Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
1.
Leukemia ; 20(2): 212-7, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16357841

ABSTRACT

Epigenetic mechanisms underlying tumorigenesis have recently received much attention as potential therapeutic targets of human cancer. We designed a pilot study to target DNA methylation and histone deacetylation through the sequential administration of 5-azacytidine followed by sodium phenylbutyrate (PB) in patients with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Ten evaluable patients (eight AML, two MDS) were treated with seven consecutive daily subcutaneous injections of 5-azacytidine at 75 mg/m2 followed by 5 days of sodium PB given intravenously at a dose of 200 mg/kg. Five patients (50%) were able to achieve a beneficial clinical response (partial remission or stable disease). One patient with MDS proceeded to allogeneic stem cell transplantation and is alive without evidence of disease 39 months later. The combination regimen was well tolerated with common toxicities of injection site skin reaction (90% of the patients) from 5-azacytidine, and somnolence/fatigue from the sodium PB infusion (80% of the patients). Correlative laboratory studies demonstrated the consistent reacetylation of histone H4, although no relationship with the clinical response could be demonstrated. Results from this pilot study demonstrate that a combination approach targeting different mechanisms of transcriptional modulation is clinically feasible with acceptable toxicity and measurable biologic and clinical outcomes.


Subject(s)
Antineoplastic Agents/therapeutic use , Azacitidine/therapeutic use , Leukemia, Myeloid/drug therapy , Myelodysplastic Syndromes/drug therapy , Phenylbutyrates/therapeutic use , Transcription, Genetic/drug effects , Acetylation/drug effects , Acute Disease , Adult , Aged , Antimetabolites, Antineoplastic/therapeutic use , DNA Methylation , Drug Administration Schedule , Drug Therapy, Combination , Female , Histones/drug effects , Histones/metabolism , Humans , Leukemia, Myeloid/diagnosis , Leukemia, Myeloid/genetics , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Pilot Projects , Treatment Outcome
2.
Leukemia ; 18(7): 1258-69, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15116119

ABSTRACT

AP-1060 is a newly established acute promyelocytic leukemia (APL) cell line from a multiple-relapse patient clinically resistant to both all-trans retinoic acid (ATRA) and arsenic trioxide (ATO). The line was initially derived as a granulocyte colony-stimulating factor-dependent strain that underwent replicative senescence and, following ethylnitrosourea treatment, as a phenotypically similar immortalized line. Immortalization was associated with broadened cytokine sensitivity but not growth autonomy, in contrast to three previously derived APL lines. Both the AP-1060 strain and line had shortened telomeres and low telomerase activity, while the line had higher expression of many genes associated with macromolecular synthesis. The karyotype was 46,XY,t(3;14)(p21.1;q11.2),t(15;17)(q22;q11)[100%]; the unique t(3;14) was observed in 4/9 t(15;17)-positive metaphase cells at previous relapse on ATRA therapy. The PML-RARalpha mRNA harbored a missense mutation in the RARalpha-region ligand-binding domain (Pro900Ser). This was associated with a right-shift and sharpening of the ATRA-induced maturation response compared to ATRA-sensitive NB4 cells, which corresponded to the transcriptional activation by PML-RARalphaPro900Ser of a cotransfected ATRA-targeted reporter vector in COS-1 cells. AP-1060 also manifested relative resistance to ATO-induced apoptosis at >/=1 microM, while 0.25 microM ATO stimulated limited atypical maturation. These findings suggest that AP-1060 will be useful for further assessing molecular elements involved in APL progression and drug response/resistance.


Subject(s)
Cell Line, Tumor , Drug Resistance, Neoplasm , Leukemia, Promyelocytic, Acute/pathology , Arsenic Trioxide , Arsenicals/pharmacology , Cell Culture Techniques/methods , Cytokines/pharmacology , Humans , Karyotyping , Mutation, Missense , Oxides/pharmacology , Receptors, Retinoic Acid/genetics , Retinoic Acid Receptor alpha , Telomerase/metabolism , Telomere/ultrastructure , Tretinoin/pharmacology
3.
J Clin Invest ; 108(9): 1321-30, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11696577

ABSTRACT

Acute promyelocytic leukemia (APL) is associated with chromosomal translocations, invariably involving the retinoic acid receptor alpha (RAR alpha) gene fused to one of several distinct loci, including the PML or PLZF genes, involved in t(15;17) or t(11;17), respectively. Patients with t(15;17) APL respond well to retinoic acid (RA) and other treatments, whereas those with t(11;17) APL do not. The PML-RAR alpha and PLZF-RAR alpha fusion oncoproteins function as aberrant transcriptional repressors, in part by recruiting nuclear receptor-transcriptional corepressors and histone deacetylases (HDACs). Transgenic mice harboring the RAR alpha fusion genes develop forms of leukemia that faithfully recapitulate both the clinical features and the response to RA observed in humans with the corresponding translocations. Here, we investigated the effects of HDAC inhibitors (HDACIs) in vitro and in these animal models. In cells from PLZF-RAR alpha/RAR alpha-PLZF transgenic mice and cells harboring t(15;17), HDACIs induced apoptosis and dramatic growth inhibition, effects that could be potentiated by RA. HDACIs also increased RA-induced differentiation. HDACIs, but not RA, induced accumulation of acetylated histones. Using microarray analysis, we identified genes induced by RA, HDACIs, or both together. In combination with RA, all HDACIs tested overcame the transcriptional repression exerted by the RAR alpha fusion oncoproteins. In vivo, HDACIs induced accumulation of acetylated histones in target organs. Strikingly, this combination of agents induced leukemia remission and prolonged survival, without apparent toxic side effects.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Remission Induction , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Blotting, Northern , Blotting, Western , Cell Cycle , Cell Differentiation , Cell Division , DNA, Complementary/metabolism , Humans , Hydroxamic Acids/pharmacology , In Situ Nick-End Labeling , Mice , Mice, Transgenic , Microscopy, Fluorescence , Models, Chemical , Oligonucleotide Array Sequence Analysis , Phenylbutyrates/pharmacology , Protein Binding , Receptors, Retinoic Acid/genetics , Retinoic Acid Receptor alpha , Time Factors , Transcription, Genetic , Transcriptional Activation , Tumor Cells, Cultured , Up-Regulation , Vorinostat
4.
Blood ; 98(9): 2651-6, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11675334

ABSTRACT

The t(15;17) translocation in acute promyelocytic leukemia (APL) yields a PML/RAR-alpha fusion messenger RNA species that can be detected by reverse transcription-polymerase chain reaction (RT-PCR) amplification. Breakpoints within intron 3 of PML produce a short PML/RAR-alpha isoform, whereas breakpoints within intron 6 result in a longer form. Using RT-PCR, serial evaluations were performed on the bone marrow of 82 patients with APL (median follow-up, > 63 months) who received retinoic acid (RA) induction followed by postremission treatment with chemotherapy, RA, and biologic agents. Sixty-four patients attained a clinical complete remission and had at least 2 RT-PCR assays performed after completing therapy. Forty of 47 patients (85%) with newly diagnosed APL who were induced using RA had residual disease detectable by RT-PCR before additional therapy. After 3 cycles of consolidation therapy, residual disease was found in only 4 of 40 evaluable patients (10%). Among newly diagnosed patients who had 2 or more negative RT-PCR assays, only 3 of 41 (7%) had a relapse, whereas all 4 patients (100%) who had 2 or more positive results had a relapse. Among 63 newly diagnosed patients, those who expressed the short isoform appeared to have shorter disease-free and overall survival durations than patients who expressed the long isoform. These data indicate that 2 or more negative RT-PCR assays on bone marrow, performed at least 1 month apart after completing therapy, are strongly associated with long-term remissions. Conversely, a confirmed positive test is highly predictive of relapse.


Subject(s)
Leukemia, Promyelocytic, Acute/diagnosis , Neoplasm Proteins/genetics , Neoplasm, Residual/genetics , Oncogene Proteins, Fusion/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Bone Marrow/metabolism , Child , Follow-Up Studies , Humans , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/therapy , Middle Aged , Neoplasm, Residual/diagnosis , Predictive Value of Tests , Prognosis , Prospective Studies , Protein Isoforms/genetics , RNA, Messenger/analysis , Recurrence , Remission Induction , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis
5.
J Clin Oncol ; 19(18): 3852-60, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11559723

ABSTRACT

PURPOSE: To determine the safety and efficacy of arsenic trioxide (ATO) in patients with relapsed acute promyelocytic leukemia (APL). PATIENTS AND METHODS: Forty patients experiencing first (n = 21) or > or = second (n = 19) relapse were treated with daily infusions of ATO to a maximum of 60 doses or until all leukemic cells in bone marrow were eliminated. Patients who achieved a complete remission (CR) were offered one consolidation course of ATO that began 3 to 4 weeks later. Patients who remained in CR were eligible to receive further cycles of ATO therapy on a maintenance study. RESULTS: Thirty-four patients (85%) achieved a CR. Thirty-one patients (91%) with CRs had posttreatment cytogenetic tests negative for t(15;17). Eighty-six percent of the patients who were assessable by reverse transcriptase polymerase chain reaction converted from positive to negative for the promyelocytic leukemia/retinoic acid receptor-alpha transcript by the completion of their consolidation therapy. Thirty-two patients received consolidation therapy, and 18 received additional ATO as maintenance. Eleven patients underwent allogeneic (n = 8) or autologous (n = 3) transplant after ATO treatment. The 18-month overall and relapse-free survival (RFS) estimates were 66% and 56%, respectively. Twenty patients (50%) had leukocytosis (> 10,000 WBC/microL) during induction therapy. Ten patients developed signs or symptoms suggestive of the APL syndrome and were effectively treated with dexamethasone. Electrocardiographic QT prolongation was common (63%). One patient had an absolute QT interval of > 500 msec and had an asymptomatic 7-beat run of torsades de pointe. Two patients died during induction, neither from drug-related causes. CONCLUSION: This study establishes ATO as a highly effective therapy for patients with relapsed APL.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenicals/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Oxides/therapeutic use , Adolescent , Adult , Arsenic Trioxide , Arsenicals/adverse effects , Electrocardiography , Female , Humans , Leukemia, Promyelocytic, Acute/blood , Leukemia, Promyelocytic, Acute/pathology , Leukocytosis/chemically induced , Male , Middle Aged , Nervous System Diseases/chemically induced , Oxides/adverse effects , Pilot Projects , Platelet Count , Remission Induction , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Syndrome
6.
Curr Oncol Rep ; 2(6): 519-23, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11122887

ABSTRACT

Acute promyelocytic leukemia (APL) is the most potentially curable type of acute myeloid leukemia. It is characterized by the chromosomal translocation t(15;17), which results in the fusion gene PML-RAR-alpha. The introduction of all-trans- retinoic acid (ATRA) was a major advance in treatment of this disease. This agent induces terminal differentiation of malignant myeloid cells to mature neutrophils, and its side effects are usually well tolerated in children. ATRA does not eradicate the malignant myeloid clone in APL and, eventually, resistance develops. Arsenic trioxide induces nonterminal differentiation of malignant promyelocytes and promotes apoptosis. APL patients treated with ATRA or arsenic trioxide have rapid resolution of their coagulopathy. Because both of these drugs are well tolerated in children and their synergy has been shown in animal models, the possibility of combining ATRA and arsenic trioxide in front-line therapy for children with APL is being considered.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Oxides/pharmacology , Tretinoin/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Arsenic Trioxide , Arsenicals/administration & dosage , Cell Differentiation , Child , Disease Models, Animal , Drug Therapy, Combination , Humans , Oxides/administration & dosage , Tretinoin/administration & dosage
7.
Semin Oncol ; 27(5): 495-501, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11049017

ABSTRACT

Arsenic trioxide (AT) has been the object of renewed interest as a therapeutic since studies in China in the late 1980s confirmed its efficacy in the treatment of acute promyelocytic leukemia (APL). These studies have been replicated in the West, with complete remissions achieved in 80% to 90% of patients with refractory or relapsed APL. The drug has been relatively well tolerated. The dose used for treatment of APL (0.15 mg/kg/d) is approximately 50% of the maximum-tolerated dose (MTD). Common side effects have included fatigue, rash, fluid retention, and QTc-interval prolongation on electrocardiogram. A "retinoic acid syndrome," similar in its manifestations to that noted after administration of all-trans retinoic acid (RA), has been observed in APL patients. Recent studies have included dose-ranging trials to determine pharmacokinetics and the optimum schedule of administration, and studies of possible mechanisms of action. Promising future trials include combining AT with RA in the treatment of newly diagnosed APL, and broadening the range of AT therapy to other leukemias, lymphomas, multiple myeloma and some solid tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenicals/therapeutic use , Hematologic Neoplasms/drug therapy , Oxides/therapeutic use , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Apoptosis , Arsenic Trioxide , Arsenicals/adverse effects , Arsenicals/pharmacokinetics , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Oxides/adverse effects , Oxides/pharmacokinetics , Tumor Cells, Cultured
8.
Cancer Chemother Pharmacol ; 46(4): 313-8, 2000.
Article in English | MEDLINE | ID: mdl-11052629

ABSTRACT

Retinoids have shown promising activity for both cancer chemoprevention and as a treatment for emphysema. However, chronic oral administration of these drugs is limited by systemic side effects, including hepatic dysfunction, skeletal malformations, hyperlipidemia. hypercalcemia, and other reactions. In order to improve the pulmonary targeting of this potentially useful therapy, we developed a system for aerosolization of retinoids that substantially increased their local bioavailability. We compared the biodistribution and pharmacokinetics of an inhaled formulation of all-trans-retinoic acid (all-trans-RA), which was packaged in a metered dose inhaler, following both intratracheal (IT) and intravenous (IV) administration in male Sprague-Dawley rats. After drug administration, anesthetized animals were killed at 5 min, and at 1, 2, 4, 6 and 24 h. Plasma and emulsified samples of liver and lung tissues were dissected, extracted, and frozen prior to measurement of all-trans-RA concentration by high-performance liquid chromatography (HPLC). Aerosolization and IT injection of all-trans-RA resulted in a significantly longer pulmonary half-life of the drug (both 5-17 h), lower peak serum concentrations (aerosol 71 +/- 31 ng/ml, IT 68 +/- 50 ng/ml), and lower liver levels (aerosol 111 +/- 28 ng/g, IT 753 +/- 350 ng/g) than the same dose administered IV (2 h, 838 +/- 56 ng/ml, 4,258 +/- 1,006 ng/g, respectively; P < 0.05 for each comparison). Histologic examination of lungs and trachea showed no focal irritation attributable to the drug after single-dose administration. These results suggest that aerosolization of retinoids may offer a practical alternative to systemic oral administration for chemoprevention trials or treatment of lung diseases. This method may substantially increase the therapeutic index of these compounds by reducing systemic complications associated with long-term dosing.


Subject(s)
Lung/metabolism , Tretinoin/administration & dosage , Administration, Inhalation , Aerosols , Animals , Male , Rats , Rats, Sprague-Dawley , Tretinoin/pharmacokinetics , Tretinoin/toxicity
9.
Proc Natl Acad Sci U S A ; 97(18): 10173-8, 2000 Aug 29.
Article in English | MEDLINE | ID: mdl-10954752

ABSTRACT

Acute promyelocytic leukemia (APL) is associated with chromosomal translocations always involving the RARalpha gene, which variably fuses to one of several distinct loci, including PML or PLZF (X genes) in t(15;17) or t(11;17), respectively. APL in patients harboring t(15;17) responds well to retinoic acid (RA) treatment and chemotherapy, whereas t(11;17) APL responds poorly to both treatments, thus defining a distinct syndrome. Here, we show that RA, As(2)O(3), and RA + As(2)O(3) prolonged survival in either leukemic PML-RARalpha transgenic mice or nude mice transplanted with PML-RARalpha leukemic cells. RA + As(2)O(3) prolonged survival compared with treatment with either drug alone. In contrast, neither in PLZF-RARalpha transgenic mice nor in nude mice transplanted with PLZF-RARalpha cells did any of the three regimens induce complete disease remission. Unexpectedly, therapeutic doses of RA and RA + As(2)O(3) can induce, both in vivo and in vitro, the degradation of either PML-RARalpha or PLZF-RARalpha proteins, suggesting that the maintenance of the leukemic phenotype depends on the continuous presence of the former, but not the latter. Our findings lead to three major conclusions with relevant therapeutic implications: (i) the X-RARalpha oncoprotein directly determines response to treatment and plays a distinct role in the maintenance of the malignant phenotype; (ii) As(2)O(3) and/or As(2)O(3) + RA combination may be beneficial for the treatment of t(15;17) APL but not for t(11;17) APL; and (iii) therapeutic strategies aimed solely at degrading the X-RARalpha oncoprotein may not be effective in t(11;17) APL.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenicals/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Oxides/therapeutic use , Tretinoin/therapeutic use , Animals , Apoptosis/drug effects , Arsenic Trioxide , Arsenicals/pharmacology , Cell Differentiation/drug effects , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 15 , Chromosomes, Human, Pair 17 , Humans , Leukemia, Promyelocytic, Acute/pathology , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Proteins/drug effects , Oncogene Proteins, Fusion/drug effects , Oxides/pharmacology , Translocation, Genetic , Transplantation, Heterologous , Tretinoin/pharmacology
10.
Blood ; 96(4): 1525-30, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10942401

ABSTRACT

Arsenic trioxide (As(2)O(3)) has recently been used successfully in the treatment of acute promyelocytic leukemia and has been shown to induce partial differentiation and apoptosis of leukemic cells in vitro. However, the mechanism by which As(2)O(3) exerts its antileukemic effect remains uncertain. Emerging data suggest that the endothelium and angiogenesis play a seminal role in the proliferation of liquid tumors, such as leukemia. We have shown that activated endothelial cells release cytokines that may stimulate leukemic cell growth. Leukemic cells, in turn, can release endothelial growth factors, such as vascular endothelial growth factor (VEGF). On the basis of these observations, we hypothesized that As(2)O(3) may interrupt a reciprocal loop between leukemic cells and the endothelium by direct action on both cell types. We have shown that treatment of proliferating layers of human umbilical vein endothelial cells (HUVECs) with a variety of concentrations of As(2)O(3) results in a reproducible dose- and time-dependent sequence of events marked by change to an activated morphology, up-regulation of endothelial cell adhesion markers, and apoptosis. Also, treatment with As(2)O(3) caused inhibition of VEGF production in the leukemic cell line HEL. Finally, incubation of HUVECs with As(2)O(3) prevented capillary tubule and branch formation in an in vitro endothelial cell-differentiation assay. In conclusion, we believe that As(2)O(3 )interrupts a reciprocal stimulatory loop between leukemic cells and endothelial cells by causing apoptosis of both cell types and by inhibiting leukemic cell VEGF production. (Blood. 2000;96:1525-1530)


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arsenicals/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Oxides/pharmacology , Arsenic Trioxide , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Leukemia/drug therapy , Leukemia/pathology , Neovascularization, Pathologic/drug therapy , Time Factors
11.
J Clin Oncol ; 18(13): 2620-5, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10893295

ABSTRACT

PURPOSE: Arsenic trioxide, like all-trans-retinoic acid (RA), induces differentiation of acute promyelocytic leukemia (APL) cells in vivo. Treatment of APL patients with all-trans RA is commonly associated with leukocytosis, and approximately 50% of patients develop the RA syndrome. We reviewed our clinical experience with arsenic trioxide to determine the incidence of these two phenomena. PATIENTS AND METHODS: Twenty-six patients with relapsed or refractory APL were treated with arsenic trioxide for remission induction at daily doses that ranged from 0.06 to 0.17 mg/kg. RESULTS: Twenty-three patients (88%) achieved complete remission. Leukocytosis was observed in 15 patients (58%). The median baseline leukocyte count for patients with leukocytosis was 3,900 cells/microL (range, 1,200 to 72,300 cells/microL), which was higher than that for patients who did not develop leukocytosis (2,100 cells/microL; range, 500 to 5,400 cells/microL; P =.01). No other cytotoxic therapy was administered, and the leukocytosis resolved in all cases. The RA syndrome was observed in eight patients (31%). Patients who developed leukocytosis were significantly more likely to develop the RA syndrome (P <.001), and no patient without a peak leukocyte count greater than 10,000 cells/microL developed the syndrome. Among the patients with leukocytosis, there was no observed relation between the leukocyte peak and the probability of developing the syndrome (P =.37). CONCLUSION: Induction therapy of APL with all-trans RA and arsenic trioxide is associated with leukocytosis and the RA syndrome. These clinical effects seem to be intrinsically related to the biologic responsiveness and the differentiation process induced by these new agents.


Subject(s)
Antineoplastic Agents/adverse effects , Arsenicals/adverse effects , Leukemia, Promyelocytic, Acute/drug therapy , Leukocytosis/chemically induced , Oxides/adverse effects , Tretinoin/adverse effects , Arsenic Trioxide , Dyspnea/chemically induced , Fever/chemically induced , Humans , Pleural Effusion/chemically induced , Syndrome
12.
Clin Cancer Res ; 6(5): 1731-5, 2000 May.
Article in English | MEDLINE | ID: mdl-10815891

ABSTRACT

Retinoids mediate their biological response by binding to specific nuclear receptors, including retinoic acid receptors and/or retinoid X receptors. LGD1550 is a high-affinity ligand for all three retinoic acid receptors (alpha, beta, and gamma isoforms) and a potent inhibitor of AP-1, a protein that is closely linked with trophic responses and malignant transformation. We conducted a dose ranging study to evaluate the pharmacokinetics, safety, clinical tolerance, and potential efficacy of this drug in patients with advanced cancer. Twenty-seven patients received oral doses of LGD1550 once per day at doses ranging from 20-400 microg/m2. Skin toxicity was the dose-limiting reaction at the 400 microg/m2 daily dose level. Less prominent reactions included nausea and headache. No major antitumor effects were observed. Pharmacokinetic studies in 15 patients at five dose levels showed that the peak plasma concentration (Cmax) and areas under the plasma concentration-time curve on day 1 were dose-proportional and were similar to values obtained on days 15, 29, and 84. Unlike other retinoids, LGD1550 did not induce its own metabolism, and there was little evidence of drug accumulation. The t1/2 was approximately 5 h after both the initial and repeated doses. We conclude that once-daily doses of LGD1550 of up to 300 microg/m2 are relatively well tolerated. Additional clinical explorations are warranted, especially in patients with cancers of the prostate, thyroid, head and neck, and cervix.


Subject(s)
Fatty Acids, Unsaturated/therapeutic use , Neoplasms/drug therapy , Receptors, Retinoic Acid/agonists , Adult , Aged , Area Under Curve , Binding, Competitive , Dose-Response Relationship, Drug , Fatty Acids, Unsaturated/adverse effects , Fatty Acids, Unsaturated/pharmacokinetics , Humans , Ligands , Middle Aged , Neoplasms/pathology , Treatment Outcome
13.
Cancer Chemother Pharmacol ; 44(5): 417-21, 1999.
Article in English | MEDLINE | ID: mdl-10501916

ABSTRACT

Inorganic arsenic trioxide (As(2)O(3)) induces a high proportion of complete remissions in relapsed patients with acute promyelocytic leukemia (APL). Previously, we have shown that both As(2)O(3 )and melarsoprol, an organic arsenical used for the treatment of trypanosomiasis, exhibit broad antileukemic activity against both chronic and acute myeloid and lymphoid leukemia cell lines. Given the breadth of this activity, we initiated a clinical study to evaluate the pharmacokinetics, safety, and potential efficacy of melarsoprol in patients with refractory or resistant leukemia. Using the antitrypanosomal dose and schedule, patients received escalating intravenous doses daily for 3 days, repeated weekly for 3 weeks. Doses were 1 mg/kg on day 1, 2 mg/kg on day 2, and 3.6 mg/kg on day 3 and on all days thereafter, up to a maximum daily dose of 200 mg. Eight patients [6 AML (2 morphologic APL), 1 CML, 1 CLL] were treated. Mean peak plasma concentrations of the parent drug were obtained immediately after injection, ranged from 1.2 microg/ml on day 1 to 2.4 microg/ml on day 3, were dose proportional, and decayed with a t(1/2) congruent with 15 min. A minor clinical response (regression of splenomegaly and lymphadenopathy) was observed in a patient with chronic lymphocytic leukemia. Central nervous system (CNS) toxicity proved limiting on this dose and schedule. Three patients experienced generalized grand mal seizures during the second week of therapy. We concluded that this dose and schedule of melarsoprol is associated with excessive CNS toxicity and that verification of the striking preclinical activity in patients with leukemia will require developing an alternative dose and schedule.


Subject(s)
Leukemia/drug therapy , Melarsoprol/adverse effects , Adult , Aged , Area Under Curve , Female , Humans , Injections, Intravenous , Leukemia/blood , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Male , Melarsoprol/administration & dosage , Melarsoprol/blood , Melarsoprol/pharmacokinetics , Middle Aged
15.
N Engl J Med ; 339(19): 1341-8, 1998 Nov 05.
Article in English | MEDLINE | ID: mdl-9801394

ABSTRACT

BACKGROUND: Two reports from China have suggested that arsenic trioxide can induce complete remissions in patients with acute promyelocytic leukemia (APL). We evaluated this drug in patients with APL in an attempt to elucidate its mechanism of action. METHODS: Twelve patients with APL who had relapsed after extensive prior therapy were treated with arsenic trioxide at doses ranging from 0.06 to 0.2 mg per kilogram of body weight per day until visible leukemic cells were eliminated from the bone marrow. Bone marrow mononuclear cells were serially monitored by flow cytometry for immunophenotype, fluorescence in situ hybridization, reverse-transcription-polymerase-chain-reaction (RT-PCR) assay for PML-RAR-alpha fusion transcripts, and Western blot analysis for expression of the apoptosis-associated proteins caspases 1, 2, and 3. RESULTS: Of the 12 patients studied, 11 achieved complete remission after treatment that lasted from 12 to 39 days (range of cumulative doses, 160 to 495 mg). Adverse effects were relatively mild and included rash, lightheadedness, fatigue, and musculoskeletal pain. Cells that expressed both CD11b and CD33 (antigens characteristic of mature and immature cells, respectively), and which were found by fluorescence in situ hybridization to carry the t(15;17) translocation, increased progressively in number during treatment and persisted in the early phase of complete remission. Eight of 11 patients who initially tested positive for the PML-RAR-alpha fusion transcript by the RT-PCR assay later tested negative; 3 other patients, who persistently tested positive, relapsed early. Arsenic trioxide induced the expression of the proenzymes of caspase 2 and caspase 3 and activation of both caspase 1 and caspase 3. CONCLUSIONS: Low doses of arsenic trioxide can induce complete remissions in patients with APL who have relapsed. The clinical response is associated with incomplete cytodifferentiation and the induction of apoptosis with caspase activation in leukemic cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenicals/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Oxides/therapeutic use , Adolescent , Adult , Aged , Antigens, CD/analysis , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Apoptosis , Arsenic Trioxide , Arsenicals/administration & dosage , Arsenicals/adverse effects , Bone Marrow Cells/immunology , Caspases , Cell Differentiation , Child , Humans , Immunophenotyping , Leukemia, Promyelocytic, Acute/immunology , Leukemia, Promyelocytic, Acute/pathology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Middle Aged , Neoplasm Proteins/analysis , Neoplasm Proteins/drug effects , Oncogene Proteins, Fusion/analysis , Oncogene Proteins, Fusion/drug effects , Oxides/administration & dosage , Oxides/adverse effects , Recurrence , Remission Induction/methods
16.
J Natl Cancer Inst ; 90(21): 1621-5, 1998 Nov 04.
Article in English | MEDLINE | ID: mdl-9811311

ABSTRACT

BACKGROUND: Acetylation of DNA-associated histones is linked to activation of gene transcription, whereas histone deacetylation is associated with transcriptional repression. Recent studies have shown that inhibitors of histone deacetylases can relieve transcriptional repression caused by the products of certain oncogenes. We tested whether these findings could be applied clinically to a patient with highly resistant acute promyelocytic leukemia. METHODS: A patient who had experienced multiple relapses was treated with all-trans-retinoic acid alone and in combination with sodium phenylbutyrate, an inhibitor of histone deacetylases. Immunohistochemistry and western blot analysis were used to assay for histone hyperacetylation in mononuclear cells from the patient's blood and bone marrow. Marrow mononuclear cells and reverse transcription-polymerase chain reaction (RT-PCR) analysis of messenger RNA encoded by the PML/RAR-alpha oncogene were used to assess minimal residual disease. RESULTS: The patient proved clinically resistant to treatment with all-trans-retinoic acid alone. However, 23 days after sodium phenylbutyrate was added to the treatment regimen, visible leukemic cells had been eliminated from her bone marrow, and she achieved a complete clinical and cytogenetic remission shortly thereafter. With a second treatment course, analysis for minimal residual disease by RT-PCR proved negative. Immunofluorescence and western blot analysis showed that phenylbutyrate caused a time-dependent increase in histone acetylation in blood and bone marrow mononuclear cells. CONCLUSIONS: Clinical treatment with an inhibitor of histone deacetylase induces histone hyperacetylation in target cells and may restore sensitivity to the anti-leukemic effects of all-trans-retinoic acid in acute promyelocytic leukemia. Similar therapy may prove useful in other neoplastic diseases that are associated with oncogenic repression of gene transcription due to recruitment of histone deacetylases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Histone Deacetylases/drug effects , Leukemia, Promyelocytic, Acute/drug therapy , Transcription, Genetic/drug effects , Acetylation/drug effects , Adolescent , Blotting, Western , Drug Administration Schedule , Female , Fluorescent Antibody Technique , Humans , Leukemia, Promyelocytic, Acute/enzymology , Phenylbutyrates/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , Tretinoin/administration & dosage
17.
Leukemia ; 12(10): 1518-21, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9766493

ABSTRACT

The use of all-trans retinoic acid (RA) for remission induction markedly increases survival of patients with acute promyelocytic leukemia (APL) compared to patients treated solely with cytotoxic chemotherapy. However, clinical resistance to this agent develops rapidly, which has been associated with a progressive decline in plasma drug concentrations. Previous studies suggested that 9-cis RA, a retinoid receptor 'pan agonist' did not induce its own catabolism to the same extent as all-trans RA. Therefore, we conducted a dose-ranging study of this compound in patients with both relapsed and newly diagnosed APL. We treated 18 patients with morphologically diagnosed APL (13 relapsed, five newly diagnosed). The daily dose of 9-cis RA ranged from 30 to 230 mg/m2/day given as a single oral dose. Four of 12 (33%) relapsed patients (three of whom were previously treated with all-trans RA) and four of five (80%) newly diagnosed patients achieved complete remission. The sole failure in the newly diagnosed group died early from an intracranial hemorrhage. One other patient with t(9;12) translocation had substantial hematologic improvement. The drug was generally well tolerated; headache and dry skin were the most common adverse reactions. Three patients were treated with corticosteroids for signs of incipient 'RA syndrome.' These preliminary data suggest that 9-cis RA is an effective agent for remission induction and deserves further investigation in patients with retinoid-sensitive APL.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Tretinoin/therapeutic use , Adolescent , Adult , Aged , Alitretinoin , Antineoplastic Agents/adverse effects , Child , Child, Preschool , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 9 , Dose-Response Relationship, Drug , Female , Humans , Infant , Leukemia, Promyelocytic, Acute/blood , Leukemia, Promyelocytic, Acute/genetics , Leukocyte Count/drug effects , Male , Middle Aged , Platelet Count/drug effects , Recurrence , Translocation, Genetic , Tretinoin/adverse effects
18.
Blood ; 92(5): 1497-504, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9716575

ABSTRACT

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-RARalpha 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-RARalpha 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-RARalpha 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-RARalpha 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-RARalpha 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.


Subject(s)
Apoptosis/drug effects , Arsenicals/pharmacology , Leukemia, Myeloid/pathology , Melarsoprol/pharmacology , Neoplasm Proteins/physiology , Nuclear Proteins , Oxides/pharmacology , Receptors, Retinoic Acid/physiology , Transcription Factors/physiology , Animals , Antineoplastic Agents/pharmacology , Arsenic Trioxide , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Division/drug effects , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Humans , Leukemia, Promyelocytic, Acute/pathology , Mice , Neoplasm Proteins/analysis , Promyelocytic Leukemia Protein , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Retinoic Acid/analysis , Transcription Factors/analysis , Tumor Cells, Cultured , Tumor Suppressor Proteins
19.
Cancer ; 80(8 Suppl): 1680-5, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9362436

ABSTRACT

Gallium nitrate was originally developed as an antineoplastic agent; however, further studies have revealed that this drug has extremely potent effects on turnover of bone, and that low doses can be used to reduce bone resorption. Like the bisphosphonates, gallium nitrate has been studied in both malignant and in nonmalignant conditions. The results of randomized double blind studies have suggested that this drug has superior clinical efficacy relative to etidronate, calcitonin, and pamidronate for the acute control of cancer-related hypercalcemia. In patients with Paget's disease, low doses of gallium nitrate reduce biochemical parameters of accelerated bone turnover, including urinary excretion of calcium, hydroxyproline, and urinary collagen cross-linked N-telopeptides. Preliminary studies showed similar effects in patients with bone involvement from a wide variety of tumor types. Based on this high degree of clinical potency revealed in clinical studies, two randomized Phase III studies have been initiated in patients with bone metastases from breast carcinoma and bone involvement due to multiple myeloma. Both studies employ cyclic therapy with low dose gallium nitrate (i.e., 40 mg administered as a subcutaneous injection once daily for 2 weeks, followed by 2 weeks off treatment, recycled monthly). The endpoints of both studies are to document reductions in time to "morbid skeletal events," such as palliative skeletal radiotherapy, stabilizing orthopedic surgery, or pathologic fractures, as well as decreases in pain and analgesic requirements and improvements in mobility and other aspects of quality of life. These trials should provide definitive evidence of whether this agent is safe and effective as a treatment for bone metastases.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Bone Resorption/prevention & control , Bone and Bones/drug effects , Gallium/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Gallium/pharmacology , Humans , Rats
20.
Blood ; 90(2): 562-70, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9226155

ABSTRACT

Inorganic arsenic trioxide (As2O3) was recently shown to induce apoptosis in NB4 promyelocytic leukemic cells. We have investigated the effects of the organic arsenical, melarsoprol (a drug used for treatment of trypanosomiasis), upon induction of apoptosis in cell lines representative of chronic B-cell lymphoproliferative disorders. An Epstein-Barr virus (EBV)-transformed B-prolymphocytic cell line (JVM-2), an EBV-transformed B-cell chronic lymphocytic leukemia (B-CLL) cell line (I83CLL), and one non-EBV-transformed B-CLL cell line (WSU-CLL) were used as targets. Dose-response experiments with melarsoprol (10(-7) to 10(-9) mol/L) were performed over 96 hours. Unexpectedly, we found that melarsoprol caused a dose- and time-dependent inhibition of survival and growth in all three cell lines. In contrast, As2O3 at similar concentrations had no effect on either viability or growth. After 24 hours, all three cell lines treated with melarsoprol (10(-7) mol/L) exhibited morphologic characteristics of apoptosis. We also observed prominent concentration-dependent downregulation of bcl-2 mRNA after 24 hours of exposure to melarsoprol in WSU-CLL, I83CLL, and JVM-2 cells. Decrease of bcl-2 protein expression was also observed in all three cell lines, whereas As2O3 had no effect on this parameter. We conclude that melarsoprol may inhibit the growth of lymphoid leukemic cell by promoting programmed cell death. Results of these studies suggest that melarsoprol shows promising therapeutic activity in these diseases, and a study to evaluate clinical effects of this drug has been initiated.


Subject(s)
Antineoplastic Agents/toxicity , Arsenic Poisoning , Arsenicals , Melarsoprol/toxicity , Oxides/toxicity , Apoptosis/drug effects , Arsenic Trioxide , Cell Division/drug effects , Cell Line, Transformed , Cell Survival/drug effects , DNA Damage , DNA Fragmentation , Drug Screening Assays, Antitumor , Herpesvirus 4, Human , Humans , Kinetics , Leukemia, Lymphocytic, Chronic, B-Cell , Proto-Oncogene Proteins c-bcl-2/biosynthesis , RNA, Messenger/biosynthesis , Time Factors , Transcription, Genetic/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...