• NAMPT is a therapeutic vulnerability that can be pharmacologically targeted in high-risk, TP53-deficient −7/del(7q) myeloid malignancies.

  • CRISPR screening and data mining identify collateral lethal genes in cancers characterized by recurrent chromosomal arm–level deletions.

Abstract

Monosomy 7 (−7) and deletions of chromosome arm 7q (del(7q)) are prevalent high-risk cytogenetic abnormalities that often co-occur with del(17p) (harboring TP53). To identify novel targeted therapies based on specific vulnerabilities in high-risk myeloid malignancies, we investigated druggable, chromosome 7–encoded essential genes that are monoallelically deleted in the context of −7/del(7q), that is, collateral lethal genes. By mining genome-wide CRISPR-Cas9 screen data sets, we identified nicotinamide phosphoribosyltransferase (NAMPT) on 7q22.3 as a specific susceptibility in 81.5% of −7/del(7q) malignancies. Human acute myeloid leukemia (AML) cell lines with partial loss of NAMPT and primary samples from patients with −7 AML demonstrated heightened sensitivity to the NAMPT inhibitor KPT-9274 compared to control samples. Notably, NAMPT inhibitors were equally effective in NAMPT-deficient samples with TP53 loss. Furthermore, combining NAMPT and poly (ADP-ribose) polymerase (PARP) inhibitors, which augment DNA damage, resulted in synergistic therapeutic effects in NAMPT-deficient AML cells. These findings indicate that NAMPT heterozygosity is a therapeutic vulnerability in high-risk myeloid malignancies with −7/del(7q) and recommend NAMPT levels as a biomarker for NAMPT inhibitor sensitivity. This study also establishes a data-driven framework for identifying collateral lethal genes in cancers with recurrent chromosomal deletions.

Cytogenetic abnormalities occur in >50% of newly diagnosed acute myeloid leukemia (AML) cases and are a main predictor of patient outcomes.1 Abnormalities involving chromosome 7, including deletions (del(7q)) and monosomy 7 (−7), fall under the high-risk category and are seen across various myeloid disorders.2-4 Current treatments for high-risk patients with myelodysplastic syndrome, such as intensive chemotherapy with the demethylating azacitidine, are only effective in ∼39% of patients, with most experiencing relapse and modest survival improvements.5 For the 40% to 50% of patients who do not respond to azacitidine, the median overall survival is ∼6 months.6 Problematically, −7/del(7q) frequently coexists with additional unfavorable cytogenetic abnormalities, such as del(17p) (harboring TP53) and a complex karyotype (CK; with ≥3 independent cytogenetic abnormalities). Genetic lesions of TP53, which are independently associated with poor outcomes, occur in ∼43% to 56% of −7/del(7q) cases.7,8 Patients with CK and −7 or del(7q) demonstrate significantly worse survival than those CK patients without −7/del7q.8 Therefore, these patients urgently require alternative, targeted, and effective treatments.

Loss of tumor suppressor genes on deleted chromosomal arms drives leukemogenesis, and efforts to restore their function have been largely unsuccessful.9,10 An innovative approach to developing targeted therapy is to exploit the unique biological characteristics of −7/del(7q) cancerous cells that make them more vulnerable. Collateral lethality occurs when an essential passenger gene is codeleted with nearby tumor suppressors, causing haploinsufficiency and cancer-specific vulnerability.11,12 These genes have been termed “CYCLOPS” (copy number alterations yielding cancer liabilities owing to partial loss) genes.12 For example, CSNK1A1 on 5q32 is a haploinsufficient gene in del(5q) myelodysplastic syndrome. Its heterozygosity enables lenalidomide to selectively target malignant cells by degrading CK1α protein.13 

In this study, we hypothesized that losing an essential chromosome 7 gene makes −7/del(7q) cells highly dependent on the remaining allele's protein product. We discovered nicotinamide phosphoribosyltransferase (NAMPT) as an AML essential gene located on chromosome band 7q22.3 using publicly available genome-wide CRISPR/Cas9 essentiality screens.14-18,NAMPT encodes the rate-limiting enzyme in the NAD salvage pathway, converting nicotinamide to nicotinamide mononucleotide, a precursor to NAD. NAD is crucial for cellular energy, DNA repair, and genome integrity, supporting proteins such as PARP1 and SIRT1.19 CRISPR editing of a single allele of NAMPT in human AML cell lines led to lower NAMPT protein expression and increased dependence on residual NAMPT for AML survival, implicating NAMPT as a collateral lethal gene. Remarkably, we found that the TP53 mutational status did not attenuate the observed effects of NAMPT inhibition on cell death, indicating that NAMPT inhibitors are equally effective in −7 patients with or without TP53 lesions. Pharmacological inhibition of NAMPT was similarly efficacious in primary samples from patients with −7/del(7q) AML with and without TP53 defects. Finally, given the prior evidence for defects in DNA damage recognition and repair in −7/del(7q) malignancies,9 we evaluated the cooperative effects of NAMPT and PARP inhibitors and found synergy in the drug combination.

Identification of CYCLOPS genes on chromosome 7

Haploinsufficient gene candidates were identified using publicly available CRISPR-Cas9 screen data from 23 AML cell lines (CRISPR_gene_dependency.csv; Dependency Map [DepMap]).17 DepMap classifies a cell line as dependent on a gene if the dependency probability exceeds 0.5. Gene knockout effects were quantified using Chronos scores, with values less than –0.5 indicating depletion and less than –1 indicating strong lethality. Additional CRISPR screen data from AML cell lines15,16,18 were obtained from BioGRID ORCS (https://orcs.thebiogrid.org/). Pan-essential genes (CRISPRInferredEssentials.csv) from DepMap were used to distinguish essential from nonessential genes. Of 711 chromosome 7 genes evaluated, 552 were expressed in human hematopoietic stem cells.20 Chronos scores were correlated with messenger RNA (mRNA) expression to identify CYCLOPS genes. Expression, relative, and absolute copy number data were downloaded from Cancer Cell Line Encyclopedia (CCLE) via DepMap21 (https://depmap.org/portal/download/custom/).

Identification of druggable chromosome 7 CYCLOPS genes as therapeutic targets

To unbiasedly identify hematopoietic essential genes on chromosome 7, we included only the genes that are expressed in human stem and progenitor cells (n = 552 genes; Figure 1A, step 1), the origin of myeloid neoplasms. Using the previously identified pan-essential gene list from cancer DepMap CRISPR screen data, we excluded nonessential genes on chromosome 7 (Figure 1A, step 2). Then, we analyzed genome-wide CRISPR-Cas9 screen data from 23 AML cell lines retrieved from the DepMap database14,17 and identified 170 chromosome 7 essential genes (Figure 1A, step 3; supplemental Table 1). To verify the reproducibility of our DepMap data–mining results, we compared our findings with 22 independently published CRISPR screens involving 15 different AML cell lines15,16,18 and identified 264 essential genes on chromosome 7 (Figure 1A, step 4; supplemental Table 1). By focusing on screens done in AML cell lines, we identified and differentiated genes on chromosome 7 that are pan-essential vs hematopoietic cells/AML essential.

Figure 1.

Genome-wide CRISPR screens identify Chr 7 CYCLOPS genes as new targets for therapy. (A) Workflow showing the identification of Chr 7 essential and CYCLOPS genes using 23 DepMap (green text) and 22 independently published screens (blue text) in AML cell lines. (B) Venn diagram showing overlap of essential genes between the 2 methods of in silico data mining for Chr 7 essential genes. Pearson χ2 test was used to calculate the statistical significance between overlaps. (C) Volcano plot showing Pearson correlations between CRISPR dependency (DepMap) and mRNA expression (CCLE) for Chr 7 essential genes. The genes whose expression levels were significantly correlated (adjusted P < .05) with either increased or decreased sensitivity to CRISPR-Cas9 knockout were categorized as ARGuS genes (red) and CYCLOPS genes (blue), respectively. Chr, chromosome.

Figure 1.

Genome-wide CRISPR screens identify Chr 7 CYCLOPS genes as new targets for therapy. (A) Workflow showing the identification of Chr 7 essential and CYCLOPS genes using 23 DepMap (green text) and 22 independently published screens (blue text) in AML cell lines. (B) Venn diagram showing overlap of essential genes between the 2 methods of in silico data mining for Chr 7 essential genes. Pearson χ2 test was used to calculate the statistical significance between overlaps. (C) Volcano plot showing Pearson correlations between CRISPR dependency (DepMap) and mRNA expression (CCLE) for Chr 7 essential genes. The genes whose expression levels were significantly correlated (adjusted P < .05) with either increased or decreased sensitivity to CRISPR-Cas9 knockout were categorized as ARGuS genes (red) and CYCLOPS genes (blue), respectively. Chr, chromosome.

Close modal

Of the 170 genes from the DepMap set, 55 were pan-essentials, and 115 were AML essentials. Of these 115 AML essentials, 47 were a hit in 21 of 23 AML cell lines evaluated, suggesting that these are true positives. Thirty-five of the 115 AML essentials were called in only 1 of the 23 screens, suggesting that these could be false positives (supplemental Figure 1A; supplemental Table 1). Of the 264 genes from the literature-based screen data, 70 were pan-essentials, and 21% of the hits (n = 56/264) occurred in 14 of the 22 screens, suggesting that these are likely true positives (supplemental Figure 1B; supplemental Table 1). We reasoned that targeting AML-specific CYCLOPS genes would minimize any negative systemic effects. There was a significant overlap between the genes identified in the 2 independent data sets, DepMap and literature-based screen analyses, yielding 127 essential genes, which we prioritized for downstream analysis (Figure 1A, step 5; Figure 1B).

CYCLOPS genes are essential genes with reduced expression in cancer cells, making them uniquely vulnerable to further suppression due to their reliance on limited gene products for survival. Conversely, our analysis revealed a complementary vulnerability in a distinct class of genes we refer to as ARGuS (Amplification-Related Gain Of Sensitivity) genes, inspired by the many-eyed giant Argus Panoptes of Greek mythology. In this case, cells with amplification or overexpression of essential genes become paradoxically more dependent on their continued activity. To classify essential chromosome 7 genes in these 2 categories, we correlated the expression levels of each candidate gene from CCLE to the dependency score for that gene across cell lines using DepMap data (Figure 1A, step 6; supplemental Table 2). Thirty of the 127 essential genes fit CYCLOPS criteria, where the gene expression level is significantly and negatively correlated with the gene essentiality score (Figure 1A, step 7; Figure 1C, blue). Among these 30 genes was PSMC2, a previously identified CYCLOPS gene on chromosome 7, validating our approach.12 We identified a significant positive correlation between gene expression and dependency for 21 genes, thus fitting the ARGuS criteria (Figure 1A, step 8; Figure 1C, red). CDK6 was one of the most significant ARGuS genes, demonstrating that higher gene expression correlates with increased dependency, as shown in a prior study.22 Our data show that integrating CRISPR-Cas9 screen data with gene expression reveals cancer cell–specific liabilities from chromosomal loss.

NAMPT is a druggable CYCLOPS gene in −7/del(7q) myeloid malignancies

We then investigated whether US Food and Drug Administration (FDA)–approved drugs or small-molecule inhibitors exist that could decrease the activity of any of the 30 identified CYCLOPS genes using data from the National Institutes of Health Illuminating the Druggable Genome project (supplemental Table 3).23 Illuminating the druggable genome classifies each gene into 1 of 4 druggability levels, ranging from targets with unknown biological functions to targets with FDA-approved drugs. Although NDUFA5 comes up as a hit with an approved FDA drug, metformin, it is not directly bound and inhibited by the drug. Therefore, we eliminated it from the downstream analysis. Most genes (25/30) had no compounds targeting their protein functions. Only one of the CYCLOPS proteins, NAMPT, had a direct small-molecule inhibitor, which was pursued in downstream analyses. (supplemental Figure 2A; supplemental Table 3).

First, we confirmed NAMPT as a CYCLOPS gene by analyzing its correlation with DNA copy number and transcript levels across DepMap cell lines (Figure 2A; supplemental Figure 2B-C). NAMPT expression and copy number were significantly correlated, particularly in hematopoietic and myeloid cells. Cell lines with low NAMPT expression (bottom 25% of mRNA expression across all cancers) were more dependent on it than those with high expression (top 25%; Figure 2B). CRISPR dependency scores showed NAMPT is selectively essential in hematopoietic cancer cell lines (supplemental Figure 2D), likely due to lower transcript levels than solid tumors (Figure 2C).

Figure 2.

NAMPT is a CYCLOPS and druggable essential gene in −7/del(7q) myeloid malignancies. (A) Spearman correlation between relative NAMPT CN and NAMPT mRNA expression in 58 myeloid cancer cell lines based on CCLE relative CN data and expression data downloaded from DepMap portal. (B) Comparison of NAMPT Chronos scores (DepMap) between cell lines expressing higher (top 25% [n = 223]) and lower (bottom 25% [n = 278]) NAMPT mRNA (CCLE). (C) NAMPT mRNA (CCLE) levels in solid (n = 1231) vs hematopoietic (n = 219, myeloid and lymphoid) cancer cell lines. One-way analysis of variance (ANOVA) with Dunnett correction for multiple comparison tests was performed for statistical analysis. (D) SNP array data (left) summarized in a pie chart (right) from 27 −7/del(7q) patients. Of 27 −7/del(7q) patients, 22 (∼81%) were NAMPT haploinsufficient. (E) Comparison of NAMPT mRNA expression (z score RPKM) of TCGA patients with AML with normal karyotype (n = 87) vs −7/del(7q) patients haploinsufficient for NAMPT (n = 21). An unpaired 2-tailed t test was performed for statistical analysis. The results shown here are based on data generated by the TCGA research network. CN, copy number; RPKM, reads per kilobase per million mapped reads; TPM, transcripts per million.

Figure 2.

NAMPT is a CYCLOPS and druggable essential gene in −7/del(7q) myeloid malignancies. (A) Spearman correlation between relative NAMPT CN and NAMPT mRNA expression in 58 myeloid cancer cell lines based on CCLE relative CN data and expression data downloaded from DepMap portal. (B) Comparison of NAMPT Chronos scores (DepMap) between cell lines expressing higher (top 25% [n = 223]) and lower (bottom 25% [n = 278]) NAMPT mRNA (CCLE). (C) NAMPT mRNA (CCLE) levels in solid (n = 1231) vs hematopoietic (n = 219, myeloid and lymphoid) cancer cell lines. One-way analysis of variance (ANOVA) with Dunnett correction for multiple comparison tests was performed for statistical analysis. (D) SNP array data (left) summarized in a pie chart (right) from 27 −7/del(7q) patients. Of 27 −7/del(7q) patients, 22 (∼81%) were NAMPT haploinsufficient. (E) Comparison of NAMPT mRNA expression (z score RPKM) of TCGA patients with AML with normal karyotype (n = 87) vs −7/del(7q) patients haploinsufficient for NAMPT (n = 21). An unpaired 2-tailed t test was performed for statistical analysis. The results shown here are based on data generated by the TCGA research network. CN, copy number; RPKM, reads per kilobase per million mapped reads; TPM, transcripts per million.

Close modal

To determine the frequency of NAMPT deletions in human −7/del(7q) myeloid diseases, we assessed the samples from patients with −7/del(7q) AML within the The Cancer Genome Atlas (TCGA) Single Nucleotide Polymorphism (SNP) array data.24 (https://www.cancer.gov/tcga). One allele of NAMPT was deleted in 81.5% of −7/del(7q) cases (n = 22) in the TCGA patient cohort (Figure 2D). The normalized z score of NAMPT mRNA expression was significantly lower (51%) in samples with NAMPT deletions than in patients with diploid chromosome 7 status (Figure 2E). Together, these data suggest that a reduced NAMPT DNA copy number, which correlates with decreased NAMPT transcripts, represents a potential collaterally lethal liability in most −7/del(7q) patients.

Pharmacological inhibition of NAMPT leads to increased cytotoxicity in NAMPT-deficient cells

We next investigated the relationship between NAMPT expression and response to NAMPT inhibitors in AML. First, we used the Genomics of Drug Sensitivity in Cancer data set 2, which contains drug sensitivity data for 349 cancer cell lines treated with the NAMPT inhibitor, FK-866.25 A total of 255 of 349 cell lines also had transcriptome profiling in the CCLE database.21 We found a significant positive correlation between NAMPT expression and FK-866 area under the curve in 256 pan-cancer (supplementary Figure 3A) and 43 hematopoietic cancer cell lines (Figure 3A), indicating that cells with reduced NAMPT are more sensitive to NAMPT inhibition.

Figure 3.

Pharmacological inhibition of NAMPT leads to increased cytotoxicity in NAMPT-deficient cells. (A) Spearmon correlation coefficient between FK-866 AUC (GDSC2) and NAMPT mRNA expression (CCLE) in 43 cell lines of hematopoietic origin. (B) Representative western blot (left) and normalized protein densitometry analysis (right) showing NAMPT1 (guide NAMPT [gNAMPT]) clone 1 [mean, 0.45; standard deviation (SD), 0.072] and gNAMPT clone 2 [mean, 0.2507; SD, 0.11]) and β-ACTIN protein levels in control (HPRT) and NAMPT-deficient clones (n = 2 clones per genotype; n = 3 replicates per clone) of the OCI-AML3 cell line, which is triploid for NAMPT. (C) Representative dose-response curves of OCI-AML3 control (HPRT) and NAMPT-deficient clonal cell lines treated with the KPT-9274 for 24 hours. (D) Average AUC and IC50 values of OCI-AML3 control (HPRT) and NAMPT-deficient clonal cell lines treated with the KPT-9274 for 24 hours (n = 7). (E) Annexin V/7-ADD flow cytometric analysis of cell death of OCI-AML3 control (HPRT) and NAMPT-deficient clonal cell lines treated with 25-nM KPT-9274 for 48 hours (n = 3). (F) Annexin V/7-AAD flow cytometric analysis of cell death of OCI-AML3 control (HPRT) and NAMPT-deficient cells treated with 25-nM KPT-9274 for 48 hours in the presence or absence of Z-VAD-FMK (50 μM), Fer-1 (1 μM), or Nec-1 (10 μM). Data were normalized to 0-nM (dimethyl sulfoxide [DMSO]) condition of each genotype. One-way ANOVA with Dunnett or Sidak correction for multiple comparison tests was used for statistical analysis among 3 independent groups. AUC, area under the curve; Fer-1, ferrostatin-1; TPM, transcripts per million.

Figure 3.

Pharmacological inhibition of NAMPT leads to increased cytotoxicity in NAMPT-deficient cells. (A) Spearmon correlation coefficient between FK-866 AUC (GDSC2) and NAMPT mRNA expression (CCLE) in 43 cell lines of hematopoietic origin. (B) Representative western blot (left) and normalized protein densitometry analysis (right) showing NAMPT1 (guide NAMPT [gNAMPT]) clone 1 [mean, 0.45; standard deviation (SD), 0.072] and gNAMPT clone 2 [mean, 0.2507; SD, 0.11]) and β-ACTIN protein levels in control (HPRT) and NAMPT-deficient clones (n = 2 clones per genotype; n = 3 replicates per clone) of the OCI-AML3 cell line, which is triploid for NAMPT. (C) Representative dose-response curves of OCI-AML3 control (HPRT) and NAMPT-deficient clonal cell lines treated with the KPT-9274 for 24 hours. (D) Average AUC and IC50 values of OCI-AML3 control (HPRT) and NAMPT-deficient clonal cell lines treated with the KPT-9274 for 24 hours (n = 7). (E) Annexin V/7-ADD flow cytometric analysis of cell death of OCI-AML3 control (HPRT) and NAMPT-deficient clonal cell lines treated with 25-nM KPT-9274 for 48 hours (n = 3). (F) Annexin V/7-AAD flow cytometric analysis of cell death of OCI-AML3 control (HPRT) and NAMPT-deficient cells treated with 25-nM KPT-9274 for 48 hours in the presence or absence of Z-VAD-FMK (50 μM), Fer-1 (1 μM), or Nec-1 (10 μM). Data were normalized to 0-nM (dimethyl sulfoxide [DMSO]) condition of each genotype. One-way ANOVA with Dunnett or Sidak correction for multiple comparison tests was used for statistical analysis among 3 independent groups. AUC, area under the curve; Fer-1, ferrostatin-1; TPM, transcripts per million.

Close modal

To test the relationship between NAMPT levels and sensitivity to NAMPT inhibitors, we genetically modified human AML cell lines to reduce NAMPT DNA copy number. We used OCI-AML3 and MOLM-13, which are triploid and diploid, respectively, based on CCLE data for NAMPT. Via CRISPR-Cas9 editing, we inactivated 2 (OCI-AML3) or 1 (MOLM-13) NAMPT alleles and confirmed that these edits partially reduced NAMPT levels compared to isogenic controls (Figure 3B; supplemental Figure 3B-C). We generated a clonal control cell line using a guide targeting intron 2 of HPRT. We assessed the viability of these cell lines treated with increasing doses of KPT-9274, a NAMPT inhibitor currently in a phase 1 clinical trial (ClinicalTrials.gov identifier: NCT04914845). Half-maximal inhibitory concentration (IC50) values were significantly lower in the NAMPT-edited clones than the control line (Figure 3C-D; supplemental Figure 3D). KPT-9274 inhibits both NAMPT and p21-activated kinase 4 (PAK4).26 To exclude off-target effects, we used a NAMPT-specific inhibitor, FK-866. Similar results were observed with FK-866 in the OCI-AML3 cell line (supplemental Figure 3E). These findings using genetic tools reveal the on-target efficacy of NAMPT inhibitors in AML cells with reduced NAMPT expression.

The depletion of NAD via NAMPT inhibition leads to cell death, and the pathways driving this process include apoptosis, autophagy, and oncosis.27-29 Flow cytometry following annexin V and 7-AAD staining showed that KPT-9274 treatment resulted in a significant increase in the percentage of annexin V–positive and 7-AAD–negative (apoptotic) and annexin V and 7-aminoactinomycin D (7-AAD) dual positive (necrotic) staining in NAMPT-deficient cells compared to control cells (Figure 3E). Next, we used a pan-caspase inhibitor (Z-VAD-FMK), necroptosis inhibitor (necrostatin-1 [Nec-1]), and ferroptosis inhibitor (ferrostatin-1) to determine the mode of death mediated by NAMPT inhibition in our study. As expected, Z-VAD-FMK completely rescued caspase-dependent cell death (Figure 3F, left) in NAMPT-deficient cells but only partially rescued total cell death (Figure 3F, right). Ferroptosis inhibition did not alter NAMPT-mediated cytotoxicity. The significant increase in cell death we observe in response to the Nec-1 inhibitor (Figure 3F, left) might be due to Nec-1’s role in enhancing other forms of cell death while inhibiting necroptosis, as reported previously.30,31 Together, these data suggest that NAMPT inhibition results in higher cytotoxic effects via both caspase-dependent and independent cell death pathways on cells expressing lower levels of NAMPT.

The cytotoxic effects of KPT-9274 are independent of TP53 in NAMPT-deficient cells

Although −7/del(7q) is an independent high-risk factor, it often co-occurs with other cytogenetic abnormalities and somatic mutations. TP53 abnormalities occur in 43% to 56% of −7/del(7q) patients.7,8 In the −7/del(7q) TCGA cohort, we found that TP53 is mutated or deleted in 12 of 22 cases (54.5%) of −7/del(7q) with partial NAMPT loss (Figure 4A). Therefore, we next tested NAMPT inhibition in TP53- and NAMPT-deficient cell lines.

Figure 4.

Cytotoxic effects of KPT-9274 are independent of TP53 mutational status in cells with NAMPT deficiency. (A) Venn diagram showing TP53 mutational or deletion status in cases of −7/del(7q) NAMPT haploinsufficient patients from the TCGA data set. (B) Western blot showing NAMPT, TP53, β-ACTIN, and lamin protein levels in control (HPRT), TP53-null, NAMPT-deficient, TP53-null, and TP53-null and NAMPT-deficient clonal cell lines of OCI-AML3. (C) Representative dose-response curves (left) and AUC and IC50 graphs (right) of control, TP53-null, NAMPT-deficient, and TP53-null NAMPT-deficient clonal OCI-AML3 cell lines treated with the KPT-9274 for 24 hours (n = 4). (D) Annexin V/7-ADD flow cytometric analysis of cell death control, TP53-null, NAMPT-deficient, and TP53-null NAMPT-deficient clonal OCI-AML3 cell lines treated with 25 nM KPT-9274 for 48 hours (n = 3). (E) Western blot showing cleaved caspase 3 and β-ACTIN levels in control (HPRT), TP53-null, NAMPT-deficient, and TP53-null NAMPT-deficient OCI-AML3 whole-cell extracts. Cells were treated with 25-nM KPT-9274 for 24 and 48 hours. A control (HPRT) cell line treated with 1-μM staurosporine for 6 hours was used as a positive control for apoptosis. One-way ANOVA with Dunnett correction for multiple comparison tests were performed for statistical analysis among 4 independent groups.

Figure 4.

Cytotoxic effects of KPT-9274 are independent of TP53 mutational status in cells with NAMPT deficiency. (A) Venn diagram showing TP53 mutational or deletion status in cases of −7/del(7q) NAMPT haploinsufficient patients from the TCGA data set. (B) Western blot showing NAMPT, TP53, β-ACTIN, and lamin protein levels in control (HPRT), TP53-null, NAMPT-deficient, TP53-null, and TP53-null and NAMPT-deficient clonal cell lines of OCI-AML3. (C) Representative dose-response curves (left) and AUC and IC50 graphs (right) of control, TP53-null, NAMPT-deficient, and TP53-null NAMPT-deficient clonal OCI-AML3 cell lines treated with the KPT-9274 for 24 hours (n = 4). (D) Annexin V/7-ADD flow cytometric analysis of cell death control, TP53-null, NAMPT-deficient, and TP53-null NAMPT-deficient clonal OCI-AML3 cell lines treated with 25 nM KPT-9274 for 48 hours (n = 3). (E) Western blot showing cleaved caspase 3 and β-ACTIN levels in control (HPRT), TP53-null, NAMPT-deficient, and TP53-null NAMPT-deficient OCI-AML3 whole-cell extracts. Cells were treated with 25-nM KPT-9274 for 24 and 48 hours. A control (HPRT) cell line treated with 1-μM staurosporine for 6 hours was used as a positive control for apoptosis. One-way ANOVA with Dunnett correction for multiple comparison tests were performed for statistical analysis among 4 independent groups.

Close modal

We assessed NAMPT dependency by comparing Chronos scores between TP53 wild-type and mutant cell lines and observed no significant difference (supplemental Figure 4A). Furthermore, GDSC2 drug response and CCLE mutational data showed no significant difference in FK-866 response between wild-type and mutant TP53 cell lines (supplemental Figure 4B).

To experimentally validate our data mining, we used CRISPR-Cas9 to knockout TP53 in OCI-AML3 and MOLM-13 cell lines with and without NAMPT deficiency (Figure 4B). NAMPT-deficient cell lines with wild-type TP53 had comparable IC50 values to those without TP53 when treated with increasing doses of KPT-9274 (Figure 4C). Next, we examined cell death induced by KPT-9274 in TP53 null and NAMPT-deficient cell lines. Surprisingly, annexin V/7-AAD staining revealed that the absence of TP53 protein did not diminish the effects of NAMPT inhibition on the death of cell lines with reduced NAMPT (Figure 4D). Edited OCI-AML3 cells treated with KPT-9274 showed no cleaved caspase 3 activation at 24 or 48 hours, unlike staurosporine-treated cells (Figure 4E), suggesting NAMPT inhibitor cytotoxicity may occur via a TP53- and caspase-3–independent cell death process. These data align with previous findings that NAMPT inhibition-induced NAD depletion causes cell death via oncosis, independent of TP53 and caspase-3.32 However, all cell death mechanisms in our study remain unexplored. In summary, our data indicate that the cytotoxic effects of KPT-9274 in cancer cells with reduced NAMPT levels persist despite the TP53 mutational status, highlighting the potential to exploit NAMPT inhibitors for the treatment of −7/del7q patients with TP53 deficiency.

Pharmacological inhibition of NAMPT is cytotoxic in −7 patient cells

To establish the clinical relevance of our findings, we assessed the effects of NAMPT pharmacological inhibition in primary patient samples (supplemental Table 4). We first compared the relative viability of samples from patients with AML with and without −7 after treatment with KPT-9274 in liquid cultures. Samples from patients with −7 AML were significantly more responsive to NAMPT inhibition than AML samples diploid for chromosome 7 (Figure 5A). In addition, −7 patient cells with P53 inactivation were equally responsive to NAMPT inhibition (Figure 5A). There was no significant viability difference among vehicle-treated groups at 0 and 48 hours, indicating that observed differences are not due to varying culture conditions for different genetic subtypes (supplemental Figure 5A). Next, we determined the effects of KPT-9274 on normal hematopoietic cells in liquid cultures. Up to 500 nM of KPT-9274 showed no significant cytotoxicity in these cells at 24 and 48 hours, indicating selective killing of −7 cells while sparing normal CD34+ hematopoietic cells (supplemental Figure 5B; Figure 5B), consistent with previous studies.33-35 

Figure 5.

Pharmacological inhibition of NAMPT leads to increased cytotoxicity in primary −7 patient cells. (A) Relative viability of primary human samples from patients with AML with and without −7 after treatment with 100 nM of KPT-9274 or DMSO control for 48 hours in liquid culture (n = 3-5 patients per group). (B) Relative viability of normal CD34+ hematopoietic samples treated with increasing concentrations of KPT-9274 for 48 hours in liquid culture (n = 3). (C) Total CFU numbers of primary human samples from patients with AML with and without −7 after treatment with 100 nM of KPT-9274 or DMSO vehicle control for 14 days in MethoCult H4434 (n = 3-4 patients per group). One-way ANOVA with Dunnett correction for multiple comparison tests was performed for statistical analysis among 4 independent groups.

Figure 5.

Pharmacological inhibition of NAMPT leads to increased cytotoxicity in primary −7 patient cells. (A) Relative viability of primary human samples from patients with AML with and without −7 after treatment with 100 nM of KPT-9274 or DMSO control for 48 hours in liquid culture (n = 3-5 patients per group). (B) Relative viability of normal CD34+ hematopoietic samples treated with increasing concentrations of KPT-9274 for 48 hours in liquid culture (n = 3). (C) Total CFU numbers of primary human samples from patients with AML with and without −7 after treatment with 100 nM of KPT-9274 or DMSO vehicle control for 14 days in MethoCult H4434 (n = 3-4 patients per group). One-way ANOVA with Dunnett correction for multiple comparison tests was performed for statistical analysis among 4 independent groups.

Close modal

Previous studies have validated in vitro colony-forming unit (CFU) assays as predictors of in vivo drug toxicity in normal cells and efficacy on leukemic cells in preclinical models.36-38 Therefore, we used CFU assays to determine the effectiveness of pharmacological NAMPT inhibition in samples from patients with −7 AML with and without TP53 inactivation. Samples from patients with −7 AML treated with KPT-9274 formed significantly fewer colonies than vehicle-treated samples. Remarkably, TP53-deficient −7 samples showed the same sensitivity to KPT-9274 as −7 samples alone (Figure 5C). Notably, −7 samples with TP53 inactivation were also CK (supplemental Table 4). These findings confirm that NAMPT haploinsufficiency in −7 and some del(7q) patients, with or without TP53 inactivation or CK, presents a targetable therapeutic vulnerability.

KPT-9274 combines synergistically with PARP inhibition to effectively target NAMPT-deficient AML

PARP1 plays a critical role in chromatin organization and DNA repair. PARP1 requires oxidized NAD (NAD+) as a substrate for adenosine 5′-diphosphate ribosylation in DNA damage repair, and NAMPT inhibition reduces NAD+ available for PARP, leading to increased DNA damage.39 Recent studies have shown that NAMPT inhibitors synergize with PARP inhibition in Ewing sarcoma.40 We and others have shown that genes involved in DNA damage recognition and repair pathways, such as CUX1,41,EZH2,42,KMT2C,43 and KMT2E,44 are frequently deleted in −7 and del(7q) conditions,9 suggesting that DNA repair factor inhibitors could be a potential treatment in these cancers. Moreover, NAD is also generated via Preiss-Handler and Kynurenine (de novo) synthesis pathways in addition to the NAMPT-mediated salvage pathway. An increase in expression or function of the key players of those pathways, such as nicotinic acid phosphoribosyl transferase and quinolinate phosphoribosyl transferase, can replace NAD, resulting in NAMPT inhibitor resistance.45,46 For these reasons, we explored whether combining PARP inhibitors with NAMPT inhibitors could enhance the cytotoxic effects in NAMPT haploinsufficient AML conditions and be used to overcome potential resistance. Incremental doses of KPT-9274 and the PARP inhibitor niraparib were administered based on the IC50 value of each drug in NAMPT-deficient OCI-AML3 cell lines with or without P53 inactivation. We assessed drug synergy using the SynergyFinder application.47 We observed significant synergy between KPT-9274 and niraparib in inhibiting cell viability in NAMPT-deficient AML cells (mean Zero interaction potency [ZIP] score, 10.24) and in NAMPT plus TP53-deficient AML cells, which displayed greater synergy (mean ZIP score, 17.10; Figure 6A-B). Remarkably, TP53-null cell lines also exhibited synergy when treated with the combination therapy (supplemental Figure 6; mean ZIP score, 10.99), suggesting that molecular pathways other than NAD synthesis might play a role in synergy. These results provide supporting evidence for PARP and NAMPT inhibitor combination therapy in −7/del(7q) and TP53-mutant patients with AML.

Figure 6.

KPT-9274 combines synergistically with niraparib to effectively target NAMPT-deficient AML. (A) Dose-response matrix (top) and analysis of 3-dimensional (3D) graph of ZIP synergy scores (bottom) after KPT-9274 is combined with nirpaparib for 72 hours in NAMPT-deficient OCI-AML3 clonal cell line (n = 3) (B) Dose-response matrix (top) and analysis of 3D graph of ZIP synergy scores (bottom) after KPT-9274 is combined with niraparib for 72 hours in P53 null NAMPT-deficient OCI-AML3 clonal cell line (n = 3; synergy score less than –10, antagonistic interaction; more than –10 and <10, additive interaction; and >10, synergistic interaction). (C) Model; NAMPT haploinsufficiency is a collateral lethal therapeutic vulnerability in high-risk −7/del(7q) myeloid malignancies.

Figure 6.

KPT-9274 combines synergistically with niraparib to effectively target NAMPT-deficient AML. (A) Dose-response matrix (top) and analysis of 3-dimensional (3D) graph of ZIP synergy scores (bottom) after KPT-9274 is combined with nirpaparib for 72 hours in NAMPT-deficient OCI-AML3 clonal cell line (n = 3) (B) Dose-response matrix (top) and analysis of 3D graph of ZIP synergy scores (bottom) after KPT-9274 is combined with niraparib for 72 hours in P53 null NAMPT-deficient OCI-AML3 clonal cell line (n = 3; synergy score less than –10, antagonistic interaction; more than –10 and <10, additive interaction; and >10, synergistic interaction). (C) Model; NAMPT haploinsufficiency is a collateral lethal therapeutic vulnerability in high-risk −7/del(7q) myeloid malignancies.

Close modal

Despite molecular and genetic insights, no targeted therapies exist for −7/del(7q) myeloid malignancies. Previous studies have identified cancer-specific vulnerabilities using RNA interference (RNAi) and CRISPR screens.48 In this study, we used DepMap genome-wide CRISPR-Cas9 screen, gene expression, and copy number data to identify NAMPT as a targetable, codeleted, and essential passenger gene in −7/del(7q) myeloid neoplasms (Figure 6C). Our study provides a data-driven framework for uncovering collateral lethal genes in other cancers with recurrent chromosomal arm–level deletions using CRISPR screens and gene expression data. In addition to identifying known CYCLOPS dependencies, our study uncovered several ARGuS genes. These findings suggest that the inhibition of ARGuS genes could present a novel therapeutic strategy, particularly in tumors in which these genes are amplified or aberrantly expressed at high levels. By exploiting the addictive dependence that some cancer cells develop on overexpressed genes, ARGuS vulnerabilities may expand the landscape of precision oncology targets, especially in cancers lacking targetable mutations but characterized by gene amplification or transcriptional upregulation.22,49 

Although we found 30 essential genes on chromosome 7 that met the criteria for a CYCLOPS gene, NAMPT was the only gene with existing small-molecule inhibitors directly targeting it. We confirmed that cellular NAMPT levels determine sensitivity to pharmacological inhibition by using genetically modified cellular models of NAMPT haploinsufficiency. Congruently, primary samples from patients with −7 AML were more sensitive to NAMPT inhibition than other AML subtypes. This aligns with recent findings by Eldfors et al50 that −7/del(7q) samples were more responsive to NAMPT inhibitors in an AML drug screen. Although Eldfors et al50 demonstrated that primary −7/del(7q) samples were sensitive to NAMPT inhibitors, the relationship between NAMPT expression and sensitivity to NAMPT inhibitors was not established. In addition, we used CRISPR-Cas9 genetic editing to reduce NAMPT levels in AML cell lines and establish NAMPT inhibitor on-target effects. Moreover, we performed CFU assays using patient samples, a clinically relevant tool proven to effectively evaluate the toxic effects of drugs, to show that NAMPT inhibitors were effective in primary −7/del(7q) patient samples. Our work described herein firmly establishes NAMPT as a therapeutic vulnerability in −7/del(7q) myeloid neoplasms. Furthermore, it provides a strong rationale for designing clinical trials to target NAMPT-deficient cancers with NAMPT inhibitors.

We also demonstrated that the cytotoxic effects of the dual inhibitor KPT-9274 in −7 AML are not attributable to off-target PAK4 inhibition. In AML, inhibiting PAK4 has been associated with maintaining leukemic stem cell quiescence.51 However, functional studies in AML cell lines show that altering PAK4 expression does not affect proliferation or sensitivity to KPT-9274.33 Using CRISPR-edited NAMPT-deficient cells and the NAMPT-specific inhibitor FK866, we confirmed that NAMPT is the primary driver of KPT-9274–associated cytotoxicity in AML. Moreover, PAK4 is not differentially expressed in −7 AML compared to other karyotypes in our patient cohort (data not shown).52 Thus, we identify NAMPT, rather than PAK4, as the primary driver of KPT-9274–associated cytotoxicity in −7 AML.

TP53 is the most frequently mutated gene in cancer, and more than half of 7/del(7q) patients display P53 inactivation, resulting in worse patient survival outcomes.7,8 Despite advancements in cancer therapeutics, P53 inactivation remains a common cause of drug resistance, leading to poor prognoses.10 We observed that TP53 status did not affect NAMPT inhibitor sensitivity in cell lines and primary patient samples. We believe the P53-independent nature of NAMPT inhibitor–induced cell death widens the clinical applicability of NAMPT inhibitors. Moreover, 50% of patients with therapy-related myeloid neoplasm (t-MN) with −7/del7q, often combined with TP53 loss, face poor outcomes.7 Successful application of NAMPT inhibitors in the clinic has the potential for a substantial impact on the growing number of cancer survivors at risk of t-MN.

Studies have shown that NAMPT inhibitors mediate cell death via apoptosis, autophagy, and oncosis.27-29 Although we found that NAMPT inhibitor treatment leads to P53-independent cell death, possibly via oncosis, we also saw an increase in annexin V–positive/7-AAD–negative staining, commonly used as a marker for early apoptosis. However, reports have shown that annexin V staining is not specific to apoptotic cell death. Indeed, oncosis also results in the exposure of phosphatidylserine residues on the outside layer of the plasma membrane, detected by annexin V,53 which may explain our observed results of annexin V–positive cells without caspase 3 activation at 48 hours. However, because we observed partial rescue of cell death upon using a pan-caspase inhibitor, we predict that other caspase-dependent, nonapoptotic cell death pathways are at play. The exact cell death mechanisms in high-risk −7 and P53-inactive AML upon NAMPT inhibition will require systematic future investigations.

Previous studies have investigated NAMPT inhibitors as potential cancer therapeutics. Despite promising preclinical results, first-generation NAMPT inhibitors (FK866 and CHS828) failed in human clinical trials because of their limited efficacy and severe side effects.54,55 In preclinical assessments, second-generation NAMPT inhibitors KPT-9274 and OT-82 have shown more promise in hematologic malignancies33-35,56 and are undergoing phase 1 clinical trials (ClinicalTrials.gov identifiers: NCT04914845 and NCT03921879). OT-82 lacked cardiac, neurological, or retinal toxicities seen in other NAMPT inhibitors and did not affect mouse aging or longevity.56 Our findings show that −7/del(7q) AML samples with NAMPT haploinsufficiency are highly sensitive to NAMPT inhibition. Low, nontoxic doses of KPT-9274 and OT-82 could be effective in −7/del(7q) patients while sparing normal hematopoietic cells. These lower doses are safe for normal hematopoietic cells, as shown in our study and others.33,34 

Although NAMPT inhibitors are effective as single agents at low concentrations in −7 AML, resistance may limit their long-term efficacy. Previous studies have shown enhanced activity when combined with venetoclax or stearoyl-Coenzyme A desaturase inhibitors (CAY10566; MF-438).33-35,50 Combining NAMPT inhibitors with the FDA-approved PARP inhibitor, niraparib, enhanced cytotoxicity in NAMPT-deficient AML cells, regardless of TP53 status, broadening therapeutic potential. Notably, PARP inhibitors alone show limited efficacy in patients with −7/del(7q) abnormalities,50 making the observed sensitivity in TP53-null cells particularly compelling. Given that both PARP and sirtuins rely on NAD, NAMPT inhibition is expected to affect their function. Supporting this, prior work showed synergy between NAMPT and SIRT6 inhibition in AML.57 These findings highlight the potential of NAMPT-based combination therapies in hematologic malignancies with NAMPT or DNA repair pathway deficiencies.

The authors acknowledge Amittha Wickrema for the gift of CD34+ primary human hematopoietic cells and Angela Stoddart for her feedback on this manuscript. The authors thank the University of Pennsylvania Stem Cell and Xenograft core (RRID: SCR_010035) staff for sample acquisition and shipment, the University of Chicago’s DNA Sequencing Facility staff for assistance with Sanger sequencing, and the Cytometry and Antibody Technology Facility staff for assistance with cell sorting. The University of Chicago DNA Sequencing Facility (RRID: SCR_019196) and Cytometry and Antibody Technology (RRID: SCR_017760) Facilities receive financial support from the Cancer Center Support Grant P30CA014599 from the National Institutes of Health (NIH)/National Cancer Institute (NCI).

This work was supported by grants from the NIH/NCI; R01CA231880 (M.E.M.); T32CA009594 (H.Z.B.); NIH/National Heart, Lung, and Blood Institute (R01HL142782 and R01HL166184 [M.E.M.]; F32 HL156600 [M.D.S.]); the V Foundation for Cancer Research Pediatric Cancer Research V Scholar All-Star Award (M.E.M.); and the Cancer Research Foundation Fletcher Scholar Award (M.E.M.). M.E.M. is a Scholar of the Leukemia & Lymphoma Society. M.P.C. is supported by U54CA283759 from the NCI and Veterans Merit Award (I01BX000918).

Contribution: M.D.S. designed and performed the experiments, analyzed and interpreted data, and wrote the manuscript; H.Z.B. designed and performed CD34 viability assays and cell death inhibitor experiments, analyzed and interpreted data, and contributed to writing methods and editing of the manuscript; S.K. provided technical assistance for cell culture, DNA sequencing, and immunoblotting; S.J.S. and M.P.C. provided primary material from patients with AML, protocols for primary human AML colony-forming unit assays, and feedback on the manuscript; and M.E.M. conceived the study, assisted in experimental design and data interpretation, supervised the study, edited the manuscript, and provided research infrastructure.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Megan E. McNerney, Department of Pathology, Knapp Center for Biomedical Discovery, The University of Chicago, 5-128 900 East 57th St, Chicago, IL 60637; email: megan.mcnerney@bsd.uchicago.edu.

1.
Fröhling
S
,
Schlenk
RF
,
Kayser
S
, et al
.
Cytogenetics and age are major determinants of outcome in intensively treated acute myeloid leukemia patients older than 60 years: results from AMLSG trial AML HD98-B
.
Blood
.
2006
;
108
(
10
):
3280
-
3288
.
2.
Bejar
R
,
Levine
R
,
Ebert
BL
.
Unraveling the molecular pathophysiology of myelodysplastic syndromes
.
J Clin Oncol
.
2011
;
29
(
5
):
504
-
515
.
3.
Luna-Fineman
S
,
Shannon
KM
,
Atwater
SK
, et al
.
Myelodysplastic and myeloproliferative disorders of childhood: a study of 167 patients
.
Blood
.
1999
;
93
(
2
):
459
-
466
.
4.
Smith
SM
,
Le Beau
MM
,
Huo
D
, et al
.
Clinical-cytogenetic associations in 306 patients with therapy-related myelodysplasia and myeloid leukemia: the University of Chicago series
.
Blood
.
2003
;
102
(
1
):
43
-
52
.
5.
Fenaux
P
,
Mufti
GJ
,
Hellstrom-Lindberg
E
, et al
.
Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study
.
Lancet Oncol
.
2009
;
10
(
3
):
223
-
232
.
6.
Prébet
T
,
Gore
SD
,
Esterni
B
, et al
.
Outcome of high-risk myelodysplastic syndrome after azacitidine treatment failure
.
J Clin Oncol
.
2011
;
29
(
24
):
3322
-
3327
.
7.
McNerney
ME
,
Brown
CD
,
Peterson
AL
, et al
.
The spectrum of somatic mutations in high-risk acute myeloid leukaemia with -7/del(7q)
.
Br J Haematol
.
2014
;
166
(
4
):
550
-
556
.
8.
Mori
M
,
Kubota
Y
,
Durmaz
A
, et al
.
Genomics of deletion 7 and 7q in myeloid neoplasm: from pathogenic culprits to potential synthetic lethal therapeutic targets
.
Leukemia
.
2023
;
37
(
10
):
2082
-
2093
.
9.
Jotte
MRM
,
McNerney
ME
.
The significance of CUX1 and chromosome 7 in myeloid malignancies
.
Curr Opin Hematol
.
2022
;
29
(
2
):
92
-
102
.
10.
Hassin
O
,
Oren
M
.
Drugging p53 in cancer: one protein, many targets
.
Nat Rev Drug Discov
.
2023
;
22
(
2
):
127
-
144
.
11.
Frei
E
.
Gene deletion: a new target for cancer chemotherapy
.
Lancet
.
1993
;
342
(
8872
):
662
-
664
.
12.
Nijhawan
D
,
Zack
TI
,
Ren
Y
, et al
.
Cancer vulnerabilities unveiled by genomic loss
.
Cell
.
2012
;
150
(
4
):
842
-
854
.
13.
Krönke
J
,
Fink
EC
,
Hollenbach
PW
, et al
.
Lenalidomide induces ubiquitination and degradation of CK1α in del(5q) MDS
.
Nature
.
2015
;
523
(
7559
):
183
-
188
.
14.
Meyers
RM
,
Bryan
JG
,
McFarland
JM
, et al
.
Computational correction of copy number effect improves specificity of CRISPR-Cas9 essentiality screens in cancer cells
.
Nat Genet
.
2017
;
49
(
12
):
1779
-
1784
.
15.
Wang
T
,
Yu
H
,
Hughes
NW
, et al
.
Gene essentiality profiling reveals gene networks and synthetic lethal interactions with oncogenic Ras
.
Cell
.
2017
;
168
(
5
):
890
-
903.e15
.
16.
Tzelepis
K
,
Koike-Yusa
H
,
De Braekeleer
E
, et al
.
A CRISPR dropout screen identifies genetic vulnerabilities and therapeutic targets in acute myeloid leukemia
.
Cell Rep
.
2016
;
17
(
4
):
1193
-
1205
.
17.
Dempster
JM
,
Rossen
J
,
Kazachkova
M
, et al
.
Extracting biological insights from the project achilles genome-scale CRISPR screens in cancer cell lines
.
bioRxiv
.
Preprint posted online 31 July 2019
.
18.
Behan
FM
,
Iorio
F
,
Picco
G
, et al
.
Prioritization of cancer therapeutic targets using CRISPR-Cas9 screens
.
Nature
.
2019
;
568
(
7753
):
511
-
516
.
19.
Garten
A
,
Schuster
S
,
Penke
M
,
Gorski
T
,
de Giorgis
T
,
Kiess
W
.
Physiological and pathophysiological roles of NAMPT and NAD metabolism
.
Nat Rev Endocrinol
.
2015
;
11
(
9
):
535
-
546
.
20.
An
N
,
Khan
S
,
Imgruet
MK
, et al
.
Gene dosage effect of CUX1 in a murine model disrupts HSC homeostasis and controls the severity and mortality of MDS
.
Blood
.
2018
;
131
(
24
):
2682
-
2697
.
21.
Ghandi
M
,
Huang
FW
,
Jané-Valbuena
J
, et al
.
Next-generation characterization of the Cancer Cell Line Encyclopedia
.
Nature
.
2019
;
569
(
7757
):
503
-
508
.
22.
Zhang
Z
,
Golomb
L
,
Meyerson
M
.
Functional genomic analysis of CDK4 and CDK6 gene dependency across human cancer cell lines
.
Cancer Res
.
2022
;
82
(
11
):
2171
-
2184
.
23.
Sharma
KR
,
Colvis
CM
,
Rodgers
GP
,
Sheeley
DM
.
Illuminating the druggable genome: pathways to progress
.
Drug Discov Today
.
2024
;
29
(
3
):
103805
.
24.
Cerami
E
,
Gao
J
,
Dogrusoz
U
, et al
.
The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data
.
Cancer Discov
.
2012
;
2
(
5
):
401
-
404
.
25.
Yang
W
,
Soares
J
,
Greninger
P
, et al
.
Genomics of drug sensitivity in cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells
.
Nucleic Acids Res
.
2013
;
41
(
Database issue
):
D955
-
D961
.
26.
Abu Aboud
O
,
Chen
CH
,
Senapedis
W
,
Baloglu
E
,
Argueta
C
,
Weiss
RH
.
Dual and specific inhibition of NAMPT and PAK4 By KPT-9274 decreases kidney cancer growth
.
Mol Cancer Ther
.
2016
;
15
(
9
):
2119
-
2129
.
27.
Hasmann
M
,
Schemainda
I
.
FK866, a highly specific noncompetitive inhibitor of nicotinamide phosphoribosyltransferase, represents a novel mechanism for induction of tumor cell apoptosis
.
Cancer Res
.
2003
;
63
(
21
):
7436
-
7442
.
28.
Cea
M
,
Cagnetta
A
,
Fulciniti
M
, et al
.
Targeting NAD+ salvage pathway induces autophagy in multiple myeloma cells via mTORC1 and extracellular signal-regulated kinase (ERK1/2) inhibition
.
Blood
.
2012
;
120
(
17
):
3519
-
3529
.
29.
Del Nagro
C
,
Xiao
Y
,
Rangell
L
,
Reichelt
M
,
O'Brien
T
.
Depletion of the central metabolite NAD leads to oncosis-mediated cell death
.
J Biol Chem
.
2014
;
289
(
51
):
35182
-
35192
.
30.
Han
W
,
Xie
J
,
Li
L
,
Liu
Z
,
Hu
X
.
Necrostatin-1 reverts shikonin-induced necroptosis to apoptosis
.
Apoptosis
.
2009
;
14
(
5
):
674
-
686
.
31.
Jie
H
,
He
Y
,
Huang
X
, et al
.
Necrostatin-1 enhances the resolution of inflammation by specifically inducing neutrophil apoptosis
.
Oncotarget
.
2016
;
7
(
15
):
19367
-
19381
.
32.
Ortega-Forte
E
,
Hernández-García
S
,
Vigueras
G
, et al
.
Potent anticancer activity of a novel iridium metallodrug via oncosis
.
Cell Mol Life Sci
.
2022
;
79
(
10
):
510
.
33.
Mitchell
SR
,
Larkin
K
,
Grieselhuber
NR
, et al
.
Selective targeting of NAMPT by KPT-9274 in acute myeloid leukemia
.
Blood Adv
.
2019
;
3
(
3
):
242
-
255
.
34.
Jones
CL
,
Stevens
BM
,
Pollyea
DA
, et al
.
Nicotinamide metabolism mediates resistance to venetoclax in relapsed acute myeloid leukemia stem cells
.
Cell Stem Cell
.
2020
;
27
(
5
):
748
-
764.e4
.
35.
Subedi
A
,
Liu
Q
,
Ayyathan
DM
, et al
.
Nicotinamide phosphoribosyltransferase inhibitors selectively induce apoptosis of AML stem cells by disrupting lipid homeostasis
.
Cell Stem Cell
.
2021
;
28
(
10
):
1851
-
1867.e8
.
36.
Clarke
E
,
Pereira
C
,
Chaney
R
,
Woodside
S
,
Eaves
AC
,
Damen
J
.
Toxicity testing using hematopoietic stem cell assays
.
Regen Med
.
2007
;
2
(
6
):
947
-
956
.
37.
Volpe
DA
,
Warren
MK
.
Myeloid clonogenic assays for comparison of the in vitro toxicity of alkylating agents
.
Toxicol In Vitro
.
2003
;
17
(
3
):
271
-
277
.
38.
Lai
D
,
Chen
M
,
Su
J
, et al
.
PP2A inhibition sensitizes cancer stem cells to ABL tyrosine kinase inhibitors in BCR-ABL+ human leukemia
.
Sci Transl Med
.
2018
;
10
(
427
):
eaan8735
.
39.
Ray Chaudhuri
A
,
Nussenzweig
A
.
The multifaceted roles of PARP1 in DNA repair and chromatin remodelling
.
Nat Rev Mol Cell Biol
.
2017
;
18
(
10
):
610
-
621
.
40.
Gibson
AE
,
Yeung
C
,
Issaq
SH
, et al
.
Inhibition of nicotinamide phosphoribosyltransferase (NAMPT) with OT-82 induces DNA damage, cell death, and suppression of tumor growth in preclinical models of Ewing sarcoma
.
Oncogenesis
.
2020
;
9
(
9
):
80
.
41.
Imgruet
MK
,
Lutze
J
,
An
N
, et al
.
Loss of a 7q gene, CUX1, disrupts epigenetically driven DNA repair and drives therapy-related myeloid neoplasms
.
Blood
.
2021
;
138
(
9
):
790
-
805
.
42.
Koyen
AE
,
Madden
MZ
,
Park
D
, et al
.
EZH2 has a non-catalytic and PRC2-independent role in stabilizing DDB2 to promote nucleotide excision repair
.
Oncogene
.
2020
;
39
(
25
):
4798
-
4813
.
43.
Chang
A
,
Liu
L
,
Ashby
JM
, et al
.
Recruitment of KMT2C/MLL3 to DNA damage sites mediates DNA damage responses and regulates PARP inhibitor sensitivity in cancer
.
Cancer Res
.
2021
;
81
(
12
):
3358
-
3373
.
44.
Tasdogan
A
,
Kumar
S
,
Allies
G
, et al
.
DNA damage-induced HSPC malfunction depends on ROS accumulation downstream of IFN-1 signaling and bid mobilization
.
Cell Stem Cell
.
2016
;
19
(
6
):
752
-
767
.
45.
Piacente
F
,
Caffa
I
,
Ravera
S
, et al
.
Nicotinic acid phosphoribosyltransferase regulates cancer cell metabolism, susceptibility to NAMPT inhibitors, and DNA repair
.
Cancer Res
.
2017
;
77
(
14
):
3857
-
3869
.
46.
Guo
J
,
Lam
LT
,
Longenecker
KL
, et al
.
Identification of novel resistance mechanisms to NAMPT inhibition via the de novo NAD+ biosynthesis pathway and NAMPT mutation
.
Biochem Biophys Res Commun
.
2017
;
491
(
3
):
681
-
686
.
47.
Zheng
S
,
Wang
W
,
Aldahdooh
J
, et al
.
SynergyFinder plus: toward better interpretation and annotation of drug combination screening datasets
.
Genomics Proteomics Bioinformatics
.
2022
;
20
(
3
):
587
-
596
.
48.
Vazquez
F
,
Sellers
WR
.
Are CRISPR screens providing the next generation of therapeutic targets?
.
Cancer Res
.
2021
;
81
(
23
):
5806
-
5809
.
49.
Jiang
J
,
Yuan
J
,
Hu
Z
, et al
.
Systematic illumination of druggable genes in cancer genomes
.
Cell Rep
.
2022
;
38
(
8
):
110400
.
50.
Eldfors
S
,
Saad
J
,
Ikonen
N
, et al
.
Monosomy 7/del(7q) cause sensitivity to inhibitors of nicotinamide phosphoribosyltransferase in acute myeloid leukemia
.
Blood Adv
.
2024
;
8
(
7
):
1621
-
1633
.
51.
Kaufmann
KB
,
Garcia-Prat
L
,
Liu
Q
, et al
.
A stemness screen reveals C3orf54/INKA1 as a promoter of human leukemia stem cell latency
.
Blood
.
2019
;
133
(
20
):
2198
-
2211
.
52.
McNerney
ME
,
Brown
CD
,
Wang
X
, et al
.
CUX1 is a haploinsufficient tumor suppressor gene on chromosome 7 frequently inactivated in acute myeloid leukemia
.
Blood
.
2013
;
121
(
6
):
975
-
983
.
53.
Lecoeur
H
,
Prévost
MC
,
Gougeon
ML
.
Oncosis is associated with exposure of phosphatidylserine residues on the outside layer of the plasma membrane: a reconsideration of the specificity of the annexin V/propidium iodide assay
.
Cytometry
.
2001
;
44
(
1
):
65
-
72
.
54.
Wei
Y
,
Xiang
H
,
Zhang
W
.
Review of various NAMPT inhibitors for the treatment of cancer
.
Front Pharmacol
.
2022
;
13
:
970553
.
55.
Goldinger
SM
,
Gobbi Bischof
S
,
Fink-Puches
R
, et al
.
Efficacy and safety of APO866 in patients with refractory or relapsed cutaneous T-cell lymphoma: a phase 2 clinical trial
.
JAMA Dermatol
.
2016
;
152
(
7
):
837
-
839
.
56.
Korotchkina
L
,
Kazyulkin
D
,
Komarov
PG
, et al
.
OT-82, a novel anticancer drug candidate that targets the strong dependence of hematological malignancies on NAD biosynthesis
.
Leukemia
.
2020
;
34
(
7
):
1828
-
1839
.
57.
Zhang
P
,
Brinton
LT
,
Williams
K
, et al
.
Targeting DNA damage repair functions of two histone deacetylases, HDAC8 and SIRT6, sensitizes acute myeloid leukemia to NAMPT inhibition
.
Clin Cancer Res
.
2021
;
27
(
8
):
2352
-
2366
.

Author notes

Data are available on request from the corresponding author, Megan E. McNerney (megan.mcnerney@bsd.uchicago.edu).

The full-text version of this article contains a data supplement.