Abstract
Over the last 17 years, clinical trials conducted worldwide have demonstrated the efficacy of arsenic trioxide (As2O3) in the treatment of relapsed acute promyelocytic leukemia (APL). Currently, the role of As2O3 in front-line therapy is under investigation. Recent trials in the US have demonstrated that the addition of As2O3 to standard treatment regimens improves survival outcomes in patients with APL and may allow a reduction in cytotoxic chemotherapy exposure. As2O3 has also shown efficacy in other malignancies, particularly multiple myeloma and myelodysplastic syndromes. Therapeutic doses of As2O3 are well tolerated, with no evidence of long-term toxicity. Adverse events include APL differentiation syndrome, electrocardiographic abnormalities, and mild elevations in liver enzymes. This review highlights trials investigating the role of As2O3 in induction and consolidation for newly diagnosed APL, as well as its role in other hematologic malignancies. The chemistry, mechanisms of action, and clinical side effects of As2O3 are also discussed.
Keywords: Arsenic trioxide (As2O3), Acute promyelocytic leukemia (APL), Differentiation, Apoptosis, Reactive oxygen species, Induction and consolidation chemotherapy, Multiple myeloma, T-cell lymphotropic virus type I (HTLV-I)-associated adult T-cell leukemia-lymphoma (ATL), Myelodysplastic syndromes (MDS)
Introduction
Rediscovery of an old drug
Arsenic has been used in medicine for more than 2400 years for a variety of ailments including ulcers, the plague, and malaria.1 In 1878, potassium arsenite was reported to have an anti-leukemic effect and was used for this purpose in the late 19th and early 20th centuries until it was replaced by busulfan in the 1950s.2–4
In the modern era, interest in arsenic as a chemotherapy was rekindled after it was identified as an active ingredient in traditional medicines in China.5 Researchers evaluated arsenic compounds for the treatment of various cancers and in 1992 published the results of a trial in which intravenous administration of arsenic trioxide (As2O3) produced a complete response (CR) in 21 (66%) of 32 patients with acute promyelocytic leukemia (APL).5,6 In two subsequent studies, Zhang et al reported that As2O3 induced a CR in 22 (73%) of 30 newly diagnosed and 22 (52%) of 42 relapsed APL patients,5,7 and Shen et al. observed a CR in nine (90%) of 10 relapsed APL patients.8
As2O3 for relapsed APL
Based on the results from the Chinese studies, a US pilot study was conducted in patients who had relapsed after one or more courses of all-trans-retinoic acid (ATRA) and anthracycline-based chemotherapy.9 Most patients had experienced multiple relapses and 58% were resistant to retinoid therapy. Eleven of 12 patients achieved a CR with As2O3 alone. Eight of these 11 responders also achieved a molecular remission, as defined by the absence of detectable PML-RARα fusion gene expression.
Following the success of the pilot study, a larger, multicenter, single-arm trial was conducted in patients who had relapsed after ATRA-based therapy. More than one third of these patients had multiple relapses and were heavily pretreated (including five patients with prior BMT). CR was achieved in 34 (85%) of 40 patients (Table 1).10 All patients who achieved a CR also showed evidence of elimination of the t(15;17), as measured either directly by traditional cytogenetics or by assays using FISH or RT-PCR for PML/RAR-α. Many of the remissions were durable. The estimated two-year overall survival (OS) and relapse-free survival (RFS) rates for patients in first relapse were 77% and 58%, respectively. These results are somewhat confounded because eleven patients underwent bone marrow transplant in remission after induction or consolidation with As2O3. Among the 27 patients who did not receive transplant, one-third remained alive with duration of remission ranging 22 to 42 months. This pivotal trial provided support for the approval of As2O3 in patients with APL who failed to respond to or relapsed following ATRA/anthracycline therapy.
Table 1.
Current therapies for APL
Study | N | Regimen | CR, % | CIR, % | EFS, % | DFS, % | OS, % |
---|---|---|---|---|---|---|---|
Initial therapy | |||||||
PETHEMA – LPA9911 |
251 | I: ATRA + CT (IDA) | 90 | 8.7, 3-yr | NR | 90, 3-yr | 85, 3-yr |
C: 3 courses CT (IDA, MXN, IDA; low-risk pts) or 3 courses ATRA + CT (intermediate/high-risk pts) | |||||||
M: Low-dose CT (MCP/ MTX) + intermittent ATRA (every 3 mos) for 2 yrs | |||||||
European APL Group – APL 200012 |
340 | I: ATRA + CT (DNR ± ara-C) | 96 | 8.4, 2-yr | 84.5, 2-yr | NR | 91.9, 2-yr |
C: 2 courses CT (DNR ± ara-C) | |||||||
M: Low-dose CT (MCP/ MTX) + intermittent ATRA (every 3 mos) for 2 yrs | |||||||
JALSG – APL9713 | 283 |
I: ATRA (low-risk pts) or ATRA + CT (IDA + ara-C; intermediate/high-risk pts) |
94 | NR | NR | 68.5, 6-yr | 83.9, 6-yr |
C: 3 courses CT (MXN + ara-C, ara-C+ etoposide + DNR, ara-C + IDA) | |||||||
M: Randomized to observation or intensified maintenance CT (6 courses CT every 6 wks) | |||||||
Second-line therapy | |||||||
Soignet et al.10 | 40 | I: As2O3 (daily until BM remission or 60 days) | 85 | NR | NR | 56, 1.5-yr (RFS) |
66, 1.5-yr |
C: As2O3 (25 doses in 35 days) | |||||||
M: Up to 4 additional consolidation courses of As2O3 |
Abbreviations: APL, acute promyelocytic leukemia; ara-C, cytarabine; As2O3, arsenic trioxide; ATRA, all-trans-retinoic acid; BM, bone marrow; C, consolidation therapy; CIR, cumulative incidence of relapse; CR, complete response; CT, chemotherapy; DFS, disease-free survival; DNR, daunorubicin; EFS, event-free survival; I, induction therapy; IDA, idarubicin; JALSG, Japan Adult Leukemia Study Group; M, maintenance therapy; MCP, mercaptopurine; MXN, mitoxantrone; MTX, methotrexate; NR, not reported; OS, overall survival; PETHEMA, Programme de Estudio y Tratamiento de las Hamopatias Malignas; RFS, relapse-free survival.
Mechanisms of action
As2O3 affects multiple cellular functions via different molecular targets (summarized in Fig. 1). Although the fundamental mechanism is the favorable chemical reaction between arsenic and thiol groups within a protein, the final outcome depends on the cell type as well as the dose and duration of arsenite exposure. For example, in APL cells, As2O3 at low concentrations (<0.5 µM) induces differentiation; at higher concentrations (0.5–2.0 µM) it causes apoptosis.14,15
Figure 1. As2O3 targets multiple cellular pathways.
In APL cells, arsenic trioxide (As2O3) restores differentiation by degrading the PML-RARα fusion protein. However, As2O3 has additional targets that are present in multiple cancer cell types. As2O3 targets the mitochondria, decreasing the mitochondrial membrane potential (ΔΨm) via multiple specific targets including Bcl-2 and the PTPC. This change in potential results in the release of cytochrome C, which activates the caspase cascade. It also results in increased release of ROS from the mitochondria. ROS levels are increased further by As2O3 inhibition of the antioxidant enzyme GPx. As2O3 also inhibits activation of the cell-survival factor NFκB via inhibition of IKK, the kinase responsible for releasing NFκB that is sequestered in the cytoplasm.
Abbreviations: APAF, apoptotic peptidase activating factor; GPx, glutathione peroxidase; IKK, IκB kinase; PTPC, permeability transition pore complex; ROS, reactive oxygen species.
Chemistry of As2O3
Trivalent arsenic (AsIII) in As2O3 or arsenite (As[OH]3) and pentavalent arsenic (AsV) in arsenate (HAsO42−) are the two biologically significant forms of arsenic. Although AsV disrupts cellular processes as a phosphate (HPO42−) mimic, the interaction of AsIII with the thiol (or sulfhydryl) groups (-SH) of proteins with a high cysteine content is the basic reaction that underlies the multiple mechanisms of action of this chemotherapeutic agent.16 In this reaction, the valence orbitals of arsenic (As) have a better overlap and energy match with those of sulfur (S) than with those of oxygen (O), leading to the formation of an AsIII-thiolate bond and the release of water, as demonstrated in the following equation17:
Vicinal thiols (i.e., thiols bonded to adjacent carbon atoms) have a high affinity for AsIII (Fig. 2). Thus, proteins that contain cysteine residues that are conformationally constrained and favorably positioned will have a higher affinity for AsIII.18
Figure 2.
Reaction of vicinal sulfhydryl groups in a protein structure with trivalent arsenic.
Stimulation of differentiation
Empirically, treatment of APL cells with As2O3 leads to their terminal differentiation in vitro and in vivo. APL cells are uniquely sensitive to As2O3 due to the expression of the PML-RARα fusion protein; however, the mechanism by which arsenic trioxide treatment induces terminal differentiation remains somewhat speculative. In normal myeloid cells, PML protein is localized to macromolecular structures in the nucleus (nuclear bodies), where PML antagonizes many processes required for the initiation and progression of malignancy.19 In leukemic cells, the PML-RARα fusion protein blocks the expression of genes required for normal differentiation. The fusion protein disrupts the nuclear bodies, and the PML protein is dispersed into smaller organelles.20 PML contains a cysteine-rich region that is hypothesized to interact with AsIII, resulting in the degradation of PML-RARα fusion protein.21 Furthermore, As2O3-induced histone acetylation has been reported to promote differentiation via alteration in gene transcription.22
Induction of apoptosis
Caspases are intracellular cysteine proteases that are key components in classic apoptosis. Caspase activation occurs in response to various types of mitochondrial damage and proapoptotic stimuli, which cause cytochrome c (normally sequestered between the mitochondrial inner and outer membranes) to be released into the cytosol, where it binds and activates Apaf-1, which in turn activates procaspase-9.23–25 Caspase-9 cleaves procaspase-3, and downstream of caspase-3, the apoptotic program branches into a multitude of subprograms, the sum of which results in the ordered dismantling and removal of the cell.26
As2O3 induces dose-dependent apoptosis in APL and hematopoietic non-APL as well as tumor and non-malignant cell lines.27,28 The activation of the caspase cascade, the decrease of the mitochondrial membrane potential ΔΨm, and the production of reactive oxygen species (ROS) all play roles in induction of As2O3-induced apoptosis.29 As2O3 also promotes apoptosis via down-regulation of Bcl-2 expression30 by prolongation of the cell cycle and cell cycle arrest of malignant cells31,32 and by opening the permeability transition pore complex (PTPC) in mitochondrial membranes by directly binding to thiol groups in the PTPC.33
Accumulation of ROS
ROS are damaging to DNA, RNA, proteins, and lipids, and include free radicals such as hydroxyl (OH• ) or superoxide (O2−•) and molecules such as hydrogen peroxide (H2O2). Mutations in nuclear or mitochondrial genes encoding components of the mitochondrial electron transport chain can lead to the accumulation of electrons along the chain. These electrons are captured by O2 to form superoxide, which is converted to hydrogen peroxide. The protective buffering systems against ROS include glutathione (GSH), thioredoxin, superoxide dismutase, catalase, and glutathione peroxidase.34
As2O3 can increase cellular levels of ROS via several targets. It can prevent ROS detoxification by inhibiting antioxidant enzymes like glutathione peroxidase, which has a thiol group required for its activation. The level of intracellular GSH, which titrates arsenic by forming As(GS)3 complexes, as well as the activities of related antioxidant enzymes, are major factors for sensitivity to As2O3. The sensitivity of APL to As2O3-induced apoptosis is closely related to its relatively poor antioxidant capacity.35 Furthermore, proteins with high sulfhydryl-disulfide oxidation potentials may be particularly susceptible to redox regulation, and the affinity of As2O3 for sulfhydryl groups on these redox-sensitive proteins explains many of its ROS-related effects.36 Finally, through the use of gene expression profiling, interference RNA, and genetically engineered cells, Chou and colleagues reported that NADPH oxidase, an enzyme complex required for the normal antibacterial function of white blood cells, is a major target of arsenic-induced ROS production.37
Inhibition of NFκB
NFκB is a transcriptional factor promoting cell survival with an important role in many cancer cells. Activation of NFκB depends on the integrity of the IκB kinase (IKK); upon phosphorylation by IKK, the inhibitory protein IκB releases NFκB for translocation to the nucleus. As2O3 has been shown to inhibit IKK by binding to cysteine-179 in the activation loop of the enzyme catalytic subunit. Although cysteine-179 is not located in the vicinity of another cysteine within the IKK primary structure, it has been suggested that another cysteine may come within a critical distance of cysteine-179 upon the folding of the polypeptide chain or the dimerization of the catalytic subunits, thus providing a high-affinity target for arsenite.38
As2O3 selectivity for APL cells
To understand the selective cytotoxicity of As2O3 against human APL, Dilda et al. screened the yeast deletion strains for sensitivity or resistance to the drug.39 A prominent sensitive mutant was missing Hog1, a MAP kinase. The most resistant mutant lacked the plasma-membrane glycerol and arsenite transporter, Fps1. Hog1 and Fps1 control the response to osmotic stress in yeast through regulation of glycerol production and plasma membrane flux, respectively. Based on these results, the investigators tested the APL cell line NB4 for impaired osmoregulation and found that the APL cells did not produce glycerol in response to osmotic stress and underwent apoptotic cell death. Moreover, the glycerol content of NB4 and differentiated NB4 cells correlated with the level of As2O3 uptake and the sensitivity of the cells. Additionally, NB4 cells accumulated more As2O3 than did non-APL cells and were generally more sensitive to the drug. The investigators concluded that the selectivity of As2O3 for APL cells relates, at least in part, to impaired osmoregulation and control of drug uptake.
Efficacy of As2O3 in newly diagnosed APL
The standard of care for newly diagnosed APL is differentiation therapy with ATRA plus anthracycline-based chemotherapy.40 This combination results in high response rates and prolonged survival (Table 1).11–13 Incorporating As2O3 into the initial treatment of APL may further reduce the relapse rate and provide a more tolerable treatment option for patients who are not candidates for chemotherapy.
As2O3 as induction therapy
As2O3 has demonstrated clinical activity in patients with newly diagnosed APL, producing CR in 83–86% of patients and 3-year OS rates of 79–86% (Table 2).41,42 Demonstrated efficacy, combined with evidence of synergy between As2O3 and ATRA, has fueled interest in combination therapy. A small randomized study in China evaluated the efficacy and safety of induction with ATRA, As2O3, or ATRA plus As2O3, followed by consolidation with three courses of conventional chemotherapy (Table 2).43 Response rates were similar between treatment groups; however, relapse was significantly less frequent in patients receiving combination therapy as compared to either agent alone (P = .038). This improved response translated into a 2-year disease-free survival (DFS) advantage. After completion of the randomized phase of the study, the ATRA-plus-As2O3 arm remained open and long-term efficacy and safety data were collected (Table 2).44 Among 80 patients who achieved CR, four (5%) had relapsed after a median follow-up of 70 months; all four were central nervous system (CNS) relapses.
Table 2.
Clinical studies of arsenic trioxide in first-line therapy for APLa
Study | N | Regimen | CR | EFS | DFS (or RFS) | OS |
---|---|---|---|---|---|---|
As2O3 in induction | ||||||
Mathews et al.41 | 72 | I: As2O3 (daily until CR or 60 days) | 86% | 75%, 3-yr | 87%, 3-yr | 86%, 3-yr |
C: As2O3 (daily for 4 wks) | ||||||
M: As2O3 10 d per mo for 6 mos | ||||||
Ghavamzadeh et al.42 |
193 | I: As2O3 (daily until CR or 60 days) | 83% | NR | 69%, 3-yr | 79%, 3-yr |
C and M: 1 or 4 courses As2O3 (daily for 4 wks) | ||||||
Shen et al.43 |
61 |
I: (A) ATRA or (B) As2O3 or (C) ATRA + As2O3 until CR | A: 95% B: 90% C: 95% |
NR | A: 68%, 2-yr B: 89%, 2-yr C: 100%, 2-yr |
NR |
C: (all patients) 3 courses CTb | ||||||
M: (A) ATRA or (B) As2O3 or (C) ATRA then As2O3 followed by low-dose CT (MCP or MTX) for 5 cycles | ||||||
Hu et al.44 | 85 | I: ATRA + As2O3 (daily until CR) | 94% | 89%, 5-yr | 95%, 5-yr (RFS) |
92%, 5-yr |
C: 3 courses CTb | ||||||
M: ATRA then As2O3 followed by low-dose CT (MCP or MTX) for 5 cycles | ||||||
Ravandi et al.45 | 82 |
I: ATRA + As2O3 (+ GO in high-risk patients) until BM remission or 85 days |
91% (CR/CRi) |
83%, 5-yr | NR | 84%, 5-yr |
C and M: 7 cycles ATRA + 4 cycles As2O3 | ||||||
As2O3 in consolidation | ||||||
Powell et al.46 (C9710) |
481 | I: ATRA + CT (DNR + ara-C) | A: 89% B: 89% |
A: 81%, 3-yr B: 66%, 3-yr P = .0007 |
NR | A: 86%, 3-yr B: 79%,3-yr P = .063 |
C: (randomized) (A) 2 courses As2O3, then 2 courses ATRA + CT (DNR) or (B) 2 courses ATRA + CT (DNR) | ||||||
M: (randomized) ATRA ± low-dose CT (MCP + MTX) | ||||||
Gore et al.47 | 45 | I: ATRA + CT (DNR) for 60 days | 91% | 76%, 2.7-yr | 90%, 2.7-yr | 88%, 2.7-yr |
C: 1 course As2O3 + CT (DNR + ara-C) | ||||||
M: ATRA (low/intermediate-risk patients) or ATRA + low- dose CT (MCP + MTX) (high-risk patients) |
Abbreviations: APL, acute promyelocytic leukemia; As2O3, arsenic trioxide; ATRA, all-trans-retinoic acid; ara-C, cytarabine; BM, bone marrow; C, consolidation therapy; CR, complete response; CRi, complete response without full platelet recovery; CT, chemotherapy; DFS, disease-free survival; DNR, daunorubicin; EFS, event-free survival; GO, gemtuzumab ozogamicin; I, induction therapy; M, maintenance therapy; MCP, mercaptopurine; MTX, methotrexate; NR, not reported; OS, overall survival; RFS, relapse-free survival.
All patients were newly diagnosed with APL.
Each course consisted of 3 consecutive regimens: DA (DNA + ara-C), ara-C pulse, HA (homogarringtonine + ara-C).
A US phase 2 trial examined whether combination therapy with ATRA and As2O3 could replace anthracycline-based chemotherapy for newly diagnosed APL.45 Low-risk patients (white blood cell [WBC] count <10 × 109/L) received ATRA plus As2O3 as induction and post-remission therapy; high-risk patients (WBC count ≥10 × 109/L) also received a single dose of gemtuzumab ozogamicin. It was hypothesized that this approach might be advantageous for patients who are unable to tolerate conventional chemotherapy, particularly elderly patients. Outcomes were similar to those observed with standard ATRA and chemotherapy combinations (Table 2). The overall response rate was 92% and responses were durable. After a median follow-up period of 23 months, relapse was documented in three (4%) of 75 responders, all of whom had high-risk disease. In patients aged 60 years and older, a CR rate of 83% was observed, compared with 95% in patients younger than 60 years (P = .17).
As2O3 in consolidation
The only phase 3 trial of As2O3 (the North American Intergroup protocol C9710, presented to date only in abstract form) evaluated the addition of As2O3 in first CR prior to standard consolidation therapy for newly diagnosed 537 eligible adults and pediatrics patients with APL.46 This study demonstrated that administration of As2O3 (0.15 mg/kg/d for 5 days each week for 5 week for two cycles, cycle 2 after two weeks rest), as the first consolidation, prior to subsequent consolidation with ATRA (45 mg/m2 × 7d) and chemotherapy (daunorubicin 50 mg/m2 × 3d; 2d for age < 15 yr) significantly improved event-free survival (EFS) (81% vs 66%, p=0.0007) in adults compared to consolidation with ATRA and chemotherapy only. Three-year OS was higher in the As2O3 group, albeit not statistically significant (86% vs 79%, p=0.063). These improvements were presumably due to a decrease in the relapse rate, although DFS has not yet been reported. It is noteworthy to mention that in this study patients who did not receive As2O3 appear to have had lower EFS and OS than historical controls treated with ATRA and chemotherapy; indeed, the survival rate in the As2O3 arm was similar to the best published data using ATRA plus chemotherapy. Full analysis of this critical study will require publication of the final manuscript.
Based on the preliminary report, there was remarkably no significant difference in DFS between patients with WBC count greater or less than 10,000/mL in As2O3 group. On the other hand, patients with WBC > 10,000/mL, who did not receive As2O3 had significantly worse DFS compared with patients with WBC < 10,000/mL (p=0.0016). This finding suggests a major advantage of As2O3-based consolidation compared to non-As2O3 containing regimens in which patients with high WBC are much more likely to relapse. There were no differences in grade 3 or 4 hematologic or non-hematologic toxicities between the two groups.
A recent phase 2 trial in the US assessed whether the incorporation As2O3 into consolidation therapy would allow a reduction in chemotherapy exposure without compromising patient outcomes.47 Enrolled patients (45 analyzed) received a single consolidation cycle with As2O3 (0.15 mg/kg/day, Monday – Friday, beginning on day 8, for 30 doses), daunorubicin (60 mg/m2/day days 1–3), and cytarabine (0.667 mg/m2/day continuous infusion days 1–3) after achieving CR with ATRA plus chemotherapy. Survival outcomes (EFS, DFS, and OS) were comparable to other treatment regimens that included more extensive chemotherapy, including the As2O3 treatment arm of the C9710 phase 3 trial (Table 2). Of 37 patients who received consolidation therapy, only one (3%) patient suffered a relapse (in the CNS) after a median follow-up of 1.8 years. Thus, OS was 88% ± 5% and leukemia-free survival was 90 ± 6%. Secondary myelodysplastic syndromes (MDS) or acute myeloid leukemia (AML) were not reported, although the longest follow-up in this study is only 5.5 years and median follow-up 2.7 years. These results indicate that a reduction in anthracycline exposure and associated toxicities may be possible while maintaining a low incidence of relapse.
Based on the current data from the As2O3-based clinical trials, which indicate that first-line arsenic trioxide therapy markedly decreases the incidence of relapse in de novo APL, and high DFS can be achieved with much lower doses of anthracycline than currently being used, we strongly recommend the routine incorporation of As2O3 into the first-line consolidation therapy of de novo APL.
As2O3 in maintenance
In the study by Soignet et al 10, those patients who remained in CR after receiving their consolidation course of As2O3 were given the option to receive up to four additional cycles of As2O3 therapy on a dose schedule similar to consolidation on a separate protocol. Eighteen patients received additional As2O3 as maintenance. Nevertheless, to date there is no published randomized trial evaluating the effect of As2O3 in maintenance setting. Meanwhile, until further studies are designed and completed, maintenance therapy for 1 to 2 years with intermittent ATRA with or without 6-mercaptopurine and methotrexate is recommended by us and many others, particularly in patients with high-risk disease.
Efficacy of As2O3 in other malignancies
Multiple myeloma (MM)
In preclinical studies, As2O3 inhibited the proliferation of MM cell lines at pharmacological (micromolar) concentrations.31,48 As2O3 treatment of cultured bone marrow mononuclear cells from MM patients selectively induced apoptosis in myeloma cells while sparing most myeloid cells.48 Initial clinical studies of As2O3 as a single agent in heavily pretreated relapsed or refractory patients demonstrated a ≥25% reduction in serum levels of M-protein in 21–33% of patients, with durations of response ranging from 1 to 74+ weeks.49–51 Disease stabilization occurred in an additional 7–40% of patients. This modest clinical efficacy of As2O3 as a single agent prompted trials in combination with other therapies.
As discussed in the mechanism of action section, sensitivity to As2O3 in vitro is inversely proportional to cellular levels of the antioxidant GSH, which can attenuate the effects of increased ROS.35 Pharmacological depletion of GSH by ascorbic acid (AA) enhances the cytotoxic effects of As2O3 in MM cell lines, providing a rationale for combination therapy.52 A phase 1 trial demonstrated that As2O3 plus AA was tolerable and demonstrated potential clinical activity (two of six patients had partial responses [PR]).53 Further clinical studies are necessary to determine the efficacy of this combination, particularly in light of more recent in vitro evidence suggesting that AA may protect cells against arsenic cytotoxicity.54
Several trials have evaluated As2O3 in combination with existing MM therapies, including melphalan, dexamethasone, and bortezomib, in relapsed patients. In a small study of 10 patients treated with As2O3, ascorbic acid, and melphalan (MAC), a response (≥25% reduction in serum M-protein) was observed in all patients, with treatment durations of 13–104 weeks.55 Notably, six of these 10 patients had received melphalan previously: four as conditioning for stem cell transplantation (SCT) and two as part of a melphalan-prednisone regimen during which disease progression occurred. In a subsequent phase 2 study, MAC treatment produced a response in 31 (48%) of 65 patients who had failed at least two prior regimens; two CR, 15 PR, and 14 minor responses were observed.56 Median progression-free survival (PFS) was 7 months and median OS was 19 months. In another randomized phase 2 trial, the addition of As2O3 to AA and high-dose melphalan was safe and well tolerated as a preparative regimen for autologous hematopoietic stem cell transplantation.57 This study was not powered to detect differences in efficacy between arms; overall, a response was observed in 41 (85%) of 48 patients including 12 (25%) CR, with a median PFS of 25 months. The combination of As2O3, AA, and dexamethasone has demonstrated efficacy in patients with relapsed or refractory MM in two phase 2 trials. In a US study, six (30%) of 20 patients achieved a PR, including two near-CR (>90% decrease in M protein); 80% of patients demonstrated at least stable disease.58 Similarly, a European trial showed a response in eight (40%) of 20 patients, including two PR.59 Median PFS was 4 months and median OS was 11 months. The combination of As2O3, AA, and the proteasome inhibitor bortezomib was examined in a phase 2 trial for relapsed MM patients, following a report of in vitro synergy.60,61 Six (27%) of 22 patients showed a clinical response to the combination, including two PR; an additional nine (41%) patients had stable disease. Median PFS was 5 months, with 1-year PFS and OS rates of 34% and 74%, respectively. The specific contribution of As2O3 to the efficacy of these combination therapies cannot be isolated without randomized trials.
Myelodysplastic syndromes (MDS)
Several in vitro studies have demonstrated apoptosis in MDS cells exposed to As2O3. 27,62,63 MDS cells are under increased oxidative stress,64 which may confer As2O3 sensitivity. In two phase 2 clinical studies involving 191 patients, single-agent As2O3 was associated with hematologic improvement in 26–34% of patients with lower-risk (low or intermediate-1 international prognostic scoring system [IPSS] risk groups) MDS and 6–17% of those with higher-risk (intermediate-2 or high IPSS risk groups) MDS.65,66 Schiller et al. observed one (3%) CR and Vey et al. observed one (<1%) CR and one (<1%) PR in higher-risk patients. Major responses were observed in all hematologic lineages in both trials.
As2O3 plus thalidomide was investigated based on the hypothesis that this combination would target both the MDS clone and the bone marrow microenvironment. This combination was evaluated in 28 patients with MDS (12 lower-risk, 16 higher-risk).67 Seven patients (25%) responded, including one CR and two trilineage responses. Three of five patients with high baseline levels of the poor prognostic marker EVI1 responded to the combination; this observation was supported by experiments in vitro, which demonstrated greater As2O3 sensitivity in cells expressing high levels of EVI1. A subsequent trial of As2O3, thalidomide, and retinoic acid in 21 higher-risk patients demonstrated responses in 10 patients (48%), including one CR and one PR.68
Other hematologic malignancies
Several trials have demonstrated promising results in AML and human T-cell lymphotropic virus type I (HTLV-I)-associated adult T-cell leukemia-lymphoma (ATL). A phase 1/2 trial examined As2O3 in combination with low-dose cytarabine in 61 previously untreated AML patients aged 60 years or older.69 A CR was achieved in 21 patients (34%), including 15 (30%) of 50 patients with secondary AML. Median survival was 5.2 months, with a median follow-up of 9.8 months. A randomized phase 3 trial of this combination versus low-dose cytarabine alone is under way (www.clinicaltrials.gov). For relapsed/refractory ATL patients, As2O3 in combination with interferon alpha (IFNα) was examined in a small (N = 7) phase 2 trial.70 Despite significant toxicity that prevented completion of therapy, one patient achieved a CR lasting more than 32 months, and three patients achieved transient PR (median duration 1 month). In a separate report involving four patients with relapsed/refractory ATL, As2O3 plus IFNα produced one PR (duration of 8 months) and one stable disease, while As2O3 alone produced no response.71 In a recent phase 2 trial of As2O3 and IFNα plus zidovudine in 10 newly diagnosed ATL patients, a 100% response rate was observed, including seven CR.72 Most toxicities were grade 1 or 2; grade 3 neutropenia occurred in 3 patients and grade 3 thrombocytopenia was observed in 2 patients. As2O3 has demonstrated limited activity in non-Hodgkin lymphoma and chronic lymphocytic leukemia,73 and no clinical benefit in patients with acute lymphoblastic leukemia.74
Solid tumors
As2O3 is under investigation as treatment for a variety of solid tumors including bladder cancer, glioma, breast cancer, hepatocellular carcinoma (HCC), cervical cancer, colorectal cancer, esophageal cancer, germ cell tumors, liver cancer, lung cancer, and melanoma (www.clinicaltrials.gov). Limited clinical activity as a single agent has been reported in a small number of patients with HCC,75 melanoma,76,77 and renal cell carcinoma78; As2O3 in combination with chemotherapy has shown promising activity in osteosarcoma and Ewing sarcoma.79
Safety and toxicity
Arsenic is well known as a toxic agent. Inorganic arsenic has been classified by the US Department of Health and Human Services, the International Agency for Research on Cancer, and the US Environmental Protection Agency as a known carcinogen. Chronic exposure to low levels of environmental arsenic has been reported to increase the incidence of skin, liver, bladder, and lung cancers.80 Other potential signs of arsenic poisoning include peripheral neuropathy, cardiomyopathy, and renal failure.81
Despite its reputation as a poison, as a therapeutic entity As2O3 has been generally well tolerated. When administered intravenously at a dosage of 0.15 mg/kg/day, leukocytosis, gastrointestinal disorders (e.g., nausea, vomiting, diarrhea), fatigue, fever, headache, cough, and dyspnea are commonly observed. Common potentially serious toxicities include APL differentiation syndrome (APLDS) and electrocardiogram (ECG) abnormalities.82
APL differentiation syndrome
Previously known as “retinoic-acid syndrome,” APLDS may present during remission induction with ATRA or As2O3 therapies as a complex of signs and symptoms, including fever, dyspnea, hypotension, weight gain, acute renal failure, and lung infiltrates, and is usually treated with high-dose corticosteroids.83 In the pivotal trial of As2O3 in APL, APLDS occurred in 10 (25%) patients; in three of these patients, APLDS was considered to be serious.10 Therapy with As2O3 was briefly interrupted (for 1–5 days) in eight patients. Notably, all patients affected by APLDS achieved a CR. A similar incidence and severity of APLDS was reported with combined ATRA and As2O3; in the M. D. Anderson phase 2 trial, 13 patients (16%) developed APLDS, and all cases were successfully managed by withholding ATRA and administering corticosteroids.45 In a study comparing As2O3, ATRA, and the combination, the incidence of APLDS-associated hyperleukocytosis was not increased with As2O3 plus ATRA; however, one death in the combination arm was attributed to APLDS.43,44
Cardiac events
ECG abnormalities, including prolonged QT interval and complete atrioventricular block, have been reported with As2O3 treatment. QT prolongation (defined as ≥450 msec for males and ≥470 msec for females) was seen in 63% of patients in the pivotal trial and led to temporary discontinuation of As2O3 therapy in one patient (3%).10 In the phase 2 study by Ravandi et al., As2O3 was discontinued in five patients (6%) due to adverse cardiac events including atrial arrhythmias and myocardial infarction.45 ECG and electrolyte monitoring is recommended prior to and during arsenic therapy. Serum potassium levels should be kept above 4 meq/L and magnesium concentrations above 1.8 mg/dL.
Liver and kidney impairment
No significant hepatotoxicity was reported in the pivotal trial in relapsed patients.10 Hu et al. reported transient grade 1 or 2 liver dysfunction in 75% of patients during induction with ATRA and As2O3; no grade 3 or 4 toxicity was observed, and treatment was not discontinued in any patient.44 Ravandi et al. noted grade 3 elevations in liver enzymes in two of 82 patients (2%), but treatment was not discontinued.45 Grade 3 renal failure occurred in one (3%) patient during the pivotal trial10 and was reported in four (5%) patients by Ravandi et al.45
Secondary malignancies
Carcinogenesis is a major concern associated with long-term exposure to arsenic. In the study published in December 2008 in the Proceedings of the National Academy of Sciences (PNAS) on long-term efficacy and safety of ATRA/As2O3-based therapy in newly diagnosed APL patients who started being accrued to the trial in 200144, no secondary carcinoma, including skin cancer, was observed. One male transiently tested positive for carcinoembryogenic antigen (CEA), and a mild, unsustained increase in CA125 in a female patient was recorded. Moreover, arsenic concentrations in the urine of patients who had ceased As2O3 treatment for 24 months were below the safety limits recommended by government agencies in several countries or regions.
Secondary malignancies were not reported in the short-term trials US registration trial10 or the phase 2 trial of As2O3 plus ATRA.45 Reduced exposure to or avoidance of chemotherapy in As2O3 patients may reduce the risk of secondary MDS and AML, which have been previously associated with APL treatment.84
Arsenic retention and long-term toxicity
The trial by Hu et al. included extensive screening of APL patients for long-term effects of arsenic exposure following completion of therapy.44 Laboratory studies (blood counts, electrolyte panels, urinalysis, liver function tests, and serum tumor marker analysis) and physical examinations (ECGs, echocardiograms, chest x-rays, dermatologic and neurologic consults, and nerve conduction velocity tests) were performed in 33 patients during therapy and at a minimum of 2 years after receiving the last dose of As2O3. At the final follow-up, physical exam and laboratory results were similar between patients and healthy controls. No tumors or skin lesions were detected. ECGs, echocardiograms, and chest x-rays were normal. Toxicology analyses in these patients found that As2O3 levels in plasma and urine dropped after termination of As2O3 treatment to levels only slightly higher than those in healthy controls, and were within recommended US safety limits.
Pharmacokinetics
In a study of 12 Japanese patients receiving As2O3 (0.15 mg/kg IV over 2 hours), the mean maximum plasma concentration of inorganic arsenic (AsIII and AsV) of 22.6 ± 11.4 ng/mL occurred at completion of the infusion, then declined biphasically, with an initial distribution phase followed by an elimination phase with a terminal half-life of 17 hours.85 The volume of distribution was large (55.9 L/kg), indicating extensive distribution throughout the body. In a separate case report of a patient with relapsed APL and meningeal infiltration, As2O3 was observed to cross the blood-brain barrier when administered intravenously; arsenic levels in cerebrospinal fluid were approximately 14% of levels in blood.86
The major arsenic metabolites identified in humans are the methylated arsenics, methylarsonic acid (MAAV) and dimethylarsinic acid (DMAAV).87–89 In the Japanese study, plasma levels of MAAV and DMAAV increased gradually over 24 hours after the first administration of As2O3, reaching peak levels of 3.1 ± 1.6 ng/mL and 5.4 ± 2.9 ng/mL, respectively.85 The contribution of these metabolites to the therapeutic effect of As2O3 is unknown. In APL, chronic lymphocytic leukemia, and leukemia/lymphoma cell lines, methylated arsenic derivates were shown to induce apoptosis, though they did not induce differentiation in APL cells.90 Plasma concentrations of inorganic arsenic did not increase with repeated administration; however, the concentrations of both major metabolites were approximately four times higher during week 4 of treatment compared with day 1.85 Arsenic content in hair and nails also increased gradually with repeated treatment and reached levels 5–7 times higher than pretreatment levels.8
Approximately 60% of the daily dose of arsenic is excreted in the urine, including inorganic and methylated arsenic; other pathways of excretion, such as biliary, may also contribute to As2O3 elimination.85,87
Conclusions
As2O3 has demonstrated remarkable efficacy in APL. The use of As2O3 in induction or consolidation strategies reduces the relapse rate and improves survival in patients with APL, especially in high-risk patients.43,44,46 The addition of As2O3 to ATRA/chemotherapy regimens may allow a reduction in chemotherapy exposure and associated toxicities without compromising cure rates.47 In select patients, chemotherapy may be eliminated altogether, providing an alternative treatment option for patients who cannot tolerate anthracyclines.45 Although As2O3 has demonstrated less robust efficacy in other malignancies, promising activity has been demonstrated when As2O3 is combined with other agents. As2O3 is generally well tolerated, both as a single agent and in combination, with manageable adverse effects and no documented evidence of long-term toxicities. Additional comparative trials will be necessary to determine which combinations of available therapies provide the greatest benefit with the least toxicity in patients with APL.
Practice Points
At the Sidney Kimmel Comprehensive Cancer Center (SKCCC) at Johns Hopkins University, patients with APL are treated as follows:
Whenever possible, they are enrolled in clinical trials.
Outside of clinical trials, patients should be treated according to a published treatment protocol, from induction through maintenance and post-maintenance monitoring.
Remission can be achieved with ATRA plus an anthracycline. The use of 60 days of ATRA (rather than administration of ATRA until normalization of hemogram) may lead to a higher level of PML/RARα molecular negativity.
If WBC ≥20 × 109/L, hydroxyurea is added to maintain WBC <20 × 109/L, with dexamethasone (10 mg IV, twice daily for 14 days) as prophylaxis against APLDS.
At SKCCC, we believe that an As2O3-containing post-remission (i.e., consolidation) chemotherapy regimen results in high relapse-free and overall survivals (Table 2).
In patients unable to tolerate anthracyclines, remission can be induced with ATRA plus As2O3 daily until absence of leukemia in bone marrow is documented, or for a maximum of 60 days.
Maintenance therapy appears important and should be completed according to the elected treatment protocol.
Treatment of relapsed disease (determined by two consecutive positive PML/RARα findings by qualitative RT-PCR within 4 weeks) should be individualized per patient.
Research Agenda
There are 87 recently completed or ongoing clinical trials listed on www.clinicaltrials.gov evaluating As2O3 alone or in combination with other agents for treatment of APL, AML, multiple myeloma, MDS, primary myelofibrosis, hepatocellular carcinoma, metastatic melanoma, CNS tumors, and breast, lung, colorectal, and kidney cancers.
In APL, the role of chemotherapeutic CNS prophylaxis, in all patients or high-risk patients, needs to be addressed in clinical trials.
In APL, the role of maintenance therapy, particularly in patients who are treated with As2O3, also needs to be addressed by future research.
Acknowledgements
The authors thank Jill Luer, PharmD, and Janis Leonoudakis, PhD, of Helix for providing medical writing assistance supported by Cephalon, Inc. The authors, however, are fully responsible for content and editorial decisions for this manuscript.
Footnotes
Conflict of interest statement
Dr. Gore: Received As2O3 from Cephalon, Inc., as part of a phase 2 trial of As2O3 in consolidation therapy of APL.
Dr. Emadi declares no conflict of interest.
References
- 1.Waxman S, Anderson KC. History of the development of arsenic derivatives in cancer therapy. Oncologist. 2001;6 Suppl 2:3–10. doi: 10.1634/theoncologist.6-suppl_2-3. [DOI] [PubMed] [Google Scholar]
- 2.Kwong YL, Todd D. Delicious poison: arsenic trioxide for the treatment of leukemia. Blood. 1997;89:3487–3488. [PubMed] [Google Scholar]
- 3.Sears DA. History of the treatment of chronic myelocytic leukemia. Am J Med Sci. 1988;296:85–86. doi: 10.1097/00000441-198808000-00001. [DOI] [PubMed] [Google Scholar]
- 4.Cutler EG, Bradford EH. Action of iron, cod-liver oil, and arsenic on the globular richness of the blood. Am J Med Sci. 1878;75:74–84. [Google Scholar]
- 5.Wang ZY, Chen Z. Acute promyelocytic leukemia: from highly fatal to highly curable. Blood. 2008;111:2505–2515. doi: 10.1182/blood-2007-07-102798. [DOI] [PubMed] [Google Scholar]
- 6.Sun HD, Ma L, Hu Z, et al. Arsenic trioxide treated 32 cases of acute promyelocytic leukemia. Chin J Integrated Tradit West Med. 1992;12:170–172. [Google Scholar]
- 7.Zhang P, Wang SY, Hu XH. Arsenic trioxide treated 72 cases of acute promyelocytic leukemia. Chin J Hematol. 1996;17:58–62. [Google Scholar]
- 8.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;89:3354–3360. [PubMed] [Google Scholar]
- 9.Soignet SL, Maslak P, Wang ZG, et al. Complete remission after treatment of acute promyelocytic leukemia with arsenic trioxide. N Engl J Med. 1998;339:1341–1348. doi: 10.1056/NEJM199811053391901. [DOI] [PubMed] [Google Scholar]
- 10.Soignet SL, Frankel SR, Douer D, et al. United States multicenter study of arsenic trioxide in relapsed acute promyelocytic leukemia. J Clin Oncol. 2001;19:3852–3860. doi: 10.1200/JCO.2001.19.18.3852. [DOI] [PubMed] [Google Scholar]
- 11.Sanz MA, Martin G, Gonzalez M, et al. Risk-adapted treatment of acute promyelocytic leukemia with all-trans-retinoic acid and anthracycline monochemotherapy: a multicenter study by the PETHEMA group. Blood. 2004;103:1237–1243. doi: 10.1182/blood-2003-07-2462. [DOI] [PubMed] [Google Scholar]
- 12.Ades L, Chevret S, Raffoux E, et al. Is cytarabine useful in the treatment of acute promyelocytic leukemia? Results of a randomized trial from the European Acute Promyelocytic Leukemia Group. J Clin Oncol. 2006;24:5703–5710. doi: 10.1200/JCO.2006.08.1596. [DOI] [PubMed] [Google Scholar]
- 13.Asou N, Kishimoto Y, Kiyoi H, et al. A randomized study with or without intensified maintenance chemotherapy in patients with acute promyelocytic leukemia who have become negative for PML-RARalpha transcript after consolidation therapy: the Japan Adult Leukemia Study Group (JALSG) APL97 study. Blood. 2007;110:59–66. doi: 10.1182/blood-2006-08-043992. [DOI] [PubMed] [Google Scholar]
- 14.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;89:3345–3353. [PubMed] [Google Scholar]
- 15.Miller WH, Jr, Schipper HM, Lee JS, et al. Mechanisms of action of arsenic trioxide. Cancer Res. 2002;62:3893–3903. [PubMed] [Google Scholar]
- 16.Aposhian HV. Biochemical toxicology of arsenic. In: Hodgson E, Bend JR, Philpot RM, editors. Reviews in Biochemical Toxicology. Vol. 10. New York, NY: Elsevier Science Publishing; 1989. pp. 265–299. [Google Scholar]
- 17.Spuches AM, Kruszyna HG, Rich AM, et al. Thermodynamics of the As(III)-thiol interaction: arsenite and monomethylarsenite complexes with glutathione, dihydrolipoic acid, and other thiol ligands. Inorg Chem. 2005;44:2964–2972. doi: 10.1021/ic048694q. [DOI] [PubMed] [Google Scholar]
- 18.Chakraborti PK, Garabedian MJ, Yamamoto KR, et al. Role of cysteines 640, 656, and 661 in steroid binding to rat glucocorticoid receptors. J Biol Chem. 1992;267:11366–11373. [PubMed] [Google Scholar]
- 19.Wang ZG, Ruggero D, Ronchetti S, et al. PML is essential for multiple apoptotic pathways. Nat Genet. 1998;20:266–272. doi: 10.1038/3073. [DOI] [PubMed] [Google Scholar]
- 20.Dyck JA, Maul GG, Miller WH, Jr, et al. A novel macromolecular structure is a target of the promyelocyte-retinoic acid receptor oncoprotein. Cell. 1994;76:333–343. doi: 10.1016/0092-8674(94)90340-9. [DOI] [PubMed] [Google Scholar]
- 21.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;88:1052–1061. [PubMed] [Google Scholar]
- 22.List A, Beran M, DiPersio J, et al. Opportunities for Trisenox ® (arsenic trioxide) in the treatment of myelodysplastic syndromes. Leukemia. 2003;17:1499–1507. doi: 10.1038/sj.leu.2403021. [DOI] [PubMed] [Google Scholar]
- 23.Kluck RM, Bossy-Wetzel E, Green DR, et al. The release of cytochrome c from mitochondria: a primary site for Bcl-2 regulation of apoptosis. Science. 1997;275:1132–1136. doi: 10.1126/science.275.5303.1132. [DOI] [PubMed] [Google Scholar]
- 24.Zou H, Li Y, Liu X, et al. An APAF-1.cytochrome c multimeric complex is a functional apoptosome that activates procaspase-9. J Biol Chem. 1999;274:11549–11556. doi: 10.1074/jbc.274.17.11549. [DOI] [PubMed] [Google Scholar]
- 25.Saleh A, Srinivasula SM, Acharya S, et al. Cytochrome c and dATP-mediated oligomerization of Apaf-1 is a prerequisite for procaspase-9 activation. J Biol Chem. 1999;274:17941–17945. doi: 10.1074/jbc.274.25.17941. [DOI] [PubMed] [Google Scholar]
- 26.Hengartner MO. The biochemistry of apoptosis. Nature. 2000;407:770–776. doi: 10.1038/35037710. [DOI] [PubMed] [Google Scholar]
- 27.Rojewski MT, Baldus C, Knauf W, et al. Dual effects of arsenic trioxide (As2O3) on non-acute promyelocytic leukaemia myeloid cell lines: induction of apoptosis and inhibition of proliferation. Br J Haematol. 2002;116:555–563. doi: 10.1046/j.0007-1048.2001.03298.x. [DOI] [PubMed] [Google Scholar]
- 28.Kanzawa T, Kondo Y, Ito H, et al. Induction of autophagic cell death in malignant glioma cells by arsenic trioxide. Cancer Res. 2003;63:2103–2108. [PubMed] [Google Scholar]
- 29.Rojewski MT, Korper S, Schrezenmeier H. Arsenic trioxide therapy in acute promyelocytic leukemia and beyond: from bench to bedside. Leuk Lymphoma. 2004;45:2387–2401. doi: 10.1080/10428190412331272686. [DOI] [PubMed] [Google Scholar]
- 30.Qin DY, Huang R, Wu T. In vitro arsenic trioxide induces apoptosis in T cells of asthmatic patients by a Bcl-2 related mechanism. Yao Xue Xue Bao. 2008;43:35–43. [PubMed] [Google Scholar]
- 31.Park WH, Seol JG, Kim ES, et al. Arsenic trioxide-mediated growth inhibition in MC/CAR myeloma cells via cell cycle arrest in association with induction of cyclin-dependent kinase inhibitor, p21, and apoptosis. Cancer Res. 2000;60:3065–3071. [PubMed] [Google Scholar]
- 32.Park JW, Choi YJ, Jang MA, et al. Arsenic trioxide induces G2/M growth arrest and apoptosis after caspase-3 activation and bcl-2 phosphorylation in promonocytic U937 cells. Biochem Biophys Res Commun. 2001;286:726–734. doi: 10.1006/bbrc.2001.5416. [DOI] [PubMed] [Google Scholar]
- 33.Larochette N, Decaudin D, Jacotot E, et al. Arsenite induces apoptosis via a direct effect on the mitochondrial permeability transition pore. Exp Cell Res. 1999;249:413–421. doi: 10.1006/excr.1999.4519. [DOI] [PubMed] [Google Scholar]
- 34.Nishikawa M. Reactive oxygen species in tumor metastasis. Cancer Lett. 2008;266:53–59. doi: 10.1016/j.canlet.2008.02.031. [DOI] [PubMed] [Google Scholar]
- 35.Dai J, Weinberg RS, Waxman S, et al. Malignant cells can be sensitized to undergo growth inhibition and apoptosis by arsenic trioxide through modulation of the glutathione redox system. Blood. 1999;93:268–277. [PubMed] [Google Scholar]
- 36.Sen CK. Redox signaling and the emerging therapeutic potential of thiol antioxidants. Biochem Pharmacol. 1998;55:1747–1758. doi: 10.1016/s0006-2952(97)00672-2. [DOI] [PubMed] [Google Scholar]
- 37.Chou WC, Jie C, Kenedy AA, et al. Role of NADPH oxidase in arsenic-induced reactive oxygen species formation and cytotoxicity in myeloid leukemia cells. Proc Natl Acad Sci U S A. 2004;101:4578–4583. doi: 10.1073/pnas.0306687101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.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;275:36062–36066. doi: 10.1074/jbc.M007204200. [DOI] [PubMed] [Google Scholar]
- 39.Dilda PJ, Perrone GG, Philp A, et al. Insight into the selectivity of arsenic trioxide for acute promyelocytic leukemia cells by characterizing Saccharomyces cerevisiae deletion strains that are sensitive or resistant to the metalloid. Int J Biochem Cell Biol. 2008;40:1016–1029. doi: 10.1016/j.biocel.2007.11.002. [DOI] [PubMed] [Google Scholar]
- 40.National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Acute Myeloid Leukemia. Version 1.2009. Available at: http://www.nccn.org/professionals/physician_gls/PDF/aml.pdf.
- 41.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;107:2627–2632. doi: 10.1182/blood-2005-08-3532. [DOI] [PubMed] [Google Scholar]
- 42.Ghavamzadeh A, Alimoghaddam K, Ghaffari SH, et al. Treatment of new cases of acute promyelocytic leukemia by arsenic trioxide without ATRA +/− chemotherapy and long term follow up of patients. Blood (ASH Annual Meeting Abstracts) 2008;112:140. [Google Scholar]
- 43.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;101:5328–5335. doi: 10.1073/pnas.0400053101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.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;106:3342–3347. doi: 10.1073/pnas.0813280106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ravandi F, Estey E, Jones D, et al. Effective treatment of acute promyelocytic leukemia with all-trans-retinoic acid, arsenic trioxide, and gemtuzumab ozogamicin. J Clin Oncol. 2009;27:504–510. doi: 10.1200/JCO.2008.18.6130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Powell BL, Moser B, Stock W, et al. Effect of consolidation with arsenic trioxide (As2O3) on event-free survival (EFS) and overall survival (OS) among patients with newly diagnosed acute promyelocytic leukemia (APL): North American Intergroup Protocol C9710. J Clin Oncol. 2007;25:18S. abstract 2. [Google Scholar]
- 47.Gore SD, Gojo I, Morris L, et al. Single cycle of Arsenic Trioxide-based consolidation chemotherapy spares anthracycline exposure in the primary management of acute promyelocytic leukemia. J Clin Oncol. 2010;28:1047–1053. doi: 10.1200/JCO.2009.25.5158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Rousselot P, Labaume S, Marolleau JP, et al. Arsenic trioxide and melarsoprol induce apoptosis in plasma cell lines and in plasma cells from myeloma patients. Cancer Res. 1999;59:1041–1048. [PubMed] [Google Scholar]
- 49.Munshi NC, Tricot G, Desikan R, et al. Clinical activity of arsenic trioxide for the treatment of multiple myeloma. Leukemia. 2002;16:1835–1837. doi: 10.1038/sj.leu.2402599. [DOI] [PubMed] [Google Scholar]
- 50.Hussein MA, Saleh M, Ravandi F, et al. Phase 2 study of arsenic trioxide in patients with relapsed or refractory multiple myeloma. Br J Haematol. 2004;125:470–476. doi: 10.1111/j.1365-2141.2004.04941.x. [DOI] [PubMed] [Google Scholar]
- 51.Rousselot P, Larghero J, Arnulf B, et al. A clinical and pharmacological study of arsenic trioxide in advanced multiple myeloma patients. Leukemia. 2004;18:1518–1521. doi: 10.1038/sj.leu.2403424. [DOI] [PubMed] [Google Scholar]
- 52.Grad JM, Bahlis NJ, Reis I, et al. Ascorbic acid enhances arsenic trioxide-induced cytotoxicity in multiple myeloma cells. Blood. 2001;98:805–813. doi: 10.1182/blood.v98.3.805. [DOI] [PubMed] [Google Scholar]
- 53.Bahlis NJ, Cafferty-Grad J, Jordan-McMurry I, et al. Feasibility and correlates of arsenic trioxide combined with ascorbic acid-mediated depletion of intracellular glutathione for the treatment of relapsed/refractory multiple myeloma. Clin Cancer Res. 2002;8:3658–3668. [PubMed] [Google Scholar]
- 54.Karasavvas N, Carcamo JM, Stratis G, et al. Vitamin C protects HL60 and U266 cells from arsenic toxicity. Blood. 2005;105:4004–4012. doi: 10.1182/blood-2003-03-0772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Borad MJ, Swift R, Berenson JR. Efficacy of melphalan, arsenic trioxide, and ascorbic acid combination therapy (MAC) in relapsed and refractory multiple myeloma. Leukemia. 2005;19:154–156. doi: 10.1038/sj.leu.2403541. [DOI] [PubMed] [Google Scholar]
- 56.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;135:174–183. doi: 10.1111/j.1365-2141.2006.06280.x. [DOI] [PubMed] [Google Scholar]
- 57.Qazilbash MH, Saliba RM, Nieto Y, et al. Arsenic trioxide with ascorbic acid and high-dose melphalan: results of a phase II randomized trial. Biol Blood Marrow Transplant. 2008;14:1401–1407. doi: 10.1016/j.bbmt.2008.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.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;23:263–272. doi: 10.1385/MO:23:2:263. [DOI] [PubMed] [Google Scholar]
- 59.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;91:1722–1723. [PubMed] [Google Scholar]
- 60.Campbell RA, Chen H, Zhu D, et al. Arsenic trioxide shows synergistic anti-myeloma effects when combined with bortezomib and melphalan in vitro and helps overcome resistance of multiple myeloma cells to these treatments in vivo. Blood (ASH Annual Meeting Abstracts) 2004;104:2467. [Google Scholar]
- 61.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;13:1762–1768. doi: 10.1158/1078-0432.CCR-06-1812. [DOI] [PubMed] [Google Scholar]
- 62.Zhang W, Ohnishi K, Shigeno K, et al. The induction of apoptosis and cell cycle arrest by arsenic trioxide in lymphoid neoplasms. Leukemia. 1998;12:1383–1391. doi: 10.1038/sj.leu.2401112. [DOI] [PubMed] [Google Scholar]
- 63.Zhu XH, Shen YL, Jing YK, et al. Apoptosis and growth inhibition in malignant lymphocytes after treatment with arsenic trioxide at clinically achievable concentrations. J Natl Cancer Inst. 1999;91:772–778. doi: 10.1093/jnci/91.9.772. [DOI] [PubMed] [Google Scholar]
- 64.Peddie CM, Wolf CR, McLellan LI, et al. Oxidative DNA damage in CD34+myelodysplastic cells is associated with intracellular redox changes and elevated plasma tumour necrosis factor-alpha concentration. Br J Haematol. 1997;99:625–631. doi: 10.1046/j.1365-2141.1997.4373247.x. [DOI] [PubMed] [Google Scholar]
- 65.Schiller GJ, Slack J, Hainsworth JD, et al. Phase II multicenter study of arsenic trioxide in patients with myelodysplastic syndromes. J Clin Oncol. 2006;24:2456–2464. doi: 10.1200/JCO.2005.03.7903. [DOI] [PubMed] [Google Scholar]
- 66.Vey N, Bosly A, Guerci A, et al. Arsenic trioxide in patients with myelodysplastic syndromes: a phase II multicenter study. J Clin Oncol. 2006;24:2465–2471. doi: 10.1200/JCO.2005.03.9503. [DOI] [PubMed] [Google Scholar]
- 67.Raza A, Buonamici S, Lisak L, et al. Arsenic trioxide and thalidomide combination produces multi-lineage hematological responses in myelodysplastic syndromes patients, particularly in those with high pre-therapy EVI1 expression. Leuk Res. 2004;28:791–803. doi: 10.1016/j.leukres.2003.11.018. [DOI] [PubMed] [Google Scholar]
- 68.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;32:251–254. doi: 10.1016/j.leukres.2007.05.025. [DOI] [PubMed] [Google Scholar]
- 69.Roboz GJ, Ritchie EK, Curcio T, et al. Arsenic trioxide and low-dose cytarabine in older patients with untreated acute myeloid leukemia, excluding acute promyelocytic leukemia. Cancer. 2008;113:2504–2511. doi: 10.1002/cncr.23855. [DOI] [PubMed] [Google Scholar]
- 70.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;5:130–134. doi: 10.1038/sj.thj.6200374. [DOI] [PubMed] [Google Scholar]
- 71.Ishitsuka K, Suzumiya J, Aoki M, et al. Therapeutic potential of arsenic trioxide with or without interferon-alpha for relapsed/refractory adult T-cell leukemia/lymphoma. Haematologica. 2007;92:719–720. doi: 10.3324/haematol.10703. [DOI] [PubMed] [Google Scholar]
- 72.Kchour G, Tarhini M, Kooshyar MM, et al. Phase II study of the efficacy and safety of the combination of arsenic trioxide, interferon alpha, and zidovudine in newly diagnosed chronic ATL. Blood. 2009;113:6528–6532. doi: 10.1182/blood-2009-03-211821. [DOI] [PubMed] [Google Scholar]
- 73.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;27:11–16. doi: 10.1002/hon.870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Litzow MR, Lee S, Bennett JM, et al. A phase II trial of arsenic trioxide for relapsed and refractory acute lymphoblastic leukemia. Haematologica. 2006;91:1105–1108. [PubMed] [Google Scholar]
- 75.Lin CC, Hsu C, Hsu CH, et al. Arsenic trioxide in patients with hepatocellular carcinoma: a phase II trial. Invest New Drugs. 2007;25:77–84. doi: 10.1007/s10637-006-9004-9. [DOI] [PubMed] [Google Scholar]
- 76.Kim KB, Bedikian AY, Camacho LH, et al. A phase II trial of arsenic trioxide in patients with metastatic melanoma. Cancer. 2005;104:1687–1692. doi: 10.1002/cncr.21386. [DOI] [PubMed] [Google Scholar]
- 77.Tarhini AA, Kirkwood JM, Tawbi H, et al. Safety and efficacy of arsenic trioxide for patients with advanced metastatic melanoma. Cancer. 2008;112:1131–1138. doi: 10.1002/cncr.23284. [DOI] [PubMed] [Google Scholar]
- 78.Vuky J, Yu R, Schwartz L, et al. Phase II trial of arsenic trioxide in patients with metastatic renal cell carcinoma. Invest New Drugs. 2002;20:327–330. doi: 10.1023/a:1016270206374. [DOI] [PubMed] [Google Scholar]
- 79.Guo W, Tang XD, Tang S, et al. [Preliminary report of combination chemotherapy including Arsenic trioxide for stage III osteosarcoma and Ewing sarcoma] Zhonghua Wai Ke Za Zhi. 2006;44:805–808. [PubMed] [Google Scholar]
- 80.Agency for Toxic Substances and Disease Registry. Public Health Statement: Arsenic. Atlanta, GA: Centers for Disease Control and Prevention, US Dept of Health and Human Services Division of Toxicology and Environmental Medicine; 2007. Publication CAS# 7440-38-2. [Google Scholar]
- 81.Agency for Toxic Substances, Disease Registry. Case Studies In Environmental Medicine (CSEM): Arsenic Toxicity. Atlanta, GA: Centers for Disease Control and Prevention, US Dept of Health and Human Services Division of Toxicology and Environmental Medicine; 2000. Publication Case studies in environmental medicine (CSEM): arsenic toxicity. [Google Scholar]
- 82.Trisenox™ (arsenic trioxide) [prescribing information] Frazer, PA: Cephalon, Inc; [Google Scholar]
- 83.Sanz MA, Grimwade D, Tallman MS, et al. Management of acute promyelocytic leukemia: Recommendations from an expert panel on behalf of the European LeukemiaNet. Blood. 2009;113:1875–1891. doi: 10.1182/blood-2008-04-150250. [DOI] [PubMed] [Google Scholar]
- 84.Garcia-Manero G, Kantarjian HM, Kornblau S, et al. Therapy-related myelodysplastic syndrome or acute myelogenous leukemia in patients with acute promyelocytic leukemia (APL) Leukemia. 2002;16:1888. doi: 10.1038/sj.leu.2402616. [DOI] [PubMed] [Google Scholar]
- 85.Fujisawa S, Ohno R, Shigeno K, et al. Pharmacokinetics of arsenic species in Japanese patients with relapsed or refractory acute promyelocytic leukemia treated with arsenic trioxide. Cancer Chemother Pharmacol. 2007;59:485–493. doi: 10.1007/s00280-006-0288-4. [DOI] [PubMed] [Google Scholar]
- 86.Knipp S, Gattermann N, Schapira M, et al. Arsenic in the cerebrospinal fluid of a patient receiving arsenic trioxide for relapsed acute promyelocytic leukemia with CNS involvement. Leuk Res. 2007;31:1585–1587. doi: 10.1016/j.leukres.2007.03.007. [DOI] [PubMed] [Google Scholar]
- 87.Wang Z, Zhou J, Lu X, et al. Arsenic speciation in urine from acute promyelocytic leukemia patients undergoing arsenic trioxide treatment. Chem Res Toxicol. 2004;17:95–103. doi: 10.1021/tx0341714. [DOI] [PubMed] [Google Scholar]
- 88.Fukai Y, Hirata M, Ueno M, et al. Clinical pharmacokinetic study of arsenic trioxide in an acute promyelocytic leukemia (APL) patient: speciation of arsenic metabolites in serum and urine. Biol Pharm Bull. 2006;29:1022–1027. doi: 10.1248/bpb.29.1022. [DOI] [PubMed] [Google Scholar]
- 89.Aposhian HV. Enzymatic methylation of arsenic species and other new approaches to arsenic toxicity. Annu Rev Pharmacol Toxicol. 1997;37:397–419. doi: 10.1146/annurev.pharmtox.37.1.397. [DOI] [PubMed] [Google Scholar]
- 90.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;63:1853–1859. [PubMed] [Google Scholar]