Conventional treatment of acute leukemia involves the use of cytotoxic agents (chemotherapy), but other strategies have been explored. All-trans retinoic acid (ATRA) and arsenic have clearly been effective in the treatment of acute promyelocytic leukemia (APL), which creates the possibility that other types of acute leukemia can be conquered by selectively inducing differentiation and/or apoptosis. A great number of investigations have been performed to elucidate the mechanisms and search for effective agents in the treatment of other types of acute leukemia by these new strategies. Progress at the molecular level has been achieved in explaining the mechanisms of action of ATRA and arsenic compounds, and several new agents have emerged, although their clinical effectiveness remains to be confirmed. Mechanism-/gene-based targeted therapy and a combination of different strategies will improve the treatment of acute leukemia.

Selective induction of differentiation and apoptosis is a new strategy in the treatment of acute leukemia. Successful treatment of acute promyelocytic leukemia (APL) with all-trans retinoic acid (ATRA) and arsenic compounds prompts us to further investigate, particularly at the molecular level, the mechanism of differentiation and apoptosis and to seek new differentiation- and apoptosis-inducing agents.1 The purpose of this article is to review the state-of-the-art in this area.

Treatment of APL with ATRA

Clinical effectiveness. ATRA is the drug of first choice in the treatment of newly diagnosed APL. ATRA was first introduced to clinical use for the treatment of APL in 1986.2 In 1988, we reported that among 24 APL patients treated with ATRA, 23 achieved complete remission (CR).3 The results were confirmed by Castaigne et al in France,4 who treated 22 APL patients (4 newly diagnosed and 18 at first to third relapse or refractory to chemotherapy) with ATRA from 1988 to July 1989, and 14 attained CR. Since then, randomized studies in many centers around the world document a rising CR rate, a decrease in severe adverse effects, and a prolongation of remission duration. Table 1  summarizes the CR rate obtained in most large series of patients (> 50) since 1990.5–,18 ATRA combined with anthracycline-based chemotherapy can achieve CR in 90%–95% of patients with APL and cure the disease in 70%–75% of the cases. Combination therapy with ATRA and chemotherapeutic agents should now be considered as a standard treatment of APL.19–,21 The conventional daily dose of ATRA is 45 mg/m2, administered orally until remission. A course of the treatment usually requires 28–32 days, and in rare cases up to 42 days. A lower ATRA dose of 30 mg/m2 can provide a similar response and is indicated in cases of drug intolerance or among elderly patients.22 An intravenous liposomal ATRA is now available for APL patients who are unable to swallow or absorb the medication. In 56 evaluated patients receiving 4 or more doses of intravenous liposomal ATRA, the CR was 87% (20/23) for the newly diagnosed APL patients, 78% for patients in first relapse (14/18), and 23% for patients in second relapse (3/13).23 Long-term follow-up confirms the benefit of ATRA incorporation in the treatment regimen of APL, even during maintenance therapy.21,24 

The major life-threatening adverse effect in APL treated with ATRA is the occurrence of retinoic acid syndrome (RAS). Its frequency can be as high as 20%–25%.1 The addition of chemotherapy at the beginning of the treatment has significantly reduced the incidence to 5%–7%.25,26 The effect of ATRA on early amelioration of coagulopathy and its possible mode of action are described in Wang and Chen1 and Degos and Wang.20 

Mechanism of action of ATRA in differentiation therapy. Over the last decade, tremendous efforts have been made to elucidate the molecular genesis of APL, as well as the mechanism of action of ATRA. The classic chromosome abnormality of APL is a translocation t(15;17)(q22;q21) resulting in the generation of fusion gene and protein PML-RARα, which plays a central role in APL pathogenesis. Experimental evidence has revealed that PML-RARα protein has the following activities: (1) it creates a complex with retinoid x receptor (RXR), nuclear corepressors (N-CoR), Sin3A, and histone deacetylase (HDAC) that represses the transcriptional expression of target genes;27 (2) it acts in a dominant negative manner on the retinoic acid-signaling pathway,28 blocking the differentiation of myeloid cells; and (3) it forms a heterodimer with wild-type PML protein and thereby disrupts the PML nuclear body or PML oncogenic domains (PODs).29 The function of PML as a growth inhibitor and regulator of apoptosis is disturbed when incorporated into PML-RARα complex. The mechanisms of action of ATRA can be summarized as follows: (1) The binding of ATRA to RAR receptors causes degradation of PML-RARα protein through the ubiquitin-protosome and caspase system,30,31 leading to restoration of terminal differentiation of promyelocytes; (2) Exposure of APL cells to ATRA in vitro or in vivo induces relocalization of PML and restores the normal structure of PODs;28 and (3) Under the action of ATRA at pharmacological concentration (1 μM), CoR is dissociated from the repressive complex, whereas CoA (coactivator) is recruited to the complex.27 As a result, the repression of transcriptional activation of target genes is relieved and the differentiation of promyelocytes’ process is restored. To further clarify the molecular mechanism of ATRA-induced differentiation, the gene expression profile in the APL cell line NB4 was studied before and after treatment with ATRA using complementary DNA array, suppression-subtractive hybridization, and differential-display polymerase chain reaction (PCR). In a study conducted by the Shanghai group, 169 genes were modulated by ATRA; among them, 100 were upregulated and 69 downregulated. These genes involve transcription factors, DNA synthesis/repair and recombination proteins, cytokines and chemokines, signal transduction modulators and effectors, interferon signaling, cell cycle regulation, apoptosis-related proteins, cell structure/mobility proteins, and cell adhesion proteins, and others. It is interesting to note that the chronology of up- or downregulation of these genes accords with the process of terminal differentiation of the leukemic cell.32 Recently, using a complementary DNA microarray platform containing 12,630 clones, in conjunction with bioinformatics analysis such as self-organizing map (SOM) and component plane presentation, we profiled gene change in the ANB4 cell line treated with a pharmacological dose of ATRA at 6, 12, 24, and 48 hours. The results showed that ATRA-induced differentiation was a complex and well-organized process. At the early stage, it was manifested by cell cycle arrest, cell proliferation suppression, and antagonism of apoptosis. The ubiquitin-proteasome degradation system was activated, chronologically correlated with the degradation of PML-RARα fusion protein and the assembly of PODs. With the process of differentiation and maturation in granulocytes, genes underlying the process of apoptosis were modulated. In addition to genes involved in the protein kinase C (PKC), protein kinase A/cyclic adenosine monophosphate (PKA-cAMP), and Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways, a large number of genes were found to be involved in other pathways, such as insulin receptor signaling and calcium signaling, that were regulated during ATRA-induced differentiation. In particular, the importance of calcium signaling was detailed in this study. Interestingly, specific members of the histone family were also found to be significantly regulated, suggesting that a chromosome remodeling process occurs during differentiation. Additionally regulated genes included a number of potential hematopoietic regulators involved in various translocations in hematopoietic malignancies. The next steps will be to clarify the relationship between molecular pathways implicated in the differentiation process and the relationship with hematopoietic malignancies (J Zhang et al, unpublished data).

Treatment of APL with arsenic compounds

Clinical effectiveness. Treatment of APL by arsenic compounds represents a successful example of apoptosis induction therapy of acute leukemia (Table 2 ). Arsenic (As) was used for more than 500 years in traditional Chinese medicine (TCM).41 In the early 1970s, a group of investigators from Harbin Medical University in the northeastern region of China reported that a crude solution of arsenic trioxide (As2O3) Ailin-1 could be used to treat APL, according to the TCM principle of using a toxic agent against something toxic. A paper first appeared in 1992,42 and a pure solution of As2O3 has been used in clinic since 1996.43 Two groups of clinical investigators reported the results obtained with pure 1% As2O3 solution. In one study, 22 of 30 (73.3%) cases of newly diagnosed APL and 22 of 42 (52.4%) cases of relapsed or refractory APL achieved CR.43 In our study, 15 of 16 APL patients in relapse entered CR, and dual effects (apoptosis and differentiation-inducing effect) on APL cells in vitro and in vivo were demonstrated.44,45 Use of a TCM compound called Indigo naturalis, which consists of realgar (containing mainly tetra-arsenic tetra-sulfide [As4S4]), Baphicacanthus cusia, Radix salviae miltiorrhizae, and Radix pseudostellariae achieved CR rates as high as 96% in 60 APL patients.46 In 1999, purified As4S4 was reported to be effective in APL treatment.36,Table 2  shows recent reports of CR rates in APL patients treated with arsenic compounds; CR rates of 85% to 90% were attained in newly diagnosed APL patients treated with As2O3. The CR rate may be higher in relapsed cases.35,40 

An important question is whether As2O3 can achieve molecular remission. When we used a very sensitive nested reverse transcriptase (RT)-PCR system, molecular remission was observed in only 1 of 10 patients who had received a single course of As2O3 and were tested immediately after CR.44 In a recent report by Lazo et al, 7 of 10 evaluated patients achieved a molecular remission.40 Soignet et al found that after 2 courses of treatment, 8 of 11 patients were converted to PML-RARα negative,47 and 31 patients (91%) with continued remission after postremission therapy had a negative result for t(15;17).38 When oral As4S4 was used, 14 of 16 patients achieved molecular remission.48 Therefore, after CR is achieved by arsenic compounds, a molecular remission is obtainable either with arsenic compounds or with ATRA and chemotherapy as consolidation treatment.

Another key question is whether clinical use of arsenic compounds can raise the 5-year survival rate. If yes, how can they be used in postremission treatment of APL? In our study, 33 relapsed APL patients treated with As2O3 who achieved CR were followed for 8 to 48 months; the estimated disease-free survival (DFS) rates at 1 and 2 years were 63.6% and 41.6%, respectively.35 Ma et al reported that overall survival (OS) for 7 years was 58.5% with chemotherapy as consolidation therapy.34 Zhang et al37 reported that the 5- and 7-year survival rates were 92.02% and 76.69%, respectively, for patients achieving CR and receiving As2O3 and/or chemotherapy as maintenance therapy. In the US multicenter study, As2O3 was used in relapsed cases. Among 32 patients achieving CR, 18 received additional As2O3 treatment and 11 underwent allogeneic or autologous bone marrow transplantation. The estimated 18-month OS and relapse-free survival rates were 66% and 56%, respectively.38 Mathews et al reported that 10 newly diagnosed APL patients treated with As2O3 and achieving CR remained in CR at a median follow-up of 15 months.39 Lu et al48 recently treated 103 APL patients in hematological CR with As4S4 orally as maintenance therapy. DFS rates for 1 and 6 years were 96.7% and 87.4%, respectively. Therefore, it seems likely that arsenic compounds appropriately used in postremission therapy could prevent recurrence and achieve a longer survival time. Randomized prospective studies will be necessary to define the arsenic compound regimen that best prevents APL relapse.

Adverse effects of arsenic compounds, which are usually mild and tolerable, include nausea, vomiting, and abdominal pain. Nevertheless, severe hepatic toxicity can occur, particularly in newly diagnosed cases.35 Cardiac damage with electrocardiographic QT prolongation was observed in 63% of patients in one report.38 The incidence of RAS-like manifestations accompanied by hyperleukocytosis may be as high as 25%, but it effectively responds to the dexamethasone treatment.38 

Mechanism of action of As2O3. As2O3 exerts dual effects on APL cells. Studies in vitro with NB4 cells showed that a higher concentration of As2O3 (0.5–1.0 μM) induced apoptosis with typical morphological changes, DNA laddering on agarose gel electrophoresis, appearance of an apoptotic peak on flow cytometric analysis, and increased expression of annexin V on the cell surface membrane. Studies on the mechanism of apoptosis revealed that it was due to the collapse of mitochondrial transmembrane potential, increase in reactive oxygen species generation, release of cytochrome c and apoptosis-inducing factor (AIF) into cytoplasm, activation of caspases, and decreased expression of Bcl-2.41,49 As2O3 acts through activation of Jun N-terminal kinase (JNK) and activator protein-1, inhibition of dual-specificity phosphatases,50 CD95-independent caspase 8 activation,51 and inhibition of nuclear factor (NF)-κB.52 In addition, As2O3 could potently enhance phosphoacetylation of serine 10 of histone H3 and phosphoacetylation at the chromatin of caspase-10.53 The most important finding is that As2O3 is able to degrade the PML-RARα oncoprotein at a wide range of concentrations (0.1–1.0 μM).45,54 As2O3 causes PML to localize into the nuclear matrix, where PML proteins become sumoylated and are degraded after recruitment of proteosomes.55 

At lower concentrations, As2O3 can induce APL cells to partially differentiate along the granulocytic pathway, as evidenced by increase in CD11b and CD14 expression and decline in CD33 expression. The mechanism of arsenic-induced differentiation is not clear. Degradation of PML-RARα in the presence of lower concentrations of As2O3, although slower in kinetics, favors the release of differentiation arrest. In addition, acetylation of histones 3 and 4 probably contributes to the mechanism of the differentiation process.41 

Treatment of APL by combining ATRA and As2O3

Studies in animal models demonstrate that synergy might exist between ATRA and As2O3. The combination of the 2 drugs may also bring clinical advantages or even the possibility of eradicating the leukemia clone in APL patients.56,57 Three relapsed APL patients achieved CR when treated with ATRA in conjunction with As2O3.58 At our institute, 31 newly diagnosed APL patients were treated with ATRA in combination with As2O3, and 29 (93.5%) entered CR. No relapses occurred in a preliminary follow-up study during a median time of 8 months.59 To further compare the combination therapy with monotherapy, we conducted a prospective study in which remission induction was performed using either ATRA, As2O3, or ATRA/As2O3 in combination. Fifty-nine newly diagnosed APL patients entered this trial: 20 cases were treated with ATRA, 18 with As2O3, and 21 with a combination of ATRA and As2O3. The CR rates were 90.5%, 88.9%, and 95.2%, respectively, without significant difference (P > .05). Of note, the time to CR was shorter—25.4 ± 5.0 days in the combination group as compared to 40.2 ± 10.5 and 32.6 ± 3.5 days in the ATRA (P = .0003) and As2O3 (P = .003) groups, respectively. Although liver damage was more frequent, it was tolerable and the treatment could continue after dose reduction. Real-time RT-PCR tests revealed that the copy number of PML-RARα transcript was reduced a median of 118.9-fold (n = 20) in the combination group, while the median reduction fold was 32.1 and 6.7 in the groups using As2O3 (n = 16) and ATRA (n = 19), respectively. These differences were statistically significant (combination group vs As2O3 group, P =.009, vs ATRA group, P = .041). Most importantly, at a median follow-up duration of 13 months, no relapse was observed in the combination group, as compared to the ATRA (21.0%; P = .02) and As2O3 (12.5%; P = .104) groups (ZZ Shi et al, unpublished data).

Other differentiation-inducing agents

Dozens of differentiation-inducing agents have been studied as potential differentiation therapy of acute leukemia. They can be divided into the following groups60:

  1. vitamin analogs: retinoids and vitamin D derivatives

  2. cytokines: granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), interferons, tumor necrosis factor (TNF)

  3. polar-aplanar compounds: hexamethylene bisacetate (HMBA)

  4. histone deacetylase inhibitors: trichostatin (TSA), phenylbutyrate,61 apicidin,62 depsipeptide (FR901228)63 

  5. inhibitors of DNA methylation: 5-aza-2′-deoxycytidine64 

  6. cyclic AMP analogs: 8-Cl-cAMP, dibutyl cAMP

  7. chemotherapeutic agents: aclarubicin, cytosine arabinoside (Ara-C), hydroxyurea

  8. medicinal plant-derived products and plant growth regulators: meisoindigo (derivative of indirubin, ingredient of Indigofera suffruticosa Mill),65 flavonoids from Morus alba leaves,66 cotylenin A (a plant growth regulator),67 tanshinone IIA (a component of Salvia miltiorrhiza),68 pyranocoumarins (isolated from Peucedanum praeruptorum Dunn),69 intermedeol (isolated from the leaves of Ligularia fischeri var. spiciformis),70 and magnolialide (a 1beta-hydroxyeudesmanolide isolated from Cichorium intybus).71 

Even though many new differentiation-inducing agents have been explored during the past few years, the majority of them have only been studied in vitro on only leukemia cell lines, such as NB4, H-60, U937, K562, or Kasumi-2, or in a primary leukemia cells culture assay. Therefore, the results of clinical trials of their effectiveness are limited. The following reports show some positive results in differentiation therapy for acute leukemia other than APL with ATRA. First, among 41 cases of acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) treated with HMBA, 3 patients achieved CR and 6 partial remission (PR), with a median duration of CR of 6.8 months (1.3–16 months).72 Second, an APL patient who had had multiple relapses and was resistant to ATRA treatment achieved clinical and cytogenetic CR after being treated with sodium phenylbutyrate. Immunofluorescence and Western blot analysis revealed that phenylbutyrate caused a time-dependent increase in histone acetylation in blood and marrow mononuclear cells.73 Third, CR was achieved in a case of M2a subtype of AML treated with ATRA alone.74 Fourth, low-dose cytarabine was used for the treatment of AML in elderly patients; 24/48 (50%) achieved CR with a median remission duration of 8 months.75 However, this schedule is not advised in patients with monocytic leukemia or in those with hypercellular marrow.76 Fifth, a combination of different inducers of differentiation could yield a higher remission rate, as illustrated in a report that concurrent administration of G-CSF, low-dose cytarabine (10 mg/m2/d, for 14 days), and aclarubicin (10–14 mg/m2/d, for 4 days) in 18 patients with relapsed AML, achieved a CR rate of 83% (15/18).77 A protocol combining low-dose cytarabine and harringtonine was tried in 10 AML patients, and 8 out of the 10 achieved CR.78 When ATRA was used in conjunction with low-dose Ara-C in the treatment of “poor prognosis” AML, 48% (16/33) of the patients entered CR, and the CR rate was 88% in 17 patients with < 50% blasts in bone marrow.79 It is noteworthy that a cytotoxic adverse effect cannot be excluded when low-dose chemotherapeutic drugs are used as differentiation-inducing agents.

Other apoptosis-inducing agents based on modulation of distinct pathways

A great number of new apoptosis-inducing agents are being studied. Table 3  displays some new compounds (other than the the arsenic compounds previously described) reported recently, and Table 4  shows medicinal plant–derived or other natural products. The mechanisms of apoptosis induction implicated for these agents can be grouped into the following well-clarified or not-yet-well-clarified categories:

  1. activation of death pathway receptors (FAS, TNF) and signaling, as with DT(338) GM-CSF,99 tryptanthrin103 

  2. calcium-dependent apoptosis signal pathway, as with calphostin C81 

    (3) activation of caspases, as with se-methyl selenocysteine,88 novel retinoid CD437 and its nonretinoidal analog MM002,96 curcumin,107 diallyl disulfide,97 and resveratrol87 

  3. increase of reactive oxygen species (ROS) production or ROS-dependent apoptosis, as with falconensone A dioxime,86 β2 microglobulin,98 and baicalin111 

  4. disruption or modification of mitochondrial transmembrane potential (Δψμ) or cytochrome c, AIF release, as with potassium antimonyl tartrate,92 homoharringtonine,106 theasinensin A,101 artepillin C,102 carnosol,104 curcumin,107 baicalin,111 sophoranone,109 dolichyl monophosphate,93 farnesylpyridinium,95 lovastatin83 and bis(4,7-dimethyl-1,10 phenanthroline) sulfatooxovanadium (IV)[VO(SO4)(Me2-Phen)2]91 

  5. modulation of Bcl-2/Bax, or Bcl-2, as with carnosol,104 erianin,105 and momordin I108 

  6. modulation of cell cycle, inhibition of cyclin D kinase (CDK), as with arginine deiminase84 and monensin94 

  7. decrease or inactivation of antiapoptosis activation gene or protein, such as XIAP processing with STI571 + TRAIL89 and NF-κB with arsenic + interferon (IFN)-α85 

  8. caspase-3 activation and related to SAPK/JNK, as with squamocin112 

For the majority of apoptosis-inducing agents, the pathways implicated in the mechanism of apoptosis are usually not unique as shown in Table 3  and Table 4  but may involve much more complex networks of modulation.

In general, arsenic compounds are not effective in the treatment of acute leukemia other than APL. The majority of apoptosis-inducing agents or products listed in Tables 3  and 4 are based on in vitro studies. However, the apoptosis-inducing CD19-directed tyrosine kinase inhibitor B43-genistein has been studied in a clinical trial.82 Seven children and 8 adults with CD19+ B-lineage acute lymphoblastic leukemia (ALL), and 1 adult with chronic lymphocytic leukemia were treated with this drug. All patients had failed to respond to previous chemotherapy, and 6 had relapsed after bone marrow transplantation; there were 2 transient responses and only 1 durable CR. In spite of few positive results from clinical trials, the treatment of acute leukemia by triggering apoptosis is still a promising new area deserving further exploration.

Apoptosis-inducing activity in chemotherapeutic agents

A number of known cytotoxic drugs can induce an apoptotic pathway. Treatment of human acute leukemia cells HL-60, U937, and Jurkat cells with etoposide, Ara-C, or doxorubicin can upregulate DR5 levels in a p53-independent manner and sensitize them to TRAIL (Apo-2L)-induced apoptosis.113 Another mechanism in etoposide-induced apoptosis is the engagement of the mitochondrial pathway by releasing cytochrome c, in which caspase 2 is considered to play an important role.114 There is evidence that doxorubicin-induced apoptosis in HL-60 cells is ROS- and proteinase-3-dependent, which is downregulated in its doxorubicin-resistant variant.115 DNA damage caused by doxorubicin can lead to activation of the p53-survivin signaling pathway, inducing cell cycle arrest and apoptosis in childhood ALL cells. Survivin is a novel member of the inhibitors of apoptosis protein.116 Mitoxantrone and anisomycin are able to induce apoptosis in HL-60 cells by stimulating JNK/SAPK activity in a time- and dose-dependent manner.117 Adriamycin is capable of inducing apoptosis in the T-cell leukemia line, H9, probably through activating JNK.118 Cladribine, cytarabine, cisplastin, and 5-fluorouracil have apoptosis- and necrosis-inducing potential in human leukemia cell lines HSB2 and Jurkat.119 Homoharringtonine (HHT), extracted from a medicinal plant (Cephalotaxus hainanensis Li) in China, has long been used as a cytotoxic agent in the treatment of acute leukemia.120 An in vitro study on HL-60 and K562 cells revealed that HHT exerts its action by inducing apoptosis with downregulation of BCR/ABL gene expression. Laser scanning confocal microscopy examination disclosed that there is an alteration of intracellular calcium distribution in apoptosis of HL-60 cells induced by harringtonine (another active component isolated from C hainanensis Li).121 The apoptotic response to HHT in the human wild-type p53 leukemic cell line MOLT-3 is independent of ROS generation, while Bax translocation, mitochondrial cytochrome c release, and caspases activation have been implicated.106 

Oncogene- or mechanism-based targeted treatment

ATRA and arsenic compounds are very effective in the treatment of APL, providing a model of success in the treatment of APL through induction of differentiation and apoptosis. However, there is an interesting question: Why do they have limited or even no effect at all in the treatment of other types of acute leukemia? The aforementioned findings may help us to understand the reasons. APL has a unique and specific chromosome aberration t(15;17) resulting in the fusion gene and protein PML-RARα, which plays a crucial role in APL genesis with arrest of differentiation and impairment of apoptosis. Although the mechanism of action of ATRA and arsenic compounds has not yet completely been explained, the data mentioned above suggest a common pharmacological activity of these 2 agents: to target the fusion protein and to cause its degradation. The success in the treatment of APL with ATRA or arsenic compounds furnishes a model of mechanism- or oncogene-based targeted therapy. A second example of considerable success through molecularly based targeted therapy is offered by chronic myelogenous leukemia treated with Gleevec, a specific inhibitor of tyrosine kinase located on ABL of the BCR-ABL fusion protein, resulting from a specific aberration of chromosome translocation t(9;22). A third candidate for study of oncogene-based targeted therapy is M2b AML. This subtype of AML accounts for about 20% to 25% of acute myeloid leukemia and is characterized by a chromosome translocation t(8;21)(q22;q22). The result of this chromosomal aberration is to generate a chimeric fusion product AML1-ETO that plays a central role in genesis of this leukemia. Similar to PML-RARa in APL, the fusion protein recruits a transcription repression complex containing histone deacetylase. A potent histone deacetylase inhibitor, TSA, and a less specific inhibitor, phenylbutyrate, partially reversed ETO-mediated repression,122 but the clinical result was not as promising. A new candidate is the FLT3 gene. In up to 41% of patients with AML, there are constitutively activating internal tandem duplication (ITD) and point mutations of the receptor tyrosine kinase FLT3.123 An inhibitor of FLT3-targeted tyrosine kinase CEP-70 has been developed, and the compound is able to inhibit FLT3 in primary leukemia blasts from AML patients harboring the FLT3/ITD mutation. This drug can prolong survival in a mouse leukemia model bearing FLT3/ITD.123 Recently, SIH-10, isolated from a Chinese medicinal herb, has been shown to induce apoptosis of Kasumi cell line and primary M2 AML (GP Zhou et al, unpublished data). A preliminary in vivo study demonstrated that intraperitoneal administration of this compound into Nod/Scid mice bearing M2 AML leukemia model can induce apoptosis of leukemia cells and prolong the life span of the animal by 44%. It seems likely that this new compound could bring benefit in gene/mechanism target-based treatment of M2 AML. Further studies are in progress.

Rational combination of differentiation treatment, apoptosis-inducing treatment, and chemotherapy

The superiority of combining differentiation and/or apoptosis inducers and chemotherapy has already been proved in the treatment of APL. With this strategy, the CR rates are higher, the survival time is longer, and the risk of adverse effects is lower; even drug resistance can be overcome. In vitro studies show that combination of a potent D3 analogue (KH 1060) with 9-cis retinoic acid can irreversibly inhibit clonal growth, decrease Bcl-2 expression, and induce apoptosis in HL-60 cells.124 IFN-γ is able to sensitize human myeloid leukemia cells to death receptor–induced and mitochondria-mediated apoptosis.125 Susceptibility to TRAIL-induced apoptosis is augmented by doxorubicin in some primary leukemia cells.126 A synergic effect is observed in APL cell differentiation through combining arsenic trioxide with cAMP, subtending a new signaling pathway.127 Arsenic is capable of inducing apoptosis in multidrug-resistant human myeloid leukemia cells that express BCR-ABL or overexpress MDR, MRP, Bcl-2, or Bcl-x(L).128 Treatment of BCR-ABL-positive leukemia cells with As2O3 combined with STI-571 can potentially induce apoptosis and differentiation.129 Studies on the treatment of acute leukemia by combining different strategies should yield promising results. As indicated above, the treatment of APL with ATRA in conjunction with As2O3 could achieve not only a high CR rate with shorter time for remission induction but also a better reduction of leukemic burden, as shown by a much higher reduction of copy number of PML-RARα mRNA transcripts.

In conclusion, the success in the treatment of APL by inducing differentiation and apoptosis with ATRA and arsenic compounds offers convincing evidence to support further investigation of new medications in the treatment of acute leukemia by these 2 new approaches. The key problem is how to find the oncogenes or genes and related proteins involved in the genesis of the leukemia and to elucidate the mechanism by which the leukemia develops. If this were accomplished, it will be possible to design and create new compounds based on oncogene or mechanism-targeted principles. The rational combination of different strategies based on regimens defined by prospective randomized studies will bring benefits in future therapies that might help us conquer the disease.

Table 1.

Complete remission rate in APL patients treated with ATRA (in series including more than 50 cases).

YearResearchersProtocolCR n(%)
Abbreviations: APL, acute promyelocytic leukemia; Ara-C, cytosine arabinoside; ATRA, all-trans retinoic acid; chemo, chemotherapy; CR, complete remission; ext, extended; HD, high dose. 
1991 Chen et al5  ATRA 50 94.0 
1992 Chinese Coop Study Group6  ATRA 400 85.0 
  ATRA + chemo 144 76.4 
1993 Shanghai Coop Study Group7  ATRA 91 81.3 
1994 Warrell et al8  ATRA 79 84.8 
1995 Kanamaru et al9  ATRA ± chemo 109 89.0 
1997 Tallman et al10  ATRA 172 72.1 
1997 Soignet et al11  ATRA ± chemo 95 83.2 
1997 Asou et al12  ATRA 62 95.2 
  ATRA ± chemo 196 88.3 
1997 Mandelli et al13  ATRA + chemo 240 95.4 
1999 Fenaux et al14  ATRA ± chemo 413 92.0 
1999 Burnett et al15  ATRA (short) + chemo 119 70.0 
  ATRA (ext) + chemo 120 87.0 
1999 Hu et al16  ATRA + chemo 120 88.4 
2000 Lengfelder et al17  ATRA + HD Ara-C 51 92.0 
2001 Asou et al18  ATRA ± chemo 369 90.0 
  ATRA alone  94.0 
  ATRA + initial chemo  89.0 
  ATRA + later chemo  88.0 
  ATRA + initial, later chemo  86.0 
YearResearchersProtocolCR n(%)
Abbreviations: APL, acute promyelocytic leukemia; Ara-C, cytosine arabinoside; ATRA, all-trans retinoic acid; chemo, chemotherapy; CR, complete remission; ext, extended; HD, high dose. 
1991 Chen et al5  ATRA 50 94.0 
1992 Chinese Coop Study Group6  ATRA 400 85.0 
  ATRA + chemo 144 76.4 
1993 Shanghai Coop Study Group7  ATRA 91 81.3 
1994 Warrell et al8  ATRA 79 84.8 
1995 Kanamaru et al9  ATRA ± chemo 109 89.0 
1997 Tallman et al10  ATRA 172 72.1 
1997 Soignet et al11  ATRA ± chemo 95 83.2 
1997 Asou et al12  ATRA 62 95.2 
  ATRA ± chemo 196 88.3 
1997 Mandelli et al13  ATRA + chemo 240 95.4 
1999 Fenaux et al14  ATRA ± chemo 413 92.0 
1999 Burnett et al15  ATRA (short) + chemo 119 70.0 
  ATRA (ext) + chemo 120 87.0 
1999 Hu et al16  ATRA + chemo 120 88.4 
2000 Lengfelder et al17  ATRA + HD Ara-C 51 92.0 
2001 Asou et al18  ATRA ± chemo 369 90.0 
  ATRA alone  94.0 
  ATRA + initial chemo  89.0 
  ATRA + later chemo  88.0 
  ATRA + initial, later chemo  86.0 
Table 2.

Complete remission rate induced by arsenic compounds (reports from 1998–2003).

YearArsenic AuthorsCompoundDisease StatusnCR (%)
Abbreviations: As2O3, arsenic trioxide; As4S4, tetra-arsenic tetra-sulfide; CR, complete remission. 
1998 Huang et al33  As2O3 Relapse + refractory 57 
1998 Ma et al34  As2O3 De novo 98 87.1 
1999 Niu et al35  As2O3 De novo 11 72.7 
   Relapse 47 85.1 
1999 Lu et al36  As4S4 De novo + relapse 100 84.9 
2000 Zhang et al37  As2O3 De novo 124 87.9 
   De novo + relapse + refractory 242 74.8 
2001 Soignet et al38  As2O3 Relapse + refractory 40 85 
2002 Mathews et al39  As2O3 De novo 11 91.0 
2003 Lazo et al40  As2O3 Relapse 12 100.0 
YearArsenic AuthorsCompoundDisease StatusnCR (%)
Abbreviations: As2O3, arsenic trioxide; As4S4, tetra-arsenic tetra-sulfide; CR, complete remission. 
1998 Huang et al33  As2O3 Relapse + refractory 57 
1998 Ma et al34  As2O3 De novo 98 87.1 
1999 Niu et al35  As2O3 De novo 11 72.7 
   Relapse 47 85.1 
1999 Lu et al36  As4S4 De novo + relapse 100 84.9 
2000 Zhang et al37  As2O3 De novo 124 87.9 
   De novo + relapse + refractory 242 74.8 
2001 Soignet et al38  As2O3 Relapse + refractory 40 85 
2002 Mathews et al39  As2O3 De novo 11 91.0 
2003 Lazo et al40  As2O3 Relapse 12 100.0 
Table 3.

Apoptosis-inducing compounds for acute leukemia cells.

Authors (Year)Apoptosis-Inducing AgentsIn Vitro Study Cell LinesMechanism
Abbreviations: AIF, apoptosis-inducing factor; ALL, acute lymphoblastic leukemia; ATRA, all-trans retinoic acid; mRNA, messenger RNA; RA, retinoic acid; NC, not clear; AML,acute myeloid leukemia; CLL, chronic lymphoid leukemia; ROS, reactive oxygen species; Dym, mitochondrial transmembrane potential; DD, death domain; TRAIL, tumor necrosis factor alpha-related apoptosis-inducing factor; IFN, interferon; NF, nuclear factor; XIAP, x chromosome-linked inhibitor of apoptosis protein 
Fujimura et al80 (1998) ATRA 9-cis RA 13-cis RA ATL p21Waf1/Cipl protein↑pRb hypophosphorylation 
Zhu et al81 (1998) Calphostin C ALL-1 (pre-pre-B)
 RS4;11 (pro-B)
 NALM-6 (pre-B)
 Daudi (B-ALL)
 Molt-3 (T-ALL)
 Jurkat (T-ALL) Calcium-dependent apoptotic signal pathway 
Uckun et al82 (1999) B43-genistein CD19+ B-ALL CLL CD19-receptor-directed tyrosine kinase inhibitor 
Dimitroulakos et al83 (1999) Lovastatin AML cell lines fresh cells Δψμ↓, glutathione↓ 
Gong et al84 (2000) Arginine deiminase ALL Cell cycle arrest in G1 
El-Sabban et al85 (2000) As + IFN-α HTLV-1 transformed cells Tax downregulation, inactivation of NF-κB pathway 
Takahashi et al86 (2000) Falconensone A, dioxime HL-60 Intracellular ROS↑ 
Dorrie et al87 (2001) Resveratrol HL-60 Independent of CD95 signaling, mitochondria/caspase 9 pathway 
Kim et al88 (2001) Se-methyl- selenocysteine HL-60 Activation of caspase 3 
Nimmanapalli et al89 (2001) STI-571 + TRAIL bcr-abl+/HL-60 Enhance Apo-2L/ TRAIL induced-apoptosis, increase of caspases 9,3,XIAP processing 
Ogata et al90 (2001) Picolinic acid- related comp HL-60 NC 
Narla et al91 (2001) VO(SO4)(Me2- Phen)2 NALM-6 leukemia cells Δψμ↓ ROS 
Lecureur et al92 (2002) K antimonyl tartrate HL-60 ROS↑, Dym↓ 
Yasugi et al93 (2002) Dolichyl monophosphate U937 Δψμ↓, AIF translocation, activation caspase 3-like protease 
Park et al94 (2002) Monensin Na+ ionophore 10 AML cells including HL-60 CDK6 Cyclin D1↓ Cyclin A↓, p27↑, changes in Δψμ, caspase 3, 8, Bax 
Hamada et al95 (2002) Farnesylpyridinium HL-60 cytochrome c↑, cytochalasin-like effect 
Zhang et al96 (2002) Novel retinoid CD437, MM002 WSU-B-CLL primary B-CLL ALL Caspase 2, 3↑, cleavage of Bcl-XL, activation of caspase 2, 3 
Kwon97 (2002) Diallyl disulfide HL-60 Caspase 3 hydrogen peroxide generation increased 
Gordon98 (2003) β2-microglobulin CCRF-HSB-2 ALL cell line Caspase, ROS dependent, release of cytochrome c, AIF 
Thorburn99 (2003) DT(388)GM-CSF U937 Activation of FAS-associated DD protein 
Authors (Year)Apoptosis-Inducing AgentsIn Vitro Study Cell LinesMechanism
Abbreviations: AIF, apoptosis-inducing factor; ALL, acute lymphoblastic leukemia; ATRA, all-trans retinoic acid; mRNA, messenger RNA; RA, retinoic acid; NC, not clear; AML,acute myeloid leukemia; CLL, chronic lymphoid leukemia; ROS, reactive oxygen species; Dym, mitochondrial transmembrane potential; DD, death domain; TRAIL, tumor necrosis factor alpha-related apoptosis-inducing factor; IFN, interferon; NF, nuclear factor; XIAP, x chromosome-linked inhibitor of apoptosis protein 
Fujimura et al80 (1998) ATRA 9-cis RA 13-cis RA ATL p21Waf1/Cipl protein↑pRb hypophosphorylation 
Zhu et al81 (1998) Calphostin C ALL-1 (pre-pre-B)
 RS4;11 (pro-B)
 NALM-6 (pre-B)
 Daudi (B-ALL)
 Molt-3 (T-ALL)
 Jurkat (T-ALL) Calcium-dependent apoptotic signal pathway 
Uckun et al82 (1999) B43-genistein CD19+ B-ALL CLL CD19-receptor-directed tyrosine kinase inhibitor 
Dimitroulakos et al83 (1999) Lovastatin AML cell lines fresh cells Δψμ↓, glutathione↓ 
Gong et al84 (2000) Arginine deiminase ALL Cell cycle arrest in G1 
El-Sabban et al85 (2000) As + IFN-α HTLV-1 transformed cells Tax downregulation, inactivation of NF-κB pathway 
Takahashi et al86 (2000) Falconensone A, dioxime HL-60 Intracellular ROS↑ 
Dorrie et al87 (2001) Resveratrol HL-60 Independent of CD95 signaling, mitochondria/caspase 9 pathway 
Kim et al88 (2001) Se-methyl- selenocysteine HL-60 Activation of caspase 3 
Nimmanapalli et al89 (2001) STI-571 + TRAIL bcr-abl+/HL-60 Enhance Apo-2L/ TRAIL induced-apoptosis, increase of caspases 9,3,XIAP processing 
Ogata et al90 (2001) Picolinic acid- related comp HL-60 NC 
Narla et al91 (2001) VO(SO4)(Me2- Phen)2 NALM-6 leukemia cells Δψμ↓ ROS 
Lecureur et al92 (2002) K antimonyl tartrate HL-60 ROS↑, Dym↓ 
Yasugi et al93 (2002) Dolichyl monophosphate U937 Δψμ↓, AIF translocation, activation caspase 3-like protease 
Park et al94 (2002) Monensin Na+ ionophore 10 AML cells including HL-60 CDK6 Cyclin D1↓ Cyclin A↓, p27↑, changes in Δψμ, caspase 3, 8, Bax 
Hamada et al95 (2002) Farnesylpyridinium HL-60 cytochrome c↑, cytochalasin-like effect 
Zhang et al96 (2002) Novel retinoid CD437, MM002 WSU-B-CLL primary B-CLL ALL Caspase 2, 3↑, cleavage of Bcl-XL, activation of caspase 2, 3 
Kwon97 (2002) Diallyl disulfide HL-60 Caspase 3 hydrogen peroxide generation increased 
Gordon98 (2003) β2-microglobulin CCRF-HSB-2 ALL cell line Caspase, ROS dependent, release of cytochrome c, AIF 
Thorburn99 (2003) DT(388)GM-CSF U937 Activation of FAS-associated DD protein 
Table 4.

Medicinal plant-derived apoptosis-inducing agents and other products.

Authors (Year)ProductsOriginIn Vitro Study Cell LineMechanism
* Unpublished data Abbreviations: HHT, homoharringtonine; L, lymphoid; M, myeloid; ALL, acute lymphoblastic leukemia; ROS, reactive oxygen species; SAPK/JNK, stress-activated protein kinase/JUN IV-terminal kinase; 
Tan et al100 (2000) Extract Sophora flavescens HL-60 Apoptosis 5–20 mg/L Differentiation 1.5 mg/L 
Pan et al101 (2000) Polyphenol theasinensin A Oolong tea U937 Caspase 9↑, release of cytochrome c, ROS↑Δψμ↓ 
Kimoto et al102 (2001) Artepillin C Brazilian propolis 7 T-cell 5 B-cell myeloid, monocytic non-L, M leukemia cell lines FAS antigen expression Δψμ↓ 
Kimoto et al103 (2001) Tryptanthrin Polygonumtinctorium Lour U937 HL-60 Enhances FAS-induced apoptosis, caspase 3↑ 
Dorrie et al104 (2001) Carnosol Herb rosemary pro-B ALL pre-B ALL Bcl-2↓ Δψμ↓ 
Li et al105 (2001) Erianin Dendrobium HL-60 Bcl-2 gene↓, Bax gene↑ 
Cai et al106 (2001) HHT Cephatotaxushainanensis Li Molt3 Cytochrome c release, translocation of Bax, activation of caspase 
Anto et al107 (2002) Curcumin Curcuma longa HL-60 Caspase 8↑, cleavage of BID, cytochrome c release 
Kim et al108 (2002) Momordin I Ampelopsisjaponica HL-60 Bcl-2/Bax↓, activation of caspase 3 
Kajimoto et al109 (2002) Sophoranone Sophora tonkinensisgagnep U937 Opening of mitochondrial permeability transition pores, ROS↑ 
Candra et al110 (2002) Saponins 7,8 Liliacae L1210 Not clear 
Ueda et al111 (2002) Baicalin Scrutellarisbaicalensisgeorgi Jurkat cells Caspase 3↑, ROS↑ cytochrome c release, Δψμ↓ 
Zhu et al112 (2002) Squamocin Annonaceae (AnnonaceousacetogeninsHL-60 Activation of caspase 3 related to SAPK/JNK 
Zhou et al (2003)* SIH-10 Medicinal herb Kasumi-1 Δψμ↓, caspase 3↑ 
Authors (Year)ProductsOriginIn Vitro Study Cell LineMechanism
* Unpublished data Abbreviations: HHT, homoharringtonine; L, lymphoid; M, myeloid; ALL, acute lymphoblastic leukemia; ROS, reactive oxygen species; SAPK/JNK, stress-activated protein kinase/JUN IV-terminal kinase; 
Tan et al100 (2000) Extract Sophora flavescens HL-60 Apoptosis 5–20 mg/L Differentiation 1.5 mg/L 
Pan et al101 (2000) Polyphenol theasinensin A Oolong tea U937 Caspase 9↑, release of cytochrome c, ROS↑Δψμ↓ 
Kimoto et al102 (2001) Artepillin C Brazilian propolis 7 T-cell 5 B-cell myeloid, monocytic non-L, M leukemia cell lines FAS antigen expression Δψμ↓ 
Kimoto et al103 (2001) Tryptanthrin Polygonumtinctorium Lour U937 HL-60 Enhances FAS-induced apoptosis, caspase 3↑ 
Dorrie et al104 (2001) Carnosol Herb rosemary pro-B ALL pre-B ALL Bcl-2↓ Δψμ↓ 
Li et al105 (2001) Erianin Dendrobium HL-60 Bcl-2 gene↓, Bax gene↑ 
Cai et al106 (2001) HHT Cephatotaxushainanensis Li Molt3 Cytochrome c release, translocation of Bax, activation of caspase 
Anto et al107 (2002) Curcumin Curcuma longa HL-60 Caspase 8↑, cleavage of BID, cytochrome c release 
Kim et al108 (2002) Momordin I Ampelopsisjaponica HL-60 Bcl-2/Bax↓, activation of caspase 3 
Kajimoto et al109 (2002) Sophoranone Sophora tonkinensisgagnep U937 Opening of mitochondrial permeability transition pores, ROS↑ 
Candra et al110 (2002) Saponins 7,8 Liliacae L1210 Not clear 
Ueda et al111 (2002) Baicalin Scrutellarisbaicalensisgeorgi Jurkat cells Caspase 3↑, ROS↑ cytochrome c release, Δψμ↓ 
Zhu et al112 (2002) Squamocin Annonaceae (AnnonaceousacetogeninsHL-60 Activation of caspase 3 related to SAPK/JNK 
Zhou et al (2003)* SIH-10 Medicinal herb Kasumi-1 Δψμ↓, caspase 3↑ 

The author would like to thank Professor Z Chen for his kind advice in the preparation of this manuscript and the staff and students of the Shanghai Institute of Hematology who have made contributions to this research.

1
Wang ZY, Chen Z. Differentiation and apoptosis induction therapy in acute promyelocytic leukemia [review].
Lancet Oncol
.
2000
;
1
:
101
–106.
2
Huang ME, Ye YC, Wang ZY. Treatment of 4 APL patients with all-trans retinoic acid.
Chin J Intern Med
.
1987
;
26
:
330
–322.
3
Huang ME, Ye YC, Chen SR, et al. Use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia.
Blood
.
1988
;
72
:
567
–572.
4
Castaigne S, Chomienne C, Daniel MT, et al. All-trans retinoic acid as a differentiation therapy for acute promyelocytic leukemia, I: clinical results.
Blood
.
1990
;
76
:
1704
–1709.
5
Chen ZX, Xue YQ, Zhang R, et al. A clinical and experimental study on all-trans retinoic acid-treated acute promyelocytic leukemia.
Blood
.
1991
;
78
:
1413
–1419.
6
Sun GL, Huang YG, Chang XF, et al, Chinese Cooperative Study Group on APL. Clinical study on all-trans retinoic acid in the treatment of 544 patients with acute promyelocytic leukemia.
Chin J Hematol
.
1992
;
13
:
135
–137.
7
Sun GL, OuYang RR, Chen SJ, et al. Shanghai Cooperative Study Group on APL. Treatment of acute promyelocytic leukemia with all-trans retinoic acid: a five year experience.
Chin Med J
.
1993
;
106
:
743
–748.
8
Warrell RP Jr, Maslak P, Eardley A et al. Treatment of acute promyelocytic leukemia with all-trans retinoic acid: an update of the New York experience.
Leukemia
.
1994
;
8
:
929
–933.
9
Kanamaru A, Takemoto Y, Tanimoto M, et al. All-trans retinoic acid for the treatment of newly diagnosed acute promyelocytic leukemia.
Blood
.
1995
;
85
:
1202
–1206.
10
Tallman MS, Andersen JW, Schiffer CA, et al. All-trans retinoic acid in acute promyelocytic leukemia.
New Engl J Med
.
1997
;
337
:
1021
–1028.
11
Soignet SL, Fleishauer A, Polyak T, et al. All-trans retinoic acid significantly increases 5-year survival in patients with acute promyelocytic leukemia: long-term follow-up of the New York Study.
Cancer-Chemother-Pharmacol
.
1997
;
40
(suppl):
S25
–S29.
12
Asou N, Adachi K, Tamura J, et al. All-trans retinoic acid therapy for newly diagnosed acute promyelocytic leukemia: comparison with intensive chemotherapy. The Japan Adult Leukemia Study Group (JALSG).
Cancer-Chemother-Pharmacol
.
1997
;
40
(suppl):
S30
–S35.
13
Mandelli F, Diverio D, Avvisati G, et al. Molecular remission in PML/RAR alpha-positive acute promyelocytic leukemia by combined all-trans retinoic and idarubicin (AIDA) therapy. Gruppo Italiono-Malattie Emutologiche Maligne dell’Adulto and Associazine Italian di Ematologia ed Oncologia Pediatrica Cooperative Groups.
Blood
.
1997
;
90
:
1014
–1021.
14
Fenaux P, Chastag C, Chevret S, et al. A randomized comparison of all-trans retinoic acid (ATRA) followed by chemotherapy and ATRA plus chemotherapy and the role of maintenance therapy in newly diagnosed acute promyelocytic leukemia.
Blood
.
1999
;
94
:
1192
–1200.
15
Burnett AK, Grimwade D, Solomon E, et al. Presenting white blood cell count and kinetics of molecular remission predict prognosis in acute promyelocytic leukemia treated with all-trans retinoic acid: result of the randomized MRC trial.
Blood
.
1999
;
93
:
4131
–4143.
16
Hu J, Shen ZX, Sun GL, et al. Long-term survival and prognostic study in acute promyelocytic leukemia treated with all-trans retinoic acid, chemotherapy and As2O3: an experience of 120 patients at a single institution.
Int J Hematol
.
1999
;
70
:
248
–260.
17
Lengfelder E, Reichert A, Schoch C, et al. Double induction strategy including high dose cytarabine in combination with all-trans retinoic acid: effects in patients with newly diagnosed acute promyelocytic leukemia. German AML Cooperative Group.
Leukemia
.
2000
;
14
:
1362
–1370.
18
Asou N, Adachi K, Tamura U, et al. Analysis of prognostic factors in newly diagnosed patients with APL: the APL92 study of the Japan Adult Leukemia Study Group (JALSG).
Cancer Chemother Pharmacol
.
2001
;
48
(suppl 1):
S65
–S71.
19
Bruserud O, Gjertsen BT. New strategies for the treatment of acute myelogenous leukemia: differentiation induction, present use and future possibilities [review].
Stem Cell
.
2000
;
18
:
157
–165.
20
Degos L, Wang ZY. All-trans retinoic acid in acute pro-myelocytic leukemia [review].
Oncogene
.
2001
;
20
:
7140
–7145.
21
Fenaux P, Chevret S, Guerci A, et al. Long-term follow-up confirms the benefit of all-trans retinoic acid in acute promyelocytic leukemia. European APL Group.
Leukemia
.
2000
;
14
:
1371
–1377.
22
Chen GQ, Shen ZX, Wu F, et al. Pharmacokinetics and efficacy of low-dose all-trans retinoic acid in the treatment of acute promyelocytic leukemia.
Leukemia
.
1996
;
10
:
825
–828.
23
Douer D, Estey E, Santillana S, et al. Treatment of newly diagnosed and relapsed acute promyelocytic leukemia with intravenous liposomal all-trans retinoic acid.
Blood
.
2001
;
97
:
73
–80.
24
Tallman MS, Andersen JW, Schiffer CA, et al. All-trans retinoic acid in acute promyelocytic leukemia: long-term outcome and prognostic factor analysis from the North American Intergroup protocol.
Blood
.
2002
;
100
:
4298
–4302.
25
Fenaux P, Chomienne C, Degos L. All-trans retinoic acid and chemotherapy in the treatment of acute promyelocytic leukemia [review].
Semin Hematol
.
2001
;
38
:
13
–25.
26
de Botton S, Chevret S, Coiteux V, et al. Early onset of chemotherapy can reduce the incidence of ATRA syndrome in newly diagnosed acute promyelocytic leukemia (APL) with low blood cell counts: results from APL 93 trial.
Leukemia
.
2003
;
17
:
339
–342.
27
Collins SJ. Acute promyelocytic leukemia: relieving repression induces remission.
Blood
.
1998
;
91
:
2631
–2633.
28
Melnick A, Licht JD. Deconstructing a disease: RARalpha, its fusion partners, and their role in the pathogenesis of acute promyelocytic leukemia.
Blood
.
1999
;
93
:
3167
–3215.
29
Koken MHM, Puvion-Dutilleul F, Guillemin MC, et al. The t(15;17) translocation alters a nuclear body in a retinoic acid-reversible fashion.
EMBO J
.
1994
;
13
:
1073
–1083.
30
Nervi C, Ferrara FF, Fanelli M, et al. Caspases mediate retinoic acid-induced degradation of acute promyelocytic leukemia PML/RARα fusion protein.
Blood
.
1998
;
92
:
2244
–2251.
31
Zhu J, Gianni M, Kopf E, et al. Retinoic acid induces proteosome-dependent degradation of retinoic acid receptor α (RARα) and oncogenic RARα fusion proteins.
Proc Natl Acad Sci U S A
.
1999
;
96
:
14807
–14812.
32
Liu TX, Zhang JW, Tao J, et al. Gene expression networks underlying retinoic acid-induced differentiation of acute promyelocytic leukemia cells.
Blood
.
2000
;
96
:
1496
–1504.
33
Huang SY, Chang CS, Tang JL, et al. Acute and chronic arsenic poisoning associated with treatment in acute promyelocytic leukemia.
Br J Haematol
.
1998
;
103
:
1092
–1095.
34
Ma J, Liu JW, Chen LY, et al. Clinical observations on arsenic trioxide (As2O3) and all-trans retinoic acid (ATRA) in the treatment of acute promyelocytic leukemia (APL) [abstract 1991].
Blood
.
1998
;
92
(suppl 1):
483a
.
35
Niu C, Yan H, Yu T, et al. Studies on treatment of acute promyelocytic leukemia with arsenic trioxide: remission induction, follow-up, and molecular monitoring in 11 newly diagnosed and 47 relapsed acute promyelocytic leukemia patients.
Blood
.
1999
;
94
:
3315
–3324.
36
Lu DP, Qiu JY, Jiang B, et al. Effective treatment of acute promyelocytic leukemia (APL) with tetra-arsenic tetra-sulfide [abstract 3085].
Blood
.
1999
;
94
(suppl):
698a
.
37
Zhang P, Wang SY, Hu LH, et al. Seven year’s summary report on the treatment of acute promyelocytic leukemia with arsenic trioxide: an analysis of 242 cases.
Chin J Hematol
.
2000
;
21
:
67
–70.
38
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.
39
Mathews V, Balasubramanian P, Shaji RV, et al. Arsenic trioxide in the treatment of newly diagnosed acute promyelocytic leukemia.
Am J Hematol
.
2002
;
70
:
292
–299.
40
Lazo G, Kantarjian H, Estey E, et al. Use of arsenic trioxide (As2O3) in the treatment of patients with acute promyelocytic leukemia.
Cancer
.
2003
;
97
:
2218
–2224.
41
Chen Z, Chen GQ, Shen ZX, et al. Treatment of acute promyelocytic leukemia with arsenic trioxide compounds: in vitro and in vivo studies [review].
Semin Hematol
.
2001
;
38
:
26
–36.
42
Sun HD, Ma L, Hu XC, et al. Treatment of acute promyelocytic leukemia by Al-1 therapy.
Chin J Integrat Chin Trad Med West Med
.
1992
;
12
:
170
–172.
43
Zhang P, Wang SY, Lu LH, et al. Arsenic trioxide-treated 72 cases of acute promyelocytic leukemia.
Chin J Hematol
.
1996
;
17
:
58
–60.
44
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.
45
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 effect on APL cells in vitro and in vivo.
Blood
.
1997
;
89
:
3345
–3353.
46
Huang SL, Guo AI, Xiang Y, et al. Clinical study on the treatment of acute promyelocytic leukemia mainly with composite indigo naturalis tablets.
Chin J Hematol
.
1995
;
16
:
26
–28.
47
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.
48
Lu DP, Qiu JY, Jiang B, et al. Tetra-arsenic tetra-sulfide for the treatment of acute promyelocytic leukemia: a pilot study.
Blood
.
2002
;
99
:
3136
–3143.
49
Cai X, Shen YL, Zhu Q, et al. Arsenic trioxide-induced apoptosis and differentiation are associated respectively with mitochondrial transmembrane potential collapse and retinoic acid pathways in acute promyelocytic leukemia.
Leukemia
.
2000
;
14
:
262
–270.
50
Davison K, Mann KK, Miller WH Jr. Arsenic trioxide: mechanisms of action [review].
Semin Hematol
.
2002
;
39
(2 suppl 1):
3
–7.
51
Kitamura K, Minami Y, Yamamoto K, et al. Involvement of CD95-independent caspase 8 activation in arsenic trioxide-induced apoptosis.
Leukemia
.
2000
;
14
:
1743
–1750.
52
Mathas S, Lietz A, Janz M, et al. Inhibition of NF-kappaB essentially contributes to arsenic trioxide-induced apoptosis.
Blood
.
2003
April 3. [epub ahead of print]
53
Li J, Chen P, Sinogeeva N, et al. Arsenic trioxide promotes histone H3 phosphoacetylation at the chromatin of caspase-10 in acute promyelocytic leukemia.
J Biol Chem
.
2002
;
277
:
49504
–49510.
54
Zhu J, Koken MH, Quignon F, et al. Arsenic-induced PML targeting into nuclear bodies: implication for the treatment of APL.
Proc Natl Acad Sci U S A
.
1997
;
94
:
3978
–3983.
55
Lallemand-Breitenbach V, Zhu J, Puvion F, et al. Role of promyelocytic leukemia (PML) sumoylation in nuclear body formation, 11S proteosome recruitment and As203-induced PML or PML-retinoic acid receptor α degradation.
J Exp Med
.
2001
;
193
:
1361
–1372.
56
Lallemand-Breitenbach V, Guillemin MC, Janin A, et al. Retinoic acid and arsenic synergize to eradicate leukemic cells in a mouse model of acute promyelocytic leukemia.
J Exp Med
.
1999
;
189
:
1043
–1052.
57
Jing Y, Wang L, Xia LJ, et al. Combined effect of all-trans retinoic acid and arsenic trioxide in acute promyelocytic leukemia cells in vitro and in vivo.
Blood
.
2001
;
97
:
264
–269.
58
An WY, Chim CS, Lie AK et al. Combined arsenic trioxide and all-trans retinoic acid treatment for acute promyelocytic leukemia recurring from previous relapses successfully treated using arsenic trioxide.
Br J Haematol
.
2002
;
117
:
130
–132.
59
Liu YF, Shen ZX, Chen X, et al. Clinical observation of the short-term efficacy of the treatment with combination of all-trans retinoic acid (ATRA) and arsenic trioxide (As2O3) in newly diagnosed acute promyelocytic leukemia (APL).
Chin J Hematol
.
2003
;
24
:
25
–27.
60
Miller WH Jr, Waxman S. Differentiation induction as a treatment for hematologic malignancies [review].
Oncogene
.
2002
;
21
:
3496
–3506.
61
Wang J, Saunthararajah Y, Redner RL, Liu JM. Inhibitors of histone deacetylase relieve ETO-mediated repression and induce differentiation of AML1-leukemia cells.
Cancer Res
.
1999
;
59
:
2766
–2769.
62
Hong J, Ishihara K, Yamaki K, et al. Apicidin, a histone deacetylase inhibitor, induces differentiation of HL-60 cells.
Cancer Lett
.
2003
;
189
:
197
–206.
63
Klisovic MI, Maghraby EA, Parthun MR, et al. Depsipeptide (FR 901228) promotes histone acetylation, gene transcription, and apoptosis and its activity is enhanced by DNA methyltransferase inhibitors in AMI/ETO-positive leukemic cells.
Leukemia
.
2003
;
17
:
350
–358.
64
Shaker S, Bernstein M, Momparler LF, Momparler RL. Preclinical evaluation of antineoplastic activity of inhibitors of DNA methylation (5-aza-2′-deoxycytidine) and histone deacetylation (trichostatin A, depsipeptide) in combination against myeloid leukemic cells.
Leuk Res
.
2003
;
27
:
437
–444.
65
Liu XM, Wang LG, Li HY, Ji XJ. Induction of differentiation and down-regulation of c-myb gene expression in ML-1 human myeloblastic leukemia cells by the clinically effective anti-leukemia agent meisoindigo.
Biochem Pharmacol
.
1996
;
51
:
1545
–1551.
66
Kim SY, Gao JJ, Kang HK. Two flavonoids from the leaves of Morus alba induce differentiation of the human promyelocytic leukemia (HL-60) cell line.
Biol Pharm Bull
.
2000
;
23
:
451
–455.
67
Honma Y. Cotylenin A—a plant growth regulator as a differentiation-inducing agent against myeloid leukemia.
Leuk Lymphoma
.
2002
;
43
:
1169
–1178.
68
Liang Y, Yang YM, Yuan SL, et al. Acute promyelocytic leukemia cell differentiation induced by tanshinone IIA and its molecular mechanism.
Chin J Hematol
.
2000
;
21
:
23
–26.
69
Zhang JX, Fong WF, Wu JY, et al. Pyranocoumarins isolated from Peucedanum praeruptorum as differentiation inducers in human leukemic HL-60 cells.
Planta Med
.
2003
;
69
:
223
–229.
70
Jeong SH, Koo SJ, Choi JH, et al. Intermedeol isolated from the leaves of Ligularia fischeri var. spiciformis induces the differentiation of human acute promyelocytic leukemia HJ-60 cells.
Planta Med
.
2002
;
68
:
881
–885.
71
Lee KT, Kim JI, Park HJ, et al. Differentiation-inducing effect of magnolialide, a 1 beta-hydroxyeudesmanolide isolated from Cichorium intybus, on human leukemia cells.
Biol Pharm Bull
.
2000
;
23
:
1005
–1007.
72
Andreeff M, Stone R, Michaeli J, et al. Hexamethylene bisacetate in myelodysplastic syndrome and acute myelogenous leukemia: a phase II clinical trial with differentiation-inducing agent.
Blood
.
1992
;
80
:
2604
–2609.
73
Warrell RP Jr, He LZ, Richon V, et al. Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase.
J Natl Cancer Inst
.
1998
;
90
:
1621
–1625.
74
Chen ZX, Wang Y, Wang W, et al. All-trans retinoic acid as a single agent induces complete remission in a patient with acute leukemia of M2a subtype.
Chin Med J
.
2002
;
115
:
58
–61.
75
Tilly H, Castaigne S, Sigaux F, Degos L. Treatment of acute myeloblastic leukemia in elderly patients with low-dose cytosine arabinoside.
Presse Med
.
1985
;
14
:
827
–831.
76
Pascarella A, Marrani C, Leoni F, et al. Low-dose cytosine arabinoside in patients with acute myeloid leukemia not eligible for standard chemotherapy.
Leuk Lymphoma
.
1995
;
18
:
465
–469.
77
Yamada K, Furusawa S, Saito K, et al. Concurrent use of granulocyte colony-stimulation factor with low-dose cytosine arabinoside and aclarubicin for previously treated acute myelogenous leukemia: a pilot study.
Leukemia
.
1995
;
9
:
10
–14.
78
Huang CL, Deng ML, Guo RJ, et al. A study on the induction of differentiation of human leukemic cells by harringtonine combined with cytarabine.
Leukemia
.
1988
;
2
:
518
–522.
79
Venditti A, Stasi R, Del Poeta G, et al. All-trans retinoic acid and low-dose cytosine arabinoside for the treatment of “poor prognosis” acute myeloid leukemia.
Leukemia
.
1995
;
9
:
1121
–1125.
80
Fujimura S, Suzumiya J, Anzai K, et al. Retinoic acids induce growth inhibition and apoptosis in adult T-cell leukemia (ATL) cell lines.
Leuk Res
.
1998
;
22
:
611
–618.
81
Zhu DM, Narla RK, Fang WH, et al. Calphostin C triggers calcium-dependent apoptosis in human acute lymphoblastic leukemia cells.
Clin Cancer Res
.
1998
;
4
:
2967
–2976.
82
Uckun FM, Messinger Y, Chen CL, et al. Treatment of therapy-refractory B-lineage acute lymphoblastic leukemia with an apoptosis-inducing CD19-directed tyrosine kinase inhibitor.
Clin Cancer Res
.
1999
;
5
:
3906
–3913.
83
Dimitroulakos J, Nohynek D, Backway KL, et al. Increased sensitivity of acute myeloid leukemias to lovastatin-induced apoptosis: a potential therapeutic approach.
Blood
.
1999
;
93
:
1308
–1318.
84
Gong H, Zolzer F, von Recklinghausen G, et al. Arginine deiminase inhibits proliferation of human leukemia cells more potently than asparaginase by inducing cell cycle arrest and apoptosis.
Leukemia
.
2000
;
14
:
826
–829.
85
El-Sabban ME, Nasr R, Dbaibo G, et al. Arsenic-interferon-alpha-triggered apoptosis in HTLV-1 transformed cells is associated with tax down-regulation and reversal of NF-kappa B activation.
Blood
.
2000
;
96
:
2849
–2855.
86
Takahashi N, Kubo Y, Iwahori A, et al. Induction of apoptosis in the human promyelocytic leukemia cell line HL-60 by falconensone A and its derivatives, new polyenes.
Biol Pharm Bull
.
2000
;
23
:
748
–754.
87
Dorrie J, Gerauer H, Wachter Y, Zunino SJ. Resveratrol induces extensive apoptosis by depolarizing mitochondrial membranes and activating caspase-9 in acute lymphoblastic leukemia cells.
Cancer Res
.
2001
;
61
:
4731
–4739.
88
Kim T, Jung U, Cho DY, Chung AS. Se-methylselenocysteine induces apoptosis through caspase activation in HL-60 cells.
Carcinogenesis
.
2001
;
22
:
559
–565.
89
Nimmanapalli R, Porosnicu M, Nguyen D, et al. Cotreatment with STI-571 enhances tumor necrosis factor alpha-related apoptosis-inducing ligand (Trail or apo-L2)-induced apoptosis for Bcr-Abl-positive human acute leukemia cells.
Clin Cancer Res
.
2001
;
7
:
350
–357.
90
Ogata S, Inoue K, Iwata K, et al. Apoptosis induced by picolinic acid-related compounds in HL-60 cells.
Biosci Biotechnol Biochem
.
2001
;
65
:
2337
–2339.
91
Narla RK, Dong Y, Uckun FM. Apoptosis inducing novel anti-leukemic agent, bis(4,7-dimethyl-1,10 phenanthroline) sulfatooxovanadium (IV) [VO(SO4)(Me2-Phen)2] depolarizes mitochondrial membranes.
Leuk Lymphoma
.
2001
;
41
:
625
–634.
92
Lecureur V, Lagadic-Gossman D, Fardel O. Potassium antimonyl tartrate induced reactive oxygen species-related apoptosis in human myeloid leukemia HL-60 cells.
Int J Oncol
.
2002
;
20
:
1071
–1076.
93
Yasugi E, Uemura I, Kumagai T, et al. Disruption of mitochondria is an early event during dolichyl monophosphate-induced apoptosis in U937 cells.
Zoolog Sci
.
2002
;
19
:
7
–13.
94
Park WH, Lee MS, Park K, et al. Monensin-mediated growth inhibition in acute myelogenous leukemia cells via cell cycle arrest and apoptosis.
Int J Cancer
.
2002
;
101
:
235
–242.
95
Hamada M, Nishio K, Doe M, et al. Farnesylpyridinium, an analog of isoprenoid farnesol, induces apoptosis but suppresses apoptotic body formation in human promyelocytic leukemia cells.
FEBS Lett
.
2002
;
514
:
250
–254.
96
Zhang Y, Dawson MI, Mohammad R, et al. Induction of apoptosis of human B-CLL and ALL cells by a novel retinoid and its nonretinoidal analog.
Blood
.
2002
;
100
:
2917
–2925.
97
Kwon KB, Yoo SJ, Ryu DG, et al. Induction of apoptosis by diallyl disulfide through activation of caspase-3 in human leukemia HL-60 cells.
Biochem Pharmacol
2002
;
63
:
41
–47.
98
Gordon J, Wu CH, Rastegar M, Safa AR. Beta2-microglobulin induces caspase-dependent apoptosis in the CCRF-HSB-2 human leukemia cell line independently of the caspase-3-8 and -9 pathways but through increased reactive oxygen species.
Int J Cancer
.
2003
;
103
:
316
–327.
99
Thorburn J, Frankel AE, Thorburn A. Apoptosis by leukemia cell-targeted Diphtheria toxin occurs via receptor-independent activation of Fas-associated death domain protein.
Clin Cancer Res
.
2003
;
9
:
861
–865.
100
Tan MQ, Luo ZY, Wang QR, Jiang DZ. The anti-leukemia effect of Sophora flavescens and its mechanism.
Bull Hunan Med Univ
.
2000
;
25
:
443
–445.
101
Pan MH, Liang YC, Lin-Shiau SY, et al. Induction of apoptosis by the oolong tea polyphenol theasinensin A through cytochrome c release and activation of caspase-9 and caspase-3 in human U937 cells.
J Agric Food Chem
.
2000
;
48
:
6337
–6346.
102
Kimoto T, Aga M, Hino K, et al. Apoptosis of human leukemia cells induced by Artepillin C, an active ingredient of Brazilian propolis.
Anticancer Res
.
2001
;
21
(1A):
221
–228.
103
Kimoto T, Hino K, Koya-Miyata S, et al. Cell differentiation and apoptosis of monocytic and promyelocytic leukemia cells (U-937 and HL-60) by tryptanthrin, an active ingredient of Polygonum tinctorium Lour.
Pathol Int
.
2001
;
51
:
315
–325.
104
Dorrie J, Sapala K, Zunino SJ. Carnosol-induced apoptosis and down-regulation of Bcl-2 in B lineage leukemia cells.
Cancer Lett
.
2001
;
170
:
33
–39.
105
Li YM, Wang HY, Liu GQ. Erianin induces apoptosis in human leukemia HL-60 cells.
Acta Pharmacol Sin
.
2001
;
22
:
1018
–1022.
106
Cai Z, Lin M, Wuchter C, et al. Apoptotic response to homoharringtonine in human wt p53 leukemic cells is independent of reactive oxygen species generation and implicates BAX translocation, mitochondrial cytochrome c release and caspase activation.
Leukemia
.
2001
;
15
:
567
–574.
107
Anto RJ, Mukhopadhyay A, Denning K, Aggarwal BB. Curcumin (diferuloylmethane) induces apoptosis through activation of caspase-8, BID cleavage and cytochrome c release: its suppression by ectopic expression of Bcl-2 and Bcl-xL.
Carcinogenesis
.
2002
;
23
:
143
–150.
108
Kim JH, Ju EM, Lee DK, Hwang HJ. Induction of apoptosis by momordin I in promyelocytic leukemia (HL-60) cells.
Anticancer Res
.
2002
;
22
:
1885
–1889.
109
Kajimoto S, Takanashi N, Kajimoto T, et al. Sophoranone, extracted from a traditional Chinese medicine Shan Dou Gen, induces apoptosis in human leukemia U937 cells via formation of reactive oxygen species and opening of mitochondrial permeability transition pores.
Int J Cancer
.
2002
;
99
:
879
–890.
110
Candra E, Matsunaga K, Fujiwara H, et al. Potent apoptotic effects of saponins from Liliaceae plants in L1210 cells.
J Pharm Pharmacol
.
2002
;
54
:
257
–262.
111
Ueda S, Nakamura H, Masutani H, et al. Baicalin induces apoptosis via mitochondrial pathway as prooxidant.
Mol Immunol
.
2002
;
38
:
781
–791.
112
Zhu XF, Liu ZC, Xie BF, et al. Involvement of caspase-3 activation in squamocin-induced apoptosis in leukemia cell line HL-60.
Life Sci
.
2002
;
70
:
1259
–1269.
113
Wen J, Ramadevi N, Nguyen D, et al. Antileukemic drugs increase death receptor 5 levels and enhance Apo-2L-induced apoptosis of human acute leukemia cells.
Blood
.
2000
;
96
:
3900
–3906.
114
Robertson JD, Enoksson M, Suomela M, et al. Caspase-2 acts upstream of mitochondria to promote cytochrome c release during etoposide-induced apoptosis.
J Biol Chem
.
2002
;
277
:
29803
–29809.
115
Wu CH, Gordon J, Rastegar M, et al. Proteinase-3, a serine protease which mediates doxorubicin-induced apoptosis in the HL-60 leukemia cell line, is downregulated in the doxorubicin-resistant variant.
Oncogene
.
2002
;
21
:
5160
–5174.
116
Zhou M, Gu L, Li F, et al. DNA damage induced a novel p53-survivin signaling pathway regulating cell cycle and apoptosis in acute lymphoblastic leukemia cells.
J Pharmacol Exp Ther
.
2002
;
303
:
124
–131.
117
Stadheim TA, Kucera GL. C-Jun N-terminal kinase/stress-activated protein kinase (JUN/SAPK) is required for mitoxantrone-and anisomycin-induced apoptosis in HL-60 cells.
Leuk Res
.
2002
;
26
:
55
–65.
118
Yu R, Shtil AA, Tan TH, et al. Adriamycin activates c-jun N terminal kinase in human leukemia cells: a relevance to apoptosis.
Cancer Lett
.
1996
;
107
:
73
–81.
119
Guchelaar HJ, Vermes I, Koopmans RP, et al. Apoptosis- and necrosis-inducing potential of cladribine, cytarabine, cisplastin, and 5-fluorouracil in vitro: a quantitative pharmacodynamic model.
Cancer Chemother Pharmacol
.
1998
;
42
:
77
–83.
120
Kantarjian HM, Talpaz M, Santini V, et al. Homoharringtonine: history, current research, and future direction [review].
Cancer
.
2001
;
92
:
1591
–1605.
121
Fang M, Zhang H, Xue S, et al. Intracellular calcium distribution in apoptosis of HL-60 cells induced by harringtonine: intranuclear accumulation and regionalization.
Cancer Lett
.
1998
;
127
:
113
–121.
122
Wang J, Saunthararajah Y, Redner RL, Liu JM. Inhibitors of histone deacetylase relieve ETO-mediated repression and induce differentiation of AML1-ETO leukemia cells.
Cancer Res
.
1999
;
59
:
2766
–2769.
123
Levis M, Allebach J, Tse KF, et al. A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo.
Blood
.
2002
;
99
:
3885
–3891.
124
Elsner E, Linker-Israeli M, Umiel T, et al. Combination of a potent 20-epi-vitamin D3 analogue (KH 1060) with 9-cis-retinoic acid irreversibly inhibits clonal growth, decreases bcl-2 expression, and induces apoptosis in HL-60 leukemic cells.
Cancer Res
.
1996
;
56
:
3570
–3576.
125
Varela N, Munoz-Pinedo C, Ruiz-Ruiz C, et al. Interferon-gamma sensitizes human myeloid leukemia cells to death-receptor-mediated apoptosis by a pleiotropic mechanism.
J Biol Chem
.
2001
;
276
:
17779
–17787.
126
Wuchter C, Krappmann D, Cai Z, et al. In vitro susceptibility to TRAIL-induced apoptosis of acute leukemia cells in the context of TRAIL receptor gene expression and constitutive NF-kappa B activity.
Leukemia
.
2001
;
15
:
921
–928.
127
Zhu Q, Zhang JW, Zhu HQ, et al. Synergic effects of arsenic trioxide and cAMP during acute promyelocytic leukemia cell maturation subtends a novel signaling cross-talk.
Blood
.
2002
;
99
:
1014
–1022.
128
Perkins C, Kim CN, Fang G, Bhalla KN. Arsenic induces apoptosis of multidrug-resistant human myeloid leukemia cells that express Bcr-Abl or overexpress MDR, MRP, Bcl-2, or Bcl-x(L).
Blood
.
2000
;
95
:
1014
–1022.
129
Porosnicu M, Nimmanapalli R, Nguyen D, et al. Co-treatment with As2O3 enhances selective cytotoxic effects of STI-571 against Bcr-Abl-positive acute leukemia cells.
Leukemia
.
2001
;
15
:
772
–778.

Author notes

*

Shanghai Second Medical University, Shanghai Institute of Hematology, 197 Rui-jin Er Road, Shanghai, 200025, China This work was supported by the Chinese National Key Program for Basic Research (973), the Chinese High-Tech Program (863), the National Natural Science Foundation of China, the Shanghai Commission for Science and Technology, and the Samuel Waxman Cancer Research and Clyde Wu Foundations.