Skip to main content
Redox Biology logoLink to Redox Biology
. 2023 Apr 2;62:102692. doi: 10.1016/j.redox.2023.102692

Emerging drugs targeting cellular redox homeostasis to eliminate acute myeloid leukemia stem cells

Rafaela GA Costa a, Suellen LR Silva a, Ingrid RSB Dias a, Maiara de S Oliveira a, Ana Carolina B da C Rodrigues a, Rosane B Dias a,b, Daniel P Bezerra a,
PMCID: PMC10119960  PMID: 37031536

Abstract

Acute myeloid leukemia (AML) is a very heterogeneous group of disorders with large differences in the percentage of immature blasts that presently are classified according to the specific mutations that trigger malignant proliferation among thousands of mutations reported thus far. It is an aggressive disease for which few targeted therapies are available and still has a high recurrence rate and low overall survival. The main reason for AML relapse is believed to be due to leukemic stem cells (LSCs) that have unlimited self-renewal capacity and long residence in a quiescent state, which promote greater resistance to traditional therapies for this cancer. AML LSCs have low oxidative stress levels, which appear to be caused by a combination of low mitochondrial activity and high activity of ROS-removing pathways. In this sense, oxidative stress has been thought to be an important new potential target for the treatment of AML patients, targeting the eradication of AML LSCs. The aim of this review is to discuss some drugs that induce oxidative stress to direct new goals for future research focusing on redox imbalance as an effective strategy to eliminate AML LSCs.

Keywords: Acute myeloid leukemia, Leukemia stem cells, Oxidative stress

1. Introduction

Acute myeloid leukemia (AML) is a very heterogeneous group of disorders with large differences in the percentage of immature blasts that presently are classified according to the specific mutations that trigger malignant proliferation among thousands of mutations reported thus far [[1], [2], [3], [4], [5]]. The American Cancer Society estimated 20,380 new cases of AML and 11,310 deaths in the United States for 2023 [6].

According to the French-American-British (FAB) classification, AML can be subclassified into undifferentiated acute myeloblastic leukemia, acute myeloblastic leukemia with minimal maturation, acute myeloblastic leukemia with maturation, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute myelomonocytic leukemia with eosinophilia, acute monocytic leukemia, acute erythroid leukemia and acute megakaryoblastic leukemia [[7], [8], [9], [10]].

In the most recent update of the World Health Organization (WHO) AML classification, there is a separation of AML with defining genetic abnormalities from AML defined by differentiation. AML with defining genetic abnormalities includes acute promyelocytic leukemia with PML::RARA fusion; AML with RUNX1::RUNX1T1 fusion; AML with CBFB::MYH11 fusion; AML with DEK::NUP214 fusion; AML with RBM15::MRTFA fusion; AML with BCR::ABL1 fusion; AML with KMT2A rearrangement; AML with MECOM rearrangement; AML with NUP98 rearrangement; AML with NPM1 mutation; AML with CEBPA mutation; AML myelodysplasia-related; and AML with other defined genetic alterations. AML defined by differentiation includes AML with minimal differentiation, AML without maturation, AML with maturation, acute basophilic leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroid leukemia, and acute megakaryoblastic leukemia [11].

AML can affect individuals of any age; although it is the most common malignancy in children aged 15 years or younger, it becomes more common with aging. Generally, adults correspond to the group of individuals who are most affected by this type of neoplasm, which reduces the success of therapeutic alternatives due to age. Thus, the disease has a poor prognosis when compared to younger individuals affected by AML [4,[12], [13], [14]].

AML is an aggressive disease for which few targeted therapies are available. The standard protocol for treatment has remained almost unchanged for more than four decades and basically consists of the administration of cytarabine (100 mg/m2/day by continuous infusion for 7 days) combined with anthracycline (daunorubicin 60 mg/m2/day or idarubicin 12 mg/m2/day for 3 days), where blasts can often be eliminated, thus achieving remission [15].

Usually, the patient undergoes induction therapy with the aim of achieving complete remission, and all of the leukemic cells must be destroyed. Although most patients are successful in remission, in many cases, minimal residual disease (MRD) remains, consequently increasing the rate of disease relapse. In this context, AML currently still has a high recurrence rate and low overall survival [[16], [17], [18], [19]]. The main reason for AML relapse is believed to be due to a subpopulation of leukemia cells called leukemia stem cells (LSCs) or leukemia initiating cells (LICs) [3,[20], [21], [22], [23], [24]].

AML LSCs are a rare blast immature population that are normally in a quiescent state and can become active as they have unlimited self-renewal capacity that continuously propagates leukemic cell clones and a large number of mature AML blast cells. Due to their unlimited self-renewal capacity, these cells are characterized by their high repopulation potential and long residence in a quiescent state of the G0 phase, playing a key role in AML relapse and refractoriness [3,[24], [25], [26], [27]].

AML LSCs have very similar characteristics to normal hematopoietic stem cells (HSCs). The cell cycle quiescence and the high capacity for self-renewal make it possible to use similar molecular mechanisms for their self-renewal and maintenance of an undifferentiated state that allows their differentiation into more developed progenitor cells. Both AML LSCs and HSCs express a CD34+CD38 phenotype [23,[28], [29], [30], [31], [32]]. In addition, other markers, including CD123 [33], CD96 [34], CLL-1 [35], TIM-3 [36], CD93 [37], and CD99 [38], have also been found to be expressed in AML LSCs.

Studies have shown that high levels of oxidative stress are found in AML, which seems to affect the proliferation, differentiation and self-renewal capacity of HSCs, consequently impairing cell growth. This can be explained by the fact that oxidative stress causes a redox imbalance, which induces cell damage [39,40]. Under normal conditions, controlled levels of oxidative stress play an important role in the self-renewal, regeneration and proliferation of HSCs. On the other hand, elevated levels of oxidative stress induce fatal oxidative damage to DNA and cellular proteins, thus contributing to the differentiation of leukemic blasts and favoring the development of AML [41,42].

Interestingly, AML LSCs have low oxidative stress levels, which appear to be caused by a combination of low mitochondrial activity and high activity of ROS-removing pathways. LSCs are highly dependent on mitochondrial regulatory mechanisms and are sensitive to oxidative stress as a result of their low activity [42,43]. In this sense, oxidative stress is thought of as an important new potential target for the treatment of AML, including AML LSCs. The aim of this review is to discuss some drugs that induce oxidative stress and are able to eliminate AML LSCs to direct new goals for future research. Searches were carried out in the scientific database PubMed comprising all papers in English published until November 2022.

1.1. Cellular redox homeostasis

Aerobic organisms use oxygen for cellular respiration, but this process produces free radicals, which are molecular fragments that can exist independently and attempt to bond with other molecules, atoms, or even individual electrons to form a stable compound. The transfer of one or more electrons from a donor to an acceptor is referred to as an oxidative process [[44], [45], [46], [47]].

Cellular redox homeostasis is maintained by the balance between oxidizing agents and the cellular antioxidant defense system. These oxidizing agents are reactive species, which include reactive oxygen species (ROS), reactive nitrogen species (RNS), and less commonly reactive sulfur species (RSS), and their excess in cells is known as oxidative stress [[47], [48], [49], [50], [51], [52]]. These reactive species can be free radicals or nonfree radical compounds (Table 1). In living organisms, nonfree radical species easily initiate free radical reactions, and oxidative stress is usually caused by excess ROS [47]. Most biological processes are regulated by redox interactions, which play an important role in the regulation of cell signaling, development, health and pathologies [53].

Table 1.

Examples of ROS, RNS and RSS of biological interest.

Free radical
Nonfree radical
Name Symbol Name Symbol
ROS
Oxygen (biradical) O2●● Hydrogen peroxide H2O2
Superoxide ion O2 Ozone O3
Hydroxyl OH Singlet oxygen 1O2
Hydroperoxyl HO2 Hypobromous acid HOBr
Carbonate CO3 Hypochlorous acid HOCl
Peroxyl ROO Hypoiodous acid HOI
Alkoxyl RO Organic peroxides ROOH
Carbon dioxide radical CO2 Peroxynitrite ONOO
Peroxynitrate O2NOO
Peroxynitrous acid ONOOH
Peroxomono-carbonate HOOCO2‾
Carbon monoxide CO
RNS
Nitric oxide NO Nitrosyl cation NO+
Nitrogen dioxide NO2 Nitrous acid HNO2
Nitrate radical NO3 Nitroxyl anion NO
Dinitrogen trioxide N2O3
Dinitrogen tetroxide N2O4
Dinitrogen pentoxide N2O5
Alkyl peroxynitrites ROONO
Alkyl peroxynitrates RO2ONO
Nitryl chloride NO2Cl
Peroxyacetyl nitrate CH3C(O)OONO2
RSS
Thiyl radical S RS Hydrogen sulfide H2S
Disulfide RSSR
Disulfide-S-monoxide RS(O)SR
Disulfide-S-dioxide RS(O)2SR
Sulfenic acid RSOH
Thiol/sulfide RSR

Oxidizing compounds can come from endogenous and exogenous sources. In mammals, the primary source of ROS is mitochondrial energy production during ATP production by the mitochondrial oxidative phosphorylation system (OXPHOS). ROS are produced by reactions in the electron transport chain (ETC) [42,47,[54], [55], [56]]. Exogenous sources of oxidizing compounds include radiation, chemical reagents, certain foods and environmental pollutants, and drugs and their metabolites [47,57].

Free radicals can affect all cellular macromolecules and cause cell and tissue damage. As a result, these radicals may cause damage and activate redox signaling pathways, and excess free radicals can lead to cell death. Under physiological conditions, the effects of these compounds are seen only locally where they are produced [[57], [58], [59], [60]].

Antioxidants inhibit oxidation and counteract oxidative stress. These antioxidants can be endogenous or exogenous [61]. Endogenous antioxidants can be enzymatic or nonenzymatic. Enzymatic antioxidants include superoxide dismutases (SODs), superoxide reductases (SORs), catalase, glutathione peroxidase (GPx), glutathione reductase (GRx), glutathione-S-transferase (GST), thioredoxin reductase (TrxR) and peroxiredoxins (PRXs). Lipoic acid, glutathione, thioredoxin and l-arginine are considered nonenzymatic. On the other hand, some antioxidant nutrients cannot be produced by the body and must be obtained exogenously through foods [47,58,[62], [63], [64], [65]].

The summarized mechanism by which these molecules carry out their antioxidant action is shown in Fig. 1. Interestingly, antioxidant enzymes that control ROS homeostasis are altered in several diseases, including cancer [66].

Fig. 1.

Fig. 1

Conversion of superoxide to water and oxygen. The enzyme superoxide dismutase converts superoxide to hydrogen peroxide. It can be detoxified to water and oxygen using catalase or glutathione peroxidase (GSH, reduced glutathione; GSSG, oxidized glutathione). Hydrogen peroxide spontaneously degrades to the reactive hydroxyl radical (OH).

SODs convert peroxide into hydrogen peroxide and oxygen. There are five types of SODs that differ by the ion in their catalytic site, including Fe, Mn, Ni, Cu or Cu and Zn [67]. In mammals, the main SOD family is SOD1, with Cu and Zn in the cytoplasm. SOD2 with Mn is in the mitochondrial matrix, and both have tumor suppressor activity [68]. SOD2 protects cells against oxidative stress, and its loss leads to defective hematopoiesis [69,70]. Polymorphisms in SODs lead to high levels of ROS, which may contribute to the malignant transformation of cells [71].

SORs reduce superoxide and are found in various organisms. They have a unique iron ion catalytic site and need electron donors to regenerate. They are classified by metallic centers and extra domains [67,72,73]. Aside from their antioxidant activity in reducing ROS, little is known about the role of SORs in leukemia and LSCs.

Catalase enzymes convert H2O2 into O2 and H2O. They can be classified into three types based on structure and function, for example, typical catalases, peroxidase catalases and manganese catalases. Human catalase is a typical catalase with an iron protoporphyrin IX active center and NADPH cofactor [[74], [75], [76]]. CATs are crucial in protecting cells against oxidative stress and are found in peroxisomes in normal cells. In cancer cells, they can impede apoptosis. CAT's role in tumors is complex and can act as both a survival enhancer and tumor suppressor. Studies show that elevated catalase expression in AML is associated with a positive therapy response, and genetic polymorphisms can alter catalase expression and susceptibility to oxidative damage [66,71,75,76].

Glutathione is known as the most abundant antioxidant in the cell [77]. Glutathiones exist in oxidized and reduced forms: glutathione in its oxidized (GSSG) and reduced (GSH) states. GST is an enzyme that helps detoxify by conjugating GSH to xenobiotics. It helps metabolize and detoxify various harmful substances and protect tissues against oxidative damage [[78], [79], [80]].

GRx is a GST helper enzyme; together, GST and GRx use NADPH to regenerate GSH from GSSG (also known as the glutathione antioxidant system). Thus, glutathione functions as a direct scavenger of ROS, and polymorphisms in the GST gene have been related to failures in enzymatic activity, implying greater susceptibility to leukemia [70,71].

The maintenance of redox homeostasis can be guaranteed by the stability of GSH/GSSG, which enables normal biological processes; on the other hand, the accumulation of GSSG/GSH under abnormal conditions can result in cell death mediated by oxidative stress [56].

GPXs have selenocysteine in their active sites and four isoforms: GPX1-4. GPX1 is the most abundant and codes for mitochondrial peroxide glutathione. GPX1 gene polymorphism can decrease enzyme activity and increase cancer risk in Caucasians [71,81]. GPX4 has been shown to be an important enzyme in the detoxification of lipid hydroperoxides [70].

TrxR is part of the thioredoxin antioxidant system, which includes NADPH, TrxR, thioredoxin and TXNIP. TXNIP inhibits thioredoxin activity by binding to it. TrxR is a selenoenzyme that uses NADPH to keep thioredoxin reduced [[82], [83], [84]]. Mammals have three TrxR isoforms with different locations and expression levels: TrxR 1 in the cytoplasm, TrxR 2 in mitochondria and TrxR 3 in testis. Two thioredoxin isoforms (Trx1 and Trx2) are also found in the cytoplasm and mitochondria [[82], [83], [84]]. In particular, overexpression of thioredoxin 1 enhanced the antitumor function of T cells [85].

PRXs are a family of six small antioxidant proteins with essential cysteine residues that scavenge cellular ROS. Cysteine residues can be oxidized and generate intermolecular disulfides with nearby proteins, which are reduced by thioredoxin. PRXs I-IV have sequences for CDK phosphorylation and may regulate the cell cycle [70,[86], [87], [88]]. PRX I is produced during serum stimulation and oxidative stress. Its loss can cause hemolytic anemia and increased malignancies. PRX II loss causes hemolytic anemia compensated by increased erythroid progenitor activity. PRX III is mitochondrial-specific and converts hydrogen peroxide to water. Its deficiency causes ROS accumulation and may regulate mitochondria-mediated apoptosis [70,[88], [89], [90]].

1.2. Pleiotropic effect of oxidative stress in AML

Oxidative stress has both tumor-promoting and tumor-suppressing functions [65]. Furthermore, the physiological level of ROS is important for the self-renewal, differentiation and proliferation of HSCs [42,91,92].

HSCs reside in bone marrow in low-oxygen environments known as stem cell niches. These hypoxic conditions cause metabolic changes such as low mitochondrial activity and low membrane potential [[93], [94], [95], [96]]. Quiescent HSCs primarily generate energy through anaerobic glycolysis, which reduces the burden of ROS-mediated oxidative stress [42,97].

Moreover, HSCs also use the cellular antioxidant system, including antioxidant molecules (e.g., glutathione, thioredoxin, NADPH) and antioxidant enzymes (e.g., catalase, SOD, glutathione peroxidase). Other ROS regulatory mechanisms used by HSCs include the interactions of cells with their microenvironment [98] and mitophagy [99], a subtype of autophagy where selective degradation of mitochondria occurs, which controls mitochondrial quantity and quality.

On the other hand, an increase in ROS (moderate level of ROS) is critical during HSC differentiation, which occurs with an increase in mitochondrial activity through OXPHOS [42,92,93]. Therefore, HSCs can self-renew (when ROS levels are reduced) or differentiate (when ROS levels are moderate/higher) depending on the ROS level [100,101].

Nevertheless, HSCs are susceptible to ROS accumulation [70]. Thus, when there is a high concentration of ROS, there can be damage to cell functions and structures that can result in cell death or abnormal hematopoiesis, with malignant transformation to LSCs being a crucial event for the progression of hematological malignancies such as AML and its aggressiveness [40,102]. Studies suggest that ROS accumulation actively participates in the conversion of normal HSCs into leukemic cells [70,103].

Interestingly, the amount of intracellular ROS also differs in AML cell subpopulations, where bulk AML cells have a high level of ROS and AML LSCs, such as HSCs, have a low level of ROS [42,104]. AML blasts present high levels of oxygen-derived radicals and thus greater endogenous oxidative stress than normal leukocytes. The antioxidant system in these cells is normally deficient, so high and persistent levels of ROS produce DNA damage leading to mutagenesis [40,71].

In AML cells, the main form of ROS is the superoxide anion (O2•-) [40]. In primary human AML cells, where the intrinsic property is the altered metabolism of glutathione, it is possible to observe oxidative stress that contributes to leukemogenesis [105]. Reduced catalase expression may also be responsible for H2O2 accumulation in leukemic cells [69]. Furthermore, constitutive activation of NADPH oxidase 2 (NOX2) has been shown to favor overproduction of ROS and is present in more than 60% of AML patients [106].

On the other hand, AML LSCs that are characterized by low ROS levels exhibit low cellular oxidative status and aberrantly overexpress the BCL-2 gene. In this respect, BCL-2 inhibition is able to reduce OXPHOS, thus selectively eradicating quiescent LSCs [107]. Thus, in AML, a low level of ROS is related to LSC quiescence, while a high level favors blast proliferation [108,109]. Fig. 2 shows the behavior of LSCs against different levels of ROS.

Fig. 2.

Fig. 2

The behavior of LSCs against different levels of ROS.

In addition to promoting leukemogenesis in AML, ROS can promote chemotherapy resistance. An example is arsenic trioxide, an antileukemic drug used for the treatment of AML. Although it is commonly used, many patients have high therapeutic resistance because some signaling pathways, such as JAK2/STAT3, reduce the antileukemic activity of arsenic trioxide, reducing the amount of ROS and protecting the cell from apoptosis. Based on this, a study demonstrated that the use of potent inhibitors of the JAK2/STAT3 pathway is an alternative to increase the sensitivity of AML cells to combined treatment with arsenic trioxide by increasing ROS and DNA damage. In this work, the authors demonstrated that the increase in ROS increased the rate of apoptosis in AML cells treated with the combination of arsenic trioxide and ruxolitinib, thus revealing that the antileukemic effect of arsenic trioxide is mediated by the elevation of ROS and DNA damage [110].

Regarding the tumor-suppressing properties of oxidative stress, elevated oxidative damage and enhanced ROS-dependent death signaling can also successfully stop some stages of tumorigenesis and make tumor cells more susceptible to cell death [65,111]. Increased oxidative stress associated with cancer progression causes metabolic reprogramming of cells, allowing them to tolerate the negative effects of increased ROS [112,113]. As a result, tumor cells become more dependent on the cellular antioxidant system. Furthermore, ROS accumulation can lead to senescence and various forms of cell death, especially ferroptosis, which is caused by iron-dependent lipid peroxidation [[114], [115], [116]].

1.3. Emerging drugs targeting oxidative stress to eliminate AML LSCs

Some drugs have been reported as pro-oxidant agents causing selective cell death against AML LSCs and have been discussed in this section as emerging drug therapies in AML patients. Table 2 summarizes the selected drugs. Among them, venetoclax is the only one in clinical use to treat AML patients, and navitoclax, chidamide, fenretinide, hydroxychloroquine, karonudib, niclosamide and triptolide are under clinical trials (Table 3).

Table 2.

Drugs that target oxidative stress to eliminate AML LSCs.

Drugs Oxidative stress-associated mechanism References
2-methoxyestradiol HIF-1α inhibition [154]
3-Deazaneplanocin A Histone methyltransferase inhibition
Thioredoxin inhibition
[134]
4-benzyl,2-methyl,1,2,4-thiadiazolidine GSK3b inhibition
PKC inhibition
FLT3 inhibition
[141]
4-hydroxy-2-nonenal NF-kB inhibition [153]
ABT-737 BCL-2 inhibition [107]
Avocatin B Fatty acid β-oxidation inhibition [150]
Celastrol NF-kB inhibition [153]
Chidamide Histone deacetylase inhibition
JNK inhibition
NF-kB inhibition
[135]
Diosmetin Estrogen receptor inhibition [155]
Disulfiram/copper JNK activation
NF-kB inhibition
[131]
Fenretinide NF-κB inhibition
Wnt inhibition
[139,140]
GNPIPP12MA FTO inhibition [133]
Hydroxychloroquine Autophagy inhibition [138]
Karonudib (also known as TH1579) MTH1 inhibition [108]
Mefloquine Lysosomal disruption [142]
Micheliolide NF-kB inhibition [149]
Navitoclax (also known as ABT-263) BCL-2 inhibition [107]
Niclosamide NF-kB inhibition [132]
Oridonin Thioredoxin inhibition
Thioredoxin reductase inhibition
[159]
Parthenolide NF-κB inhibition / p53 activation [144]
Peperomin E Thioredoxin reductase inhibition [157]
Piplartine Glutathione inhibition [142]
Triptolide HIF-1α inhibition [152]
Venetoclax (also known as ABT-199) BCL-2 inhibition [117]

Table 3.

Drugs that target oxidative stress to eliminate AML LSCs and are tested in clinical trials as anti-AML therapy.

ID Study Title Conditions Interventions Phase First Posted Current Status
NCT05305859 Venetoclax Combining Chidamide and Azacitidine (VCA) in the Treatment of R/R AML Leukemia, Myeloid, Acute
Relapsed Adult AML
Refractory Leukemia
Drug: venetoclax combining chidamide and azacitidine (VCA) Phase 2 March 31, 2022 Recruiting
NCT05330364 Study of Chidamide Combined With Cladribine in Refractory/Relapsed Acute Myeloid Leukemia Acute Myeloid Leukemia Drug: Chidamide Drug: Cladribine Phase 2 April 15, 2022 Recruiting
NCT05603884 VCA Regimen Followed by D-MAG Regimen on the Treatment of Elderly Patients With Newly Diagnosed Acute Myeloid Leukemia (AML) Leukemia, Myeloid, Acute
AML Stage, Adult
Drug: Venetoclax Combining Chidamide and Azacitidine (VCA) regimen followed by dicitabine combined with liposome mitoxantrone, cytarabine, and G-CSF (D-MAG) regimen Phase 2 November 3, 2022 Not yet recruiting
NCT02886559 Chidamide Plus DCAG for Relapsed/Refractory AML AMLRelapse Drug: Chidamide plus DCAG regimen Phase 1 Phase 2 September 1, 2016 Unknown
NCT03031262 Efficacy and Safety of Chidamide in CBF Leukemia AML Drug: Cytarabine Drug: Chidamide Phase 1 Phase 2 January 25, 2017 Unknown
NCT03985007 CDIAG Regimen in the Treatment of Relapsed or Refractory Acute Myeloid Leukemia Acute Myeloid Leukemia, Relapsed, Adult Drug: CDIAG regimen Phase 2 June 13, 2019 Completed
NCT05029141 New Double Epigenetic Regimen in the Treatment of Relapsed or Refractory Acute Myeloid Leukemia Acute Myeloid Leukemia
Refractory Acute Leukemia
Relapsed Adult AML
Drug: CAHAG regimen
Drug: Placebo regimen
Phase 2 August 31, 2021 Active, not recruiting
NCT05659992 Clinical Study of Venetoclax Combined With CACAG Regimen in the Treatment of Newly Diagnosed Acute Myeloid Leukemia Acute Myeloid Leukemia Drug: azacytidine; cytarabine; aclamycin; Chidamide; venetoclax; granulocyte Phase 1 December 21, 2022 Recruiting
NCT03453255 DCHA as Postremission Therapy for AML With t(8; 21) Chemotherapy Drug: Chemotherapy Phase 1 Phase 2 March 5, 2018 Unknown
NCT05682755 Chidamide Prevents Recurrence of High-risk AML After Allo-HSCT Leukemia, Myeloid, Acute Drug: Chidamide Phase 1 Phase 2 January 12, 2023 Recruiting
NCT05270200 Single Arm Study of Post-transplant Azacitidine and Chidamide for Prevention of Acute Myelogenous Leukemia Relapse Leukemia, Myeloid, Acute Drug: Azacitidine Drug: Chidamide Phase 1 Phase 2 March 8, 2022 Recruiting
NCT01187810 Fenretinide in Children With Recurrent/Resistant ALL, AML, and NHL Acute Myelogenous Leukemia
Acute Lymphoblastic Leukemia
Non-Hodgkin's Lymphoma
Drug: Fenretinide Drug: Cytarabine Drug: Methotrexate Phase 1 August 24, 2010 Terminated (drug supply)
NCT00104923 Fenretinide in Treating Patients With Refractory or Relapsed Hematologic Cancer Chronic Myeloproliferative Disorders
Leukemia
Lymphoma
Multiple Myeloma and Plasma Cell Neoplasm
Drug: fenretinide Phase 1 March 4, 2005 Completed
NCT02631252 Phase I Study of Mitoxantrone and Etoposide Combined With Hydroxychloroquine, for Relapsed Acute Myelogenous Leukemia Leukemia, Acute Myelogenous Drug: Hydroxychloroquine
Drug: Mitoxantrone
Drug: Etoposide
Phase 1 December 16, 2015 Terminated (Inability to accrue)
NCT04077307 A Study in Leukemia Patients With Karonudib (MAATEO) Leukemia Drug: Karonudib Phase 1 September 4, 2019 Unknown
NCT05222984 Navitoclax, Venetoclax, and Decitabine for the Treatment of Relapsed or Refractory Acute Myeloid Leukemia Previously Treated With Venetoclax Recurrent Acute Myeloid Leukemia
Refractory Acute Myeloid Leukemia
Drug: Decitabine Biological: Navitoclax Drug: Venetoclax Phase 1 February 3, 2022 Recruiting
NCT05455294 Combination Navitoclax, Venetoclax and Decitabine for Advanced Myeloid Neoplasms Myeloid Malignancy
Myelodysplastic Syndromes
Myelofibrosis
Acute Myeloid Leukemia
Myeloproliferative Neoplasm
Drug: Navitoclax
Drug: Venetoclax
Drug: Decitabine
Phase 1 July 13, 2022 Recruiting
NCT05188170 Niclosamide in Pediatric Patients With Relapsed and Refractory AML Acute Myeloid Leukemia Drug: Niclosamide Phase 1 January 12, 2022 Recruiting
NCT03347994 Minnelide in Adult Patients With Relapsed or Refractory Acute Myeloid Leukemia Acute Myeloid Leukemia Drug: Minnelide Phase 1 November 20, 2017 Withdrawn (Discordance in contractual language and terms)

*All studies were accessed at www.clinicaltrials.gov on March 28th, 2023 by using the search term "AML" and the drugs that are presented in Table 2 (with exception of venetoclax that is already in clinical use).

1.4. Synthetic small molecules

1.4.1. BCL-2 inhibitors

As mentioned above, the different ROS levels found in AML LSCs interfere with some cellular characteristics as well as drug responsiveness. CD34+ AML cells exhibit low levels of ROS, showing increased expression of genes related to stress, drug resistance and poor prognosis [117]. Furthermore, CD34+ AML cells with low ROS levels exhibited increased sensitivity to BCL2 inhibitors, including venetoclax (also known as ABT-199), navitoclax (also known as ABT-263) and ABT-737. These small molecules reduce OXPHOS, increase ROS and selectively eliminate AML LSCs [107,117,118].

Venetoclax is a United States Food and Drug Administration (US-FDA)-approved highly selective molecule that was designed to specifically target the BH3 domain of BCL2 [119]. Although venetoclax alone has only modest clinical efficacy in AML, it achieves high response rates when combined with azacitidine or low-dose cytarabine [120,121]. Many AML patients, on the other hand, do not respond or develop resistance to venetoclax treatment because their AML LSCs can also use fatty acid oxidation (FAO) to feed OXPHOS [[122], [123], [124]].

More recently, Hoang et al. [125] reported that venetoclax combined with arsenic trioxide enhanced apoptosis in AML LSCs derived from both venetoclax-sensitive and venetoclax-resistant primary AML cells. In particular, Cho et al. [126] reported that arsenic trioxide synergistically promotes venetoclax anti-AML LSC effects by downregulating Mcl-1.8-Chloro-adenosine [122,127] and CDK7 inhibitors [128] have also been reported to enhance the anti-AML LSC activity of venetoclax.

Lachowiez et al. [129] reported a clinical trial evaluating the efficacy and safety of the intensive chemotherapy and venetoclax combination regimen compared with the intensive chemotherapy alone regimen in patients with AML. After a 30-month follow-up, the venetoclax plus intensive chemotherapy cohort had an MRD-negative composite complete response rate of 64 (86%) of 74 patients, compared to 86 (61%) of 140 patients in the intensive chemotherapy cohort. Although the overall survival did not significantly differ between these two cohorts, venetoclax plus intensive chemotherapy improved event-free survival.

1.4.2. Disulfiram/copper

Disulfiram is a U.S. FDA-approved drug to treat alcoholism that acts as an aldehyde dehydrogenase inhibitor. The action of disulfiram combined with copper was studied in three AML cell lines, KG-1, NB4 and U-937. This combination caused disruption of ROS balance, cell cycle arrest and apoptosis in AML cells along with increased expression of the tumor suppressor FOXO and decreased expression of the MYC oncogene [130].

Xu et al. [131] demonstrated the antileukemic activity of disulfiram in combination with copper, where these compounds exhibited the ability to target CD34+CD38 LSC-like cells sorted from AML KG-1a and Kasumi-1 cell lines, as well as primary CD34+ AML samples. Apoptotic cell death induction and suppression of colony formation were observed in disulfiram/copper-treated AML cells but not in their normal counterparts. These effects were associated with oxidative stress by activating the ROS-JNK pathway and inhibiting NF-kB signaling.

1.4.3. Niclosamide

Niclosamide is an orally bioavailable chlorinated salicylanilide approved by the US-FDA as an anthelmintic agent. In a study performed by Jin et al. [132], niclosamide was able to induce cell death in the CD34+CD38 subpopulation from AML primary cells, preserving those from normal bone marrow. Increased ROS and inhibition of NF-kB signaling were also found in niclosamide-treated AML HL-60 cells. Elimination of ROS by the glutathione precursor N-acetylcysteine decreased niclosamide-induced apoptosis, suggesting that niclosamide increased ROS levels to induce apoptosis in AML HL-60 cells. In nu/nu BALB/c mice bearing HL-60 xenografts, niclosamide (40 mg/kg) inhibited tumor growth and NF-kB signaling.

1.4.4. GNPIPP12MA

A study developed a treatment strategy against AML LSCs based on bioprinted GSH nanocomposites loaded with an N6-methyladenosine (m6A) fat mass and obesity-associated (FTO) demethylase inhibitor (GNPIPP12MA) with the aim of achieving synergistic GSH depletion of the FTO/m6A pathway and consequently targeting leukemia development. GNPIPP12MA was able to target AML Kasumi-1 and CD34+CD38 AML cells, leading to cell death by ferroptosis in a ROS-dependent mechanism with low effect in cells from normal bone morrow. It was also effective in the AML C1498 mouse model and increased the effect of the anti-PDL-1 antibody in this in vivo model [133].

1.4.5. 3-Deazaneplanocin A

3-Deazaneplanocin A is a histone methyltransferase inhibitor that induces disruption in polycomb-repressive complex 2 (PRC2), causing apoptosis in cancer cells. Zhou et al. [134] demonstrated that 3-deazaneplanocin induces apoptosis in both AML cell lines and patient-derived CD34+CD38 AML-enriched subpopulations. Inhibition of thioredoxin activity and augmentation of ROS were also found in 3-deazaneplanocin A-treated AML cells.

1.4.6. Chidamide

Chidamide, a benzamide-type selective histone deacetylase (HDAC) inhibitor developed in China, induces cell cycle arrest and apoptosis in cancer cells. It induced apoptosis in AML cell lines and primary AML CD34+ cells. Chidamide caused ROS-mediated apoptotic cell death by modulation of antiapoptotic and proapoptotic proteins in the BCL2 family, activation of caspase-3 and cleavage of PARP, along with downregulation of the JNK and NF-κB pathways through activation of CD40 [135].

1.4.7. Hydroxychloroquine

Hydroxychloroquine is a U.S. FDA-approved drug to treat malaria and acute and chronic inflammatory diseases, such as rheumatoid arthritis and systemic lupus erythematosus. It is also an autophagy inhibitor with the ability to induce ROS-mediated apoptosis in cancer cells [136,137]. In a study with AML LSCs performed by Folkerts et al. [138], MOLM13 and NB4 AML cell lines and primary AML CD34+ cells were more sensitive to hydroxychloroquine than normal bone marrow CD34+ cells. Next, primary AML CD34+ cells were separated into low ROS and high ROS subtractions. Primary AML CD34+ cells with low ROS levels showed higher basal autophagy and were more sensitive to hydroxychloroquine than AML CD34+ cells with high ROS levels.

1.4.8. Karonudib

Karonudib (also known as TH1579) is an oral inhibitor of MTH1, an enzyme preventing oxidized dNTP incorporation in DNA. It was able to eliminate both primary blast AML and primary CD34+CD38 AML cells while sparing normal cells. Karonudib caused mitotic arrest, increased ROS levels and improved oxidative DNA damage in AML HL-60 cells. Karonudib (45 and 90 mg/kg twice daily, three times a week) inhibited AML development and enhanced survival in NSG mice transplanted intravenously with AML HL60Adp luciferase-expressing cells [108].

1.4.9. Fenretinide

Fenretinide is a synthetic retinoid derivative with action in cancer cells. It was able to induce cell death and inhibit the formation of colonies in primary AML CD34+ cells without affecting their normal counterparts. Fenretinide also caused the generation of ROS, increased stress responses and apoptosis-related gene transcripts and reduced NF-κB and Wnt signaling. It also inhibited the in vivo engraftment of AML LSCs but not HSCs [139].

Zhao et al. [140] demonstrated that it induces ROS production and NF-κB inhibition in AML MV4-11 cells. In addition, primary AML CD34+ cells with FMS-like tyrosine kinase 3 (FLT3)-internal tandem duplication (ITD) mutations were more sensitive to fenretinide than those without FLT3-ITD mutations.

1.4.10. 4-Benzyl,2-methyl,1,2,4-thiadiazolidine

4-Benzyl,2-methyl,1,2,4-thiadiazolidine, a member of the thiadiazolidine class, is a non-ATP competitive inhibitor of glycogen synthase kinase 3β (GSK3b). It caused cell death in different subtypes of leukemia, including primary CD34+CD38 AML cells, without affecting HSCs. Induction of oxidative stress and inhibition of the PKC and FLT3 signaling pathways were also found in 4-benzyl,2-methyl,1,2,4-thiadiazolidine-treated primary AML cells [141].

1.4.11. Mefloquine

Mefloquine is a U.S. FDA-approved antimalarial agent. Similar to chloroquine and hydroxychloroquine, it is an autophagy inhibitor. Mefloquine induced cell death in AML cell lines, primary bulk AML cells and primary AML progenitor cells more effectively than in normal hematopoietic progenitor cells. Induction of oxidative stress and lysosomal disruption were found in mefloquine-treated TEX and OCI-AML2 cells. Mefloquine also showed antileukemic activity on mouse leukemic MDAY-D2 cells and human leukemic OCI-AML2 or K562 cells (50 mg/kg daily by oral gavage), as well as on primary human AML cells (100 mg/kg daily by oral gavage) growing in xenografted mice [142].

1.5. Natural product compounds

1.5.1. Parthenolide and analogs

Parthenolide, a natural component extracted from the species Tanacetum parthenium, has been identified to inhibit the ability of LSCs to respond to oxidative stress by making LSCs sensitive to cell death [143]. It induced robust apoptosis in primary human AML cells and blast crisis CML cells while sparing HSCs. When compared to cytarabine chemotherapy, parthenolide is much more specific for leukemia cells. It also induced NF-κB inhibition, proapoptotic activation of the tumor suppressor p53 and increased production of ROS, suggesting that this molecule activity triggers LSC-specific apoptosis [144]. Furthermore, Kim et al. [145] demonstrated that parthenolide activity is greater in cell lines with high myeloperoxidase leukemia than in cell lines with low myeloperoxidase leukemia.

In contrast, parthenolide has poor pharmacological properties, limiting its potential clinical use. Therefore, many parthenolide analogs have been synthesized and evaluated against leukemia cells. Dimethylamino-parthenolide, an orally bioavailable parthenolide analog, caused cell death in human primary LSCs from both myeloid and lymphoid leukemias and was cytotoxic to bulk leukemic cells. It also induces oxidative stress, inhibits NF-kB signaling and activates the p53 pathway [146]. A C1–C10-modified parthenolide analog was also found to inhibit AML LSCs along with improved ROS induction and less toxicity to healthy bone marrow cells [147]. Darwish et al. [148] developed PLGA-antiCD44-parthenolide nanoparticles with enhanced bioavailability and selectivity to target AML cells.

1.5.2. Micheliolide

Micheliolide is a guaianolide sesquiterpene lactone isolated from Michelia compressa and Michelia champaca that is structurally related to parthenolide. It caused apoptosis in AML KG-1a cells, primary bulk AML cells and primary CD34+CD38 AML cells but not in HSCs. These effects were associated with inhibition of NF-κB signaling and generation of intracellular ROS. Micheliolide derivative DMAMCL administered orally at doses of 25, 50 and 100 mg/kg for seven treatments every other day increased survival in mice xenotransplanted with primary human AML cells [149].

1.5.3. Avocatin B

Avocatin B is an avocado-derived lipid mixture containing avocadene and avocadyne (two 17-carbon polyhydroxylated fatty alcohols). It is a fatty acid β-oxidation inhibitor that induces cell death in AML cell lines, primary bulk AML and progenitor AML cells without affecting HSCs. Avocatin B inhibited fatty acid oxidation and decreased NADPH levels in AML TEX cells, resulting in ROS-dependent apoptotic cell death. These effects were related to the CPT1 enzyme, which facilitates the lipid transport of mitochondria [150].

1.5.4. Triptolide

Triptolide, a diterpenoid triepoxide from the medicinal plant Tripterygium wilfordii Hook F, has attracted extensive exploration due to its multiple biological activities [151]. Triptolide demonstrated the ability to induce apoptosis in CD34+CD38 LSC-like cells sorted from AML KG-1a cells. Moreover, triptolide enhanced the proapoptotic effect of idarubicin, increased the levels of ROS and decreased the colony-forming ability of CD34+CD38 LSC-like cells from AML KG-1a cells. A reduction in the expression of Nrf2 and HIF-1α was also observed in cells treated with the combination of triptolide and idarubicin [152].

1.5.5. Celastrol

Celastrol (also known as tripterine) is a pentacyclic nortriterpene quinone isolated from the root of Tripterygium wilfordii and Tripterygium regelii. Celastrol was selected by in silico study due its similar properties to parthenolide. It was able to eliminate bulk primary AML as well primary CD34+CD38 AML cells with less effect on HSCs. Celastrol also impaired engraftment of primary AML cells in immunocompromised mice. Induction of apoptosis associated with oxidative stress, Nrf2 activation and NF-kB signaling inhibition were also observed in celastrol-treated AML cells [153].

1.5.6. 4-hydroxy-2-nonenal

4-Hydroxy-2-nonenal is an α,β-unsaturated hydroxyalkenal produced in cells during oxidative stress by lipid peroxidation. Similar to celastrol, 4-hydroxy-2-nonenal was selected as an anti-AML compound for the in silico study. It also exhibited toxicity to primary AML CD34+CD38cells and impaired engraftment of primary AML cells in immunocompromised mice. 4-Hydroxy-2-nonenal also caused oxidative stress-mediated apoptosis, Nrf2 activation and inhibition of NF-kB signaling in primary AML cells [153].

1.5.7. 2-methoxyestradiol

2-Methoxyestradiol is a 17-estradiol endogenous metabolite that interacts with estrogen receptors and microtubules. Zhe et al. [154] reported that 2-methoxyestradiol is able to induce apoptosis in HL-60 and Kasumi-1 AML cell lines along with inhibition of HIF-1α and its target genes VEGF, GLUT1 and HO-1. It also reduced the CD34+ cell subpopulation of bone marrow from AML patients, with a lower effect on CD34+ cells from normal bone marrow, in a superior manner to cytarabine. This molecule also increased intracellular ROS, and its effect was prevented using the antioxidant N-acetylcysteine.

1.5.8. Diosmetin

Diosmetin is a flavonoid molecule from citrus fruits that has a variety of pharmacological properties. Diosmetin presented in vitro anti-AML activity against a panel of AML cell lines (KG-1a, U937, OCI-AML2 and TEX) and impaired engraftment of TEX cells in immunocompromised mice. Mechanistically, diosmetin is an estrogen receptor β (ERβ) agonist. In particular, primary AML cells with high levels of ERβ were more sensitive than primary AML cells with low ERβ. Interestingly, knockdown of ERβ induced resistance to diosmetin cytotoxicity, while ERβ overexpression improved sensitivity to diosmetin. This effect was associated with the activation of ROS signaling. Moreover, diosmetin (50 mg/kg every other day) administered for 6 weeks reduced engraftment of primary AML cells but not HSCs in immunocompromised mice [155].

1.5.9. Peperomin E

Peperomin E, a secolignan isolated from the Chinese medicinal plant Peperomia dindygulensis (Piperaceae), has been reported to selectively and effectively target and initiate oxidative stress-induced apoptosis in KG-1a CD34+CD38 cells [156].

Peperomin E and its orally bioavailable analog 6-methyl(hydroxyethyl)amino-2,6-dihydropeperomin E also eliminate KG-1a CD34+ cells and primary CD34+ AML cells. 6-Methyl(hydroxyethyl)amino-2,6-dihydropeperomin E (25 and 50 mg/kg/day for 5 weeks) reduced the growth of KG-1a CD34+ cells in immunocompromised mice. It also induced oxidative stress-mediated apoptosis in KG-1a CD34+ cells through inhibition of thioredoxin reductase [157].

1.5.10. Oridonin

Oridonin, a diterpene extracted from the species Rabdosia rubescens, caused apoptotic cell death in t(8; 21) leukemia cells, induced cleavage of the AML1-ETO oncoprotein resulting from t(8; 21) and reduced tumor growth in immunocompromised mice inoculated with t(8; 21)-harboring Kasumi-1 cells [158]. Oridonin also selectively induced ROS-mediated apoptosis by directly binding to glutathione and inhibiting thioredoxin/thioredoxin reductase in t(8; 21) AML cells. Inhibition of c-Kit + AML LSCs in bone marrow from AML1-ETO9a leukemia mice was also observed after treatment with oridonin in vitro and in vivo [159].

1.5.11. Piplartine

Piplartine, also known as piperlongumine, is a natural molecule from the genus Piper with multiple activities [[160], [161], [162], [163], [164], [165], [166]]. Investigating a targeted therapy based on the eradication of CD34+ AML cells that have acquired aberrant glutathione metabolism, Pei et al. [143] used agents known to act directly on the aberrant glutathione pathway, such as piplartine and/or piplartine, in combination with cytarabine and idarubicin in AML cells. Their work demonstrated that these compounds were able to induce glutathione depletion and lead to a reduction in the primary AML CD34+CD38subpopulation.

2. Conclusion

As we discuss in this review, oxidative stress has both tumor-promoting and tumor-suppressing functions. Under low conditions, ROS participate in the self-renewal of HSCs, while an increase in the level of ROS is necessary for the proliferation and differentiation of HSCs, but excessive levels of ROS induce oxidative damage to cellular DNA and proteins, resulting in cell death or favoring the development of AML. In AML, a low level of ROS is related to the quiescence of LSCs, while a high level of ROS is found in AML blasts and participates in cell proliferation. In both cases, the increase in ROS in AML bulk and/or AML LSCs can induce apoptotic death and suppress AML cell proliferation. Currently, there are some potential drugs able to eliminate AML LSCs via ROS increase and, consequently, reduce the probability of disease recurrence. Among them, navitoclax, chidamide, fenretinide, hydroxychloroquine, karonudib, niclosamide, and triptolide are under clinical trials, and venetoclax is a drug approved by the US-FDA to treat patients with AML. Future research targeting oxidative stress as an efficient approach to eradicate AML LSCs should be undertaken.

Authors' contributions

RGAC, SLRS and IRSBD drafted the content of the cellular redox homeostasis subtopic. RGAC, SLRS, IRSBD and ACBCR drafted the content of the pleiotropic effect of oxidative stress in the AML subtopic. RGAC, SLRS and MSO drafted the content of emerging drugs targeting oxidative stress to eliminate AML LSCs subtopic. RBD and DPB planned this review and reviewed the whole text. All authors read and approved the final manuscript.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

RGAC, SLRS, IRSBD MSO and RBD receive personal scholarship from Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES, Brazil), ACBCR receives personal scholarship from Fundação de Amparo à Pesquisa do Estado da Bahia (FAPESB, Brazil) and DPB receives personal scholarship from Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq, Brazil).

Abbreviations:

AML

acute myeloid leukemia

CAT

catalase

CDKs

cyclin-dependent kinases

ERβ

estrogen receptor β

ETC

electron transport chain

FAB

French-American-British

FAO

fatty acid oxidation

FLT3

FMS-like tyrosine kinase 3

FTO

fat mass and obesity associated

GPx

glutathione peroxidase

GRx

glutathione reductase

GSH

reduced glutathione

GSK3b

glycogen synthase kinase 3β

GSSG

oxidized glutathione

GST

glutathione-S-transferase

HDAC

histone deacetylase

HSCs

hematopoietic stem cells

ITD

internal tandem duplication

LSCs

leukemia stem cells

m6A

N6-methyladenosine

MRD

minimal residual disease

NADPH

nicotinamide adenine dinucleotide phosphate

NOX2

NADPH oxidase 2

OXPHOS

oxidative phosphorylation system

PRC2

polycomb-repressive complex 2

PRXs

peroxiredoxins

RNS

reactive nitrogen species

ROS

reactive oxygen species

RSS

reactive sulfur species

SODs

superoxide dismutases

SORs

superoxide reductases

TrxR

thioredoxin reductase

TXNIP

thioredoxin-interacting protein

US-FDA

United States Food and Drug Administration

WHO

World Health Organization

Data availability

No data was used for the research described in the article.

References

  • 1.Papaemmanuil E., Gerstung M., Bullinger L., Gaidzik V.I., Paschka P., Roberts N.D., Potter N.E., Heuser M., Thol F., Bolli N., Gundem G., Van Loo P., Martincorena I., Ganly P., Mudie L., McLaren S., O'Meara S., Raine K., Jones D.R., Teague J.W., Butler A.P., Greaves M.F., Ganser A., Döhner K., Schlenk R.F., Döhner H., Campbell P.J. Genomic classification and prognosis in acute myeloid leukemia. N. Engl. J. Med. 2016;374:2209–2221. doi: 10.1056/NEJMoa1516192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Short N.J., Rytting M.E., Cortes J.E. Acute myeloid leukaemia. Lancet. 2018;392:593–606. doi: 10.1016/s0140-6736(18)31041-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Rodrigues A.C.B.D.C., Costa R.G.A., Silva S.L.R., Dias I.R.S.B., Dias R.B., Bezerra D.P. Cell signaling pathways as molecular targets to eliminate AML stem cells. Crit. Rev. Oncol. Hematol. 2021;160 doi: 10.1016/j.critrevonc.2021.103277. [DOI] [PubMed] [Google Scholar]
  • 4.Granroth G., Khera N., Arana Yi C. Progress and challenges in survivorship after acute myeloid leukemia in adults. Curr Hematol Malig Rep. 2022;17:243–253. doi: 10.1007/s11899-022-00680-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Döhner H., Wei A.H., Appelbaum F.R., Craddock C., DiNardo C.D., Dombret H., Ebert B.L., Fenaux P., Godley L.A., Hasserjian R.P., Larson R.A., Levine R.L., Miyazaki Y., Niederwieser D., Ossenkoppele G., Röllig C., Sierra J., Stein E.M., Tallman M.S., Tien H.F., Wang J., Wierzbowska A., Löwenberg B. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood. 2022;140:1345–1377. doi: 10.1182/blood.2022016867. [DOI] [PubMed] [Google Scholar]
  • 6.American Cancer Society. Cancer Statistics Center. Accessed on March 28, 2023.
  • 7.Bennett J.M., Catovsky D., Daniel M.T., Flandrin G., Galton D.A., Gralnick H.R., Sultan C. Proposals for the classification of the acute leukaemias. French-American-British (FAB) co-operative group. Br. J. Haematol. 1976;33:451–458. doi: 10.1111/j.1365-2141.1976.tb03563.x. [DOI] [PubMed] [Google Scholar]
  • 8.Bennett J.M., Catovsky D., Daniel M.T., Flandrin G., Galton D.A., Gralnick H.R., Sultan C. Proposed revised criteria for the classification of acute myeloid leukemia. A report of the French-American-British Cooperative Group. Ann. Intern. Med. 1985;103:620–625. doi: 10.7326/0003-4819-103-4-620. [DOI] [PubMed] [Google Scholar]
  • 9.Bennett J.M., Catovsky D., Daniel M.T., Flandrin G., Galton D.A., Gralnick H.R., Sultan C. Criteria for the diagnosis of acute leukemia of megakaryocyte lineage (M7). A report of the French-American-British Cooperative Group. Ann. Intern. Med. 1985;103:460–462. doi: 10.7326/0003-4819-103-3-460. [DOI] [PubMed] [Google Scholar]
  • 10.Bennett J.M., Catovsky D., Daniel M.T., Flandrin G., Galton D.A., Gralnick H.R., Sultan C. Proposal for the recognition of minimally differentiated acute myeloid leukaemia (AML-MO) Br. J. Haematol. 1991;78:325–329. doi: 10.1111/j.1365-2141.1991.tb04444.x. [DOI] [PubMed] [Google Scholar]
  • 11.Khoury J.D., Solary E., Abla O., Akkari Y., Alaggio R., Apperley J.F., Bejar R., Berti E., Busque L., Chan J.K.C., Chen W., Chen X., Chng W.J., Choi J.K., Colmenero I., Coupland S.E., Cross N.C.P., De Jong D., Elghetany M.T., Takahashi E., Emile J.F., Ferry J., Fogelstrand L., Fontenay M., Germing U., Gujral S., Haferlach T., Harrison C., Hodge J.C., Hu S., Jansen J.H., Kanagal-Shamanna R., Kantarjian H.M., Kratz C.P., Li X.Q., Lim M.S., Loeb K., Loghavi S., Marcogliese A., Meshinchi S., Michaels P., Naresh K.N., Natkunam Y., Nejati R., Ott G., Padron E., Patel K.P., Patkar N., Picarsic J., Platzbecker U., Roberts I., Schuh A., Sewell W., Siebert R., Tembhare P., Tyner J., Verstovsek S., Wang W., Wood B., Xiao W., Yeung C., Hochhaus A. The 5th edition of the World health organization classification of haematolymphoid tumours: myeloid and histiocytic/dendritic neoplasms. Leukemia. 2022;36:1703–1719. doi: 10.1038/s41375-022-01613-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bower H., Andersson T.M., Björkholm M., Dickman P.W., Lambert P.C., Derolf Å.R. Continued improvement in survival of acute myeloid leukemia patients: an application of the loss in expectation of life. Blood Cancer J. 2016 Feb 5;6(2) doi: 10.1038/bcj.2016.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bosshard R., O'Reilly K., Ralston S., Chadda S., Cork D. Systematic reviews of economic burden and health-related quality of life in patients with acute myeloid leukemia. Cancer Treat Rev. 2018;69:224–232. doi: 10.1016/j.ctrv.2018.07.0053. [DOI] [PubMed] [Google Scholar]
  • 14.Shallis R.M., Wang R., Davidoff A., Ma X., Zeidan A.M. Epidemiology of acute myeloid leukemia: recent progress and enduring challenges. Blood Rev. 2019 Jul;36:70–87. doi: 10.1016/j.blre.2019.04.005. [DOI] [PubMed] [Google Scholar]
  • 15.Bazinet A., Assouline S. A review of FDA-approved acute myeloid leukemia therapies beyond '7 + 3. Expet Rev. Hematol. 2021;14(2):185–197. doi: 10.1080/17474086.2021.1875814. [DOI] [PubMed] [Google Scholar]
  • 16.Ding L., Ley T., Larson D. Clonal evolution in relapse myeloid leukemia revealed by whole genome sequencing. Nature. 2012;481:506–510. doi: 10.1038/nature10738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Short N.J., Zhou S., Fu C., Berry D.A., Walter R.B., Freeman S.D., Hourigan C.S., Huang X., Nogueras Gonzalez G., Hwang H., Qi X., Kantarjian H., Ravandi F. Association of measurable residual disease with survival outcomes in patients with acute myeloid leukemia: a systematic review and meta-analysis. JAMA Oncol. 2020 Dec 1;6(12):1890–1899. doi: 10.1001/jamaoncol.2020.4600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ngai L.L., Kelder A., Janssen J.J.W.M., Ossenkoppele G.J., Cloos J. MRD tailored therapy in AML: what we have learned so far. Front. Oncol. 2021 Jan 15;10 doi: 10.3389/fonc.2020.603636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Heuser M., Freeman S.D., Ossenkoppele G.J., Buccisano F., Hourigan C.S., Ngai L.L., Tettero J.M., Bachas C., Baer C., Béné M.C., Bücklein V., Czyz A., Denys B., Dillon R., Feuring-Buske M., Guzman M.L., Haferlach T., Han L., Herzig J.K., Jorgensen J.L., Kern W., Konopleva M.Y., Lacombe F., Libura M., Majchrzak A., Maurillo L., Ofran Y., Philippe J., Plesa A., Preudhomme C., Ravandi F., Roumier C., Subklewe M., Thol F., van de Loosdrecht A.A., van der Reijden B.A., Venditti A., Wierzbowska A., Valk P.J.M., Wood B.L., Walter R.B., Thiede C., Döhner K., Roboz G.J., Cloos J. 2021 Update on MRD in acute myeloid leukemia: a consensus document from the European LeukemiaNet MRD Working Party. Blood. 2021 Dec 30;138(26):2753–2767. doi: 10.1182/blood.2021013626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lapidot T., Sirard C., Vormoor J., Murdoch B., Hoang T., Caceres-Cortes J., Minden M., Paterson B., Caligiuri M.A., Dick J.E. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994 Feb 17;367(6464):645–648. doi: 10.1038/367645a0. [DOI] [PubMed] [Google Scholar]
  • 21.Ho T.C., LaMere M., Stevens B.M., Ashton J.M., Myers J.R., O'Dwyer K.M., Liesveld J.L., Mendler J.H., Guzman M., Morrissette J.D., Zhao J., Wang E.S., Wetzler M., Jordan C.T., Becker M.W. Evolution of acute myelogenous leukemia stem cell properties after treatment and progression. Blood. 2016 Sep 29;128(13):1671–1678. doi: 10.1182/blood-2016-02-695312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Laverdière I., Boileau M., Neumann A.L., Frison H., Mitchell A., Ng S., Wang J., Minden M.D., Eppert K. Leukemic stem cell signatures identify novel therapeutics targeting acute myeloid leukemia. Blood Cancer J. 2018;8(6):52. doi: 10.1038/s41408-018-0087-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Moshaver B., Wouters R.F., Kelder A., Ossenkoppele G.J., Westra G., Kwidama Z., Rutten A.R., Kaspers G., Zweegman S., Cloos J., Schuurhuis G.J. Relationship between CD34/CD38 and side population (SP) defined leukemia stem cell compartments in acute myeloid leukemia. Leuk. Res. 2019;81:27–34. doi: 10.1016/j.leukres.2019.04.004. [DOI] [PubMed] [Google Scholar]
  • 24.Niu J., Peng D., Liu L. Drug resistance mechanisms of acute myeloid leukemia stem cells. Front. Oncol. 2022 Jul 5;12 doi: 10.3389/fonc.2022.896426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hope K.J., Jin L.Q., Dick J.E. Acute myeloid leukemia originates from a hierarchy of leukemic stem cell classes that differ in self-renewal capacity. Nat. Immunol. 2004;5(7):738–743. doi: 10.1038/ni1080. [DOI] [PubMed] [Google Scholar]
  • 26.Saito Y., Uchida N., Tanaka S., Suzuki N., Tomizawa-Murasawa M., Sone A., Najima Y., Takagi S., Aoki Y., Wake A., Taniguchi S., Shultz L.D., Ishikawa F. Induction of cell cycle entry eliminates human leukemia stem cells in a mouse model of AML. Nat. Biotechnol. 2010 Mar;28(3):275–280. doi: 10.1038/nbt.1607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Phan T.G., Croucher P.I. The dormant cancer cell life cycle. Nat. Rev. Cancer. 2020;20(7):398–411. doi: 10.1038/s41568-020-0263-0. [DOI] [PubMed] [Google Scholar]
  • 28.Carlesso N., Cardoso A.A. Stem cell regulatory niches and their role in normal and malignant hematopoiesis. Curr. Opin. Hematol. 2010;17:281–286. doi: 10.1097/MOH.0b013e32833a25d8. [DOI] [PubMed] [Google Scholar]
  • 29.Tabe Y., Konopleva M. Advances in understanding the leukaemia microenvironment. Br. J. Haematol. 2014;164:767–778. doi: 10.1111/bjh.12725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Mendelson A., Frenette P.S. Hematopoietic stem cell niche maintenance during homeostasis and regeneration. Nat. Med. 2014;20:833–846. doi: 10.1038/nm.3647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Pollyea D.A., Jordan C.T. Therapeutic targeting of acute myeloid leukemia stem cells. Blood. 2017;129(12):1627–1635. doi: 10.1182/blood-2016-10-696039. [DOI] [PubMed] [Google Scholar]
  • 32.Lee G.Y., Jeong S.Y., Lee H.R., Oh I.H. Age-related differences in the bone marrow stem cell niche generate specialized microenvironments for the distinct regulation of normal hematopoietic and leukemia stem cells. Sci. Rep. 2019;9(1):1007. doi: 10.1038/s41598-018-36999-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Jordan C.T., Upchurch D., Szilvassy S.J., Guzman M.L., Howard D.S., Pettigrew A.L., Meyerrose T., Rossi R., Grimes B., Rizzieri D.A., Luger S.M., Phillips G.L. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia. 2000 Oct;14(10):1777–1784. doi: 10.1038/sj.leu.2401903. [DOI] [PubMed] [Google Scholar]
  • 34.Hosen N., Park C.Y., Tatsumi N., Oji Y., Sugiyama H., Gramatzki M., Krensky A.M., Weissman I.L. CD96 is a leukemic stem cell-specific marker in human acute myeloid leukemia. Proc. Natl. Acad. Sci. U. S. A. 2007 Jun 26;104(26):11008–11013. doi: 10.1073/pnas.0704271104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.van Rhenen A., van Dongen G.A., Kelder A., Rombouts E.J., Feller N., Moshaver B., Stigter-van Walsum M., Zweegman S., Ossenkoppele G.J., Jan Schuurhuis G. The novel AML stem cell associated antigen CLL-1 aids in discrimination between normal and leukemic stem cells. Blood. 2007 Oct 1;110(7):2659–2666. doi: 10.1182/blood-2007-03-083048. [DOI] [PubMed] [Google Scholar]
  • 36.Kikushige Y., Shima T., Takayanagi S., Urata S., Miyamoto T., Iwasaki H., Takenaka K., Teshima T., Tanaka T., Inagaki Y., Akashi K. TIM-3 is a promising target to selectively kill acute myeloid leukemia stem cells. Cell Stem Cell. 2010 Dec 3;7(6):708–717. doi: 10.1016/j.stem.2010.11.014. [DOI] [PubMed] [Google Scholar]
  • 37.Iwasaki M., Liedtke M., Gentles A.J., Cleary M.L. CD93 marks a non-quiescent human leukemia stem cell population and is required for development of MLL-rearranged acute myeloid leukemia. Cell Stem Cell. 2015;17(4):412–421. doi: 10.1016/j.stem.2015.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chung S.S., Eng W.S., Hu W., Khalaj M., Garrett-Bakelman F.E., Tavakkoli M., Levine R.L., Carroll M., Klimek V.M., Melnick A.M., Park C.Y. CD99 is a therapeutic target on disease stem cells in myeloid malignancies. Sci. Transl. Med. 2017 Jan 25;9(374) doi: 10.1126/scitranslmed.aaj2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Pilo F., Angelucci E. A storm in the niche: iron, oxidative stress and haemopoiesis. Blood Rev. 2018;32(1):29–35. doi: 10.1016/j.blre.2017.08.005. [DOI] [PubMed] [Google Scholar]
  • 40.Sun Y., Liu X., Wang L., Xu L., Liu K., Xu L., Shi F., Zhang Y., Gu N., Xiong F. High-performance SOD mimetic enzyme Au@Ce for arresting cell cycle and proliferation of acute myeloid leukemia. Bioact. Mater. 2021 Aug 18;10:117–130. doi: 10.1016/j.bioactmat.2021.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ray P.D., Huang B.-W., Tsuji Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling Cell. Signal. 2012;24:981–990. doi: 10.1016/j.cellsig.2012.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Mattes K., Vellenga E., Schepers H. Differential redox-regulation and mitochondrial dynamics in normal and leukemic hematopoietic stem cells: a potential window for leukemia therapy. Crit. Rev. Oncol. Hematol. 2019 Dec;144 doi: 10.1016/j.critrevonc.2019.102814. [DOI] [PubMed] [Google Scholar]
  • 43.Pei S., Minhajuddin M., Adane B., Khan N., Stevens B.M., Mack S.C., Lai S., Rich J.N., Inguva A., Shannon K.M., Kim H., Tan A.C., Myers J.R., Ashton J.M., Neff T., Pollyea D.A., Smith C.A., Jordan C.T. AMPK/FIS1-Mediated mitophagy is required for self-renewal of human AML stem cells. Cell Stem Cell. 2018 Jul 5;23(1):86–100.e6. doi: 10.1016/j.stem.2018.05.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Finkel T., Holbrook N.J. Oxidants, oxidative stress and the biology of ageing. Nature. 2000 Nov 9;408(6809):239–247. doi: 10.1038/35041687. [DOI] [PubMed] [Google Scholar]
  • 45.Halliwell B., Gutteridge J.M.C. Oxford University Press; 2015. Free Radicals in Biology and Medicine. [DOI] [Google Scholar]
  • 46.Radi R. Oxygen radicals, nitric oxide, and peroxynitrite: redox pathways in molecular medicine. Proc. Natl. Acad. Sci. U. S. A. 2018 Jun 5;115(23):5839–5848. doi: 10.1073/pnas.1804932115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Martemucci G., Costagliola C., Mariano M., D’andrea L., Napolitano P., D'Alessandro A.G. Free radical properties, source and targets, antioxidant consumption and health. Oxygen. 2022;2(2):48–78. doi: 10.3390/oxygen2020006. [DOI] [Google Scholar]
  • 48.Sies H. Oxidative stress: a concept in redox biology and medicine. Redox Biol. 2015;4:180–183. doi: 10.1016/j.redox.2015.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Santolini J., Wootton S.A., Jackson A.A., Feelisch M. The redox architecture of physiological function. Curr Opin Physiol. 2019;9:34–47. doi: 10.1016/j.cophys.2019.04. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Olson K.R. Are reactive sulfur species the new reactive oxygen species? Antioxidants Redox Signal. 2020 doi: 10.1089/ars.2020.8132. [DOI] [PubMed] [Google Scholar]
  • 51.Hayes J.D., Dinkova-Kostova A.T., Tew K.D. Oxidative stress in cancer. Cancer Cell. 2020 Aug 10;38(2):167–197. doi: 10.1016/j.ccell.2020.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Perillo B., Di Donato M., Pezone A., Di Zazzo E., Giovannelli P., Galasso G., Castoria G., Migliaccio A. ROS in cancer therapy: the bright side of the moon. Exp. Mol. Med. 2020 Feb;52(2):192–203. doi: 10.1038/s12276-020-0384-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Le Gal K., Schmidt E.E., Sayin V.I. Cellular redox homeostasis. Antioxidants. 2021;10(9):1377. doi: 10.3390/antiox10091377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Bleier L., Wittig I., Heide H., Steger M., Brandt U., Dröse S. Generator-specific targets of mitochondrial reactive oxygen species. Free Radic. Biol. Med. 2015 Jan;78:1–10. doi: 10.1016/j.freeradbiomed.2014.10.511. [DOI] [PubMed] [Google Scholar]
  • 55.Brand M.D. Mitochondrial generation of superoxide and hydrogen peroxide as the source of mitochondrial redox signaling. Free Radic. Biol. Med. 2016;100:14–31. doi: 10.1016/j.freeradbiomed.2016.04.001. [DOI] [PubMed] [Google Scholar]
  • 56.Liu X., Zhang Y., Zhuang L., Olszewski K., Gan B. NADPH debt drives redox bankruptcy: SLC7A11/xCT-mediated cystine uptake as a double-edged sword in cellular redox regulation. Genes Dis. 2020 Nov 25;8(6):731–745. doi: 10.1016/j.gendis.2020.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Sies H., Jones D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020 Jul;21(7):363–383. doi: 10.1038/s41580-020-0230-3. [DOI] [PubMed] [Google Scholar]
  • 58.Sies H., Berndt C., Jones D.P. Oxidative stress. Annu. Rev. Biochem. 2017 Jun 20;86:715–748. doi: 10.1146/annurev-biochem-061516-045037. [DOI] [PubMed] [Google Scholar]
  • 59.Hawkins C.L., Davies M.J. Detection, identification, and quantification of oxidative protein modifications. J. Biol. Chem. 2019;294:19683–19708. doi: 10.1074/jbc.REV119.006217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Zhang H., Gong W., Wu S., Perrett S. Hsp70 in redox homeostasis. Cells. 2022 Feb 28;11(5):829. doi: 10.3390/cells11050829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Fuloria S., Subramaniyan V., Karupiah S., Kumari U., Sathasivam K., Meenakshi D.U., Wu Y.S., Sekar M., Chitranshi N., Malviya R., Sudhakar K., Bajaj S., Fuloria N.K. Comprehensive review of methodology to detect reactive oxygen species (ROS) in mammalian species and establish its relationship with antioxidants and cancer. Antioxidants. 2021 Jan 18;10(1):128. doi: 10.3390/antiox10010128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Aquilano K., Baldelli S., Ciriolo M.R. Glutathione: new roles in redox signaling for an old antioxidant. Front. Pharmacol. 2014 Aug 26;5:196. doi: 10.3389/fphar.2014.00196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Tretter V., Hochreiter B., Zach M.L., Krenn K., Klein K.U. Understanding cellular redox homeostasis: a challenge for precision medicine. Int. J. Mol. Sci. 2021 Dec 22;23(1):106. doi: 10.3390/ijms23010106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hasan A.A., Kalinina E., Tatarskiy V., Shtil A. The thioredoxin system of mammalian cells and its modulators. Biomedicines. 2022 Jul 21;10(7):1757. doi: 10.3390/biomedicines10071757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Cheung E.C., Vousden K.H. The role of ROS in tumour development and progression. Nat. Rev. Cancer. 2022 May;22(5):280–297. doi: 10.1038/s41568-021-00435-0. [DOI] [PubMed] [Google Scholar]
  • 66.Galasso M., Dalla Pozza E., Chignola R., Gambino S., Cavallini C., Quaglia F.M., Lovato O., Dando I., Malpeli G., Krampera M., Donadelli M., Romanelli M.G., Scupoli M.T. The rs1001179 SNP and CpG methylation regulate catalase expression in chronic lymphocytic leukemia. Cell. Mol. Life Sci. 2022 Sep 16;79(10):521. doi: 10.1007/s00018-022-04540-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Martins M.C., Romão C.V., Folgosa F., Borges P.T., Frazão C., Teixeira M. How superoxide reductases and flavodiiron proteins combat oxidative stress in anaerobes. Free Radic. Biol. Med. 2019;140:36–60. doi: 10.1016/j.freeradbiomed.2019.01.051. [DOI] [PubMed] [Google Scholar]
  • 68.Griess B., Tom E., Domann F., Teoh-Fitzgerald M. Extracellular superoxide dismutase and its role in cancer. Free Radic. Biol. Med. 2017;112:464–479. doi: 10.1016/j.freeradbiomed.2017.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Maiti G.P., Sinha S., Mahmud H., Boysen J., Mendez M.T., Vesely S.K., Holter-Chakrabarty J., Kay N.E., Ghosh A.K. SIRT3 overexpression and epigenetic silencing of catalase regulate ROS accumulation in CLL cells activating AXL signaling axis. Blood Cancer J. 2021 May 17;11(5):93. doi: 10.1038/s41408-021-00484-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Ghaffari S. Oxidative stress in the regulation of normal and neoplastic hematopoiesis. Antioxidants Redox Signal. 2008;10(11):1923–1940. doi: 10.1089/ars.2008.2142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bănescu C., Iancu M., Trifa A.P., Cândea M., Benedek Lazar E., Moldovan V.G., Duicu C., Tripon F., Crauciuc A., Dobreanu M. From six gene polymorphisms of the antioxidant system, only GPX Pro198Leu and GSTP1 Ile105Val modulate the risk of acute myeloid leukemia. Oxid. Med. Cell. Longev. 2016;2016 doi: 10.1155/2016/2536705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Sheng Y., Abreu I.A., Cabelli D.E., Maroney M.J., Miller A.F., Teixeira M., Valentine J.S. Superoxide dismutases and superoxide reductases. Chem. Rev. 2014 Apr 9;114(7):3854–3918. doi: 10.1021/cr4005296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Pinto A.F., Romão C.V., Pinto L.C., Huber H., Saraiva L.M., Todorovic S., Cabelli D., Teixeira M. Superoxide reduction by a superoxide reductase lacking the highly conserved lysine residue. J. Biol. Inorg. Chem. 2015 Jan;20(1):155–164. doi: 10.1007/s00775-014-1222-6. [DOI] [PubMed] [Google Scholar]
  • 74.Kirkman H.N., Gaetani G.F. Mammalian catalase: a venerable enzyme with new mysteries. Trends Biochem. Sci. 2007 Jan;32(1):44–50. doi: 10.1016/j.tibs.2006.11.003. [DOI] [PubMed] [Google Scholar]
  • 75.Eras N., Türkoz G., Tombak A., Tiftik N., Yalin S., Berkoz M., Erden S., Akbas E. An investigation of the relation between catalase C262T gene polymorphism and catalase enzyme activity in leukemia patients. Arch. Med. Sci. 2019 Nov 12;17(4):928–933. doi: 10.5114/aoms.2019.89692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Galasso M., Gambino S., Romanelli M.G., Donadelli M., Scupoli M.T. Browsing the oldest antioxidant enzyme: catalase and its multiple regulation in cancer. Free Radic. Biol. Med. 2021;172:264–272. doi: 10.1016/j.freeradbiomed.2021.06.010. [DOI] [PubMed] [Google Scholar]
  • 77.Asantewaa G., Harris I.S. Glutathione and its precursors in cancer. Curr. Opin. Biotechnol. 2021 Apr;68:292–299. doi: 10.1016/j.copbio.2021.03.001. [DOI] [PubMed] [Google Scholar]
  • 78.Wu G., Fang Y.Z., Yang S., Lupton J.R., Turner N.D. Glutathione metabolism and its implications for health. J. Nutr. 2004 Mar;134(3):489–492. doi: 10.1093/jn/134.3.489. [DOI] [PubMed] [Google Scholar]
  • 79.Deponte M. Glutathione catalysis and the reaction mechanisms of glutathione-dependent enzymes. Biochim. Biophys. Acta. 2013 May;1830(5):3217–3266. doi: 10.1016/j.bbagen.2012.09.018. [DOI] [PubMed] [Google Scholar]
  • 80.Franchina D.G., Kurniawan H., Grusdat M., Binsfeld C., Guerra L., Bonetti L., Soriano-Baguet L., Ewen A., Kobayashi T., Farinelle S., Minafra A.R., Vandamme N., Carpentier A., Borgmann F.K., Jäger C., Chen Y., Kleinewietfeld M., Vasiliou V., Mittelbronn M., Hiller K., Lang P.A., Brenner D. Glutathione-dependent redox balance characterizes the distinct metabolic properties of follicular and marginal zone B cells. Nat. Commun. 2022 Apr 4;13(1):1789. doi: 10.1038/s41467-022-29426-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Handy D.E., Loscalzo J. The role of glutathione peroxidase-1 in health and disease. Free Radic. Biol. Med. 2022 Aug 1;188:146–161. doi: 10.1016/j.freeradbiomed.2022.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Mustacich D., Powis G. Thioredoxin reductase. Biochem. J. 2000 Feb 15;346(Pt 1):1–8. Pt 1. [PMC free article] [PubMed] [Google Scholar]
  • 83.Arnér E.S. Focus on mammalian thioredoxin reductases--important selenoproteins with versatile functions. Biochim. Biophys. Acta. 2009 Jun;1790(6):495–526. doi: 10.1016/j.bbagen.2009.01.014. [DOI] [PubMed] [Google Scholar]
  • 84.Muri J., Kopf M. The thioredoxin system: balancing redox responses in immune cells and tumors. Eur. J. Immunol. 2022 Oct 25 doi: 10.1002/eji.202249948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chakraborty P., Chatterjee S., Kesarwani P., Thyagarajan K., Iamsawat S., Dalheim A., Nguyen H., Selvam S.P., Nasarre P., Scurti G., Hardiman G., Maulik N., Ball L., Gangaraju V., Rubinstein M.P., Klauber-DeMore N., Hill E.G., Ogretmen B., Yu X.Z., Nishimura M.I., Mehrotra S. Thioredoxin-1 improves the immunometabolic phenotype of antitumor T cells. J. Biol. Chem. 2019 Jun 7;294(23):9198–9212. doi: 10.1074/jbc.RA118.006753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kang S.W., Chae H.Z., Seo M.S., Kim K., Baines I.C., Rhee S.G. Mammalian peroxiredoxin isoforms can reduce hydrogen peroxide generated in response to growth factors and tumor necrosis factor-alpha. J. Biol. Chem. 1998 Mar 13;273(11):6297–6302. doi: 10.1074/jbc.273.11.6297. [DOI] [PubMed] [Google Scholar]
  • 87.Chang T.S., Jeong W., Choi S.Y., Yu S., Kang S.W., Rhee S.G. Regulation of peroxiredoxin I activity by Cdc2-mediated phosphorylation. J. Biol. Chem. 2002 Jul 12;277(28):25370–25376. doi: 10.1074/jbc.M110432200. [DOI] [PubMed] [Google Scholar]
  • 88.Neumann C.A., Krause D.S., Carman C.V., Das S., Dubey D.P., Abraham J.L., Bronson R.T., Fujiwara Y., Orkin S.H., Van Etten R.A. Essential role for the peroxiredoxin Prdx1 in erythrocyte antioxidant defence and tumour suppression. Nature. 2003 Jul 31;424(6948):561–565. doi: 10.1038/nature01819. [DOI] [PubMed] [Google Scholar]
  • 89.Lee T.H., Kim S.U., Yu S.L., Kim S.H., Park D.S., Moon H.B., Dho S.H., Kwon K.S., Kwon H.J., Han Y.H., Jeong S., Kang S.W., Shin H.S., Lee K.K., Rhee S.G., Yu D.Y. Peroxiredoxin II is essential for sustaining life span of erythrocytes in mice. Blood. 2003 Jun 15;101(12):5033–5038. doi: 10.1182/blood-2002-08-2548. [DOI] [PubMed] [Google Scholar]
  • 90.Chang T.S., Cho C.S., Park S., Yu S., Kang S.W., Rhee S.G. Peroxiredoxin III, a mitochondrion-specific peroxidase, regulates apoptotic signaling by mitochondria. J. Biol. Chem. 2004 Oct 1;279(40):41975–41984. doi: 10.1074/jbc.M407707200. [DOI] [PubMed] [Google Scholar]
  • 91.Yu W.M., Liu X., Shen J., Jovanovic O., Pohl E.E., Gerson S.L., Finkel T., Broxmeyer H.E., Qu C.K. Metabolic regulation by the mitochondrial phosphatase PTPMT1 is required for hematopoietic stem cell differentiation. Cell Stem Cell. 2013 Jan 3;12(1):62–74. doi: 10.1016/j.stem.2012.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Takubo K., Nagamatsu G., Kobayashi C.I., Nakamura-Ishizu A., Kobayashi H., Ikeda E., Goda N., Rahimi Y., Johnson R.S., Soga T., Hirao A., Suematsu M., Suda T. Regulation of glycolysis by Pdk functions as a metabolic checkpoint for cell cycle quiescence in hematopoietic stem cells. Cell Stem Cell. 2013 Jan 3;12(1):49–61. doi: 10.1016/j.stem.2012.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Maryanovich M., Zaltsman Y., Ruggiero A., Goldman A., Shachnai L., Zaidman S.L., Porat Z., Golan K., Lapidot T., Gross A. An MTCH2 pathway repressing mitochondria metabolism regulates haematopoietic stem cell fate. Nat. Commun. 2015 Jul 29;6:7901. doi: 10.1038/ncomms8901. [DOI] [PubMed] [Google Scholar]
  • 94.Simsek T., Kocabas F., Zheng J., Deberardinis R.J., Mahmoud A.I., Olson E.N., Schneider J.W., Zhang C.C., Sadek H.A. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell. 2010 Sep 3;7(3):380–390. doi: 10.1016/j.stem.2010.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Vannini N., Girotra M., Naveiras O., Nikitin G., Campos V., Giger S., Roch A., Auwerx J., Lutolf M.P. Specification of haematopoietic stem cell fate via modulation of mitochondrial activity. Nat. Commun. 2016 Oct 12;7 doi: 10.1038/ncomms13125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Sukumar M., Liu J., Mehta G.U., Patel S.J., Roychoudhuri R., Crompton J.G., Klebanoff C.A., Ji Y., Li P., Yu Z., Whitehill G.D., Clever D., Eil R.L., Palmer D.C., Mitra S., Rao M., Keyvanfar K., Schrump D.S., Wang E., Marincola F.M., Gattinoni L., Leonard W.J., Muranski P., Finkel T., Restifo N.P. Mitochondrial membrane potential identifies cells with enhanced stemness for cellular therapy. Cell Metabol. 2016 Jan 12;23(1):63–76. doi: 10.1016/j.cmet.2015.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Suda T., Takubo K., Semenza G.L. Metabolic regulation of hematopoietic stem cells in the hypoxic niche. Cell Stem Cell. 2011 Oct 4;9(4):298–310. doi: 10.1016/j.stem.2011.09.010. [DOI] [PubMed] [Google Scholar]
  • 98.Ishikawa E.T., Gonzalez-Nieto D., Ghiaur G., Dunn S.K., Ficker A.M., Murali B., Madhu M., Gutstein D.E., Fishman G.I., Barrio L.C., Cancelas J.A. Connexin-43 prevents hematopoietic stem cell senescence through transfer of reactive oxygen species to bone marrow stromal cells. Proc. Natl. Acad. Sci. U. S. A. 2012 Jun 5;109(23):9071–9076. doi: 10.1073/pnas.1120358109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ito K., Turcotte R., Cui J., Zimmerman S.E., Pinho S., Mizoguchi T., Arai F., Runnels J.M., Alt C., Teruya-Feldstein J., Mar J.C., Singh R., Suda T., Lin C.P., Frenette P.S., Ito K. Self-renewal of a purified Tie2+ hematopoietic stem cell population relies on mitochondrial clearance. Science. 2016 Dec 2;354(6316):1156–1160. doi: 10.1126/science.aaf5530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Jang Y.Y., Sharkis S.J. A low level of reactive oxygen species selects for primitive hematopoietic stem cells that may reside in the low-oxygenic niche. Blood. 2007 Oct 15;110(8):3056–3063. doi: 10.1182/blood-2007-05-087759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Golan K., Kumari A., Kollet O., Khatib-Massalha E., Subramaniam M.D., Ferreira Z.S., Avemaria F., Rzeszotek S., García-García A., Xie S., Flores-Figueroa E., Gur-Cohen S., Itkin T., Ludin-Tal A., Massalha H., Bernshtein B., Ciechanowicz A.K., Brandis A., Mehlman T., Bhattacharya S., Bertagna M., Cheng H., Petrovich-Kopitman E., Janus T., Kaushansky N., Cheng T., Sagi I., Ratajczak M.Z., Méndez-Ferrer S., Dick J.E., Markus R.P., Lapidot T. Daily onset of light and darkness differentially controls hematopoietic stem cell differentiation and maintenance. Cell Stem Cell. 2018 Oct 4;23(4):572–585.e7. doi: 10.1016/j.stem.2018.08.002. [DOI] [PubMed] [Google Scholar]
  • 102.Prieto-Bermejo R., Romo-González M., Pérez-Fernández A., Ijurko C., Hernández-Hernández Á Reactive oxygen species in haematopoiesis: leukaemic cells take a walk on the wild side. J. Exp. Clin. Cancer Res. 2018;37(1):125. doi: 10.1186/s13046-018-0797-0. Published 2018 Jun 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Naka K., Muraguchi T., Hoshii T., Hirao A. Regulation of reactive oxygen species and genomic stability in hematopoietic stem cells. Antioxidants Redox Signal. 2008;10:1883–1894. doi: 10.1089/ars.2008.2114. [DOI] [PubMed] [Google Scholar]
  • 104.Stelmach P., Trumpp A. Leukemic stem cells and therapy resistance in acute myeloid leukemia. Haematologica. 2023 Feb 1;108(2):353–366. doi: 10.3324/haematol.2022.280800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wei J., Xie Q., Liu X., Wan C., Wu W., Fang K., Yao Y., Cheng P., Deng D., Liu Z. Identification the prognostic value of glutathione peroxidases expression levels in acute myeloid leukemia. Ann. Transl. Med. 2020;8(11):678. doi: 10.21037/atm-20-3296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Hole P.S., Zabkiewicz J., Munje C., Newton Z., Pearn L., White P., Marquez N., Hills R.K., Burnett A.K., Tonks A., Darley R.L. Overproduction of NOX-derived ROS in AML promotes proliferation and is associated with defective oxidative stress signaling. Blood. 2013 Nov 7;122(19):3322–3330. doi: 10.1182/blood-2013-04-491944. [DOI] [PubMed] [Google Scholar]
  • 107.Lagadinou E.D., Sach A., Callahan K., Rossi R.M., Neering S.J., Minhajuddin M., Ashton J.M., Pei S., Grose V., O'Dwyer K.M., Liesveld J.L., Brookes P.S., Becker M.W., Jordan C.T. 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]
  • 108.Sanjiv K., Calderón-Montaño J.M., Pham T.M., Erkers T., Tsuber V., Almlöf I., Höglund A., Heshmati Y., Seashore-Ludlow B., Nagesh Danda A., Gad H., Wiita E., Göktürk C., Rasti A., Friedrich S., Centio A., Estruch M., Våtsveen T.K., Struyf N., Visnes T., Scobie M., Koolmeister T., Henriksson M., Wallner O., Sandvall T., Lehmann S., Theilgaard-Mönch K., Garnett M.J., Östling P., Walfridsson J., Helleday T., Warpman Berglund U. MTH1 inhibitor TH1579 induces oxidative DNA damage and mitotic arrest in acute myeloid leukemia. Cancer Res. 2021 Nov 15;81(22):5733–5744. doi: 10.1158/0008-5472.CAN-21-0061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Dakik H., El Dor M., Leclerc J., Kouzi F., Nehme A., Deynoux M., Debeissat C., Khamis G., Ducrocq E., Ibrik A., Stasia M.J., Raad H., Rezvani H.R., Gouilleux F., Zibara K., Herault O., Mazurier F. Characterization of NADPH oxidase expression and activity in acute myeloid leukemia cell lines: a correlation with the differentiation status. Antioxidants. 2021;10:498. doi: 10.3390/antiox10030498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Mesbahi Y., Zekri A., Ghaffari S.H., Tabatabaie P.S., Ahmadian S., Ghavamzadeh A. Blockade of JAK2/STAT3 intensifies the anti-tumor activity of arsenic trioxide in acute myeloid leukemia cells: novel synergistic mechanism via the mediation of reactive oxygen species. Eur. J. Pharmacol. 2018;834:65–76. doi: 10.1016/j.ejphar.2018.07.010. [DOI] [PubMed] [Google Scholar]
  • 111.Gorrini C., Harris I.S., Mak T.W. Modulation of oxidative stress as an anticancer strategy. Nat. Rev. Drug Discov. 2013;12:931–947. doi: 10.1038/nrd4002. [DOI] [PubMed] [Google Scholar]
  • 112.Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 113.Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12:31–46. doi: 10.1158/2159-8290.CD-21-1059. [DOI] [PubMed] [Google Scholar]
  • 114.Hangauer M.J., Viswanathan V.S., Ryan M.J., Bole D., Eaton J.K., Matov A., Galeas J., Dhruv H.D., Berens M.E., Schreiber S.L., McCormick F., McManus M.T. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature. 2017;551:247–250. doi: 10.1038/nature24297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kuang F., Liu J., Tang D., Kang R. Oxidative damage and antioxidant defense in ferroptosis. Front. Cell Dev. Biol. 2020;8 doi: 10.3389/fcell.2020.586578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Jiang X., Stockwell B.R., Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 2021;22:266–282. doi: 10.1038/s41580-020-00324-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Mattes K., Gerritsen M., Folkerts H., Geugien M., van den Heuvel F.A., Svendsen A.F., Yi G., Martens J., Vellenga E. CD34+ acute myeloid leukemia cells with low levels of reactive oxygen species show increased expression of stemness genes and can be targeted by the BCL2 inhibitor venetoclax. Haematologica. 2020;105:e399–e403. doi: 10.3324/haematol.2019.229997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Grundy M., Balakrishnan S., Fox M., Seedhouse C.H., Russell N.H. Genetic biomarkers predict response to dual BCL-2 and MCL-1 targeting in acute myeloid leukaemia cells. Oncotarget. 2018;9:37777–37789. doi: 10.18632/oncotarget.26540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Souers A.J., Leverson J.D., Boghaert E.R., Ackler S.L., Catron N.D., Chen J., Dayton B.D., Ding H., Enschede S.H., Fairbrother W.J., Huang D.C., Hymowitz S.G., Jin S., Khaw S.L., Kovar P.J., Lam L.T., Lee J., Maecker H.L., Marsh K.C., Mason K.D., Mitten M.J., Nimmer P.M., Oleksijew A., Park C.H., Park C.M., Phillips D.C., Roberts A.W., Sampath D., Seymour J.F., Smith M.L., Sullivan G.M., Tahir S.K., Tse C., Wendt M.D., Xiao Y., Xue J.C., Zhang H., Humerickhouse R.A., Rosenberg S.H., Elmore S.W. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat. Med. 2013;19:202–208. doi: 10.1038/nm.3048. [DOI] [PubMed] [Google Scholar]
  • 120.Jones C.L., Stevens B.M., D'Alessandro A., Reisz J.A., Culp-Hill R., Nemkov T., Pei S., Khan N., Adane B., Ye H., Krug A., Reinhold D., Smith C., DeGregori J., Pollyea D.A., Jordan C.T. Inhibition of amino acid metabolism selectively targets human leukemia stem cells. Cancer Cell. 2018;34:724–740.e4. doi: 10.1016/j.ccell.2018.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Pollyea D.A., Stevens B.M., Jones C.L., Winters A., Pei S., Minhajuddin M., D'Alessandro A., Culp-Hill R., Riemondy K.A., Gillen A.E., Hesselberth J.R., Abbott D., Schatz D., Gutman J.A., Purev E., Smith C., Jordan C.T. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat. Med. 2018;24:1859–1866. doi: 10.1038/s41591-018-0233-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Buettner R., Nguyen L.X.T., Morales C., Chen M.H., Wu X., Chen L.S., Hoang D.H., Hernandez Vargas S., Pullarkat V., Gandhi V., Marcucci G., Rosen S.T. Targeting the metabolic vulnerability of acute myeloid leukemia blasts with a combination of venetoclax and 8-chloro-adenosine. J. Hematol. Oncol. 2021;14:70. doi: 10.1186/s13045-021-01076-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.DiNardo C.D., Jonas B.A., Pullarkat V., Thirman M.J., Garcia J.S., Wei A.H., Konopleva M., Döhner H., Letai A., Fenaux P., Koller E., Havelange V., Leber B., Esteve J., Wang J., Pejsa V., Hájek R., Porkka K., Illés Á., Lavie D., Lemoli R.M., Yamamoto K., Yoon S.S., Jang J.H., Yeh S.P., Turgut M., Hong W.J., Zhou Y., Potluri J., Pratz K.W. Azacitidine and Venetoclax in previously untreated acute myeloid leukemia. N. Engl. J. Med. 2020;383:617–629. doi: 10.1056/NEJMoa2012971. [DOI] [PubMed] [Google Scholar]
  • 124.Stevens B.M., Jones C.L., Pollyea D.A., Culp-Hill R., D'Alessandro A., Winters A., Krug A., Abbott D., Goosman M., Pei S., Ye H., Gillen A.E., Becker M.W., Savona M.R., Smith C., Jordan C.T. Fatty acid metabolism underlies venetoclax resistance in acute myeloid leukemia stem cells. Nat. Can. (Que.) 2020;1:1176–1187. doi: 10.1038/s43018-020-00126-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Hoang D.H., Buettner R., Valerio M., Ghoda L., Zhang B., Kuo Y.H., Rosen S.T., Burnett J., Marcucci G., Pullarkat V., Nguyen L.X.T. Arsenic trioxide and venetoclax synergize against AML progenitors by ROS induction and inhibition of Nrf2 activation. Int. J. Mol. Sci. 2022 Jun 12;23(12):6568. doi: 10.3390/ijms23126568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Cho H., Jang J.E., Eom J.I., Jeung H.K., Chung H., Kim J.S., Cheong J.W., Min Y.H. Arsenic trioxide synergistically promotes the antileukaemic activity of venetoclax by downregulating Mcl-1 in acute myeloid leukaemia cells. Exp. Hematol. Oncol. 2021;10:28. doi: 10.1186/s40164-021-00221-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Hoang D.H., Morales C., Rodriguez I.R., Valerio M., Guo J., Chen M.H., Wu X., Horne D., Gandhi V., Chen L.S., Zhang B., Pullarkat V., Rosen S.T., Marcucci G., Buettner R., Nguyen L.X.T. Synergy of venetoclax and 8-chloro-adenosine in AML: the interplay of rRNA inhibition and fatty acid metabolism. Cancers. 2022;14:1446. doi: 10.3390/cancers14061446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.He L., Arnold C., Thoma J., Rohde C., Kholmatov M., Garg S., Hsiao C.C., Viol L., Zhang K., Sun R., Schmidt C., Janssen M., MacRae T., Huber K., Thiede C., Hébert J., Sauvageau G., Spratte J., Fluhr H., Aust G., Müller-Tidow C., Niehrs C., Pereira G., Hamann J., Tanaka M., Zaugg J.B., Pabst C. CDK7/12/13 inhibition targets an oscillating leukemia stem cell network and synergizes with venetoclax in acute myeloid leukemia. EMBO Mol. Med. 2022;14 doi: 10.15252/emmm.202114990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Lachowiez C.A., Reville P.K., Kantarjian H., Jabbour E., Borthakur G., Daver N., Loghavi S., Furudate K., Xiao L., Pierce S., Short N.J., Maiti A., Yilmaz M., Sasaki K., Takahashi K., Konopleva M., Pemmaraju N., Popat U., Shpall E., Garcia-Manero G., Ravandi F., DiNardo C.D., Kadia T.M. Venetoclax combined with induction chemotherapy in patients with newly diagnosed acute myeloid leukaemia: a post hoc, propensity score-matched, cohort study. Lancet Haematol. 2022;9:e350–e360. doi: 10.1016/S2352-3026(22)00076-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Hassani S., Ghaffari P., Chahardouli B., Alimoghaddam K., Ghavamzadeh A., Alizadeh S., Ghaffari S.H. Disulfiram/copper causes ROS levels alteration, cell cycle inhibition, and apoptosis in acute myeloid leukaemia cell lines with modulation in the expression of related genes. Biomed. Pharmacother. 2018;99:561–569. doi: 10.1016/j.biopha.2018.01.109. [DOI] [PubMed] [Google Scholar]
  • 131.Xu B., Wang S., Li R., Chen K., He L., Deng M., Kannappan V., Zha J., Dong H., Wang W. Disulfiram/copper selectively eradicates AML leukemia stem cells in vitro and in vivo by simultaneous induction of ROS-JNK and inhibition of NF-κB and Nrf2. Cell Death Dis. 2017;8:e2797. doi: 10.1038/cddis.2017.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Jin Y., Lu Z., Ding K., Li J., Du X., Chen C., Sun X., Wu Y., Zhou J., Pan J. Antineoplastic mechanisms of niclosamide in acute myelogenous leukemia stem cells: inactivation of the NF-kappaB pathway and generation of reactive oxygen species. Cancer Res. 2010;70:2516–2527. doi: 10.1158/0008-5472.CAN-09-3950. [DOI] [PubMed] [Google Scholar]
  • 133.Cao K., Du Y., Bao X., Han M., Su R., Pang J., Liu S., Shi Z., Yan F., Feng S. Glutathione-bioimprinted nanoparticles targeting of N6-methyladenosine FTO demethylase as a strategy against leukemic stem cells. Small. 2022;18 doi: 10.1002/smll.202106558. [DOI] [PubMed] [Google Scholar]
  • 134.Zhou J., Bi C., Cheong L.L., Mahara S., Liu S.C., Tay K.G., Koh T.L., Yu Q., Chng W.J. The histone methyltransferase inhibitor, DZNep, up-regulates TXNIP, increases ROS production, and targets leukemia cells in AML. Blood. 2011;118:2830–2839. doi: 10.1182/blood-2010-07-294827. [DOI] [PubMed] [Google Scholar]
  • 135.Li Y., Chen K., Zhou Y., Xiao Y., Deng M., Jiang Z., Ye W., Wang X., Wei X., Li J., Liang J., Zheng Z., Yao Y., Wang W., Li P., Xu B. A new strategy to target acute myeloid leukemia stem and progenitor cells using chidamide, a histone deacetylase inhibitor. Curr. Cancer Drug Targets. 2015;15:493–503. doi: 10.2174/156800961506150805153230. [DOI] [PubMed] [Google Scholar]
  • 136.Ferreira P.M.P., Sousa R.W.R., Ferreira J.R.O., Militão G.C.G., Bezerra D.P. Chloroquine and hydroxychloroquine in antitumor therapies based on autophagy-related mechanisms. Pharmacol. Res. 2021;168 doi: 10.1016/j.phrs.2021.105582. [DOI] [PubMed] [Google Scholar]
  • 137.Chen J., Pan Q., Bai Y., Chen X., Zhou Y. Hydroxychloroquine induces apoptosis in cholangiocarcinoma via reactive oxygen species accumulation induced by autophagy inhibition. Front. Mol. Biosci. 2021;8 doi: 10.3389/fmolb.2021.720370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Folkerts H., Hilgendorf S., Wierenga A.T.J., Jaques J., Mulder A.B., Coffer P.J., Schuringa J.J., Vellenga E. Inhibition of autophagy as a treatment strategy for p53 wild-type acute myeloid leukemia. Cell Death Dis. 2017;8:e2927. doi: 10.1038/cddis.2017.317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Zhang H., Mi J.Q., Fang H., Wang Z., Wang C., Wu L., Zhang B., Minden M., Yang W.T., Wang H.W., Li J.M., Xi X.D., Chen S.J., Zhang J., Chen Z., Wang K.K. Preferential eradication of acute myelogenous leukemia stem cells by fenretinide. Proc. Natl. Acad. Sci. U. S. A. 2013;110:5606–5611. doi: 10.1073/pnas.1302352110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Zhao X.Y., Zhang R.R., Ye Q., Qiu F., Xu H.Y., Wei F.G., Zhang H. 4-Hydroxyphenyl retinamide preferentially targets FLT3 mutated acute myeloid leukemia via ROS induction and NF-κB inhibition. Curr Med Sci. 2020;40:810–816. doi: 10.1007/s11596-020-2259-0. [DOI] [PubMed] [Google Scholar]
  • 141.Guzman M.L., Li X., Corbett C.A., Rossi R.M., Bushnell T., Liesveld J.L., Hébert J., Young F., Jordan C.T. Rapid and selective death of leukemia stem and progenitor cells induced by the compound 4-benzyl, 2-methyl, 1,2,4-thiadiazolidine, 3,5 dione (TDZD-8) Blood. 2007;110:4436–4444. doi: 10.1182/blood-2007-05-088815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Sukhai M.A., Prabha S., Hurren R., Rutledge A.C., Lee A.Y., Sriskanthadevan S., Sun H., Wang X., Skrtic M., Seneviratne A., Cusimano M., Jhas B., Gronda M., MacLean N., Cho E.E., Spagnuolo P.A., Sharmeen S., Gebbia M., Urbanus M., Eppert K., Dissanayake D., Jonet A., Dassonville-Klimpt A., Li X., Datti A., Ohashi P.S., Wrana J., Rogers I., Sonnet P., Ellis W.Y., Corey S.J., Eaves C., Minden M.D., Wang J.C., Dick J.E., Nislow C., Giaever G., Schimmer A.D. Lysosomal disruption preferentially targets acute myeloid leukemia cells and progenitors. J. Clin. Invest. 2013;123:315–328. doi: 10.1172/JCI64180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Pei S., Minhajuddin M., Callahan K.P., Balys M., Ashton J.M., Neering S.J., Lagadinou E.D., Corbett C., Ye H., Liesveld J.L., O'Dwyer K.M., Li Z., Shi L., Greninger P., Settleman J., Benes C., Hagen F.K., Munger J., Crooks P.A., Becker M.W., Jordan C.T. Targeting aberrant glutathione metabolism to eradicate human acute myelogenous leukemia cells. J. Biol. Chem. 2013;288:33542–33558. doi: 10.1074/jbc.M113.511170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Guzman M.L., Rossi R.M., Karnischky L., Li X., Peterson D.R., Howard D.S., Jordan C.T. The sesquiterpene lactone parthenolide induces apoptosis of human acute myelogenous leukemia stem and progenitor cells. Blood. 2005;105:4163–4169. doi: 10.1182/blood-2004-10-4135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Kim Y.R., Eom J.I., Kim S.J., Jeung H.K., Cheong J.W., Kim J.S., Min Y.H. Myeloperoxidase expression as a potential determinant of parthenolide-induced apoptosis in leukemia bulk and leukemia stem cells. J. Pharmacol. Exp. Therapeut. 2010;335:389–400. doi: 10.1124/jpet.110.169367. [DOI] [PubMed] [Google Scholar]
  • 146.Guzman M.L., Rossi R.M., Neelakantan S., Li X., Corbett C.A., Hassane D.C., Becker M.W., Bennett J.M., Sullivan E., Lachowicz J.L., Vaughan A., Sweeney C.J., Matthews W., Carroll M., Liesveld J.L., Crooks P.A., Jordan C.T. An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells. Blood. 2007;110:4427–4435. doi: 10.1182/blood-2007-05-090621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Kempema A.M., Widen J.C., Hexum J.K., Andrews T.E., Wang D., Rathe S.K., Meece F.A., Noble K.E., Sachs Z., Largaespada D.A., Harki D.A. Synthesis and antileukemic activities of C1-C10-modified parthenolide analogues. Bioorg. Med. Chem. 2015;23:4737–4745. doi: 10.1016/j.bmc.2015.05.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Darwish N., Sudha T., Godugu K., Bharali D.J., Elbaz O., El-Ghaffar H., Azmy E., Anber N., Mousa S.A. Novel targeted nano-parthenolide molecule against NF-kB in acute myeloid leukemia. Molecules. 2019;24:2103. doi: 10.3390/molecules24112103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Ji Q., Ding Y.H., Sun Y., Zhang Y., Gao H.E., Song H.N., Yang M., Liu X.L., Zhang Z.X., Li Y.H., Gao Y.D. Antineoplastic effects and mechanisms of micheliolide in acute myelogenous leukemia stem cells. Oncotarget. 2016;7:65012–65023. doi: 10.18632/oncotarget.11342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Lee E.A., Angka L., Rota S.G., Hanlon T., Mitchell A., Hurren R., Wang X.M., Gronda M., Boyaci E., Bojko B., Minden M., Sriskanthadevan S., Datti A., Wrana J.L., Edginton A., Pawliszyn J., Joseph J.W., Quadrilatero J., Schimmer A.D., Spagnuolo P.A. Targeting mitochondria with avocatin b induces selective leukemia cell death. Cancer Res. 2015;75:2478–2488. doi: 10.1158/0008-5472.CAN-14-2676. [DOI] [PubMed] [Google Scholar]
  • 151.Zhou Z.L., Yang Y.X., Ding J., Li Y.C., Miao Z.H. Triptolide: structural modifications, structure-activity relationships, bioactivities, clinical development and mechanisms. Nat. Prod. Rep. 2012;29:457–475. doi: 10.1039/c2np00088a. [DOI] [PubMed] [Google Scholar]
  • 152.Liu Y., Chen F., Wang S., Guo X., Shi P., Wang W., Xu B. Low-dose triptolide in combination with idarubicin induces apoptosis in AML leukemic stem-like KG1a cell line by modulation of the intrinsic and extrinsic factors. Cell Death Dis. 2013;4:e948. doi: 10.1038/cddis.2013.467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Hassane D.C., Guzman M.L., Corbett C., Li X., Abboud R., Young F., Liesveld J.L., Carroll M., Jordan C.T. Discovery of agents that eradicate leukemia stem cells using an in silico screen of public gene expression data. Blood. 2008;111:5654–5662. doi: 10.1182/blood-2007-11-126003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Zhe N., Chen S., Zhou Z., Liu P., Lin X., Yu M., Cheng B., Zhang Y., Wang J. HIF-1α inhibition by 2-methoxyestradiol induces cell death via activation of the mitochondrial apoptotic pathway in acute myeloid leukemia. Cancer Biol. Ther. 2016;17:625–634. doi: 10.1080/15384047.2016.1177679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Rota S.G., Roma A., Dude I., Ma C., Stevens R., MacEachern J., Graczyk J., Espiritu S., Rao P.N., Minden M.D., Kreinin E., Hess D.A., Doxey A.C., Spagnuolo P.A. Estrogen receptor β is a novel target in acute myeloid leukemia. Mol. Cancer Therapeut. 2017;16:2618–2626. doi: 10.1158/1535-7163.MCT-17-0292. [DOI] [PubMed] [Google Scholar]
  • 156.Gao M., Wang X.Z., Shu Y.T., Liang J., Chen L., Liu R., Liang J.Y., Wen H.M. Peperomin E and its synthetic amino derivatives: potent agents targeting leukaemia stem cells. RSC Adv. 2017;7:52200–52209. doi: 10.1039/C7RA09928B. [DOI] [Google Scholar]
  • 157.Wang X., Gao M., Zhang J., Ma Y., Qu W., Liang J., Wu H., Wen H. Peperomin E and its orally bioavailable analog induce oxidative stress-mediated apoptosis of acute myeloid leukemia progenitor cells by targeting thioredoxin reductase. Redox Biol. 2019;24 doi: 10.1016/j.redox.2019.101153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Zhou G.B., Kang H., Wang L., Gao L., Liu P., Xie J., Zhang F.X., Weng X.Q., Shen Z.X., Chen J., Gu L.J., Yan M., Zhang D.E., Chen S.J., Wang Z.Y., Chen Z. Oridonin, a diterpenoid extracted from medicinal herbs, targets AML1-ETO fusion protein and shows potent antitumor activity with low adverse effects on t(8;21) leukemia in vitro and in vivo. Blood. 2007;109:3441–3450. doi: 10.1182/blood-2006-06-032250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Zhen T., Wu C.F., Liu P., Wu H.Y., Zhou G.B., Lu Y., Liu J.X., Liang Y., Li K.K., Wang Y.Y., Xie Y.Y., He M.M., Cao H.M., Zhang W.N., Chen L.M., Petrie K., Chen S.J., Chen Z. Targeting of AML1-ETO in t(8;21) leukemia by oridonin generates a tumor suppressor-like protein. Sci. Transl. Med. 2012;4:127ra38. doi: 10.1126/scitranslmed.3003562. [DOI] [PubMed] [Google Scholar]
  • 160.Bezerra D.P., Militão G.C., de Castro F.O., Pessoa C., de Moraes M.O., Silveira E.R., Lima M.A., Elmiro F.J., Costa-Lotufo L.V. Piplartine induces inhibition of leukemia cell proliferation triggering both apoptosis and necrosis pathways. Toxicol. Vitro. 2007;21:1–8. doi: 10.1016/j.tiv.2006.07.007. [DOI] [PubMed] [Google Scholar]
  • 161.Bezerra D.P., Pessoa C., de Moraes M.O., Saker-Neto N., Silveira E.R., Costa-Lotufo L.V. Overview of the therapeutic potential of piplartine (piperlongumine) Eur. J. Pharmaceut. Sci. 2013;48:453–463. doi: 10.1016/j.ejps.2012.12.003. https://doi: 10.1016/j.ejps.2012.12.003 [DOI] [PubMed] [Google Scholar]
  • 162.Adams D.J., Dai M., Pellegrino G., Wagner B.K., Stern A.M., Shamji A.F., Schreiber S.L. Synthesis, cellular evaluation, and mechanism of action of piperlongumine analogs. Proc. Natl. Acad. Sci. U. S. A. 2012;109:15115–15120. doi: 10.1073/pnas.1212802109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Costa C.O.S., Araujo Neto J.H., Baliza I.R.S., Dias R.B., Valverde L.F., Vidal M.T.A., Sales C.B.S., Rocha C.A.G., Moreira D.R.M., Soares M.B.P., Batista A.A., Bezerra D.P. Novel piplartine-containing ruthenium complexes: synthesis, cell growth inhibition, apoptosis induction and ROS production on HCT116 cells. Oncotarget. 2017;8:104367–104392. doi: 10.18632/oncotarget.22248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Baliza I.R.S., Silva S.L.R., Santos L. dS., Neto J.H.A., Dias R.B., Sales C.B.S., Rocha C.A.G., Soares M.B.P., Batista A.A., Bezerra D.P. Ruthenium complexes with piplartine cause apoptosis through MAPK signaling by a p53-dependent pathway in human colon carcinoma cells and inhibit tumor development in a xenograft model. Front. Oncol. 2019;9:582. doi: 10.3389/fonc.2019.00582. https://doi: 10.3389/fonc.2019.00582 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Oliveira M.S., Barbosa M.I.F., de Souza T.B., Moreira D.R.M., Martins F.T., Villarreal W., Machado R.P., Doriguetto A.C., Soares M.B.P., Bezerra D.P. A novel platinum complex containing a piplartine derivative exhibits enhanced cytotoxicity, causes oxidative stress and triggers apoptotic cell death by ERK/p38 pathway in human acute promyelocytic leukemia HL-60 cells. Redox Biol. 2019;20:182–194. doi: 10.1016/j.redox.2018.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Dartora V.F.C., Salata G.C., Passos J.S., Branco P.C., Silveira E., Steiner A.A., Costa-Lotufo L.V., Lopes L.B. Hyaluronic acid nanoemulsions improve piplartine cytotoxicity in 2D and 3D breast cancer models and reduce tumor development after intraductal administration. Int. J. Biol. Macromol. 2022;219:84–95. doi: 10.1016/j.ijbiomac.2022.07.162. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No data was used for the research described in the article.


Articles from Redox Biology are provided here courtesy of Elsevier

RESOURCES