Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
2.
J Clin Oncol ; 42(1): 90-102, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37883734

RESUMO

PURPOSE: Long-term survival in high-risk neuroblastoma (HRNB) is approximately 50%, with mortality primarily driven by relapse. Eflornithine (DFMO) to reduce risk of relapse after completion of immunotherapy was investigated previously in a single-arm, phase II study (NMTRC003B; ClinicalTrials.gov identifier: NCT02395666) that suggested improved event-free survival (EFS) and overall survival (OS) compared with historical rates in a phase III trial (Children Oncology Group ANBL0032; ClinicalTrials.gov identifier: NCT00026312). Using patient-level data from ANBL0032 as an external control, we present new analyses to further evaluate DFMO as HRNB postimmunotherapy maintenance. PATIENTS AND METHODS: NMTRC003B (2012-2016) enrolled patients with HRNB (N = 141) after standard up-front or refractory/relapse treatment who received up to 2 years of continuous treatment with oral DFMO (750 ± 250 mg/m2 twice a day). ANBL0032 (2001-2015) enrolled patients with HRNB postconsolidation, 1,328 of whom were assigned to dinutuximab (ch.14.18) treatment. Selection rules identified 92 NMTRC003B patients who participated in (n = 87) or received up-front treatment consistent with (n = 5) ANBL0032 (the DFMO/treated group) and 852 patients from ANBL0032 who could have been eligible for NMTRC003B after immunotherapy, but did not enroll (the NO-DFMO/control group). The median follow-up time for DFMO/treated patients was 6.1 years (IQR, 5.2-7.2) versus 5.0 years (IQR, 3.5-7.0) for NO-DFMO/control patients. Kaplan-Meier and Cox regression compared EFS and OS for overall groups, 3:1 (NO-DFMO:DFMO) propensity score-matched cohorts balanced on 11 baseline demographic and disease characteristics with exact matching on MYCN, and additional sensitivity analyses. RESULTS: DFMO after completion of immunotherapy was associated with improved EFS (hazard ratio [HR], 0.50 [95% CI, 0.29 to 0.84]; P = .008) and OS (HR, 0.38 [95% CI, 0.19 to 0.76]; P = .007). The results were confirmed with propensity score-matched cohorts and sensitivity analyses. CONCLUSION: The externally controlled analyses presented show a relapse risk reduction in patients with HRNB treated with postimmunotherapy DFMO.


Assuntos
Eflornitina , Neuroblastoma , Criança , Humanos , Eflornitina/efeitos adversos , Pontuação de Propensão , Recidiva Local de Neoplasia/tratamento farmacológico , Neuroblastoma/tratamento farmacológico , Recidiva , Intervalo Livre de Doença
3.
Cancer Rep (Hoboken) ; 5(11): e1616, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35355452

RESUMO

BACKGROUND: Survival for patients with high-risk neuroblastoma (HRNB) remains poor despite aggressive multimodal therapies. AIMS: To study the feasibility and safety of incorporating a genomic-based targeted agent to induction therapy for HRNB as well as the feasibility and safety of adding difluoromethylornithine (DFMO) to anti-GD2 immunotherapy. METHODS: Twenty newly diagnosed HRNB patients were treated on this multicenter pilot trial. Molecular tumor boards selected one of six targeted agents based on tumor-normal whole exome sequencing and tumor RNA-sequencing results. Treatment followed standard upfront HRNB chemotherapy with the addition of the selected targeted agent to cycles 3-6 of induction. Following consolidation, DFMO (750 mg/m2 twice daily) was added to maintenance with dinutuximab and isotretinoin, followed by continuation of DFMO alone for 2 years. DNA methylation analysis was performed retrospectively and compared to RNA expression. RESULTS: Of the 20 subjects enrolled, 19 started targeted therapy during cycle 3 and 1 started during cycle 5. Eighty-five percent of subjects met feasibility criteria (receiving 75% of targeted agent doses). Addition of targeted agents did not result in toxicities requiring dose reduction of chemotherapy or permanent discontinuation of targeted agent. Following standard consolidation, 15 subjects continued onto immunotherapy with DFMO. This combination was well-tolerated and resulted in no unexpected adverse events related to DFMO. CONCLUSION: This study demonstrates the safety and feasibility of adding targeted agents to standard induction therapy and adding DFMO to immunotherapy for HRNB. This treatment regimen has been expanded to a Phase II trial to evaluate efficacy.


Assuntos
Antineoplásicos , Neuroblastoma , Humanos , Eflornitina/efeitos adversos , Projetos Piloto , Quimioterapia de Indução , Estudos Retrospectivos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/genética , Imunoterapia , Antineoplásicos/uso terapêutico , Fatores Imunológicos , Genômica , RNA/uso terapêutico
4.
Biomolecules ; 11(5)2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-34068700

RESUMO

Polyamine spermidine is essential for the proliferation of eukaryotic cells. Administration of polyamine biosynthesis inhibitor α-difluoromethylornithine (DFMO) induces cytostasis that occurs in two phases; the early phase which can be reversed by spermidine, spermine, and some of their analogs, and the late phase which is characterized by practically complete depletion of cellular spermidine pool. The growth of cells at the late phase can be reversed by spermidine and by very few of its analogs, including (S)-1-methylspermidine. It was reported previously (Witherspoon et al. Cancer Discovery 3(9); 1072-81, 2013) that DFMO treatment leads to depletion of cellular thymidine pools, and that exogenous thymidine supplementation partially prevents DFMO-induced cytostasis without affecting intracellular polyamine pools in HT-29, SW480, and LoVo colorectal cancer cells. Here we show that thymidine did not prevent DFMO-induced cytostasis in DU145, LNCaP, MCF7, CaCo2, BT4C, SV40MES13, HepG2, HEK293, NIH3T3, ARPE19 or HT-29 cell lines, whereas administration of functionally active mimetic of spermidine, (S)-1-methylspermidine, did. Thus, the effect of thymidine seems to be specific only for certain cell lines. We conclude that decreased polyamine levels and possibly also distorted pools of folate-dependent metabolites mediate the anti-proliferative actions of DFMO. However, polyamines are necessary and sufficient to overcome DFMO-induced cytostasis, while thymidine is generally not.


Assuntos
Citostáticos/farmacologia , Eflornitina/efeitos adversos , Neoplasias/tratamento farmacológico , Poliaminas/farmacologia , Timidina/farmacologia , Animais , Células Cultivadas , Humanos , Camundongos , Neoplasias/metabolismo , Neoplasias/patologia , Inibidores da Ornitina Descarboxilase/efeitos adversos , Inibidores da Ornitina Descarboxilase/farmacologia
5.
N Engl J Med ; 383(11): 1028-1039, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32905675

RESUMO

BACKGROUND: The efficacy and safety of combination therapy with eflornithine and sulindac, as compared with either drug alone, in delaying disease progression in patients with familial adenomatous polyposis are unknown. METHODS: We evaluated the efficacy and safety of the combination of eflornithine and sulindac, as compared with either drug alone, in adults with familial adenomatous polyposis. The patients were stratified on the basis of anatomical site with the highest polyp burden and surgical status; the strata were precolectomy (shortest projected time to disease progression), rectal or ileal pouch polyposis after colectomy (longest projected time), and duodenal polyposis (intermediate projected time). The patients were then randomly assigned in a 1:1:1 ratio to receive 750 mg of eflornithine, 150 mg of sulindac, or both once daily for up to 48 months. The primary end point, assessed in a time-to-event analysis, was disease progression, defined as a composite of major surgery, endoscopic excision of advanced adenomas, diagnosis of high-grade dysplasia in the rectum or pouch, or progression of duodenal disease. RESULTS: A total of 171 patients underwent randomization. Disease progression occurred in 18 of 56 patients (32%) in the eflornithine-sulindac group, 22 of 58 (38%) in the sulindac group, and 23 of 57 (40%) in the eflornithine group, with a hazard ratio of 0.71 (95% confidence interval [CI], 0.39 to 1.32) for eflornithine-sulindac as compared with sulindac (P = 0.29) and 0.66 (95% CI, 0.36 to 1.24) for eflornithine-sulindac as compared with eflornithine. Among 37 precolectomy patients, the corresponding values in the treatment groups were 2 of 12 patients (17%), 6 of 13 (46%), and 5 of 12 (42%) (hazard ratios, 0.30 [95% CI, 0.07 to 1.32] and 0.20 [95% CI, 0.03 to 1.32]); among 34 patients with rectal or ileal pouch polyposis, the values were 4 of 11 patients (36%), 2 of 11 (18%), and 5 of 12 (42%) (hazard ratios, 2.03 [95% CI, 0.43 to 9.62] and 0.84 [95% CI, 0.24 to 2.90]); and among 100 patients with duodenal polyposis, the values were 12 of 33 patients (36%), 14 of 34 (41%), and 13 of 33 (39%) (hazard ratios, 0.73 [95% CI, 0.34 to 1.52] and 0.76 [95% CI, 0.35 to 1.64]). Adverse and serious adverse events were similar across the treatment groups. CONCLUSIONS: In this trial involving patients with familial adenomatous polyposis, the incidence of disease progression was not significantly lower with the combination of eflornithine and sulindac than with either drug alone. (Funded by Cancer Prevention Pharmaceuticals; ClinicalTrials.gov number, NCT01483144; EudraCT number, 2012-000427-41.).


Assuntos
Polipose Adenomatosa do Colo/tratamento farmacológico , Progressão da Doença , Eflornitina/uso terapêutico , Sulindaco/uso terapêutico , Adulto , Quimioterapia Combinada , Eflornitina/efeitos adversos , Feminino , Humanos , Análise de Intenção de Tratamento , Estimativa de Kaplan-Meier , Masculino , Sulindaco/efeitos adversos , Resultado do Tratamento
6.
PLoS One ; 15(2): e0228996, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32053698

RESUMO

BACKGROUND: The plenteous resistance to and undesirable consequences of the existing antipiroplasmic therapies have emphasized the urgent need for new chemotherapeutics and drug targets for both prophylaxis and chemotherapy. Hydroxyurea (HYD) is an antineoplastic agent with antitrypanosomal activity. Eflornithine (α-difluoro-methyl ornithine, DFMO) is the best choice therapy for the treatment of late-stage Gambian human African trypanosomiasis. METHODS: In this study, the inhibitory and combination efficacy of HYD and DFMO with existing babesicidal drugs (diminazene aceturate (DA), atovaquone (ATV), and clofazimine (CLF)) deoxyribonucleotide in vitro against the multiplication of Babesia and Theileria. As well as, their chemotherapeutic effects were assessed on B. microti strain that infects rodents. The Cell Counting Kits-8 (CCK-8) test was used to examine their cytotoxicity on human foreskin fibroblast (HFF), mouse embryonic fibroblast (NIH/3T3), and Madin-Darby bovine kidney (MDBK) cells. FINDINGS: HYD and DFMO suppressed the multiplication of all tested species (B. bigemina, B. bovis, B. caballi, B. divergens, and T. equi) in a dose-related manner. HFF, NIH/3T3, or MDBK cell viability was not influenced by DFMO at 1000 µM, while HYD affected the MDBK cell viability at EC50 value of 887.5±14.4 µM. The in vitro combination treatments of DFMO and HYD with CLF, DA, and ATV exhibited synergistic and additive efficacy toward all tested species. The in vivo experiment revealed that HYD and DFMO oral administration at 100 and 50 mg/kg inhibited B. microti multiplication in mice by 60.1% and 78.2%, respectively. HYD-DA and DFMO-DA combined treatments showed higher chemotherapeutic efficacy than their monotherapies. CONCLUSION: These results indicate the prospects of HYD and DFMO as drug candidates for piroplasmosis treatment, when combined mainly with DA, ATV, and CLF. Therefore, further studies are needed to combine HYD or DFMO with either ATV or CLF and examine their impact on B. microti infection in mice.


Assuntos
Babesia/efeitos dos fármacos , Eflornitina/efeitos adversos , Eflornitina/farmacologia , Hidroxiureia/efeitos adversos , Hidroxiureia/farmacologia , Theileria/efeitos dos fármacos , Animais , Antineoplásicos , Antiprotozoários/administração & dosagem , Antiprotozoários/farmacologia , Atovaquona/efeitos adversos , Atovaquona/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Clofazimina/efeitos adversos , Clofazimina/farmacologia , Diminazena/efeitos adversos , Diminazena/análogos & derivados , Diminazena/farmacologia , Cães , Prepúcio do Pênis/citologia , Humanos , Masculino , Camundongos , Células NIH 3T3
7.
Sci Rep ; 8(1): 14445, 2018 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-30262852

RESUMO

High risk neuroblastoma (HRNB) accounts for 15% of all pediatric cancer deaths. Despite aggressive therapy approximately half of patients will relapse, typically with only transient responses to second-line therapy. This study evaluated the ornithine decarboxylase inhibitor difluoromethylornithine (DFMO) as maintenance therapy to prevent relapse following completion of standard therapy (Stratum 1) or after salvage therapy for relapsed/refractory disease (Stratum 2). This Phase II single agent, single arm multicenter study enrolled from June 2012 to February 2016. Subjects received 2 years of oral DFMO (750 ± 250 mg/m2 twice daily). Event free survival (EFS) and overall survival (OS) were determined on an intention-to-treat (ITT) basis. 101 subjects enrolled on Stratum 1 and 100 were eligible for ITT analysis; two-year EFS was 84% (±4%) and OS 97% (±2%). 39 subjects enrolled on Stratum 2, with a two-year EFS of 54% (±8%) and OS 84% (±6%). DFMO was well tolerated. The median survival time is not yet defined for either stratum. DFMO maintenance therapy for HRNB in remission is safe and associated with high EFS and OS. Targeting ODC represents a novel therapeutic mechanism that may provide a new strategy for preventing relapse in children with HRNB.


Assuntos
Eflornitina/administração & dosagem , Quimioterapia de Manutenção , Neuroblastoma/tratamento farmacológico , Neuroblastoma/mortalidade , Pré-Escolar , Intervalo Livre de Doença , Eflornitina/efeitos adversos , Feminino , Humanos , Masculino , Taxa de Sobrevida
8.
CNS Oncol ; 7(2): CNS16, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29378419

RESUMO

This review covers the literature between 1989 and 2007 on studies relevant to the neuro-oncology usage of eflornithine (α-difluoromethylornithine), an oral agent that irreversibly inhibits the enzyme ornithine decarboxylase. It covers the use of eflornithine, alone or in combination, to treat high-grade gliomas. In addition, we provide an update on overall survival from The University of Texas MD Anderson Cancer Center Community Clinical Oncology Program and Clinical Trials Data Office that demonstrates a meaningful benefit in overall survival for eflornithine as a single agent and in combination with nitrosourea-based therapies for anaplastic gliomas. We also provide a framework for understanding the basis and study design of the ongoing pivotal, registrational Phase III multicenter trial for recurrent/progressive anaplastic astrocytoma.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Eflornitina/uso terapêutico , Glioma/tratamento farmacológico , Animais , Antineoplásicos/efeitos adversos , Ensaios Clínicos como Assunto , Eflornitina/efeitos adversos , Humanos
9.
BMC Gastroenterol ; 16(1): 87, 2016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27480131

RESUMO

BACKGROUND: Molecular studies suggest inhibition of colorectal mucosal polyamines (PAs) may be a promising approach to prevent colorectal cancer (CRC). Inhibition of ornithine decarboxylase (ODC) using low-dose eflornithine (DFMO, CPP-1X), combined with maximal PA export using low-dose sulindac, results in greatly reduced levels of normal mucosal PAs. In a clinical trial, this combination (compared with placebo) reduced the 3-year incidence of subsequent high-risk adenomas by >90 %. Familial Adenomatous Polyposis (FAP) is characterized by marked up-regulation of ODC in normal intestinal epithelial and adenoma tissue, and therefore PA reduction might be a potential strategy to control progression of FAP-related intestinal polyposis. CPP FAP-310, a randomized, double-blind, Phase III trial was designed to examine the safety and efficacy of sulindac and DFMO (alone or in combination) for preventing a clinically relevant FAP-related progression event in individuals with FAP. METHODS: Eligible adults with FAP will be randomized to: CPP-1X 750 mg and sulindac 150 mg, CPP-1X placebo and sulindac 150 mg, or CPP-1X 750 mg and sulindac placebo once daily for 24 months. Patients will be stratified based on time-to-event prognosis into one of the three treatment arms: best (ie, longest time to first FAP-related event [rectal/pouch polyposis]), intermediate (duodenal polyposis) and worst (pre-colectomy). Stage-specific, "delayed time to" FAP-related events are the primary endpoints. Change in polyp burden (upper and/or lower intestine) is a key secondary endpoint. DISCUSSION: The trial is ongoing. As of February 1, 2016, 214 individuals have been screened; 138 eligible subjects have been randomized to three treatment groups at 15 North American sites and 6 European sites. By disease strata, 26, 80 and 32 patients are included for assessment of polyp burden in the rectum/pouch, duodenal polyposis and pre-colectomy groups, respectively. Median age is 40 years; 59 % are men. The most common reasons for screening failure include minimal polyp burden (n = 22), withdrawal of consent (n = 9) and extensive polyposis requiring immediate surgical intervention (n = 9). Enrollment is ongoing. TRIAL REGISTRATION: This trial is registered at ClinicalTrials.gov ( NCT01483144 ; November 21, 2011) and the EU Clinical Trials Register( EudraCT 2012-000427-41 ; May 15, 2014).


Assuntos
Polipose Adenomatosa do Colo/tratamento farmacológico , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Eflornitina/uso terapêutico , Sulindaco/uso terapêutico , Polipose Adenomatosa do Colo/metabolismo , Adulto , Antineoplásicos/efeitos adversos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Progressão da Doença , Método Duplo-Cego , Neoplasias Duodenais/tratamento farmacológico , Neoplasias Duodenais/metabolismo , Eflornitina/efeitos adversos , Feminino , Humanos , Mucosa Intestinal/metabolismo , Masculino , Poliaminas/antagonistas & inibidores , Poliaminas/metabolismo , Sulindaco/efeitos adversos
10.
PLoS One ; 10(5): e0127246, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26018967

RESUMO

BACKGROUND: Neuroblastoma (NB) is the most common cancer in infancy and most frequent cause of death from extracranial solid tumors in children. Ornithine decarboxylase (ODC) expression is an independent indicator of poor prognosis in NB patients. This study investigated safety, response, pharmacokinetics, genetic and metabolic factors associated with ODC in a clinical trial of the ODC inhibitor difluoromethylornithine (DFMO) ± etoposide for patients with relapsed or refractory NB. METHODS AND FINDINGS: Twenty-one patients participated in a phase I study of daily oral DFMO alone for three weeks, followed by additional three-week cycles of DFMO plus daily oral etoposide. No dose limiting toxicities (DLTs) were identified in patients taking doses of DFMO between 500-1500 mg/m2 orally twice a day. DFMO pharmacokinetics, single nucleotide polymorphisms (SNPs) in the ODC gene and urinary levels of substrates for the tissue polyamine exporter were measured. Urinary polyamine levels varied among patients at baseline. Patients with the minor T-allele at rs2302616 of the ODC gene had higher baseline levels (p=0.02) of, and larger decreases in, total urinary polyamines during the first cycle of DFMO therapy (p=0.003) and had median progression free survival (PFS) that was over three times longer, compared to patients with the major G allele at this locus although this last result was not statistically significant (p=0.07). Six of 18 evaluable patients were progression free during the trial period with three patients continuing progression free at 663, 1559 and 1573 days after initiating treatment. Median progression-free survival was less among patients having increased urinary polyamines, especially diacetylspermine, although this result was not statistically significant (p=0.056). CONCLUSIONS: DFMO doses of 500-1500 mg/m2/day are safe and well tolerated in children with relapsed NB. Children with the minor T allele at rs2302616 of the ODC gene with relapsed or refractory NB had higher levels of urinary polyamine markers and responded better to therapy containing DFMO, compared to those with the major G allele at this locus. These findings suggest that this patient subset may display dependence on polyamines and be uniquely susceptible to therapies targeting this pathway. TRIAL REGISTRATION: Clinicaltrials.gov NCT#01059071.


Assuntos
Eflornitina/farmacologia , Neuroblastoma/tratamento farmacológico , Inibidores da Ornitina Descarboxilase/farmacologia , Fenótipo , Poliaminas/metabolismo , Adolescente , Criança , Pré-Escolar , Eflornitina/efeitos adversos , Eflornitina/farmacocinética , Eflornitina/uso terapêutico , Etoposídeo/efeitos adversos , Etoposídeo/farmacologia , Etoposídeo/uso terapêutico , Feminino , Humanos , Lactente , Masculino , Neuroblastoma/enzimologia , Neuroblastoma/genética , Neuroblastoma/urina , Ornitina Descarboxilase/metabolismo , Inibidores da Ornitina Descarboxilase/efeitos adversos , Inibidores da Ornitina Descarboxilase/farmacocinética , Inibidores da Ornitina Descarboxilase/uso terapêutico , Poliaminas/urina , Recidiva , Segurança , Resultado do Tratamento
11.
Adv Gerontol ; 25(2): 293-300, 2012.
Artigo em Russo | MEDLINE | ID: mdl-23130522
12.
Pharmacol Biochem Behav ; 100(3): 464-73, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22024160

RESUMO

Recent evidence suggests that polyamines putrescine, spermidine and spermine are essential in maintaining normal cellular function. The present study investigated the effects of chronic treatment of difluoromethylornithine (DFMO, 3% in drinking water), a potent inhibitor of putrescine synthesis, for 54 consecutive days on animals'behavior and neurochemical levels in the CA1, CA2/3 and dentate gyrus sub-regions of the hippocampus and the prefrontal cortex. The DFMO group showed performance impairments in the place navigation and the probe test conducted 24 h after the training in the reference memory version of the water maze task, but not in the elevated plus maze, open field, object recognition, cued navigation and the working memory version of the water maze task when compared to the control group (drinking water only). DFMO treatment resulted in approximately 80-90% and 20% of reductions in the putrescine and spermidine levels, respectively, in the four brain regions examined, and a small reduction in agmatine level in the CA2/3, with no effects on spermine, glutamate and γ-aminobutyrate. The DFMO group showed decreased body weight relative to the control one. However, there were no significant differences between groups in the normalized brain, kidney and liver weights. The present study demonstrates that chronic treatment of DFMO depletes putrescine and decreases spermidine levels in the brain, inhibits growth, and impairs spatial learning and memory in the reference memory version of the water maze specifically. These findings merit further investigation to fully understand the functional role of endogenous polyamines in learning and memory.


Assuntos
Eflornitina/efeitos adversos , Inibidores Enzimáticos/efeitos adversos , Hipocampo/efeitos dos fármacos , Deficiências da Aprendizagem/induzido quimicamente , Transtornos da Memória/induzido quimicamente , Inibidores da Ornitina Descarboxilase , Córtex Pré-Frontal/efeitos dos fármacos , Agmatina/metabolismo , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Comportamento Animal/efeitos dos fármacos , Eflornitina/administração & dosagem , Inibidores Enzimáticos/administração & dosagem , Hipocampo/metabolismo , Masculino , Aprendizagem em Labirinto , Memória/efeitos dos fármacos , Especificidade de Órgãos , Córtex Pré-Frontal/metabolismo , Putrescina/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Comportamento Espacial/efeitos dos fármacos , Espermidina/metabolismo , Testes de Toxicidade Crônica
13.
Cancer Prev Res (Phila) ; 1(1): 32-8, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18841250

RESUMO

Preclinical studies of chemoprevention drugs given in combination at low doses show remarkable efficacy in preventing adenomas with little additional toxicities, suggesting a strategy to improve risk to benefit ratios for preventing recurrent adenomas. Three hundred seventy-five patients with history of resected (> or =3 mm) adenomas were randomly assigned to receive oral difluoromethylornithine (DFMO) 500 mg and sulindac 150 mg once daily or matched placebos for 36 months, stratified by use of low-dose aspirin (81 mg) at baseline and clinical site. Follow-up colonoscopy was done 3 years after randomization or off-study. Colorectal adenoma recurrence was compared among the groups with log-binomial regression. Comparing the outcome in patients receiving placebos to those receiving active intervention, (a) the recurrence of one or more adenomas was 41.1% and 12.3% (risk ratio, 0.30; 95% confidence interval, 0.18-0.49; P < 0.001); (b) 8.5% had one or more advanced adenomas, compared with 0.7% of patients (risk ratio, 0.085; 95% confidence interval, 0.011-0.65; P < 0.001); and (c) 17 (13.2%) patients had multiple adenomas (>1) at the final colonoscopy, compared with 1 (0.7%; risk ratio, 0.055; 0.0074-0.41; P < 0.001). Serious adverse events (grade > or =3) occurred in 8.2% of patients in the placebo group, compared with 11% in the active intervention group (P = 0.35). There was no significant difference in the proportion of patients reporting hearing changes from baseline. Recurrent adenomatous polyps can be markedly reduced by a combination of low oral doses of DFMO and sulindac and with few side effects.


Assuntos
Adenoma/prevenção & controle , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Colorretais/prevenção & controle , Eflornitina/administração & dosagem , Sulindaco/administração & dosagem , Adenoma/diagnóstico , Adulto , Idoso , Algoritmos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Colonoscopia , Neoplasias Colorretais/diagnóstico , Método Duplo-Cego , Eflornitina/efeitos adversos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Placebos , Sulindaco/efeitos adversos , Resultado do Tratamento
14.
Cancer Prev Res (Phila) ; 1(7): 514-21, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19139001

RESUMO

A phase III clinical trial assessed the recurrence of adenomatous polyps after treatment for 36 months with difluoromethylornithine (DFMO) plus sulindac or matched placebos. Temporary hearing loss is a known toxicity of treatment with DFMO, thus a comprehensive approach was developed to analyze serial air conduction audiograms. The generalized estimating equation method estimated the mean difference between treatment arms with regard to change in air conduction pure tone thresholds while accounting for within-subject correlation due to repeated measurements at frequencies. Based on 290 subjects, there was an average difference of 0.50 dB between subjects treated with DFMO plus sulindac compared with those treated with placebo (95% confidence interval, -0.64 to 1.63 dB; P = 0.39), adjusted for baseline values, age, and frequencies. In the normal speech range of 500 to 3,000 Hz, an estimated difference of 0.99 dB (-0.17 to 2.14 dB; P = 0.09) was detected. Dose intensity did not add information to models. There were 14 of 151 (9.3%) in the DFMO plus sulindac group and 4 of 139 (2.9%) in the placebo group who experienced at least 15 dB hearing reduction from baseline in 2 or more consecutive frequencies across the entire range tested (P = 0.02). Follow-up air conduction done at least 6 months after end of treatment showed an adjusted mean difference in hearing thresholds of 1.08 dB (-0.81 to 2.96 dB; P = 0.26) between treatment arms. There was no significant difference in the proportion of subjects in the DFMO plus sulindac group who experienced clinically significant hearing loss compared with the placebo group. The estimated attributable risk of ototoxicity from exposure to the drug is 8.4% (95% confidence interval, -2.0% to 18.8%; P = 0.12). There is a <2 dB difference in mean threshold for patients treated with DFMO plus sulindac compared with those treated with placebo.


Assuntos
Pólipos Adenomatosos/prevenção & controle , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Audiometria de Tons Puros , Neoplasias do Colo/prevenção & controle , Audição/efeitos dos fármacos , Método Duplo-Cego , Eflornitina/administração & dosagem , Eflornitina/efeitos adversos , Perda Auditiva/induzido quimicamente , Humanos , Recidiva Local de Neoplasia/prevenção & controle , Sulindaco/administração & dosagem , Sulindaco/efeitos adversos
15.
Cancer Epidemiol Biomarkers Prev ; 13(7): 1250-2, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15247138

RESUMO

Difluoromethylornithine (DFMO) is a potent, irreversible inhibitor of ornithine decarboxylase, the rate-limiting enzyme in the synthesis of polyamines that promote cellular proliferation. DFMO has been tested as a potential cancer therapeutic and chemopreventive agent in clinical trials. Reversible hearing loss is a recognized toxicity of DFMO that usually occurs at doses above 2 g/m(2)/d, and generally when the cumulative dose exceeds 250 g/m(2). In a recently completed Barrett's esophagus chemoprevention trial, a participant developed a 15-dB decrease in hearing at frequencies of 250, 2,000, and 3,000 Hz in the right ear and a > or =20-dB decrease in hearing at 4,000 to 6,000 Hz in the left ear after taking 0.5 g/m(2)/d DFMO for approximately 13 weeks (cumulative dose of 45 g/m(2)). The threshold shifts persisted 7 months after DFMO was discontinued. There was no obvious impact on the participant's clinical hearing, but these findings were consistent with irreversible hearing loss. This is the first case reported of irreversible ototoxicity in a clinical trial participant receiving DFMO and, thus, trial participants should be made aware of this small but important risk.


Assuntos
Antineoplásicos/efeitos adversos , Esôfago de Barrett/prevenção & controle , Eflornitina/efeitos adversos , Perda Auditiva/induzido quimicamente , Antineoplásicos/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Quimioprevenção/efeitos adversos , Eflornitina/administração & dosagem , Humanos , Masculino , Pessoa de Meia-Idade , Inibidores da Ornitina Descarboxilase , Ensaios Clínicos Controlados Aleatórios como Assunto/efeitos adversos
16.
Parasitol Res ; 90 Supp 1: S49-52, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12811548

RESUMO

Eflornithine is the only new molecule registered for the treatment of human African trypanosomiasis over the last 50 years. It is the drug used mainly as a back-up for melarsoprol refractory Trypanosoma brucei gambiense cases. The most commonly used dosage regimen for the treatment of T. b. gambiensesleeping sickness consists of 100 mg kg(-1) body weight at intervals of 6 h for 14 days (150 mg kg(-1) body weight in children) of eflornithine given as short infusions. Its efficacy against Trypanosoma brucei rhodesiense is limited due to the innate lack of susceptibility of this parasite based on a higher ornithine decarboxylase turnover. Adverse drug reactions during eflornithine therapy are frequent and the characteristics are similar to other cytotoxic drugs for the treatment of cancer. Their occurrence and intensity increase with the duration of treatment and the severity of the general condition of the patient. Generally, adverse reactions to eflornithine are reversible after the end of treatment. They include convulsions (7%), gastrointestinal symptoms like nausea, vomiting and diarrhea (10%-39%), bone marrow toxicity leading to anemia, leucopenia and thrombocytopenia (25-50%), hearing impairment (5% in cancer patients) and alopecia (5-10%). The drug arrests embryonic development in mice, rats and rabbits but the extent of excretion into breast milk is unknown. The mean half-life is around 3-4 h and the volume of distribution in the range of 0.35 l kg(-1). Renal clearance is about 2 ml min kg(-1) (i.v.) and accounts for more than 80% of drug elimination. Bioavailability of an orally administered 10 mg kg(-1) dose was estimated at 54%. One of the major determinants of successful treatment seems to be the cerebrospinal fluid drug level reached during treatment, and it was shown that levels above 50 micro mol l(-1) must be reached to attain the consistent clearance of parasites. Based on its trypanostatic rather than trypanocidal mode of action, it is a rather slow-acting drug.


Assuntos
Eflornitina/uso terapêutico , Tripanossomicidas/uso terapêutico , Tripanossomíase Africana/tratamento farmacológico , Animais , Eflornitina/efeitos adversos , Eflornitina/farmacocinética , Eflornitina/farmacologia , Humanos , Tripanossomicidas/efeitos adversos , Tripanossomicidas/farmacocinética , Tripanossomicidas/farmacologia , Trypanosoma brucei gambiense/efeitos dos fármacos , Trypanosoma brucei gambiense/fisiologia , Trypanosoma brucei rhodesiense/efeitos dos fármacos , Trypanosoma brucei rhodesiense/fisiologia
17.
Cancer Epidemiol Biomarkers Prev ; 10(6): 657-61, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11401916

RESUMO

Individuals who receive life-saving organ transplants and the required immunosuppression often develop secondary cancers. One of the most common secondary cancers is nonmelanoma skin cancer in sun-exposed areas. Attempts to prevent these cancers have not been successful. Difluoromethylornithine (DFMO), a suicide inhibitor of ornithine decarboxylase (ODC), is a known experimental cancer prevention agent that is being evaluated in a number of human cancer prevention trials. This report describes a Phase I trial in 18 organ transplant recipients, randomized to 1.0 and 0.5 g of DFMO or a placebo, designed to look at short-term toxicities over 28 days as well as the impact of DFMO on two biological parameters, skin polyamines and 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced ODC activity. Blood levels of DFMO were also measured. The results indicate that DFMO was well tolerated over the 28-day period. The TPA-induced ODC activity in 3-mm skin biopsies was significantly lowered by 80 and 67% at the two dose levels. Polyamine levels were not affected significantly except for putrescine at the 0.5-g level. Blood levels of DFMO were about two times higher than expected, based on our prior pharmacokinetic studies. Our studies indicate that DFMO is a reasonable agent that should be tested further in larger Phase 2b trials in this population as a chemopreventive agent. TPA-induced ODC activity appears to be a relevant intermediate biological assay.


Assuntos
Antineoplásicos/farmacologia , Eflornitina/farmacologia , Transplante de Órgãos , Neoplasias Cutâneas/prevenção & controle , Adulto , Idoso , Antineoplásicos/efeitos adversos , Quimioprevenção , Eflornitina/efeitos adversos , Feminino , Humanos , Imunossupressores/efeitos adversos , Masculino , Pessoa de Meia-Idade , Ornitina Descarboxilase/análise , Ornitina Descarboxilase/metabolismo , Placebos
18.
Laryngoscope ; 111(5): 781-5, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11359155

RESUMO

OBJECTIVES/HYPOTHESIS: Difluoromethylornithine (DFMO) is an anticancer experimental drug that is ototoxic. The objectives of these three experiments were to: 1) determine a dose and dosing schedule of DFMO that produces significant hearing loss (HL) in newborn gerbils; 2) compare the HL level for control and newborn gerbils receiving daily subcutaneous injections of DFMO; and 3) to determine if DFMO-related HL is significantly reversible following discontinuation of DFMO treatment. STUDY DESIGN: Prospective, non-randomized experimental design with placebo controls. METHODS: Click-evoked auditory brainstem response (ABR) testing was performed for 21-day-old Mongolian gerbils following daily subcutaneous injections of DFMO or saline. Three experiments were carried out using different injection schedules and doses of DFMO. In experiment 3, animals were retested at 42 days of age following a 3-week recovery from DFMO. RESULTS: Animals administered an 18-day regimen of DFMO at 1 g/kg per day (from day 3 to day 20) had click thresholds of 25 to 65 dB nHL, whereas animals receiving daily injections of saline had thresholds of 5 to 20 dB nHL. Animals retested after 3 weeks of recovery from DFMO treatment had thresholds ranging from 5 to 20 dB nHL. Differences were statistically significant. CONCLUSIONS: DFMO causes mild to moderate HL in neonatal gerbils that recovers after discontinuation of the drug.


Assuntos
Antineoplásicos/efeitos adversos , Surdez/induzido quimicamente , Eflornitina/efeitos adversos , Animais , Animais Recém-Nascidos , Surdez/fisiopatologia , Eflornitina/toxicidade , Potenciais Evocados Auditivos do Tronco Encefálico , Gerbillinae , Estudos Prospectivos
19.
Clin Cancer Res ; 5(11): 3438-44, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10589756

RESUMO

DFMO (alpha-difluoromethylornithine) is an oral irreversible inhibitor of ornithine decarboxylase, the first rate-limiting enzyme in polyamine synthesis. DFMO has been shown to have antiproliferative effects against several human cancers, and some studies have suggested that DFMO may have pro-apoptotic and anti-invasive properties as well. DFMO is well tolerated with minimal toxicity but has been associated with ototoxicity with prolonged daily administration. We conducted a Phase I/II tolerability, pharmacokinetic, and efficacy study of high-dose DFMO in metastatic breast cancer patients. Twenty-one patients were treated with 4800 mg of DFMO p.o. three times a day for 14 days, followed by a 2-week drug holiday on a 28-day cycle. Urinary polyamine and blood DFMO levels were measured at multiple time points during therapy. High-dose DFMO was well tolerated, and no clinically significant ototoxicity was noted. No patient achieved an objective antitumor response; however, one patient with heavily pretreated liver metastases has achieved stable disease for 18 months to date on DFMO. Putrescine, spermine, and spermidine urinary levels were suppressed with DFMO treatment and remained low during the 2-week drug holiday. High-dose DFMO on a schedule of 2 weeks on treatment followed by 2 weeks off is well tolerated, is not associated with ototoxicity, and leads to sustained suppression of urinary polyamine levels. Although not an active cytotoxic agent for metastatic breast cancer, the intriguing prolonged growth arrest of liver metastases in one patient highlights the potential clinical growth inhibitory properties of DFMO. We believe that DFMO is worthy of study as adjuvant therapy in primary breast cancer patients and as a chemopreventive agent.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Eflornitina/uso terapêutico , Poliaminas/urina , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Neoplasias da Mama/patologia , Eflornitina/efeitos adversos , Eflornitina/farmacocinética , Feminino , Humanos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Putrescina/urina , Receptores de Estrogênio/análise , Espermidina/urina , Espermina/urina , Fatores de Tempo
20.
Clin Cancer Res ; 5(5): 945-51, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10353725

RESUMO

D,L-alpha-difluoromethylornithine (DFMO) was synthesized over 20 years ago. It was hoped that this enzyme-activated, irreversible inhibitor of ornithine decarboxylase, the first enzyme in polyamine synthesis, would be effective as a chemotherapy for hyperproliferative diseases, including cancer and/or infectious processes. DFMO was generally found to exert cytostatic effects on mammalian cells and tissues, and its effectiveness as a therapeutic agent has been modest. DFMO was also found to cause treatment-limiting (but reversible) ototoxicity at high doses. This side effect, along with its minimal therapeutic activity, contributed to the loss of interest by many clinicians in further developing DFMO as a cancer therapeutic agent. However, DFMO was subsequently shown to inhibit carcinogen-induced cancer development in a number of rodent models, and interest in developing this compound as a preventive agent has increased. The rationale for the inhibition of ornithine decarboxylase as a cancer chemopreventive agent has been strengthened in recent years because this enzyme has been shown to be transactivated by the c-myc oncogene in certain cell/tissue types and to cooperate with the ras oncogene in malignant transformation of epithelial tissues. Recent clinical cancer chemoprevention trials, using dose de-escalation designs, indicate that DFMO can be given over long periods of time at low doses that suppress polyamine contents in gastrointestinal and other epithelial tissues but cause no detectable hearing loss or other side effects. Current clinical chemoprevention trials are investigating the efficacy of DFMO to suppress surrogate end point biomarkers (e.g., colon polyp recurrence) of carcinogenesis in patient populations at elevated risk for the development of specific epithelial cancers, including colon, esophageal, breast, cutaneous, and prostate malignancies.


Assuntos
Anticarcinógenos/uso terapêutico , Eflornitina/uso terapêutico , Neoplasias/prevenção & controle , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Anticarcinógenos/efeitos adversos , Anticarcinógenos/toxicidade , Divisão Celular/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Neoplasias do Colo/prevenção & controle , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Eflornitina/efeitos adversos , Eflornitina/toxicidade , Perda Auditiva/induzido quimicamente , Doenças Hematológicas/induzido quimicamente , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/metabolismo , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/prevenção & controle , Inibidores da Ornitina Descarboxilase , Poliaminas/metabolismo , Lesões Pré-Cancerosas/tratamento farmacológico , Roedores , Células Tumorais Cultivadas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA