Skip to main content
Blood logoLink to Blood
. 2011 Mar 21;117(24):6425–6437. doi: 10.1182/blood-2010-11-283598

From an old remedy to a magic bullet: molecular mechanisms underlying the therapeutic effects of arsenic in fighting leukemia

Sai-Juan Chen 1,2,, Guang-Biao Zhou 3, Xiao-Wei Zhang 1,2, Jian-Hua Mao 1, Hugues de Thé 4, Zhu Chen 1,2,
PMCID: PMC3123014  PMID: 21422471

Abstract

Arsenic had been used in treating malignancies from the 18th to mid-20th century. In the past 3 decades, arsenic was revived and shown to be able to induce complete remission and to achieve, when combined with all-trans retinoic acid and chemotherapy, a 5-year overall survival of 90% in patients with acute promyelocytic leukemia driven by the t(15;17) translocation-generated promyelocytic leukemia–retinoic acid receptor α (PML-RARα) fusion. Molecularly, arsenic binds thiol residues and induces the formation of reactive oxygen species, thus affecting numerous signaling pathways. Interestingly, arsenic directly binds the C3HC4 zinc finger motif in the RBCC domain of PML and PML-RARα, induces their homodimerization and multimerization, and enhances their interaction with the SUMO E2 conjugase Ubc9, facilitating subsequent sumoylation/ubiquitination and proteasomal degradation. Arsenic-caused intermolecular disulfide formation in PML also contributes to PML-multimerization. All-trans retinoic acid, which targets PML-RARα for degradation through its RARα moiety, synergizes with arsenic in eliminating leukemia-initiating cells. Arsenic perturbs a number of proteins involved in other hematologic malignancies, including chronic myeloid leukemia and adult T-cell leukemia/lymphoma, whereby it may bring new therapeutic benefits. The successful revival of arsenic in acute promyelocytic leukemia, together with modern mechanistic studies, has thus allowed a new paradigm to emerge in translational medicine.

Introduction

Arsenic is the 20th most abundant element in the earth's crust.1 It is a chemical analog of phosphorus and lies directly below P in the periodic table. A unique feature of arsenic is its extremely paradoxical abilities: it is toxic to humans, animals, and plants, but is used instead of phosphorus by a recently isolated bacterium2; it kills people, but saves lives; it can cause some cancers but cures others such as the acute promyelocytic leukemia (APL)3; it is one of the oldest drugs in the world but was progressively revived between the 1970s and 1990s, because of its striking efficacy on APL, which represented the most malignant type of acute leukemia.3 These paradoxical effects of arsenic reflect its unique metabolism and multiple properties.4 In this review, we address how arsenic rebuilds its reputation from a notorious poison to a new lease of life by mechanistic molecular and translational medicine studies.

Arsenic in nature

Arsenic is the 33rd element in the periodic table and exists ubiquitously in either inorganic or organic forms, pure metallic arsenic being rarely found in nature. Arsenic occurs in 5 different valence states, +V (arsenate), +III (arsenite), +I (arsonium metal), 0 (arsenic), and −III (arsine).5 Most arsenic compounds have no smell or special taste and are white or colorless powders that do not evaporate. Arsenic gets into air when contaminated materials are burned. Inorganic arsenic occurs naturally in soils and sedimentary rocks such as minerals and ores containing copper or lead, and it occurs in combination with many other elements especially oxygen, chlorine, and sulfur. There are 3 inorganic arsenic forms, namely, red arsenic (As4S4, also known as realgar), yellow arsenic (As2S3, also known as orpiment), and white arsenic (arsenic trioxide, ATO; As2O3). ATO is made by burning realgar or orpiment.6 Organic arsenic is arsenic compounds containing carbon that can be found in nature in water, natural gas, and shale oil.5 Examples of organic arsenic are methylarsine (CH3AsH2), dimethylarsine [(CH3)2AsH], trimethylarsine [(CH3)3As], monomethylarsonic acid [CH3AsO(OH)2, MMAV], monomethylarsenous acid [CH3As(OH)2, MMAIII], dimethylarsinic acid [(CH3)2AsO(OH), DMAV], dimethylarsenous acid [(CH3)2AsOH, DMAIII], trimethylarsinic oxide [(CH3)3AsO, TMAO], tetramethylarsonium ion [(CH3)4As+, TMA+], arsenobetaine [(CH3)3As+CH2COO, AB], arsenocholine [(CH3)As+CH2 CH2OH, AC], and others.5,79 Darinaparsin is an organic arsenic composed of dimethylated arsenic linked to glutathione.10

Regarding arsenic metabolism, an arsenic methyltransferase, encoded by a gene that was originally annotated as Cyt19 and subsequently As3mt, was identified in rat that catalyzes the methylation of arsenite, with S-adenosyl-L-methionine as the methyl donor.11,12 Inorganic arsenic is metabolized by a sequential process involving a 2-electron reduction of pentavalent arsenic to trivalent arsenic, followed by oxidative methylation to pentavalent organic arsenic.13,14 The postulated scheme is as follows: iAsV → iAsIII → MAsV → MAsIII → DMAsV → DMAsIII → TMAsV → TMAsIII.1316 Of note, derivatives of MAsIII and DMAsIII are more toxic than either iAsV or iAsIII.16 Methylarsine oxide (MAsIIIO) and to a lesser extent iododimethylarsine are more potent growth inhibitors and apoptotic inducers than iAsIII in leukemia cells, and apoptosis is associated with greater hydrogen peroxide accumulation and inhibition of glutathione peroxidase activity. In vivo hepatic methylation of iAsIII may contribute to ATO-induced apoptosis but not differentiation of APL cells.15

Arsenic is toxic to human health in that it can induce skin lesions, hemorrhagic gastroenteritis, cardiac arrhythmia, psychiatric disease, and cancers.9,17,18 Generally, inorganic arsenic species are more toxic than organic forms to living organisms, and arsenite is usually more toxic than arsenate. Exposure to ATO by ingestion of 70-80 mg has been reported to be fatal for humans.7,19 The World Health Organization set the first International Drinking Water Standard for arsenic concentration at 200 μg/L in 1958, recommended lowering the standard to 50 μg/L in 1963, and further lowered the standard to 10 μg/L in 1993.20 Yet millions of people worldwide ingest drinking water contaminated with arsenic at levels > 100 μg/L.5,7,20

The toxicity of trivalent arsenic is related to its high affinity for the sulfhydryl groups of biomolecules such as glutathione and lipoic acid and the cysteinyl residues of many proteins and enzymes.7,21 Arsenic up-regulates glutathione-related genes and enzyme activities and binds to sulfhydryl groups.22,23 The formation of AsIII-sulfur bonds results in various harmful effects by inhibiting the activities of enzymes such as glutathione reductase, glutathione peroxidases, thioredoxin reductase, and thioredoxin peroxidase.4,7,21 Because all of these enzymes regulate cellular redox status by providing antioxidant defense, arsenic exposure leads to production of reactive oxygen species (ROS).4 Similarly, flavin enzymes such as NAD(P)H oxidase and NO synthase isozymes have been proposed to be involved in the generation of ROS associated with arsenic exposure.4 Arsenic also alters global histone H3 methylation.24 Consequently, arsenic affects many signal transduction cascades (eg, activation of the epidermal growth factor receptor [EGFR] signal pathway25) and activates (or inactivates) transcription factors such as activated protein-126 and nuclear factor–erythroid 2-related factor 2.27,28 Biomarkers of arsenic exposure include the total arsenic in urine,29 clastogenicity in peripheral lymphocytes, micronuclei in oral mucosa and bladder cells, and induction of heme oxygenase. A potential susceptibility biomarker is variability in arsenic metabolism, which reflects polymorphisms in the genes that encode the arsenic-metabolizing enzymes.29

Arsenic as an old remedy

Arsenic is one of the oldest drugs in the world. It was first mentioned by Hippocrates (460-370 BC) who used realgar and orpiment pastes to treat ulcers in Western medicine. In China, arsenic pills for the treatment of periodic fever were recorded in the Chinese Nei Jing Treaty (263 BC).30 Si-Miao Sun (581-682 AD) purified a medicine composed of realgar, orpiment, and ATO in treating malaria,31 whereas Shi-Zhen Li (1518-1593 AD)32 in the Ming Dynasty described the use of ATO as a remedy for a variety of diseases in his pharmacopedia. Arsenic therapy was introduced to Europe by Avicennes (980-1037 AD) and Paracelsus (1493-1541 AD). In 1774, Lefébure introduced an arsenic-containing paste proposed to be an “established remedy to radically cure all cancers.”6 Fowler solution (1% potassium arsenite, KAsO2) was first described in 1845 and was used to treat anemia and rheumatism, psoriasis, eczematous eruptions, dermatitis herpetiformis, asthma, cholera, and syphilis. In 1865, Fowler solution was the first chemotherapeutic agent used in the treatment of leukemia that produced some transient improvement.3335 In 1931 Forkner and Scott, at Boston City Hospital, “rediscovered” Fowler solution for the treatment of chronic myeloid leukemia (CML), and arsenicals and irradiation remained the treatment of choice until busulphan was introduced in 1953.3335

Arsenic in treating APL

APL, the M3 subtype of acute myeloid leukemia (AML M3), is characterized by the accumulation of abnormal promyelocytes in blood and bone marrow, the occurrence of fibrinogenopenia and disseminated intravascular coagulation, and the specific chromosomal translocation t(15;17)(q22;q21).3638 The t(15;17) fuses the retinoic acid receptor α (RARα) gene on chromosome 17 to the promyelocytic leukemia (PML) gene on 15q, yielding the PML-RARα fusion protein that is the key driver of APL leukemogenesis.3638 APL was firstly described by Hillestad in 19573 and was considered at the time the most fatal type of acute leukemia.3,36,38 The past 5 decades have witnessed tremendous advances in improving APL outcome from highly fatal to highly curable (Figure 1).35,36,38,39 Although chemotherapy (anthracyclines) shines the first light of hope,40 all-trans retinoic acid (ATRA), which triggers terminal differentiation of APL cells, achieves a complete remission (CR) rate of 90%38,4144 (and references within these articles). Although ATRA alone rarely cures APL, its combination with anthracyclines allowed a significant number of cures. ATO further prolongs survival of patients with APL, especially those with relapsed or refractory disease, and cures a number of them as a single agent. Moreover, combined use of ATRA and ATO not only markedly enhances clearance of PML-RARα transcript, but allows the 5-year overall survival (OS) to reach 91.7%38,39,45 (Figure 1). Mechanistically, both ATRA and ATO trigger catabolism of the PML-RARα fusion protein. However, ATO has no effect on APL driven by promyelocytic leukemia zinc finger (PLZF)–RARα, which is generated by t(11;17) and accounts for 1%-2% of patients with APL.4651 Thus, the story of APL can serve as a model for the development of targeted therapies and curative approaches for malignant disease.51

Figure 1.

Figure 1

A historical view of the treatment outcome (represented by 5-year overall survival) of APL.

Clinical efficacy

In the early 1970s, a group from Harbin Medical University in northeastern China tested Ailing-1 containing 1% ATO and a trace amount of mercury chloride in a variety of cancers by intravenous administration. In the 1990s, Sun et al52 showed that Ailing-1 induced CR in 21 of 32 patients with APL with an impressive 10-year survival rate of 30%. The efficacy of pure ATO in treating relapsed APL was then reported by Shanghai Institute of Hematology (SIH) in 1996-1999.53,54 Shen et al54 evaluated the therapeutic effect of ATO in the treatment of 15 patients with APL at relapse after ATRA-induced and chemotherapy-maintained CR. ATO was administered intravenously at the dose of 10 mg/d. They showed that CR was achieved in 9 of 10 patients (90%) treated with ATO alone and in the remaining 5 patients with ATO in combination with low-dose chemotherapeutic drugs or ATRA. No bone marrow depression was encountered during ATO treatment.54 Niu et al53 reported that clinical CR was obtained in 8 of 11 newly diagnosed cases (72.7%) and in 40 of 47 relapsed patients (85.1%). They recommended that ATRA is used as first choice for remission induction in newly diagnosed APL cases, whereas ATO can be either used as a rescue for relapsed cases or included into multidrug consolidation/maintenance clinical trials. Furthermore, after CR achieved with the use of ATO alone, a molecular remission is obtainable in a relatively high proportion of the patients, from 72%55 to 91%56 in different multicenter studies, showing that ATO is an effective drug for APL. With the use of ATO as a single agent, a good long-term remission can be obtained, as evidenced by a 5-year event-free survival (EFS) of 69%57 to 72.7%58 in 2 recent reports (Table 1).

Table 1.

Outcome of patients with APL treated with ATO-based regimens since 2006

Year Author No. Regimen CR, % EFS, %* DFS, %* OS, %*
2010 Powell et al59 244 (237 standard controls) Induction: ATRA/CT; consolidation: ATRA/CT/ATO 90 80 (3 y) 90 (3 y) 86 (3 y)
2010 Zhou et al58 19 (age ≤ 15) ATO 89.5 72.7 NR 83.9
2010 Mathews et al57 72 ATO 86.1 69 80 74.2
2009 Dai et al60 90 ATO/ATRA 93.3 92.2 (3 y)
2006 Ghavamzadeh et al61 111 ATO 85.6 NR 63.7 (2 y) 87.6 (3 y)
2008 Hu et al39 85 ATO/ATRA/CT 94.1 89.2 NR 91.7
2007 Wu et al62 114 As4S4/ATRA or As4S4/CT 94 (4 y)

NR indicates not reported.

*

Five years, unless otherwise indicated.

The efficacy of As4S4 in APL was also investigated. In 1995, Huang et al63 reported remarkable results of the Realgar-Indigo Naturalis Formula, in which As4S4 is the principle ingredient, in treating 60 patients with APL, including 43 newly diagnosed cases. In 2002, Lu et al64 showed that of the 129 patients with APL receiving pure As4S4,103 cases (79.8%) achieved CR. In the newly diagnosed group (19 patients), the estimated disease-free survival rates for 1 and 3 years were 86.1% and 76.6%, respectively, with a median follow-up time of 13.5 months. They further showed that in 114 patients receiving As4S4 in combination with ATRA or chemotherapy (mitoxantrone or hydroxyurea or both), the estimated 4-year disease-free survival was 94%.62

Mechanisms of action

PML-RARα as a direct arsenic target.

That ATO exerts drastic therapeutic effects against APL, but not other subtypes of AML (including variant APL driven by the PLZF-RARα fusion), suggests a crucial link between its mechanism of action and PML-RARα, a potent transcriptional regulator that alters expression of ATRA or non-ATRA target genes.38,6567 Indeed, it was rapidly shown that arsenic efficiently triggers the degradation of PML-RARα through its PML moiety.6870

The wild-type PML and the PML moiety in fusion protein harbor the RBCC domain,69,70 which contains 1 RING and 2 B boxes (B box 1 and B box 2) motifs capable of binding metal (physiologically zinc) ions, and a coiled-coil (CC) motif mediating homodimer formation.71,72 Figure 2A is a schematic representation of major domains of PML, RARα, and PML-RARα. In normal cells, PML proteins are the main components of spherical nuclear organelles designated nuclear bodies (NBs), which play a key role in regulation of apoptosis, epigenetic control of chromatin, and transcriptional expression, as well as storage/modulation of certain nuclear proteins.7375 NB structures are disrupted in APL cells, because of formation of PML/PML-RARα heterocomplex. This yields a much larger number of tiny dots by immunofluoresence analysis, showing the disorganization of this nuclear domain.76 The ATRA and arsenic-triggered degradation process is intimately coupled to changes in PML/PML-RARα localization.46,68,70,7779 Indeed, in ATO-treated APL cells or cells transfected with PML, PML-RARα,79 both PML-RARα and wild-type PML are quickly translocated to the nuclear matrix, sumoylated, ubiquitinated, and subsequently degraded by the proteasome. In parallel, in arsenic-treated APL cells, dots of PML-containing proteins are aggregated to form larger particles at the nuclear matrix, before their ultimate disappearance (Figure 2B) and the cells are committed to apoptosis or partial differentiation (Figure 2C).

Figure 2.

Figure 2

Effects of arsenic on APL cells. (A) Schematic represents the structure of PML, RARα, and PML-RARα. (B) The NB4 cells were treated with 1μM ATO for indicated time points and were assessed by immunofluorescence staining with an anti-PML antibody (green). Stainings were analyzed using a Leica TCS SP5 confocal microscope equipped with a 63×/1.4 NA oil objective (Leica Microsystems). Images were processed using Leica AF lite software. (C) Arsenic induces dual effects on APL cells. The NB4 cells were treated with indicated concentration for 48 hours and stained with the Wright stain. Stainings were analyzed using an Olympus BX51 research microscope equipped with a 100×/1.30 NA oil objective (Olympus). Images were processed using Adobe Photoshop CS (Adobe Systems). Original magnification, ×1000. (D) Colocalization of PML and PML-RARα with the fluorescent organic arsenical ReAsH in NB4 cells. Images were visualized and processed using the equipment described for panel B. (E) The schematic diagram of the structure of PML RING coordinated with zinc or arsenic. (F) Predicted structure of PML RING/UBC9 complex. (G) A working model of the mechanism by which arsenic controls the fate of PML and PML-RARα.

Because the wild-type PML and the PML moiety in the fusion protein harbor a number of adjacently located cysteine residues with metal-binding ability in their RBCC domain,71,72 we hypothesized that ATO might target PML/PML-RARα at this domain. We tested this possibility by biotin-arsenic/streptavidin pull-down affinity assay and a red fluorescent organic arsenic compound ReAsH/immunofluorescent analysis. Zhang et al79 and Jeanne et al46 showed that ReAsH colocalized with PML/PML-RARα (Figure 2D) and that arsenic could directly bind the wild-type and fusion proteins. Consistently, deletion of the RING domain in PML abolished the ReAsH colocalization signal. The PML RING peptide containing C3HC4 (aa 57-91) zinc finger (ZF) was expressed and purified for refined arsenic-binding analyses. With the use of matrix-assisted laser desorption ionization– time of flight mass spectrometry, it was found that 1 PML RING molecule without metal ions (apo-PML RING) could bind 2 arsenics at ATO concentrations of 1-2μM.79 Arsenic bound PML RING through thiol groups of cysteines with the formation of arsenic-sulphur bonds, as evidenced by near-ultraviolet absorbance spectrometry assays. With the use of x-ray absorption spectra assay, including extended x-ray absorption fine structure and x-ray absorption near-edge structure, the local structures of PML RING around metal ions within ∼ 6 Å were obtained, and it was found that trivalent arsenics could coordinate to PML RING each by 3 conserved cysteines, in ZF1 with C60, C77, and C80 and in ZF2 with C72, C88, and C91, compared with the coordination by zinc in ZF1 with C57, C60, C77, C80 and in ZF2 with C72, H74, C88, and C91 (Figure 2E). In addition, the PML B box 2 domain also directly bound arsenic in vitro, whereas its deletion led to a significant reduction of ReASH colocalization signals in cells.46,79 Of note, arsenic was able to competitively replace zinc in PML RING coordinated with zinc (zinc-PML RING) according to the nuclear magnetic resonance heteronuclear single-quantum coherence spectra.79 On binding to arsenic, PML RING underwent conformational changes and aggregation, most probably because of the formation of homodimer by intermolecular As-S bonds, followed by oligomerization among these homodimers.46,79 In in vitro reconstituted conditions and mammalian 2-hybrid assay in cells, arsenic binding to PML facilitated its interaction with the unique small ubiquitin-like protein (SUMO) E2 conjugase UBC9, leading to sumoylation at K65 and K160.80 K160 sumoylation also mediated subsequent recruitment of 11S proteasome.78,81 A 3-dimensional structure modeling shows that the 2 ZFs in PML RING motif locate at the interface with UBC9, providing a structural basis for the formation of PML/UBC9 complex in the presence of arsenic (Figure 2F). Recent studies indicate that RNF4, a ubiquitin E3 ligase containing SUMO interaction motifs,77,82 could recruit sumoylated PML/PML-RARα and promote their proteasomal degradation.77,82 These results show that ATO controls the fate of the PML-RARα by directly binding PML (Figure 2G) and, at least partially, explaining why ATO is effective for APL.79

In addition to formation of arsenic-cysteine bonds that favor aggregation, arsenic-induced ROS also initiate intermolecular disulfide formation.46 Disulfide-linked PML or PML-RARα multimers become nuclear matrix-associated and form NBs. Thus, PML oxidation regulated NB biogenesis. In that respect, non–arsenical oxidants also elicited PML-RARα multimerization, NB association, degradation, and leukemia response in vivo. Critically, oxidants did not affect PLZF-RARα–driven APL, a genetic demonstration that PML is the key target.46 Arsenic can also bind other proteins, including the ubiquitin E3 ligases c-CBL (Casitas B-lineage lymphoma) and SIAH1,83 both harboring RING finger motifs (Table 2).

Table 2.

Arsenic binding proteins

Proteins and references Disease or cell lines Category Functions
PML79 APL Phosphoprotein; scaffold protein Tumor suppressor; probable transcription factor
c-CBL83 CML, K562 cells Ubiquitin E3 ligase Proteolysis
SIAH183 CML, K562 cells Ubiquitin E3 ligase Proteolysis
Trx R84 MCF-7 cells Oxidoreductase Redox regulation
GSR85 Arsenic toxication Oxidoreductase Redox regulation
TPX-2 II86 Ovary cells Peroxidase Redox regulation
PDI87 Fibrosarcoma cells Oxidoreductase Redox regulation
MTs88 Arsenic detoxication Metallothioneins Binding heavy metals
MTF189 Arsenic detoxication Transcription factor Activation of metallothionein transcription
Keap127 hepa1c1c7 cells Phosphoprotein Transcription regulation
Tubulins9092 K562 cells Cell skeleton proteins Structural subunit of microtubules
β-Actin9193 K562 and MCF-7 cells Cell skeleton proteins Cytoskeleton
PPM1D94 Malignancies Phosphatase; oncoprotein Protein serine/threonine phosphatase; regulation of cell cycle
JNK phosphatase26 Carcinogenesis Dual specificity protein phosphatase Cellular signaling
Iκ B95 Inflammation and carcinogenesis Protein kinase Regulation of NFκB pathway
Galectin-186,96 Ovary cells Pyruvate kinase Regulation of NFκB pathway
PKM291 MCF-7 cells Phosphoprotein Glycolysis
Hemoglobin97 Red blood cells Globin family Oxygen transport

Elimination of leukemia-initiating cells.

Leukemia-initiating cells (LICs) are pluripotent, self-renewing, phenotypically primitive and mitotically quiescent cells that have been identified in acute and chronic myeloid and lymphoid leukemia subtypes. Their noncycling status and inherent or acquired drug resistance mechanisms allow them to escape conventional and targeted therapies that effectively kill proliferating leukemia cells.98 In APL, PML-RARα is required, and even a minute amount of the oncoprotein allows LICs self-renewal in vivo.99 Zheng et al100 showed that in Sca1+/lin murine hematopoietic stem cells retrovirally transduced with PML-RARα and LICs from PML-RARα mice, ex vivo treatment with arsenic overcomes the aberrant stem cell capacity of PML-RARα+ LICs. Whereas transcriptional activation of PML-RARα on effect of ATRA probably controls differentiation, only the catabolism of the fusion protein triggers LIC eradication and long-term remission of mouse APL.99 ATRA induces differentiation of PLZF-RARα–driven mouse APL, but neither LIC clearance nor disease remission was achieved, explaining the clinical ATRA resistance of this rare APL subtype. Importantly, the ATRA/ATO combination rapidly clears PML-RARα+ LICs, resulting in APL eradication in murine models and patients.99 Because anthracyclines101 produce ROS, the ATRA and idarubicin regimen102 may induce PML-RARα degradation and hence promote LIC clearance, resulting in dramatically prolonged survival.102

Arsenic targeting of PML may also be important in the non-APL setting. Indeed, Ito et al103 showed that PML was required for hematopoietic stem cell maintenance, and in CML it appeared to be the factor that enabled LICs to maintain their quiescence—the inert state that prevented them from being destroyed by cancer therapies. Interestingly, ATO could reversibly decrease PML expression in LICs, suggesting that this agent may be of broader interest than previously thought. In gliomas, ATO seemed able to inhibit the Notch pathway and deplete the cancer stem-like cell population.104 ATO was shown to antagonize the Hedgehog pathway by preventing ciliary accumulation and reducing stability of the Gli2 transcriptional effector.105 ATO also represses NFκB106 and β-catenin,107 facilitating elimination of LICs (Figure 3).

Figure 3.

Figure 3

Arsenic targets critical pathways for the leukemia-initiating cells. Arsenic induces generation of ROS, perturbation of some signal pathways, and modulation of transcriptional factors. Arsenic also activates MEK1/ERK pathway, whereas combination of MEK1 inhibitor (MEKi, red filled circle) and arsenic results in synergistic antitumor effects.

Cell differentiation, apoptosis, and autophagy.

At the cellular level, ATO exerts dose-dependent dual effects on APL cells70 in that it induces apoptosis through activating the mitochondria-mediated intrinsic apoptotic pathway at high concentrations (1-2μM) and promotes cell differentiation at low concentrations (0.25-0.5μM; Figure 2C). The mechanisms of proapoptotic activity of ATO were scrutinized by many groups at gene/protein levels,38,108 and a large body of information has been gathered, including histone H3 phosphoacetylation at the CASP-10109; the involvement of JNK signaling110; anion exchanger 2111 and GSTP1-1112; the suppression of human telomerase reverse transcriptase (hTERT), C17, and C-MYC through Sp1 oxidation113; the repression of NFκB activation106; and the down-regulation of the WT1 gene.114 A pathway composed of ataxia telangiectasia mutated and Rad3-related, PML, CHK2, and p53 has been proposed to mediate ATO-induced apoptosis.115 ATO up-regulates a set of genes such as NADPH oxidase116 and 8-hydroxy-2′-deoxyguanosine (8-OHdG),117 and causes generation of ROS, which play a role as a mediator to induce apoptosis through release of cytochrome c to cytosol and activation of caspases. However, although all those mechanisms probably contribute to chronic or acute arsenic toxicity, it is unclear what is their contribution to arsenic response in APL, because they are unlikely to be specific for APL cells.

Recent studies further showed that in APL cells both ATRA and ATO induce autophagy by the mammalian target of rapamycin (mTOR)118 or MEK/ERK pathway,119 and autophagic degradation contributes significantly to proteolysis of PML-RARα. Inhibitors of autophagy or molecular targeting of BECN1 or ATG7 results in reversal of the suppressive effects of ATO on leukemic cells.119

Arsenic-based combinatory therapeutic regimens

ATO/ATRA combination.

Rationales.

Combination treatment regimens have been carefully designed to conquer APL.38 In a PML-RARα mouse model and a human NB4 APL cell line–based ascites/leukemia mouse model, ATRA/ATO combination dramatically prolongs survival or even eradicates the disease.120,121 Both ATO99,100 and ATRA99 induce LIC loss in PML-RARα mouse APL and the combination of the two agents results in synergistic clearance of LICs through cooperative PML-RARα degradation. Moreover, ATRA was shown to be able to increase the expression of the cell membrane arsenic transporter AQP9, which facilitated arsenic uptake.122

Although ATO alone induces partial differentiation,70 it synergizes with cAMP analog 8-CPT-cAMP in inducing full maturation of ATRA-sensitive and ATRA-resistance cells.123,124 However, activation of cAMP signaling was shown to enhance LIC loss by ATRA.99,125,126 Interestingly, ATRA could rapidly trigger a marked increase in the intracellular cAMP level and cAMP-dependent protein kinase (PKA) activity.127 Therefore, a cross talk may exist between ATO and ATRA signaling pathways through the cAMP/PKA node.

Moreover, ATRA potentiates ATO-induced RXRα phosphorylation and cooperates with ATO to induce apoptosis,128 and it was shown that ATRA induced degradation, whereas ATO antagonized catabolism of IκB.106 In the maturation-resistant NB4-R1 cells, ATRA exhibited antiproliferative properties through down-regulation of telomerase,129 and ATO enhanced this effect.130 When transcriptome/proteome approaches were used, Zheng et al131 reported that, although ATRA mainly caused transcriptional remodeling, ATO induced a deeper change of proteome pattern. ATRA/ATO combination amplified RA signaling, as highlighted by molecules involving IFN, calcium, cAMP/PKA, MAPK/JNK/p38, G-CSF, and TNF pathways. ATRA/ATO combination strongly activated the ubiquitin-proteasome pathway and significantly repressed genes/proteins promoting cell cycle or enhancing cell proliferation. Interestingly, the ATRA/ATO combination did not enhance the expression of stress response–related genes, including HSPA8, HSPCA, and AHSAI.131 Taken together, these results suggest that the ATRA/ATO combination may cause a synergy in therapeutic efficacy but not adverse effects.

Clinical efficacy.

A randomized clinical trial comparing ATRA/ATO combination and monotherapy was conducted in SIH from April 2001 to February 2003.38,45,132 Sixty-one APL cases were randomly assigned into 3 groups treated, respectively with ATRA, ATO, and the combination of the 2. It was reported in 2004 that the 3 groups achieved the same results in terms of CR rates (≥ 90%), although the combination therapy group needed the shortest time duration for remission induction. An obvious advantage of the combination therapy was that it generated much less minimal residual disease after consolidation than the 2 other therapeutic approaches as measured with real-time quantitative RT-PCR for PML-RARα. After a follow-up of 8-30 months all patients in the combination therapy group were in good clinical remission, whereas 7 of 37 cases in the 2 monotherapy groups relapsed (P < .05), showing the superiority of the combination therapy.45 Under this circumstance, the investigators from the SIH decided, from the ethical point of view, a termination of the randomized grouping and only the arm of combination therapy should be extended. In 2009, SIH reported the results of 85 patients administrated ATRA/ATO with a median follow-up of 70 months. Eighty patients (94.1%) entered CR.39 Kaplan-Meier estimates of the 5-year EFS and OS for all patients were 89.2% ± 3.4% and 91.7% ± 3.0%, respectively, and the 5-year relapse-free survival and OS for patients who achieved CR (n = 80) were 94.8% ± 2.5% and 97.4% ± 1.8%, respectively. On ATRA/ATO, prognosis was not influenced by initial white blood cell count, distinct PML-RARα types, or FLT3 mutations. The toxicity profile was mild and reversible (see “Adverse effects of arsenic”). The results were confirmed by recent long-term follow-up studies.133,134 Powell et al59 reported that of the 244 patients who received ATRA/chemotherapy as induction and ATRA/chemotherapy plus ATO as consolidation therapies, 195 cases (80%) achieved a 3-year EFS. Compared with the SIH trial that used ATRA/ATO/chemotherapy as induction therapy,39 the slightly lower EFS rate of this study might be because of the multicenter nature of the trial or could reflect the advantage of incorporating ATO into induction remedy for newly diagnosed APL. Taken together, ATRA/ATO/chemotherapy combinatory regimen transforms APL from a highly fatal to a highly curable disease.

Realgar-Indigo Naturalis Formula.

ATRA/As4S4 combination also showed enhanced therapeutic efficacy in APL.62 In traditional Chinese medicine, combination therapy containing multiple drugs with distinct but related mechanisms has been advocated for > 2500 years by prescriptions called formulas to amplify therapeutic efficacies of each agent and to minimize adverse effects.30,135 On the basis of traditional Chinese medicine theories, a patented Realgar-Indigo Naturalis Formula (RIF) was designed in the 1980s,63 in which a mined ore realgar was the principle element, whereas indigo naturalis, Salvia miltiorrhiza, and radix pseudostellariae were adjuvant components to assist the effects of realgar. Multicenter clinical trials showed that a CR rate of 96.7%136 to 98%63 and a 5-year OS rate of 86.88%137 were achieved in patients with APL receiving RIF, with moderate gastrointestinal discomfort and rash as the main adverse effects. Realgar in combination with indigo also exhibited an extent of anti-APL activity.138 The mechanisms of action of RIF were carefully dissected with the use of As4S4 (A), indirubin (I), and tanshinone IIA (T) as representatives of realgar, indigo naturalis, and Salvia miltiorrhiza, respectively,139 and it was shown that ATI combination yielded enhanced therapeutic efficacies against APL in murine model. ATI combination caused synergic effects and resulted in a much more profound differentiation of APL cells, potentiated ubiquitination and degradation of PML-RARα oncoprotein, stronger reprogramming of myeloid differentiation regulators, and enhanced G1/G0 arrest compared with cells treated with monoagents or biagents. Furthermore, T and I up-regulated AQP9 and facilitated transportation of arsenic into malignant promyelocytes, which in turn intensified arsenic-mediated PML-RARα degradation and therapeutic efficacies (Figure 4). These results open a new window for a better understanding of the therapeutic strategies of other traditional formulas.

Figure 4.

Figure 4

Mechanisms of action of representative components of RIF in treating APL. As, As4S4; Ind, indirubin; Tan, tanshinone IIA.

ATO in combination with MEK1 inhibition.

Studies have shown that activation of ERK1/2 as well as of the kinases immediately upstream of ERK, known as MAP/ERK kinases or MEKs can confer a drug-resistant phenotype to cancer cells. For example, rapamycin and its analogs activate the MAPK pathway in solid tumor,140 imatinib increases the activity of p42/44 MAPK in CML CD34+ cells that contributes to incomplete elimination of CML progenitors,141 and FLT3 inhibitor-resistant cells show continued activation of PI3K/AKT and/or RAS/MEK/MAPK signaling pathways.142 Accordingly, MAPK/MEK inhibitors may be helpful to overcome drug resistance in leukemic cells. Altman et al143 showed that in leukemia cells on ATO treatment, the AKT kinase is phosphorylated/activated to regulate downstream engagement of mTOR and its effectors. Targeted disruption of AKT1/AKT2 genes or inhibition of mTOR strongly enhances ATO's effects on leukemia cells.143 Treatment with ATO induces a MAPK-mediated PML phosphorylation144 that is associated with subsequent ubiquitination and proteasomal degradation.46 Lunghi et al145 reported that APL cells exploited the RAS-MAPK pathway to inactivate the proapoptotic protein BAD by phosphorylation at Ser112 and to delay ATO-induced apoptosis. MEK1 inhibitors suppressed ERK1/2, dephosphorylated BAD, and inhibited the ATO-induced increase of Bcl-xL, resulting in enhanced apoptosis and overcoming drug resistance.145 Combined use of ATO and MEK1 inhibitors leads to induction of the p53AIP1 (p53-regulated apoptosis-inducing protein 1) in NB4 and K562 cell lines146 and primary cells from patients with AML,137 and inhibition of tumor growth and elongation of survival in a human xenograft multiple myeloma model. These studies provide the framework for testing MEK1 inhibitor/ATO combination in patients with hematologic malignancies.148

Arsenic in treating CML

CML, a malignant myeloproliferative disease originated from pluripotential hematopoietic stem cells, is characterized by the Philadelphia chromosome formed by translocation t(9;22)(q34;q11) that generates a chimeric fusion protein BCR-ABL with constitutively activated tyrosine kinase activity.149 Imatinib mesylate (IM; or Gleevec, Glivec, or STI571), a rationally designed BCR-ABL inhibitor, has shown remarkable clinical efficacy that achieved an estimated 5-year OS of 89% in 553 patients with CML.150 However, IM151,152 and dasatinib152,153 do not deplete LICs, whereas a proportion of patients develop IM resistance,152,154156 and patients with advanced stage disease respond initially but then relapse.152,157 Moreover, cardiotoxicity of IM was also reported.158,159

Historically, ATO therapy was the first chemotherapeutic intervention for CML. Fowler solution was used to treat CML in the 19th century and became the mainstream therapeutic reagent for leukemia.160 In the 1930s, the efficacy of arsenic in the treatment of CML established it as a primary therapeutic agent for this disease.160 Until the advent of modern chemotherapy, arsenic and radiation were the mainstays of treatment for patients with CML. Arsenic was shown to be able to target PML and to eradicate quiescent LICs in CML.103 ATO inhibited translation of mRNA of BCR/ABL, resulting in attenuation of BCR/ABL levels and apoptosis of human leukemia cells.161 Zhang et al162 reported that arsenic targets BCR-ABL by ubiquitination of key lysine residues, leading to its proteasomal degradation. Recently, Mao et al83 showed that arsenic could directly bind c-CBL, the E3 ligase of BCR-ABL, by the conserved cysteines, including C381 at RING finger domain (Figure 5), resulting in inhibition of c-CBL's self-ubiquitination at K389 and subsequent proteasomal degradation. Consistent with these results, substitution of cysteine at 381 or lysine at 389 by alanine abrogated arsenic binding and c-CBL's self-ubiquitination, respectively. Consequently, elevated c-CBL promoted ubiquitination of BCR-ABL at K1517, leading to degradation of the aberrant kinase (Figure 5).

Figure 5.

Figure 5

Effects of arsenic on c-CBL and BCR-ABL in CML cells. (A) Without arsenic treatment, c-CBL is self-ubiquitinated and degraded in proteasome. (B) Arsenic treatment inhibits c-CBL self-ubiquitination and proteasomal degradation, and triggers ubiquitination of BCR-ABL at K1517 followed by degradation in the proteasome.

Arsenic exerts synergistic effects with IM in inducing apoptosis of CML cells and in prolonging survival of mice inoculated with CML cells.162164 It was shown that arsenic and IM induce cell cycle arrest at G2/M and G1 phases, respectively. Arsenic and IM synergistically activate the endogenous and exogenous endoplasmic reticulum (ER) stress, leading to enhanced cell apoptosis.162,163,165 These discoveries provide rationales for a clinical trial to test the arsenic/IM combination therapy in CML.

Arsenic in treating other malignancies

Arsenic has been used in treating multiple myeloma, myelodysplasia syndrome, and lymphoid malignancies, including non-Hodgkin lymphoma, and has displayed beneficial effects in some cases (Table 3). In adult T-cell leukemia/lymphoma (ATL)–derived cells, ATO reportedly synergized with IFNα to induce cell-cycle arrest and apoptosis176 through down-regulation of the HTLV-1 oncoprotein Tax and inactivation of NF-κB.177,178 Clinically, arsenic/IFN therapy exhibited some efficacy in 7 patients with refractory aggressive ATL,170 whereas in 10 cases of newly diagnosed chronic ATL arsenic/IFN/zidovudine combination showed an impressive 100% response rate.171 Recent animal studies in Lck-Tax transgenics that develop an ATL-like disease have recapitulated the therapeutic action of the arsenic/IFNα association, strongly suggesting that the latter is actually targeting Tax for degradation.179 Moreover, transplantation studies have shown that Tax degradation is accompanied by loss of leukemia-initiating activity, but not short-term growth, providing a striking parallel with APL and suggesting that arsenic may promote catabolism of specific classes of oncoproteins.

Table 3.

Clinical studies of arsenic in treating other malignancies

Disease Year Authors No. Regimen Response
MM 2006 Berenson et al166 65 ATO + AA + melphan 2 CR; 15 PR; 14 MR
2006 Abou-Jawde et al167 20 ATO + AA + dexamethasone 6 PR
2006 Wu et al168 20 ATO + AA + dexamethasone 2 PR; 6 MR
2007 Berenson et al169 22 ATO + AA + bortezomib 2 PR; 4 MR
ATL 2004 Hermine et al170 7 (Relapsed/refractory) ATO + IFN 1 CR; 3 PR
2009 Kchour et al171 10 (Newly diagnosed) ATO + IFN + zidovudine 9 CR; 1 PR
MDS 2006 Schiller et al172 76 ATO 1 CR; 13 HI
2006 Vey et al173 115 ATO 1 CR; 1 PR; 22 HI
2008 Zheng et al174 21 ATO + RA + thalidomide 1 CR; 1 PR; 3 HI
Lymphoid malignancies 2009 Chang et al175 16 ATO + AA 1 response

MM indicates multiple myeloma; AA, ascorbic acid; CR, complete response; PR, partial response; MR, minor response; ATL, adult T-cell leukemia/lymphoma; MDS, myelodysplasia syndrome; HI, hematologic improvement; and RA, retinoic acid.

There are 111 recently completed or ongoing clinical trials listed on www.clinicaltrials.gov that evaluate ATO alone or in combination with other agents for treatment of cancers, excluding APL. ATO is under investigation as treatment for a variety of solid tumors, including lung cancer, hepatocellular carcinoma, and colorectal cancer. Limited clinical activity as a single agent has been reported in a small number of patients with hepatocellular carcinoma, melanoma, and renal cell carcinoma; ATO in combination with chemotherapy has shown promising activity in osteosarcoma and Ewing sarcoma180 (and references in this review article).

Adverse effects of arsenic

Although arsenic seems to be synonymous with poison, nearly all recent clinical trial results suggest that arsenic at therapeutic concentrations is generally well tolerated. No bone marrow depression and chemotherapy-associated secondary malignancy were observed with arsenic treatment.52,54

Sudden death was recorded in one study,181 and severe liver impairment was documented.53 These toxicities might be because of a genetic basis with exceptional susceptibilities to arsenic toxicity in rare patients, exposure to anthracyclines or other cardiotoxic agents before ATO therapy, and abnormal electrolyte levels, or other unidentified factors.53,57,181,182 Hyperleukocytosis, a retinoic acid syndrome (or differentiation syndrome)–like clinical entity was also reported and was shown to be driven by chemokine production induced by ATO or ATRA as a single agent or in combination.183 In long-term studies,39,57,58,182 the toxicity profile of arsenic was mild, and 24 months after the last dose of ATRA/ATO patients had urine arsenic concentrations well below the safety limit. Mathews et al57 reported that despite counseling against pregnancy after therapy, in view of the absence of data on effect of prior ATO therapy and teratogenicity, 7 patients (4 women and 3 men) have had 8 normal babies.

Perspectives

As a traditional poison, inappropriate use of arsenic may kill people; as one of the oldest drugs in the world, its appropriate application cures some cancer types and saves lives. These facts clearly suggest that when considering how to control an emerging “bad factor,” one might try to find out its other side and the safe translation.

Arsenic is the most potent single agent against APL. The revival of arsenic by its application in treating APL is a unique story in cancer research. It also highlights some of the essential concepts in pharmacology, such as the key importance of the therapeutic ratio between normal cells and the target. It illustrates the power of combinations. Indeed, in APL, ATO/ATRA combination has exhibited drastically enhanced therapeutic efficacy compared with either single agent, transforming the fate of an otherwise highly fatal disease. In treating other types of malignancies, including solid tumors, rational combinatory regimens could be designed to improve clinical outcome. For example, because arsenic binds and activates c-CBL that controls signaling of EGFR, the therapeutic efficacy of ATO in combination with EGFR inhibitor could be tested in non–small cell lung cancer and other related human malignancies.

Acknowledgments

We apologize to our many colleagues whose work could not be cited because of space restrictions. We thank Prof Zhen-Yi Wang at SIH for his long term support, Dr Laurent Degos from Hospital Saint Louis in Paris, and Dr Samuel Waxman from Mount Sinai Medical Center in New York for friendly long-term collaboration.

This work was supported in part by the Chinese National Key Program for Basic Research (973; 2010CB529200) and National High Tech Program (863), National Natural Science Foundation of China, Shanghai Municipal Commission for Science and Technology, Shanghai Municipal Commission for Education, and Samuel Waxman Cancer Research Foundation.

Authorship

Contribution: S.-J.C., G.-B.Z., X.-W.Z., J.-H.M., H.d.T., and Z.C. all contributed to the writing of this manuscript.

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

Correspondence: Sai-Juan Chen, Shanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Rd II, Shanghai, 200025, China; e-mail: sjchen@stn.sh.cn; or Zhu Chen, Shanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Rd II, Shanghai, 200025, China; e-mail: zchen@stn.sh.cn.

References

  • 1.National Research Council (NRC) Arsenic. Washington, DC: The National Academies Press; 1977. [Google Scholar]
  • 2.Wolfe-Simon F, Blum JS, Kulp TR, et al. A bacterium that can grow by using arsenic instead of phosphorus [published online ahead of print December 2, 2010]. Science. doi: 10.1126/science.1197258. [Google Scholar]
  • 3.Hillestad LK. Acute promyelocytic leukemia. Acta Med Scand. 1957;159(3):189–194. [PubMed] [Google Scholar]
  • 4.Kumagai Y, Sumi D. Arsenic: signal transduction, transcription factor, and biotransformation involved in cellular response and toxicity. Annu Rev Pharmacol Toxicol. 2007;47(1):243–262. doi: 10.1146/annurev.pharmtox.47.120505.105144. [DOI] [PubMed] [Google Scholar]
  • 5.ISSI Consulting Group. Arsenic Occurrence in Public Drinking Water Supplies. Washington, DC: U.S. Environmental Protection Agency; 2000. EPA publication no. 815-R-00-023. [Google Scholar]
  • 6.Zhu J, Chen Z, Lallemand-Breitenbach V, de The H. How acute promyelocytic leukaemia revived arsenic. Nat Rev Cancer. 2002;2(9):705–713. doi: 10.1038/nrc887. [DOI] [PubMed] [Google Scholar]
  • 7.Sharma VK, Sohn M. Aquatic arsenic: toxicity, speciation, transformations, and remediation. Environ Int. 2009;35(4):743–759. doi: 10.1016/j.envint.2009.01.005. [DOI] [PubMed] [Google Scholar]
  • 8.Wang JP, Qi L, Moore MR, Ng JC. A review of animal models for the study of arsenic carcinogenesis. Toxicol Lett. 2002;133(1):17–31. doi: 10.1016/s0378-4274(02)00086-3. [DOI] [PubMed] [Google Scholar]
  • 9.Ng JC. Environmental contamination of arsenic and its toxicological impact on humans. Environ Chem. 2005;2(3):146–160. [Google Scholar]
  • 10.Mann KK, Wallner B, Lossos IS, Miller WH., Jr Darinaparsin: a novel organic arsenical with promising anticancer activity. Expert Opin Investig Drugs. 2009;18(11):1727–1734. doi: 10.1517/13543780903282759. [DOI] [PubMed] [Google Scholar]
  • 11.Lin S, Shi Q, Nix FB, et al. A novel S-adenosyl-L-methionine:arsenic(III) methyltransferase from rat liver cytosol. J Biol Chem. 2002;277(13):10795–10803. doi: 10.1074/jbc.M110246200. [DOI] [PubMed] [Google Scholar]
  • 12.Wood TC, Salavagionne OE, Mukherjee B, et al. Human arsenic methyltransferase (AS3MT) pharmacogenetics: gene resequencing and functional genomics studies. J Biol Chem. 2006;281(11):7364–7373. doi: 10.1074/jbc.M512227200. [DOI] [PubMed] [Google Scholar]
  • 13.Del Razo LM, Styblo M, Cullen WR, Thomas DJ. Determination of trivalent methylated arsenicals in biological matrices. Toxicol Appl Pharmacol. 2001;174(3):282–293. doi: 10.1006/taap.2001.9226. [DOI] [PubMed] [Google Scholar]
  • 14.Kitchin KT. Recent advances in arsenic carcinogenesis: modes of action, animal model systems, and methylated arsenic metabolites. Toxicol Appl Pharmacol. 2001;172(3):249–261. doi: 10.1006/taap.2001.9157. [DOI] [PubMed] [Google Scholar]
  • 15.Chen GQ, Zhou L, Styblo M, et al. Methylated metabolites of arsenic trioxide are more potent than arsenic trioxide as apoptotic but not differentiation inducers in leukemia and lymphoma cells. Cancer Res. 2003;63(8):1853–1859. [PubMed] [Google Scholar]
  • 16.Styblo M, Del Razo LM, Vega L, et al. Comparative toxicity of trivalent and pentavalent inorganic and methylated arsenicals in rat and human cells. Arch Toxicol. 2000;74(6):289–299. doi: 10.1007/s002040000134. [DOI] [PubMed] [Google Scholar]
  • 17.Smith AH, Hopenhayn-Rich C, Bates MN, et al. Cancer risks from arsenic in drinking water. Environ Health Perspect. 1992;97:259–267. doi: 10.1289/ehp.9297259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hughes MF. Arsenic toxicity and potential mechanisms of action. Toxicol Lett. 2002;133(1):1–16. doi: 10.1016/s0378-4274(02)00084-x. [DOI] [PubMed] [Google Scholar]
  • 19.Vallee BL, Ulmer DD, Wacker WE. Arsenic toxicology and biochemistry. A M A Arch Ind Health. 1960;21:132–151. [Google Scholar]
  • 20.The World Bank. Environment and Social Unit-South Asia Region. Towards a More Effective Operational Response. Arsenic Contamination of Groundwater in South and East Asian Countries. Washington, DC: The World Bank; 2005. Vol II technical report. [Google Scholar]
  • 21.Aposhian HV, Aposhian MM. Arsenic toxicology: five questions. Chem Res Toxicol. 2006;19(1):1–15. doi: 10.1021/tx050106d. [DOI] [PubMed] [Google Scholar]
  • 22.Teixeira MC, Ciminelli VST, Dantas MSS, Diniz SF, Duarte HA. Raman spectroscopy and DFT calculations of As(III) complexation with a cysteine-rich biomaterial. J Colloid Interface Sci. 2007;315(1):128–134. doi: 10.1016/j.jcis.2007.06.041. [DOI] [PubMed] [Google Scholar]
  • 23.Schuliga M, Chouchane S, Snow ET. Upregulation of glutathione-related genes and enzyme activities in cultured human cells by sublethal concentrations of inorganic arsenic. Toxicol Sci. 2002;70(2):183–192. doi: 10.1093/toxsci/70.2.183. [DOI] [PubMed] [Google Scholar]
  • 24.Zhou X, Sun H, Ellen TP, Chen H, Costa M. Arsenite alters global histone H3 methylation. Carcinogenesis. 2008;29(9):1831–1836. doi: 10.1093/carcin/bgn063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Andrew AS, Mason RA, Memoli V, Duell EJ. Arsenic activates EGFR pathway signaling in the lung. Toxicol Sci. 2009;109(2):350–357. doi: 10.1093/toxsci/kfp015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cavigelli M, Li WW, Lin A, et al. The tumor promoter arsenite stimulates AP-1 activity by inhibiting a JNK phosphatase. EMBO J. 1996;15(22):6269–6279. [PMC free article] [PubMed] [Google Scholar]
  • 27.He X, Ma Q. Critical cysteine residues of Kelch-like ECH-associated protein 1 in arsenic sensing and suppression of nuclear factor erythroid 2-related factor 2. J Pharmacol Exp Ther. 2010;332(1):66–75. doi: 10.1124/jpet.109.160465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Pi J, Qu W, Reece JM, Kumagai Y, Waalkes MP. Transcription factor Nrf2 activation by inorganic arsenic in cultured keratinocytes: involvement of hydrogen peroxide. Exp Cell Res. 2003;290(2):234–245. doi: 10.1016/s0014-4827(03)00341-0. [DOI] [PubMed] [Google Scholar]
  • 29.Hughes MF. Biomarkers of exposure: a case study with inorganic arsenic. Environ Health Perspect. 2006;114(11):1790–1796. doi: 10.1289/ehp.9058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Beijing, China: Chinese Medical Ancient Books Publishing House; 2003. The Inner Canon of Emperor Huang (Originally published in the Spring and Autumn and the Warring States Periods of China, 722 BC-221 BC). [Google Scholar]
  • 31.Huang B, Wang Y. Thousand Formulas and Thousand Herbs of Traditional Chinese Medicine. Harbin, China: Heilongjiang Education Press; 1993. [Google Scholar]
  • 32.Li SZ. The Compendium of Materia Medica (Originally published in the Ming Dynasty of China, 1578) Beijing, China: People's Medical Publishing House; 1982. [Google Scholar]
  • 33.Jolliffe DM. A history of the use of arsenicals in man. J R Soc Med. 1993;86(5):287–289. doi: 10.1177/014107689308600515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sears DA. History of the treatment of chronic myelocytic leukemia. Am J Med Sci. 1988;296(2):85–86. doi: 10.1097/00000441-198808000-00001. [DOI] [PubMed] [Google Scholar]
  • 35.Zhou G, Zhang J, Wang Z, Chen S, Chen Z. Treatment of acute promyelocytic leukaemia with all-trans retinoic acid and arsenic trioxide: a paradigm of synergistic molecular targeting therapy. Phil Trans R Soc B. 2007;362(1482):959–971. doi: 10.1098/rstb.2007.2026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Degos L. The history of acute promyelocytic leukaemia. Br J Haematol. 2003;122(4):539–553. doi: 10.1046/j.1365-2141.2003.04460.x. [DOI] [PubMed] [Google Scholar]
  • 37.Zhou GB, Zhao WL, Wang ZY, Chen SJ, Chen Z. Retinoic acid and arsenic for treating acute promyelocytic leukemia. PLoS Med. 2005;2(1):33–38. doi: 10.1371/journal.pmed.0020012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wang ZY, Chen Z. Acute promyelocytic leukemia: from highly fatal to highly curable. Blood. 2008;111(5):2505–2515. doi: 10.1182/blood-2007-07-102798. [DOI] [PubMed] [Google Scholar]
  • 39.Hu J, Liu YF, Wu CF, et al. Long-term efficacy and safety of all-trans retinoic acid/arsenic trioxide-based therapy in newly diagnosed acute promyelocytic leukemia. Proc Natl Acad Sci U S A. 2009;106(9):3342–3347. doi: 10.1073/pnas.0813280106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Bernard J, Weil M, Boiron M, et al. Acute promyelocytic leukemia: results of treatment by daunorubicin. Blood. 1973;41(4):489–496. [PubMed] [Google Scholar]
  • 41.Huang ME, Ye YC, Chen SR, et al. Use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia. Blood. 1988;72(2):567–572. doi: 10.1182/blood-2016-11-750182. [DOI] [PubMed] [Google Scholar]
  • 42.Ades L, Guerci A, Raffoux E, et al. Very long-term outcome of acute promyelocytic leukemia after treatment with all-trans retinoic acid and chemotherapy: the European APL Group experience. Blood. 2010;115(9):1690–1696. doi: 10.1182/blood-2009-07-233387. [DOI] [PubMed] [Google Scholar]
  • 43.Creutzig U, Zimmermann M, Dworzak M, et al. Favourable outcome of patients with childhood acute promyelocytic leukaemia after treatment with reduced cumulative anthracycline doses. Br J Haematol. 2010;149(3):399–409. doi: 10.1111/j.1365-2141.2010.08107.x. [DOI] [PubMed] [Google Scholar]
  • 44.Wang ZY. Ham-Wasserman lecture: treatment of acute leukemia by inducing differentiation and apoptosis. Hematology Am Soc Hematol Educ Program. 2003:1–13. doi: 10.1182/asheducation-2003.1.1. [DOI] [PubMed] [Google Scholar]
  • 45.Shen ZX, Shi ZZ, Fang J, et al. All-trans retinoic acid/As2O3 combination yields a high quality remission and survival in newly diagnosed acute promyelocytic leukemia. Proc Natl Acad Sci U S A. 2004;101(15):5328–5335. doi: 10.1073/pnas.0400053101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Jeanne M, Lallemand-Breitenbach V, Ferhi O, et al. PML/RARA oxidation and arsenic binding initiate the antileukemia response of As2O3. Cancer Cell. 2010;18(1):88–98. doi: 10.1016/j.ccr.2010.06.003. [DOI] [PubMed] [Google Scholar]
  • 47.Rego EM, He LZ, Warrell RP, Jr, Wang ZG, Pandolfi PP. Retinoic acid (RA) and As2O3 treatment in transgenic models of acute promyelocytic leukemia (APL) unravel the distinct nature of the leukemogenic process induced by the PML-RARalpha and PLZF-RARalpha oncoproteins. Proc Natl Acad Sci U S A. 2000;97(18):10173–10178. doi: 10.1073/pnas.180290497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Koken MH, Daniel MT, Gianni M, et al. Retinoic acid, but not arsenic trioxide, degrades the PLZF/RARalpha fusion protein, without inducing terminal differentiation or apoptosis, in a RA-therapy resistant t(11;17)(q23;q21) APL patient. Oncogene. 1999;18(4):1113–1118. doi: 10.1038/sj.onc.1202414. [DOI] [PubMed] [Google Scholar]
  • 49.Licht JD, Chomienne C, Goy A, et al. Clinical and molecular characterization of a rare syndrome of acute promyelocytic leukemia associated with translocation (11;17). Blood. 1995;85(4):1083–1094. [PubMed] [Google Scholar]
  • 50.Chen Z, Brand NJ, Chen A, et al. Fusion between a novel Kruppel-like zinc finger gene and the retinoic acid receptor-alpha locus due to a variant t(11;17) translocation associated with acute promyelocytic leukaemia. EMBO J. 1993;12(3):1161–1167. doi: 10.1002/j.1460-2075.1993.tb05757.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.de The H, Chen Z. Acute promyelocytic leukaemia: novel insights into the mechanisms of cure. Nat Rev Cancer. 2010;10(11):775–783. doi: 10.1038/nrc2943. [DOI] [PubMed] [Google Scholar]
  • 52.Sun HD, Ma L, Hu XC, Zhang TD. Ai-Lin I treated 32 cases of acute promyelocytic leukemia. Chin J Integrat Chin West Med. 1992;12:170–171. [Google Scholar]
  • 53.Niu C, Yan H, Yu T, et al. Studies on treatment of acute promyelocytic leukemia with arsenic trioxide: remission induction, follow-up, and molecular monitoring in 11 newly diagnosed and 47 relapsed acute promyelocytic leukemia patients. Blood. 1999;94(10):3315–3324. [PubMed] [Google Scholar]
  • 54.Shen ZX, Chen GQ, Ni JH, et al. Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL), II: clinical efficacy and pharmacokinetics in relapsed patients. Blood. 1997;89(9):3354–3360. [PubMed] [Google Scholar]
  • 55.Shigeno K, Naito K, Sahara N, et al. Arsenic trioxide therapy in relapsed or refractory Japanese patients with acute promyelocytic leukemia: updated outcomes of the phase II study and postremission therapies. Int J Hematol. 2005;82(3):224–229. doi: 10.1532/IJH97.05044. [DOI] [PubMed] [Google Scholar]
  • 56.Soignet SL, Frankel SR, Douer D, et al. United States multicenter study of arsenic trioxide in relapsed acute promyelocytic leukemia. J Clin Oncol. 2001;19(18):3852–3860. doi: 10.1200/JCO.2001.19.18.3852. [DOI] [PubMed] [Google Scholar]
  • 57.Mathews V, George B, Chendamarai E, et al. Single-agent arsenic trioxide in the treatment of newly diagnosed acute promyelocytic leukemia: long-term follow-up data. J Clin Oncol. 2010;28(24):3866–3871. doi: 10.1200/JCO.2010.28.5031. [DOI] [PubMed] [Google Scholar]
  • 58.Zhou J, Zhang Y, Li J, et al. Single-agent arsenic trioxide in the treatment of children with newly diagnosed acute promyelocytic leukemia. Blood. 2010;115(9):1697–1702. doi: 10.1182/blood-2009-07-230805. [DOI] [PubMed] [Google Scholar]
  • 59.Powell BL, Moser B, Stock W, et al. Arsenic trioxide improves event-free and over-all survival for adults with acute promyelocytic leukemia: North American Leukemia Intergroup Study C9710. Blood. 2010;116(19):3751–3757. doi: 10.1182/blood-2010-02-269621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Dai CW, Zhang GS, Shen JK, et al. Use of all-trans retinoic acid in combination with arsenic trioxide for remission induction in patients with newly diagnosed acute promyelocytic leukemia and for consolidation/maintenance in CR patients. Acta Haematol. 2009;121(1):1–8. doi: 10.1159/000204472. [DOI] [PubMed] [Google Scholar]
  • 61.Ghavamzadeh A, Alimoghaddam K, Ghaffari SH, et al. Treatment of acute promyelocytic leukemia with arsenic trioxide without ATRA and/or chemotherapy. Ann Oncol. 2006;17(1):131–134. doi: 10.1093/annonc/mdj019. [DOI] [PubMed] [Google Scholar]
  • 62.Wu T, Zhao J, Jiang B, et al. Tetra-arsenic tetra-sulfide containing triple-agent regimen as the first line therapy for acute promyelocytic leukemia: expeditiously consecutive complete remission and improved disease-free survival [abstract]. Blood. 2007;110(11) Abstract 591. [Google Scholar]
  • 63.Huang SL, Guo AX, Xiang Y, et al. Clinical study on the treatment of acute promyelocytic leukemia with Composite Indigo Naturalis tablets. Chin J Hematol. 1995;16:26–28. [Google Scholar]
  • 64.Lu DP, Qiu JY, Jiang B, et al. Tetra-arsenic tetra-sulfide for the treatment of acute promyelocytic leukemia: a pilot report. Blood. 2002;99(9):3136–3143. doi: 10.1182/blood.v99.9.3136. [DOI] [PubMed] [Google Scholar]
  • 65.Wang K, Wang P, Shi J, et al. PML/RARalpha targets promoter regions containing PU.1 consensus and RARE half sites in acute promyelocytic leukemia. Cancer Cell. 2010;17(2):186–197. doi: 10.1016/j.ccr.2009.12.045. [DOI] [PubMed] [Google Scholar]
  • 66.Melnick A, Licht JD. Deconstructing a disease: RARalpha, its fusion partners, and their roles in the pathogenesis of acute promyelocytic leukemia. Blood. 1999;93(10):3167–3215. [PubMed] [Google Scholar]
  • 67.Kamashev D, Vitoux D, de The H. PML-RARA-RXR oligomers mediate retinoid and rexinoid/cAMP cross-talk in acute promyelocytic leukemia cell differentiation. J Exp Med. 2004;199(8):1163–1174. doi: 10.1084/jem.20032226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zhu J, Koken MH, Quignon F, et al. Arsenic-induced PML targeting onto nuclear bodies: implications for the treatment of acute promyelocytic leukemia. Proc Natl Acad Sci U S A. 1997;94(8):3978–3983. doi: 10.1073/pnas.94.8.3978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Chen GQ, Zhu J, Shi XG, et al. In vitro studies on cellular and molecular mechanisms of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia: As2O3 induces NB4 cell apoptosis with downregulation of Bcl-2 expression and modulation of PML-RAR alpha/PML proteins. Blood. 1996;88(3):1052–1061. [PubMed] [Google Scholar]
  • 70.Chen GQ, Shi XG, Tang W, et al. Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL), I: As2O3 exerts dose-dependent dual effects on APL cells. Blood. 1997;89(9):3345–3353. [PubMed] [Google Scholar]
  • 71.Borden KL, Boddy MN, Lally J, et al. The solution structure of the RING finger domain from the acute promyelocytic leukaemia proto-oncoprotein PML. EMBO J. 1995;14(7):1532–1541. doi: 10.1002/j.1460-2075.1995.tb07139.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Borden KL, Lally JM, Martin SR, et al. In vivo and in vitro characterization of the B1 and B2 zinc-binding domains from the acute promyelocytic leukemia protooncoprotein PML. Proc Natl Acad Sci U S A. 1996;93(4):1601–1606. doi: 10.1073/pnas.93.4.1601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Butler JT, Hall LL, Smith KP, Lawrence JB. Changing nuclear landscape and unique PML structures during early epigenetic transitions of human embryonic stem cells. J Cell Biochem. 2009;107(4):609–621. doi: 10.1002/jcb.22183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lallemand-Breitenbach V, de The H. PML nuclear bodies. Cold Spring Harb Perspect Biol. 2010;2(5):a000661. doi: 10.1101/cshperspect.a000661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Luciani JJ, Depetris D, Usson Y, et al. PML nuclear bodies are highly organised DNA-protein structures with a function in heterochromatin remodelling at the G2 phase. J Cell Sci. 2006;119(Pt 12):2518–2531. doi: 10.1242/jcs.02965. [DOI] [PubMed] [Google Scholar]
  • 76.Daniel MT, Koken M, Romagne O, et al. PML protein expression in hematopoietic and acute promyelocytic leukemia cells. Blood. 1993;82(6):1858–1867. [PubMed] [Google Scholar]
  • 77.Lallemand-Breitenbach V, Jeanne M, Benhenda S, et al. Arsenic degrades PML or PML-RARalpha through a SUMO-triggered RNF4/ubiquitin-mediated pathway. Nat Cell Biol. 2008;10(5):547–555. doi: 10.1038/ncb1717. [DOI] [PubMed] [Google Scholar]
  • 78.Lallemand-Breitenbach V, Zhu J, Puvion F, et al. Role of promyelocytic leukemia (PML) sumolation in nuclear body formation, 11S proteasome recruitment, and As2O3-induced PML or PML/retinoic acid receptor alpha degradation. J Exp Med. 2001;193(12):1361–1371. doi: 10.1084/jem.193.12.1361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zhang XW, Yan XJ, Zhou ZR, et al. Arsenic trioxide controls the fate of the PML-RAR{alpha} oncoprotein by directly binding PML. Science. 2010;328(5975):240–243. doi: 10.1126/science.1183424. [DOI] [PubMed] [Google Scholar]
  • 80.Zhu J, Zhou J, Peres L, et al. A sumoylation site in PML/RARA is essential for leukemic transformation. Cancer Cell. 2005;7(2):143–153. doi: 10.1016/j.ccr.2005.01.005. [DOI] [PubMed] [Google Scholar]
  • 81.Chen Z, Zhao WL, Shen ZX, et al. Arsenic trioxide and acute promyelocytic leukemia: clinical and biological. Curr Top Microbiol Immunol. 2007;313:129–144. doi: 10.1007/978-3-540-34594-7_8. [DOI] [PubMed] [Google Scholar]
  • 82.Tatham MH, Geoffroy MC, Shen L, et al. RNF4 is a poly-SUMO-specific E3 ubiquitin ligase required for arsenic-induced PML degradation. Nat Cell Biol. 2008;10(5):538–546. doi: 10.1038/ncb1716. [DOI] [PubMed] [Google Scholar]
  • 83.Mao JH, Sun XY, Liu JX, et al. As4S4 targets RING-type E3 ligase c-CBL to induce degradation of BCR-ABL in chronic myelogenous leukemia. Proc Natl Acad Sci U S A. 2010;107(50):21683–21688. doi: 10.1073/pnas.1016311108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Lu J, Chew EH, Holmgren A. Targeting thioredoxin reductase is a basis for cancer therapy by arsenic trioxide. Proc Natl Acad Sci U S A. 2007;104(30):12288–12293. doi: 10.1073/pnas.0701549104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Rodriguez VM, Del Razo LM, Limon-Pacheco JH, et al. Glutathione reductase inhibition and methylated arsenic distribution in Cd1 mice brain and liver. Toxicol Sci. 2005;84(1):157–166. doi: 10.1093/toxsci/kfi057. [DOI] [PubMed] [Google Scholar]
  • 86.Chang KN, Lee TC, Tam MF, et al. Identification of galectin I and thioredoxin peroxidase II as two arsenic-binding proteins in Chinese hamster ovary cells. Biochem J. 2003;371(Pt 2):495–503. doi: 10.1042/BJ20021354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Donoghue N, Yam PT, Jiang XM, Hogg PJ. Presence of closely spaced protein thiols on the surface of mammalian cells. Protein Sci. 2000;9(12):2436–2445. doi: 10.1110/ps.9.12.2436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ngu TT, Stillman MJ. Arsenic binding to human metallothionein. J Am Chem Soc. 2006;128(38):12473–12483. doi: 10.1021/ja062914c. [DOI] [PubMed] [Google Scholar]
  • 89.He X, Ma Q. Induction of metallothionein I by arsenic via metal-activated transcription factor 1: critical role of C-terminal cysteine residues in arsenic sensing. J Biol Chem. 2009;284(19):12609–12621. doi: 10.1074/jbc.M901204200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Li YM, Broome JD. Arsenic targets tubulins to induce apoptosis in myeloid leukemia cells. Cancer Res. 1999;59(4):776–780. [PubMed] [Google Scholar]
  • 91.Zhang X, Yang F, Shim JY, et al. Identification of arsenic-binding proteins in human breast cancer cells. Cancer Lett. 2007;255(1):95–106. doi: 10.1016/j.canlet.2007.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Menzel DB, Hamadeh HK, Lee E, et al. Arsenic binding proteins from human lymphoblastoid cells. Toxicol Lett. 1999;105(2):89–101. doi: 10.1016/s0378-4274(98)00380-4. [DOI] [PubMed] [Google Scholar]
  • 93.Izdebska M, Grzanka A, Ostrowski M, Zuryn A, Grzanka D. Effect of arsenic trioxide (Trisenox) on actin organization in K-562 erythroleukemia cells. Folia Histochem Cytobiol. 2009;47(3):453–459. doi: 10.2478/v10042-009-0080-5. [DOI] [PubMed] [Google Scholar]
  • 94.Yoda A, Toyoshima K, Watanabe Y, et al. Arsenic trioxide augments Chk2/p53-mediated apoptosis by inhibiting oncogenic Wip1 phosphatase. J Biol Chem. 2008;283(27):18969–18979. doi: 10.1074/jbc.M800560200. [DOI] [PubMed] [Google Scholar]
  • 95.Kapahi P, Takahashi T, Natoli G, et al. Inhibition of NF-kappa B activation by arsenite through reaction with a critical cysteine in the activation loop of Ikappa B kinase. J Biol Chem. 2000;275(46):36062–36066. doi: 10.1074/jbc.M007204200. [DOI] [PubMed] [Google Scholar]
  • 96.Lin CH, Huang CF, Chen WY, et al. Characterization of the interaction of galectin-1 with sodium arsenite. Chem Res Toxicol. 2006;19(3):469–474. doi: 10.1021/tx0503348. [DOI] [PubMed] [Google Scholar]
  • 97.Lu M, Wang H, Li XF, et al. Binding of dimethylarsinous acid to cys-13alpha of rat hemoglobin is responsible for the retention of arsenic in rat blood. Chem Res Toxicol. 2007;20(1):27–37. doi: 10.1021/tx060195+. [DOI] [PubMed] [Google Scholar]
  • 98.Huntly BJ, Gilliland DG. Leukaemia stem cells and the evolution of cancer-stem-cell research. Nat Rev Cancer. 2005;5(4):311–321. doi: 10.1038/nrc1592. [DOI] [PubMed] [Google Scholar]
  • 99.Nasr R, Guillemin MC, Ferhi O, et al. Eradication of acute promyelocytic leukemia-initiating cells through PML-RARA degradation. Nat Med. 2008;14(12):1333–1342. doi: 10.1038/nm.1891. [DOI] [PubMed] [Google Scholar]
  • 100.Zheng X, Seshire A, Ruster B, et al. Arsenic but not all-trans retinoic acid overcomes the aberrant stem cell capacity of PML/RAR{alpha}-positive leukemic stem cells. Haematologica. 2007;92(3):323–331. doi: 10.3324/haematol.10541. [DOI] [PubMed] [Google Scholar]
  • 101.Berthiaume JM, Wallace KB. Adriamycin-induced oxidative mitochondrial cardiotoxicity. Cell Biol Toxicol. 2007;23(1):15–25. doi: 10.1007/s10565-006-0140-y. [DOI] [PubMed] [Google Scholar]
  • 102.Lo-Coco F, Avvisati G, Vignetti M, et al. Front-line treatment of acute promyelocytic leukemia with AIDA induction followed by risk-adapted consolidation for adults younger than 61 years: results of the AIDA-2000 trial of the GIMEMA Group. Blood. 2010;116(17):3171–3179. doi: 10.1182/blood-2010-03-276196. [DOI] [PubMed] [Google Scholar]
  • 103.Ito K, Bernardi R, Morotti A, et al. PML targeting eradicates quiescent leukaemia-initiating cells. Nature. 2008;453(7198):1072–1078. doi: 10.1038/nature07016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Zhen Y, Zhao S, Li Q, Li Y, Kawamoto K. Arsenic trioxide-mediated Notch pathway inhibition depletes the cancer stem-like cell population in gliomas. Cancer Lett. 2010;292(1):64–72. doi: 10.1016/j.canlet.2009.11.005. [DOI] [PubMed] [Google Scholar]
  • 105.Kim J, Lee JJ, Kim J, Gardner D, Beachy PA. Arsenic antagonizes the Hedgehog pathway by preventing ciliary accumulation and reducing stability of the Gli2 transcriptional effector. Proc Natl Acad Sci U S A. 2010;107(30):13432–13437. doi: 10.1073/pnas.1006822107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Mathieu J, Besancon F. Arsenic trioxide represses NF-kappaB activation and increases apoptosis in ATRA-treated APL cells. Ann N Y Acad Sci. 2006;1090:203–208. doi: 10.1196/annals.1378.022. [DOI] [PubMed] [Google Scholar]
  • 107.Yu J, Qian H, Li Y, et al. Arsenic trioxide (As2O3) reduces the invasive and metastatic properties of cervical cancer cells in vitro and in vivo. Gynecol Oncol. 2007;106(2):400–406. doi: 10.1016/j.ygyno.2007.04.016. [DOI] [PubMed] [Google Scholar]
  • 108.Miller WH, Jr, Schipper HM, Lee JS, Singer J, Waxman S. Mechanisms of action of arsenic trioxide. Cancer Res. 2002;62(14):3893–3903. [PubMed] [Google Scholar]
  • 109.Li J, Chen P, Sinogeeva N, et al. Arsenic trioxide promotes histone H3 phosphoacetylation at the chromatin of CASPASE-10 in acute promyelocytic leukemia cells. J Biol Chem. 2002;277(51):49504–49510. doi: 10.1074/jbc.M207836200. [DOI] [PubMed] [Google Scholar]
  • 110.Davison K, Mann KK, Miller WH., Jr Arsenic trioxide: mechanisms of action. Semin Hematol. 2002;39(2 suppl 1):3–7. doi: 10.1053/shem.2002.33610. [DOI] [PubMed] [Google Scholar]
  • 111.Pan XY, Chen GQ, Cai L, Buscemi S, Fu GH. Anion exchanger 2 mediates the action of arsenic trioxide. Br J Haematol. 2006;134(5):491–499. doi: 10.1111/j.1365-2141.2006.06224.x. [DOI] [PubMed] [Google Scholar]
  • 112.Bernardini S, Nuccetelli M, Noguera NI, et al. Role of GSTP1-1 in mediating the effect of As2O3 in the acute promyelocytic leukemia cell line NB4. Ann Hematol. 2006;85(10):681–687. doi: 10.1007/s00277-006-0139-8. [DOI] [PubMed] [Google Scholar]
  • 113.Chou WC, Chen HY, Yu SL, et al. Arsenic suppresses gene expression in promyelocytic leukemia cells partly through Sp1 oxidation. Blood. 2005;106(1):304–310. doi: 10.1182/blood-2005-01-0241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Glienke W, Chow KU, Bauer N, Bergmann L. Down-regulation of wt1 expression in leukemia cell lines as part of apoptotic effect in arsenic treatment using two compounds. Leuk Lymphoma. 2006;47(8):1629–1638. doi: 10.1080/10428190600625398. [DOI] [PubMed] [Google Scholar]
  • 115.Joe Y, Jeong JH, Yang S, et al. ATR, PML, and CHK2 play a role in arsenic trioxide-induced apoptosis. J Biol Chem. 2006;281(39):28764–28771. doi: 10.1074/jbc.M604392200. [DOI] [PubMed] [Google Scholar]
  • 116.Chen YC, Lin-Shiau SY, Lin JK. Involvement of reactive oxygen species and caspase 3 activation in arsenite-induced apoptosis. J Cell Physiol. 1998;177(2):324–333. doi: 10.1002/(SICI)1097-4652(199811)177:2<324::AID-JCP14>3.0.CO;2-9. [DOI] [PubMed] [Google Scholar]
  • 117.Ninomiya M, Kajiguchi T, Yamamoto K, et al. Increased oxidative DNA products in patients with acute promyelocytic leukemia during arsenic therapy. Haematologica. 2006;91(11):1571–1572. [PubMed] [Google Scholar]
  • 118.Isakson P, Bjoras M, Boe SO, Simonsen A. Autophagy contributes to therapy-induced degradation of the PML/RARA oncoprotein. Blood. 2010;116(13):2324–2331. doi: 10.1182/blood-2010-01-261040. [DOI] [PubMed] [Google Scholar]
  • 119.Goussetis DJ, Altman JK, Glaser H, et al. Autophagy is a critical mechanism for the induction of the antileukemic effects of arsenic trioxide. J Biol Chem. 2010;285(39):29989–29997. doi: 10.1074/jbc.M109.090530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Jing Y, Wang L, Xia L, et al. Combined effect of all-trans retinoic acid and arsenic trioxide in acute promyelocytic leukemia cells in vitro and in vivo. Blood. 2001;97(1):264–269. doi: 10.1182/blood.v97.1.264. [DOI] [PubMed] [Google Scholar]
  • 121.Lallemand-Breitenbach V, Guillemin MC, Janin A, et al. Retinoic acid and arsenic synergize to eradicate leukemic cells in a mouse model of acute promyelocytic leukemia. J Exp Med. 1999;189(7):1043–1052. doi: 10.1084/jem.189.7.1043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Leung J, Pang A, Yuen WH, Kwong YL, Tse EWC. Relationship of expression of aquaglyceroporin 9 with arsenic uptake and sensitivity in leukemia cells. Blood. 2007;109(2):740–746. doi: 10.1182/blood-2006-04-019588. [DOI] [PubMed] [Google Scholar]
  • 123.Guillemin MC, Raffoux E, Vitoux D, et al. In vivo activation of cAMP signaling induces growth arrest and differentiation in acute promyelocytic leukemia. J Exp Med. 2002;196(10):1373–1380. doi: 10.1084/jem.20021129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Zhu Q, Zhang JW, Zhu HQ, et al. Synergic effects of arsenic trioxide and cAMP during acute promyelocytic leukemia cell maturation subtends a novel signaling cross-talk. Blood. 2002;99(3):1014–1022. [PubMed] [Google Scholar]
  • 125.Nasr R, Lallemand-Breitenbach V, Zhu J, Guillemin MC, de The H. Therapy-induced PML/RARA proteolysis and acute promyelocytic leukemia cure. Clin Cancer Res. 2009;15(20):6321–6326. doi: 10.1158/1078-0432.CCR-09-0209. [DOI] [PubMed] [Google Scholar]
  • 126.Nasr R, de The H. Eradication of acute promyelocytic leukemia-initiating cells by PML/RARA-targeting. Int J Hematol. 2010;91(5):742–747. doi: 10.1007/s12185-010-0582-0. [DOI] [PubMed] [Google Scholar]
  • 127.Zhao Q, Tao J, Zhu Q, et al. Rapid induction of cAMP/PKA pathway during retinoic acid-induced acute promyelocytic leukemia cell differentiation. Leukemia. 2004;18(2):285–292. doi: 10.1038/sj.leu.2403226. [DOI] [PubMed] [Google Scholar]
  • 128.Tarrade A, Bastien J, Bruck N, et al. Retinoic acid and arsenic trioxide cooperate for apoptosis through phosphorylated RXR alpha. Oncogene. 2005;24(14):2277–2288. doi: 10.1038/sj.onc.1208402. [DOI] [PubMed] [Google Scholar]
  • 129.Pendino F, Flexor M, Delhommeau F, et al. Retinoids down-regulate telomerase and telomere length in a pathway distinct from leukemia cell differentiation. Proc Natl Acad Sci U S A. 2001;98(12):6662–6667. doi: 10.1073/pnas.111464998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Tarkanyi I, Dudognon C, Hillion J, et al. Retinoid/arsenic combination therapy of promyelocytic leukemia: induction of telomerase-dependent cell death. Leukemia. 2005;19(10):1806–1811. doi: 10.1038/sj.leu.2403923. [DOI] [PubMed] [Google Scholar]
  • 131.Zheng PZ, Wang KK, Zhang QY, et al. Systems analysis of transcriptome and proteome in retinoic acid/arsenic trioxide-induced cell differentiation/apoptosis of promyelocytic leukemia. Proc Natl Acad Sci U S A. 2005;102(21):7653–7658. doi: 10.1073/pnas.0502825102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Liu YF, Zhu YM, Shi ZZ, et al. Long-term follow-up confirms the benefit of all-trans retinoic acid (ATRA) and arsenic trioxide (As2O3) as front line therapy for newly diagnosed acute promyelocytic leukemia [abstract]. Blood. 2006;108(11) Abstract 565. [Google Scholar]
  • 133.Estey E, Garcia-Manero G, Ferrajoli A, et al. Use of all-trans retinoic acid plus arsenic trioxide as an alternative to chemotherapy in untreated acute promyelocytic leukemia. Blood. 2006;107(9):3469–3473. doi: 10.1182/blood-2005-10-4006. [DOI] [PubMed] [Google Scholar]
  • 134.Wang G, Li W, Cui J, et al. An efficient therapeutic approach to patients with acute promyelocytic leukemia using a combination of arsenic trioxide with low-dose all-trans retinoic acid. Hematol Oncol. 2004;22(2):63–71. doi: 10.1002/hon.728. [DOI] [PubMed] [Google Scholar]
  • 135.Beijing, China: Culture Relics Press; 1979. Formularies for 52 Kinds of Disorders (Originally published in the spring and autumn and the warring states periods of China, 600 BC-400 BC). Reproduced by: Group for Management of the Silk Book in Mawangdui Han Dynasty Tomb. [Google Scholar]
  • 136.The Cooperation Group of Phase II Clinical Trial of Compound Huangdai Tablet. Phase II clinical trial of compound Huangdai tablet in newly diagnosed acute promyelocytic leukemia. Chin J Hematol. 2006;27:801–804. [Google Scholar]
  • 137.Xiang Y, Huang SL, Guo A, et al. The influence on long-term survey of the patients with acute promyelocytic leukemia treated with compound huangdai tablets and chemotherapy. Chin J Clin Hematol. 2007;16(5):204–206. [Google Scholar]
  • 138.Zhou A. Qing-Huang-San (Realgar in combination with Indigo) in treatment of leukemia. Chin J Integr Tradit West Med. 1998;18:582–583. [Google Scholar]
  • 139.Wang L, Zhou GB, Liu P, et al. Dissection of mechanisms of Chinese medicinal formula Realgar-Indigo naturalis as an effective treatment for promyelocytic leukemia. Proc Natl Acad Sci U S A. 2008;105(12):4826–4831. doi: 10.1073/pnas.0712365105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Carracedo A, Ma L, Teruya-Feldstein J, et al. Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J Clin Invest. 2008;118(9):3065–3074. doi: 10.1172/JCI34739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Chu S, Holtz M, Gupta M, Bhatia R. BCR/ABL kinase inhibition by imatinib mesylate enhances MAP kinase activity in chronic myelogenous leukemia CD34+ cells. Blood. 2004;103(8):3167–3174. doi: 10.1182/blood-2003-04-1271. [DOI] [PubMed] [Google Scholar]
  • 142.Piloto O, Wright M, Brown P, et al. Prolonged exposure to FLT3 inhibitors leads to resistance via activation of parallel signaling pathways. Blood. 2007;109(4):1643–1652. doi: 10.1182/blood-2006-05-023804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Altman JK, Yoon P, Katsoulidis E, et al. Regulatory effects of mammalian target of rapamycin-mediated signals in the generation of arsenic trioxide responses. J Biol Chem. 2008;283(4):1992–2001. doi: 10.1074/jbc.M705227200. [DOI] [PubMed] [Google Scholar]
  • 144.Hayakawa F, Privalsky ML. Phosphorylation of PML by mitogen-activated protein kinases plays a key role in arsenic trioxide-mediated apoptosis. Cancer Cell. 2004;5(4):389–401. doi: 10.1016/s1535-6108(04)00082-0. [DOI] [PubMed] [Google Scholar]
  • 145.Lunghi P, Tabilio A, Lo-Coco F, Pelicci PG, Bonati A. Arsenic trioxide (ATO) and MEK1 inhibition synergize to induce apoptosis in acute promyelocytic leukemia cells. Leukemia. 2005;19(2):234–244. doi: 10.1038/sj.leu.2403585. [DOI] [PubMed] [Google Scholar]
  • 146.Lunghi P, Costanzo A, Levrero M, Bonati A. Treatment with arsenic trioxide (ATO) and MEK1 inhibitor activates the p73-p53AIP1 apoptotic pathway in leukemia cells. Blood. 2004;104(2):519–525. doi: 10.1182/blood-2003-08-2743. [DOI] [PubMed] [Google Scholar]
  • 147.Lunghi P, Costanzo A, Salvatore L, et al. MEK1 inhibition sensitizes primary acute myelogenous leukemia to arsenic trioxide-induced apoptosis. Blood. 2006;107(11):4549–4553. doi: 10.1182/blood-2005-07-2829. [DOI] [PubMed] [Google Scholar]
  • 148.Lunghi P, Giuliani N, Mazzera L, et al. Targeting MEK/MAPK signal transduction module potentiates ATO-induced apoptosis in multiple myeloma cells through multiple signaling pathways. Blood. 2008;112(6):2450–2462. doi: 10.1182/blood-2007-10-114348. [DOI] [PubMed] [Google Scholar]
  • 149.Ren R. Mechanisms of BCR-ABL in the pathogenesis of chronic myelogenous leukaemia. Nat Rev Cancer. 2005;5(3):172–183. doi: 10.1038/nrc1567. [DOI] [PubMed] [Google Scholar]
  • 150.Druker BJ, Guilhot F, O'Brien SG, et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med. 2006;355(23):2408–2417. doi: 10.1056/NEJMoa062867. [DOI] [PubMed] [Google Scholar]
  • 151.Michor F, Hughes TP, Iwasa Y, et al. Dynamics of chronic myeloid leukaemia. Nature. 2005;435(7046):1267–1270. doi: 10.1038/nature03669. [DOI] [PubMed] [Google Scholar]
  • 152.Gorre ME, Mohammed M, Ellwood K, et al. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science. 2001;293(5531):876–880. doi: 10.1126/science.1062538. [DOI] [PubMed] [Google Scholar]
  • 153.Copland M, Hamilton A, Elrick LJ, et al. Dasatinib (BMS-354825) targets an earlier progenitor population than imatinib in primary CML but does not eliminate the quiescent fraction. Blood. 2006;107(11):4532–4539. doi: 10.1182/blood-2005-07-2947. [DOI] [PubMed] [Google Scholar]
  • 154.Azam M, Latek RR, Daley GQ. Mechanisms of autoinhibition and STI-571/imatinib resistance revealed by mutagenesis of BCR-ABL. Cell. 2003;112(6):831–843. doi: 10.1016/s0092-8674(03)00190-9. [DOI] [PubMed] [Google Scholar]
  • 155.Shah NP, Nicoll JM, Nagar B, et al. Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia. Cancer Cell. 2002;2(2):117–125. doi: 10.1016/s1535-6108(02)00096-x. [DOI] [PubMed] [Google Scholar]
  • 156.Donato NJ, Wu JY, Stapley J, et al. BCR-ABL independence and LYN kinase overexpression in chronic myelogenous leukemia cells selected for resistance to STI571. Blood. 2003;101(2):690–698. doi: 10.1182/blood.V101.2.690. [DOI] [PubMed] [Google Scholar]
  • 157.Druker BJ, Sawyers CL, Kantarjian H, et al. Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. N Engl J Med. 2001;344(14):1038–1042. doi: 10.1056/NEJM200104053441402. [DOI] [PubMed] [Google Scholar]
  • 158.Kerkela R, Grazette L, Yacobi R, et al. Cardiotoxicity of the cancer therapeutic agent imatinib mesylate. Nat Med. 2006;12(8):908–916. doi: 10.1038/nm1446. [DOI] [PubMed] [Google Scholar]
  • 159.Hu Z, Pan XF, Wu FQ, et al. Synergy between proteasome inhibitors and imatinib mesylate in chronic myeloid leukemia. PLoS ONE. 2009;4(7):e6257. doi: 10.1371/journal.pone.0006257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.O'Dwyer M. Multifaceted approach to the treatment of bcr-abl-positive leukemias. Oncologist. 2002;7(Suppl 1):30–38. doi: 10.1634/theoncologist.7-suppl_1-30. [DOI] [PubMed] [Google Scholar]
  • 161.Nimmanapalli R, Bali P, O'Bryan E, et al. Arsenic trioxide inhibits translation of mRNA of bcr-abl, resulting in attenuation of Bcr-Abl levels and apoptosis of human leukemia cells. Cancer Res. 2003;63(22):7950–7958. [PubMed] [Google Scholar]
  • 162.Zhang QY, Mao JH, Liu P, et al. A systems biology understanding of the synergistic effects of arsenic sulfide and imatinib in BCR/ABL-associated leukemia. Proc Natl Acad Sci U S A. 2009;106(9):3378–3383. doi: 10.1073/pnas.0813142106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Yin T, Wu YL, Sun HP, et al. Combined effects of As4S4 and imatinib on chronic myeloid leukemia cells and BCR-ABL oncoprotein. Blood. 2004;104(13):4219–4225. doi: 10.1182/blood-2004-04-1433. [DOI] [PubMed] [Google Scholar]
  • 164.La Rosee P, Johnson K, O'Dwyer ME, Druker BJ. In vitro studies of the combination of imatinib mesylate (Gleevec) and arsenic trioxide (Trisenox) in chronic myelogenous leukemia. Exp Hematol. 2002;30(7):729–737. doi: 10.1016/s0301-472x(02)00836-6. [DOI] [PubMed] [Google Scholar]
  • 165.Du Y, Wang K, Fang H, et al. Coordination of intrinsic, extrinsic, and endoplasmic reticulum-mediated apoptosis by imatinib mesylate combined with arsenic trioxide in chronic myeloid leukemia. Blood. 2006;107(4):1582–1590. doi: 10.1182/blood-2005-06-2318. [DOI] [PubMed] [Google Scholar]
  • 166.Berenson JR, Boccia R, Siegel D, et al. Efficacy and safety of melphalan, arsenic trioxide and ascorbic acid combination therapy in patients with relapsed or refractory multiple myeloma: a prospective, multicentre, phase II, single-arm study. Br J Haematol. 2006;135(2):174–183. doi: 10.1111/j.1365-2141.2006.06280.x. [DOI] [PubMed] [Google Scholar]
  • 167.Abou-Jawde RM, Reed J, Kelly M, et al. Efficacy and safety results with the combination therapy of arsenic trioxide, dexamethasone, and ascorbic acid in multiple myeloma patients: a phase 2 trial. Med Oncol. 2006;23(2):263–272. doi: 10.1385/MO:23:2:263. [DOI] [PubMed] [Google Scholar]
  • 168.Wu KL, Beksac M, van Droogenbroeck J, et al. Phase II multicenter study of arsenic trioxide, ascorbic acid and dexamethasone in patients with relapsed or refractory multiple myeloma. Haematologica. 2006;91(12):1722–1723. [PubMed] [Google Scholar]
  • 169.Berenson JR, Matous J, Swift RA, et al. A Phase I/II study of arsenic trioxide/bortezomib/ascorbic acid combination therapy for the treatment of relapsed or refractory multiple myeloma. Clin Cancer Res. 2007;13(6):1762–1768. doi: 10.1158/1078-0432.CCR-06-1812. [DOI] [PubMed] [Google Scholar]
  • 170.Hermine O, Dombret H, Poupon J, et al. Phase II trial of arsenic trioxide and alpha interferon in patients with relapsed/refractory adult T-cell leukemia/lymphoma. Hematol J. 2004;5(2):130–134. doi: 10.1038/sj.thj.6200374. [DOI] [PubMed] [Google Scholar]
  • 171.Kchour G, Tarhini M, Kooshyar MM, et al. Phase 2 study of the efficacy and safety of the combination of arsenic trioxide, interferon alpha, and zidovudine in newly diagnosed chronic adult T-cell leukemia/lymphoma (ATL). Blood. 2009;113(26):6528–6532. doi: 10.1182/blood-2009-03-211821. [DOI] [PubMed] [Google Scholar]
  • 172.Schiller GJ, Slack J, Hainsworth JD, et al. Phase II multicenter study of arsenic trioxide in patients with myelodysplastic syndromes. J Clin Oncol. 2006;24(16):2456–2464. doi: 10.1200/JCO.2005.03.7903. [DOI] [PubMed] [Google Scholar]
  • 173.Vey N, Bosly A, Guerci A, et al. Arsenic trioxide in patients with myelodysplastic syndromes: a phase II multicenter study. J Clin Oncol. 2006;24(16):2465–2471. doi: 10.1200/JCO.2005.03.9503. [DOI] [PubMed] [Google Scholar]
  • 174.Zheng WL, Zhang GS, Xu YX, et al. Arsenic trioxide, thalidomide and retinoid acid combination therapy in higher risk myelodysplastic syndrome patients. Leuk Res. 2008;32(2):251–254. doi: 10.1016/j.leukres.2007.05.025. [DOI] [PubMed] [Google Scholar]
  • 175.Chang JE, Voorhees PM, Kolesar JM, et al. Phase II study of arsenic trioxide and ascorbic acid for relapsed or refractory lymphoid malignancies: a Wisconsin Oncology Network study. Hematol Oncol. 2009;27(1):11–16. doi: 10.1002/hon.870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Bazarbachi A, El-Sabban ME, Nasr R, et al. Arsenic trioxide and interferon-alpha synergize to induce cell cycle arrest and apoptosis in human T-cell lymphotropic virus type I-transformed cells. Blood. 1999;93(1):278–283. [PubMed] [Google Scholar]
  • 177.Nasr R, Rosenwald A, El-Sabban ME, et al. Arsenic/interferon specifically reverses 2 distinct gene networks critical for the survival of HTLV-1-infected leukemic cells. Blood. 2003;101(11):4576–4582. doi: 10.1182/blood-2002-09-2986. [DOI] [PubMed] [Google Scholar]
  • 178.El-Sabban ME, Nasr R, Dbaibo G, et al. Arsenic-interferon-alpha-triggered apoptosis in HTLV-I transformed cells is associated with tax down-regulation and reversal of NF-kappa B activation. Blood. 2000;96(8):2849–2855. [PubMed] [Google Scholar]
  • 179.El Hajj H, El-Sabban M, Hasegawa H, et al. Therapy-induced selective loss of leukemia-initiating activity in murine adult T cell leukemia. J Exp Med. 2010;207(13):2785–2792. doi: 10.1084/jem.20101095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Emadi A, Gore SD. Arsenic trioxide–an old drug rediscovered. Blood Rev. 2007;24(4–5):191–199. doi: 10.1016/j.blre.2010.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Westervelt P, Brown RA, Adkins DR, et al. Sudden death among patients with acute promyelocytic leukemia treated with arsenic trioxide. Blood. 2001;98(2):266–271. doi: 10.1182/blood.v98.2.266. [DOI] [PubMed] [Google Scholar]
  • 182.Mathews V, George B, Lakshmi KM, et al. Single-agent arsenic trioxide in the treatment of newly diagnosed acute promyelocytic leukemia: durable remissions with minimal toxicity. Blood. 2006;107(7):2627–2632. doi: 10.1182/blood-2005-08-3532. [DOI] [PubMed] [Google Scholar]
  • 183.Luesink M, Pennings JLA, Wissink WM, et al. Chemokine induction by all-trans retinoic acid and arsenic trioxide in acute promyelocytic leukemia: triggering the differentiation syndrome. Blood. 2009;114(27):5512–5521. doi: 10.1182/blood-2009-02-204834. [DOI] [PubMed] [Google Scholar]

Articles from Blood are provided here courtesy of The American Society of Hematology

RESOURCES