Introduction

Tyrosine kinase inhibitors (TKIs) represent the frontline treatment against chronic myeloid leukemia (CML), characterized by the t(9:22) chromosomal translocation, resulting in the breakpoint cluster region-Abelson (BCR-ABL)1 fusion oncogene with constitutively activated Abl tyrosine kinase activity. Despite TKI efficacy, resistant mutants turn CML into a controllable yet persisting chronic disease, interfering with the patient’s quality of life [1,2,3]. Moreover, one-third of newly diagnosed patients develop primary or secondary resistance toward approved TKIs, and clinical relapse occurs [4]. Imatinib resistance originates from BCR-ABL-dependent and -independent mechanisms, including leukemia stem cell (LSC) treatment insensitivity. LSCs and TKI/therapy-resistant cells depend on mitochondrial metabolism and oxidative phosphorylation (OXPHOS) to meet their energy demands [5, 6], in contrast to mature CML cells or normal hematopoietic stem cells (HSCs).

The goal of therapeutic approaches for CML treatment is to eliminate LSCs by identifying and targeting genetic alterations and molecular pathways that contribute to their survival and resistance to apoptosis. Efforts must be undertaken to develop combination therapies targeting tyrosine kinases with novel drug candidates that specifically target genes or molecules necessary for the survival mechanisms of CML LSCs [7]. Accordingly, identifying and targeting rewired energy metabolism is a relevant strategy for develo** non-ABL-related therapeutic options for treating TKI-resistant CML patients, preventing the appearance of TKI-resistant cells, and LSC persistence.

Nicotinamide adenine dinucleotide (NAD) is a substrate for NAD+-dependent enzymes and a major coenzyme in bioenergetic processes. The NAD+/NADH ratio modulates metabolic processes, including oxidative phosphorylation (OXPHOS), tricarboxylic acid (TCA) cycle, fatty acid oxidation (FAO), and glycolysis. NADH serves as a central hydride donor to mitochondrial ATP synthesis. Maintaining NAD+ levels and the NAD+/NADH ratio is crucial for mitochondrial function, ATP production, and homeostasis. Hence, NAD metabolism is a master regulator linking metabolic processes to OXPHOS [8]. Most cells replenish their NAD+ pool primarily via the NAD+ salvage pathway by recycling nicotinamide (NAM), generated by NAD+-consuming enzymes predominantly by poly (ADP-ribose) polymerase 1 (PARP1) and sirtuin (SIRT)1 [9, 18].

In addition to the cytotoxic activity, whether by apoptosis or controlled necrosis, the activation of the immune-mediated destruction of tumor cells is deeply involved in the curative effect of a given anticancer agent. There is a growing interest in promoting anti-tumor immune responses as a potential alternative strategy for cancer treatment [19]. Accordingly, the induction of immunogenic cell death (ICD) is an attractive approach to fighting cancer malignancies [20]. Pharmacological mediators of ICD stimulate the host’s immune system via the release of damage‐associated molecular patterns (DAMPs), eventually triggering a persistent immune response [21].

We investigated a TKI-independent, immunogenic cell death mechanism triggered by a novel, methylated indolequinone (MAC681). This compound targets PARP1 degradation, exacerbating mitochondrial dysfunction and inducting a non-canonical, necroptotic-like cell death. We used MAC681-treated, dying, or dead cells to demonstrate immunogenic vaccination against myeloid leukemia.

Materials and methods

Computational analysis of public CML datasets

This study utilized several publicly available datasets, which were processed as follows:

  1. A)

    GSE5550 [22]: 17 patients’ bone marrow samples sorted for CD34+ hematopoietic stem and progenitor cells, 8 healthy donors, and 9 untreated CML patients in chronic phase were downloaded from GEO. The samples were processed and normalized using the robust Multi-Array Average (RMA) expression measure from the “affy” package in R [23]. All the probes were analyzed separately; if multiple probes representing the same gene were present, only the most significant probe was kept.

  2. B)

    GSE97562 [24] and GSE47927 [25]: A total of 19 patients’ bone marrow samples sorted for CD34+, 8 healthy donors, and 11 untreated CML patients in chronic phase raw CEL files were downloaded from GEO (Supp. Table S1A). Samples were processed and normalized using the RMA expression measure from the “affy” package in R [23]. To correct for the batch effect between datasets, we employed the combat function of the "sva" R package [26].

  3. C)

    The Microarray Innovations in LEukemia (MILE) study, GSE13159 [27]: A total of 66 untreated CML patients’ bone marrow mononuclear cells raw CEL files were downloaded from GEO. Samples were processed and normalized using the robust RMA expression measure from the “affy” package in R [23]. Samples were divided into 4 groups based on the quartiles of PARP1 expression. The high and low quartiles were subsequently used for the differential expression analysis.

  4. D)

    The Cancer Cell Line Encyclopedia (CCLE) dataset: Gene expression transcript per million (TPM) normalized reads from all the cancer cell lines (n = 1165) were downloaded from the Depmap portal website (https://depmap.org/portal/download/all/) [28].

Differential expression analysis

To identify differentially expressed genes (DEGs) between healthy and CML CD34+ cells or between CML patients with high and low expression of PARP1, we utilized the “limma” R package [29]. We applied a threshold of |log2(FC)|> 0.3 and a false discovery rate (FDR) < 0.05. The resulting DEGs were visualized using volcano and generated using the “ggplot2” R package [30] (https://ggplot2.tidyverse.org).

Pathways and enrichment analysis

We performed functional enrichment analysis using GO terms, GSE analysis (GSEA), and Reactome pathways to analyze the identified DEGs comprehensively. GO terms and GSEA analysis were performed using “fgsea” and “msigdb” R packages. Reactome enrichments were performed on the web-based application [31] (https://reactome.org/). Pathways were considered significant when the FDR ≤ 0.05. We re-grouped each pathway for its higher hierarchical annotation in Reactome and calculated a weighted percentage of pathways enriched. Genes belonging to the NAD+ metabolome pathway were collected from **ing on the nucleus (24 h), a hallmark of necroptosis, are seen (top panel). Mitochondrial alterations (lower panel) induced by MAC681 are also visualized. G Time-dependent induction of mitochondrial membrane potential loss (left panel) and cell death induction (right panel) by MAC681 (5 µM). MMP was analyzed after incubation with MitoTracker Red (100 nM) by flow cytometry. Mitochondrial oxidative phosphorylation uncoupler CCCP (50 µM, 20 min) was used as a positive control. Flow cytometry analyzed cell death induction by propidium iodide (PI) uptake after 15 min staining with PI (0.5 µg/mL). DMSO (50%, 1 min) was used as a positive control (one-way ANOVA, Dunnett's multiple comparisons test *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001). H Cells were treated with MAC681 (5 µM, 6 h) and analyzed by fluorescence microscopy after staining with Rhod2-AM (Rhod, 2.5 µM), MitoTracker Green (MTG, 100 nM), and Hoechst. The intensity of the Rhod2-AM signal was quantified in regions stained with MitoTracker Green, and the co-occurrence and correlation were quantified by Manders’, Spearman’s, and Pearson’s coefficients. Double positive cells were analyzed, and the average of three independent experiments was shown (double-sided unpaired t-test ***p ≤ 0.001). I Cytosolic Ca2+ levels of cells treated with MAC681 (5 µM) for indicated time points, stained by Fluo4-AM, were measured by flow cytometry (double-sided unpaired t-test *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001). J Cytosolic Ca2+ levels of cells treated with MAC681 (5 µM) for indicated time points in the presence of EGTA (650 µM), followed by TSG (10 nM) treatment, were measured by Fluo4-AM every 15 s by flow cytometry, (AUC, double-sided unpaired t-test **p ≤ 0.01, ***p ≤ 0.001). K Colony formation assay with K-562 cells treated with MAC681 at the indicated concentrations. Images are representative of three independent experiments (One-way ANOVA, Dunnett's multiple comparisons test **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001). L In vivo zebrafish xenografts model of tumor mass formation after injection of fluorescent CellTracker™ CM-DiI Dye-stained K-562 cells pre-treated for the indicated time. M MAC681 decreases the viability of CML patients’ PBMCs in dose- and time-dependent manners. CML primary cells were treated ex-vivo with indicated concentrations of MAC681, and cell viability was evaluated after 4, 12, and 24 h by trypan blue exclusion test. The histogram corresponds to the mean ± SD (n = 2 CML patients). N Analysis of ALDH activity in K-562 cells. Cells were treated with MAC681 (5 µM, 12 h) or Imatinib (0.25 µM, 24 h), and the ALDH inhibitor diethylaminobenzaldehyde (DEAB) was used to distinguish cell subpopulations with low and high ALDH activity. Representative dot plots showing the percentage of ALDH-positive cells and corresponding quantifications (right panel) of three independent experiments are presented (left panel) (one-way ANOVA, Tukey’s multiple comparisons test *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001)