ABSTRACT
Acute lymphocytic leukemia (ALL) is the most common leukemia in children, with the T-cell subtype (T-ALL) accounting for 15% of those cases. Despite advancements in the treatment of T-ALL, patients still face a dismal prognosis following their first relapse. Relapse can be attributed to the inability of chemotherapy agents to eradicate leukemia stem cells (LSC), which possess self-renewal capabilities and are responsible for the long-term maintenance of the disease. Mitochondria have been recognized as a therapeutic vulnerability for cancer stem cells, including LSCs. Mitocans have shown promise in T-ALL both in vitro and in vivo, with some currently in early-phase clinical trials. However, due to challenges in studying LSCs in T-ALL, our understanding of how mitochondrial function influences self-renewal remains limited. This review highlights the emerging literature on targeting mitochondria in diverse T-ALL models, emphasizing specific mitochondrial vulnerabilities linked to LSC self-renewal and their potential to significantly improve T-ALL treatment.
KEYWORDS: Mitochondria, OXPHOS, metabolism, self-renewal, leukemia stem cells, cancer stem cells, T-ALL
1. Introduction
Leukemia is the most prevalent pediatric cancer, representing one-third of all childhood cancers. Acute lymphocytic leukemia (ALL) accounting for more than 80% of those cases.1 T-cell acute lymphoblastic leukemia (T-ALL), characterized by the malignant transformation of T-lymphocytes precursor cells, represents 10–15% of pediatric ALL.2
The treatment of ALL has seen significant advancements; a once fatal cancer with a survival rate of only 11% in the 1960s3 now boasts a 90% cure rate for B-cell ALL (B-ALL).4 However, cure rates of T-ALL patients remain lower, at approximately 72%.4,5 Following the first relapse, T-ALL patients often develop multi-drug resistance and become refractory to second-line treatments.6 These patients face a grim outlook. Relapsed T-ALL patients have a 5-year overall survival (OS) rate of 20–40%,7 with a median OS of just 4–5 months.8
Relapse in leukemia is often attributed to the failure of cytotoxic chemotherapy agents to eradicate leukemia stem cells (LSCs), which may be quiescent or have inherent drug-resistance mechanisms.9 The cancer stem cell (CSC) hypothesis posits that a small subset of cells within a tumor possesses an indefinite ability to self-renew, is responsible for the long-term maintenance of tumor growth, and can potentially repopulate the cancer from just a single cell.10 Research into LSCs in T-ALL has advanced significantly. Leukemia-initiating activity being identified in a distinct subpopulation of cells in patient samples,11 in T-ALL mouse12 and zebrafish models.13 These cells can induce leukemia after transplantation to a secondary host, retain the immunophenotypic characteristics of the primary leukemia, and maintain the ability to self-renew after serial transplantation.14,15 Consequently, the eradication of LSCs holds great promise to achieve completed disease regression and improved clinical outcomes for T-ALL.16
Currently, no approved therapies are specifically designed to target the self-renewal processes of leukemia stem cells in T-ALL. Existing therapeutic strategies under development for T-ALL predominantly focus on the modulation of dysregulated signaling pathways, including Notch, mTOR, and PI3K.17 However, these pathways are also important in the development and function of the normal hematopoietic system, and their utilization to target LSCs has resulted in side effects limiting their clinical applications.18 Hence, there is a need for innovative therapeutic approaches to effectively target LSCs in T-ALL with an enhanced safety profile and increased potential for a successful transition to clinical development.
The role of the mitochondria conferring stemness to cancer stem cells19 and their contribution to drug resistance20 has been increasingly recognized. This positions mitochondrial function as a pivotal target for therapies aimed at eliminating CSCs.21 The mitochondria in CSCs exhibit distinct characteristics compared to those in the rest of the tumor. Those unique features may play a central role in maintaining stemness and regulating cell proliferation and apoptosis.19 Such features include mitochondrial morphology,22 subcellular localization,23 the amount of mitochondrial DNA (mtDNA)24 and mitochondrial metabolism.25 These characteristics have been extensively studied in CSCs and compared to their differentiated counterparts in various cancers, such as liver,26 breast27 and oral28 cancers.
In T-ALL, targeting the mitochondria is emerging as a promising strategy for anti-leukemia therapies, either as a standalone treatment or in combination with conventional chemotherapy agents.29 However, investigating mitochondrial alterations specifically associated with stemness in T-ALL presents challenges, primarily due to the lack of definitive surface markers for LSCs, necessitating functional assays to interrogate self-renewal.30 Despite these obstacles, the specific contributions of mitochondria to LSC self-renewal are becoming more apparent. This review will explore the current understanding and emerging insights into the role of mitochondria in self-renewal, as well as the potential mitochondrial vulnerabilities of LSCs.
2. T-ALL: epidemiology and current landscape
2.1. Genetic insights and treatment approaches
T-ALL accounts for 10–15% of pediatric ALL cases, with an incidence 2–3 times higher in boys than in girls. These patients often present with high initial white blood cell count, a higher frequency of mediastinal mass, and neurological abnormalities.31 Genetic and epigenetic abnormalities in immature thymocytes can lead to T-ALL or T-cell lymphoblastic lymphoma (T-LBL).1,32 Predominantly, three pathways are dysregulated in T-ALL. First, over 90% of T-ALL cases can be classified into subgroups based on the deregulation of T-ALL transcription factors such as TAL1, TAL2, and LMO2/LYL1. Second, the aberrant activation of NOTCH1 signaling, either through activating mutations (in more than 75% of cases) or mutations that inhibit the negative regulator FBXW7, leads to high Myc expression and uncontrolled cell proliferation. Finally, there is often a deletion of the tumor suppressor genes CDKN2A/CDKN2B.33
Treatment for newly diagnosed T-ALL patients involves intensive chemotherapy structured in three phases: induction, consolidation, and maintenance. The goal of induction therapy is to restore normal blood cell levels, and it typically includes vincristine, corticosteroids, and asparaginase with or without anthracycline for 4–6 weeks. Following that, consolidation therapy aims to eliminate the residual leukemia cells, which is achieved using a combination of high-dose chemotherapy agents, including methotrexate. Maintenance therapy lasts for 1–2 years and includes 6-mercaptopurine (6-MP) and methotrexate to prevent relapse.34 Hematopoietic stem cell transplantation is reserved for adult T-ALL patients at high risk for relapse.35
2.2. Addressing treatment challenges in T-ALL
Pediatric patients exhibit unique drug absorption and metabolic profiles compared to adults, making toxicity a crucial consideration in intensive chemotherapy protocols.36 Despite high cure rates, treatment-related morbidity and adverse event rates in pediatric T-ALL remain significant. Summers et al. evaluated 120 children aged 1–21 with T-ALL to assess clinically relevant adverse effects (AEs) resulting from chemotherapy. The majority of patients, 70% during induction and 80% during consolidation, experienced AEs.37 The chemotherapy-induced gonadotoxic effects of chemotherapy pose another significant concern. Close et al. categorized different leukemia treatment protocols according to the risk they pose for gonadal dysfunction/infertility. More than half (54%) of the ALL treatment protocols present a high risk, particularly for male patients, who comprise the higher proportion of the affected demographic.38
The emergence of Chimeric Antigen Receptors (CAR)-T cell therapy (CAR T-cell) marked a significant breakthrough in the treatment of hematological malignancies, offering a possible solution for the challenges associated with conventional chemotherapies.39 CAR-T cells are engineered to identify antigens presented by cancer cells and initiate a potent anti-tumor response. In B-ALL, the US Food and Drug Administration (FDA) approval of two CD19-targeted CAR-T cell therapies has been particularly transformative for patients refractory to initial chemotherapy or experiencing relapse,40 with phase II clinical trials in pediatric and adolescent patients reporting an 81% remission rate at day 28 post-infusion. These high remission rates have been confirmed by real-world data.41 However, despite the promise of CAR-T cell therapies, they are associated with significant limitations, including severe side effects such as cytokine release syndrome and neurotoxicity.42 For instance, 23–46% of patients treated with CAR-T therapies experienced supraphysiological cytokine production, leading to massive T-cell expansion, Macrophage Activation Syndrome, renal failure, pulmonary edema, and immune effector cell-associated neurotoxicity.43 Additionally, the high cost of FDA-approved CAR T-cell therapies raises serious concerns about feasibility and affordability for the general patient population.44 Finally, no approved CAR-T is available for standard clinical care for T-ALL patients. CD7 CAR-T cell therapy is currently under investigation for T-ALL, with ongoing clinical trials evaluating its safety and efficacy in patients with CD7-positive malignancies.45–47 This gap in available treatments highlights the urgent need for continued research and development of effective therapies for T-ALL.
2.3. Prognostic indicators and relapse challenges
The most pressing challenge in the treatment of T-ALL across pediatric and adult populations is the occurrence of relapse.48 Minimal residual disease (MRD), defined as the persistence of cancer cells post-treatment, is the most important prognostic factor for assessing relapse risks.49 MRD levels are typically assessed by flow cytometry, which has a detection threshold of 0.01%.50 More recently, clonoSEQ has been approved as a next-generation sequencing (NGS) platform for MRD measurement in ALL.51 This assay utilizes multiplex PCR and NGS techniques to identify leukemia-specific B or T cell receptor gene rearrangements.52 The NGS-based MRD testing boasts a high sensitivity, estimated at 10−7.53 Momen et al. reported that MRD detected exclusively by clonoSEQ but not by flow cytometry corresponded to lower MRD values.54 Based on the MRD scores at specific time points, patients are stratified into standard, intermediate, and high-risk groups, enhancing the predictive accuracy of patient outcomes.34 The presence of MRD is now considered the strongest predictor of patient relapse.55 Yet, the limited tolerability of cytotoxic chemotherapies makes achieving remission after a relapse difficult, often leading to poor outcomes. For instance, in the E2993/UKALL12 study, only 8 out of 123 adult T-ALL patients who relapsed survived after a median follow-up of 5.2 years.56 A similar retrospective analysis of relapsed pediatric T-ALL patients showed a 10-year overall survival rate ranging from 13% to 29%,57 highlighting the grim prognosis for patients post-relapse.
In total, the field of pediatric T-ALL treatment faces three critical challenges: the morbidities associated with chemotherapy, the limitations of still-developing CAR T-cell therapies for T-cell malignancies, and the bleak prognosis for patients who relapse. These issues highlight a significant gap in effective treatment strategies. To overcome these challenges, there is a pressing need for innovative therapies that target the biological underpinnings of the disease.
3. Targeting leukemia stem cells for improved clinical outcomes
Intensive, cytotoxic chemotherapy remains the standard of care in treating T-ALL and effectively eliminates proliferating cells. However, these therapies often target the bulk of leukemia cells indiscriminately, which can result in a dramatic initial response but may fail to induce a complete and long-term remission.58 This outcome is particularly evident in patients with high MRD values, who are at greatest risk of relapse. The ability of MRD cells to reform leukemia from low cell numbers can be understood through the cancer stem cell model. In T-ALL, this model highlights leukemia stem cells (LSCs) as a critical subpopulation.59 These cells possess an indefinite capacity for self-renew and are essential for sustaining long-term tumor growth.60 Importantly, only the LSC subset can reestablish leukemia, while the other, more differentiated sub-populations have a limited capacity for long-term self-renewal and proliferation (Figure 1a).10 Targeting and eliminating these LSCs is essential for patients to achieve durable remission (Figure 1b).59
Figure 1.
Leukemia stem cells are responsible for the long-term maintenance of the disease and patient relapse. a: Leukemia stem cells are a subset of cells within T-ALL that have the capacity to self-renew and regenerate disease from low cell number, compared to the rest of the population that have limited cell divisions. b: Conventional chemotherapy drugs are targeted toward the rapidly proliferating leukemia cells and may not always eradicate LSCs, resulting in patient relapse. Treatment strategies that combine LSC targeting therapies with conventional chemotherapy can eradicate the entire disease and prevent recurrence.
3.1. Challenges in defining leukemia stem cell markers in T-ALL
A particular challenge in targeting T-ALL self-renewal involves the lack of robust surface markers for LSCs in this disease. Cox et al. utilized pediatric leukemia patient samples in engraftment studies with NOD/SCID mice and demonstrated the ability of CD34+/CD4− and CD34+/CD7− subfractions to populate leukemia after serial passaging.61 In contrast, Gerby et al. reported that the CD34+/CD7- subpopulation isolated from the peripheral blood of T-ALL patients contained normal human progenitors that underwent normal hematopoietic differentiation in vitro and in vivo. Instead, they described the enrichment of LSCs in the CD34+/CD7+ fraction.11 In addition, Chiu et al. showed that a subset of cells that are CD7+CD1a− from primary T-ALL patient samples are enriched for LSC by xenograft studies in immune-deficient mice and noted CD34 as a reliable stemness marker for some but not all of the samples tested.62 Advanced single-cell sequencing technologies have also revealed significant heterogeneity within the LSC population in T-ALL. Zhu et al. analysis used single-cell transcriptomics and showed that cells expressing high levels of TIM-3/HAVCR2 were enriched for pathways associated with hematopoietic stem cells, late hematopoietic progenitors, and cellular quiescence. These cells also demonstrated the highest leukemia-initiating capacity, confirmed by a limiting dilution assay in immune-deficient mice. Almost every cell in this high TIM-3/HAVCR2 expression group was able to induce T-ALL development compared to 1 in 28 in the low-expression group.63 In a related study, Panelli et al. reported that CD117 and CD82 characterized a distinct T-ALL subpopulation enriched in MRD. This observation was further validated by injecting CD117+CD82+ T-ALL cells into immune-deficient mice at limiting dilution and finding enrichment in the leukemia-initiating capacity in this population of cells in two independent primary T-ALL samples.64 Taken together, these findings imply that the LSC activity in T-ALL might originate from different subpopulations of cells and underscore the heterogeneity within LSCs. Until cell surface makers can be reliably defined for LSCs in T-ALL, there is increasing dependence on functional methodologies to assess LSC self-renewal capabilities. Techniques such as the limiting dilution transplantation studies in immune-deficient mice are widely used,65,66 and more recently, similar studies have been conducted in syngeneic clonal zebrafish models.67,68 Continued research is needed to better understand LSC distribution in T-ALL and to validate newly described surface markers across different T-ALL animal models and patient samples. Ultimately, the identification of robust LSC surface makers will be critical for advancing self-renewal-focused drug screens and the discovery of novel therapeutic targets for T-ALL.
3.2. Mechanisms of leukemia stem cell survival and drug resistance
Intriguingly, the prevalence of LSCs within the leukemia population is closely linked to disease pathogenesis. Ho et al. monitored a cohort of acute myelogenous leukemia (AML) patients using limiting dilution analysis to quantity LSC frequency. They observed a dramatic 9- to 90-fold increase in LSC frequency between diagnosis and relapse,69 underscoring the critical role LSCs play in disease progression and recurrence. Another study found that the presence of LSCs in AML, identified with a CD34+/CD38− immune phenotype, strongly predicted patient outcomes.70 Patients with higher LSC frequencies (>3.5%) had a median relapse-free survival of 5.6 months compared to 16 months for those with lower frequencies of these cells,71 highlighting the significant impact of LSC burden on clinical prognosis.
The relationship between LSC prevalence and patient prognosis is evident. A deeper understanding of LSC biology is crucial to addressing their role in disease progression and treatment resistance, particularly their mechanisms of chemoresistance and survival. Work in AML, where LSCs have been well-characterized, shows that this cell type possesses several unique properties that allow it to evade conventional cytotoxic chemotherapy (Figure 2).72 Notably, LSCs have higher expression of ATP-binding cassette (ABC) transporters, which are key players in drug resistance. These include the permeability glycoprotein (P-gp), breast cancer resistance protein (BCRP), and multidrug resistance-associated protein 1 (MRP1).73 Additionally, LSCs often exist in a state of dormancy or slowed growth, allowing them to circumvent the effects of antiproliferative chemotherapy agents.74 In T-ALL patient-derived xenograft (PDX) mouse models, cells demonstrating the slowest growth rate and highest retention of the cell proliferation stain CFSE were associated with stemness, drug resistance, and high similarity to MRD cells.75 LSCs maintain dormancy by activating pathways central to self-renewal and stemness, such as the Wnt/β-catenin signaling pathway. In T-ALL patients, about 85%of the patients have an upregulation in the expression of β-Catenin and subsequent high expression of Wnt target genes, including c-MYC, TCF, and LEF.76 Single-cell sequencing of MRD samples in T-ALL patients also identified high expression of β-Catenin in this drug-resistant cell population.77 LSCs also display defects in apoptosis-related signaling pathways, such as the upregulation of the anti-apoptotic protein BCL-2.78
Figure 2.
LSCs have unique mechanisms in place to overcome chemotherapy. Leukemia stem cells develop multiple mechanisms to escape conventional chemotherapy treatment including the expression of drugs efflux pumps, cell cycle arrest, their interaction with the bone marrow micromovement and the altered signaling and apoptosis regulation pathways.
While these intrinsic mechanisms are pivotal, the tumor microenvironment (TME) also plays an essential role in maintaining LSC survival and drug resistance. In normal tissues, niches provide essential cues to regulate stem cell differentiation and quiescence,79 and in cancer, these niches support cancer stem cells by promoting self-renewal and recruiting stromal and immune cells to drive tumor progression.80 In leukemia, the bone marrow niche, which tightly regulates hematopoietic stem cells (HSCs),81 is co-opted by LSCs to gain survival advantages and evade therapy.82 This niche-driven support includes mitochondrial transfer from stromal cells to LSCs,83 cytokine-mediated signaling,84 and the release of extracellular vesicles,85 which collectively modulates drug resistance pathways. By leveraging these niche-specific interactions, LSCs evade therapeutic pressures, contributing to MRD and increasing the likelihood of relapse.
3.3. Therapeutic strategies targeting leukemia stem cells
The intrinsic survival mechanisms and niche-driven support that sustain leukemia stem cells (LSCs) highlight the challenges of effectively targeting these cells. Building on this understanding, researchers have developed therapeutic strategies aimed at disrupting LSC survival and self-renewal pathways. Notable success has been achieved with all-trans retinoic acid (ATRA) in the treatment of acute promyelocytic leukemia (APL). ATRA induces terminal differentiation of APL blasts into functional granulocytes.86 This approach has successfully been translated into clinical settings; patients receiving a combination therapy of ATRA and arsenic trioxide (ATO) showed superior outcomes compared to those on ATO monotherapy. All patients (n = 20) treated with this combination remained in remission, while 19% (n = 37) of those receiving only ATO relapsed within a median follow-up time of 18 months.87
Similarly, targeting LSCs has gained traction in AML, with the development of therapies designed to disrupt survival pathways in these cells. Venetoclax, a BCL-2 inhibitor, successfully activates apoptotic pathways in LSCs and offers a new therapeutic strategy for patients with high-risk or refractory AML88,89
In T-ALL, there are currently no therapies approved specifically for targeting the LSC population. However, several investigational therapeutics focus on modulating pathways critical to T-ALL LSC activity (Table 1).17 For example, Notch signaling, a central pathway in T-ALL development, has long been an attractive target.104 Early efforts involved γ-secretase inhibitors (GSIs), which block a critical step in Notch activation.105 The γ-secretase complex mediates a secondary cleavage of the Notch1 receptor to facilitate the release of intracellular Notch, which is subsequently translocated to the nucleus to activate transcription of Notch target genes.106 Several GSIs such as MK0752,107 PF-03084014108 and BMS-906024109 have undergone clinical trials in T-ALL patients.17 Other GSIs, including MRK-560,91 MRK-003,90 and DAPT92 have shown promise in pre-clinical studies, reducing LSC frequency and inhibiting leukemia engraftment in murine models. For instance, Habets et al. reported that inhibition of a subclass of γ-secretase complexes with MRK-560 significantly reduced leukemia burden in four PDX models in vivo .91 However, despite these advances, the translation of GSIs to standard clinical practice has been hindered by significant gastrointestinal toxicities.110
Table 1.
Inhibitors targeting canonical self-renewal pathways in T-ALL.
Drug | Pathway | Specific effect | The model used in testing | Effect on LSC | Drug development stage* | Reference |
---|---|---|---|---|---|---|
γ-secretase inhibitor: MRK-003 | Notch1 | Notch pathway inhibition | Tal1/Lmo2 transgenic mice model | Treatment reduced LSC frequency; all transplanted mice failed to develop the disease | Preclinical | 90 |
γ-secretase inhibitor: MRK-560 | Notch pathway inhibition | Leukemia mice model; Human T-ALL PDX mice | Treatment strongly mitigated leukemia development | Preclinical | 91 | |
γ-secretase inhibitor: DAPT | Notch pathway inhibition | Primary T-ALL cells; Human T-ALL PDX-mice | Lower T-ALL engraftment in mice transplanted with cells drug-treated | Preclinical | 92 | |
FDA-approved compounds and 2-ME2 | MYC and SCL protein levels decrease | Thymocytes reprogrammed by the SCL and LMO1 oncogenic transcription factors into self-renewing pre-LSCs; Transgenic mice model | 2-ME2 abrogated pre-LSC viability in vitro and self-renewal activity in vivo | Preclinical | 93 | |
γ-secretase inhibitor: DAPT | LYL1 and LMO2 expression inhibition | Thymocytes reprogrammed by the SCL and LMO1 oncogenic transcription factors into self-renewing pre-LSCs; Transgenic mice model | Lower T-ALL engraftment in mice transplanted with cells drug-treated | Preclinical | 94 | |
γ-secretase inhibitor: DAPT | Notch pathway inhibition | T cell lymphoma line: CUTLL1 | Decrease of cell viability | Preclinical | 95 | |
Rapamycin, JQ1, and VX-680 |
MYC and PI3K-Akt | MYC and PI3K/Akt pathway inhibition | T-ALL cell lines: Jurkat, MOLT-3, MOLT-4, MOLT-16, CCRF-CEM; T cell lymphoma line: CUTLL1; PTEN null mice model | Treatment eliminated PTEN null LSCs | JQ1, and VX-680: Preclinical Rapamycin: Phase I, II (completed) |
96 |
MK-2206 | PI3K-Akt | Allosteric Akt inhibitor | T-ALL cell lines: MOLT-4, CCRF-CEM, CEM-R | Treatment induced apoptosis in a T-ALL patient cell subset (CD34þ/CD4/CD7), which is enriched in LSCs | Phase I, II (completed) |
97 |
Rapamycin | mTOR complex 1 inhibition | PTEN null mice model | Long treatment of pre-leukemic PTEN-deleted mice blocked the formation of LSCs and prevented disease development | Phase I, II (completed) | 98 | |
BKM120 | PIK3 inhibition | Primary T-ALL cells; T-ALL cell lines: Jurkat, RPMI-8402, BE-13, HPB-ALL, PF-382, P12-Ichikawa, DND-41, MOLT-4, CCRF-CEM, CEM-R | Treatment-induced apoptosis in the side population (SP) of cells that is enriched in LSCs | Phase I, II (completed) |
99 | |
JQ1 | MYC | c-Myc pathway inhibition | Tal1/Lmo2 transgenic mice; Primary mouse T-ALL cells; Primary human T-ALL cells | Treatment reduced LSC frequency | Preclinical | 100 |
10058-F4, MYCi975 | MYC-MAX | MYC–MAX complex formation | T-ALL cell lines: KOPT-K1 and Jurkat | Treatment induced cytotoxicity in T-ALL cells in a dose-dependent manner and down-regulated the expression of NOTCH1 | Preclinical | 101 |
PROTAC: ARV-825 | Notch1-MYC-CD44 | Downregulation of the Notch1-MYC-CD44 axis |
T-ALL cell lines: CCRF-CEM, HPB-ALL, KOPT-K1, Loucy, MOLT4, SUP-T1; PTEN null mice model; Human T-ALL PDX-mice | Treatment reduced LSC frequency, induced cell cycle arrest and apoptosis; extended survival of mice engrafted with T-ALL from ARV-825 treated mice | Preclinical | 102 |
Erlotinib | Wnt/β-catenin | Wnt/β-catenin inhibition | T-ALL cell lines: Jurkat; 6xTCF/LEF-miniP:sGFP transgenic zebrafish; T-ALL CG1 zebrafish model | Treatment reduced self-renewal of T-ALL cell line, reduced LSC frequency and delayed disease formation in zebrafish models | Phase I, II (completed) | 67 |
XAV-939 | Wnt/β-catenin inhibition | T-ALL cell lines: HPBALL, RPMI8402; Human PDX cells | XAV-939 induced cytotoxicity in T-ALL cells in a dose-dependent manner | Preclinical | 103 |
*Clinical studies focused specifically on leukemia.
Monoclonal antibodies targeting Notch, such as OMP-52M51, represent an alternative approach with fewer toxicities. Pre-clinical studies using PDX models demonstrated that treatment with the anti-Notch antibody significantly delayed the engraftment of T-ALL cells. This includes PDX derived from patients whose disease did not respond well to chemotherapy or who relapsed. For example, mice treated with anti-Notch1-treated mice were healthy or had low leukemia burden compared to control animals, suggesting decreased LSC function.111 Preliminary results from the phase I clinical trial on OMP-52M51 in patients with hematologic malignancies, including T-ALL, suggest a moderate efficacy and an acceptable safety profile, with diarrhea noted as the most common side effect.112
The PI3K/Akt/mTOR signaling pathway is also implicated in the self-renewal of LSCs in T-ALL.13 This pathway is negatively regulated by PTEN, a tumor suppressor gene altered in 11–27% of pediatric T-ALL cases.113 Therapeutic inhibition of this pathways has shown promise in targeting LSC-enriched populations. For instance, BKM120, a pan-PI3K inhibitor, preferentially induced apoptosis in the side population (SP) of cells enriched in LSCs, with a lesser effect on non-SP cells in primary T-ALL patient samples.99 Similarly, the Akt inhibitor MK-2206 induced apoptosis in a specific subset of cells (CD34+/CD4-/CD7-), which is enriched in LSCs,97 further demonstrating the potential of targeting this pathways.
In pre-clinical models, Guo et al. reported that the long-term treatment (3–4 months) of pre-leukemic PTEN-deleted mice with the mTOR inhibitor rapamycin blocked the LSC self-renewal and prevented disease development.98 Schubbert et al. demonstrated that treating mice engrafted with PTEN-null T-ALL cells with a combination of rapamycin and the MYC inhibitor JQ1 resulted in marked reduction of splenomegaly and significantly decreases in leukemia burden within seven days. Importantly, this combination treatment was found to significantly decrease the percent of Lin−;CD3+;c-kitmid LSC-enriched subpopulation in the bone marrow.96
However, targeting the PI3K/Akt/mTOR pathway presents challenges due to its crosstalk with the Notch signaling pathway. Inhibition of PI3K/mTOR can inadvertently activate Notch signaling, which may lead to treatment resistance.96,114 This necessitates the need for combination therapies to simultaneously address interconnected signaling networks and effectively manage disease.
The NOTCH and PI3K/Akt/mTOR signaling pathways are also directly involved in regulating MYC expression, creating a synergistic network critical for leukemic progression. Indirect targeting of MYC has been vigorously investigated for decades.115 One promising strategy involves inhibiting MYC gene transcription, with JQ1 as a prominent example. JQ1 selectively targets the acetyl-lysine recognition motif of the BET family protein BRD4, leading to the suppression of MYC transcription and a reduction in leukemic stem cell frequency.100 Another approach targets MYC-MAX complex formation, which is essential for MYC-mediated transcriptional programs. Small molecule inhibitors 10,058-F4 and MYCi975 have demonstrated efficacy in disrupting MYC – MAX dimerization, resulting in suppressed growth of T-ALL cell lines.101 However, while these approaches show potential, relatively few compounds have been investigated in the context of LSCs in T-ALL, highlighting the need for further exploration in this area.
The Wnt signaling pathway is crucial for self-renewal in both normal116 and cancer stem cells10 and represents another significant therapeutic target in T-ALL. Inhibition of Wnt signaling in hematopoietic stem cells reduces their capacity to self-renew and regenerate the hematopoietic system.117 Dysregulated Wnt signaling is implicated in the malignant transformation and the maintenance of LSCs in AML and chronic myeloid leukemia (CML).118,119 In T-ALL, Giambra et al. observed activated Wnt signaling in a subpopulation of cells enriched for LSCs in NOTCH1-ΔE induced leukemia mouse model. Their findings showed an 85-fold decrease in LSC frequency following the transduction of leukemia cells with a dominant negative TCF (dnTCF) that inhibited β-catenin binding and prevented canonical activation of the pathway.
Pharmacological inhibition of Wnt signaling has shown pre-clinical efficacy. For example, XAV-939 demonstrated ability to inhibit the growth and proliferation of various T-ALL cell lines and human PDX cells in vitro.103 However, the role of Wnt signaling in normal tissue regeneration and hematopoiesis presents a significant challenge for clinical translation due to associated side effects.120 To overcome this challenge, our group utilized a zebrafish line that harbors a Wnt/β-catenin reporter (6XTCF/LEF-miniP:dGFP) to repurpose FDA-approved compounds with established safety profiles as Wnt/β-catenin inhibitors. Erlotinib emerged as a top hit and was found to inhibit the nuclear localization of β-catenin and reduce the expression of Wnt targets. Erlotinib inhibited the self-renewal of human T-ALL cells in vitro and reduced LSC frequency and delayed disease formation in zebrafish models.67
Despite significant progress in targeting T-ALL LSCs in cell cultures and pre-clinical models, clinical application of most of the inhibitors listed in Table 1 may be limited due to the dual role of these signaling pathways in both LSC maintenance and normal stem cell self-renewal. This overlap poses a major challenge, as therapeutic interventions often result in significant toxicity, making it difficult to exploit a therapeutic window. Identifying novel, LSC-specific targets that regulate self-renewal and survival is critical to advancing the translatability of research findings into clinical practice for T-ALL.
4. The mitochondria as a target organelle in T-ALL
Overcoming challenges associated with targeting traditional signaling pathways in T-ALL has prompted increased investigation into cancer cell metabolism, which has emerged as a promising area of research.121 Mitochondria are indispensable organelles responsible for energy production, metabolic regulation, and cell fate determination.122 In cancer, mitochondria are not only the hub of metabolic reprogramming but also key players in sustaining tumor survival, promoting proliferation, and mediating drug resistance. Traditionally, cancer cells have been characterized by their reliance on glucose for energy, a phenomenon known as the Warburg effect.123 However, increasing evidence describes the central role of mitochondria in regulating the metabolic switch between aerobic glycolysis and oxidative phosphorylation (OXPHOS), enabling the high rates of anabolism and bioenergetics necessary for oncogenic transformation.124
In addition to energy metabolism, mitochondria regulate apoptosis through calcium signaling. Tumors can modulate their sensitivity to apoptosis by altering calcium flux between mitochondria and the endoplasmic reticulum (ER) via proteins like the voltage-dependent anion channel (VDAC),125 disrupting this critical signaling axis. Mitochondria-generated reactive oxygen species (ROS) also play a pivotal role in cancer progression, activating survival and metastatic pathways, including nuclear factor−κB (NF-κB), PI3K/Akt, and the mitogen-activated protein kinase (MAPK)/extracellular-regulated kinase 1/2 (ERK1/2) pathways.126,127 Therapeutic strategies targeting mitochondrial ROS can overwhelm antioxidant defenses, destabilize mitochondrial membrane potential, and induce apoptosis.128,129
Drugs targeting mitochondrial metabolism have demonstrated success in leukemias such as AML, CML, and CLL, where they have overcome chemoresistance,130–132 induced apoptosis,131 and promoted cellular differentiation.133 Recent research in T-ALL has revealed similarly critical roles for mitochondrial function. T-ALL cell lines exhibit increased expression of OXPHOS genes compared to normal T cells and other cancer cell lines.134 A metabolism-focused CRISPR-Cas9 screen by Kong et al. identified mitochondrial complex I as essential for T-ALL cell survival, with loss of these genes proving lethal, especially in the presence of the mitochondria-targeted antioxidants like mito-vitamin E. Subsequently, the use of Phenformin, an inhibitor of mitochondrial complex I and OXPHOS, significantly reduced T-ALL burden in mouse models.135 These findings highlight the potential of targeting mitochondrial processes as a therapeutic strategy for T-ALL.
4.1. Mitocans as a new treatment strategy for T-ALL
With the established importance of mitochondrial pathways in T-ALL, the emergence of Mitocans – small molecule compounds that precisely target and destabilize mitochondrial functions – opens new avenues for effective treatment strategies.136 These compounds disrupt key mitochondrial processes, including mitochondrial ATP and DNA synthesis, the balance between OXPHOS and glycolysis, redox homeostasis, and mitochondrial-dependent apoptosis.137
Numerous mitochondrial targets in T-ALL have been identified and investigated for their therapeutic potential. Hexokinase, a key enzyme linking glycolysis and mitochondrial metabolism, is one such target, and its inhibition induces mitochondrial dysfunction and apoptosis in T-ALL cells.138,139 Pro-apoptotic Bcl2 family proteins, which regulate mitochondrial membrane permeability, are also critical targets, with studies showing their modulation can sensitize T-ALL cells to apoptosis.140–145 The mitochondrial voltage-dependent anion channel (VDAC) plays a role in maintaining mitochondrial integrity and regulating metabolite exchange, making it another promising target.146 Additionally, therapeutic approaches that destabilize mitochondrial redox homeostasis have been shown to increase ROS levels in T-ALL, leading to mitochondrial damage and cell death.147–149
Mitocans are effective through several mechanisms, including cytochrome c release,150 loss of mitochondrial membrane potential,151 disruption of the electron transport chain, and ultimately, the failure of ATP production.139 These effective collectively impair mitochondrial function and drive apoptosis, offering a powerful approach to overcoming drug resistance and eradicating leukemia cells. A comprehensive summary of these studies is provided in Table 2.
Table 2.
Summary of mitochondrial-targeting agents tested in T-ALL.
Drug | Mitochondrial target/Vulnerability | The model used in testing | General Outcome | Drug development stage* | Reference |
---|---|---|---|---|---|
Poricoic acid A | ↑ROS levels; ↓Complex I; ↓Cristae, and ↑mitochondria membrane density | T-ALL cell lines: Jurkat, MOLT-3, ALL-SIL, RPMI-8402 |
Autophagy, Ferroptosis | Preclinical | 152 |
11- methoxytabersonine (11-MT) |
↑ROS levels; ↓MMP; ↓PI3K/Akt/mTOR signaling pathway | T-ALL cell lines: MOLT-4, Jurkat, CCRF-CEM, CEM/C1 | Apoptosis | Preclinical | 151 |
Minocycline | ↑ROS levels; ↓MMP; ↑PINK1 and PARKIN | T-ALL cell line: Jurkat | Apoptosis | Preclinical | 153 |
Cannabinoid CP55940 | ↑ROS levels; ↓MMP; ↑PINK1 and PARKIN | T-ALL cell line: Jurkat | Apoptosis | Preclinical | 154 |
WP1130 | ↓MMP; ↓Mcl-1 | T-ALL cell lines: Jurkat, MOLT-4, HPB, CCRF-CEM | Apoptosis | Preclinical | 155 |
Vorinostat/Quinacrine | ↓MMP; ↓Mcl-1; ↓Bcl-2/Bax ratio; ↑PARKIN | T-ALL cell lines: Jurkat, MOLT-4; PDX mouse model | Apoptosis, Mitophagy blockage, and Tumor size decrease | Vorinostat: Phase I, II (completed and recruiting) Quinacrine or Combination of drugs: Preclinical |
140 |
Cannabidiol | ↓MMP; Cytochrome c release, ↑Ca2+, | T-ALL cell lines: MOLT-3, Jurkat, CCRF-CEM | Apoptosis | Preclinical | 156 |
Tipifarnib | ↓SHMT2, MTHFD1 and CTPS1 | 25 T-ALL cell lines | Cytotoxicity | Phase I, II, III (completed) | 157 |
Brusatol | ↑ROS levels; ↓MMP | T-ALL cell lines: MOLT-4, CCRF-CEM | Cytotoxicity | Preclinical | 147 |
Oxamate | LDH inhibition; ↑ROS levels; ↓MMP | T-ALL cell lines: Jurkat, DU528 | Apoptosis | Preclinical | 158 |
α-Pinene | ↑ROS levels; ↓MMP; ↓ Basal and maximal OCR; ↑Glucose consumption and lactate production; ↑ EGR1-p53-Bax/Bcl-2-caspase cascade | T-ALL cell lines: EL-4, MOLT-44; PDX mouse model | Apoptosis, Tumor size decrease | Preclinical | 142 |
Ferrocene derivatives | ↑ROS levels; ↓MMP; ↓PI3K/Akt/mTOR signaling pathway | T-ALL cell lines: Jurkat, CEM-T4, RAJI, CA46, SNT8, SNK6 | Apoptosis | Preclinical | 159 |
SHIN2 | Inhibition of mitochondrial isozyme SHMT2 | T-ALL cell lines: MOLT-4, MOLT-3, Jurkat, KOPT-K1; PDX mouse model | Cytotoxicity, Sensitization to methotrexate, Tumor size decrease | Preclinical | 160 |
PKHB1 | ↑ROS levels; ↓MMP; ↑ATP release | T-ALL cell lines: MOLT-4, CCRF-CEM, L5178Y-R | Apoptosis | Preclinical | 148 |
ABT-199/Gemcitabine | ↓MMP; ↓Bcl2 and Bcl-xL;↑ Bim and Puma; Cytochrome c release, PARP cleavage | T-ALL cell lines: MOLT-4, Jurkat | Apoptosis | Venetoclax: Phase I, II, III (completed and recruiting) Phase IV (recruiting) Gencitabine: Phase I, II (completed and recruiting) Combination of drugs: Preclinical |
141 |
MI-2 | PARP cleavage; ↓Survivin, Bcl-xL, Bcl-2 and NF-ƙB; ↑Bax | T-ALL cell lines: MOLT-4, CCRF-CEM | Apoptosis | Preclinical | 143 |
Resveratrol, Quercetin, Genistein, Curcumin | ↓MMP | T-ALL cell line: MOLT-4 | Cytotoxicity | Preclinical | 161 |
Melittin | ↓OCR | T-ALL cell line: Jurkat | Cytotoxicity | Preclinical | 162 |
Cannabidiol, Curcumin, Quercetin | ↓MMP; ↑ROS levels; ↑Ca2+, | T-ALL cell line: Jurkat | Cytotoxicity | Preclinical | 163 |
Cannabidiol, Tamoxifen | ↓MMP; ↑Ca2+; Cytochrome c release | T-ALL cell lines: Jurkat, CCRF-CEM | Cytotoxicity | Preclinical | 150 |
Xestospongin B | InsP3R inhibition resulting in a decrease of InsP3R-mediated Ca2+ transfer from the endoplasmic reticulum to mitochondria; ↓Basal and maximal OCR | T-ALL cell lines: Jurkat, CCRF-CEM | Cytotoxicity | Preclinical | 164 |
Tamoxifen | ↓MMP | T-ALL cell line: Jurkat | Apoptosis, Autophagy, Sensitization to dexamethasone | Preclinical | 165 |
Apatinib and Chidamide | ↓ basal and maximal OCR, ↓Enzymes involved in the citric acid cycle and oxidative phosphorylation, ↑Mitochondria-mediated apoptosis pathways | T-ALL cell lines: MOLT-4, Jurkat; PDX mouse model | Apoptosis, Tumor size decrease | Apatinib: Preclinical Chidamide: Phase I, II (recruiting) Combination of drugs: Preclinical |
166 |
Decitabine | Mitochondrial morphology changes; ↓PI3K/Akt/mTOR signaling pathway | T-ALL cell line: MOLT-4 | Apoptosis | Phase I, II and III (completed and recruiting) | 167 |
Quercetin and Autophagy inhibitors | ↓MMP; ↑ROS levels; ↓BAG3, Mcl-1;↑Bak, Caspase-9 -3 -8; PARP cleavage | T-ALL cell lines: Jurkat clones (J/Neo and J/BCL-XL) |
Apoptosis | Preclinical | 144 |
Ginsenoside 24-hydroxy-ginsengdiol | ↓MMP; ↑ROS levels; ↑Bax, Caspase-9–3; Cytochrome c release | T-ALL cell lines: CCRF-CEM; PDX mouse model | Apoptosis, Tumor size decrease | Preclinical | 145 |
Chidamide | ↓MMP;↓ cFLI PL, HDAC 1 and HDAC 3 |
T-ALL cell lines: Jurkat, HUT-78 | Apoptosis and Necroptosis | Phase I, II (recruiting) | 168 |
Diacetyl hexamethylene diamine | ↓MMP; ↓Bcl-2/Bax ratio; ↓Akt | T-ALL cell line: Jurkat | Apoptosis | Preclinical | 169 |
TPEN and TPGS agents (T2 combo) | ↓MMP; ↑caspase 3 | T-ALL cell line: Jurkat | Cytotoxicity | Preclinical | 170 |
VDAC1-derived peptides | Cytochrome c release, HK dissociation from mitochondria | T-ALL cell lines: MOLT-4, Jurkat | Cytotoxicity | Preclinical | 146 |
ABT-737 | ↓MMP; ↑Bax, Bid and Caspase; Cytochrome c release; | T-ALL cell lines: MOLT-3, MOLT-4, CCRF-CEM, COG-VV-317 | Apoptosis, Sensitization to chemotherapies | Preclinical | 171 |
ABT-737 | ↓MMP; ↓Bcl2 and Mcl-1; Cytochrome c release. | T-ALL cell line: CEM-c1 | Apoptosis | Preclinical | 172 |
ABT-263 | ↓Pro-survival Bcl2 family proteins | PDX mouse model | Tumor size decrease, Sensitization to chemotherapies | Phase I, II (completed) | 173 |
Methyl jasmonate | ↓ATP; Cytochrome c release; HK dissociation from mitochondria | T-ALL cell line: MOLT-4 | Cytotoxicity | Preclinical | 139 |
2-DG, LND, 3-BrPA | HK inhibition | T-ALL cell lines: MOLT-4, Jurkat | Sensitization to glucocorticoids | Preclinical | 138 |
Resveratrol | ↓MMP; ↑ROS levels | T-ALL cell line: Jurkat | Apoptosis | Preclinical | 174 |
Arsenic trioxide | ↑ROS; ↑Bad; ↓Akt | T-ALL cell lines: MOLT-4, Jurkat, CEM-C7, CEM-C1 | Apoptosis | Phase I, II, III, IV (completed) Phase I, II (recruiting) |
149 |
Vitamin K3, Vitamin C | ↓MMP; ↑ROS levels; ↑Nf-kB, p53, c-Jun, Caspase-3 | T-ALL cell line: Jurkat | Apoptosis | Preclinical | 175 |
Tigecycline | ↓Basal and maximal OCR; ↓ATP; ↑ROS levels | T-ALL cell lines: CCRF-CEM, DND-41, MOLT-4; PDX mouse model | Apoptosis, Sensitization to chemotherapies, Tumor size decrease | Phase I (completed) | 176 |
Ara-C | ↑ROS levels | T-ALL cell lines: MOLT-4, Jurkat | Apoptosis | Phase I, II, III, IV (completed and recruiting) | 177 |
ROS- Reactive oxygen species, MMP- Mitochondria membrane potential, OCR- Oxygen consumption rate, HK- hexokinase, PDX- Patient-derived xenograft.
*Clinical studies focused specifically on leukemia.
5. The mitochondria as key driver of self-renewal in T-ALL
The extensive investigation of mitochondrial function across various cancers, including in T-ALL, has identified numerous mitochondrial targets that have shown anti-cancer activity in cell lines and animal models.29,178 This section focuses on advancing the understanding of the specific contribution of mitochondria to LSCs in T-ALL. While mitochondria are well established as a general target in T-ALL, the evolving understanding of metabolic heterogeneity in cancer underscores the importance of targeting mitochondrial vulnerabilities specific to distinct tumor subpopulations, such as cancer stem cells (CSCs) and LSCs.179
These subpopulations exhibit unique metabolic preferences influenced by several factors such as their driving mutations, cell of origin, epigenetic regulations, and tumor microenvironment.180 There is also increasing recognition of bioenergetic heterogeneity among CSCs. Specifically, CSCs of different tumors display different energy preferences (glycolytic vs. OXPHOS) depending on their spatial location, oxygen levels in the tissue, and prior exposure to chemotherapy.181
A large body of literature has evaluated the metabolic energy preferences for LSCs in AML.182 Proteomics analysis comparing LSCs with healthy age-matched hematopoietic stem and progenitor cells (HSPCs) revealed a shift from glycolysis to predominantly OXPHOS in LSCs.183 Various studies have targeted LSCs by inhibiting their OXPHOS metabolism. For instance, Lagadinou et al. demonstrated that when OXPHOS was inhibited by treatment with the mitochondrial inhibitors oligomycin and FCCP, LSCs failed to switch to glycolytic metabolism, indicating metabolic inflexibility, and then used ABT-263 to induce death in the LSC subpopulation.78 This finding was supported by Jones et al., who showed that LSCs utilize nutrients such as amino acids and fatty acids to fuel OXPHOS for energy production, and successfully targeted LSCs using a combination of venetoclax and azacytidine to inhibit both mitochondrial function and amino acid metabolism simultaneously.184 The investigation of mitochondrial function in self-renewal pathways in T-ALL offers a promising avenue for the development of LSC-specific targeted therapies similar to those seen in AML.
In T-ALL, maintaining a critical mitochondrial mass may be necessary for leukemia cells to mitigate the oxidative stress induced by chemotherapy. Burt et al. observed the transfer of mitochondria from mesenchymal stromal cells to T-ALL cells in a co-culture model via tunneling nanotube structures. Inhibiting these structures and thus depleting mitochondrial mass led to excessive ROS accumulation in T-ALL cells. Authors suggested that the protection of leukemia cells within a specific niche may also explain the failure of ROS-inducing therapies such as cytarabine (AraC) and daunorubicin to completely eradicate the disease in T-ALL patients.185
Expanding upon these insights into the importance of mitochondria in T-ALL, Zhong et al. investigated the role of the oxysterol-binding protein ORP4L in maintaining mitochondrial respiration in T-ALL by regulating calcium homeostasis. They found that primary T-ALL cells had increased ATP production rates, basal respiration, and ROS production compared to normal T cells. In addition, primary T-ALL cells and T-ALL cell lines demonstrated a reduced capacity to switch to aerobic glycolysis when treated with the mitochondrial inhibitor oligomycin. Short hairpin RNA (shRNA) targeting ORP4L reduced ATP production and disturbed the calcium mitochondrial signaling axis, resulting in low engraftment of primary T-ALL cells in mice, indicative of impaired self-renewal capabilities.186 Subsequently, ORP4L was shown to be selectively expressed on LSCs in AM, where knockdown of ORP4L resulted in decreased colony formation and energy production.187 Most recently, it was discovered that ORP4L deletion can prevent the induction of T-cell leukemogenesis by human T-cell leukemia virus 1.188 Together, these findings suggest a central role of mitochondrial metabolism and its regulators in the development and maintenance of T-ALL.
Another key facet of mitochondrial function explored in T-ALL is OXPHOS. Baran et al. described a functional link between activating NOTCH1 mutations in T-ALL and OXPHOS. Bioinformatics analysis of T-ALL data sets indicated the Notch-1 bound target genes were predominantly associated with the OXPHOS pathway and mitochondrial electron transport chain. This connection was biologically validated using pre-LSCs from the murine SCL-LMO1 transgenic model, where activation of Notch transformed these cells into hypercompetitive leukemia-propagating cells, leading to aggressive T-ALL without any latency. Moreover, in the presence of the activating Notch1 ligand DL4, inhibition of OXPHOS with the small molecule IACS-010759 effectively suppressed T-ALL development in a dose-dependent manner. Notably, cells were insensitive to treatment in the absence of the ligand, further linking Notch activation to OXPHOS in T-ALL. IACS-010759 also significantly reduced the basal and maximal oxygen consumption rates, as well as ATP production, across 11 different T-ALL cell lines, with a higher statistical significance in NOTCH-mutant cells compared to normal T lymphocytes. The bioenergetic changes correlated with decreased viability in T-ALL cell lines, patient-derived xenografts, and patient-derived primary samples. These findings emphasize the critical role of mitochondrial metabolism in the activation of pre-LSCs and the maintenance of disease in the context of activated Notch, one of the most prevalent driver mutations in T-ALL.134
Conversely, other studies have indicated a different mitochondrial adaptation in T-ALL, where there is a shift toward glycolysis for maintaining self-renewal. Fahy et al. examined T-ALL cells within the context of the bone marrow microenvironment to explore mechanisms of therapy resistance. LSCs are thought to predominantly reside in the hypoxic bone marrow niche.103 Under hypoxic conditions designed to mimic the bone marrow microenvironment, T-ALL cells entered a state of growth arrest, or dormancy, and exhibited increased chemotherapy resistance. These traits are characteristic of the LSC phenotype. When exposed to low oxygen levels, primary T-ALL cells had reduced retention of the MitoTracker stain, indicating reduced mitochondrial content, and increased levels of mitochondrial depolarization, indicative of decreased mitochondrial activity. In addition, elevated lactate levels in the cell culture media suggested a shift toward anaerobic glycolysis. These data suggest a significant metabolic rewiring of mitochondria in response to hypoxic conditions, such as may be found in the LSC bone marrow niche.189
Building on these insights into metabolic adaptation, Gachet et al. examined genetic factors contributing to poor prognosis in T-ALL associated with chromosome 6q deletion and found a ribosomal-mitochondrial axis involved in leukemogenesis. Their integrated genomic approaches identified SYNCRIP and SNHG5 as candidate haploinsufficient genes. Silencing these genes significantly accelerated the Tal1/Lmo1/Notch1-induced T-ALL development in mice. CRISPR/Cas9 mediated deletion of those two genes in the CCRF-CEM human T-ALL cell line led to global downregulation of ribosomal pathways and oxidative phosphorylation. Clones lacking these genes had reduced mitochondrial respiration and enhanced glycolysis compared to non-edited controls, indicative of profound metabolic shifts. Moreover, to directly assess the impact of reduced OXPHOS on leukemogenesis, PDX-derived human T-ALL cells were treated with tigecycline, an inhibitor of mitochondrial protein translation, followed by an in vitro limiting dilution assay and immunotyping. Tigecycline treatment induced a shift toward the greater CD34 and lower CD8 expression indicative of increased clonogenicity.190 These findings highlight the complex interplay between genetic mutations and alterations in the cell’s energy metabolism, in this case through the ribosomal-mitochondrial axis, that impact the self-renewal properties of the LSC.
While the role of mitochondrial metabolism in T-ALL self-renewal is increasingly recognized, there remains a lack of consensus on the specific contributions of mitochondria, as evidenced by the conflicting findings summarized in Table 3. One potential explanation for these discrepancies is that many findings related to altered mitochondrial metabolism were byproducts of research focused on another primary target rather than from direct interrogation of mitochondrial functions in T-ALL self-renewal. An unbiased, systematic investigation of key targets known to regulate mitochondrial processes, followed by functional assays of self-renewal, could conclusively link mitochondrial function to LSC frequency or other indicators of self-renewal. Additionally, metabolic heterogeneity within different T-ALL models may contribute to inconsistent findings, leading to diverse interpretations of metabolic alterations. Ultimately, addressing these discrepancies through comprehensive and targeted research could lead to significant advancements in mitochondrial-focused therapies in T-ALL, enhancing treatment strategies for this challenging leukemia subtype.
Table 3.
Mitochondrial vulnerabilities that affect self-renewal in T-ALL.
Mitochondrial Vulnerability | Mechanism of Action in Mitochondria | Models Used for Testing | General Outcome | Drug(s) Used | Reference |
---|---|---|---|---|---|
Mitochondrial respiration | ALL EVs were enriched with cholesterol, which accelerated the mitochondrial metabolism and the loss of quiescence in targeted healthy HSPC |
PDX mouse model | ALL cells produced EVs that target endogenous murine HSPC in bone marrow disturbing their quiescence and maintenance |
None | 191 |
Mitochondrial calcium concentration |
ORP4L expression in T-ALL cells mediates G protein-dependent signaling and leads to translocation and activation of PLCβ3 to maintain calcium homeostasis and bioenergetics |
Primary T-ALL cells and PDX mouse model |
ORP4L supports mitochondrial oxidative phosphorylation for T-ALL cell survival |
None | 186 |
Mitochondrial respiration | Deletion of chromosome 6q results in downregulation of genes SYNCRIP and SNHG5 involved in mitochondrial function. Also, it reduced basal and maximal mitochondrial respiration | Primary T-ALL cells; T-ALL cell line: CCRF-CEM; T-ALL mouse model; PDX mouse model | Deletion of chromosome 6q inactivated SYNCRIP and SNHG5 genes results in metabolic re-wiring and increased leukemia-initiating activity | Tigecycline | 190 |
Mitochondrial content and membrane potential | Hypoxia-induced low mitochondrial mass and depolarization of the mitochondria membrane | Primary T-ALL cells; PDX mouse model | Hypoxia-induced HIF-1α expression and slowed down T-ALL growth through increased quiescence of leukemic cells, inducing drug resistance | Sensitization to chemotherapies | 189 |
Mitochondrial respiration | IACS-010759, an inhibitor of mitochondrial complex I, disrupts oxidative phosphorylation and redox balance in NOTCH1-mutated T-ALL and pre-leukemic cells | T-ALL cell lines: Jurkat, PF-382, 1301, TALL-1, Loucy, P12-Ichikawa, MOLT-3, MOLT-4, CCRF-CEM, SUPT1 and KOPT-K1; T-ALL primary cells; PDX mouse model | OXPHOS downstream of Notch1 is essential for preleukemic and leukemic stem cell function. OXPHOS pathway blockade induced profound metabolic shutdown in T-ALL cells with NOTCH1 mutations, in turn resulting in apoptosis | IACS-010759 | 134 |
EVs- Extracellular Vesicles, PDX- Patient-derived xenograft, HSPC- Hematopoietic Stem and Progenitor Cells, OXPHOS- Oxidative phosphorylation
6. Conclusion
Poor post-relapse outcomes remain a significant challenge in treating T-ALL. Targeting leukemia stem cells holds considerable promise for eliminating relapse and improving patient outcomes. However, targeting the canonical signaling pathways central to LSC function, such as Notch, Akt, and Wnt, is complicated by dose-limiting side effects due to their importance in normal tissue development and homeostasis.
Metabolic heterogeneity within tumors is increasingly recognized, with cancer stem cells, including LSCs, often exhibiting metabolic phenotypes distinct from the bulk tumor. This highlights the potential of targeting metabolic pathways to specifically inhibit self-renewal and LSC function. In this review, we analyzed the role of the mitochondria in the context of T-ALL self-renewal. Although Mitocans are widely used as a general T-ALL target, few studies detail the specific contributions of mitochondria to T-ALL LSC maintenance and behavior. This gap in knowledge can be partly explained by the current lack of robust surface markers for LSCs in T-ALL and the need for functional assays to accurately assess LSC enrichment.
Moving forward, there is an opportunity to systemically dissect the role of various mitochondrial processes in T-ALL self-renewal. Such investigations could provide promising avenues for therapy development, leading to the identification of targeted agents that specifically disrupt self-renewal in T-ALL and possibly other cancer stem cells with similar metabolic dependencies.
Funding Statement
This research was funded by the National Cancer Institute (R37CA227656 to JSB) and the Kentucky Pediatric Cancer Research Foundation (research grant to JSB).
Disclosure statement
No potential conflict of interest was reported by the author(s).
Author Contributions
Writing – original draft preparation, M.A.A; writing – review and editing, J.S.B; visualization, M.A.A., E.W; funding acquisition, J.S.B. All authors have read and agreed to the published version of the manuscript.
References
- 1.Karrman K, Johansson B.. Pediatric T‐cell acute lymphoblastic leukemia. Genes Chromosomes Cancer. 2017;56(2):89–18. doi: 10.1002/gcc.22416. [DOI] [PubMed] [Google Scholar]
- 2.Van Vlierberghe P, Ferrando A. The molecular basis of T cell acute lymphoblastic leukemia. J Clin Investigation. 2012;122(10):3398–3406. doi: 10.1172/JCI61269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Pui C-H, Evans WE. A 50-year journey to cure childhood acute lymphoblastic leukemia. In: Seminars in hematology. Vol. 50. WB Saunders; 2013. p. 185–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Pui C-H, Yang JJ, Hunger SP, Pieters R, Schrappe M, Biondi A, Vora A, Baruchel A, Silverman LB, Schmiegelow K, et al. Childhood acute lymphoblastic leukemia: progress through collaboration. J Clin Oncol. 2015;33(27):2938. doi: 10.1200/JCO.2014.59.1636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Pui C-H, Robison LL, Look AT. Acute lymphoblastic leukaemia. Lancet. 2008;371(9617):1030–1043. doi: 10.1016/S0140-6736(08)60457-2. [DOI] [PubMed] [Google Scholar]
- 6.Bhojwani D, Pui C-H. Relapsed childhood acute lymphoblastic leukaemia. lancet oncology. 2013;14(6):e205–e217. doi: 10.1016/S1470-2045(12)70580-6. [DOI] [PubMed] [Google Scholar]
- 7.Sive JI, Buck G, Fielding A, Lazarus HM, Litzow MR, Luger S, Marks DI, McMillan A, Moorman AV, Richards SM, et al. Outcomes in older adults with acute lymphoblastic leukaemia (ALL): results from the international MRC UKALL XII/ECOG 2993 trial. Br J Haematol. 2012;157(4):463–471. doi: 10.1111/j.1365-2141.2012.09095.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Oriol A, Vives S, Hernandez-Rivas JM, Tormo M, Heras I, Rivas C, Bethencourt C, Moscardo F, Bueno J, Grande C, et al. Outcome after relapse of acute lymphoblastic leukemia in adult patients included in four consecutive risk-adapted trials by the PETHEMA study Group. Haematologica. 2010;95(4):589. doi: 10.3324/haematol.2009.014274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zhou B-BS, Zhang H, Damelin M, Geles KG, Grindley JC, Dirks PB. Tumour-initiating cells: challenges and opportunities for anticancer drug discovery. Nat Rev Drug Discov. 2009;8(10):806–823. doi: 10.1038/nrd2137. [DOI] [PubMed] [Google Scholar]
- 10.Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell STEM Cell. 2014;14(3):275–291. doi: 10.1016/j.stem.2014.02.006. [DOI] [PubMed] [Google Scholar]
- 11.Gerby B, Clappier E, Armstrong F, Deswarte C, Calvo J, Poglio S, Soulier J, Boissel N, Leblanc T, Baruchel A, et al. Expression of CD34 and CD7 on human T-cell acute lymphoblastic leukemia discriminates functionally heterogeneous cell populations. Leukemia. 2011;25(8):1249–1258. doi: 10.1038/leu.2011.93. [DOI] [PubMed] [Google Scholar]
- 12.McCormack MP, Young LF, Vasudevan S, de Graaf CA, Codrington R, Rabbitts TH, Jane SM, Curtis DJ. The Lmo2 oncogene initiates leukemia in mice by inducing thymocyte self-renewal. Science. 2010;327(5967):879–883. doi: 10.1126/science.1182378. [DOI] [PubMed] [Google Scholar]
- 13.Blackburn JS, Liu S, Wilder J, Dobrinski K, Lobbardi R, Moore F, Martinez S, Chen E, Lee C, Langenau D. Clonal evolution enhances leukemia-propagating cell frequency in T cell acute lymphoblastic leukemia through Akt/mTORC1 pathway activation. Cancer Cell. 2014;25(3):366–378. doi: 10.1016/j.ccr.2014.01.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3(7):730–737. doi: 10.1038/nm0797-730. [DOI] [PubMed] [Google Scholar]
- 15.Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367(6464):645–648. doi: 10.1038/367645a0. [DOI] [PubMed] [Google Scholar]
- 16.Tamiro F, Weng AP, Giambra V. Targeting leukemia-initiating cells in acute lymphoblastic leukemia. Cancer Res. 2021;81(16):4165–4173. doi: 10.1158/0008-5472.CAN-20-2571. [DOI] [PubMed] [Google Scholar]
- 17.Zhang Z, Yang K, Zhang H. Targeting leukemia-initiating cells and leukemic niches: the next therapy station for T-cell acute lymphoblastic leukemia? Cancers. 2022;14(22):5655. doi: 10.3390/cancers14225655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Fransecky L, Mochmann LH, Baldus CD. Outlook on PI3K/AKT/mTOR inhibition in acute leukemia. Mol Cellular Therapies. 2015;3(1):1–17. doi: 10.1186/s40591-015-0040-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zheng X-X, Chen J-J, Sun Y-B, Chen T-Q, Wang J, Yu S-C. Mitochondria in cancer stem cells: Achilles heel or hard armor. Trends Cell Biol. 2023;33(8):708–727. doi: 10.1016/j.tcb.2023.03.009. [DOI] [PubMed] [Google Scholar]
- 20.Naik PP, Mukhopadhyay S, Panda PK, Sinha N, Das CK, Mishra R, Patil S, Bhutia SK. Autophagy regulates cisplatin-induced stemness and chemoresistance via the upregulation of CD 44, ABCB 1 and ADAM 17 in oral squamous cell carcinoma. Cell Proliferation. 2018;51(1):e12411. doi: 10.1111/cpr.12411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Sotgia F, Ozsvari B, Fiorillo M, De Francesco EM, Bonuccelli G, Lisanti MP. A mitochondrial based oncology platform for targeting cancer stem cells (CSCs): MITO-ONC-RX. Cell Cycle. 2018;17(17):2091–2100. doi: 10.1080/15384101.2018.1515551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Praharaj PP, Panigrahi DP, Bhol CS, Patra S, Mishra SR, Mahapatra KK, Behera BP, Singh A, Patil S, Bhutia SK. Mitochondrial rewiring through mitophagy and mitochondrial biogenesis in cancer stem cells: a potential target for anti-csc cancer therapy. Cancer Lett. 2021;498:217–228. doi: 10.1016/j.canlet.2020.10.036. [DOI] [PubMed] [Google Scholar]
- 23.Ye XQ, Li Q, Wang G-H, Sun F-F, Huang G-J, Bian X-W, Yu S-C, Qian G-S. Mitochondrial and energy metabolism-related properties as novel indicators of lung cancer stem cells. Int J Cancer. 2011;129(4):820–831. doi: 10.1002/ijc.25944. [DOI] [PubMed] [Google Scholar]
- 24.Liu T, Ma Q, Li W, Hu Y, Yang J, Yao Q. Ubiquilin 1 suppresses the cancer stem cell–like traits of non-small cell lung cancer cells by regulating reactive oxygen species homeostasis. Bioengineered. 2021;12(1):7132–7144. doi: 10.1080/21655979.2021.1979353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Dando I, Dalla Pozza E, Biondani G, Cordani M, Palmieri M, Donadelli M. The metabolic landscape of cancer stem cells. IUBMB Life. 2015;67(9):687–693. doi: 10.1002/iub.1426. [DOI] [PubMed] [Google Scholar]
- 26.Chang C-W, Lo J-F, Wang XW. Roles of mitochondria in liver cancer stem cells. Differentiation. 2019;107:35–41. doi: 10.1016/j.diff.2019.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Peiris-Pagès M, Martinez-Outschoorn UE, Pestell RG, Sotgia F, Lisanti MP. Cancer stem cell metabolism. Breast Cancer Res. 2016;18(1):1–10. doi: 10.1186/s13058-016-0712-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Olmedo I, Martínez D, Carrasco-Rojas J, Jara JA. Mitochondria in oral cancer stem cells: unraveling the potential drug targets for new and old drugs. Life Sci. 2023;331:122065. doi: 10.1016/j.lfs.2023.122065. [DOI] [PubMed] [Google Scholar]
- 29.Olivas-Aguirre M, Pottosin I, Dobrovinskaya O. Mitochondria as emerging targets for therapies against T cell acute lymphoblastic leukemia. J Leukocyte Biol. 2019;105(5):935–946. doi: 10.1002/JLB.5VMR0818-330RR. [DOI] [PubMed] [Google Scholar]
- 30.Tan SH, Bertulfo FC, Sanda T. Leukemia-initiating cells in T-cell acute lymphoblastic leukemia. Front Oncol. 2017;7:218. doi: 10.3389/fonc.2017.00218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lato MW, Przysucha A, Grosman S, Zawitkowska J, Lejman M. The new therapeutic strategies in pediatric T-cell acute lymphoblastic leukemia. Int J Mol Sci. 2021;22(9):4502. doi: 10.3390/ijms22094502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Khanam T, Sandmann S, Seggewiss J, Ruether C, Zimmermann M, Norvil AB, Bartenhagen C, Randau G, Mueller S, Herbrueggen H, et al. Integrative genomic analysis of pediatric T-cell lymphoblastic lymphoma reveals candidates of clinical significance. Am J Hematol, Blood. 2021;137(17):2347–2359. doi: 10.1182/blood.2020005381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Inaba H, Mullighan CG. Pediatric acute lymphoblastic leukemia. Haematologica. 2020;105(11):2524. doi: 10.3324/haematol.2020.247031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kato M, Manabe A. Treatment and biology of pediatric acute lymphoblastic leukemia. Pediatrics Int. 2018;60(1):4–12. doi: 10.1111/ped.13457. [DOI] [PubMed] [Google Scholar]
- 35.Prockop S, Wachter F. The current landscape: Allogeneic hematopoietic stem cell transplant for acute lymphoblastic leukemia. Best Pract Res Clin Haematol. 2023;36(3):101485. doi: 10.1016/j.beha.2023.101485. [DOI] [PubMed] [Google Scholar]
- 36.Śliwa-Tytko P, Kaczmarska A, Lejman M, Zawitkowska J. Neurotoxicity associated with treatment of acute lymphoblastic leukemia chemotherapy and immunotherapy. Int J Mol Sci. 2022;23(10):5515. doi: 10.3390/ijms23105515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Summers RJ. High burden of clinically significant adverse events associated with contemporary therapy for pediatric T-cell acute lymphoblastic leukemia/lymphoma. Pediatr Blood Cancer. 2023;70(11):e30571. doi: 10.1002/pbc.30571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Close A. Fertility preservation in pediatric leukemia and lymphoma: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2023; e30407. doi: 10.1002/pbc.30407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Sheykhhasan M, Manoochehri H, Dama P. Use of CAR T-cell for acute lymphoblastic leukemia (ALL) treatment: a review study. Cancer Gene Ther. 2022;29(8):1080–1096. doi: 10.1038/s41417-021-00418-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hucks G, Rheingold SR. The journey to CAR T cell therapy: the pediatric and young adult experience with relapsed or refractory B-ALL. Blood Cancer J. 2019;9(2):10. doi: 10.1038/s41408-018-0164-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Testa U, Sica S, Pelosi E, Castelli G, Leone G. CAR-T cell therapy in B-cell acute lymphoblastic leukemia. Mediterr J Hematol Infect Dis. 2024;16(1):e2024010. doi: 10.4084/MJHID.2024.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Frey NV, Porter DL. Cytokine release syndrome with novel therapeutics for acute lymphoblastic leukemia. Am Soc Hematol Educ Program Book. 2016;2016(1):567–572. doi: 10.1182/asheducation-2016.1.567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 2021;11(4):69. doi: 10.1038/s41408-021-00459-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Fiorenza S, Ritchie DS, Ramsey SD, Turtle CJ, Roth JA. Value and affordability of CAR T-cell therapy in the United States. Bone Marrow Transplant. 2020;55(9):1706–1715. doi: 10.1038/s41409-020-0956-8. [DOI] [PubMed] [Google Scholar]
- 45.Anti-CD7 CAR-T Cell Therapy for Relapse and Refractory CD7 Positive T Cell Malignancies . https://classic.clinicaltrials.gov/show/NCT05290155.
- 46.Chiesa R, Georgiadis C, Ottaviano G, Syed F, Braybrook T, Etuk A, Zhan H, Gkazi SA, Preece R, Adams S, et al. Tvt CAR7: phase 1 clinical trial of base-edited “universal” CAR7 T cells for paediatric relapsed/refractory T-ALL. Blood. 2022;140(Supplement 1):4579–4580. doi: 10.1182/blood-2022-169114. [DOI] [Google Scholar]
- 47.CD7-CAR-T Cells in Pediatric Relapsed/Refractory CD7+ T-ALL/LL . https://classic.clinicaltrials.gov/show/NCT06064903.
- 48.Hunger SP, Raetz EA. How I treat relapsed acute lymphoblastic leukemia in the pediatric population. Blood. 2020;136(16):1803–1812. doi: 10.1182/blood.2019004043. [DOI] [PubMed] [Google Scholar]
- 49.Saygin C, Cannova J, Stock W, Muffly L. Measurable residual disease in acute lymphoblastic leukemia: methods and clinical context in adult patients. Haematologica. 2022;107(12):2783. doi: 10.3324/haematol.2022.280638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Theunissen P, Mejstrikova E, Sedek L, van der Sluijs-Gelling AJ, Gaipa G, Bartels M, Sobral da Costa E, Kotrová M, Novakova M, Sonneveld E, et al. Standardized flow cytometry for highly sensitive MRD measurements in B-cell acute lymphoblastic leukemia. Blood. 2017;129(3):347–357. doi: 10.1182/blood-2016-07-726307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Monter A, Nomdedéu JF. ClonoSEQ assay for the detection of lymphoid malignancies. Expert Rev Mol Diagn. 2019;19(7):571–578. doi: 10.1080/14737159.2019.1627877. [DOI] [PubMed] [Google Scholar]
- 52.Ching T, Duncan ME, Newman-Eerkes T, McWhorter MME, Tracy JM, Steen MS, Brown RP, Venkatasubbarao S, Akers NK, Vignali M, et al. Analytical evaluation of the clonoSEQ Assay for establishing measurable (minimal) residual disease in acute lymphoblastic leukemia, chronic lymphocytic leukemia, and multiple myeloma. BMC Cancer. 2020;20(1):1–15. doi: 10.1186/s12885-020-07077-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Hansen MH, Cédile O, Larsen TS, Abildgaard N, Nyvold CG. Perspective: sensitive detection of residual lymphoproliferative disease by NGS and clonal rearrangements—how low can you go? Exp Hematol. 2021;98:14–24. doi: 10.1016/j.exphem.2021.03.005. [DOI] [PubMed] [Google Scholar]
- 54.Momen N, Tario J, Fu K, Qian Y-W. Multiparameter flow cytometry and ClonoSEQ correlation to evaluate precursor B-lymphoblastic leukemia measurable residual disease. J Hematopathol. 2023;16(2):85–94. doi: 10.1007/s12308-023-00544-9. [DOI] [PubMed] [Google Scholar]
- 55.Othus M, Wood BL, Stirewalt DL, Estey EH, Petersdorf SH, Appelbaum FR, Erba HP, Walter RB. Effect of measurable (‘minimal’) residual disease (MRD) information on prediction of relapse and survival in adult acute myeloid leukemia. Leukemia. 2016;30(10):2080–2083. doi: 10.1038/leu.2016.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Marks DI, Paietta EM, Moorman AV, Richards SM, Buck G, DeWald G, Ferrando A, Fielding AK, Goldstone AH, Ketterling RP, et al. T-cell acute lymphoblastic leukemia in adults: clinical features, immunophenotype, cytogenetics, and outcome from the large randomized prospective trial (UKALL XII/ECOG 2993). Blood. 2009;114(25):5136–5145. doi: 10.1182/blood-2009-08-231217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Reismüller B, Attarbaschi A, Peters C, Dworzak MN, Pötschger U, Urban C, Fink F-M, Meister B, Schmitt K, Dieckmann K, et al. Long-term outcome of initially homogenously treated and relapsed childhood acute lymphoblastic leukaemia in Austria – a population-based report of the Austrian Berlin-Frankfurt-Münster (BFM) Study Group. Br J Haematol. 2009;144(4):559–570. doi: 10.1111/j.1365-2141.2008.07499.x. [DOI] [PubMed] [Google Scholar]
- 58.Belmonte M, Hoofd C, Weng AP, Giambra V. Targeting leukemia stem cells: which pathways drive self-renewal activity in T-cell acute lymphoblastic leukemia? Curr Oncol. 2016;23(1):34–41. doi: 10.3747/co.23.2806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Senft D, Jeremias I. A rare subgroup of leukemia stem cells harbors relapse-inducing potential in acute lymphoblastic leukemia. Exp Hematol. 2019;69:1–10. doi: 10.1016/j.exphem.2018.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Trumpp A, Haas S. Cancer stem cells: the adventurous journey from hematopoietic to leukemic stem cells. Cell. 2022;185(8):1266–1270. doi: 10.1016/j.cell.2022.03.025. [DOI] [PubMed] [Google Scholar]
- 61.Cox CV, Martin HM, Kearns PR, Virgo P, Evely RS, Blair A. Characterization of a progenitor cell population in childhood T-cell acute lymphoblastic leukemia. Blood. 2007;109(2):674–682. doi: 10.1182/blood-2006-06-030445. [DOI] [PubMed] [Google Scholar]
- 62.Chiu PP, Jiang H, Dick JE. Leukemia-initiating cells in human T-lymphoblastic leukemia exhibit glucocorticoid resistance. Blood. 2010;116(24):5268–5279. doi: 10.1182/blood-2010-06-292300. [DOI] [PubMed] [Google Scholar]
- 63.Zhu H, Zhang L, Wu Y, Dong B, Guo W, Wang M, Yang L, Fan X, Tang Y, Liu N, et al. T-ALL leukemia stem cell’stemness’ is epigenetically controlled by the master regulator SPI1. Elife. 2018;7:e38314. doi: 10.7554/eLife.38314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Panelli P, De Santis E, Colucci M, Tamiro F, Sansico F, Miroballo M, Murgo E, Padovano C, Gusscott S, Ciavarella M, et al. Noncanonical β-catenin interactions promote leukemia-initiating activity in early T-cell acute lymphoblastic leukemia. Blood. 2023;141(13):1597–1609. doi: 10.1182/blood.2022017079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Agro L, O’Brien CA. In vitro and in vivo limiting dilution assay for colorectal cancer. Bio-protocol. 2015;5(22):e1659–e1659. doi: 10.21769/BioProtoc.1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Drake AC, Khoury M, Leskov I, Iliopoulou BP, Fragoso M, Lodish H, Chen J. Human CD34+ CD133+ hematopoietic stem cells cultured with growth factors including Angptl5 efficiently engraft adult NOD-SCID Il2rγ−/−(NSG) mice. PLOS ONE. 2011;6(4):e18382. doi: 10.1371/journal.pone.0018382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Al-Hamaly MA. Zebrafish drug screening identifies Erlotinib as an inhibitor of Wnt/β-catenin signaling and self-renewal in T-cell acute lymphoblastic leukemia. Biomed Pharmacother. 2024;170:116013. doi: 10.1016/j.biopha.2023.116013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Blackburn JS, Liu S, Langenau DM. Quantifying the frequency of tumor-propagating cells using limiting dilution cell transplantation in syngeneic zebrafish. JoVE (J Visualized Experiments). 2011;2011(53):e2790. doi: 10.3791/2790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Ho T-C, LaMere M, Stevens BM, Ashton JM, Myers JR, O’Dwyer KM, Liesveld JL, Mendler JH, Guzman M, Morrissette JD, et al. Evolution of acute myelogenous leukemia stem cell properties after treatment and progression. Am J Hematol, Blood. 2016;128(13):1671–1678. doi: 10.1182/blood-2016-02-695312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Hanekamp D, Cloos J, Schuurhuis GJ. Leukemic stem cells: identification and clinical application. Int J Hematol. 2017;105(5):549–557. doi: 10.1007/s12185-017-2221-5. [DOI] [PubMed] [Google Scholar]
- 71.Van Rhenen A, Feller N, Kelder A, Westra AH, Rombouts E, Zweegman S, van der Pol MA, Waisfisz Q, Ossenkoppele GJ, Schuurhuis GJ, et al. High stem cell frequency in acute myeloid leukemia at diagnosis predicts high minimal residual disease and poor survival. Clin Cancer Res. 2005;11(18):6520–6527. doi: 10.1158/1078-0432.CCR-05-0468. [DOI] [PubMed] [Google Scholar]
- 72.Li Y, Wang Z, Ajani JA, Song S. Drug resistance and Cancer stem cells. Cell Commun Signaling. 2021;19(1):1–11. doi: 10.1186/s12964-020-00627-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Vasconcelos FC, de Souza PS, Hancio T, de Faria FCC, Maia RC. Update on drug transporter proteins in acute myeloid leukemia: Pathological implication and clinical setting. Crit Rev Oncol/Hematol. 2021;160:103281. doi: 10.1016/j.critrevonc.2021.103281. [DOI] [PubMed] [Google Scholar]
- 74.Vetrie D, Helgason GV, Copland M. The leukaemia stem cell: similarities, differences and clinical prospects in CML and AML. Nat Rev Cancer. 2020;20(3):158–173. doi: 10.1038/s41568-019-0230-9. [DOI] [PubMed] [Google Scholar]
- 75.Ebinger S, Özdemir EZ, Ziegenhain C, Tiedt S, Castro Alves C, Grunert M, Dworzak M, Lutz C, Turati VA, Enver T, et al. Characterization of rare, dormant, and therapy-resistant cells in acute lymphoblastic leukemia. Cancer Cell. 2016;30(6):849–862. doi: 10.1016/j.ccell.2016.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ng O, Erbilgin Y, Firtina S, Celkan T, Karakas Z, Aydogan G, Turkkan E, Yildirmak Y, Timur C, Zengin E, et al. Deregulated WNT signaling in childhood T-cell acute lymphoblastic leukemia. Blood Cancer J. 2014;4(3):e192–e192. doi: 10.1038/bcj.2014.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Panelli P, Elisabetta DS, Mattia C, Francesco T, Francesca S, Mattia M, Emanuele M. Noncanonical β-catenin interactions promote leukemia-initiating activity in early T-cell acute lymphoblastic leukemia. Blood. 2023;141(13):1597–1609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Lagadinou ED, Sach A, Callahan K, Rossi R, Neering S, Minhajuddin M, Ashton J, Pei S, Grose V, O’Dwyer K, et al. BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell STEM Cell. 2013;12(3):329–341. doi: 10.1016/j.stem.2012.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Plaks V, Kong N, Werb Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell STEM Cell. 2015;16(3):225–238. doi: 10.1016/j.stem.2015.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Oskarsson T, Batlle E, Massagué J. Metastatic stem cells: sources, niches, and vital pathways. Cell STEM Cell. 2014;14(3):306–321. doi: 10.1016/j.stem.2014.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Behrmann L, Wellbrock J, Fiedler W. Acute myeloid leukemia and the bone marrow niche—take a closer look. Front Oncol. 2018;8:444. doi: 10.3389/fonc.2018.00444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Niu J, Peng D, Liu L. Drug resistance mechanisms of acute myeloid leukemia stem cells. Front Oncol. 2022;12:896426. doi: 10.3389/fonc.2022.896426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Moschoi R, Imbert V, Nebout M, Chiche J, Mary D, Prebet T, Saland E, Castellano R, Pouyet L, Collette Y, et al. Protective mitochondrial transfer from bone marrow stromal cells to acute myeloid leukemic cells during chemotherapy. Blood. 2016;128(2):253–264. doi: 10.1182/blood-2015-07-655860. [DOI] [PubMed] [Google Scholar]
- 84.Abdul-Aziz AM, Sun Y, Hellmich C, Marlein CR, Mistry J, Forde E, Piddock RE, Shafat MS, Morfakis A, Mehta T, et al. Acute myeloid leukemia induces protumoral p16INK4a-driven senescence in the bone marrow microenvironment. Blood. 2019;133(5):446–456. doi: 10.1182/blood-2018-04-845420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Bouvy C, Wannez A, Laloy J, Chatelain C, Dogné J-M. Transfer of multidrug resistance among acute myeloid leukemia cells via extracellular vesicles and their microRNA cargo. Leukemia Res. 2017;62:70–76. doi: 10.1016/j.leukres.2017.09.014. [DOI] [PubMed] [Google Scholar]
- 86.Nasr R, Guillemin M-C, Ferhi O, Soilihi H, Peres L, Berthier C, Rousselot P, Robledo-Sarmiento M, Lallemand-Breitenbach V, Gourmel B, et al. Eradication of acute promyelocytic leukemia-initiating cells through PML-RARA degradation. Nat Med. 2008;14(12):1333–1342. doi: 10.1038/nm.1891. [DOI] [PubMed] [Google Scholar]
- 87.Shen Z-X, Shi Z-Z, Fang J, Gu B-W, Li J-M, Zhu Y-M, Shi J-Y, Zheng P-Z, Yan H, Liu Y-F, et al. All-trans retinoic acid/As2O3 combination yields a high quality remission and survival in newly diagnosed acute promyelocytic leukemia. Proc Natl Acad Sci. 2004;101(15):5328–5335. doi: 10.1073/pnas.0400053101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Pollyea DA, Stevens BM, Jones CL, Winters A, Pei S, Minhajuddin M, D’Alessandro A, Culp-Hill R, Riemondy KA, Gillen AE, et al. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat Med. 2018;24(12):1859–1866. doi: 10.1038/s41591-018-0233-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Vazquez R, Breal C, Zalmai L, Friedrich C, Almire C, Contejean A, Barreau S, Grignano E, Willems L, Deau-Fischer B, et al. Venetoclax combination therapy induces deep AML remission with eradication of leukemic stem cells and remodeling of clonal haematopoiesis. Blood Cancer J. 2021;11(3):62. doi: 10.1038/s41408-021-00448-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Tatarek J, Cullion K, Ashworth T, Gerstein R, Aster JC, Kelliher MA. Notch1 inhibition targets the leukemia-initiating cells in a Tal1/Lmo2 mouse model of T-ALL0. Blood. 2011;118(6):1579–1590. doi: 10.1182/blood-2010-08-300343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Habets RA, de Bock CE, Serneels L, Lodewijckx I, Verbeke D, Nittner D, Narlawar R, Demeyer S, Dooley J, Liston A, et al. Safe targeting of T cell acute lymphoblastic leukemia by pathology-specific NOTCH inhibition. Sci Transl Med. 2019;11(494):eaau6246. doi: 10.1126/scitranslmed.aau6246. [DOI] [PubMed] [Google Scholar]
- 92.Armstrong F, de la Grange PB, Gerby B, Rouyez M-C, Calvo J, Fontenay M, Boissel N, Dombret H, Baruchel A, Landman-Parker J, et al. NOTCH is a key regulator of human T-cell acute leukemia initiating cell activity. Blood. 2009;113(8):1730–1740. doi: 10.1182/blood-2008-02-138172. [DOI] [PubMed] [Google Scholar]
- 93.Gerby B, Veiga DFT, Krosl J, Nourreddine S, Ouellette J, Haman A, Lavoie G, Fares I, Tremblay M, Litalien V, et al. High-throughput screening in niche-based assay identifies compounds to target preleukemic stem cells. J Clin Investigation. 2016;126(12):4569–4584. doi: 10.1172/JCI86489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Gerby B, Tremblay CS, Tremblay M, Rojas-Sutterlin S, Herblot S, Hébert J, Sauvageau G, Lemieux S, Lécuyer E, Veiga DFT, et al. SCL, LMO1 and Notch1 reprogram thymocytes into self-renewing cells. PLOS Genet. 2014;10(12):e1004768. doi: 10.1371/journal.pgen.1004768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Colangelo T, Panelli P, Mazzarelli F, Tamiro F, Melocchi V, De Santis E, Cuttano R, Palumbo O, Rossi G, Bianchi F, et al. Extracellular vesicle microRNAs contribute to Notch signaling pathway in T-cell acute lymphoblastic leukemia. Mol Cancer. 2022;21(1):226. doi: 10.1186/s12943-022-01698-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Schubbert S, Cardenas A, Chen H, Garcia C, Guo W, Bradner J, Wu H. Targeting the MYC and PI3K pathways eliminates leukemia-initiating cells in T-cell acute lymphoblastic leukemia. Cancer Res. 2014;74(23):7048–7059. doi: 10.1158/0008-5472.CAN-14-1470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Simioni C, Neri LM, Tabellini G, Ricci F, Bressanin D, Chiarini F, Evangelisti C, Cani A, Tazzari PL, Melchionda F, et al. Cytotoxic activity of the novel Akt inhibitor, MK-2206, in T-cell acute lymphoblastic leukemia. Leukemia. 2012;26(11):2336–2342. doi: 10.1038/leu.2012.136. [DOI] [PubMed] [Google Scholar]
- 98.Guo W, Schubbert S, Chen JY, Valamehr B, Mosessian S, Shi H, Dang NH, Garcia C, Theodoro MF, Varella-Garcia M, et al. Suppression of leukemia development caused by PTEN loss. Proc Natl Acad Sci. 2011;108(4):1409–1414. doi: 10.1073/pnas.1006937108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Lonetti A, Antunes IL, Chiarini F, Orsini E, Buontempo F, Ricci F, Tazzari PL, Pagliaro P, Melchionda F, Pession A, et al. Activity of the pan-class I phosphoinositide 3-kinase inhibitor NVP-BKM120 in T-cell acute lymphoblastic leukemia. Leukemia. 2014;28(6):1196–1206. doi: 10.1038/leu.2013.369. [DOI] [PubMed] [Google Scholar]
- 100.Roderick JE, Tesell J, Shultz LD, Brehm MA, Greiner DL, Harris MH, Silverman LB, Sallan SE, Gutierrez A, Look AT, et al. C-Myc inhibition prevents leukemia initiation in mice and impairs the growth of relapsed and induction failure pediatric T-ALL cells. Blood. 2014;123(7):1040–1050. doi: 10.1182/blood-2013-08-522698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Noguchi K, Itoh M, Ito C, Tohda S. Effects of MYC inhibitors on the growth of acute leukaemia cells. Anticancer Res. 2023;43(7):3023–3028. doi: 10.21873/anticanres.16473. [DOI] [PubMed] [Google Scholar]
- 102.Piya S, Yang Y, Bhattacharya S, Sharma P, Ma H, Mu H, He H, Ruvolo V, Baran N, Davis RE, et al. Targeting the NOTCH1-MYC-CD44 axis in leukemia-initiating cells in T-ALL. Leukemia. 2022;36(5):1261–1273. doi: 10.1038/s41375-022-01516-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Giambra V, Jenkins CE, Lam SH, Hoofd C, Belmonte M, Wang X, Gusscott S, Gracias D, Weng AP. Leukemia stem cells in T-ALL require active Hif1α and Wnt signaling. Blood. 2015;125(25):3917–3927. doi: 10.1182/blood-2014-10-609370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Kaushik B, Pal D, Saha S. Gamma secretase inhibitor: therapeutic target via NOTCH signaling in T cell acute lymphoblastic leukemia. Curr Drug Targets. 2021;22(15):1789–1798. doi: 10.2174/1389450122666210203192752. [DOI] [PubMed] [Google Scholar]
- 105.Samon JB, Castillo-Martin M, Hadler M, Ambesi-Impiobato A, Paietta E, Racevskis J, Wiernik PH, Rowe JM, Jakubczak J, Randolph S, et al. Preclinical analysis of the γ-secretase inhibitor PF-03084014 in combination with glucocorticoids in T-cell acute lymphoblastic leukemia. Mol Cancer Ther. 2012;11(7):1565–1575. doi: 10.1158/1535-7163.MCT-11-0938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Bray SJ. Notch signalling: a simple pathway becomes complex. Nat Rev Mol Cell Biol. 2006;7(9):678–689. doi: 10.1038/nrm2009. [DOI] [PubMed] [Google Scholar]
- 107.A Notch Signalling Pathway Inhibitor for Patients With T-cell Acute Lymphoblastic Leukemia/Lymphoma (ALL)(0752-013) . https://classic.clinicaltrials.gov/show/NCT00100152.
- 108.Papayannidis C, DeAngelo DJ, Stock W, Huang B, Shaik MN, Cesari R, Zheng X, Reynolds JM, English PA, Ozeck M, et al. A Phase 1 study of the novel gamma-secretase inhibitor PF-03084014 in patients with T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma. Blood Cancer J. 2015;5(9):e350–e350. doi: 10.1038/bcj.2015.80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Study to Evaluate the Safety and Tolerability of Weekly Intravenous (IV) Doses of BMS-906024 in Subjects With Acute T-cell Lymphoblastic Leukemia or T-cell Lymphoblastic Lymphoma . https://classic.clinicaltrials.gov/show/NCT01363817.
- 110.Baratta MG. Adjusting the focus on γ-secretase inhibition. Nat Rev Cancer. 2019;19(8):419–419. doi: 10.1038/s41568-019-0174-0. [DOI] [PubMed] [Google Scholar]
- 111.Agnusdei V, Minuzzo S, Frasson C, Grassi A, Axelrod F, Satyal S, Gurney A, Hoey T, Seganfreddo E, Basso G, et al. Therapeutic antibody targeting of Notch1 in T-acute lymphoblastic leukemia xenografts. Leukemia. 2014;28(2):278–288. doi: 10.1038/leu.2013.183. [DOI] [PubMed] [Google Scholar]
- 112.Casulo C, Ruan J, Dang NH, Gore L, Diefenbach C, Beaven AW, Castro JE, Porcu P, Faoro L, Dupont J, et al. Safety and preliminary efficacy results of a phase I first-in-human study of the novel Notch-1 targeting antibody brontictuzumab (OMP-52M51) administered intravenously to patients with hematologic malignancies. Blood. 2016;128(22):5108. doi: 10.1182/blood.V128.22.5108.5108. [DOI] [Google Scholar]
- 113.Martelli AM, Paganelli F, Fazio A, Bazzichetto C, Conciatori F, McCubrey JA. The key roles of PTEN in T-cell acute lymphoblastic leukemia development, progression, and therapeutic response. Cancers. 2019;11(5):629. doi: 10.3390/cancers11050629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Shepherd C, Banerjee L, Cheung CW, Mansour MR, Jenkinson S, Gale RE, Khwaja A. PI3K/mTOR inhibition upregulates NOTCH-MYC signalling leading to an impaired cytotoxic response. Leukemia. 2013;27(3):650–660. doi: 10.1038/leu.2012.285. [DOI] [PubMed] [Google Scholar]
- 115.Palomero T, Ferrando A. Oncogenic NOTCH1 control of MYC and PI3K: challenges and opportunities for anti-NOTCH1 therapy in T-cell acute lymphoblastic leukemias and lymphomas. Clin Cancer Res. 2008;14(17):5314–5317. doi: 10.1158/1078-0432.CCR-07-4864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.MacDonald BT, Tamai K, He X. Wnt/β-catenin signaling: components, mechanisms, and diseases. Dev Cell. 2009;17(1):9–26. doi: 10.1016/j.devcel.2009.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Reya T, Duncan AW, Ailles L, Domen J, Scherer DC, Willert K, Hintz L, Nusse R, Weissman IL. A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature. 2003;423(6938):409–414. doi: 10.1038/nature01593. [DOI] [PubMed] [Google Scholar]
- 118.Wang Y, Krivtsov AV, Sinha AU, North TE, Goessling W, Feng Z, Zon LI, Armstrong SA. The Wnt/β-catenin pathway is required for the development of leukemia stem cells in AML. Science. 2010;327(5973):1650–1653. doi: 10.1126/science.1186624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Zhao C, Blum J, Chen A, Kwon HY, Jung SH, Cook JM, Lagoo A, Reya T. Loss of β-catenin impairs the renewal of normal and CML stem cells in vivo. Cancer Cell. 2007;12(6):528–541. doi: 10.1016/j.ccr.2007.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Jung Y-S, Park J-I. Wnt signaling in cancer: therapeutic targeting of Wnt signaling beyond β-catenin and the destruction complex. Exp Mol Med. 2020;52(2):183–191. doi: 10.1038/s12276-020-0380-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Vander Heiden MG. Targeting cancer metabolism: a therapeutic window opens. Nat Rev Drug Discov. 2011;10(9):671–684. doi: 10.1038/nrd3504. [DOI] [PubMed] [Google Scholar]
- 122.Wen S, Zhu D, Huang P. Targeting cancer cell mitochondria as a therapeutic approach. Future Med Chem. 2013;5(1):53–67. doi: 10.4155/fmc.12.190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Cairns RA, Harris IS, Mak TW. Regulation of cancer cell metabolism. Nat Rev Cancer. 2011;11(2):85–95. doi: 10.1038/nrc2981. [DOI] [PubMed] [Google Scholar]
- 124.Grasso D, Zampieri LX, Capelôa T, Van de Velde JA, Sonveaux P. Mitochondria in cancer. Cell Stress. 2020;4(6):114. doi: 10.15698/cst2020.06.221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Zhang L, Qi J, Zhang X, Zhao X, An P, Luo Y, Luo J. The regulatory roles of mitochondrial calcium and the mitochondrial calcium uniporter in tumor cells. Int J Mol Sci. 2022;23(12):6667. doi: 10.3390/ijms23126667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Guo X, Yang N, Ji W, Zhang H, Dong X, Zhou Z, Li L, Shen H-M, Yao SQ, Huang W, et al. Mito-bomb: targeting mitochondria for cancer therapy. Adv Mater. 2021;33(43):2007778. doi: 10.1002/adma.202007778. [DOI] [PubMed] [Google Scholar]
- 127.Moloney JN, Cotter TG. ROS signalling in the biology of cancer. In: Seminars in cell & developmental biology. Vo. 80. Academic Press; 2018. p. 50–64. [DOI] [PubMed] [Google Scholar]
- 128.Chen Y, Zhang H, Zhou H, Ji W, Min W. Mitochondrial redox signaling and tumor progression. Cancers. 2016;8(4):40. doi: 10.3390/cancers8040040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Mani S, Swargiary G, Ralph SJ. Targeting the redox imbalance in mitochondria: a novel mode for cancer therapy. Mitochondrion. 2022;62:50–73. doi: 10.1016/j.mito.2021.11.002. [DOI] [PubMed] [Google Scholar]
- 130.Baccelli I, Gareau Y, Lehnertz B, Gingras S, Spinella J-F, Corneau S, Mayotte N, Girard S, Frechette M, Blouin-Chagnon V, et al. Mubritinib targets the electron transport chain complex I and reveals the landscape of OXPHOS dependency in acute myeloid leukemia. Cancer Cell. 2019;36(1):84–99. e8. doi: 10.1016/j.ccell.2019.06.003. [DOI] [PubMed] [Google Scholar]
- 131.De Rosa V, Monti M, Terlizzi C, Fonti R, Del Vecchio S, Iommelli F. Coordinate modulation of glycolytic enzymes and OXPHOS by imatinib in BCR-ABL driven chronic myelogenous leukemia cells. Int J Mol Sci. 2019;20(13):3134. doi: 10.3390/ijms20133134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Yuan F, Cheng C, Xiao F, Liu H, Cao S, Zhou G. Inhibition of mTORC1/P70S6K pathway by Metformin synergistically sensitizes Acute Myeloid Leukemia to Ara-C. Life Sci. 2020;243:117276. doi: 10.1016/j.lfs.2020.117276. [DOI] [PubMed] [Google Scholar]
- 133.Yehudai D, Liyanage SU, Hurren R, Rizoska B, Albertella M, Gronda M, Jeyaraju DV, Wang X, Barghout SH, MacLean N, et al. The thymidine dideoxynucleoside analog, alovudine, inhibits the mitochondrial DNA polymerase γ, impairs oxidative phosphorylation and promotes monocytic differentiation in acute myeloid leukemia. haematologica. 2019;104(5):963. doi: 10.3324/haematol.2018.195172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Baran N, Lodi A, Dhungana Y, Herbrich S, Collins M, Sweeney S, Pandey R, Skwarska A, Patel S, Tremblay M, et al. Inhibition of mitochondrial complex I reverses NOTCH1-driven metabolic reprogramming in T-cell acute lymphoblastic leukemia. Nat Commun. 2022;13(1):2801. doi: 10.1038/s41467-022-30396-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Kong H, Reczek CR, McElroy GS, Steinert EM, Wang T, Sabatini DM, Chandel NS. Metabolic determinants of cellular fitness dependent on mitochondrial reactive oxygen species. Sci Adv. 2020;6(45):eabb7272. doi: 10.1126/sciadv.abb7272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Neuzil J, Dong L-F, Rohlena J, Truksa J, Ralph SJ. Classification of mitocans, anti-cancer drugs acting on mitochondria. Mitochondrion. 2013;13(3):199–208. doi: 10.1016/j.mito.2012.07.112. [DOI] [PubMed] [Google Scholar]
- 137.Dong L, Gopalan V, Holland O, Neuzil J. Mitocans revisited: mitochondrial targeting as efficient anti-cancer therapy. Int J Mol Sci. 2020;21(21):7941. doi: 10.3390/ijms21217941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Hulleman E, Kazemier KM, Holleman A, VanderWeele DJ, Rudin CM, Broekhuis MJC, Evans WE, Pieters R, Den Boer ML. Inhibition of glycolysis modulates prednisolone resistance in acute lymphoblastic leukemia cells. Blood. 2009;113(9):2014–2021. doi: 10.1182/blood-2008-05-157842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Goldin N, Arzoine L, Heyfets A, Israelson A, Zaslavsky Z, Bravman T, Bronner V, Notcovich A, Shoshan-Barmatz V, Flescher E, et al. Methyl jasmonate binds to and detaches mitochondria-bound hexokinase. Oncogene. 2008;27(34):4636–4643. doi: 10.1038/onc.2008.108. [DOI] [PubMed] [Google Scholar]
- 140.Jing B, Jin J, Xiang R, Liu M, Yang L, Tong Y, Xiao X, Lei H, Liu W, Xu H, et al. Vorinostat and quinacrine have synergistic effects in T-cell acute lymphoblastic leukemia through reactive oxygen species increase and mitophagy inhibition. Cell Death Disease. 2018;9(6):589. doi: 10.1038/s41419-018-0679-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Xiufeng Z, Haijun Z, Silei B, Manman D, Yong Z, Lian Y, Zhihong F, Bing X. Co-operation of ABT-199 and gemcitabine in impeding DNA damage repair and inducing cell apoptosis for synergistic therapy of T-cell acute lymphoblastic leukemia. Anti-Cancer Drugs. 2019;30(2):138–148. doi: 10.1097/CAD.0000000000000702. [DOI] [PubMed] [Google Scholar]
- 142.Abe M, Noboru A, Maiko K, Chie F, Daisuke Y, Ziyi W, Masayuki M. Antitumor activity of α‐pinene in T‐cell tumors. Canc Sci. 2024;115(4):1317–1332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Wang R, Zhang H, Xu J, Zhang N, Pan T, Zhong X, Zhang H, Yin L, Yao Y, Wu Q, et al. MALT1 inhibition as a therapeutic strategy in T-cell acute lymphoblastic leukemia by blocking Notch1-induced nf-κB activation. Front Oncol. 2020;10:558339. doi: 10.3389/fonc.2020.558339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Ha EJ, Ki, YK, Chae, EK, Do, YJ, Young, HK. Enhancement of quercetin‐induced apoptosis by cotreatment with autophagy inhibitor is associated with augmentation of bak‐dependent mitochondrial pathway in Jurkat T cells. Oxidative Medicine and Cellular Longevity; 2019. p. 7989276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Li Q, Chen Y, Zhao X, Lu B, Qu T, Tang L, Zheng Q. Ginsenoside 24-OH-PD from red ginseng inhibits acute T-lymphocytic leukaemia by activating the mitochondrial pathway. PLOS ONE. 2023;18(5):e0285966. doi: 10.1371/journal.pone.0285966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Shteinfer-Kuzmine A, Amsalem Z, Arif T, Zooravlov A, Shoshan-Barmatz V. Selective induction of cancer cell death by VDAC 1-based peptides and their potential use in cancer therapy. Mol Oncol. 2018;12(7):1077–1103. doi: 10.1002/1878-0261.12313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Jorge J, Magalhães N, Alves R, Lapa B, Gonçalves AC, Sarmento-Ribeiro AB. Antitumor effect of brusatol in acute lymphoblastic leukemia models is triggered by reactive oxygen species accumulation. Biomedicines. 2022;10(9):2207. doi: 10.3390/biomedicines10092207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Uscanga-Palomeque AC, Calvillo-Rodríguez KM, Gómez-Morales L, Lardé E, Denèfle T, Caballero-Hernández D, Merle-Béral H, Susin SA, Karoyan P, Martínez-Torres AC, et al. CD 47 agonist peptide PKHB 1 induces immunogenic cell death in T-cell acute lymphoblastic leukemia cells. Cancer Sci. 2019;110(1):256–268. doi: 10.1111/cas.13885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Bornhauser BC, Bonapace L, Lindholm D, Martinez R, Cario G, Schrappe M, Niggli FK, Schäfer BW, Bourquin J-P. Low-dose arsenic trioxide sensitizes glucocorticoid-resistant acute lymphoblastic leukemia cells to dexamethasone via an akt-dependent pathway. Blood. 2007;110(6):2084–2091. doi: 10.1182/blood-2006-12-060970. [DOI] [PubMed] [Google Scholar]
- 150.Olivas-Aguirre M, Torres-López L, Gómez-Sandoval Z, Villatoro-Gómez K, Pottosin I, Dobrovinskaya O. Tamoxifen sensitizes acute lymphoblastic leukemia cells to cannabidiol by targeting cyclophilin-D and altering mitochondrial Ca2+ homeostasis. Int J Mol Sci. 2021;22(16):8688. doi: 10.3390/ijms22168688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Wang Y-P, Pan F, Wang Y-D, Khan A, Liu Y-P, Yang M-L, Cao J-X, Zhao T-R, Cheng G-G. Anti-leukemic effect and molecular mechanism of 11-methoxytabersonine from Melodinus cochinchinensis via network pharmacology, ros-mediated mitochondrial dysfunction and PI3K/Akt signaling pathway. Bioorg Chem. 2022;120:105607. doi: 10.1016/j.bioorg.2022.105607. [DOI] [PubMed] [Google Scholar]
- 152.Chen L, Fang W, Liu J, Qi X, Zhao L, Wang Y, Liu Y, Kong D, Sun X, Li X, et al. Poricoic acid a (PAA) inhibits T-cell acute lymphoblastic leukemia through inducing autophagic cell death and ferroptosis. Biochem Biophys Res Commun. 2022;608:108–115. doi: 10.1016/j.bbrc.2022.03.105. [DOI] [PubMed] [Google Scholar]
- 153.Ruiz-Moreno C, Velez-Pardo C, Jimenez-Del-Rio M. Minocycline induces apoptosis in acute lymphoblastic leukemia Jurkat cells. Toxicol Vitro. 2018;50:336–346. doi: 10.1016/j.tiv.2018.03.012. [DOI] [PubMed] [Google Scholar]
- 154.Soto-Mercado V, Mendivil-Perez M, Jimenez-Del-Rio M, Fox JE, Velez-Pardo C. Cannabinoid CP55940 selectively induces apoptosis in Jurkat cells and in ex vivo T-cell acute lymphoblastic leukemia through H2O2 signaling mechanism. Leukemia Res. 2020;95:106389. doi: 10.1016/j.leukres.2020.106389. [DOI] [PubMed] [Google Scholar]
- 155.Luo H, Jing B, Xia Y, Zhang Y, Hu M, Cai H, Tong Y, Zhou L, Yang L, Yang J, et al. WP1130 reveals USP24 as a novel target in T-cell acute lymphoblastic leukemia. Cancer Cell Int. 2019;19(1):1–14. doi: 10.1186/s12935-019-0773-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Olivas-Aguirre M, Torres-López L, Valle-Reyes JS, Hernández-Cruz A, Pottosin I, Dobrovinskaya O. Cannabidiol directly targets mitochondria and disturbs calcium homeostasis in acute lymphoblastic leukemia. Cell Death Dis. 2019;10(10):779. doi: 10.1038/s41419-019-2024-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Alonso-Alonso R, Mondéjar R, Martínez N, García-Diaz N, Pérez C, Merino D, Rodríguez M, Esteve-Codina A, Fuste B, Gut M, et al. Identification of tipifarnib sensitivity biomarkers in T-cell acute lymphoblastic leukemia and T-cell lymphoma. Scientific Rep. 2020;10(1):6721. doi: 10.1038/s41598-020-63434-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Yu H, Yin Y, Yi Y, Cheng Z, Kuang W, Li R, Zhong H, Cui Y, Yuan L, Gong F, et al. Targeting lactate dehydrogenase a (LDHA) exerts antileukemic effects on T-cell acute lymphoblastic leukemia. Cancer Commun. 2020;40(10):501–517. doi: 10.1002/cac2.12080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Zeng L, Tang M, Pi C, Zheng J, Gao S, Chabanne T, Chauvin R, Cheng W, Lin H, Xu R, et al. Novel ferrocene derivatives induce apoptosis through mitochondria-dependent and cell cycle arrest via PI3K/Akt/mTOR signaling pathway in T cell acute lymphoblastic leukemia. Cancers. 2021;13(18):4677. doi: 10.3390/cancers13184677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.García-Cañaveras JC, Lancho O, Ducker GS, Ghergurovich JM, Xu X, da Silva-Diz V, Minuzzo S, Indraccolo S, Kim H, Herranz D, et al. SHMT inhibition is effective and synergizes with methotrexate in T-cell acute lymphoblastic leukemia. Leukemia. 2021;35(2):377–388. doi: 10.1038/s41375-020-0845-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Koszałka P, Stasiłojć G, Miękus-Purwin N, Niedźwiecki M, Purwin M, Grabowski S, Bączek T. The cooperative anti-neoplastic activity of polyphenolic phytochemicals on human T-cell acute lymphoblastic leukemia cell line MOLT-4 in vitro. Int J Mol Sci. 2022;23(9):4753. doi: 10.3390/ijms23094753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Gasanoff E, Liu Y, Li F, Hanlon P, Garab G. Bee venom melittin disintegrates the respiration of mitochondria in healthy cells and lymphoblasts, and induces the formation of non-bilayer structures in model inner mitochondrial membranes. Int J Mol Sci. 2021;22(20):11122. doi: 10.3390/ijms222011122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Olivas-Aguirre M, Torres-López L, Pottosin I, Dobrovinskaya O. Phenolic compounds cannabidiol, curcumin and quercetin cause mitochondrial dysfunction and suppress acute lymphoblastic leukemia cells. Int J Mol Sci. 2020;22(1):204. doi: 10.3390/ijms22010204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Cruz P, Ahumada-Castro U, Bustos G, Molgó J, Sauma D, Lovy A, Cárdenas C. Inhibition of InsP3R with Xestospongin B reduces mitochondrial respiration and induces selective cell death in T cell acute lymphoblastic leukemia cells. Int J Mol Sci. 2021;22(2):651. doi: 10.3390/ijms22020651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Torres-López L, Maycotte P, Liñán-Rico A, Liñán-Rico L, Donis-Maturano L, Delgado-Enciso I, Meza-Robles C, Vásquez-Jiménez C, Hernández-Cruz A, Dobrovinskaya O, et al. Tamoxifen induces toxicity, causes autophagy, and partially reverses dexamethasone resistance in Jurkat T cells. J Leukocyte Biol. 2019;105(5):983–998. doi: 10.1002/JLB.2VMA0818-328R. [DOI] [PubMed] [Google Scholar]
- 166.Zhong M, Lin F, Jiang Y, Pan G, Tan J, Zhou H, Lai Q, Chen Q, Deng M, Zha J, et al. Therapeutic interaction of Apatinib and Chidamide in T-Cell acute lymphoblastic Leukemia through interference with mitochondria associated biogenesis and intrinsic apoptosis. J Personalized Med. 2021;11(10):977. doi: 10.3390/jpm11100977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Zhang G, Gao X, Zhao X, Wu H, Yan M, Li Y, Zeng H, Ji Z, Guo X. Decitabine inhibits the proliferation of human T‑cell acute lymphoblastic leukemia molt4 cells and promotes apoptosis partly by regulating the PI3K/AKT/mTOR pathway. Oncol Lett. 2021;21(5):1–10. doi: 10.3892/ol.2021.12601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Chi Z, Gao H, Liu H, Wu B, Zhang B, Gu M, Yang W. Chidamide induces necroptosis via regulation of c‑flipl expression in Jurkat and HUT‑78 cells. Mol Med Rep. 2020;21(2):936–944. doi: 10.3892/mmr.2019.10873. [DOI] [PubMed] [Google Scholar]
- 169.Hong Y, Zhang J, Guo Q, Zhu M, Chen B, Luo W. Diacetyl hexamethylene diamine (CAHB) exerts pro-apoptotic and anti-proliferative function in leukemic T lymphocytes via downregulating PI3K/Akt signaling. Med Sci Monit Int Med J Exp Clin Res. 2019;25:5211. doi: 10.12659/MSM.915840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Mendivil-Perez M, Jimenez-Del-Rio M, Velez-Pardo C. Combinational treatment of TPEN and TPGS induces apoptosis in acute lymphoblastic and chronic myeloid leukemia cells in vitro and ex vivo. Med Oncol. 2022;39(7):109. doi: 10.1007/s12032-022-01697-w. [DOI] [PubMed] [Google Scholar]
- 171.Kang MH, Kang YH, Szymanska B, Wilczynska-Kalak U, Sheard MA, Harned TM, Lock RB, Reynolds CP. Activity of vincristine, L-ASP, and dexamethasone against acute lymphoblastic leukemia is enhanced by the BH3-mimetic ABT-737 in vitro and in vivo. Blood. 2007;110(6):2057–2066. doi: 10.1182/blood-2007-03-080325. [DOI] [PubMed] [Google Scholar]
- 172.Del Gaizo Moore V, Schlis KD, Sallan SE, Armstrong SA, Letai A. BCL-2 dependence and ABT-737 sensitivity in acute lymphoblastic leukemia. Blood. 2008;111(4):2300–2309. doi: 10.1182/blood-2007-06-098012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Suryani S, Carol H, Chonghaile TN, Frismantas V, Sarmah C, High L, Bornhauser B, Cowley MJ, Szymanska B, Evans K, et al. Cell and molecular determinants of in vivo efficacy of the BH3 mimetic ABT-263 against pediatric acute lymphoblastic leukemia xenografts. Clin Cancer Res. 2014;20(17):4520–4531. doi: 10.1158/1078-0432.CCR-14-0259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Sassi N, Mattarei A, Azzolini M, Bernardi P, Szabo’ I, Paradisi C, Zoratti M, Biasutto L. Mitochondria-targeted resveratrol derivatives act as cytotoxic pro-oxidants. Curr Pharm Des. 2014;20(2):172–179. doi: 10.2174/13816128113199990034. [DOI] [PubMed] [Google Scholar]
- 175.Bonilla-Porras AR, Jimenez-Del-Rio M, Velez-Pardo C. Vitamin K3 and vitamin C alone or in combination induced apoptosis in leukemia cells by a similar oxidative stress signalling mechanism. Cancer Cell Int. 2011;11(1):1–11. doi: 10.1186/1475-2867-11-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Fu X. Targeting mitochondrial respiration selectively sensitizes pediatric acute lymphoblastic leukemia cell lines and patient samples to standard chemotherapy. Am J Cancer Res. 2017;7(12):2395. [PMC free article] [PubMed] [Google Scholar]
- 177.Kanno S-I. Susceptibility to cytosine arabinoside (Ara-c)-induced cytotoxicity in human leukemia cell lines. Toxicol Lett. 2004;152(2):149–158. doi: 10.1016/j.toxlet.2004.04.014. [DOI] [PubMed] [Google Scholar]
- 178.Boukalova S, Katerina R, Jakub R, Jiri N. Mitocans: mitochondrially targeted anti-cancer drugs. Mitochondrial Biol Exp Ther. 2018;613–635. [Google Scholar]
- 179.Tong Y, Gao W-Q, Liu Y. Metabolic heterogeneity in cancer: an overview and therapeutic implications. Biochim et Biophys Acta (BBA)-Rev Cancer. 2020;1874(2):188421. doi: 10.1016/j.bbcan.2020.188421. [DOI] [PubMed] [Google Scholar]
- 180.Gentric G, Mieulet V, Mechta-Grigoriou F. Heterogeneity in cancer metabolism: new concepts in an old field. Antioxid Redox Signaling. 2017;26(9):462–485. doi: 10.1089/ars.2016.6750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Snyder V, Reed-Newman TC, Arnold L, Thomas SM, Anant S. Cancer stem cell metabolism and potential therapeutic targets. Front Oncol. 2018;8:203. doi: 10.3389/fonc.2018.00203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Peng M, Huang Y, Zhang L, Zhao X, Hou Y. Targeting mitochondrial oxidative phosphorylation eradicates acute myeloid leukemic stem cells. Front Oncol. 2022;12:899502. doi: 10.3389/fonc.2022.899502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Raffel S, Klimmeck D, Falcone M, Demir A, Pouya A, Zeisberger P, Lutz C, Tinelli M, Bischel O, Bullinger L, et al. Quantitative proteomics reveals specific metabolic features of acute myeloid leukemia stem cells. Blood. 2020;136(13):1507–1519. doi: 10.1182/blood.2019003654. [DOI] [PubMed] [Google Scholar]
- 184.Jones CL, Stevens BM, D’Alessandro A, Reisz JA, Culp-Hill R, Nemkov T, Pei S, Khan N, Adane B, Ye H, et al. Inhibition of amino acid metabolism selectively targets human leukemia stem cells. Cancer Cell. 2018;34(5):724–740.e4. doi: 10.1016/j.ccell.2018.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Burt R, Dey A, Aref S, Aguiar M, Akarca A, Bailey K, Day W, Hooper S, Kirkwood A, Kirschner K, et al. Activated stromal cells transfer mitochondria to rescue acute lymphoblastic leukemia cells from oxidative stress. Blood. 2019;134(17):1415–1429. doi: 10.1182/blood.2019001398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Zhong W, Yi Q, Xu B, Li S, Wang T, Liu F, Zhu B, Hoffmann PR, Ji G, Lei P, et al. ORP4L is essential for T-cell acute lymphoblastic leukemia cell survival. Nat Commun. 2016;7(1):12702. doi: 10.1038/ncomms12702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Zhong W, Xu M, Li C, Zhu B, Cao X, Li D, Chen H, Hu C, Li R, Luo C, et al. ORP4L extracts and presents PIP2 from plasma membrane for PLCβ3 catalysis: targeting it eradicates leukemia stem cells. Cell Rep. 2019;26(8):2166–2177.e9. doi: 10.1016/j.celrep.2019.01.082. [DOI] [PubMed] [Google Scholar]
- 188.Zhong W, Cao X, Pan G, Niu Q, Feng X, Xu M, Li M, Huang Y, Yi Q, Yan D. ORP4L is a prerequisite for the induction of T-cell leukemogenesis associated with human T-cell leukemia virus 1. Blood. 2022;139(7):1052–1065. doi: 10.1182/blood.2021013579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Fahy L, Calvo J, Chabi S, Renou L, Le Maout C, Poglio S, Leblanc T, Petit A, Baruchel A, Ballerini P, et al. Hypoxia favors chemoresistance in T-ALL through an HIF1α-mediated mTORC1 inhibition loop. Blood Adv. 2021;5(2):513–526. doi: 10.1182/bloodadvances.2020002832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Gachet S, El-Chaar T, Avran D, Genesca E, Catez F, Quentin S, Delord M, Thérizols G, Briot D, Meunier G, et al. Deletion 6q drives T-cell leukemia progression by ribosome modulation. Cancer Discov. 2018;8(12):1614–1631. doi: 10.1158/2159-8290.CD-17-0831. [DOI] [PubMed] [Google Scholar]
- 191.Georgievski A, Michel A, Thomas C, Mlamla Z, Pais de Barros J-P, Lemaire-Ewing S, Garrido C, Quéré R. Acute lymphoblastic leukemia-derived extracellular vesicles affect quiescence of hematopoietic stem and progenitor cells. Cell Death Disease. 2022;13(4):337. doi: 10.1038/s41419-022-04761-5. [DOI] [PMC free article] [PubMed] [Google Scholar]