Abstract Background In the ongoing battle against BCR-ABL+ leukemia, despite significant advances with tyrosine kinase inhibitors (TKIs), the persistent challenges of drug resistance and the enduring presence of leukemic stem cells (LSCs) remain formidable barriers to achieving a cure. Methods In this study, we demonstrated that Disulfiram (DSF) induces ferroptosis to synergize with TKIs in inhibiting BCR-ABL+ cells, particularly targeting resistant cells and LSCs, using cell models, mouse models, and primary cells from patients. We elucidated the mechanism by which DSF promotes GPX4 degradation to induce ferroptosis through immunofluorescence, co-immunoprecipitation (CO-IP), RNA sequencing, lipid peroxidation assays, and rescue experiments. Results Here, we present compelling evidence elucidating the sensitivity of DSF, an USA FDA-approved drug for alcohol dependence, towards BCR-ABL+ cells. Our findings underscore DSF’s ability to selectively induce a potent cytotoxic effect on BCR-ABL+ cell lines and effectively inhibit primary BCR-ABL+ leukemia cells. Crucially, the combined treatment of DSF with TKIs selectively eradicates TKI-insensitive stem cells and resistant cells. Of particular note is DSF’s capacity to disrupt GPX4 stability, elevate the labile iron pool, and intensify lipid peroxidation, ultimately leading to ferroptotic cell death. Our investigation shows that BCR-ABL expression induces alterations in cellular iron metabolism and increases GPX4 expression. Additionally, we demonstrate the indispensability of GPX4 for LSC development and the initiation/maintenance of BCR-ABL+ leukemia. Mechanical analysis further elucidates DSF’s capacity to overcome resistance by reducing GPX4 levels through the disruption of its binding with HSPA8, thereby promoting STUB1-mediated GPX4 ubiquitination and subsequent proteasomal degradation. Furthermore, the combined treatment of DSF with TKIs effectively targets both BCR-ABL+ blast cells and drug-insensitive LSCs, conferring a significant survival advantage in mouse models. Conclusion In summary, the dual inhibition of GPX4 and BCR-ABL presents a promising therapeutic strategy to synergistically target blast cells and drug-insensitive LSCs in patients, offering potential avenues for advancing leukemia treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-024-02162-0. Keywords: Ferroptosis, Disulfiram, BCR-ABL+ leukemia, GPX4, Tyrosine kinase inhibitor Introduction Chronic myeloid leukemia (CML) and a subset of acute lymphoblastic leukemia (ALL), collectively known as Philadelphia chromosome-positive (Ph+) leukemias, are marked by a common pathogenic feature—the Philadelphia chromosome, which arises from the t(9;22)(q34;q11) translocation [[52]1]. This translocation leads to the formation of the fusion gene BCR::ABL (BCR-ABL), producing the dysregulated BCR-ABL tyrosine kinase that serves as the primary driver of leukemogenesis and remains the central focus of targeted therapies. The advent of tyrosine kinase inhibitors (TKIs), including imatinib mesylate (IM), dasatinib (DAS), and nilotinib (NL), has ushered in a transformative era in CML management. By specifically targeting BCR-ABL kinase activity, these TKIs have shifted CML from a once-fatal disease to a more manageable condition [[53]2]. Despite substantial clinical efficacy observed in chronic phase (CP) CML, achieving complete cure remains elusive, with only approximately 10% of CP CML patients able to discontinue TKI treatment and maintain therapy-free remission. Challenges persist in addressing reduced TKI efficacy during the accelerated phase and blast crisis (BC) of CML, along with prevalent primary and acquired resistance to these compounds. Similarly, BCR-ABL+ acute lymphoblastic leukemia (ALL), sharing resemblances with the aggressive lymphoid BC of CML, confronts comparable challenges of relapse under current TKI monotherapies [[54]1]. While TKIs have significantly advanced therapy management in BCR-ABL+ leukemia, providing controlled CML and long-term remission in BCR-ABL+ B-ALL when tailored to individual conditions, their limitations are evident. TKIs fall short of achieving complete cure for BCR-ABL+ leukemia due to their inability to eradicate cancer/leukemia stem cells (CSCs/LSCs), which harbor kinase-independent survival pathways [[55]3]. Additionally, genetic instability observed in BCR-ABL+ leukemias results in TKI-resistant point mutations in the BCR-ABL kinase domain and other genetic anomalies, constraining TKI efficacy. LSCs in BCR-ABL+ leukemias carrying the BCR-ABL fusion gene are deemed the root cause of cancer initiation, resistance, and relapse [[56]4–[57]6]. These LSCs operate independently of BCR–ABL for survival, underscoring the need to identify and target kinase-independent pathways [[58]3]. Thus, selective elimination of LSCs while sparing their normal counterparts may offer a potentially curative therapy against certain cancers, warranting further investigation. Ferroptosis, a form of regulated cell death (RCD), manifests as cell demise through intracellular deposition of a lethal amount of lipid reactive oxygen species (lip-ROS) mediated by iron ions [[59]7–[60]9]. The primary mechanism of ferroptosis involves perturbation of intracellular lipid redox reactions. Glutathione peroxidase 4 (GPX4) plays a pivotal role in thwarting lipid peroxidation and ferroptosis by clearing lipid peroxides [[61]10, [62]11]. Recent studies have highlighted the importance of iron metabolism in maintaining CSCs, noting that the high concentration of iron in CSCs compared to non-CSCs underlines their iron addiction and potential vulnerability to ferroptosis. Given their increased resistance to standard therapies and tendency for relapse, CSCs’ abnormal regulation of iron metabolism-related proteins and dependence on iron make them a promising target for innovative cancer treatments that induce ferroptosis cell death by disrupting their iron metabolism [[63]12, [64]13]. Ferroptosis has thus emerged as a promising therapeutic strategy for curbing tumor progression and overcoming drug resistance [[65]14]. Targeting GPX4 offers substantial potential to enhance the efficacy of chemotherapy, particularly against drug-resistant cancer cells, including CSCs [[66]15]. Several ferroptosis-inducing agents have been identified to date, including System Xc⁻ inhibitors (e.g., erastin and sorafenib) and GPX4 inhibitors (like RSL3). These agents, especially GPX4 inhibitors, are extensively utilized in preclinical cancer research and have demonstrated significant efficacy across various cancer models [[67]16, [68]17]. However, it is important to note that these compounds may also induce ferroptotic cell death in immune cells, potentially compromising the antitumor immune response [[69]18–[70]20]. Therefore, the development of cancer-specific ferroptosis inducers is crucial to mitigate adverse effects and enhance the therapeutic efficacy of immunotherapies. Disulfiram (DSF), a long-standing treatment for alcohol dependence, boasts well-established pharmacokinetics, safety, and tolerability at USA FDA-recommended doses [[71]21]. Previous studies have demonstrated the efficacy of DSF in preventing and treating diet-induced obesity [[72]22], as well as its ability to counteract cell apoptosis and septic cell death induced by lipopolysaccharide (LPS) in murine models [[73]21]. Notably, emerging research has highlighted DSF’s potent anti-inflammatory and anticancer properties [[74]23]. Our previous study has also revealed its efficacy against T-cell acute lymphoblastic leukemia (T-ALL), while indicating that it does not adversely affect normal T cells [[75]24]. These findings suggest DSF’s promise as an effective and safer therapeutic option for cancer treatment. In this study, we demonstrate that DSF, alone or in combination with TKIs, effectively eradicates malignant cells and LSCs of BCR-ABL+ leukemias, while exerting minimal effects on normal blood cells derived from healthy individuals (HIs). Mechanistically, DSF induces ferroptosis by disrupting the interaction between HSPA8 and GPX4, which facilitates STUB1-mediated ubiquitination and subsequent proteasomal degradation of GPX4. Our findings indicate that GPX4 plays a critical role in the anti-ferroptotic mechanisms that sustain blast cells and LSCs in BCR-ABL+ leukemia, positioning it as a promising therapeutic target for DSF in the eradication of LSCs. Materials and methods Clinical samples Peripheral blood (PB) or bone marrow (BM) samples were collected from patients diagnosed with CML, Philadelphia chromosome-positive B-cell acute lymphoblastic leukemia (BCR-ABL+ B-ALL) (see Supplemental Table S1), as well as from HIs. Samples were collected from the First Affiliated Hospital of Jinan University. Written informed consent was obtained from all participants before blood collection, and the study was approved by the ethical committee of the First Affiliated Hospital of Jinan University (No.2014006). Cell culture Mouse 32D cells were purchased from the China Center for Type Culture Collection (CCTCC). The human CML cell line KBM5, K562-R, and 32D-BCR-ABL were generously provided by Prof. Jingxuan Pan, Sun Yat-Sen University. Other hematological malignancy cell lines including U266, RPMI8226, H929, CCRF-CEM, Molt4, Jurkat, NB4, MOLM-13, KG1-α, HL-60, MV-411, K562, K562-R, Ku-812, TOM-1, Kasumi-1, SUP-B15 and 293T cells were provided by the Institute of Hematology, Jinan University. SUP-B15, U266, RPMI8226, H929, CCRF-CEM, Molt4, Jurkat, NB4, MOLM-13, KG1-α, Kasumi-1, HL-60, MV-411, K562, Ku-812, TOM-1 were cultured in 90% RPMI 1640 with 10% Gibco fetal bovine serum (FBS). K562-R cells were cultured with 5µM IM under the same conditions. KBM5, and 32D-BCR-ABL were cultured in 90% Iscove’s Modified Dulbecco’s Medium (IMDM) with 10% FBS. 293T cells were cultured in 90% DMEM with FBS. All cell lines were maintained in a humidified atmosphere containing 5% CO[2] at 37 °C. Plasmocin prophylactic was added to all media to prevent potential mycoplasma contamination. CD34 + cells were isolated using the CD34 Microbeads Kit (#130-046-702, Miltenyi Biotec, Germany) according to the manufacturer’s instructions and cultured in IMDM supplemented with 10% FBS along with SCF (#300-07-500, Proteintech; 100 ng/mL), IL-3 (#213-13-100, Proteintech; 20 ng/mL), IL-6 (#216-16-100, Proteintech, 20 ng/mL), and GM-CSF (#300-03-100, Proteintech; 100 ng/mL) at 37℃ in a humidified incubator with 5% CO[2]. Reagents Disulfiram (DSF; S1680), Imatinib (IM; S1026), Dasatinib (DAS; S1021), Z-VAD-FMK (S7023), N-acetylcysteine (NAC; S1623), Ferrostatin-1 (Fer-1; S7243), Deferoxamine (DFO; S5742), MG132 (S2619), Chloroquine (CQ; S6999), Cycloheximide (CHX; C7698), Vitamin E (Vit-E; S4686), VX-765 (S2228), MG132 (S2619) and Necrosulfonamide (NSA; S8251) were purchased from Selleck Chemicals (Houston, TX, USA). 4-Diethylaminobenzaldehyde (DEAB; HY-W016645) was purchased from MedChemExpress (Monmouth Junction, NJ, USA). Z-VAD-FMK, Fer-1, DFO, Vit-E, NSA were dissolved in DMSO and added to culture media at a final concentration of 10 µM. NAC was dissolved in DMSO and added to culture media at a final concentration of 1 µM. RNA interference KBM5 cells were transfected using the Neon^® Transfection System (Invitrogen) with 100 pmol of oligonucleotides in 10 µl reactions [[76]25]. Briefly, 2 × 10^5 cells were suspended in 100 pmol of siHSPA8, siALDHA1, or siGPX4 in 10 µl reactions. After electroporation, cells were cultured in IMDM medium containing 10% FBS at 37 °C with 5% CO[2]. The transfected cells were used for subsequent experiments after 24–48 h. siRNA sequences targeting HSPA8, STUB1, BCR-ABL and GPX4/Gpx4 are listed in Supplemental Table S2. In vivo siRNA delivery The preparation of amine-terminated, generation 5 polyamidoamine (G5-PAMAM, hereafter called G5) dendrimer-siRNA nanoparticles for use in mouse models was conducted according to the protocol established by Prof. Daoguang Yan from Jinan University. Briefly, in vivo siRNA delivery was conducted using G5 (CYD-150 A), obtained from Weihai Chenyuan Molecular New Materials Co., Ltd. The N: P ratio of G5 to siRNA was set at 30:1, and the complexes were formed in PBS at room temperature with gentle vortexing for 10 min. A dosage of 1.0 mg/kg of the G5-siRNA complex was administered via tail vein injection every 3 days. Establishment of stable cell lines A total of 3 µg of the PLKO-puro-NC or PLKO-puro-sh-GPX4 plasmid along with 1.5 µg of psPAX2 and 1.5 µg of pMD2.G were transfected into 293T cells using lipofectamine 3000 (Life Technologies, USA). Virus supernatant was harvested 24 and 48 h after transfection. Subsequently, KBM5 cells and mouse lineage- cells were co-cultured with virus supernatant. Then 5 µg/mL puromycin was applied to select positively infected cells for at least 10 days. shRNA sequences targeting GPX4/Gpx4 are listed in Supplemental Table S2. Lentiviruses overexpressing HSPA8 (LV-HSPA8, GV705), GPX4 (LV-GPX4, GOSL0438191), control lentivirus (LV-NC, CON525), Flag-tagged GPX4-WT (wild type), and Flag-tagged seven cysteines-mutated GPX4 (GPX4-7CA mutant) were purchased from Genechem (Shanghai, China). KBM5 cells were transduced with LV-HSPA8 or LV-GPX4 lentivirus, and positively transduced cells were selected by treatment with 5 µg/mL puromycin for a duration of 10 days. Cell viability assay Cell viability was assessed using the Cell Counting Kit-8 (CCK-8) (#96992, Sigma) following the manufacturer’s protocol. In brief, cancer cell lines were seeded at 10,000 cells per well, and primary cells at 50,000 cells per well, in 96-well plates. After treatment, 10 µl of CCK-8 reagent was added to each well and incubated for 4 h. The absorbance was then measured using a microplate reader (Biotek Synergy4, USA). Colony forming cells (CFC) assays GFP + lineage- c-Kit + cells sorted from BCR-ABL mice were plated in M3434 methylcellulose medium (Stem Cell Technologies). Cultures were incubated at 37 °C in a humidified atmosphere of 5% CO[2] for 10–14 days. For CFC assays using BCR-ABL+ leukemia cell lines, 1,000 cells were seeded into 24-well plates after a 24-hour drug treatment. Subsequently, the cells were cultured in IMDM supplemented with 2% FBS and Methylcellulose Stock Solution (HSC001, Bio-Techne). Cultures were incubated at 37 °C in a humidified atmosphere of 5% CO[2] for 10 to 14 days before counting. Lipid peroxidation and cell death analysis BCR-ABL+ leukemia cell lines, primary cells from patients or mouse primary cells were harvested and washed three times in PBS following various treatments. Lipid peroxidation and cell death analysis were using BODIPY-C11(D3861, Invitrogen) and Annexin-V-APC/PI kit (AP107-100, MultiSciences, China), and analyzed by flow cytometry. The labile iron pool assay The FeRhoNox-1 probe (MX4558, MKBio, China) was used to assess the labile iron pool. Briefly, a 500 µl cell suspension from each treatment group was seeded into triplicate wells of a 24-well plate. Subsequently, 2.5 µM FeRhoNox-1 probe was added to each well and incubated for 20 min at 37 °C. Flow cytometry was then conducted for analysis. Malondialdehyde (MDA) assay The relative concentration of malondialdehyde (MDA), an end product of lipid peroxidation, was measured using a Lipid Peroxidation Assay Kit (S0131M, Beyotime, China) following the manufacturer’s instructions. GSH and GSSG detection The GSH and GSSG levels of BCR-ABL+ leukemia cell lines or tissue from mice were measured using a GSH and GSSG Assay Kit (S0053, Beyotime, China) according to the manufacturer’s instructions. The total GSH and GSSG concentrations were calculated using a standard curve and normalized to the total protein level in each sample. Immunofluorescence analysis Cells were washed with cold phosphate-buffered saline (PBS), fixed in 4% paraformaldehyde for 15 min at room temperature, and permeabilized with 0.1% triton X100 for 10 min at room temperature. Cells were stained with GPX4 antibody (Santa Cruz, sc-166570; 1:100) or HSPA8 antibody (ab51052, Abcam; 1:200) at room temperature for 1 h. After washing 3 times with PBS, cells were incubated with the secondary antibody of Alexa Fluor 555 labeled goat anti-rabbit IgG or Alexa Fluor 488 labeled goat anti-mouse IgG at room temperature for 30 min. 4′,6-Diamidine-2′-phenylindole dihydrochloride (DAPI) (Thermo Fisher Scientific) was used to stain the nucleus. MitoTracker Red (M7512, Invirogen) was used to stain mitochondria. Images were captured using a Leica SP8 confocal microscope (Leica Corp, Germany). Western blot analysis Cells were harvested and lysed in RIPA buffer supplemented with protease inhibitors. Proteins were then separated using a 12% SDS-PAGE Criterion X-gel (Bio-Rad) and transferred to a PVDF membrane (Bio-Rad). The membranes were blocked with QuickBlock™ Blocking Buffer (Beyotime, China) and incubated overnight at 4 °C with primary antibodies against GPX4 (#59735S, Cell Signaling Technology; ab125066, Abcam; 1:1000), HSPA8 (#8444, Cell Signaling Technology; 1:1000), Ubiquitin (ab134953, Abcam; 1:2000), and Flag (AF0036, Beyotime; 1:1000). The anti-β-actin (ACTB) antibody (A3853, SIGMA; 1:5000) was used as a control. Bands were visualized using enhanced chemiluminescence (ECL; Beyotime, China) and the UVITEC photo documenter. Coimmunoprecipitation (CO-IP) Cell protein extracts were isolated using Pierce IP Lysis Buffer (Thermo Fisher Scientific), then, cell lysates were incubated specific antibody (Anti-GPX4, sc-166570, Santa Cruz, USA) at 4 °C overnight. Dynabeads Protein A/G beads (CXB-PA/G0004-1, EeasunBio, Guangzhou) were incubated with the obtained protein complex for 6 h at 4 °C. The magnetic bead-antibody-antigen complex was washed three times with IP buffer, then eluted with loading buffer and heated at 100 °C for 10 min. The samples were resolved by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and subsequently analyzed by immunoblotting. RNA extraction and quantitative real-time RT-PCR Total RNA from BCR-ABL+ cell lines or primary cells was extracted with Trizol reagent (Invitrogen) according to the manufacturer’s instructions. RNA was reverse transcribed into cDNA using the High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems, CA, USA). Real-time PCR was conducted using SYBR Green (Tiangen, China), following a cycling program of 45 cycles at 95 °C for 30 s, 60 °C for 30 s, and 72 °C for 30 s. Relative expression levels were determined using the comparative 2-ΔΔCt method, with data normalized to ACTB mRNA levels. The primers used for real-time PCR analysis are listed in Supplemental Table S3. RNA-seq analysis KBM5 cells were treated with DMSO (Ctrl) or 0.25 µM DSF, both in the absence and presence of 0.25 µM IM for 48 h. Total RNA was extracted and assessed for RNA integrity using the RNA Integrity Number (RIN). cDNA library construction and sequencing were performed by Shanghai Biotechnology Corporation using the VAHTS Stranded mRNA-seq Library Prep Kit for Illumina^®. High-quality reads were aligned and mapped to the human reference genome (GRCh38) using Hisat2 (version 2.0.4). Gene expression levels were calculated in terms of fragments per kilobase of exon model per million mapped reads (FPKM) and are provided in Supplemental Table S4. Flow cytometric analysis BM and spleen cells harvested from recipient mice were analyzed by flow cytometry (BD LSRFortessa) following staining with the specified antibodies. For the detection of leukemia myeloid cells, cells were stained with anti-mouse Gr-1-APC (E-AB-F1120UE, Elabscience), anti-mouse Mac-1-PE (E-AB-F1081UD, Elabscience), or anti-human CD45-FITC (#368508, BioLegend). To identify lineage-negative Sca-1 + c-Kit+ (LSK) cells, cells were stained with lineage-PerCP-Cy5.5 (#561317, BD), Sca-1-APC (160904, BioLegend), and c-Kit-BV421 (#567818, BD). For the identification of long-term hematopoietic stem cells (LT-HSCs) and short-term hematopoietic stem cells (ST-HSCs), cells were stained with lineage-PerCP-Cy5.5 (#558074, BD), Sca-1-APC (#17-5981-82, eBioscience), c-Kit-BV421 (#105828, BioLegend), CD135-PE (#12-1351-83, eBioscience), CD150-PE-Cyanine7 (#25-1502-82, eBioscience), and CD48-APC-eFluor780 (#47-0481-82, Invitrogen). The gating strategies used for flow cytometry analysis are detailed in Supplemental Fig. S1A. Molecular docking The molecular structures necessary for docking studies were obtained from the Protein Data Bank ([77]https://www.rcsb.org/) and AlphaFold ([78]https://alphafold.ebi.ac.uk/). For molecular docking predictions on the Windows platform, AutoDock 4.2.6 and PyMOL 2.5.4 were employed, while AutoDock Vina and GRAMM (Global RAnge Molecular Matching, [79]https://gramm.compbio.ku.edu/) were utilized on the Ubuntu system. KBM5 mice sh-NC or sh-GPX4 KBM5 cells (2 × 10^6 cells/mouse) were transplanted into B-NDG mice via tail vein injection to establish CML-CDX mouse models (n = 8). B-NDG mice were procured from Biocytogen (Beijing, China). Seven days post-administration of KBM5 cells, flow cytometry using a BD LSRFortessa was employed to assess the expression of human CD45 + cells. The mice were subsequently divided into three groups: sh-NC, sh-GPX4-1, and sh-GPX4-2. At day 28, three mice were randomly selected from each group, and the weight and size of the spleen (n = 3) were recorded. Tumor burden in the spleen and bone marrow of each group (n = 3) was assessed, and the survival times of the remaining mice in each group (n = 5) were documented. The animal experimental protocols were approved by the Laboratory Animal Ethics Committee of Jinan University (Approval No. 20230812-12). K562 mice K562 cells stably expressing both GFP and luciferase (K562-GL, 5 × 10^6 cells/mouse, IV) were transplanted into B-NDG mice via tail vein injection to establish CML-CDX mouse models (n = 5). B-NDG mice were obtained from Biocytogen (Beijing, China). Bioluminescence imaging was performed using the IVIS imaging system one week post-injection of K562 cells, following luciferin administration. The mice were then randomly assigned to one of four treatment groups: a control group receiving vehicle, an IM monotherapy group (100 mg/kg/day), a DSF monotherapy group (100 mg/kg/day), and a combined treatment group (100 mg/kg/day IM + 100 mg/kg/day DSF). Tumor burden progression was monitored weekly via bioluminescence imaging, and the mice were observed throughout the study period to evaluate survival outcomes. The animal experimental protocols were approved by the Laboratory Animal Ethics Committee of Jinan University (Approval No. 20211109-19). BCR-ABL mice The Scl-tTa-BCR-ABL1/GFP (hereafter referred to as BCR-ABL) mice were provided by Prof. Weizhang Wang, Guangdong Pharmaceutical University. FVB-N mice were purchased from HFK Bio-Technology (Beijing, China). To evaluate the therapeutic potential of DSF and TKIs, BM cells were harvested from BCR-ABL mice after a 4-week induction of BCR-ABL expression via tetracycline withdrawal. BM GFP + cells (1 × 10^6 cells/mouse, IV) were transplanted into lethally irradiated wild-type FVB/N recipient mice, which received 900 cGy of radiation. Blood samples were collected 4 weeks post-transplantation to confirm the onset of leukemia. The mice were then subjected to various treatment regimens: IM (100 mg/kg/day, oral gavage), DSF (100 mg/kg/day, oral gavage), a combination of DSF and IM, or a control group receiving only a vehicle (0.5% CMC-NA). After 4 weeks of treatment, a subset of animals from each cohort was euthanized for post-treatment analysis, while the remaining mice were monitored for survival. To assess the impact of DSF and TKIs treatment on LSC burden, 1 × 10^6 BM GFP + cells from treated mice were transplanted into secondary (2nd) recipient mice, and engraftment rates were monitored. To evaluate the antileukemic efficacy of GPX4 deletion in maintaining BCR-ABL+ leukemia, 1 × 10^6 BM GFP + cells from 4-week Tet-off BCR-ABL mice were transplanted into lethally irradiated primary recipients. These mice were divided into two groups and treated with G5-siNC or G5-siGpx4 for 2 weeks. Circulating blasts and LSKs were assessed, and survival outcomes were compared between the two groups. To further investigate the antileukemic efficacy of GPX4 deletion during leukemogenesis, 5 × 10^5 preleukemic lineage-c-Kit + cells were sorted and transplanted into lethally irradiated recipient mice. BCR-ABL mice received G5-siGpx4 (1 mg/kg/day, IV) or G5-siNC (1 mg/kg/day, IV) for 4 weeks, starting the day after transplantation and Tet-off BCR-ABL induction. Circulating blasts, LSKs, and survival were compared between the two groups. The animal experimental protocols were approved by the Laboratory Animal Ethics Committee of Jinan University (Approval Nos. 20230313-06, 20230602-13, 20230920-01, and 20240709-01). Statistical analysis All statistical analyses were conducted in R language software (version 3.8.2, [80]https://www.r-project.org/) and GraphPad Prism (version 8.0, CA, USA), as appropriate. Differences in two independent subgroups in clinical samples were tested by Mann-Whitney-Wilcoxon. For comparisons between two groups in vivo and in vitro experiments, two-tailed unpaired or paired Student’s t test was utilized. For comparisons among three or more groups, a one-way ANOVA followed by Bonferroni’s post hoc test was used. To compare a control group with multiple experimental groups, a one-way ANOVA with Dunnett’s post hoc test was applied, unless otherwise specified. The “surv_cutpoint” function in the “survminer” package was used to determine the optimal cut-off values of GPX4. Kaplan-Meier curves were compared by log-rank test using the R package “survival”. The differentially expressed genes in RNA-seq data were identified utilizing the package “edgeR”. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were analyzed by the packages “clusterProfiler” and “org.HS.eg.db”. A two-tailed P value < 0.05 was considered statistically significant. Result The combination of DSF and TKIs synergistically promotes cell death in BCR-ABL+ leukemia cells DSF, a well-established treatment for alcohol dependence for over seven decades, exhibits established pharmacokinetics, safety, and tolerability at doses recommended by the USA FDA. Recent studies unveil DSF’s remarkable anti-cancer properties [[81]26]. To evaluate its impact on blood cancer, we subjected various cancer cell lines to varying DSF concentrations, revealing a dose-dependent inhibition of cancer cell growth. Notably, BCR-ABL+ cells exhibited heightened sensitivity to DSF (Fig. [82]1A). Subsequently, two BCR-ABL+ cell lines, KBM5 (CML) and TOM-1 (BCR-ABL+ B-ALL), demonstrated significant induction of cell death by DSF in a dose-dependent manner (Fig. [83]1B). DSF also markedly impeded colony number and size in KBM5 and TOM-1 cells (Fig. [84]1C). Further investigations explored DSF’s potential to enhance cytotoxic effects in cells from newly diagnosed BCR-ABL+ leukemia patients, consistently promoting cell death in primary BCR-ABL+ leukemia cells (Fig. [85]1D). To discern DSF’s anti-leukemia effects in cells harboring BCR-ABL relative to BCR-ABL- cells, we included BCR-ABL1-transformed 32D-BCR-ABL and parental mouse hematopoietic 32D cells. The results indicated more pronounced cell death in 32D-BCR-ABL cells compared to 32D cells under equivalent concentrations (Fig. S1B), emphasizing DSF’s robust cytotoxic effect on BCR-ABL+ leukemia cells. Fig. 1. [86]Fig. 1 [87]Open in a new tab DSF cooperates with TKIs to induce the cell death of BCR-ABL+ leukemia cells. A A panel of BCR-ABL+ leukemia cells and cancer cell lines were treated with increasing concentrations of DSF for 48 h and the cell viability was determined by CCK-8 assay (left panel), and the IC50 was calculated by CompuSyn 2.0 (right panel). B BCR-ABL+ leukemia cell lines were treated with the indicated concentrations of DSF (left panel), and the percentage of Annexin V + cells was detected by the flow cytometry (right panel). C Effect of DSF on the colony-forming ability of BCR-ABL+ leukemia cells. Representative results are shown in the left panel, with quantification from three experiments displayed in the right panel. D BM MNCs from newly diagnosed patients with CML (n = 6) or BCR-ABL+ B-ALL (n = 6) were exposed to indicated concentrations of DSF for 48 h, and the percentage of Annexin V + cells was detected by the flow cytometry. E Heatmaps of drug combination responses were generated using the online SynergyFinder software ([88]https://www.synergyfinder.fimm.fi/), assessing the synergistic effects of DSF + TKIs in KBM5 and TOM-1 cells. Synergistic effects were indicated by scores over 0, and those surpassing 10 suggested strong synergistic effects. In these heatmaps, red signals synergism, while green represents antagonism. The drug combinations exhibiting the most potent synergistic effects are highlighted with white squares (left panel). KBM5 and TOM-1 cells were treated with DSF alone, TKIs alone, or the combination of DSF and TKIs at the indicated concentrations for 48 h and the cell viability was measured by CCK8 assay (right panel). F KBM5 and TOM-1 cells were treated with DSF (0.25 µM) alone, IM (0.25 µM) for KBM5 cells, DAS (0.25 µM) for TOM-1 cells, or the combination of DSF and TKIs at the indicated time points, and cell viability was measured by CCK8 assay(two-way ANOVA test, data are presented as mean ± SD). G KBM5 (upper panel) and TOM-1 cells (lower panel) were treated with 0.25 µM DSF, 0.25 µM TKIs or the combination of DSF and TKIs, and the percentage of Annexin V + cells was determined by flow cytometry. H Representative image and quantification of the colony-forming ability of BCR-ABL+ leukemia cells treated with DSF (0.25 µM), with or without IM (0.25 µM). I Cell death in Ku-812 and SUP-B15 cells with indicated treatment (0.25 µM DSF, 0.25 µM TKIs or the combination of DSF and TKIs for 48 h) was evaluated by flow cytometry labeling with Annexin-V and PI. J-L Cell viability of BM MNCs from newly diagnosed patients with CML (n = 10) or BCR-ABL+ B-ALL (n = 5) or HI PB (n = 3) with indicated treatment (0.25 µM DSF, 0.25 µM TKIs or the combination of DSF and TKIs) for 48 h were measured by CCK-8 assay (J) and flow cytometry(K-L). Data are expressed as the mean ± SD. n = 3 or more independent biological replicates, * P  < 0.05; ** P  < 0.01**** P  ≤ 0.0001 Given that TKIs represent the current first-line therapy for BCR-ABL+ leukemia, we investigated whether DSF synergized with TKIs in BCR-ABL+ leukemia cells. We chose DAS for TOM-1 cells (BCR-ABL+ B-ALL) due to its ability to overcome various mechanisms of imatinib resistance and its enhanced activity against BCR-ABL. For KBM5 cells (CML), we selected IM, as it remains the standard treatment and is effective in achieving cytogenetic responses. Treatment of KBM5 and TOM-1 cells with TKIs alone or in combination with low dose DSF (0.25µM) induced a synergistic reduction in cell activity (Fig. [89]1E). Further experiments involving time gradients of the drugs consistently demonstrated a synergistic effect between DSF and TKIs (Fig. [90]1F-G). Importantly, combination treatment led to increased cell death and notable reduction in colony number and size (Fig. [91]1H). Similar findings were observed in two additional BCR-ABL+ cell lines, KU-812 and SUP-B15 (Fig. [92]1I and Fig. S1C). This synergistic effect was minimally observed in normal human PB mononuclear cells (MNCs) but significantly affected BM MNCs from BCR-ABL+ leukemia patients (Fig. [93]1J-L). Collectively, these results underscore the synergistic effect between DSF and TKIs. DSF induces BCR-ABL+ leukemia cell ferroptosis To delve deeper into the potential pathways underlying the effects of DSF in BCR-ABL+ leukemia cells, we analyzed the enriched pathways of differentially expressed genes in KBM5 cells treated with Ctrl (DMSO), IM, DSF, or IM + DSF for 48 h (Fig. [94]2A). Comparing the DSF + IM treated cells with the Ctrl, we observed significant enrichment changes in the oxidative stress pathway and glutathione pathway (Fig. [95]2B, left panel). Furthermore, comparing the DSF + IM treated cells with the IM treated cells, we found significant changes in pathway enrichment related to ferroptosis, suggesting that DSF may induce cell death in the KBM5 cell line through the ferroptosis pathway (Fig. [96]2B, right panel). To confirm these results, we utilized ferroptosis inhibitors Fer-1, Vit-E, and DFO, oxidative stress inhibitors NAC, and cell death inhibitors NSA, VX-765, and z-VAD-FKM on DSF-induced cell death. Notably, DSF-induced cell viability reduction could be rescued by Fer-1, Vit-E, DFO, and NAC (Fig. [97]2C), but not by Z-VAD-FMK, NSA, VX-765, indicating that DSF indeed induces leukemic cell death through the ferroptosis and oxidative stress pathways. Fig. 2. [98]Fig. 2 [99]Open in a new tab DSF induces BCR-ABL+ leukemia cell ferroptosis. A Schema of the experimental design to define the pathway in DSF-treated BCR-ABL+ leukemia cells. B Bubble diagram of KEGG pathway enrichment analysis was performed in KBM5 cells. The top 16 pathways most significantly changed in DSF + IM group compared with Ctrl group (left panel). The top 12 pathways most significantly changed in DSF + IM group compared with IM group (right panel). C The inhibitory effects of DSF (0.25 µM), IM (0.25 µM), or the combination of DSF and IM on KBM5 cells were assessed in the presence of the indicated inhibitors using the CCK-8 assay. D Fluorescence intensities of BODIPY-C11 in DSF treated KBM5 and TOM1 cells by flow cytometry. E The MDA in cell lysates of KBM5(up panel) and TOM-1(lower panel) were treated with the indicated concentrations of DSF for 36 h. F-G KBM5 and TOM-1 were treated with 0.25 µM DSF, 0.25 µM TKIs, or a combination of DSF and TKIs for 48 h. Fluorescence intensities of BODIPY-C11 (F) and ROS level (G) were determined by flow cytometry. H The MDA concentration of BM MNCs from newly diagnosed patients with CML treated with DSF (0.25 µM) or TKIs (0.25 µM) alone, or a combination of DSF and TKIs for 48 h. I The primary cells from HIs, CML patients and BCR-ABL+ B-ALL patients were treated with DSF (0.25 µM) alone, TKIs (0.25 µM) alone, or a combination of DSF and TKIs for 48 h. The levels of lipid peroxidation were determined by flow cytometry using the BODIPY-C11 probe. Representative results are shown in (left panel), and quantification are shown in (right panel). Data are expressed as the mean ± SD. n = 3 or more independent biological replicates, * P  < 0.05; ** P  < 0.01, *** P  ≤ 0.001, **** P  ≤ 0.0001 Moreover, we assessed key biochemical events of ferroptosis, including lipid peroxidation measured by fluorescence probes BODIPY-C11 and malondialdehyde (MDA), a product of lipid peroxidation, in the KBM5 and TOM-1 cell lines with graded doses of DSF. We observed a dose-dependent increase in lipid peroxidation and MDA (Fig. [100]2D-E). Flow cytometry analysis of lipid peroxidation also showed an increase in DSF-induced ferroptosis after combined treatment with TKIs (Fig. [101]2F), accompanied by an increase in reactive oxygen species (ROS) (Fig. [102]2G), MDA (Fig. S1D) and a decrease in GPX4 enzymatic activity (Fig. S1E). These results further underscore the crucial role of ferroptosis in DSF-induced cell death in BCR-ABL+ cell lines. Furthermore, we measured MDA levels in primary BM cells from CML patients treated with DSF or in combination with IM. Consistent with the cell line results, DSF combined with IM treatment led to increased MDA in CML cells (Fig. [103]2H). Additionally, we analyzed lipid peroxidation levels and found that the combination of DSF and IM induced lipid peroxidation in BCR-ABL+ leukemia cells, while having no significant impact on HIs (Fig. [104]2I). Interestingly, compared with BCR-ABL+ cell lines, DSF had no impact on lipid peroxidation of acute myeloid leukemia (AML) cells, consistent with previous studies demonstrating that DSF induces apoptosis in AML cells (Fig. S1F). These data collectively indicate that DSF induces ferroptosis in BCR-ABL+ leukemia cells. DSF induces ferroptosis by repressing GPX4 Subsequently, we investigated the expression of GPX4, closely associated with ferroptosis, in BCR-ABL+ cells following DSF treatment. While mRNA expression showed no significant changes (Fig. [105]3A), protein levels of GPX4 decreased in response to DSF alone or in combination with TKIs in KBM5 and TOM-1 cells (Fig. [106]3B). Moreover, DSF in combination with IM led to a significant reduction in GPX4 protein levels in a time-dependent manner (Fig. S2A). Confocal microscopy analysis shows that cytosolic GPx4 (cGPx4) is the key isoform dominantly expressed relative to mitochondrial GPx4 (mGPx4) and nucleolar GPx4 (nGPx4), and further confirmed decreased GPX4 protein levels in DSF and DSF + IM-treated KBM5 cells (Fig. [107]3C and Fig. S2B). Compared to BCR-ABL+ cells, we found that DSF treatment does not induce GPX4 repression in multiple myeloma cell lines with low sensitivity to DSF (Fig. S2C). These findings suggest that DSF may induce BCR-ABL+ cell ferroptosis through GPX4 suppression. Fig. 3. [108]Fig. 3 [109]Open in a new tab DSF induce ferroptosis through GPX4 targeting. A The mRNA level of GPX4 in RNA-seq data and the FPKM values were used to compare differences in gene expression among samples. B The protein level of GPX4 in BCR-ABL+ leukemia cell lines treated with the indicated concentrations of DSF, or DSF (0.25 µM) alone, TKIs (0.25 µM) alone, or a combination of DSF and TKIs for 48 h. C Representative immunofluorescence images of GPX4 protein in KBM5 cells treated with DSF (0.25 µM) alone, IM (0.25 µM) alone, or a combination of DSF and IM for 48 h. Nuclei are stained with DAPI (×10). Scale bars, 5 μm. D Western blot assay of GPX4 and ACTB proteins in KBM5 cells transfected with sh-GPX4 or its control sh-NC. E The colony formation assay was utilized to detect cell proliferation of KBM5 cells transfected with sh-GPX4 or its control sh-NC. F-H KBM5 cells transfected with sh-GPX4 were treated with 0.25 µM IM for 48 h, Cell viability (F), cell death (G), and fluorescence intensities of BODIPY-C11(H) were analyzed using the CCK8 assay or flow cytometry. I Western blot analysis of the GPX4 expression in GPX4-overexpressing KBM5 cells. Cell viability (J), cell death analysis (K), and fluorescence intensities of BODIPY-C11 (L) of GPX4-overexpressing KBM5 cells treated with 0.25 µM DSF or 0.25 µM IM for 48 h. Data are expressed as the mean ± SD. n  = 3 or more independent biological replicates, * P  < 0.05; ** P  < 0.01, *** P  ≤ 0.001, **** P  ≤ 0.0001 To explore the relationship between GPX4 and DSF, we generated KBM5 cell lines with GPX4 knockdown (KD) (Fig. [110]3D). Depletion of GPX4 markedly reduced the clonogenic potential of KBM5 cells in colony-forming assays (Fig. [111]3E), significantly impaired their proliferative capacity (Fig. [112]3F), and heightened ferroptosis in response to IM treatment (Fig. [113]3G-H). Consistent with the effect of GPX4 depletion, the combination of the ferroptosis inducer Fin56 and TKIs more effectively inhibited BCR-ABL+ leukemia cells than each treatment alone (Fig. S3A-C). Finally, we overexpressed GPX4 in KBM5 cell lines (Fig. [114]3I), and GPX4-overexpressing KBM5 cells demonstrated resistance to growth inhibition and ferroptosis by IM and DSF (Fig. [115]3J-L). The effect of DSF has previously been shown to ALDH1A1. Next, we also explored whether ALDH1A1 mediates DSF induced ferroptosis. Our results indicate that pharmacological and genetic ALDH1A1 inhibition does not significantly enhance the effects of GPX4 KD or DSF treatment on KBM5 or leukemic stem cells (Fig. S4A-C). Collectively, our data reveal that DSF induces ferroptosis by suppressing GPX4 expression. DSF sensitized TKI resistant cells and leukemia stem cells to TKIs Despite the continuous improvement in efficacy with the use of second-generation and third-generation TKIs, resistance to TKIs, disease progression, and recurrence due to resistant cells or leukemic stem cells (LSCs) remain formidable challenges in CML treatment. To address this, we investigated whether the combination of DSF and TKIs could overcome TKI resistance in cell lines. Consequently, we employed CCK8 and flow cytometry to assess changes in cell viability and cell death in IM resistant cells K562-R following the combination of DSF and IM. The results unveiled a clear synergistic effect of the two drugs on IM-resistant K562-R cells (Fig. [116]4A-B). Additionally, we measured changes in GPX4 protein levels and lipid peroxidation after combination treatment, indicating that the combination of DSF and IM could induce ferroptosis in these cells (Fig. [117]4C-D). Furthermore, sorted CD34 + cells from newly diagnosed CML patients were treated with IM and DSF. The combination of the two drugs increased cell death, inhibited cell viability, and augmented lipid peroxidation in CML CD34 + cells (Fig. [118]4E-G and Fig. S4D). Fig. 4. [119]Fig. 4 [120]Open in a new tab Dual BCR-ABL and GPX4 inhibition more effectively targets IM-resistant cells and LSCs than either treatment alone. A-D Prior to the experiment, K562-R cells were cultured in an IM-free medium for 48 h. IM-resistant cells were treated with IM (5 µM) alone, DSF (0.25 µM) alone, or a combination of DSF and IM for 48 h (A-C) or 56 h (D). Their responses were assessed using CCK8, western blot and flow cytometry to measure cell viability (A), cell death (B), the protein levels of GPX4 (C), and fluorescence intensities of BODIPY-C11 (D). E-G CD34 + cells from CML patients were treated with IM (0.25 µM) alone, DSF (0.25 µM) alone, or a combination of DSF and IM for 48 h. The effects were evaluated using CCK8 and flow cytometry to assess cell viability (E), cell death (F), and fluorescence intensities of BODIPY-C11 (G). H-I Mouse LSK cells from BCR-ABL mice 4 weeks after induction of BCR-ABL expression were treated with IM (0.25 µM) alone, DSF (0.25 µM) alone, or a combination of DSF and IM. Their responses were evaluated using CCK8 (H) and colony formation assays (I). Data are expressed as the mean ± SD. n = 3 or more independent biological replicates, * P  < 0.05; ** P  < 0.01, *** P  ≤ 0.001, **** P  ≤ 0.0001 To further validate this discovery, we utilized the stem cell promoter (Scl)-driven transgenic Scl-tTa-BCR-ABL1 (hereafter referred to as BCR-ABL) mouse model. Withdrawal of tetracycline (Tet) induces BCR-ABL expression and the development of BC CML-like disease in these mice. Mouse LSCs (LSK cells) were sorted from Tet-off BCR-ABL mice, revealing a dramatic decrease in cell viability and clonogenic potential when LSK cells were treated with IM in combination with DSF (Fig. [121]4H-I). Overall, these results indicate that the combination of DSF and TKIs can effectively promote the death of drug-resistant and DSF-insensitive cells, including drug-insensitive LSCs, in BCR-ABL+ leukemia. GPX4 is highly expressed in BCR-ABL+ leukemia, and its elevated expression is associated with a poor prognosis in BCR-ABL+ ALL Given GPX4’s essential role in inhibiting ferroptosis, our data demonstrate that DSF induces ferroptosis and decreases GPX4 expression. Based on this, we hypothesized that BCR-ABL+ leukemia cells may rely on high GPX4 levels. To investigate, we compared GPX4 protein levels in PB MNCs from CML patients, BCR-ABL+ B-ALL patients, and HIs. We found elevated GPX4 protein levels in primary BCR-ABL+ leukemia samples (Fig. [122]5A-B), consistent with observations in BCR-ABL+ leukemia cell lines compared to HIs (Fig. [123]5C). Additionally, CD34 + cells from CML patients exhibited higher GPX4 levels compared to those from HIs (Fig. [124]5D). Furthermore, in the BCR-ABL mouse model, we observed increased Gpx4 expression in BM MNCs (Fig. [125]5E, left and middle panel) and LSK cells (Fig. [126]5E, right panel) from Tet-off BCR-ABL mice. Consistently, Gpx4 was increased in 32D-BCR-ABL(p210) cells compared to their parental 32D cells (Fig. [127]5F). Overexpression of BCR-ABL1(p190) also led to increased Gpx4 protein levels (Fig. S5A), while BCR-ABL KD attenuated GPX4 expression at both mRNA and protein levels (Fig. S5B). These results suggest that BCR-ABL promotes GPX4 expression and contributes to BCR-ABL+ leukemia development. Fig. 5. [128]Fig. 5 [129]Open in a new tab GPX4 is highly expressed in BCR-ABL+ leukemia cells and is associated with a poor prognosis in BCR-ABL+ B-ALL. A-B Protein levels of GPX4 were assessed in primary cells from CML (A) and BCR-ABL+ B-ALL leukemia (B) patients and HIs. C GPX4 protein levels were examined in BCR-ABL+ leukemia cell lines, and primary cells from HIs. D RNA levels of GPX4 were evaluated in CD34 + cells obtained from HIs and CML patients. E Expression of GPX4 in cells from BCR-ABL mice before or after tetracycline withdrawal. Representative western blot (left panel) and immunofluorescence images (middle panel) of GPX4 protein in BM cells. RNA levels of GPX4 in LSK cells from BCR-ABL mice were also examined (right panel). F Protein levels of GPX4 were measured in 32D and 32D-BCR-ABL cells (left panel). Representative immunofluorescence images of GPX4 in these cells were analyzed (right panel). G Levels of lipid reactive oxygen species were determined in PB MNCs from HIs and CML patients. H Fluorescence intensities of BODIPY-C11 in 32D and 32D-BCR-ABL cells following treatment with IM (0.25 µM) alone, DSF (0.25 µM) alone, or a combination of DSF and IM for 48 h were measured. I Kaplan‒Meier analysis was performed to evaluate the associations of GPX4 expression with overall survival (OS) in patients with BCR-ABL+ B-ALL in the [130]GSE34941-GP11088. The cutoff value is 13.9. J GPX4 expression levels in 477 newly diagnosed BCR-ABL+ CML patients and 74 patients receiving TKI treatment more than 1 year. Data are expressed as the mean ± SD. n = 3 or more independent biological replicates, * P < 0.05; ** P < 0.01, *** P ≤ 0.001, **** P ≤ 0.0001 Concurrent with high GPX4 expression, CML patient BM MNCs exhibited lower lipid peroxidation levels compared to HIs, indicating ferroptosis inhibition (Fig. [131]5G). This observation was consistent in 32D-BCR-ABL(p210) and 32D cells (Fig. S5C-D). Notably, CML leukemic cells displayed significantly increased Fe2 + levels (Fig. S5E). Next, we examined the effect of DSF on BCR-ABL+ cell death. DSF alone or in combination with IM markedly induced cell death (Fig. S5F) and lipid peroxidation (Fig. [132]5H) in 32D-BCR-ABL cells with high Gpx4 expression, but not in their parental 32D cells. Next, we further investigated the relationship between GPX4 and the prognosis of BCR-ABL+ B-ALL patients. Analysis of RNA-seq data from bone marrow samples in the [133]GSE34941-GP11088 dataset revealed a notable trend: high GPX4 expression was associated with poor overall survival (OS) in BCR-ABL+ B-ALL patients (Fig. [134]5I). Although this association did not achieve statistical significance, it may be influenced by the limited sample size. Furthermore, we examined the correlation between GPX4 expression levels and clinical outcomes in BCR-ABL+ CML patients using the [135]GSE76312 dataset. The results indicated a significant decrease in GPX4 expression in CML patients after ≥ 1 year of TKI treatment (Fig. [136]5J). Collectively, these findings suggest that elevated GPX4 expression is significantly linked to adverse clinical outcomes in both BCR-ABL+ B-ALL and CML patients. Targeting GPX4 compromises BCR-ABL+ leukemia initiation/progression The results presented above demonstrate the elevated levels of GPX4 in BCR-ABL+ leukemia and the inhibitory effect of GPX4 KD on ferroptosis induction and leukemia cell growth in vitro. Subsequently, we investigated the role of GPX4 in a xenograft mouse model (Fig. [137]6A). Consistent with our previous findings, GPX4 KD via shRNAs led to a substantial reduction in the number of CML leukemia cells in the BM (Fig. [138]6B). Notably, compared to the sh-NC group, GPX4 KD resulted in significantly decreased spleen size and weight (Fig. [139]6C), ultimately leading to prolonged survival in recipient mice (Fig. [140]6D). Fig. 6. [141]Fig. 6 [142]Open in a new tab GPX4 is essential for LSC development and the maintenance of BCR-ABL+ leukemia. A Schema of the experimental strategy to define the dependence of CML cells on GPX4 in the maintenance of leukemia. (n = 8 mice per group). B Flow cytometry analysis of the CD45 + cell in the BM of CML mice. C Representative images of spleen. Statistical analysis of spleen weight is presented. D Kaplan–Meier survival curves showing effect of GPX4 KD on leukemia maintenance in immunodeficient mice. subsequent to sh-GPX4 KBM5 cell transplantation (n = 5). E Schematic overview of G5-siGpx4 nanoparticle therapy of BCR-ABL+ leukemia mice. (n = 8 mice per group for this experiment). F The frequency of LSKs, LT-HSCs, and ST-HSCs was analyzed by flow cytometry. G-H Mac-1 + Gr1 + myeloid cells in spleen (G) and BM (H). I-L Survival (I), spleen weight (J), the ratio of GSH and GSSG (K), MDA (L), and CFC (M) of the G5-siGpx4-treated vs. G5-siNC-treated mice are shown. N-R Experimental design (N). To assess the impact of G5-siGpx4 treatment on the LSC development, lineage-c-Kit + cells from BCR-ABL mice (BCR-ABL were induced for two days by Tet-off) were transplanted into recipient mice, starting on the day after transplantation, mice were treated G5-siGpx4 or G5-siNC. Spleen weight (O), Mac-1 + Gr1 + myeloid cells (P), LSK cells (Q), Survival (R), the ratio of GSH/GSSG (S) and MDA(T) of the G5-siGpx4-treated vs. G5-siNC-treated mice are shown. Data are expressed as mean ± SD from three or more independent biological replicates. Statistical significance is denoted as * P < 0.05; ** P < 0.01; *** P ≤ 0.001; **** P ≤ 0.0001 We then examined whether GPX4 deletion impacts BCR-ABL+ leukemia maintenance in the BCR-ABL mouse model (Fig. [143]6E). For gene delivery, we utilized amine-terminated, G5 (G5-PAMAM) dendrimers. After transplantation of cells from 4-week Tet-off BCR-ABL mice into lethally irradiated primary recipients, the mice were divided into two groups and treated with G5-siNC or G5-siGpx4, followed by an assessment of leukemia cell engraftment in the BM. As anticipated, a profound reduction in LSCs and quiescent LSCs (Fig. [144]6F) was observed in the BM of mice treated with G5-siGpx4 nanoparticles compared with G5-siNC. Notably, the siGpx4 nanoparticles significantly decreased myeloid cells (Gr-1 + Mac-1+) in the spleen (Fig. [145]6G) and BM (Fig. [146]6H), ultimately prolonging the survival of recipient mice (Fig. [147]6I). Additionally, compared with the G5-siNC group, the spleen size and weight were significantly decreased in the G5-siGpx4 group (Fig. [148]6J), accompanied by reduced leukemia cell infiltration in spleen and BM (Fig. S6A). Moreover, reduced Gpx4 protein levels were observed in the spleens of G5-siGpx4 group mice compared with the control group (Fig. S6B). Consistent with the in vitro findings, G5-siGpx4 nanoparticles-mediated Gpx4 KD resulted in high levels of malondialdehyde (MDA) and low levels of reduced glutathione (GSH) in the spleen and BM (Fig. [149]6K-L). Moreover, Gpx4 KD significantly inhibited colony-forming capacity in BM cells from Tet-off BCR-ABL mice (Fig. [150]6M). To explore the requirement for Gpx4 in leukemogenesis, we employed the BCR-ABL mouse model (Fig. [151]6N). In this model, BM preleukemic lineage-c-kit + cells were sorted and transplanted into lethally irradiated recipient mice. Withdrawal of Tet led to the induction of BCR-ABL expression, generating self-renewing LSCs, and causing BCR-ABL+ leukemia. Compared with the G5-siNC group, the spleen size and weight were significantly decreased in the G5-siGpx4 group (Fig. [152]6O), accompanied by reduced myeloid cells (Gr-1 + Mac-1+, Fig. [153]6P) and LSCs (LSK cells, Fig. [154]6Q). Additionally, preleukemic cells treated with G5-siGpx4 exhibited a significantly longer latency period for the onset of leukemia compared to controls (Fig. [155]6R). Consistently, the anti-leukemia effects of G5-siGpx4 nanoparticles were associated with a substantial decrease in GSH levels (Fig. [156]6S) and an increase in MDA levels (Fig. [157]6T) in the spleen and BM. Moreover, in the presence of DSF, GPX4 depletion led to the lowest survival rate of LSCs, as indicated in Fig. S6C-D. Collectively, our data suggest that GPX4 is required for the maintenance and initiation/development of BCR-ABL+ leukemia. DSF promotes GPX4 degradation by inhibiting HSPA8 binding to GPX4 We then delved into the molecular mechanism by which DSF regulates the protein level of GPX4. To explore the impact of DSF on the expressions of GPX4 upon CHX treatment, we observed a significant decrease in GPX4 protein half-life in KBM5 cells treated with DSF (Fig. [158]7A). Furthermore, we utilized MG132 (a proteasome pathway inhibitor) and CQ (an autophagy inhibitor) to treat CML cells with or without DSF treatment. Our findings revealed that MG132 could reverse the effect of DSF on GPX4 protein degradation, while CQ did not (Fig. [159]7B). Co-immunoprecipitation (Co-IP) assays revealed substantial polyubiquitination of GPX4 in DSF-treated cells (Fig. [160]7C), indicating that DSF modulates GPX4 protein stability via the ubiquitin-proteasome pathway. Utilizing UbiBrowser ([161]http://ubibrowser.bio-it.cn/ubibrowser/), we identified several potential interactors (HSPA8 and STUB1) of GPX4 (Fig. S7A). Given that HSPA8 is regulated by BCR-ABL and plays a vital role in leukemia cell survival [[162]27], we explored whether HSPA8 affects GPX4 expression. Molecular docking analysis suggested a potential interaction between HSPA8 and GPX4 (Fig. [163]7D). Co-IP analysis further indicated the GPX4-HSPA8 interaction, which was disrupted upon DSF treatment (Fig. [164]7E). Previous research has demonstrated that DSF binds to multiple cysteine residues on GPX4, disrupting its interaction with HSC70 (HSPA8) and thereby preventing erastin-induced autophagy-mediated GPX4 degradation [[165]28]. In line with these findings, we observed that GPX4 with seven cysteine site mutations reduced DSF’s effects. However, while prior studies indicated that this mutation impaired DSF’s ability to block erastin-induced autophagy-mediated degradation of GPX4, our results suggest that it instead inhibits DSF-induced ubiquitin-mediated degradation of GPX4 (Fig. [166]7F). To assess whether HSPA8 regulates the post-translational expression of GPX4, we downregulated HSPA8 in KBM5 cells and observed a significant decrease in GPX4 protein levels (Fig. [167]7G). HSPA8 KD also led to GPX4 suppression, which was reversible by MG132 (Fig. [168]7H), suggesting that HSPA8 influences GPX4 expression via the ubiquitin-proteasome pathway. Conversely, HSPA8 overexpression increased GPX4 protein levels (Fig. [169]7I) and significantly reduced GPX4 polyubiquitination, enhancing GPX4 protein stability even after DSF treatment (Fig. [170]7J-K). As seen with GPX4 KD, HSPA8 KD significantly induced cell death and lipid peroxidation in DSF-treated KBM5 cells (Fig. S7B). To explore if DSF might inhibit GPX4 through HSPA8, we knocked down GPX4 in KBM5 cells overexpressing HSPA8 (Fig. [171]7L). In these cells, HSPA8 overexpression led to reduced cell death and lipid peroxidation upon GPX4 KD (Fig. [172]7M). Notably, HSPA8 expression did not decrease with DSF treatment, indicating that DSF’s inhibitory effect on GPX4 may not involve HSPA8 downregulation (Fig. S7C-D). Recent studies have identified STUB1 as a novel ubiquitin E3 ligase targeting GPX4 [[173]29]. In line with this, Co-IP analysis confirmed the interaction between GPX4 and STUB1, which was enhanced by DSF treatment (Fig. [174]7N). Silencing STUB1 increased GPX4 levels (Fig. [175]7O) and suppressed DSF-induced cell death and lipid peroxidation (Fig. S7E-F). Furthermore, STUB1 KD significantly inhibited GPX4 ubiquitination (Fig. [176]7P). Immunoblot analysis following GPX4 IP revealed that elevated HSPA8 levels reduced the interaction between GPX4 and STUB1 (Fig. [177]7Q). These findings suggest that DSF modulates GPX4 stability via HSPA8, promoting STUB1-mediated GPX4 ubiquitination and proteasomal degradation. Fig. 7. [178]Fig. 7 [179]Open in a new tab GPX4 degradation and Ferroptosis induced by DSF was dependent on HSPA8. A KBM5 cells were treated with CHX alone (20 µM) or in combination with DSF (0.25 µM) or MG132 (0.25 µM). The protein level of GPX4 was assayed by western blots. B Western blot assay of GPX4 in KBM5 cells treated with DMSO, DSF (0.25 µM), or MG132 (0.25 µM) and CQ (25 µM) for 48 h. C Anti-Ubiquitin immunoblotting assay of GPX4 polyubiquitination in KBM5 cells treated with DSF (0.25 µM, 36 h) or control. D Molecular docking predicted a direct interaction between HSPA8 and GPX4. E Co-immunoprecipitation (Co-IP) of HSPA8 and GPX4 proteins in the whole cell lysates of KBM5 cells with or without DSF treatment (0.25 µM, 36 h). F GPX4-reconstituted cells were treated with 0.25 µM DSF for 46 h, and cells were collected for Western blot analysis. G qPCR analysis of HSPA8 (left panel) expression in KBM5 cells transfected with siHSPA8 or siNC. Western blot analysis of GPX4(right panel). H Western blot analysis of GPX4 expression in KBM5 cells transfected with siNC, siHSPA8-1, or siHSPA8-2 and treated with DMSO or MG132 (0.25 µM, 48 h). I qPCR analysis of HSPA8 (left panel) expression in HSPA8-overexpressing KBM5 cells. Western blot analysis of GPX4 (right panel) expression in these cells is also shown. J Western blot analysis of GPX4 expression in KBM5 cells transduced with LV-NC or LV-HSPA8 and treated with DSF or control. K The effect of HSPA8 on the ubiquitination modification of GPX4. L Western blot analysis of GPX4 expression in LV-NC or LV-HSPA8 transfected KBM5 cells following GPX4 KD. M Cell death (left panel) and fluorescence intensities of BODIPY-C11 (right panel) in LV-NC or LV-HSPA8 transfected KBM5 cells following GPX4 KD are displayed. N Co-IP of STUB1 and GPX4 proteins in the whole cell lysates of KBM5 cells with or without DSF treatment. O qPCR analysis of STUB1 (upper panel) expression in KBM5 cells transfected with siSTUB1 or siNC. Western blot analysis of GPX4(lower panel). P The effect of STUB1 KD on the ubiquitination modification of GPX4. Q The effect of HSPA8 on the STUB1-GPX4 binding. Data are expressed as mean ± SD from three or more independent biological replicates The combination of DSF and TKI exerts synergistic anti-leukemic effects in vivo The aforementioned results underscore the anti-leukemia efficacy of DSF alone or in combination with TKIs in BCR-ABL+ leukemia, indicating their potential to induce ferroptosis. To further evaluate their therapeutic potential, we investigated their effects in BCR-ABL+ leukemia mouse models. Firstly, K562 Xenograft models were established (Fig. [180]8A), revealing significantly smaller leukemia burden and prolonged survival in mice treated with the combination of DSF and TKIs compared to those treated with DSF or TKIs alone (Fig. [181]8B-C). Subsequently, BCR-ABL mice were utilized to assess the therapeutic potential of DSF and TKIs (Fig. [182]8D). Treatment with DSF and DSF + IM led to a notable reduction in spleen size and weight compared to control mice (Fig. [183]8E), with evident morphological improvements in BM and spleen (Fig. S8A-B). Additionally, reduced Gpx4 protein levels were observed in the spleens of mice treated with DSF plus IM compared to control and IM treatment alone (Fig. S8C). Notably, populations of myeloid leukemic cells in BM and spleen were significantly diminished in CML mice treated with DSF, further reduced in those treated with the combination of DSF and IM (Fig. [184]8F). Moreover, a lower proportion of LSK and LT-HSCs cells were observed upon treatment with DSF alone or in combination with IM (Fig. [185]8G-H). DSF treatment alone and in combination with TKIs significantly prolonged survival in murine models (Fig. [186]8I). In line with our findings, the IM and DSF combination group showed elevated levels of MDA, along with reduced levels of GSH and GPX4 (Fig. [187]8J-L). To assess the impact of combination treatment on the LSC burden, we transplanted 1 × 10^6 BM cells from mice treated with either DSF alone or DSF plus IM into 2nd recipients (Fig. S8D). The recipients of BM from DSF plus IM treated donors exhibited significantly smaller leukemia burden (Fig. S8E), decreased size and weight of the spleens (Fig. S8F). These findings collectively demonstrate that the combination of DSF and TKIs markedly exerts cytotoxicity against BCR-ABL+ leukemia cells, including stem/progenitor cells, compared to individual treatments alone. Fig. 8. [188]Fig. 8 [189]Open in a new tab Enhanced anti-leukemia activity in BCR-ABL+ leukemia by the combination of DSF and TKIs in vivo. A Schematic of xenotransplantation assay with K562 cells and B-NDG immune-deficient recipient mice. (n = 5 mice per group for this experiment). B Representative in vivo bioluminescent images of NRGS recipient mice xenotransplanted with GFP+/luciferase + K562 (K562-GL) cells treated with IM and DSF, either alone or in combination. C Kaplan–Meier survival curve following treatments with vehicle, TKIs alone, DSF alone, or a combination of DSF and TKIs (n = 5 mice per group). D Experimental design using the BCR-ABL transgenic mouse model. Treatment with DSF, IM, or DSF in combination with IM or vehicle alone (Ctrl) was initiated after transplantation and continued for 4 weeks. E-L A group of mice was sacrificed and analyzed after Treatment, and the rest for survival analysis. Images and weights of spleen (E), Mac-1 + Gr1 + myeloid cells (F), frequency of LSKs(G), LT-HSCs/ST-HSCs cells (H), survival (I), the ratio of GSH and GSSG (J), MDA (K), and representative immunofluorescence images of GPX4 protein (L) from each group are shown. M A summary of regulatory mechanisms that are significantly affected by the interaction of DSF and TKIs is shown. Data are presented as mean ± SD from three or more independent biological replicates Discussion BCR-ABL+ leukemia is characterized by the presence of small populations of LSCs, which pose a significant challenge to achieving long-term remission and cure. While BCR-ABL TKIs like IM, NL, and DAS induce remission by targeting CML progenitors, they fail to eliminate LSCs, which persist despite prolonged therapy, potentially leading to disease relapse. Hence, there is a pressing need for improved therapeutic approaches targeting CML LSCs [[190]30]. Our study offers compelling evidence for the therapeutic potential of DSF in BCR-ABL+ human leukemia. We demonstrate that DSF selectively eliminates BCR-ABL+ cell lines and effectively inhibits primary BCR-ABL+ human leukemia cells. In both in vivo and in vitro settings, DSF combined with TKIs eradicates malignant cells and LSCs in BCR-ABL+ leukemias. Our findings address the persistent challenges of drug resistance and LSC persistence, which hamper efforts towards achieving a cure. Ferroptosis, a regulated form of cell death, can be induced by factors such as increased reactive iron ions and disrupted iron pools. Various cellular pathways, including the Xc- system-GSH-GPX4 axis, NAD(P)H/FSP1/CoQ10 axis, and GCH1/BH4/DHFR axis, can suppress ferroptosis by inhibiting lipid peroxidation [[191]7]. Ferroptosis has been associated with cancer therapy resistance, and inducing ferroptosis has been shown to reverse drug resistance [[192]31]. DSF has been reported to exert potent anti-leukemic effects through various cell death mechanisms, including NETosis, pyroptosis, apoptosis, ferroptosis, and cuproptosis [[193]21, [194]23, [195]24, [196]26]. In our study, we demonstrate that DSF alone or combined with TKIs induces alterations in the ferroptosis pathway, suggesting that induction of ferroptosis contributes to its anti-leukemic effects. Indeed, the effects of DSF alone or combined with TKIs on BCR-ABL+ leukemia cells can be reversed by various ferroptosis inhibitors. Our data reveal that DSF induces ferroptosis by suppressing GPX4 expression, with this effect being most pronounced when combined with TKI treatment. Importantly, TKI treatment alone is insufficient to induce ferroptosis, likely due to only a modest reduction in GPX4 levels. In contrast, DSF exerts a more potent inhibitory effect on GPX4, and the combination of both agents leads to a synergistic depletion of GPX4, resulting in higher levels of ferroptosis (Fig. [197]8M). Recent studies have also shown that GPX4 plays a crucial role in counteracting apoptosis [[198]11]. It is therefore possible that the synergy between IM and DSF stems from IM-induced apoptosis—potentially facilitated in part by GPX4 suppression—alongside DSF-induced ferroptosis. Furthermore, our preclinical studies indicate that the combination of DSF and TKI is more effective in suppressing BCR-ABL+ leukemia cells than either agent used alone. GPX4 serves as a central regulator of ferroptosis [[199]32]. We find that GPX4 is overexpressed in BCR-ABL+ leukemia compared to normal controls. Furthermore, DSF exerts its cytotoxic effects on BCR-ABL+ cells by disrupting GPX4 stability, ultimately leading to ferroptotic cell death. This novel finding uncovers a previously unknown vulnerability in BCR-ABL+ cells that can be targeted for therapeutic purposes. Mechanistically, our findings indicate that DSF primarily promotes the ubiquitin-mediated degradation of GPX4 while simultaneously inhibiting its interaction with HSPA8. Previous study has documented an interaction between these proteins, suggesting that DSF disrupts the HSPA8-GPX4 binding, which in turn reduces erastin-induced GPX4 degradation [[200]28]. This discrepancy in results may stem from differences in the cellular reliance on distinct degradation pathways; prior research has shown that erastin induces autophagic degradation in HT1080 cells, wherein HSC70 (HSPA8) serves as an adaptor protein for chaperone-mediated autophagy [[201]33, [202]34]. In this context, the application of DSF disrupts the GPX4-HSPA8 interaction, thus inhibiting erastin-induced GPX4 degradation. In contrast, our study reveals that in BCR-ABL+ leukemia, GPX4 degradation is predominantly mediated via the ubiquitination pathway. DSF not only inhibits the GPX4-HSPA8 interaction but also facilitates GPX4’s ubiquitin-mediated degradation. Notably, DSF treatment enhances GPX4 ubiquitination, while overexpression of HSPA8 effectively inhibits this process. Importantly, a recent study supports the role of HSPA8 in stabilizing GPX4 expression, thereby enhancing its protein levels in liver cancer cells [[203]35]. Although this study did not explore the underlying mechanisms, we propose that GPX4 in liver cancer cells may similarly depend on ubiquitin-mediated degradation. Furthermore, recent investigations have identified STUB1 as a novel ubiquitin E3 ligase targeting GPX4 [[204]29]. Indeed, our results show that while DSF inhibits the binding of HSPA8 to GPX4, it concurrently enhances the interaction between GPX4 and STUB1. This shift suggests that upon DSF treatment, GPX4 preferentially associates with STUB1, thereby promoting its degradation (Fig. [205]8M). Additionally, we confirmed that STUB1 plays a crucial role in regulating GPX4 degradation and cell death in BCR-ABL+ leukemia cells. Collectively, these findings illuminate the complex interplay between GPX4, HSPA8, and STUB1, underscoring the potential of targeting these interactions in therapeutic strategies for BCR-ABL+ leukemias. Inhibition of GPX4 has been shown to sensitize therapy-resistant cancer cells to treatment [[206]15, [207]36]. Our study provides evidence that GPX4 is the target of DSF in inducing ferroptosis in BCR-ABL+ leukemia cells. We demonstrate that GPX4 is crucial for LSC development, transformation, and maintenance in BCR-ABL+ leukemia. As a pivotal regulator of ferroptosis, GPX4 plays a crucial role in maintaining cellular integrity [[208]37–[209]39], as well as in hematopoietic system [[210]40, [211]41]. Nevertheless, dietary supplementation with vitamin E has been shown to protect hematopoietic stem and progenitor cells (HSPCs) from ferroptosis-induced damage in Gpx4-depleted mice [[212]42], thereby safeguarding the functional integrity of the hematopoietic system. Conversely, inhibition of GPX4 specifically impairs the development and propagation of LSCs in mice fed natural ingredient diets, highlighting its potential as a therapeutic target in BCR-ABL+ leukemia mouse models, as demonstrated in this study. DSF has garnered attention for its potent anti-tumor activity across various cancer types, coupled with low toxicity [[213]43, [214]44]. In contrast to normal tissues, cancer cells typically exhibit elevated levels of GPX4, rendering them more tolerant to lipid reactive oxygen species (ROS) accumulation. Hence, there arises a possibility that DSF-mediated inhibition of GPX4 could enhance ferroptosis specifically in leukemic cells while sparing normal tissues. This represents a promising avenue for cancer treatment. Our findings lend support to the hypothesis that DSF treatment alone or in combination with TKIs significantly diminishes the burden of LSCs, suppresses disease progression, and enhances survival rates in murine models, underscoring its therapeutic potential in BCR-ABL+ leukemia. In conclusion, our study presents a promising therapeutic strategy for targeting drug-insensitive LSCs in BCR-ABL+ leukemia. Dual inhibition of GPX4 and BCR-ABL with DSF and TKIs, respectively, holds potential for addressing minimal residual disease and improving outcomes in preclinical mouse models. Further clinical trials are warranted to validate these findings and optimize the dosing and administration of DSF in combination with TKIs. Supplementary Information [215]Supplementary Material 1.^ (903KB, pdf) [216]Supplementary Material 2.^ (4.5MB, xlsx) [217]Supplementary Material 3.^ (2.5MB, docx) Acknowledgements