.5 nmol/L; gray bars), or GMX1778 (12.5 nmol/L; black bars) with temozolomide (200 μmol/L), olaparib (5 μmol/L), or temozolomide plus olaparib for 6 hours. *, P < 0.05 for difference between NAMPT inhibitors versus NAMPT inhibitors and olaparib. **, P < 0.05 for difference between NAMPT inhibitors plus temozolomide versus NAMPT inhibitors, temozolomide, plus olaparib. D, Relative cell viability of IDH1-mutant lines treated with no inhibitor (white bars), FK866 (12.5 nmol/L; gray bars), or GMX1778 (12.5 nmol/L; black bars) with DMSO, temozolomide (200 μmol/L), olaparib (5 μmol/L), or temozolomide plus olaparib for 24 hours. *, P < 0.05 for difference between NAMPT inhibitors versus NAMPT inhibitors and olaparib. **, P < 0.05 for difference between NAMPT inhibitors plus temozolomide versus NAMPT inhibitors, temozolomide, plus olaparib. E, Relative NAD+ levels in IDH1-mutant lines treated with no inhibitor (white bars), FK866 (12.5 nmol/L; gray bars), or GMX1778 (12.5 nmol/L; black bars) with temozolomide (200 μmol/L), NMN (1 mmol/L), or temozolomide plus NMN for 6 hours. *, P < 0.05 for difference between NAMPT inhibitors versus NAMPT inhibitors plus NMN. **, P < 0.05 for difference between NAMPT inhibitors plus temozolomide versus NAMPT inhibitors, temozolomide, plus NMN treatment. F, Relative cell viability of IDH1-mutant lines treated with no inhibitor (white bars), FK866 (12.5 nmol/L; gray bars), or GMX1778 (12.5 nmol/L; black bars) with DMSO, temozolomide (200 μmol/L), NMN (1 mmol/L), or temozolomide plus NMN for 24 hours. *, P < 0.05 for difference between NAMPT inhibitors versus NAMPT inhibitors plus NMN. **, P < 0.05 for difference between NAMPT inhibitors plus temozolomide versus NAMPT inhibitors, temozolomide, plus NMN treatment.

Figure 3.

NAD+ depletion with temozolomide (TMZ) and NAMPT inhibitors in endogenous IDH1-mutant cancers. A, Relative NAD+ levels in IDH1-mutant lines with no inhibitor (white bars), FK866 (12.5 nmol/L; gray bars), or GMX1778 (12.5 nmol/L; black bars) with/without temozolomide for 6 hours. P < 0.05 for difference between DMSO and temozolomide in no inhibitor–treated cells (*), FK866-treated cells (**), and GMX1778-treated cells (***). B, Relative NAD+ levels in IDH1-mutant lines treated with FK866 (12.5 nmol/L; red lines), GMX1778 (12.5 nmol/L; orange lines), temozolomide (200 μmol/L; blue lines), FK866 plus temozolomide (black lines), and GMX1778 plus temozolomide (green lines) at indicated time point. DMSO-treated cells were used as control. C, Relative NAD+ levels in IDH1-mutant lines treated with no inhibitor (white bars), FK866 (12.5 nmol/L; gray bars), or GMX1778 (12.5 nmol/L; black bars) with temozolomide (200 μmol/L), olaparib (5 μmol/L), or temozolomide plus olaparib for 6 hours. *, P < 0.05 for difference between NAMPT inhibitors versus NAMPT inhibitors and olaparib. **, P < 0.05 for difference between NAMPT inhibitors plus temozolomide versus NAMPT inhibitors, temozolomide, plus olaparib. D, Relative cell viability of IDH1-mutant lines treated with no inhibitor (white bars), FK866 (12.5 nmol/L; gray bars), or GMX1778 (12.5 nmol/L; black bars) with DMSO, temozolomide (200 μmol/L), olaparib (5 μmol/L), or temozolomide plus olaparib for 24 hours. *, P < 0.05 for difference between NAMPT inhibitors versus NAMPT inhibitors and olaparib. **, P < 0.05 for difference between NAMPT inhibitors plus temozolomide versus NAMPT inhibitors, temozolomide, plus olaparib. E, Relative NAD+ levels in IDH1-mutant lines treated with no inhibitor (white bars), FK866 (12.5 nmol/L; gray bars), or GMX1778 (12.5 nmol/L; black bars) with temozolomide (200 μmol/L), NMN (1 mmol/L), or temozolomide plus NMN for 6 hours. *, P < 0.05 for difference between NAMPT inhibitors versus NAMPT inhibitors plus NMN. **, P < 0.05 for difference between NAMPT inhibitors plus temozolomide versus NAMPT inhibitors, temozolomide, plus NMN treatment. F, Relative cell viability of IDH1-mutant lines treated with no inhibitor (white bars), FK866 (12.5 nmol/L; gray bars), or GMX1778 (12.5 nmol/L; black bars) with DMSO, temozolomide (200 μmol/L), NMN (1 mmol/L), or temozolomide plus NMN for 24 hours. *, P < 0.05 for difference between NAMPT inhibitors versus NAMPT inhibitors plus NMN. **, P < 0.05 for difference between NAMPT inhibitors plus temozolomide versus NAMPT inhibitors, temozolomide, plus NMN treatment.

Close modal

To investigate whether the additive effect of concurrent NAMPT inhibitor and temozolomide treatment on NAD+ levels in mutant IDH1 cells were mediated through PARP, we tested the combination with and without PARP inhibition with small-molecule inhibitors. Indeed, olaparib and veliparib were each sufficient to near-completely rescue the decrease in NAD+ levels as well as cell viability when given concurrently with NAMPT inhibitor and temozolomide in multiple IDH1-mutant cancer cells (Fig. 3C and D; Supplementary Fig. S3D and S3E). In addition, exogenous supplementation with NAD+ or nicotinamide mononucleotide, the product of the NAMPT reaction and the immediate precursor of NAD+, restored NAD+ levels and near-completely abrogated the cytotoxic effect of temozolomide and NAMPT inhibitor combination on IDH1-mutant cell viability (Fig. 3E and F; Supplementary Fig. S3F and S3G). These experiments indicate the effects of combined temozolomide and NAMPT inhibitor were both NAD+- and NAMPT specific. Together, these data indicate that combined treatment with NAMPT inhibitor and temozolomide results in additive NAD+ depletion in IDH1-mutant cancer cells.

Combination therapy with NAMPT inhibitor and temozolomide is selectively toxic to IDH1-mutant cancer cells

To experimentally confirm whether combined temozolomide and NAMPT inhibitor treatment is selective for IDH1 mutation itself, we engineered an isogenic system using the UACC257 melanoma line to stably express mutant IDH1 (UACC257-IDH1R132H) or control GFP (UACC257-GFP; Fig. 4A). There was no significant difference in proliferation between these lines (Supplementary Fig. S4A). Consistent with previous observations in a different model system (24), expression of mutant IDH1 lowered NAPRT1 expression (Fig. 4A) and steady-state NAD+ levels (Fig. 4B).

Figure 4.

Selective vulnerability of IDH1-mutant cancer cells to combination therapy with NAMPT inhibitor and temozolomide. A, Western blot analysis of IDH1R132H (left) and NAPRT1 (right) in UACC257-GFP and -IDH1R132H cells. Vinculin, loading control. B, Mean baseline absolute NAD+ levels in UACC257-GFP (1,916.8 ± 19.9 pmol/1 × 106 cells) and UACC257-IDH1R132H (1,494.2 ± 71.8 pmol/1 × 106 cells) *, P < 0.05 for difference between UACC257-GFP and-IDH1R132H cells. C, Relative cell viability of UACC257-GFP treated with DMSO (black bars) or temozolomide (200 μmol/L; orange bars), and UACC257-IDH1R132H with DMSO (blue bars) or temozolomide (200 μmol/L; red bars) with indicated doses of FK866 (left) or GMX1778 (right) for 96 hours. P < 0.05 for difference between temozolomide versus temozolomide plus NAMPT inhibitors in UACC257-GFP cells (*), DMSO versus NAMPT inhibitors in UACC257-IDH1R132H cells (**), and temozolomide versus temozolomide plus NAMPT inhibitors in UACC257-IDH1R132H cells (***). D, Western blot analysis of IDH1R132H in MGG18-IDH1-R132H cells with or without 1 μg/mL tetracycline (Tet). Vinculin, loading control. E, Relative cell viability of MGG18-IDH1-R132H (tet−) cells with DMSO (black bars) or temozolomide (200 μmol/L; orange bars), and MGG18-IDH1-R132H (tet+) cells with DMSO (blue bars), or temozolomide (200 μmol/L; red bars) with FK866 (left) or GMX1778 (right) treatment for 96 hours. P < 0.05 for difference between temozolomide versus temozolomide plus NAMPT inhibitors in MGG18-IDH1-R132H (tet−) cells (*), DMSO versus NAMPT inhibitors in MGG18-IDH1-R132H (tet+) cells (**), and temozolomide versus temozolomide plus NAMPT inhibitors in MGG18-IDH1-R132H (tet+) cells (***). F, Western blot analysis of NAPRT1 in control (nonsilencing) and NAPRT1 shRNA–transduced UACC257 cells. Vinculin, loading control. G, Mean baseline absolute NAD+ levels in nonsilencing shRNA–transduced UACC257 (1,342.5 ± 72.2 pmol/1 × 106 cells) and NAPRT1 shRNA–transduced UACC257 cells (724.8 ± 58.8 pmol/1 × 106 cells; *, P < 0.05). H, Relative cell viability of nonsilencing (NS) shRNA–transduced UACC257 with DMSO (black bars) or temozolomide (200 μmol/L; orange bars), and NAPRT1 shRNA–transduced UACC257 cells with DMSO (blue bars), or temozolomide (200 μmol/L; red bars) with FK866 (left) or GMX1778 (right) treatment for 96 hours. P < 0.05 for difference between temozolomide versus temozolomide plus NAMPT inhibitors in nonsilencing shRNA–transduced cells (*), DMSO versus NAMPT inhibitors in NAPRT1 shRNA–transduced UACC257 cells (**), temozolomide versus temozolomide plus NAMPT inhibitors in NAPRT1 shRNA–transduced UACC257 cells (***).

Figure 4.

Selective vulnerability of IDH1-mutant cancer cells to combination therapy with NAMPT inhibitor and temozolomide. A, Western blot analysis of IDH1R132H (left) and NAPRT1 (right) in UACC257-GFP and -IDH1R132H cells. Vinculin, loading control. B, Mean baseline absolute NAD+ levels in UACC257-GFP (1,916.8 ± 19.9 pmol/1 × 106 cells) and UACC257-IDH1R132H (1,494.2 ± 71.8 pmol/1 × 106 cells) *, P < 0.05 for difference between UACC257-GFP and-IDH1R132H cells. C, Relative cell viability of UACC257-GFP treated with DMSO (black bars) or temozolomide (200 μmol/L; orange bars), and UACC257-IDH1R132H with DMSO (blue bars) or temozolomide (200 μmol/L; red bars) with indicated doses of FK866 (left) or GMX1778 (right) for 96 hours. P < 0.05 for difference between temozolomide versus temozolomide plus NAMPT inhibitors in UACC257-GFP cells (*), DMSO versus NAMPT inhibitors in UACC257-IDH1R132H cells (**), and temozolomide versus temozolomide plus NAMPT inhibitors in UACC257-IDH1R132H cells (***). D, Western blot analysis of IDH1R132H in MGG18-IDH1-R132H cells with or without 1 μg/mL tetracycline (Tet). Vinculin, loading control. E, Relative cell viability of MGG18-IDH1-R132H (tet−) cells with DMSO (black bars) or temozolomide (200 μmol/L; orange bars), and MGG18-IDH1-R132H (tet+) cells with DMSO (blue bars), or temozolomide (200 μmol/L; red bars) with FK866 (left) or GMX1778 (right) treatment for 96 hours. P < 0.05 for difference between temozolomide versus temozolomide plus NAMPT inhibitors in MGG18-IDH1-R132H (tet−) cells (*), DMSO versus NAMPT inhibitors in MGG18-IDH1-R132H (tet+) cells (**), and temozolomide versus temozolomide plus NAMPT inhibitors in MGG18-IDH1-R132H (tet+) cells (***). F, Western blot analysis of NAPRT1 in control (nonsilencing) and NAPRT1 shRNA–transduced UACC257 cells. Vinculin, loading control. G, Mean baseline absolute NAD+ levels in nonsilencing shRNA–transduced UACC257 (1,342.5 ± 72.2 pmol/1 × 106 cells) and NAPRT1 shRNA–transduced UACC257 cells (724.8 ± 58.8 pmol/1 × 106 cells; *, P < 0.05). H, Relative cell viability of nonsilencing (NS) shRNA–transduced UACC257 with DMSO (black bars) or temozolomide (200 μmol/L; orange bars), and NAPRT1 shRNA–transduced UACC257 cells with DMSO (blue bars), or temozolomide (200 μmol/L; red bars) with FK866 (left) or GMX1778 (right) treatment for 96 hours. P < 0.05 for difference between temozolomide versus temozolomide plus NAMPT inhibitors in nonsilencing shRNA–transduced cells (*), DMSO versus NAMPT inhibitors in NAPRT1 shRNA–transduced UACC257 cells (**), temozolomide versus temozolomide plus NAMPT inhibitors in NAPRT1 shRNA–transduced UACC257 cells (***).

Close modal

Next, we tested the combination on cell viability in the engineered lines. Consistent with our previous findings, expression of mutant IDH1 induced sensitivity to NAMPT inhibitors. Although a minor additive cytotoxic effect was seen in control GFP-expressing cells, combination treatment potently decreased cell viability in the mutant IDH1–expressing cell line (Fig. 4C). To confirm this IDH1 genotypic selectivity in a second line, we used a tetracycline-inducible IDH1-mutant glioblastoma tumorsphere line, MGG18-IDH1-R132H (Fig. 4D; ref. 24). As a control, tetracycline did not affect MGG18 response to NAMPT inhibitor or temozolomide (Supplementary Fig. S4B). Again, when mutant IDH1 was induced, no significant difference in cell proliferation was observed (Supplementary Fig. S4C). Consistent with our aforementioned findings with the UACC257 engineered lines, MGG18-IDH1-R132H became more acutely sensitive to NAMPT inhibitor treatment and demonstrated marked additive sensitivity to combination treatment with temozolomide, while combined therapy was minimally cytotoxic without mutant IDH1 induction (Fig. 4E).

We hypothesized that the mechanism of enhanced cytotoxicity in IDH1-mutant cells, compared with wild-type cells, was mediated through the reset of basal steady-state NAD+ levels by inhibition of NAPRT1, a rate-limiting enzyme of NAD+ biosynthesis. To validate this threshold effect, we established a stable NAPRT1 knockdown line of UACC257 (Fig. 4F). Consistent with our results observed in the endogenous and enforced IDH1-mutant lines, NAPRT1 knockdown decreased steady-state levels of NAD+ (Fig. 4G). Importantly, single-agent NAMPT inhibitor and combination treatment decreased cell viability of the NAPRT1 knockdown line to a greater extent than that of controls (Fig. 4H). Altogether, these findings indicate that mutant IDH1 decreases basal NAD+ through NAPRT1 inhibition, mediating increased vulnerability to combination treatment with NAMPT inhibitor and temozolomide in a genotype selective manner.

Combined temozolomide and NAMPT inhibitor treatment induces autophagy and apoptosis, and results in cell death in IDH1-mutant cells

In parallel with ATP-based cell viability assay, we confirmed cell death from combined NAMPT inhibitor and temozolomide treatment by counting viable cells after 48-hour treatment (Fig. 5A; Supplementary Fig. S5A and S5B). We further found that combination treatment significantly inhibited clone formation of HT1080 cells (Fig. 5B), even with lower doses of temozolomide (Supplementary Fig. S5C). We observed induction of LC3-II expression, a marker of autophagy, at 48 hours after NAMPT inhibitor treatment (Supplementary Fig. S5D). Combined treatment with temozolomide more strongly induced LC3-II expression than single-agent exposure with either NAMPT inhibitor or temozolomide, and LC3-II expression was detectable as early as 6 hours posttreatment (Fig. 5C), indicative of an earlier engagement of a metabolic stress response. Furthermore, exogenous 3-methyladenine (3-MA), an autophagy inhibitor, partially rescued the cytotoxicity of the combination treatment (Fig. 5D; Supplementary Fig. S5E). In addition to autophagy, we found combination treatment induced PARP cleavage (Supplementary Fig. S5F) and increased caspase-3/7 activity (Supplementary Fig. S5G), suggesting a contribution of apoptosis to cell death. However, the pan-caspase inhibitor Z-VAD-FMK, which strongly suppressed caspase-3/7 activity (Supplementary Fig. S5H), only mildly rescued cell viability after combined temozolomide and NAMPT inhibitor treatment (Supplementary Fig. S5I). These data provide evidence that the additive cytotoxicity of temozolomide when combined with an NAMPT inhibitor in IDH-mutant cells is primarily mediated through augmentation of a metabolic stress response.

Figure 5.

Cell death mechanism in combined temozolomide (TMZ) and NAMPT inhibitor treatment of IDH1-mutant cells. A, Cell viability of IDH1-mutant MGG152 after treatment with FK866 (12.5 nmol/L), temozolomide (200 μmol/L), or FK866 plus temozolomide (top) and GMX1778 (12.5 nmol/L), temozolomide (200 μmol/L) or GMX1778 plus temozolomide (bottom) measured by ATP-based CellTiter-Glo assay (white bars) and viable cell count (Trypan blue exclusion) assay (black bars) at indicated time points. P < 0.05 for comparing NAMPT inhibitors versus NAMPT inhibitors plus temozolomide with the CellTiter-Glo assay (*), and the Trypan blue exclusion assay (**). DMSO-treated cells were used as control at each time point. B, Clonogenic assay of HT1080 cells treated for 27 hours with DMSO, temozolomide (200 μmol/L), FK866 (12.5 nmol/L), or FK866 (12.5 nmol/L) plus temozolomide (200 μmol/L) showing surviving fraction after treatment. Data are expressed as % DMSO. *, P < 0.05 for difference between DMSO versus temozolomide plus FK866. C, Western blot analysis of LC3-I and LC3-II expressions in MGG152 cells treated with DMSO, FK866 (12.5 nmol/L), temozolomide (200 μmol/L), or FK866 plus temozolomide for 6 hours. Actin, loading control. D, Relative cell viability of IDH1-mutant lines treated with no inhibitor (white bars), FK866 (12.5 nmol/L; gray bars), or GMX1778 (12.5 nmol/L; black bars) with DMSO, temozolomide (200 μmol/L), 3-MA (5 mmol/L), or temozolomide plus 3-MA for 24 hours. *, P < 0.05 for difference between NAMPT inhibitors and NAMPT inhibitors plus 3-MA. **, P < 0.05 for difference between NAMPT inhibitors plus temozolomide versus NAMPT inhibitors, temozolomide, plus 3-MA. Scale bars, SE.

Figure 5.

Cell death mechanism in combined temozolomide (TMZ) and NAMPT inhibitor treatment of IDH1-mutant cells. A, Cell viability of IDH1-mutant MGG152 after treatment with FK866 (12.5 nmol/L), temozolomide (200 μmol/L), or FK866 plus temozolomide (top) and GMX1778 (12.5 nmol/L), temozolomide (200 μmol/L) or GMX1778 plus temozolomide (bottom) measured by ATP-based CellTiter-Glo assay (white bars) and viable cell count (Trypan blue exclusion) assay (black bars) at indicated time points. P < 0.05 for comparing NAMPT inhibitors versus NAMPT inhibitors plus temozolomide with the CellTiter-Glo assay (*), and the Trypan blue exclusion assay (**). DMSO-treated cells were used as control at each time point. B, Clonogenic assay of HT1080 cells treated for 27 hours with DMSO, temozolomide (200 μmol/L), FK866 (12.5 nmol/L), or FK866 (12.5 nmol/L) plus temozolomide (200 μmol/L) showing surviving fraction after treatment. Data are expressed as % DMSO. *, P < 0.05 for difference between DMSO versus temozolomide plus FK866. C, Western blot analysis of LC3-I and LC3-II expressions in MGG152 cells treated with DMSO, FK866 (12.5 nmol/L), temozolomide (200 μmol/L), or FK866 plus temozolomide for 6 hours. Actin, loading control. D, Relative cell viability of IDH1-mutant lines treated with no inhibitor (white bars), FK866 (12.5 nmol/L; gray bars), or GMX1778 (12.5 nmol/L; black bars) with DMSO, temozolomide (200 μmol/L), 3-MA (5 mmol/L), or temozolomide plus 3-MA for 24 hours. *, P < 0.05 for difference between NAMPT inhibitors and NAMPT inhibitors plus 3-MA. **, P < 0.05 for difference between NAMPT inhibitors plus temozolomide versus NAMPT inhibitors, temozolomide, plus 3-MA. Scale bars, SE.

Close modal

Therapeutic effect of combined NAMPT inhibitor and temozolomide is independent of MMR pathway activity

The clonal emergence of resistance to alkylating chemotherapy in a population of cancer cells is known to be mediated by inactivation of genes in the MMR system (33), and an MMR deficiency is typically acquired in post-temozolomide recurrent gliomas (13–16). To assess the independence of our observed combination effect from known mediators of temozolomide genotoxic cytotoxicity, we examined whether temozolomide potentiation of NAD+ depletion in IDH1-mutant cancer cells varied in relation to MMR status. To this end, we established stable MSH6 knockdown lines of HT1080, which is MGMT unmethylated (Supplementary Table S1; Fig. 6A and B). As expected, knockdown of MSH6 (MSH6 #1 and MSH6 #2) induced resistance to temozolomide, confirming the critical role of MSH6 in MMR-mediated temozolomide sensitivity (Fig. 6C). However, MSH6 knockdown did not alter NAMPT inhibitor sensitivity in HT1080 cells (Fig. 6D). Importantly, and strikingly indicative of this second independent mechanism, temozolomide equally potentiated the effect of NAMPT inhibitor treatment in HT1080 cells with and without MSH6 knockdown (Fig. 6D). Similarly, in MGMT methylated MGG152 cells (Fig. 6A), we established stable MSH6 knockdown cells and confirmed MSH6 knockdown increased resistance to temozolomide (Fig. 6E and F). Again, irrespective of MSH6 status, NAMPT inhibitors displayed equal efficacy (Fig. 6G), and temozolomide treatment equivalently augmented NAMPT inhibitor cytotoxicity in MGG152 cells (Fig. 6G). These findings indicate the independent effectiveness of temozolomide metabolic augmentation targeting NAD+ in IDH1-mutant cells, regardless of the status of resistance determinants to temozolomide genotoxic monotherapy effect.

Figure 6.

Efficacy of combined temozolomide and NAMPT inhibitor treatment for MMR-deficient IDH1-mutant cells. A, MGMT promoter methylation status in HT1080 and MGG152 cells. MSP, methylation-specific PCR; U, unmethylated; M, methylated. B, Western blot analysis of MSH6 in nonsilencing (NS) and MSH6 shRNA–transduced HT1080 cells (MSH6 #1 and #2). Actin, loading control. C, Cell viability assay of nonsilencing (NS) and MSH6 shRNA–transduced HT1080 cells (MGMT promoter unmethylated; MSH6 #1, left; MSH6 #2, right) treated with temozolomide for 6 days. *, P < 0.05 for difference between nonsilencing versus MSH6 shRNA–transduced cells. D, Relative cell viability of nonsilencing and MSH6 shRNA (MSH6 #1 and #2)–transduced HT1080 cells with NAMPT inhibitors with or without temozolomide (200 μmol/L) for 48 hours. P < 0.05 for difference between NAMPT inhibitors versus temozolomide plus NAMPT inhibitors in nonsilencing shRNA–transduced cells (*), NAMPT inhibitors versus temozolomide plus NAMPT inhibitors in MSH6 shRNA–transduced cells (**, MSH6 #1; ***, MSH6 #2). E, Western blot analysis of MSH6 in nonsilencing shRNA and MSH6 shRNA (MSH6 #1)–transduced MGG152 cells. Actin, loading control. F, Cell viability assay of nonsilencing and MSH6 shRNA (MSH6 #1)–transduced MGG152 cells (MGMT promoter methylated) with temozolomide treatment for 6 days. *, P < 0.05 for difference between nonsilencing and MSH6 shRNA–transduced cells. G, Relative cell viability of nonsilencing and MSH6 shRNA (MSH6 #1)–transduced MGG152 cells treated with NAMPT inhibitors with or without temozolomide (200 μmol/L) for 24 hours. Scale bars, SE.

Figure 6.

Efficacy of combined temozolomide and NAMPT inhibitor treatment for MMR-deficient IDH1-mutant cells. A, MGMT promoter methylation status in HT1080 and MGG152 cells. MSP, methylation-specific PCR; U, unmethylated; M, methylated. B, Western blot analysis of MSH6 in nonsilencing (NS) and MSH6 shRNA–transduced HT1080 cells (MSH6 #1 and #2). Actin, loading control. C, Cell viability assay of nonsilencing (NS) and MSH6 shRNA–transduced HT1080 cells (MGMT promoter unmethylated; MSH6 #1, left; MSH6 #2, right) treated with temozolomide for 6 days. *, P < 0.05 for difference between nonsilencing versus MSH6 shRNA–transduced cells. D, Relative cell viability of nonsilencing and MSH6 shRNA (MSH6 #1 and #2)–transduced HT1080 cells with NAMPT inhibitors with or without temozolomide (200 μmol/L) for 48 hours. P < 0.05 for difference between NAMPT inhibitors versus temozolomide plus NAMPT inhibitors in nonsilencing shRNA–transduced cells (*), NAMPT inhibitors versus temozolomide plus NAMPT inhibitors in MSH6 shRNA–transduced cells (**, MSH6 #1; ***, MSH6 #2). E, Western blot analysis of MSH6 in nonsilencing shRNA and MSH6 shRNA (MSH6 #1)–transduced MGG152 cells. Actin, loading control. F, Cell viability assay of nonsilencing and MSH6 shRNA (MSH6 #1)–transduced MGG152 cells (MGMT promoter methylated) with temozolomide treatment for 6 days. *, P < 0.05 for difference between nonsilencing and MSH6 shRNA–transduced cells. G, Relative cell viability of nonsilencing and MSH6 shRNA (MSH6 #1)–transduced MGG152 cells treated with NAMPT inhibitors with or without temozolomide (200 μmol/L) for 24 hours. Scale bars, SE.

Close modal

Combined temozolomide and NAMPT inhibitor therapy has additive activity against IDH1-mutant xenograft tumors

The NAMPT inhibitor FK866 potently inhibits the growth of HT1080 (IDH1R132C) xenograft tumors implanted in the flank of immunocompromised mice (24). We tested whether temozolomide alteration of NAD+ metabolism could potentiate the in vivo growth inhibiting effects of NAMPT inhibitor in this IDH-mutant xenograft model. We first measured intratumoral NAD+ levels with temozolomide treatment, two doses of NAMPT inhibitor (15 or 30 mg/kg per day), and combined therapy with temozolomide and FK866 at 15 mg/kg per day. Monotherapy with temozolomide and both doses of NAMPT inhibitor significantly reduced intratumoral NAD+ levels compared with vehicle-treated tumor tissue, and there was a dose-dependent decrease in NAD+ levels with NAMPT inhibitor treatment (Fig. 7A). Notably, reduced-dose FK866 (15 mg/kg per day) plus temozolomide treatment reduced intratumoral NAD+ to the levels equivalent to that of high dose FK866 (30 mg/kg per day).

Figure 7.

In vivo therapeutic effects of combined temozolomide (TMZ) and NAMPT inhibitor treatment in an endogenous IDH1-mutant cancer model. A, Mean absolute NAD+ levels in HT1080 xenograft specimens treated with vehicle, temozolomide (50 mg/kg), FK866 (15 or 30 mg/kg), or FK866 (15 mg/kg) plus temozolomide. Data were expressed as pmol/mg protein. *, P < 0.05 for comparison between FK866 (15 mg/kg) and FK866 (30 mg/kg). **, P < 0.05 for comparison between FK866 (15 mg/kg) and FK866 (15 mg/kg) plus temozolomide (50 mg/kg). B and C, Therapeutic effects of combined temozolomide and NAMPT inhibitor treatment in subcutaneous HT1080 (B) and U87 (C) xenografts. Plot depicts tumor volume changes over time, with values being normalized to tumor volumes at day 0 when treatments began. Each point represents the average of tumor volumes in a group treated with vehicle (black line; n = 6), temozolomide (50 mg/kg; purple line; n = 6), FK866 (15 mg/kg; blue line; n = 6), or FK866 (15 mg/kg) plus temozolomide (red line; n = 6). *, P < 0.05 comparison between FK866 (15 mg/kg) and vehicle. **, P < 0.05 comparison between FK866 (15 mg/kg) plus temozolomide (50 mg/kg) and vehicle. ***, P < 0.05 comparison between FK866 (15 mg/kg) plus temozolomide (50 mg/kg) and FK866 (15 mg/kg). D, Hematoxylin and eosin (H&E), Ki-67, and LC3A/B staining of HT1080 xenograft tissues after treatment with vehicle (top) and FK866 and temozolomide (bottom). Scale bar, 100 μm. E, Bar graphs showing percent positive staining cells of Ki67 (71.0% ± 9.3% in vehicle vs. 31.0% ± 10.2% in FK866 plus temozolomide; P = 0.003), and LC3A/B (21.5% ± 1.6% in vehicle vs. 90.7% ± 2.4% in FK866 plus temozolomide; P = 0.0009). F, Bodyweight changes of animals bearing subcutaneous HT1080 xenografts during the treatment with vehicle (black line; n = 6; oral gavage), temozolomide (50 mg/kg; purple line; n = 6; oral gavage), FK866 (15 mg/kg; blue line; n = 6; i.p), or FK866 (15 mg/kg) plus temozolomide (red line; n = 6). Arrows, the time points when treatment doses were given.

Figure 7.

In vivo therapeutic effects of combined temozolomide (TMZ) and NAMPT inhibitor treatment in an endogenous IDH1-mutant cancer model. A, Mean absolute NAD+ levels in HT1080 xenograft specimens treated with vehicle, temozolomide (50 mg/kg), FK866 (15 or 30 mg/kg), or FK866 (15 mg/kg) plus temozolomide. Data were expressed as pmol/mg protein. *, P < 0.05 for comparison between FK866 (15 mg/kg) and FK866 (30 mg/kg). **, P < 0.05 for comparison between FK866 (15 mg/kg) and FK866 (15 mg/kg) plus temozolomide (50 mg/kg). B and C, Therapeutic effects of combined temozolomide and NAMPT inhibitor treatment in subcutaneous HT1080 (B) and U87 (C) xenografts. Plot depicts tumor volume changes over time, with values being normalized to tumor volumes at day 0 when treatments began. Each point represents the average of tumor volumes in a group treated with vehicle (black line; n = 6), temozolomide (50 mg/kg; purple line; n = 6), FK866 (15 mg/kg; blue line; n = 6), or FK866 (15 mg/kg) plus temozolomide (red line; n = 6). *, P < 0.05 comparison between FK866 (15 mg/kg) and vehicle. **, P < 0.05 comparison between FK866 (15 mg/kg) plus temozolomide (50 mg/kg) and vehicle. ***, P < 0.05 comparison between FK866 (15 mg/kg) plus temozolomide (50 mg/kg) and FK866 (15 mg/kg). D, Hematoxylin and eosin (H&E), Ki-67, and LC3A/B staining of HT1080 xenograft tissues after treatment with vehicle (top) and FK866 and temozolomide (bottom). Scale bar, 100 μm. E, Bar graphs showing percent positive staining cells of Ki67 (71.0% ± 9.3% in vehicle vs. 31.0% ± 10.2% in FK866 plus temozolomide; P = 0.003), and LC3A/B (21.5% ± 1.6% in vehicle vs. 90.7% ± 2.4% in FK866 plus temozolomide; P = 0.0009). F, Bodyweight changes of animals bearing subcutaneous HT1080 xenografts during the treatment with vehicle (black line; n = 6; oral gavage), temozolomide (50 mg/kg; purple line; n = 6; oral gavage), FK866 (15 mg/kg; blue line; n = 6; i.p), or FK866 (15 mg/kg) plus temozolomide (red line; n = 6). Arrows, the time points when treatment doses were given.

Close modal

On the basis of these observations, we tested whether the combination of reduced-dose FK866 with temozolomide has antitumor effect in HT1080 xenografts. Compared with vehicle treatment, FK866 (15 mg/kg) alone, but not temozolomide alone, significantly suppressed tumor growth. Indeed, combination treatment was significantly superior in inhibiting tumor growth compared with monotherapy with either agent or vehicle (Fig. 7B). In contrast, we did not observe the combination effect in IDH wild-type U87 xenografts (Fig. 7C). Histopathologic analysis demonstrated the presence of necrotic regions in tumor tissues receiving combination treatment, but not in those receiving vehicle treatment. IHC analysis revealed that combination treatment significantly decreased Ki-67 expression (Fig. 7D and E) and induced the autophagy marker LC3-A/B (Fig. 7D and E), similar to patterns previously observed in single-agent NAMPT inhibitor treatment (24). In contrast, we did not find significant differences in cleaved PARP and cleaved caspase-3 expression between combination and control groups (Supplementary Fig. S6). Importantly, we did not observe significant adverse toxicity during treatment, and no differences in body weight were detected between all the treatment groups (Fig. 7F).

Steady-state levels of cellular NAD+ are established by the balance of metabolite influx from biosynthetic enzymes, such as NAPRT1 and NAMPT, and efflux via NAD+ consumption by enzyme complexes including the PARP and SIRT proteins (28). In IDH1-mutant cancer cells, the baseline pool of NAD+ is diminished compared with that in IDH1 wild-type cells, due to reduced expression of the NAD+ biosynthetic enzyme NAPRT1 (24). In our findings herein, we observed a burst of NAD+ consumption associated with PARP activation during the initial time period after temozolomide treatment. This NAD+ consumption critically depleted NAD+ levels and introduced a window of hypervulnerability to NAMPT inhibitors. This finding provided a rationale for the therapeutic combination of temozolomide and NAMPT inhibitors, which resulted in improved efficacy, when compared with their use as single agents, in both in vitro and in vivo IDH1-mutant cancer models.

Importantly, our evidence indicates the effect is genotype selective, as we observe greater potency against IDH1-mutant cells compared with wild-type (including noncancerous) cells. Whether our findings would extend across the diverse spectrum of IDH1-mutant cancers would require further validation. Nonetheless, the efficacy of combining these two agents may allow for optimized dosing to maintain therapeutic index without compromising anticancer activity; for instance, by enabling reduced doses of agents that induce significant toxicity at doses needed for efficacy in monotherapy (34). This is particularly relevant to NAMPT inhibitors, which have been observed to have dose-limiting thrombocytopenia and gastrointestinal toxicities (35). Also, given the time dependence of temozolomide-induced PARP activation observed in vitro, future studies would be required to delineate the optimal timing window for combined administration of these agents. Analogously, we can envision strategies that combine systemic administration of one agent with local administration of the partner drug (or prodrug with CNS penetration before activation), to maximize intratumoral pharmacodynamic effect.

Furthermore, our findings may offer a new mechanistic perspective on the accumulating evidence that IDH1-mutant gliomas can display clinical responsiveness to alkylating chemotherapy. Our data raise the possibility that metabolic vulnerabilities during the acute DNA damage response could, in part, mediate this effectiveness. The discovery of an alternative mechanism of chemotherapy-induced cytotoxicity in IDH-mutant cancer cells would also provide an explanation for the paradox of why MGMT promoter methylation is not a predictive biomarker of chemotherapy response in IDH-mutant gliomas (36), which is in contrast to the evidence base derived from IDH wild-type glioblastomas (9, 37).

Finally, and notably from a clinical standpoint, our findings suggest that exploiting this mechanism of temozolomide-induced metabolic stress may allow for the recovery of an effective therapeutic index even in recurrent cancers with MMR-mediated alkylator resistance. Indeed, Sobol and colleagues have previously highlighted this ability to shift the spectrum of rate-limiting DNA damage response by modulating PARP/NAD+ consumption to restore cytotoxicity in MMR-deficient IDH wild-type glioma cells (38). On the basis of our findings here in IDH1-mutant tumors, we propose that upfront combination treatment regimens could potentially minimize or even avoid the development of this escape pathway of MMR deficiency in recurrences, improving the durability of treatment response. Further studies of other chemotherapeutic agents in clinical use, such as carmustine and lomustine, are needed to fully understand how they may exploit metabolic vulnerability for IDH1-mutant cancers. Notably, these alkylating agents generate a spectrum of DNA adducts that differ compared with temozolomide, especially with regard to N3-methyladenine and N7-methylguanine lesions (27), and therefore, their effects in combination with NAD+-reducing agents may differ as well.

In conclusion, we demonstrate the rational conception of a combined therapy with the alkylating agent temozolomide and targeted metabolic inhibition for IDH1-mutant tumors. These findings offer an evidence base for treatment strategies with the potential to minimize toxicity, allow for salvage therapy, and ultimately provide a more effective treatment for patients with these cancers.

T. Batchelor is a consultant/advisory board member for Amgen, Merck, NXDC, Proximagen, and Roche. A.J. Iafrate has ownership interest (including patents) in Amgen and is a consultant/advisory board member for Roche. D.P. Cahill has received speakers bureau honoraria from Merck. No potential conflicts of interest were disclosed by the other authors.

Conception and design: K. Tateishi, J.J. Miller, T.T. Batchelor, H. Wakimoto, A.S. Chi, D.P. Cahill

Development of methodology: K. Tateishi, F. Higuchi

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): K. Tateishi, M.V.A. Koerner, N. Lelic, A.J. Iafrate

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): K. Tateishi, J.J. Miller, N. Lelic, G.M. Shankar, A.J. Iafrate, H. Wakimoto, D.P. Cahill

Writing, review, and/or revision of the manuscript: K. Tateishi, J.J. Miller, G.M. Shankar, S. Tanaka, D.E. Fisher, T.T. Batchelor, A.J. Iafrate, H. Wakimoto, A.S. Chi, D.P. Cahill

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): K. Tateishi, M.V.A. Koerner, S. Tanaka, T.T. Batchelor, D.P. Cahill

Study supervision: H. Wakimoto, A.S. Chi, D.P. Cahill

Other (extending study to melanomas with IDH1 mutation): D.E. Fisher

The investigators thank Drs. Hang Lee and Alona Muzikansky for biostatistical input and review. The investigators would additionally like to thank their colleagues within the DFHCC for helpful discussions, and timely and careful review of the manuscript.

This work was supported by NIH P50CA165962-01A1 to T.T. Batchelor (principal investigator), D.P. Cahill, H. Wakimoto, and A.S. Chi, K24 CA125440-06 to T.T. Batchelor, a Burroughs Wellcome Fund Career Award to D.P. Cahill, Grant-Aid for Scientific Research (16K10765 to K. Tateishi), Yokohama Academic Foundation (K. Tateishi), The Yasuda Medical Foundation (K. Tateishi), and a Society of Nuclear Medicine and Molecular Imaging Wagner-Torizuka Fellowship (K. Tateishi).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Parsons
DW
,
Jones
S
,
Zhang
X
,
Lin
JC
,
Leary
RJ
,
Angenendt
P
, et al
An integrated genomic analysis of human glioblastoma multiforme
.
Science
2008
;
321
:
1807
12
.
2.
Yan
H
,
Parsons
DW
,
Jin
G
,
McLendon
R
,
Rasheed
BA
,
Yuan
W
, et al
IDH1 and IDH2 mutations in gliomas
.
N Engl J Med
2009
;
360
:
765
73
.
3.
Hartmann
C
,
Meyer
J
,
Balss
J
,
Capper
D
,
Mueller
W
,
Christians
A
, et al
Type and frequency of IDH1 and IDH2 mutations are related to astrocytic and oligodendroglial differentiation and age: a study of 1,010 diffuse gliomas
.
Acta Neuropathol
2009
;
118
:
469
74
.
4.
The Cancer Genome Atlas Research Network
,
Brat
DJ
,
Verhaak
RG
,
Aldape
KD
,
Yung
WK
,
Salama
SR
, et al
Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas
.
N Engl J Med
2015
;
372
:
2481
98
.
5.
Eckel-Passow
JE
,
Lachance
DH
,
Molinaro
AM
,
Walsh
KM
,
Decker
PA
,
Sicotte
H
, et al
Glioma groups based on 1p/19q, IDH, and TERT promoter mutations in tumors
.
N Engl J Med
2015
;
372
:
2499
508
.
6.
Louis
DN
,
Perry
A
,
Reifenberger
G
,
von Deimling
A
,
Figarella-Branger
D
,
Cavenee
WK
, et al
The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary
.
Acta Neuropathol
2016
;
131
:
803
20
.
7.
Cairncross
JG
,
Wang
M
,
Jenkins
RB
,
Shaw
EG
,
Giannini
C
,
Brachman
DG
, et al
Benefit from procarbazine, lomustine, and vincristine in oligodendroglial tumors is associated with mutation of IDH
.
J Clin Oncol
2014
;
32
:
783
90
.
8.
Buckner
JC
,
Shaw
EG
,
Pugh
SL
,
Chakravarti
A
,
Gilbert
MR
,
Barger
GR
, et al
Radiation plus procarbazine, CCNU, and vincristine in low-grade glioma
.
N Engl J Med
2016
;
374
:
1344
55
.
9.
Hegi
ME
,
Diserens
A
,
Gorlia
T
,
Hamou
M
,
de Tribolet
N
,
Weller
M
, et al
MGMT gene silencing and benefit from temozolomide in glioblastoma
.
N Engl J Med
2005
;
352
:
997
1003
.
10.
Roos
WP
,
Batista
LF
,
Naumann
SC
,
Wick
W
,
Weller
M
,
Menck
CF
, et al
Apoptosis in malignant glioma cells triggered by the temozolomide-induced DNA lesion O6-methylguanine
.
Oncogene
2007
;
26
:
186
97
.
11.
Tentori
L
,
Graziani
G
. 
Pharmacological strategies to increase the antitumor activity of methylating agents
.
Curr Med Chem
2002
;
9
:
1285
301
.
12.
Hunter
C
,
Smith
R
,
Cahill
DP
,
Stephens
P
,
Stevens
C
,
Teague
J
, et al
A hypermutation phenotype and somatic MSH6 mutations in recurrent human malignant gliomas after alkylator chemotherapy
.
Cancer Res
2006
;
66
:
3987
91
.
13.
Cahill
DP
,
Codd
PJ
,
Batchelor
TT
,
Curry
WT
,
Louis
DN
. 
MSH6 inactivation and emergent temozolomide resistance in human glioblastomas
.
Clin Neurosurg
2008
;
55
:
165
71
.
14.
Yip
S
,
Miao
J
,
Cahill
DP
,
Iafrate
AJ
,
Aldape
K
,
Nutt
CL
, et al
MSH6 mutations arise in glioblastomas during temozolomide therapy and mediate temozolomide resistance
.
Clin Cancer Res
2009
;
15
:
4622
9
.
15.
Johnson
BE
,
Mazor
T
,
Hong
C
,
Barnes
M
,
Aihara
K
,
McLean
CY
, et al
Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma
.
Science
2014
;
343
:
189
93
.
16.
van Thuijl
HF
,
Mazor
T
,
Johnson
BE
,
Fouse
SD
,
Aihara
K
,
Hong
C
, et al
Evolution of DNA repair defects during malignant progression of low-grade gliomas after temozolomide treatment
.
Acta Neuropathol
2015
;
129
:
597
607
.
17.
Losman
JA
,
Kaelin
WG
 Jr
. 
What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer
.
Genes Dev
2013
;
27
:
836
52
.
18.
Dang
L
,
White
DW
,
Gross
S
,
Bennett
BD
,
Bittinger
MA
,
Driggers
EM
, et al
Cancer-associated IDH1 mutations produce 2-hydroxyglutarate
.
Nature
2009
;
462
:
739
44
.
19.
Koivunen
P
,
Lee
S
,
Duncan
CG
,
Lopez
G
,
Lu
G
,
Ramkissoon
S
, et al
Transformation by the (R)-enantiomer of 2-hydroxyglutarate linked to EGLN activation
.
Nature
2012
;
483
:
484
8
.
20.
Izquierdo-Garcia
JL
,
Viswanath
P
,
Eriksson
P
,
Cai
L
,
Radoul
M
,
Chaumeil
MM
, et al
IDH1 mutation induces reprogramming of pyruvate metabolism
.
Cancer Res
2015
;
75
:
2999
3009
.
21.
Chesnelong
C
,
Chaumeil
MM
,
Blough
MD
,
Al-Najjar
M
,
Stechishin
OD
,
Chan
JA
, et al
Lactate dehydrogenase A silencing in IDH mutant gliomas
.
Neuro Oncol
2014
;
16
:
686
95
.
22.
Grassian
AR
,
Parker
SJ
,
Davidson
SM
,
Divakaruni
AS
,
Green
CR
,
Zhang
X
, et al
IDH1 mutations alter citric acid cycle metabolism and increase dependence on oxidative mitochondrial metabolism
.
Cancer Res
2014
;
74
:
3317
31
.
23.
Reitman
ZJ
,
Jin
G
,
Karoly
ED
,
Spasojevic
I
,
Yang
J
,
Kinzler
KW
, et al
Profiling the effects of isocitrate dehydrogenase 1 and 2 mutations on the cellular metabolome
.
Proc Natl Acad Sci U S A
2011
;
108
:
3270
5
.
24.
Tateishi
K
,
Wakimoto
H
,
Iafrate
AJ
,
Tanaka
S
,
Loebel
F
,
Lelic
N
, et al
Extreme vulnerability of IDH1 mutant cancers to NAD+ depletion
.
Cancer Cell
2015
;
28
:
773
84
.
25.
Seltzer
MJ
,
Bennett
BD
,
Joshi
AD
,
Gao
P
,
Thomas
AG
,
Ferraris
DV
, et al
Inhibition of glutaminase preferentially slows growth of glioma cells with mutant IDH1
.
Cancer Res
2010
;
70
:
8981
7
.
26.
Trivedi
RN
,
Almeida
KH
,
Fornsaglio
JL
,
Schamus
S
,
Sobol
RW
. 
The role of base excision repair in the sensitivity and resistance to temozolomide-mediated cell death
.
Cancer Res
2005
;
65
:
6394
400
.
27.
Fu
D
,
Calvo
JA
,
Samson
LD
. 
Balancing repair and tolerance of DNA damage caused by alkylating agents
.
Nat Rev Cancer
2012
;
12
:
104
20
.
28.
Cambronne
XA
,
Stewart
ML
,
Kim
D
,
Jones-Brunette
AM
,
Morgan
RK
,
Farrens
DL
, et al
Biosensor reveals multiple sources for mitochondrial NAD(+)
.
Science
2016
;
352
:
1474
7
.
29.
Wakimoto
H
,
Kesari
S
,
Farrell
CJ
,
Curry
WT
 Jr
,
Zaupa
C
,
Aghi
M
, et al
Human glioblastoma-derived cancer stem cells: establishment of invasive glioma models and treatment with oncolytic herpes simplex virus vectors
.
Cancer Res
2009
;
69
:
3472
81
.
30.
Wakimoto
H
,
Mohapatra
G
,
Kanai
R
,
Curry
WT
 Jr
,
Yip
S
,
Nitta
M
, et al
Maintenance of primary tumor phenotype and genotype in glioblastoma stem cells
.
Neuro Oncol
2012
;
14
:
132
44
.
31.
Wakimoto
H
,
Tanaka
S
,
Curry
WT
,
Loebel
F
,
Zhao
D
,
Tateishi
K
, et al
Targetable signaling pathway mutations are associated with malignant phenotype in IDH-mutant gliomas
.
Clin Cancer Res
2014
;
20
:
2898
909
.
32.
Tateishi
K
,
Iafrate
AJ
,
Ho
Q
,
Curry
WT
,
Batchelor
TT
,
Flaherty
KT
, et al
Myc-driven glycolysis is a therapeutic target in glioblastoma
.
Clin Cancer Res
2016
;
22
:
4452
65
.
33.
Branch
P
,
Aquilina
G
,
Bignami
M
,
Karran
P
. 
Defective mismatch binding and a mutator phenotype in cells tolerant to DNA damage
.
Nature
1993
;
362
:
652
4
.
34.
von Heideman
A
,
Berglund
A
,
Larsson
R
,
Nygren
P
. 
Safety and efficacy of NAD depleting cancer drugs: results of a phase I clinical trial of CHS 828 and overview of published data
.
Cancer Chemother Pharmacol
2010
;
65
:
1165
72
.
35.
Sampath
D
,
Zabka
TS
,
Misner
DL
,
O'Brien
T
,
Dragovich
PS
. 
Inhibition of nicotinamide phosphoribosyltransferase (NAMPT) as a therapeutic strategy in cancer
.
Pharmacol Ther
2015
;
151
:
16
31
.
36.
Wick
W
,
Meisner
C
,
Hentschel
B
,
Platten
M
,
Schilling
A
,
Wiestler
B
, et al
Prognostic or predictive value of MGMT promoter methylation in gliomas depends on IDH1 mutation
.
Neurology
2013
;
81
:
1515
22
.
37.
Gilbert
MR
,
Wang
M
,
Aldape
KD
,
Stupp
R
,
Hegi
ME
,
Jaeckle
KA
, et al
Dose-dense temozolomide for newly diagnosed glioblastoma: a randomized phase III clinical trial
.
J Clin Oncol
2013
;
31
:
4085
91
.
38.
Goellner
EM
,
Grimme
B
,
Brown
AR
,
Lin
YC
,
Wang
XH
,
Sugrue
KF
, et al
Overcoming temozolomide resistance in glioblastoma via dual inhibition of NAD+ biosynthesis and base excision repair
.
Cancer Res
2011
;
71
:
2308
17
.

Supplementary data