Inorganic arsenic trioxide (As2O3) and the organic arsenical, melarsoprol, were recently shown to inhibit growth and induce apoptosis in NB4 acute promyelocytic leukemia (APL) and chronic B-cell leukemia cell lines, respectively. As2O3 has been proposed to principally target PML and PML-RAR proteins in APL cells. We investigated the activity of As2O3 and melarsoprol in a broader context encompassing various myeloid leukemia cell lines, including the APL cell line NB4-306 (a retinoic acid–resistant cell line derived from NB4 that no longer expresses the intact PML-RAR fusion protein), HL60, KG-1, and the myelomonocytic cell line U937. To examine the role of PML in mediating arsenical activity, we also tested these agents using murine embryonic fibroblasts (MEFs) and bone marrow (BM) progenitors in which the PML gene had been inactivated by homologous recombination. Unexpectedly, we found that both compounds inhibited cell growth, induced apoptosis, and downregulated bcl-2 protein in all cell lines tested. Melarsoprol was more potent than As2O3 at equimolar concentrations ranging from 10−7 to 10−5 mol/L. As2O3 relocalized PML and PML-RAR onto nuclear bodies, which was followed by PML degradation in NB4 as well as in HL60 and U937 cell lines. Although melarsoprol was more potent in inhibiting growth and inducing apoptosis, it did not affect PML and/or PML-RAR nuclear localization. Moreover, both As2O3 and melarsoprol comparably inhibited growth and induced apoptosis of PML+/+ and PML−/− MEFs, and inhibited colony-forming unit erythroid (CFU-E) and CFU granulocyte-monocyte formation in BM cultures of PML+/+ and PML−/− progenitors. Together, these results show that As2O3 and melarsoprol inhibit growth and induce apoptosis independent of both PML and PML-RAR expression in a variety of myeloid leukemia cell lines, and suggest that these agents may be more broadly used for treatment of leukemias other than APL.

© 1998 by The American Society of Hematology.

THE USE OF ORGANIC arsenicals was pioneered by Paul Ehrlich, who showed that Salvarsan (arsphenamine) was an extremely effective antispirochetal agent.1 Most organic arsenicals have been superseded by other drugs, but melarsoprol remains an important therapy for certain tropical diseases, such as African sleeping sickness.2 Moreover, potassium arsenite (administered orally as Fowler’s solution) was widely used in the West for controlling leukocytosis in patients with chronic myelocytic leukemia in the early 1900s.3 However, this therapy gradually waned with the advent of radiotherapy in the 1930s and the identification of potent antileukemic agents after the Second World War.

Recently, inorganic arsenic trioxide (As2O3) was reported to induce complete remission in a high proportion of patients with refractory acute promyelocytic leukemia (APL).4-6 Chen et al showed that As2O3 induced apoptosis in the APL cell line, NB4, which may be mediated through downregulation of bcl-2, and modulation of the PML-RARα fusion protein, the specific product of the t(15;17) of APL,7-10 and/or the PML protein.11 More recently, the antileukemic effects of As2O3 were proposed to be directly mediated by its ability to induce the relocalization and degradation of PML, as well as the degradation of PML-RARα in APL cells.12-14 We found that the organic arsenical, melarsoprol, exhibited effects similar to As2O3 on chronic B-cell leukemic cell lines,15 a finding which suggested that the cytotoxic action of this drug may not be dependent on PML or PML-RARα expression.

In the present study we have explored the effects of As2O3 and melarsoprol in a variety of myeloid leukemia cell lines in which the PML-RARα fusion protein is or is not expressed. Furthermore, to examine whether PML mediated the growth inhibitory and apoptotic activities of these compounds, we also evaluated both As2O3 and melarsoprol in cells from mice in which PML had been disrupted by homologous recombination, using both murine embryonic fibroblasts (MEFs) and bone marrow (BM) progenitors from PML−/− mice.16-18 

We unexpectedly found that As2O3 and melarsoprol can inhibit growth and induce apoptosis in a PML and PML-RARα independent manner, thus suggesting that both compounds may be useful for treatment of leukemias other than APL.

Reagents.

Arsenic trioxide (As2O3) was purchased from Sigma Chemicals, Inc (Milwaukee, WI). A stock solution was made with phosphate-buffered saline (PBS) at a concentration of 10−2 mol/L. Melarsoprol (Arsobal; purchased from Rhône Poulenc Rorer, Paris, France) was dissolved in propylene glycol at a stock concentration of 0.09 mol/L (36 mg/mL) and stored at room temperature in the dark. Both solutions were diluted to working concentrations before use.

Cell culture and cell viability assays.

Cell lines, including NB4 cells, the RA-resistant NB4 subclone 306, HL60, U937, and KG-1, were cultured in RPMI 1640 medium containing 10% fetal bovine serum (FBS), 2 mmol/L L-glutamine, penicillin G (100 U/mL), and streptomycin (100 μg/mL). MEFs were isolated from wild-type and PML−/− mouse embryos at 13.5 days of gestation and cultured in Dulbecco’s Modification of Eagle’s Medium (DMEM) supplemented with 20% FBS. Early passage MEFs were used. For cell growth and viability assays, NB4, NB4.306, HL60, U937, and KG-1 cells were seeded at an initial concentration of 1 × 105 cells/mL, and incubated in RPMI 1640 medium plus 10% FBS with or without As2O3 or melarsoprol. Cell growth and viability were assessed in triplicate by an automatic counter (Zb1; Coulter Electronic, Miami, FL) and trypan blue exclusion at indicated days. The percentage of viable cells in treated samples was divided by the average viability of the untreated samples (controls). Each growth curve was repeated at least three times.

In situ terminal deoxynucleotidyl transferase mediated dUTP-biotin nick-end labeling (TUNEL).

The procedure for identification of programmed cell death in situ was performed as described by Gavrieli et al,19 with minor modifications. In brief, NB4, HL60, and U937 cells were treated with reagents described above for 24 hours, and then pelleted onto slides by centrifugation. The slides were quickly air-dried and fixed in 4% paraformaldehyde for 30 minutes at room temperature and washed with PBS. To quench endogenous peroxidase, cells were treated with 0.1% H2O2 in PBS for 15 minutes at room temperature and washed with dH2O and TdT buffer. The labeling reaction was performed with TdT-biotin dUTP labeling mix for 1 hour in a humid chamber and stopped in 2× SSC (0.3 mol/L NaCl, 0.03 mol/L sodium citrate, pH 7.0) for 15 minutes. The slides were incubated with Vectastain ABC reagent (Vector Laboratories, Burlingame, CA) for 30 minutes after blocking with 2% bovine serum albumin (BSA) in PBS. TUNEL-positive cells were visualized by 3, 3′-diaminobenzidene (DAB) staining and counterstaining with hematoxylin. The same assay was also performed with MEFs. In this case, 1 × 104 cells were plated in each chamber of Permanox slides (Nalge Nunc International, Naperville, IL) in 1 mL of DMEM medium with or without the above-mentioned reagents. After a 24-hour incubation, cells were fixed and the same procedures described above were performed. At least 300 cells were counted for each condition.

Western blot analysis.

Cells were lysed in a lysis buffer containing 20 mmol/L Tris-HCl, 1 mmol/L EGTA, 50 mmol/L NaVO4, 50 mmol/L NaF, 0.01 U/mL aprotinin, 10 μg/mL leupeptin, 10 μg/mL pepstatin, 1 mmol/L elastinal, and 1 mmol/L phenylmethylsulfonyl fluoride (PMSF) (all from Sigma). The lysates were then sonicated using a ultrasonic homogenizer (4710 series; Cole Parmer Instruments, Chicago, IL), centrifuged at 7,500g (Sorvall Instruments, Newtown, CT), and the protein content of the lysates was determined (BioRad Protein Assay Kit I, Melville, NY) at 595 nm with a BSA standard. A sample buffer containing 10% glycerol, 0.4% sodium dodecyl sulfate (SDS), 0.3% bromphenol blue, 0.2% pyronin Y, in 1× stacking buffer (Tris base 0.5 mol/L, 0.8% SDS), 20% 2-mercaptoethanol, was added to the cell lysates which were subsequently heat-denatured at 95°C for 3 minutes. Ten micrograms per lane of protein was loaded on an SDS-polyacrylamide gel containing 12.5% polyacrylamide, and size-fractionated by electrophoresis. Proteins were electroblotted onto Immobilon-P PVDF transfer membrane (Millipore, Bedford, MA) and immunostained with a mouse anti-human monoclonal bcl-2 antibody (clone 124, M0887; dilution 1:5,000; Dako, Carpinteria, CA). Bound antibody was detected using the ECL chemiluminescence detection system (Amersham, Arlington Heights, IL). Protein bands were quantified by computer densitometry.

Immunofluorescence analysis.

Cells were cultured in medium with As2O3 or melarsoprol at concentrations of 10−6 and 10−5 mol/L for 4 hours or 24 hours, and then pelleted onto slides by cytospin and fixed in 4% paraformaldehyde. A 1:100 dilution of a mouse monoclonal antibody raised against the N-terminal region of PML (clone PGM3, SC-966; Santa Cruz Biotechnology, Santa Cruz, CA) was applied to the slides for 1 hour. Subsequently, a 1:200 dilution of a Texas Red–conjugated rabbit anti-mouse antibody (115-075-003; Jackson ImmunoResearch Laboratories Inc, West Grove, PA) was used as secondary antibody to stain the PML protein.

In vitro BM culture.

BM cells from wild-type and PML−/− mice were obtained by flushing the femur and tibia BM in Iscove’s MEM containing 15% FBS and plated at a concentration of 5 × 104 cells per dish in “Complete” methycellulose medium (StemCell Technologies, Vancouver, BC, Canada) with or without 10−6 mol/L of As2O3 or melarsoprol. Colony-forming unit erythroid (CFU-E) and CFU granulocyte-monocyte (GM) were counted at days 2 and 6 after plating, respectively. The inhibition of colony formation was calculated by comparison with control.

Growth inhibition by arsenicals does not depend on PML-RARα.

We tested the effects of As2O3 and melarsoprol on the growth of NB4, HL60, U937, and KG-1 cells. At concentrations of As2O3 or melarsoprol ranging from 10−7 to 10−5 mol/L, both compounds inhibited cell growth in all cell lines tested. The effective concentration of As2O3 for growth inhibition was 10−6 mol/L, and at equimolar concentrations, melarsoprol was more potent than As2O3 in all cell lines but KG-1 (Fig 1). Inhibition of growth and survival of cell lines was not dependent on the presence or absence of PML-RARα whose expression is restricted to NB4 cells.

Fig. 1.

Growth inhibition of NB4, HL60, U937, and KG-1 cell lines after As2O3 or melarsoprol treatment. Various concentrations of As2O3 and melarsoprol were added to the media (Materials and Methods): Controls (○), 10−5 mol/L (▵), 10−6 mol/L (□), and 10−7 mol/L (•). As2O3 at a concentration of 10−4 mol/L (⊞) was also used. Representative results from one of three independent experiments are shown as the mean ± SD of triplicates. Inhibition of cell growth and loss of viability are comparable in four different cell lines in which PML-RAR fusion protein is or is not present.

Fig. 1.

Growth inhibition of NB4, HL60, U937, and KG-1 cell lines after As2O3 or melarsoprol treatment. Various concentrations of As2O3 and melarsoprol were added to the media (Materials and Methods): Controls (○), 10−5 mol/L (▵), 10−6 mol/L (□), and 10−7 mol/L (•). As2O3 at a concentration of 10−4 mol/L (⊞) was also used. Representative results from one of three independent experiments are shown as the mean ± SD of triplicates. Inhibition of cell growth and loss of viability are comparable in four different cell lines in which PML-RAR fusion protein is or is not present.

Close modal
Growth inhibition by arsenicals of the RA-resistant NB4 subclone 306.

Both drugs were then tested in the RA-resistant subclone 306 derived from NB4 cells in which the intact PML-RARα protein is no longer expressed.20 This line was sensitive to As2O3 (10−6mol/L) and melarsoprol (10−7 mol/L) (Fig 2A and B). Growth inhibition was also in this case both time and dose dependent. Surprisingly, NB4.306 seemed to respond better than the parental NB4 cell line to both compounds. These results are in keeping with the notion that inhibition of growth and survival of NB4 cell lines does not depend on the presence of PML-RARα or its degradation, and suggest that both arsenicals can be used in the treatment of both RA-sensitive and RA-resistant APL.

Fig. 2.

As2O3 and melarsoprol induce growth inhibition of the RA-resistant cell line NB4.306 cells. Concentrations of As2O3 (A, growth curve; C, viability) and melarsoprol (B, growth curve; D, viability) are the same as depicted in Fig 1. As2O3 and melarsoprol inhibit cell growth in a dose-dependent manner.

Fig. 2.

As2O3 and melarsoprol induce growth inhibition of the RA-resistant cell line NB4.306 cells. Concentrations of As2O3 (A, growth curve; C, viability) and melarsoprol (B, growth curve; D, viability) are the same as depicted in Fig 1. As2O3 and melarsoprol inhibit cell growth in a dose-dependent manner.

Close modal
Induction of apoptosis by arsenicals in NB4, HL60, and U937 cell lines.

To show whether the growth inhibition by As2O3or melarsoprol is caused by induction of apoptosis, in situ TdT labeling or DAPI staining were performed 24 hours after cells were treated with different concentrations of As2O3or melarsoprol (see Materials and Methods). TUNEL-positive cells or cells with nuclear fragmentation which represent apoptotic cells were counted. Both As2O3 and melarsoprol induced apoptosis in all cell lines tested. Maximal apoptotic effects were observed at 10−5 mol/L with both drugs; however, melarsoprol was again somewhat more potent at this concentration (Fig 3A). At 10−6 mol/L concentration, apoptotic cells accounted for only 6% to 10% of the population examined. However, percent of mortality, in certain cases, exceeded these values, suggesting that arsenicals may also induce cell death through mechanisms other than apoptosis. The in situ TdT assay, in these experiments, seemed more sensitive than the morphological analysis (Fig 3). Apoptosis induced by these two compounds was comparable among the three different cell lines tested, irrespective of the expression of the PML-RARα fusion protein.

Fig. 3.

As2O3 and melarsoprol induce apoptosis in NB4, HL60, and U937 cell lines. Apoptotic cells were identified by the in situ TdT assay, as well as morphological changes of nuclei after DAPI staining. The percentages of TUNEL-positive cells (A) and cells with nuclear fragmentation (B) were expressed as the mean ± SD. Morphology of TUNEL-positive cells are indicated with black arrows and fragmented nuclei are indicated with white arrows, as shown in panels (C) and (D).

Fig. 3.

As2O3 and melarsoprol induce apoptosis in NB4, HL60, and U937 cell lines. Apoptotic cells were identified by the in situ TdT assay, as well as morphological changes of nuclei after DAPI staining. The percentages of TUNEL-positive cells (A) and cells with nuclear fragmentation (B) were expressed as the mean ± SD. Morphology of TUNEL-positive cells are indicated with black arrows and fragmented nuclei are indicated with white arrows, as shown in panels (C) and (D).

Close modal
PML and PML-RARα are relocalized and degraded by As2O3 but not melarsoprol.

PML is typically concentrated within discrete speckled nuclear structures that have been variably named PML nuclear bodies (NBs), or Kremer bodies, ND10, or PODs (for PML oncogenic domains).21-25 The presence of PML-RARα leads to the delocalization of PML from the NBs in APL cells, where it acquires a microspeckled/granular nuclear localization. PML, RARα, RXRα, and the PML-RARα fusion protein colocalize in these aberrant structures. Upon RA treatment, these proteins reacquire their natural nuclear localization.23-25 

A previous report suggested that As2O3-induced relocalization of PML or PML-RARα onto nuclear bodies and subsequent degradation of PML or PML-RARα was key in the induction of apoptosis in NB4 cells by this compound.12 To test this hypothesis, we cultured three different leukemic cell lines in the presence of As2O3 or melarsoprol at a concentration of 10−6 (Fig 4) or 10−5 mol/L (not shown) for 24 hours, time by which both compounds already induced cell death. Indirect immunofluorescence staining of PML with a specific monoclonal antibody was performed in control and treated NB4, HL60, and U937 cells after 4 and 24 hours. In NB4 cells, the microspeckled pattern of PML staining rapidly changed to a speckled pattern upon As2O3 treatment, followed by a reduction in the number of PML NBs (Fig 4). The normal nuclear distribution of PML was seen in untreated HL60 and U937 cells, as shown in Fig 4. However, after incubation with As2O3, the number of PML NBs was dramatically reduced also in these cells. Although melarsoprol was more potent in inducing apoptosis, treatment with this drug did not alter PML or PML-RARα nuclear localization patterns in NB4, HL60, and U937 cells either after 4 hours (not shown) or after 24 hours of incubation at both 10−6 (Fig 4) or 10−5 mol/L (not shown) concentrations, suggesting that these agents do not modulate PML or PML-RARα proteins in an identical manner.

Fig. 4.

PML relocalization and degradation in APL or non-APL cell lines after As2O3 and melarsoprol treatment. As2O3 induces PML relocalization onto nuclear bodies, followed by PML degradation not only in NB4 cells but also in HL60 and U937 cell lines. Melarsoprol does not affect PML nuclear localization.

Fig. 4.

PML relocalization and degradation in APL or non-APL cell lines after As2O3 and melarsoprol treatment. As2O3 induces PML relocalization onto nuclear bodies, followed by PML degradation not only in NB4 cells but also in HL60 and U937 cell lines. Melarsoprol does not affect PML nuclear localization.

Close modal
As2O3 and melarsoprol downregulate bcl-2.

To identify what would be a possible mechanism underlying the ability of As2O3 and melarsoprol to induce apoptosis, we performed a Western blot analysis of bcl-2 expression on NB4, NB4-306, HL60, U937 cells treated with various concentrations of As2O3 and melarsoprol (Materials and Methods). Both compounds reduced the levels of bcl-2 protein, not only in NB4 cells but also in HL60, U937, and the RA-resistant NB4 subclone 306 upon 24-hour incubation with these drugs (Fig 5). These changes were dose dependent. Again, melarsoprol was more potent in this effect compared to As2O3. Thus, both As2O3and melarsoprol induce bcl-2 downregulation, and this process does not depend on the presence of PML-RARα.

Fig. 5.

Downregulation of bcl-2 by As2O3and melarsoprol in myeloid cell lines irrespective of PML-RAR expression. As2O3 and melarsoprol downregulate bcl-2 expression at concentrations of 10−6 and 10−5 mol/L in all cell lines tested.

Fig. 5.

Downregulation of bcl-2 by As2O3and melarsoprol in myeloid cell lines irrespective of PML-RAR expression. As2O3 and melarsoprol downregulate bcl-2 expression at concentrations of 10−6 and 10−5 mol/L in all cell lines tested.

Close modal
Growth inhibition and induction of apoptosis by arsenicals in cells devoid of PML.

To study the role of PML in growth inhibition or apoptosis by arsenicals, we took advantage of mice in which the PML gene had been ablated by homologous recombination.16-18 We treated primary MEFs isolated from wild-type and PML−/− mice with different concentrations of As2O3 or melarsoprol for 24 hours. Both compounds triggered apoptosis of MEFs at concentrations of 10−6 and 10−5mol/L. No differences in apoptosis between wild-type and PML−/− MEFs were observed (Fig6A and B). Next, we performed in vitro cultures with BM cells from wild-type and PML−/− mice in the presence or absence of As2O3 or melarsoprol (Materials and Methods). Formation of CFU-E and CFU-GM from wild-type and PML−/− BM progenitors were comparably inhibited by 10−6 mol/L concentration of both compounds (Fig 6C and D). Thus, both As2O3 and melarsoprol can inhibit cell growth in a PML independent manner.

Fig. 6.

As2O3 and melarsoprol induce growth inhibition and apoptosis in a PML independent manner. Wild-type and PML−/− MEFs were incubated with the indicated concentrations of As2O3 (A) and melarsoprol (B) for 24 hours, and apoptotic cells were identified by in situ TdT. For each condition, at least 300 cells were counted. No differences in apoptosis and growth inhibition induced by As2O3 and melarsoprol between wild-type and PML−/− cells were found. For in vitro BM cultures (Materials and Methods), wild-type and PML−/− cells were incubated with As2O3 and melarsoprol at a concentration of 10−6 mol/L. At day 2 and day 6 CFU-E (C) and CFU-GM (D) were counted in triplicate. The data are expressed as a percentage of colony formation of arsenicals-treated versus untreated cells: untreated = 100%. The bars indicate the mean values ± SD from one representative experiment out of three.

Fig. 6.

As2O3 and melarsoprol induce growth inhibition and apoptosis in a PML independent manner. Wild-type and PML−/− MEFs were incubated with the indicated concentrations of As2O3 (A) and melarsoprol (B) for 24 hours, and apoptotic cells were identified by in situ TdT. For each condition, at least 300 cells were counted. No differences in apoptosis and growth inhibition induced by As2O3 and melarsoprol between wild-type and PML−/− cells were found. For in vitro BM cultures (Materials and Methods), wild-type and PML−/− cells were incubated with As2O3 and melarsoprol at a concentration of 10−6 mol/L. At day 2 and day 6 CFU-E (C) and CFU-GM (D) were counted in triplicate. The data are expressed as a percentage of colony formation of arsenicals-treated versus untreated cells: untreated = 100%. The bars indicate the mean values ± SD from one representative experiment out of three.

Close modal

Clinical reports from two Chinese groups, confirmed by a preliminary study in the United States, indicate that As2O3is extremely effective for inducing complete remission in patients with APL who are resistant to both cytotoxic chemotherapy and all-trans retinoic acid.4-6 These studies have suggested that almost all patients who express PML-RARα fusion mRNA are clinically responsive. Recently, the cytotoxic effects of this agent were linked to its ability to relocalize the aberrant PML-RARα fusion protein onto a specific nuclear organelle, the PML nuclear body, which was closely followed by degradation of PML-RARα and the PML proteins.12,14 The direct implication of these results is that the antileukemic effects of As2O3 would thus be limited to APL patients who expressed the fusion protein. However, we have recently tested both As2O3 and melarsoprol (an organic arsenical) in various B-cell lymphoid cell lines and observed striking apoptotic effects,15 suggesting that in these cell lines, PML-RARα expression was not essential to the cytotoxic mechanism. Therefore, to further examine the dependency of arsenicals on PML or PML-RARα expression, we evaluated the effects of these agents in a number of myeloid cell lines that expressed wild-type PML, with or without the fusion PML-RARα, and also in cells in which wild-type PML had been knocked out by homologous recombination.

Results from our experiments show that the cytotoxic effect of both arsenicals in these cell lines is not mediated by mechanisms that are dependent on PML or PML-RARα expression. In most lines, melarsoprol was somewhat more potent compared with As2O3 in inhibiting growth and inducing apoptosis, and the effects of both drugs were dose dependent. As previously reported,12 we confirmed that As2O3 relocalized PML protein onto nuclear bodies and induced PML and PML-RARα degradation in NB4 cells while triggering apoptosis. However, similar effects were also observed in HL60 and U937 cells which do not harbor the PML-RARα fusion gene. Moreover, melarsoprol induced apoptosis in all the cell lines tested without altering PML and/or PML-RARα.

Similarly, the differentiating action of As2O3and melarsoprol, which in our hands was negligible, did not appear to depend on the expression and/or modulation of PML and/or PML-RARα either. In fact, the small effect we observed in long-term cultures (up to 2 weeks), was comparable in all the cell lines tested with both compounds (data not shown).

We also found that bcl-2 downregulation, which has been previously linked to the antileukemic effects of As2O3 in APL, was also not dependent on expression of PML-RARα protein, because it occurred in the NB4 subclone 306 in which the intact protein is not detectable. Finally, to test whether PML expression was essential to the antileukemic effects of arsenicals, we tested both agents in mouse embryonic fibroblasts and BM cells from animals wherein wild-type PML had been eliminated by homologous recombination. In these cells wholly lacking PML expression,18 both As2O3 and melarsoprol were equally effective in inhibiting growth and inducing apoptosis, and both had similar effects on normal CFU-E and CFU-GM colony formation. Moreover, no differences between wild-type and PML−/− cells were observed. Together, these data strongly support the notion that the antileukemic effects of these arsenicals occurs independently of both PML and PML-RARα expression. These results are in keeping with the medicinal history of arsenicals for diseases that are not characterized by alterations in PML protein such as, for instance, chronic myelocytic leukemia.3 

Our results indicate that both As2O3 and melarsoprol are broadly active as antileukemic agents in both myeloid and lymphoid diseases. However, recent studies in solid tumor cell lines show that this activity is not indiscriminate, and does not occur merely as a nonselective cellular poison.26-28 Recent attempts to extend the clinical utility of melarsoprol have been constrained by previously unappreciated neurotoxicity.29Although severe neurotoxicity has also been reported with doses of As2O3 approximately fivefold in excess of that recently reported to be effective in APL,30 this effect has not been observed with the lower doses used clinically in recent series.4-6 

In conclusion, our data indicate that cytotoxic activity is not mediated by the PML protein and therefore is not limited to diseases that are associated with alterations in PML expression. Thus, assuming a reasonable therapeutic index can be established clinically, arsenicals may have a potentially broader therapeutic role that is not confined to APL.

Supported in part by the National Cancer Institute, National Institutes of Health (CA-77136 to R.P.W. and CA-71692 to P.P.P.); the US Food and Drug Administration (FD-R-001364 to R.P.W.); and by a grant from the Lymphoma Foundation. P.P.P. is a Scholar of the Leukemia Society of America.

Address reprint requests to Pier Paolo Pandolfi, MD, PhD, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10021; e-mail: p-pandolfi@ski.mskcc.org

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked "advertisement" is accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Schamberg
JF
Kolmer
JA
Brown
H
Arsenoxide in relation to the toxicity and therapeutic activity of arsphenamine and neoarsphenamine.
Am J Syph Neurol
18
1934
37
2
Kuzoe
FAS
Current situation of African trypanosomiasis.
Acta Trop
54
1993
153
3
Forkner
C
McNair-Scott
TF
Arsenic as a therapeutic agent in chronic myeloid leukemia.
JAMA
97
1931
305
4
Sun
HD
Ma
L
Hu
XC
Zhang
TD
Ai-Lin I treated 32 cases of acute promyelocytic leukemia.
Chin J Integrat Chin West Med
12
1992
170
5
Shen
ZX
Chen
GQ
Ni
JH
Li
XS
Xiong
SM
Qiu
QY
Zhu
J
Tang
W
Sun
GL
Yang
KQ
Chen
Y
Zhou
L
Fang
ZW
Wang
YT
Ma
J
Zhang
P
Zhang
TD
Chen
SJ
Chen
Z
Wang
ZY
Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): II. Clinical efficacy and pharmacokinetics in relapsed patients.
Blood
89
1997
3354
6
Warrell RP Jr
Soignet
SL
Gabrilove
JL
Calleja
EM
Maslak
P
Scheinberg
DA
Chen
YW
Pandolfi
PP
Initial Western study of arsenic trioxide (As2O3) in acute promyelocytic leukemia.
Proc Am Soc Clin Oncol
17
1998
6a
7
Pandolfi
PP
Grignani
F
Alcalay
M
Mencarelli
A
Biondi
A
Lo Coco
F
Grignani
F
Pelicci
PG
Structure and origin of the acute promyelocytic leukemia myl/RARα cDNA and characterization of its retinoid-binding and transactivation properties.
Oncogene
6
1991
1285
8
Kakizuka
A
Miller WH Jr
Umesono
K
Warrell RP Jr
Frankel
SR
Murty
VVVS
Dmitrovsky
E
Evans
RM
Chromosomal translocation t(15;17) in human acute promyelocytic leukemia fuses RARα with a novel putative transcription factor, PML.
Cell
66
1991
663
9
de Thè
H
Lavau
C
Marchio
A
Chomienne
C
Degos
L
Dejean
A
The PML/RARα fusion mRNA generated by the t(15;17) translocation in acute promyelocytic leukemia encodes a functionally altered RAR.
Cell
66
1991
675
10
Pandolfi
PP
Alcalay
M
Fagioli
M
Zangrilli
D
Mencarelli
A
Diverio
D
Biondi
A
Lo Coco
F
Rambaldi
A
Grignani
F
Rochette-Egly
C
Gaube
MP
Chambon
P
Pelicci
PG
Genomic variability and alternative splicing generate multiple PML/RARα transcripts that encode aberrant PML proteins and PML/RARα isoforms in acute promyelocytic leukemia.
EMBO J
11
1992
1397
11
Chen
GQ
Zhu
J
Shi
XG
Ni
JH
Zhong
HJ
Si
GY
Jin
XL
Tang
W
Li
XS
Xong
SM
Shen
ZX
Sun
GL
Ma
J
Zhang
P
Zhang
TD
Gazin
C
Naoe
T
Chen
SJ
Wang
ZY
Chen
Z
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-RARα/PML proteins.
Blood
88
1996
1052
12
Zhu
J
Koken
MHM
Quignon
F
Chelbi-Alix
MK
Degos
L
Wang
ZY
Chen
Z
de The
H
Arsenic-induced PML targeting onto nuclear bodies: Implications for the treatment of acute promyelocytic leukemia.
Proc Natl Acad Sci USA
94
1997
3978
13
Shao
W
Fanelli
M
Ferrara
FF
Riccioni
R
Rosenauer
A
Davison
K
Lamph
WW
Waxman
S
Pelicci
PG
Lo Coco
F
Avvisati
G
Testa
U
Peschle
C
Gambacorti-Passerini
C
Nervi
C
Miller WH Jr
Arsenic trioxide as an inducer of apoptosis and loss of PML/RARα protein in acute promyelocytic leukemia cells.
J Natl Cancer Inst
90
1998
124
14
Look
AT
Arsenic and Apoptosis in the treatment of acute promyelocytic leukemia.
J Natl Cancer Inst
90
1998
86
15
König
A
Wrazel
L
Warrell RP Jr
Rivi
R
Pandolfi
PP
Jakubowski
A
Gabrilove
JL
Comparative activity of melarsoprol and arsenic trioxide in chronic B-cell leukemia lines.
Blood
90
1997
562
16
(abstr, suppl 1)
Pandolfi
PP
Rivi
R
Gaboli
M
Giorgio
M
Antoniou
M
Bygrave
A
Fagioli
M
Cordon-Cardo
C
Pelicci
PG
Luzzatto
L
Grosveld
F
Targeted disruption of the PML gene of acute promyelocytic leukemia.
Blood
86
1995
261a
17
(abstr, suppl 1)
Wang
ZG
Giorgio
M
Rivi
R
Cordon-Cardo
C
Gaboli
M
Pandolfi
PP
Growth and tumor suppressive properties of the PML gene of acute promyelocytic leukemia in PML-/- mice.
Blood
88
1996
477a
18
Wang
ZG
Delva
L
Gaboli
M
Rivi
R
Giorgio
M
Cordon-Cardo
C
Grosveld
F
Pandolfi
PP
Role of PML in cell growth and the retinoic acid pathway.
Science
297
1998
1547
19
Gavrieli
Y
Sherman
Y
Ben-Sasson
SA
Identification of programmed cell death in situ via specific labeling of nuclear DNA fragmentation.
J Cell Biol
119
1992
493
20
Benedetti
L
Grignani
F
Scicchitano
BM
Jetten
AM
Diverio
D
Lo Coco
F
Avvisati
G
Gambacorti-Passerini
C
Adamo
S
Levin
AA
Pelicci
PG
Nervi
C
Retinoid-induced differentation of acute promyelocytic leukemia involves PML-RARα–mediated increase of type II transglutaminase.
Blood
87
1996
1939
21
Dupuy-Coin
AM
Localization of nuclear functions as revealed by ultrastructural autoradiography and cytochemistry
Busch
H
The Cell Nucleus.
1974
5
Academic
New York, NY
22
Ascoli
C
Maul
GJ
Identification of a novel nuclear domain.
J Cell Biol
112
1991
785
23
Koken
MHM
Puvion-Dutilleul
F
Guillemin
MC
Viron
A
Linares-Cruz
G
Stuurman
N
de Jong
L
Szostecki
C
Calvo
F
Chomienne
C
Degos
L
Puvion
E
de Thé
H
The t(15;17) translocation alters a nuclear body in a retinoic acid-reversible fashion.
EMBO J
13
1994
1073
24
Dyck
J
Maul
GG
Miller WH Jr
Chen
JD
Kakizuka
A
Evans
RM
A novel macromolecular structure is a target of the promyelocyte-retinoic acid receptor oncoprotein.
Cell
76
1994
333
25
Weis
K
Rambaud
S
Lavau
C
Jansen
J
Carvalho
T
Carmo-Fonseca
M
Lamond
A
Dejean
A
Retinoic acid regulates aberrant nuclear localization of PML-RARalpha in acute promyelocytic leukemic cells.
Cell
76
1994
345
26
Yang
CH
Wang
TY
Chen
YC
Cytotoxcity of arsenic trioxide in cancer cell lines.
Proc Am Assoc Cancer Res
39
1998
227
27
(abstr, suppl 1)
Calleja
E
Konig
A
Warrell RP Jr
Gabrilove
JL
Arsenic trioxide induces apoptosis in K562 chronic myelogenous leukemia (CML) cells.
Blood
90
1997
3636
28
Calleja
E
Gabrilove
JL
Warrell RP Jr
Arsenic trioxide inhibits cell survival in a variety of cancer cell types.
Proc Am Soc Clin Oncol
17
1998
218a
29
(abstr, suppl 1)
Soignet
SL
Rivi
R
Konig
A
Tong
WP
Gabrilove
JL
Scheinberg
DA
Pandolfi
PP
Warrell RP Jr
Melarsoprol, an organic arsenical, in the treatment of refractory leukemia: Laboratory and clinical results.
Blood
90
1997
507a
30
(abstr, suppl 1)
Westervelt
P
Pollock
J
Huag
J
Ley
TJ
DiPersio
JF
Response and toxicity associated with dose escalation of arsenic trioxide in the treatment of resistant acute promyelocytic leukemia.
Blood
90
1997
3859a
Sign in via your Institution